You are on page 1of 322

hematology education

the education program for the annual congress of the European Hematology Association

ISSN 1872-5503

Education program for the


14th congress of the
European Hematology
Association

Berlin, Germany,
June 4-7, 2009
hematology education
the education program for the annual congress of the European Hematology Association

Education program for the


14th congress of the
European Hematology
Association

Berlin, Germany,
June 4-7, 2009
Copyright Information

©2009 by European Hematology Association. All rights reserved.


ISSN 1872-5503

Hematology Education, the education program of the annual congress of the European Hematology
Association is published by the European Hematology Association (EHA) in one volume per year.

All business correspondence and purchase and reprint requests should be addressed to the European
Hematology Association, Westblaak 71, 3012 KE Rotterdam, The Netherlands; phone: + 31 (0)10 4361
760; fax: + 31 (0)10 4361 817; e-mail: info@ehaweb.org.

Hematology Education 2009 is available on the internet at http://www.ehaweb.org

No part of this publication may be used (as hereinafter defined) in any form or by any means now or here-
after known, electronic or mechanical, without permission in writing from the Owner, the European
Hematology Association. For purpose of this notice, the term “use” includes, but is not limited to, repro-
duction, photocopying, storage in a retrieval system, translation, and educational purposes such as class-
room instruction and clinical and residency training. This publication or any part thereof may be used for
educational purposes at conferences, continuing education courses, and other educational activity, provid-
ed no fee or other compensation is charged therefore. All materials so used must acknowledge the
Owner’s copyright therein as “©2009 by the European Hematology Association.” When requesting the
Owner’s permission to use this publication or any part thereof, please contact the EHA Executive Office,
Westblaak 71, 3012 KE Rotterdam, The Netherlands; phone: +31 (0)10 4361 760; fax: +31 (0)10 4361 817;
e-mail: info@ehaweb.org.

Article Citations
Cite articles in this volume by listing Title, Author(s), Hematology Education, 2009, beginning page num-
ber-ending page number.

Hematology Education Rights and Permissions


For instructions on requesting permission to reprint or to order copies of Hematology Education manu-
scripts, figures or tables. Please follow the Right and Permission guidelines (http:// www.ehaweb.org).
Questions regarding permission for Hematology Education should be directed to Publication
(info@ehaweb.org).

The Owner disclaims responsibility for opinions expressed by the authors.


hematology education
the education program for the annual congress of the European Hematology Association

EHA Executive Board B Huntly, United Kingdom


W Fibbe, President, The Netherlands J Ingerslev, Denmark
R Foà, President Elect, Italy S Izraeli, Israel
E Hellström-Lindberg, Past President, Sweden B Lämmle, Switzerland
H Döhner, Treasurer, Germany L Malcovati, Italy
I Roberts, Secretary, United Kingdom M Manz, Switzerland
R Pieters, The Netherlands
EHA Councilors M Piris, Spain
E Berntorp, Sweden J San Miguel, Spain
C Camaschella, Italy B Schlegelberger, Germany
C Chomienne, France R Schlenk, Germany
U Jäger, Austria M Trneny, Czech Republic
C Lacombe, France A Urbano Ispizua, Spain
J Sierra, Spain A Waage, Norway
R Skoda, Switzerland
I Touw, The Netherlands EHA Education Committee
A Green, Chair, United Kingdom
EHA Local Organizing Committee 14th Congress M Arat, Turkey
R Hehlmann, Congress President, Germany E Berntorp, Sweden
B Dörken, Germany C Chomienne, France
D Niederwieser, Germany C Craddock, United Kingdom
E Thiel, Germany W Fibbe, The Netherlands
E Hellström-Lindberg, Sweden
EHA Scientific Program Committee 14th Congress D Jasmin, France
R Skoda, Chair, Switzerland F Lo Coco, Italy
H Büller, The Netherlands H Servé, Germany
C Chomienne, France
J Cools, Belgium EHA Press Committee 2009
F Lo Coco, Italy A Hagenbeek, Chair, The Netherlands
C Niemeyer, Germany W Fibbe, The Netherlands
I Peake, United Kingdom R Hehlmann, Germany
G Salles, France I Roberts, United Kingdom
M Theobald, The Netherlands R Skoda, Switzerland
S Thein, United Kingdom
EHA Publication Committee
EHA Scientific Program Committee Advisory W Fibbe, Chair, The Netherlands
Board 14th Congress M Cazzola, Italy
N Avent, United Kingdom R Foà, Italy
M Baccarani, Italy C Lacombe, France
C Beaumont, France S McCann, Ireland
E Berntorp, Sweden
J Cornelissen, The Netherlands European Hematology Association
C Craddock, United Kingdom E-mail: info@ehaweb.org,
N Cross, United Kingdom Website: www.ehaweb.org
T Enver, United Kingdom
C Gachet, France
G Gaidano, Italy
P Ghia, Italy
W Hofmann, Germany
Education Book Reviewers

We would like to thank the reviewers of the manuscripts for the Education Book.

M André, Belgium E Hellström-Lindberg, Sweden


N Avent, United Kingdom W Hofmann, Germany
M Baccarani, Italy S Izraeli, Israel
E Berntorp, Sweden M Laffan, United Kingdom
A Biondi, Italy T Lamy, France
M Björkholm, Sweden B Lämmle, Switzerland
F Caligaris-Cappio, Italy F Lo Coco, Italy
A Cohen, United Kingdom H Pahl, Germany
J Cools, Belgium R Pieters, The Netherlands
J Cornelissen, The Netherlands M Piris, Spain
C Craddock, United Kingdom P Rousselot, France
H Einsele, Germany G Salles, France
T Enver, United Kingdom J San Miguel, Spain
W Fibbe, The Netherlands R Schlenk, Germany
G Gaidano, Italy I Roberts, United Kingdom
A Gratwohl, Switzerland S Thein, United Kingdom
A Green, United Kingdom M Theobald, The Netherlands
Word of welcome

On behalf of the EHA Board and the Scientific Program Committee we would like to welcome you to
the beautiful city of Berlin. The EHA Congress is the largest and most comprehensive hematology meet-
ing in Europe with a world-class program of invited speakers and selected abstracts. The Education
Program covers a large spectrum of basic, translational and clinical research in hematology that will be pre-
sented by distinguished internationally recognized speakers. In addition to enjoying the presentations, we
hope you will find the peer-reviewed manuscripts in this Education Book a useful source of information
and references.

Radek Skoda Rüdiger Hehlmann


Chair Scientific Program Committee Congress President

© www.visitberlin.de
hematology education: the education program for the annual congress
of the European Hematology Association - 2009; volume 3, issue 1

Table of Contents

Acute lymphoblastic leukemia Clinical trial design


1-7 Genomic profiling of acute lymphoblastic leukemia 89-95 Designing and interpreting quality-of-life and other
C.G. Mullighan patient-reported outcomes in clinical trials
F. Efficace, M.A. Sprangers
8-12 Genetic and epigenetic programs in MLL-rearranged acute
lymphoblastic leukemia 96-100 The use of genomics in clinical trial design
S.A. Armstrong R. Simon
13-16 The acute lymphoblastic leukemias of Down syndrome 101-105 Do commonly-used clinical trial designs reflect
S. Izraeli clinical reality?
E. Estey

Acute myeloid leukemia


Diffuse large B-cell lymphoma
17-23 Acute myeloid leukemia biology and leukemia stem cells
E. Gudgin, B. Huntly 106-111 Molecular basis of aggressive B-cell non-Hodgkin
lymphomas
24-29 Minimal residual disease detection in acute myeloid E. Hartmann, A. Rosenwald
leukemias
G. Saglio, E. Messa, D. Cilloni 112-117 Using molecular signatures to identify rational therapeutic
targets in diffuse-large B-cell lymphomas
30-37 Risk-adapted treatment for adult acute myeloid leukemia B. Chapuy, M. Shipp
J. Sierra, S. Brunet
118-122 Treatment options for relapsing patients with diffuse large
B cell lymphoma
C. Gisselbrecht
Bleeding disorders
38-43 von Willebrand factor: its role in hemostasis
and thrombosis Hematopoiesis
S.F. De Meyer, K. Vanhoorelbeke
123-127 Anatomy of the hematopoietic stem cell niche during
44-49 Phenotype, genotype and classification development
in von Willebrand disease E. Dzierzak
A. Goodeve
128-132 Population dynamics of hematopoietic stem cells
50-54 Treatment options in inherited von Willebrand disease H. Takizawa, M.G. Manz
A.B. Federici
133-139 Dynamic interactions between the nervous and immune
systems with the microenvironment regulate
hematopoietic stem cell migration and development.
Chronic lymphocytic leukemia The essential roles of SDF-1 and CXCR4
K. Lapid, T. Lapidot
55-60 Cellular origin of chronic lymphocytic leukemia
U. Klein

61-67 Clinical implications of novel biological markers in Hodgkin’s disease


chronic lymphocytic leukemia
R. Rosenquist 140-143 Nodular lymphocyte-predominant Hodgkin’s lymphoma
in children and adults
68-71 Risk-adapted chronic lymphocytic leukemia management J. Landman-Parker
P. Hillmen
144-150 The prognostic role of positron emission tomography
scan in Hodgkin’s lymphoma
A. Gallamini
Chronic myeloid leukemia
151-154 Recent advances in the treatment of early-stage
72-77 Stem cell persistence in chronic myeloid leukemia Hodgkin’s lymphoma
M.W. Deininger D.A. Eichenauer, A. Engert

78-82 Dynamics of mutated clones in chronic myeloid


leukemia patients
A. Hochhaus
Multiple myeloma

83-88 Second generation tyrosine kinase inhibitors: 155-160 Pathobiology, genetics and clinical outcome
which and when? of myeloma
G. Morgan, K. Boyd
H. Kantarjian, J. Cortes

Education program for the 14th congress of the European Hematology Association, Berlin, Germany, June 4-7, 2009
hematology education: the education program for the annual congress
of the European Hematology Association - 2009; volume 3, issue 1

Table of Contents

161-165 Experimental agents knocking at the door of myeloma


treatment Stem cell transplantation
N.C. Munshi
242-249 Immunobiology of reduced intensity conditioning
166-171 Treatment options for myeloma in 2009 in hematopoietic stem cell transplantation
J. Bladé, L. Rosiñol B. Sandmaier, F. Baron

250-255 Reduced intensity conditioning as a platform for


immunotherapy
Myelodysplastic syndromes J. Barrett

172-176 Pathogenesis and basic aspects of myelodysplastic


syndromes 256-260 The role of reduced intensity conditioning in
N. Galili, A. Raza allogeneic hematopoietic stem cell transplantation
A. Gratwohl
177-180 Molecular pathogenesis of myelodysplastic syndromes:
identifying new therapeutic targets based on biology
S.D. Nimer
Thrombosis
181-187 Risk-adapted treatment of myelodysplastic syndromes
M. Cazzola, L. Malcovati 261-265 Genetics of thrombosis
P.H. Reitsma

266-270 Venous thrombosis: from bench to bedside and back


Myeloproliferative disorders T. Baglin

188-191 The role of somatic and hereditary genetic factors 271-275 New anticoagulants
in the pathogenesis of myeloproliferative neoplasms A. Kleinjan, H.R. Büller
R. Kralovics

192-199 Risk adapted approach to the treatment of primary


myelofibrosis Transfusion
J. Mascarenhas, R. Hoffman
276-281 Transfusion policies and practices in alloimmunized sickle
200-207 Interferon in the treatment of myeloproliferative disorders cell patients
J.-J. Kiladjian J.M. Moulds

282-287 Transfusion problems in stem cell transplant patients


D.H. Pamphilon
Pediatric hematological malignancies
288-293 Transfusion issues in neonatal and pediatric hematology
208-213 Myelodysplastic syndromes in children and adolescents
N.L.C. Luban
C.M. Niemeyer

214-220 Pediatric acute myeloid leukemia


G.J.L. Kaspers Uncommon lymphoproliferative disorders
221-226 Pediatric myeloproliferative neoplasms 294-301 Natural killer cell disorders
M.L. Randi, M.C. Putti
G. Semenzato, R. Zambello

302-307 Immunological and clinical features of


T cell large granular lymphocyte leukemia
Red cell disease
E. Matutes
227-232 Red cell membrane disorders
308-313 The treatment of hairy cell leukemia
N. Mohandas
D. Catovsky, M. Else
233-237 Predicting clinical severity in sickle cell anemia
M.H. Steinberg 314 Index of authors
238-241 Diagnosis and management of erythrocytosis
M. F. McMullin

Education program for the 14th congress of the European Hematology Association, Berlin, Germany, June 4-7, 2009
Acute lymphoblastic leukemia

Genomic profiling of acute lymphoblastic leukemia

C.G. Mullighan A B S T R A C T

Until recently, our understanding of the genetic basis of acute lymphoblastic leukemia (ALL) has
Department of Pathology, St Jude
Children’s Research Hospital, relied primarily on the identification of gross chromosomal changes. However, the full repertoire of
Memphis, Tennessee, USA genetic changes involved in leukemogenesis has been incompletely understood. Microarray profiling
of genetic alterations across the genome, coupled with gene expression profiling and cancer gene
resequencing has transformed our understanding of ALL, and has identified new prognostic markers
Hematology Education: and potential pathways for therapeutic intervention. Genetic alterations targeting key cellular path-
the education program ways are common in ALL, and are significantly associated with disease subtype. Notably, genes regu-
for the annual congress of the lating lymphoid development are mutated in over two-thirds of B-progenitor ALL cases, and con-
European Hematology Association tribute to leukemogenesis in experimental models. Specific genetic alterations are associated with
prognosis, notably deletion or sequence mutation of the early lymphoid transcription factor IKAROS
2009;3:1-7 (IKZF1). Alteration of IKZF1 is common in BCR-ABL1 lymphoid leukemia, predicts very poor outcome
in BCR-ABL1 negative leukemia, with recurring new abnormalities at the time of relapse. Future stud-
ies will utilize next-generation sequencing approaches to perform genome-wide analysis of epigentic
alterations, sequence variations, and small RNA expression. Together, this will provide a comprehen-
sive understanding of the genetic alterations in ALL, and enable the development of rationally target-
ed therapies to improve outcome in this disease.

cute lymphoblastic leukemia (ALL) is have identified additional lesions (e.g., dele-

A the commonest childhood cancer, and


although associated with a generally
favorable prognosis,1,2 a substantial minority
tion of ETV6 in B-ALL and CDKN2A/B and
NOTCH1 in T-ALL).15-18 The completion of
the human genome project and development
of children fail therapy and relapse, with a of microarray-based approaches has now
very poor outcome.3,4 ALL is associated with enabled identification of structural genetic
a much worse prognosis in adults, in part abnormalities in ALL on a genome-wide
due to the high frequency of unfavorable scale at high resolution, and this has been
alterations, such as the Philadelphia chromo- exceptionally informative in ALL.
some.5-7 Moreover, with the exception of
imatinib, existing therapies are associated Microarray analysis of genetic alterations in
with substantial short and long-term toxici- acute lymphoblastic leukemia
ties. To improve outcome in both childhood Microarray platforms for analysis of
and adult ALL, novel targeted therapies genetic alterations include bacterial artificial
against rational therapeutic targets are chromosome (BAC) and oligonucleotide
required. arrays,19 in which these probes are either
Identification of suitable pathways for spotted or synthesized directly onto the
therapeutic intervention has been limited by array. BAC arrays reliably detect large alter-
our incomplete understanding of the genetic ations and have been extensively used in
alterations contributing to leukemogenesis. studies of cancer genomics,20-34 however the
ALL is characterized by multiple recurring probes used are large (often over 100 kb),
chromosomal abnormalities detectable on and consequently BAC arrays have a limited
cytogenetic analysis, including karyotypic ability to detect focal DNA copy number
analysis and FISH, including aneuploidy alterations common in ALL.35-37 Oligonu-
(e.g., high hyperdiploidy and hypodiploidy) cleotide arrays use short (20 to 50+)
and translocations (e.g., t(12;21) ETV6- nucleotide probes and can interrogate copy
RUNX1, t(1;19) TCF3-PBX1, t(9;22) BCR- number alterations at extremely high resolu-
ABL1 and MLL rearrangement in B-progeni- tion. Two main designs are most widely
tor ALL), and rearrangement of the TLX1, used: two color oligonucleotide array-CGH
TLX3, LYL1, TAL1 and MLL genes in platforms (e.g., Agilent, Roche Nimblegen),
T-ALL.8-12 However, several of these alter- and single channel arrays that genotype sin-
ations may be detected for years prior to the gle nucleotide polymorphisms (SNPs; e.g.,
onset of leukemia, and commonly do not Affymetrix, Illumina).38,39 Both provide high
alone induce leukemia in experimental mod- quality, high-resolution data, but SNP array
els, 13,14 suggesting that additional genetic or platforms combine measurement of DNA
epigenetic alterations must contribute to copy number state in addition to SNP geno-
leukemogenesis. Candidate gene studies typing. This allows the detection of both

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 1 |
14th Congress of the European Hematology Association

copy number alterations and copy-neutral loss of het- regions of recurring genetic alterations targeting critical
erozygosity (copy-neutral LOH, also referred to as cellular pathways, including lymphoid differentiation,
acquired uniparental disomy) that may indicate redupli- cell cycle, apoptosis, tumor suppression, oncogenes,
cation of a mutated or aberrantly methylated cancer lymphoid signaling, and drug responsiveness.37,45-49 In a
gene in the region of copy-neutral LOH. The majority of study of 192 B-progenitor and 50 T-lineage ALL sam-
published analyses in ALL have reported acquired ples, we used three Affymetrix SNP arrays examining
genetic changes; however, oligonucleotide arrays are over 350,000 markers at a resolution of approximately 5
also being used to investigate the role of inherited kb.37 This identified over 50 recurring regions of copy
genetic variants, including copy number polymor- number alterations. An important finding was that,
phisms and SNP genotypes, in tumor susceptibility, unlike many solid tumors, large genomic abnormalities
treatment responsiveness, and outcome. and high level amplification were uncommon, with the
The first studies profiling genome-wide copy number exception of the large whole and sub chromosomal
alterations in ALL utilized BAC arrays, and demonstrat- gains characteristic of hyperdiploid ALL and unbalanced
ed the ability of this approach to identify sub-micro- translocations (e.g., 1q+ in TCF3-PBX1 ALL). The
scopic lesions,21,22,24-34,40 including deletions of ETV6, dele- majority of recurring copy number alterations were
tion of CDKN2A/B and 9q34 duplication in T-ALL. focal (less than 1 Mb), commonly with a single or few
They also mapped complex structural changes, such as genes in the minimal region of deletion. Few focal gains
translocation breakpoints24,29 and large amplifications were seen, notable exceptions including amplification
(e.g., intrachromosomal amplification of chromosome of MYB/AHI1, and gains flanking the NUP214-ABL1
21).24,25,27,29 The first report of SNP array analysis of ALL translocation at 9q in T-lineage ALL. The recurring dele-
examined ten cases, and identified multiple regions of tions identified targeted lymphoid development (dis-
LOH, most commonly CDKN2A/B.41 cussed below), tumor suppressors (NF1, PTEN, RB1,
Although these studies provided valuable proof of the ATM), apoptosis regulators (BTG1), putative lymphoid
utility of genome-wide profiling of ALL, a limitation signaling molecules (BTLA/CD200, TOX), micro-RNAs
was the low resolution of both array platforms used, (mir-15/16), steroid receptors (NR3C1, NR3C2), and a
which prevented comprehensive identification of copy number of genes with unknown function in oncogene-
number alterations. In the last four years, there has been sis and leukemia (C20orf94/MKKS, ADD3, DMD).
a dramatic increase in the feature density, and conse- This study examined the principle recurring cytoge-
quently in the resolution, of oligonucleotide arrays. netic subtypes of ALL (high hyperdiploid, hypodiploid,
Most reported SNP array studies in ALL have used ETV6-RUNX1, TCF3-PBX1, BCR-ABL1, MLL-
arrays incorporating 100,000 to 500,000 probes, with an rearranged, and normal/miscellaneous karyotype) and
average intermarker resolution of 5 to 25 kb. Current observed significant differences in the frequency and
platforms employ millions of probes: the SNP 6.0 plat- nature of copy number alterations across ALL subtypes.
form (Affymetrix) incorporates approximately 900,000 Overall, approximately six lesions per case were seen,
SNP probes, and 900,000 non-SNP copy number probes. suggesting that gross genomic instability is not present
Copy number data can be extracted from both probe in the majority of cases. Less than one deletion per case
types and combined to identify copy number alterations was observed in MLL-rearranged ALL, suggesting that
at sub kilobase resolution. Current oligonucleotide few additional genetic alterations are required to induce
array-CGH platforms employ over two million probes this disease. In contrast, both ETV6-RUNX1 and BCR-
(e.g., Roche NimbleGen). It is important to note that the ABL1 ALL harbored over six lesions per case, many of
SNP array platform was initially designed as a genotyp- which were recurring and distinct between these sub-
ing tool to facilitate genetic association studies, and the types, suggesting an absolute requirement for these
distribution of markers is not even across the genome, lesions in the pathogenesis of each leukemia. Deletions
resulting in suboptimal coverage of many genes, partic- involving ADD3, C20orf94, ERG, ETV6, the fragile histi-
ularly with lower resolution arrays.42 Assessment of dine triad gene FHIT, TBL1XR1, a histone cluster at
tumor DNA copy number status requires careful bioin- 6p22.22 were common in B-ALL but rare in T-ALL,
formatic analysis, in order to: (i) account for gross genet- whereas CDKN2A/B deletion was more frequent in T-
ic abnormalities, such as aneuploidy, during the normal- ALL (72%) than B-ALL (34%).37
ization process; (ii) accurately and robustly identify all A key finding was alteration of genes regulating lym-
lesions, many of which are very focal and may be phoid development in over 40% of B-progenitor ALL.
missed by visual inspection of large genomic regions; Genes involved included the transcription factors PAX5
(iii) accurately distinguish inherited from somatic copy (one third of cases), the IKAROS family (IKZF1, IKZF2,
number variants. We and others have developed and IKZF3), early B cell factor (EBF1), TCF3, LEF1, the
adapted tools to perform these analyses.37,43,44 recombinase activating genes RAG1/2, B-cell linker
Consequently, the choice of analysis approach should (BLNK) and the pre-B cell receptor gene VPREB1.
be carefully considered when performing and interpret- Several observations suggest that these alterations are
ing SNP array studies in cancer. important in leukemogenesis. B-progenitor ALL is char-
acterized by a block in differentiation at the pro- to pre-
Genetic alterations at diagnosis in acute lymphoblastic B cell stage of development, and the genes targeted by
leukemia copy number alterations are most commonly transcrip-
Several groups have now performed genome-wide tion factors required for B-cell commitment and differ-
profiling of copy number alterations and LOH in entiation. Targeted deletion of Pax5 and Ebf1 in mice
leukemic samples obtained at diagnosis in B-progenitor results in arrested lymphoid development at this pro- to
and T-lineage ALL, and have identified multiple novel pre-B stage of development. Furthermore, PAX5, which

| 2 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

is required for normal B-lymphoid commitment and dif-


ferentiation50-53 is targeted by deletions, focal internal
amplification, sequence mutations, and novel transloca-
tions,48,54-59 each of which impair the normal transactivat-
ing function of PAX5 in vitro.37 Furthermore, the frequen-
cy and pattern of lesions in this pathway is again asso-
ciated with ALL subtype. All hypodiploid ALL cases
have PAX5 deletions, and commonly concomitant PAX5
point mutations, as well as other lesions in the B cell
development pathway. Notably, hypodiploidy is associ-
ated with poor outcome, and studies in other cohorts
have observed that an increasing number of lesions tar-
geting B lymphoid development are associated with
poor outcome.60 Furthermore, deletion of IKZF1
(IKAROS) is a near obligate lesion in BCR-ABL1 lym-
phoid leukemia,46 and is associated with poor prognosis
in BCR-ABL1 negative ALL, suggesting a central role of
IKAROS in leukemogenesis and responsiveness to ther-
apy.60 Finally, experimental modeling of individual genes
in the B-cell development pathway has established a
role of these lesions in leukemogenesis. In a retroviral
bone marrow transplant model of BCR-ABL1 ALL, Pax5
haploinsufficiency increases the penetrance and reduces
the latency of ALL.61 Similarly, following chemical or
retroviral mutagenesis, Pax5 haploinsufficiency dramat-
ically increases the penetrance of B-ALL.62 In both mod-
els, the development of leukemia is accompanied by the
acquisition of additional genetic changes also observed
in the analogous human leukemias, such as deletion of
Cdkn2a/b, and deletion or sequence mutation of the sec-
ond Pax5 allele, suggesting that these lesions are
required for acquisition of the full leukemic phenotype.

Genome wide profiling of T-lineage acute lymphoblastic


leukemia Figure 1. DNA copy number alterations in ALL. (A) Log2-
ratio copy number heatmap of Affymetrix SNP array wide
Genome wide profiling has also been revealing in T- copy number data in 126 representative pediatric B-pro-
lineage ALL. BAC and oligonucleotide arrays have been genitor and T-lineage ALL cases. Blue is deletion, red is
gain. (B) Copy number heatmap of 61 pediatric ALL cases
used to identify copy number alterations accompanying with copy number alterations involving PAX5 at 9p13.2.
novel structural abnormalities, such as the NUP214- Data is taken from ALL patients reported in (37), and the
ABL1 and SET-NUP214 fusions,63,64 and focal copy num- figure is reproduced with permission from Seminars in
ber alterations involving and dysregulating TAL1, Hematology 2009;46:3-15, Copyright Elsevier.
LMO2, RB1, PTEN, FBXW7 and MYB.37,65-70 Recently, a
novel subtype was of T-ALL was identified with aber-
rant immunophenotype reminiscent of early thymic
progenitors and very poor outcome.71 These atypical T-
ALL cases have an unusually high burden of copy num-
ber alterations on SNP array analysis suggestive of
genomic instability, although the underlying genetic
defect remains to be identified.

Genomic analysis of BCR-ABL1 leukemia


An important area of research is the use of genomic
approaches to identify novel genetic alterations con-
Figure 2. Mutations in genes regulating B-lymphoid devel-
tributing to poor outcome in ALL. One example is opment in B-progenitor ALL. The differentiation of
leukemia associated with the constitutively active hematopoietic stem cells to mature B cells is regulated by
kinase, BCR-ABL1, which is a hallmark of chronic a controlled hierarchy of transcription factors that enforce
B-lineage commitment and differentiation, and suppress
myeloid leukemia (CML) and which typically responds differentiation to alternate (T-lymphoid, myeloid) lineages.
favorably to tyrosine kinase inhibitors,72 and BCR-ABL1 These transcription factors are targeted by genetic alter-
de novo ALL, which has a poor prognosis.73 The mecha- ations in over 60% of B-progenitor ALL cases.37,46,60 Notably,
nisms responsible for these distinct biologic and clinical the genetic alterations affect transcription factors control-
ling differentiation up to the pre-B cell stage of develop-
phenotypes are incompletely understood, and previous- ment, at which differentiation is arrested in many B-ALL
ly have been attributed, at least in part, to the BCR- cases. Additional known or putative B-lymphoid regulatory
ABL1 isoform (p185/p190 v. p210)74 and the lineage and genes targeted less frequently in ALL include BLNK, IKZF2,
IKZF3, MEF2C, RAG1/2 and SOX4.
maturational stage of the leukemia initiating cell.75,76

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 3 |
14th Congress of the European Hematology Association

SNP array studies of ALL and CML incorporating 43 tional finding was that the poor outcome, IKZF1 mutat-
BCR-ABL1 de novo ALL cases, and 128 samples obtained ed BCR-ABL1 negative cases in this study shared a sim-
from 90 CML patients at key stages of disease, includ- ilar gene expression profile to BCR-ABL1 positive ALL.
ing chronic phase (N=64), accelerated phase (N=15) and A similar subtype of “BCR-ABL1-like” childhood ALL
blast crisis (22 myeloid and 9 lymphoid), have identified cases with poor outcome has also been recently report-
lesions significantly associated with lineage and disease ed by den Boer et al.88 This suggests that IKZF1 is an
progression in BCR-ABL1 leukemia.77,78 Notably, few important determinant of the biology of both BCR-
copy number alterations are present in chronic phase ABL1 positive and negative disease, and suggests that
CML, and the only recurring lesions are deletions or BCR-ABL1 negative cases have hitherto unidentified
gains involving the breakpoints of BCR and ABL1. In kinase mutations. Ongoing efforts profiling genomic
contrast, deletion of IKZF1 (IKAROS) is very common alterations in adult ALL, which has a substantially
in de novo BCR-ABL1 ALL, and at the progression of worse outcome than pediatric ALL, will be of great
CML to lymphoid blast crisis, but not chronic phase interest.
CML.78 Moreover, serial analysis of CML samples The studies described above demonstrate that genet-
demonstrated that progression to lymphoid blast crisis ic alterations present in diagnosis ALL samples are
is accompanied by the acquisition of deletions also seen important in leukemogenesis, and predict risk of subse-
at similar frequency in de novo BCR-ABL1 ALL, including quent relapse. A logical next step was to compare the
deletions of CDKN2A/B, PAX5, C20orf94, RB1, MEF2C genetic alterations present at relapse with those present
and EBF1.78-79 at the time of diagnosis. We analyzed 61 matched diag-
The high frequency of IKZF1 deletion in BCR-ABL1 nosis-relapse B-progenitor and T-lineage ALL cases, and
ALL (36 of 43 cases) is notable, as prior studies had demonstrated that over 92% of patients exhibited dif-
observed a high frequency of expression of aberrant ferences in the patterns of copy number alterations from
IKZF1 transcripts in ALL.80-86 IKZF1 has two zinc finger diagnosis to relapse. Importantly, profiling of the pat-
domains: N-terminal zinc fingers that mediate DNA- tern of deletions at antigen receptor loci demonstrated a
binding, and C-terminal zinc fingers that mediate common clonal origin of the diagnosis and relapse sam-
dimerization.87 One third of ALL cases with IKZF1 dele- ples.89 Of these cases, only one third showed simple
tion have a focal lesion involving the N-terminal zinc clonal evolution, with retention of all copy number
fingers. This results in expression of an internally trun- alterations detected at diagnosis, and the acquisition of
cated isoform, Ik6, that cannot bind DNA yet retains new lesions. Over 50% of cases acquired new lesions
the C-terminal zinc fingers and may still dimerize, and and lost copy number alterations present at diagnosis,
act as a dominant negative isoform. The high frequency but had a common clonal origin. Moreover, backtrack-
of expression of this isoform in BCR-ABL1 ALL and ing of relapse-acquired lesions using genomic PCR
lymphoid blast crisis CML suggests a central role in the showed that in most cases, the relapse clone was pres-
pathogenesis and poor outcome of these diseases. ent at diagnosis, but at low levels not detectable using
the SNP arrays. This has provided important insights
Genomic profiling of high-risk leukemia into the mechanism of relapse. Specifically, many
Relapse is most frequent in specific ALL subtypes, patients harbor multiple clones at the time of diagnosis,
including BCR-ABL1 positive, MLL-rearranged and and the lesions present in each clone confer resistance to
hypodiploid ALL, but is seen across the spectrum of therapy and promote subsequent relapse. Also, among
ALL. Two approaches have been used to identify the the commonest deletions acquired at the time of relapse
genetic basis of treatment failure: genomic analysis of were those targeting CDKN2A/B and IKZF1 (and relat-
samples obtained at diagnosis from children with high- ed IKAROS family members), again suggesting that
risk ALL, and comparative profiling of matched diagno- these lesions have important roles in resistance to ther-
sis and relapsed ALL samples. apy.
In a study that incorporated SNP array profiling, gene
expression analysis, and candidate gene resequencing in Future directions
a cohort of 221 children with high-risk ALL (the High resolution, genome wide profiling of genetic
Children’s Oncology Group P9906 cohort, that exclud- alterations using microarray methodologies has provid-
ed known high-risk subtypes, such as BCR-ABL1 and ed important insights into the mechanisms of leukemo-
hypodiploid ALL), we identified a high significant asso- genesis and therapeutic resistance in ALL that had pre-
ciation between deletion or sequence mutation of viously been unsuspected from low-resolution genetic
IKZF1 and poor outcome.60 Children with IKZF1 alter- approaches. It is likely that ongoing genomic profiling
ation had a five-year cumulative incidence of relapse of studies will continue to provide valuable insights. The
73.4% compared to 25% without IKZF1 alteration. ability to identify copy number alterations using
This was confirmed in a validation cohort of St Jude microarray approaches is critically dependent on array
childhood ALL cases. Furthermore, IKZF1 alteration design and resolution. Notably, several key lesions in
was significantly associated with high levels of minimal ALL, such as deletion of IKZF1, are poorly detected
residual disease (MRD) early in therapy in both P9906 using arrays that incorporate as many as 250,000
and St Jude cohorts. Moreover, IKZF1 alteration probes, and are only robustly detected using newer,
remained significantly associated with poor outcome in multimillion feature arrays.90 Consequently, the full
multivariable analyses incorporating age, leukocyte repertoire of copy number alterations in ALL remains to
count, subtype and levels of MRD, suggesting that be defined and tiling analysis of leukemia genomes is
detection of IKZF1 alteration will be useful in the initial required. Furthermore, these data have examined only
management of newly diagnosed ALL cases. An addi- type of genetic alteration in a genome wide fashion.

| 4 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

Given the tremendous insights gained from analysis of 17. Okuda T, Shurtleff SA, Valentine MB, Raimondi SC, Head DR,
Behm F, et al. Frequent deletion of p16INK4a/MTS1 and
copy number alterations, it is now logical to perform p15INK4b/MTS2 in pediatric acute lymphoblastic leukemia.
genome-wide analysis of sequence variation, epigenetic Blood 1995;85:2321-30.
changes, and gene expression, and this is a major focus 18. Weng AP, Ferrando AA, Lee W, Morris JPt, Silverman LB,
Sanchez-Irizarry C, et al. Activating mutations of NOTCH1 in
of many cancer genomics projects. Acquisition, analy- human T cell acute lymphoblastic leukemia. Science 2004;
sis, and integration of these massive datasets will be 306:269-71.
challenging, but is becoming attainable with the rapid 19. Davies JJ, Wilson IM, Lam WL. Array CGH technologies and
their applications to cancer genomes. Chromosome Res 2005;
advances in next generation sequencing technologies 13:237-48.
that may eventually supplant microarray analysis of 20. Pinkel D, Albertson DG. Comparative genomic hybridization.
genetic alterations.91-93 Together, these studies offer the Annu Rev Genomics Hum Genet 2005;6:331-54.
promise of new targets, new therapies, and improved 21. Larramendy ML, Huhta T, Heinonen K, Vettenranta K,
Mahlamaki E, Riikonen P, et al. DNA copy number changes in
outcome in ALL. childhood acute lymphoblastic leukemia. Haematologica
1998;83:890-5.
22. Larramendy ML, Huhta T, Vettenranta K, El-Rifai W, Lundin J,
Pakkala S, et al. Comparative genomic hybridization in child-
References hood acute lymphoblastic leukemia. Leukemia 1998;12:1638-
44.
1. Pui CH, Robison LL, Look AT. Acute lymphoblastic 23. Huhta T, Vettenranta K, Heinonen K, Kanerva J, Larramendy
leukaemia. Lancet 2008;371:1030-43. ML, Mahlamaki E, et al. Comparative genomic hybridization
2. Pui CH, Sandlund JT, Pei D, Campana D, Rivera GK, Ribeiro and conventional cytogenetic analyses in childhood acute
RC, et al. Improved outcome for children with acute lym- myeloid leukemia. Leuk Lymphoma 1999;35:311-5.
phoblastic leukemia: results of Total Therapy Study XIIIB at St 24. Schoumans J, Johansson B, Corcoran M, Kuchinskaya E, Golo-
Jude Children's Research Hospital. Blood 2004;104:2690-6. vleva I, Grander D, et al. Characterisation of dic(9;20)(p11-
3. Einsiedel HG, von Stackelberg A, Hartmann R, Fengler R, 13;q11) in childhood B-cell precursor acute lymphoblastic
Schrappe M, Janka-Schaub G, et al. Long-term outcome in leukaemia by tiling resolution array-based comparative
children with relapsed ALL by risk-stratified salvage therapy: genomic hybridisation reveals clustered breakpoints at 9p13.2
results of trial acute lymphoblastic leukemia-relapse study of and 20q11.2. Br J Haematol 2006;135:492-9.
the Berlin-Frankfurt-Munster Group 87. J Clin Oncol 2005;23: 25. Strefford JC, van Delft FW, Robinson HM, Worley H,
7942-50. Yiannikouris O, Selzer R, et al. Complex genomic alterations
4. Rivera GK, Zhou Y, Hancock ML, Gajjar A, Rubnitz J, Ribeiro and gene expression in acute lymphoblastic leukemia with
RC, et al. Bone marrow recurrence after initial intensive treat- intrachromosomal amplification of chromosome 21. Proc Natl
ment for childhood acute lymphoblastic leukemia. Cancer
Acad Sci USA 2006;103:8167-72.
2005;103:368-76.
5. Gleissner B, Gokbuget N, Bartram CR, Janssen B, Rieder H, 26. van Vlierberghe P, Meijerink JP, Lee C, Ferrando AA, Look AT,
Janssen JW, et al. Leading prognostic relevance of the BCR- van Wering ER, et al. A new recurrent 9q34 duplication in
ABL translocation in adult acute B-lineage lymphoblastic pediatric T-cell acute lymphoblastic leukemia. Leukemia
leukemia: a prospective study of the German Multicenter Trial 2006;20:1245-53.
Group and confirmed polymerase chain reaction analysis. 27. Davidsson J, Andersson A, Paulsson K, Heidenblad M, Isak-
Blood 2002;99:1536-43. sson M, Borg A, et al. Tiling resolution array comparative
6. Fielding AK, Richards SM, Chopra R, Lazarus HM, Litzow genomic hybridization, expression and methylation analyses
MR, Buck G, et al. Outcome of 609 adults after relapse of of dup(1q) in Burkitt lymphomas and pediatric high hyper-
acute lymphoblastic leukemia (ALL); an MRC UKALL12 diploid acute lymphoblastic leukemias reveal clustered near-
/ECOG 2993 study. Blood 2007;109:944-50. centromeric breakpoints and overexpression of genes in 1q22-
7. Rowe JM, Buck G, Burnett AK, Chopra R, Wiernik PH, 32.3. Hum Mol Genet 2007;16:2215-25.
Richards SM, et al. Induction therapy for adults with acute 28. Kuchinskaya E, Nordgren A, Heyman M, Schoumans J,
lymphoblastic leukemia: results of more than 1500 patients Corcoran M, Staaf J, et al. Tiling-resolution array-CGH reveals
from the international ALL trial: MRC UKALL XII/ECOG the pattern of DNA copy number alterations in acute lym-
E2993. Blood 2005;106:3760-7. phoblastic leukemia with 21q amplification: the result of
8. Harrison CJ, Foroni L. Cytogenetics and molecular genetics of telomere dysfunction and breakage/fusion/breakage cycles?
acute lymphoblastic leukemia. Rev Clin Exp Hematol 2002; Leukemia 2007;21:1327-30.
6:91-113. 29. Lundin C, Heidenblad M, Strombeck B, Borg A, Hovland R,
9. Pui CH, Relling MV, Downing JR. Acute lymphoblastic Heim S, et al. Tiling resolution array CGH of dic(7;9)(p11
leukemia. N Engl J Med 2004;350:1535-48. approximately 13;p11 approximately 13) in B-cell precursor
10. Raimondi SC. Cytogenetics of acute leukemias. In: Pui CH, acute lymphoblastic leukemia reveals clustered breakpoints at
editor. Childhood Leukemias. 2nd ed. Cambridge (England): 7p11.2 approximately 12.1 and 9p13.1. Cytogenet Genome
Cambridge University Press; 2006. p. 235-71. Res 2007;118:13-8.
11. Graux C, Cools J, Michaux L, Vandenberghe P, Hagemeijer A. 30. Strefford JC, Worley H, Barber K, Wright S, Stewart AR,
Cytogenetics and molecular genetics of T-cell acute lym- Robinson HM, et al. Genome complexity in acute lym-
phoblastic leukemia: from thymocyte to lymphoblast. phoblastic leukemia is revealed by array-based comparative
Leukemia 2006;20:1496-510. genomic hybridization. Oncogene 2007;26:4306-18.
12. Harrison CJ. Cytogenetics of paediatric and adolescent acute 31. Jalali GR, An Q, Konn ZJ, Worley H, Wright SL, Harrison CJ,
lymphoblastic leukaemia. Br J Haematol 2009;144:147-56. et al. Disruption of ETV6 in intron 2 results in upregulatory
13. Andreasson P, Schwaller J, Anastasiadou E, Aster J, Gilliland and insertional events in childhood acute lymphoblastic
DG. The expression of ETV6/CBFA2 (TEL/AML1) is not suffi- leukaemia. Leukemia 2008;22:114-23.
cient for the transformation of hematopoietic cell lines in vitro 32. Kuchinskaya E, Heyman M, Nordgren A, Schoumans J, Staaf
or the induction of hematologic disease in vivo. Cancer Genet J, Borg A, et al. Array-CGH reveals hidden gene dose changes
Cytogenet 2001;130:93-104. in children with acute lymphoblastic leukaemia and a normal
14. Williams RT, Roussel MF, Sherr CJ. Arf gene loss enhances or failed karyotype by G-banding. Br J Haematol 2008;140:
oncogenicity and limits imatinib response in mouse models of 572-7.
Bcr-Abl-induced acute lymphoblastic leukemia. Proc Natl 33. Rabin KR, Man TK, Yu A, Folsom MR, Zhao YJ, Rao PH, et al.
Acad Sci USA 2006;103:6688-93. Clinical utility of array comparative genomic hybridization for
15. Hebert J, Cayuela JM, Berkeley J, Sigaux F. Candidate tumor- detection of chromosomal abnormalities in pediatric acute
suppressor genes MTS1 (p16INK4A) and MTS2 (p15INK4B) lymphoblastic leukemia. Pediatr Blood Cancer 2008;51:171-7.
display frequent homozygous deletions in primary cells from 34. Steinemann D, Cario G, Stanulla M, Karawajew L, Tauscher
T- but not from B-cell lineage acute lymphoblastic leukemias. M, Weigmann A, et al. Copy number alterations in childhood
Blood 1994;84:4038-44. acute lymphoblastic leukemia and their association with min-
16. Ogawa S, Hangaishi A, Miyawaki S, Hirosawa S, Miura Y, imal residual disease. Genes Chromosomes Cancer 2008;47:
Takeyama K, et al. Loss of the cyclin-dependent kinase 4- 471-80.
inhibitor (p16; MTS1) gene is frequent in and highly specific to 35. Hehir-Kwa JY, Egmont-Petersen M, Janssen IM, Smeets D, van
lymphoid tumors in primary human hematopoietic malignan- Kessel AG, Veltman JA. Genome-wide copy number profiling
cies. Blood 1995;86:1548-56. on high-density bacterial artificial chromosomes, single-

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 5 |
14th Congress of the European Hematology Association

nucleotide polymorphisms, and oligonucleotide microarrays: 56. Nebral K, Konig M, Harder L, Siebert R, Haas OA, Strehl S.
a platform comparison based on statistical power analysis. Identification of PML as novel PAX5 fusion partner in child-
DNA Res 2007;14:1-11. hood acute lymphoblastic leukaemia. Br J Haematol 2007;139:
36. Lo KC, Bailey D, Burkhardt T, Gardina P, Turpaz Y, Cowell JK. 269-74.
Comprehensive analysis of loss of heterozygosity events in 57. Bousquet M, Broccardo C, Quelen C, Meggetto F, Kuhlein E,
glioblastoma using the 100K SNP mapping arrays and compar- Delsol G, et al. A novel PAX5-ELN fusion protein identified in
ison with copy number abnormalities defined by BAC array B-cell acute lymphoblastic leukemia acts as a dominant nega-
comparative genomic hybridization. Genes Chromosomes tive on wild-type PAX5. Blood 2007;109:3417-23.
Cancer 2008;47:221-37. 58. An Q, Wright SL, Konn ZJ, Matheson E, Minto L, Moorman
37. Mullighan CG, Goorha S, Radtke I, Miller CB, Coustan-Smith AV, et al. Variable breakpoints target PAX5 in patients with
E, Dalton JD, et al. Genome-wide analysis of genetic alter- dicentric chromosomes: a model for the basis of unbalanced
ations in acute lymphoblastic leukaemia. Nature 2007;446: translocations in cancer. Proc Natl Acad Sci USA 2008;105:
758-64. 17050-4.
38. Matsuzaki H, Dong S, Loi H, Di X, Liu G, Hubbell E, et al. 59. Nebral K, Denk D, Attarbaschi A, Konig M, Mann G, Haas
Genotyping over 100,000 SNPs on a pair of oligonucleotide OA, et al. Incidence and diversity of PAX5 fusion genes in
arrays. Nat Methods 2004;1:109-11. childhood acute lymphoblastic leukemia. Leukemia 2009;23:
39. Peiffer DA, Le JM, Steemers FJ, Chang W, Jenniges T, Garcia F, 134-43.
et al. High-resolution genomic profiling of chromosomal aber- 60. Mullighan CG, Su X, Zhang J, Radtke I, Phillips LA, Miller CB,
rations using Infinium whole-genome genotyping. Genome et al. Deletion of IKZF1 and Prognosis in Acute Lymphoblastic
Res 2006;16:1136-48. Leukemia. N Engl J Med 2009;360:470-80.
40. Lilljebjorn H, Heidenblad M, Nilsson B, Lassen C, Horvat A, 61. Miller CB, Mullighan CG, Su X, Ma J, Wang M, Zhang J, et al.
Heldrup J, et al. Combined high-resolution array-based com- Pax5 Haploinsufficiency Cooperates with BCR-ABL1 to
parative genomic hybridization and expression profiling of Induce Acute Lymphoblastic Leukemia. Blood 2008;112: 293.
ETV6/RUNX1-positive acute lymphoblastic leukemias reveal 62. Dang J, Mullighan CG, Phillips LA, Mehta P, Downing JR.
a high incidence of cryptic Xq duplications and identify sever- Retroviral and chemical mutagenesis identifies Pax5 as a
al putative target genes within the commonly gained region. tumor suppressor in B-progenitor acute lymphoblastic
Leukemia 2007;21:2137-44. leukemia. Blood 2008;112:1798.
41. Irving JA, Bloodworth L, Bown NP, Case MC, Hogarth LA, 63. Graux C, Cools J, Melotte C, Quentmeier H, Ferrando A,
Hall AG. Loss of heterozygosity in childhood acute lym- Levine R, et al. Fusion of NUP214 to ABL1 on amplified epi-
phoblastic leukemia detected by genome-wide microarray sin- somes in T-cell acute lymphoblastic leukemia. Nat Genet
gle nucleotide polymorphism analysis. Cancer Res 2005;65: 2004;36:1084-9.
3053-8. 64. Van Vlierberghe P, van Grotel M, Tchinda J, Lee C, Beverloo
42. Xavier RJ, Rioux JD. Genome-wide association studies: a new HB, van der Spek PJ, et al. The recurrent SET-NUP214 fusion
window into immune-mediated diseases. Nat Rev Immunol as a new HOXA activation mechanism in pediatric T-cell
2008;8:631-43. acute lymphoblastic leukemia. Blood 2008;111:4668-80.
43. Pounds SB, Cheng C, Mullighan CG, Raimondi SC, Shurtleff 65. Van Vlierberghe P, van Grotel M, Beverloo HB, Lee C,
SA, Downing JR. Reference Alignment of SNP Microarray Helgason T, Buijs-Gladdines J, et al. The cryptic chromosomal
Signals for Copy Number Analysis of Tumors. Bioinformatics. deletion del(11)(p12p13) as a new activation mechanism of
2009;25:315-21. LMO2 in pediatric T-cell acute lymphoblastic leukemia. Blood
44. Olshen AB, Venkatraman ES, Lucito R, Wigler M. Circular 2006;108:3520-9.
binary segmentation for the analysis of array-based DNA 66. Maser RS, Choudhury B, Campbell PJ, Feng B, Wong KK,
copy number data. Biostatistics 2004;5:557-72. Protopopov A, et al. Chromosomally unstable mouse tumours
45. Kuiper RP, Schoenmakers EF, van Reijmersdal SV, Hehir-Kwa have genomic alterations similar to diverse human cancers.
JY, van Kessel AG, van Leeuwen FN, et al. High-resolution Nature 2007;447:966-71.
genomic profiling of childhood ALL reveals novel recurrent 67. Palomero T, Sulis ML, Cortina M, Real PJ, Barnes K, Ciofani
genetic lesions affecting pathways involved in lymphocyte M, et al. Mutational loss of PTEN induces resistance to
differentiation and cell cycle progression. Leukemia 2007; NOTCH1 inhibition in T-cell leukemia. Nat Med 2007;13:
21:1258-66. 1203-10.
46. Mullighan CG, Miller CB, Radtke I, Phillips LA, Dalton J, Ma 68. O'Neil J, Grim J, Strack P, Rao S, Tibbitts D, Winter C, et al.
J, et al. BCR-ABL1 lymphoblastic leukaemia is characterized FBW7 mutations in leukemic cells mediate NOTCH pathway
by the deletion of Ikaros. Nature 2008;453:110-4. activation and resistance to gamma-secretase inhibitors. J Exp
47. Kawamata N, Ogawa S, Zimmermann M, Kato M, Sanada M, Med 2007;204:1813-24.
Hemminki K, et al. Molecular allelokaryotyping of pediatric 69. Clappier E, Cuccuini W, Kalota A, Crinquette A, Cayuela JM,
acute lymphoblastic leukemias by high-resolution single Dik WA, et al. The C-MYB locus is involved in chromosomal
nucleotide polymorphism oligonucleotide genomic microar- translocation and genomic duplications in human T-cell acute
ray. Blood 2008;111:776-84. leukemia (T-ALL), the translocation defining a new T-ALL sub-
48. Kawamata N, Ogawa S, Zimmermann M, Niebuhr B, type in very young children. Blood 2007;110:1251-61.
Stocking C, Sanada M, et al. Cloning of genes involved in 70. Lahortiga I, De Keersmaecker K, Van Vlierberghe P, Graux C,
chromosomal translocations by high-resolution single nu- Cauwelier B, Lambert F, et al. Duplication of the MYB onco-
cleotide polymorphism genomic microarray. Proc Natl Acad gene in T cell acute lymphoblastic leukemia. Nat Genet
Sci USA 2008;105:11921-6. 2007;39:593-5.
49. Bungaro S, Dell'Orto MC, Zangrando A, Basso D, Gorletta T, 71. Coustan-Smith E, Mullighan CG, Onciu M, Behm FG,
Lo Nigro L, et al. Integration of genomic and gene expression Raimondi SC, Pei D, et al. Early T-cell precursor leukaemia: a
data of childhood ALL without known aberrations identifies subtype of very high-risk acute lymphoblastic leukaemia.
subgroups with specific genetic hallmarks. Genes Chromo- Lancet Oncol 2009.
somes Cancer 2009;48:22-38. 72. Goldman JM, Melo JV. Chronic myeloid leukemia--advances
50. Nutt SL, Heavey B, Rolink AG, Busslinger M. Commitment to in biology and new approaches to treatment. N Engl J Med
the B-lymphoid lineage depends on the transcription factor 2003;349:1451-64.
Pax5. Nature 1999;401:556-62. 73. Ribeiro RC, Abromowitch M, Raimondi SC, Murphy SB,
51. Nutt SL, Eberhard D, Horcher M, Rolink AG, Busslinger M. Behm F, Williams DL. Clinical and biologic hallmarks of the
Pax5 determines the identity of B cells from the beginning to Philadelphia chromosome in childhood acute lymphoblastic
the end of B-lymphopoiesis. Int Rev Immunol 2001;2:65-82. leukemia. Blood 1987;70:948-53.
52. Busslinger M. Transcriptional control of early B cell develop- 74. Melo JV. The diversity of BCR-ABL fusion proteins and their
ment. Annu Rev Immunol. 2004;22:55-79. relationship to leukemia phenotype. Blood 1996;88:2375-84.
53. Nutt SL, Kee BL. The transcriptional regulation of B cell line- 75. Huntly BJ, Shigematsu H, Deguchi K, Lee BH, Mizuno S,
age commitment. Immunity. 2007;26:715-25. Duclos N, et al. MOZ-TIF2, but not BCR-ABL, confers proper-
54. Cazzaniga G, Daniotti M, Tosi S, Giudici G, Aloisi A, Pogliani ties of leukemic stem cells to committed murine hematopoiet-
E, et al. The paired box domain gene PAX5 is fused to ic progenitors. Cancer Cell 2004;6:587-96.
ETV6/TEL in an acute lymphoblastic leukemia case. Cancer 76. Savona M, Talpaz M. Getting to the stem of chronic myeloid
Res 2001;61:4666-70. leukaemia. Nat Rev Cancer 2008;8:341-50.
55. Strehl S, Konig M, Dworzak MN, Kalwak K, Haas OA. 77. Mullighan CG, Miller CB, Phillips LA, Dalton JD, Ma J, Radtke
PAX5/ETV6 fusion defines cytogenetic entity dic(9;12) I, et al. BCR-ABL1 lymphoblastic leukaemia is characterized
(p13;p13). Leukemia 2003;17:1121-3. by the deletion of Ikaros. Nature 2008;453:110-4.

| 6 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

78. Mullighan CG, Radtke I, Zhang J, Phillips LA, Su X, Ma J, et al. human leukaemias using real-time quantitative polymerase
Genome-wide analysis of genetic alterations in chronic chain reaction. Br J Haematol 2000;110:826-30.
myeloid leukemia. Blood 2008;112:1089. 86. Takanashi M, Yagi T, Imamura T, Tabata Y, Morimoto A, Hibi
79. Reynaud D, Demarco IA, Reddy KL, Schjerven H, Bertolino E, S, et al. Expression of the Ikaros gene family in childhood
Chen Z, et al. Regulation of B cell fate commitment and acute lymphoblastic leukaemia. Br J Haematol 2002;117:525-
immunoglobulin heavy-chain gene rearrangements by Ikaros. 30.
Nat Immunol 2008;9:927-36. 87. Molnar A, Georgopoulos K. The Ikaros gene encodes a family
80. Sun L, Crotty ML, Sensel M, Sather H, Navara C, Nachman J, of functionally diverse zinc finger DNA-binding proteins. Mol
et al. Expression of dominant-negative Ikaros isoforms in T- Cell Biol 1994;14:8292-303.
cell acute lymphoblastic leukemia. Clin Cancer Res 1999;5: 88. Den Boer ML, van Slegtenhorst M, De Menezes RX, Cheok
2112-20. MH, Buijs-Gladdines JG, Peters ST, et al. A subtype of child-
81. Sun L, Goodman PA, Wood CM, Crotty ML, Sensel M, Sather hood acute lymphoblastic leukaemia with poor treatment out-
H, et al. Expression of aberrantly spliced oncogenic ikaros iso- come: a genome-wide classification study. Lancet Oncol
forms in childhood acute lymphoblastic leukemia. J Clin 2009;10:125-34.
Oncol 1999;17:3753-66. 89. Mullighan CG, Phillips LA, Su X, Ma J, Miller CB, Shurtleff SA,
82. Sun L, Heerema N, Crotty L, Wu X, Navara C, Vassilev A, et
et al. Genomic analysis of the clonal origins of relapsed acute
al. Expression of dominant-negative and mutant isoforms of
the antileukemic transcription factor Ikaros in infant acute lymphoblastic leukemia. Science 2008;322:1377-80.
lymphoblastic leukemia. Proc Natl Acad Sci USA 1999;96: 90. Mullighan CG, Phillips LA, Kiedrowski LA, Ma J, Williams RT,
680-5. Shurtleff SA, et al. Ultra-high resolution genomic analysis of
83. Nakase K, Ishimaru F, Avitahl N, Dansako H, Matsuo K, Fujii genomic alterations in high-risk acute lymphoblastic
K, et al. Dominant negative isoform of the Ikaros gene in leukemia. Blood 2008;112:2053.
patients with adult B-cell acute lymphoblastic leukemia. 91. Campbell PJ, Stephens PJ, Pleasance ED, O'Meara S, Li H,
Cancer Res 2000;60:4062-5. Santarius T, et al. Identification of somatically acquired
84. Nishii K, Kita K, Miwa H, Shikami M, Taniguchi M, Usui E, et rearrangements in cancer using genome-wide massively paral-
al. Expression of B cell-associated transcription factors in B-cell lel paired-end sequencing. Nat Genet 2008;40:722-9.
precursor acute lymphoblastic leukemia cells: association with 92. Mortazavi A, Williams BA, McCue K, Schaeffer L, Wold B.
PU.1 expression, phenotype, and immunogenotype. Int J Mapping and quantifying mammalian transcriptomes by
Hematol 2000;71:372-8. RNA-Seq. Nat Methods 2008;5:621-8.
85. Olivero S, Maroc C, Beillard E, Gabert J, Nietfeld W, 93. Shendure J. The beginning of the end for microarrays? Nat
Chabannon C, et al. Detection of different Ikaros isoforms in Methods 2008;5:585-7.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 7 |
Acute lymphoblastic leukemia

Genetic and epigenetic programs in MLL-rearranged


acute lymphoblastic leukemia

S.A. Armstrong A B S T R A C T

Mixed lineage leukemia (MLL)-rearranged leukemias represent a subset of human leukemias that
Division of Hematology/Oncology, often have a poor prognosis. Recent studies have focused on characterization of the aberrant gene
Children’s Hospital, Department of
Pediatric Oncology, Boston, USA expression and epigenetic programs have begun to provide insight into the mechanisms by which
MLL-translocations induce both acute lymphoblastic leukemia and acute myelogenous leukemia.
Wildtype MLL is a histone methyltransferase that modifies histone H3 on lysine 4. However, MLL-
fusion proteins generated by the MLL-translocations interact with other histone modifying enzymes
Hematology Education: to direct inappropriate histone modifications as part of the mechanism by which they induce aberrant
the education program for the
gene expression. Future studies promise to guide us to therapeutics that target the epigenetic pro-
annual congress of the European
Hematology Association grams in these difficult to treat leukemias.

2009;3:8-12

eukemias bearing translocations involv- Subsequently, over 30 different transloca-

L ing chromosome 11q23 are of particular


interest due to unique clinical and bio-
logical characteristics. 11q23 rearrangements
tions have been identified, all of which
appear to produce a fusion protein possess-
ing the NH2-terminus of MLL fused in-frame
are found in over 70% of leukemias in to the COOH-terminus of the fusion part-
infants of less than 1 year of age whether the ner.7 Of interest, a subset of t(4;11) ALLs also
immunophenotype is more consistent with express the reciprocal product (AF4-AML),
acute lymphoblastic leukemia (ALL) or acute suggesting this fusion protein might also
myelogenous leukemia (AML).1 11q23 have pathogenic importance.8 While MLL
rearrangements are also found in a subset of translocations can be found in either ALL or
ALL cases diagnosed in adults. Importantly, AML, particular translocations show lineage
infants diagnosed with ALL harboring an specificity with the t(4;11) found most often
11q23 translocation have a particularly poor in ALL and the t(9;11)(p21;q23) found most
prognosis with an overall survival of less often in AML. This points to the transloca-
than 50%, whereas children with ALL har- tion partner as having a role in the disease
boring other translocations have an overall phenotype, and to functional heterogeneity
survival of over 80%. Leukemias that devel- of MLL-fusions, but the molecular details of
op as a result of treatment with topoiso- these associations are unclear.
merase II inhibitors frequently harbor 11q23
rearrangements and have a poor prognosis.2 Mixed lineage leukemia function in
Some infant leukemias express cell surface hematopoiesis
antigens characteristic of both lymphoblasts The MLL gene encodes a 3969 amino acid
and monoblasts, and are often designated DNA-binding protein that possesses multi-
Acute Biphenotyic Leukemias (ABL). The ple recognizable protein motifs, including an
association of 11q23 rearrangements with NH2-terminal DNA binding domain, tran-
either ALL, AML, or ABL is unique in that scriptional activation and repression
most other chromosomal rearrangements domains, and a COOH-terminal SET
tend to be associated with leukemias of a domain that contains histone H3 lysine 4
particular hematopoietic lineage. These (H3K4) methyltransferase activity.9,10 Recent
observations led to the name mixed lineage biochemical studies have identified MLL as a
leukemia (MLL) for the gene that resides on member of a large multi-protein complex
11q23. that contains members involved in chro-
The most common MLL translocation matin modification/remodeling. Notably,
found in lymphoblastic leukemias is the the complex includes histone deacetylases
t(4;11)(q21;q23). Multiple groups cloned the (HDACs) and members of the Swi/Snf chro-
MLL-AF4 gene formed as a result of this matin remodeling complex.10 Also, MLL is
translocation in the early 1990s.3-6 MLL-AF4 recruited to the promoters of select cell cycle
encodes a protein of 2304 amino acids, with regulatory genes by the protein product of
the NH2-terminal with approximately 1400 the MEN1 tumor suppressor gene, suggest-
amino acids derived from MLL on chromo- ing a role for MLL in tumor suppression and
some 11, and COOH-terminal amino acids cell cycle control.11,12 These data support the
from the AF4 gene on chromosome 4. hypothesis that the MLL protein regulates

|8| Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

gene expression via chromatin modification. lower levels are associated with lymphoid identity.
Analysis of MLL knockout mice suggests that MLL Recent studies have also defined specific gene expres-
plays an important role in development and sion signatures associated with MLL-translocations in
hematopoiesis through maintenance of appropriate primary human AML blasts.21 Of interest, even though
homeotic (Hox) gene expression.13,14 Detailed studies there are clear differences in expression of lineage asso-
assessing the specific role of MLL in hematopoietic ciated genes between MLL-rearranged ALL and MLL-
development have shown that it is necessary for defini- rearranged AML, there appears to be a core gene expres-
tive hematopoiesis and expansion of hematopoietic sion profile found in all MLL-rearranged human
progenitors and stem cells found in the aorta-gonad- leukemias independent of the lineage markers.21
mesonephros (AGM) region of the developing Presumably, MLL-fusion proteins directly regulate a
embryo.15 Also, MLL plays a critical role in adult HSC.16 subset of these genes. This is further supported by the
The defect in hematopoietic progenitor expansion can fact that this MLL-associated signature consists of mul-
be rescued by re-expression of Hox genes confirming tiple highly expressed HOX genes.
the importance of MLL-mediated Hox gene expression
during hematopoiesis. Histone methylation and mixed lineage leukemia-fusions
Multiple studies have demonstrated the ability of Hox Recent data suggests that many MLL fusion partners
genes to induce leukemia in mice, and the t(7;11) belong to a network involved in transcriptional regula-
(p15;p15) translocation found in some human AML tion through histone modification.26,27 An example of
results in a fusion of the HOXA9 gene to the nucleo- this is the MLL-EEN fusion protein that requires an argi-
porin NUP98.17,18 Given the apparent importance of Hox nine methyltransferase for its transforming activity.28
genes in leukemogenesis, it seems likely that transloca- Also, the MLL fusion partner AF10 associates with the
tions involving MLL, a known regulator of HOX genes,
DOT1L histone methyltransferase that methylates
alters expression of HOX genes that are important for
lysine 79 residues in histone H3 (H3K79).29 Expression
leukemogenesis. Further support for HOX genes as cen-
of an MLL-DOT1L fusion immortalized hematopoietic
tral regulators of MLL-induced leukemogenesis comes
progenitors, an activity not observed with either MLL
from gene expression studies that have found multiple
or DOT1L alone.29 Further evidence that MLL-fusions
HOXA cluster genes more highly expressed in MLL-
rearranged myelogenous and lymphoblastic leukemias may be involved in regulation of gene expression via
as compared to MLL-germline leukemias,19-22 and exper- histone modification came from studies assessing the
iments using RNAi, which demonstrate a continued MLL-ENL fusion. Upon expression of MLL-ENL, H3K79
requirement for HOXA cluster genes and MEIS1.23,24 levels associated with the HoxA9 and Meis1 promoters
increase.30 More recent studies have also demonstrated
Gene expression programs in mixed lineage leukemia- physical association between MLL-ENL and DOT1L.31
rearranged acute lymphoblastic leukemia Thus, at least for these fusions, association with a
Gene expression studies of human MLL-rearranged B- unique histone methyltransferase appears to be impor-
precursor ALL demonstrated that hundreds of genes are tant for leukemogenic transformation. As studies have
differentially expressed when compared to other B-pre- linked methylation of H3K79 to positive transcriptional
cursor ALLs.19,20,22,25 Based on the magnitude of the differ- regulation,32,33 the loss of H3K4 methyltransferase activ-
ences in gene expression, we proposed that MLL- ity in an MLL-fusion might be compensated by acquisi-
translocations specify a unique lymphoblastic leukemia. tion of H3K79 methyltransferase activity or arginine
Other large gene expression studies have also shown methyltransferase activity. Furthermore, as different
that ALLs with distinct chromosomal rearrangements methylation marks (H3K4, H3K36, H3K79, etc.) may
have unique gene expression profiles, thus providing positively regulate transcription in unique ways,32,34 the
support for this hypothesis.20 Genes relatively highly replacement of H3K4 activity in wild-type MLL with a
expressed in MLL-rearranged B-precursor ALL are those different histone methyltransferase activity in the MLL-
associated with hematopoietic progenitors and devel- fusion complex could further perturb transcriptional
oping myeloid cells, whereas the genes expressed at control (Figure 1).

Figure 1. Histone methylation and


MLL-fusions. H3K4 methylation is
associated with transcriptional initia-
tion and other marks such as H3K79
and H3K36 are associated with tran-
scriptional elongation. Biochemical
studies have demonstrated binding of
MLL-fusion partners to the H3K79
methyltransferase DOT1L. This leads
to the hypothesis that abnormal
recruitment of DOT1L to MLL target
genes is an important part of MLL-
fusion mediated oncogenesis.
Reprinted with permission from refer-
ence 27.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 9 |
14th Congress of the European Hematology Association

H3K79 methylation in mixed lineage leukemia-AF4 ALL the control of the endogenous promoter. Mice contain-
Since AF4 also associates with the DOT1L methyl- ing an MLL-AF9 fusion gene under control of the MLL
transferase,35 we assessed whether there might be dif- promoter spontaneously develop AML with a latency of
ferences in H3K79 methylation between MLL-AF4 ALLs 4 months to over 1 year.38 This is widely interpreted as
and normal B-cell progenitors.36 Using a newly devel- a requirement for a second genetic event during leuke-
oped mouse model of MLL-AF4 ALL, we performed mogenesis. Other models, such as an MLL-Cbp knock-
ChIP-chip analysis on mouse MLL-AF4 pre-B cell ALLs in or an AML1-Eto knock-in model do not spontaneous-
and three normal pre-B cell samples. These studies ly develop leukemia.39,40 In these models, either irradia-
demonstrated an increase in H3K79 dimethylation asso- tion or chemical mutagenesis is necessary in order to
ciated with the HoxA cluster in leukemia cells, and in induce leukemias, thus clearly requiring multiple events
approximately 1100 other promoter regions, whereas for the development of leukemia.
only 285 promoter regions were associated with Multiple lines of evidence now points to a multi-step
enhanced H3K79 dimethylation in normal pre-B cells. pathogenesis of human acute leukemia. Elegant epi-
Comparison of H3K79 methylation and gene expres- demiologic studies suggest that childhood leukemias
sion demonstrated that ectopic H3K79me2 in MLL-AF4 require at least two and probably more genetic events to
leukemias is associated with enhanced gene expression. occur for the development of leukemia. In particular,
Next, we assessed whether similar differences in lymphoblastic leukemias with TEL-AML1 rearrange-
H3K79me2 exist in human MLL-rearranged ALLs and ments appear to develop after a multi-step process. TEL-
found elevated H3K79me2 associated with 1378 pro- AML1 rearrangements are often detected in blood taken
moter regions in the leukemias and elevated H3K79me2 at birth from a child who will develop ALL 3 to 5 years
associated with 562 promoter regions in normal sam- later. This suggests that TEL-AML1 rearrangements are
ples, thus demonstrating more genes associated with the first genetic event, but also that other mutations are
increased H3K79me2 in the MLL-rearranged leukemias required for the development of ALL.41 Similar studies
as compared to normal cells. Similar to the mouse have been performed on blood spots from children that
model, we found enhanced H3K79 methylation associ- would develop MLL-rearranged ALL and the MLL-
ated with the HOXA cluster in human leukemia cells translocations clearly develop in utero while the
(Figure 2). Furthermore, there is a strong correlation leukemias become apparent sometime during the first
between elevated H3K79 methylation and abnormal year of life.42
gene expression in human MLL-rearranged ALL. A Receptor tyrosine kinases are attractive candidates as
recent independent study similarly demonstrated wide- signaling molecules that may cooperate with transloca-
spread abnormal H3K79 methylation in MLL- tion-associated fusion proteins during leukemogenesis.
rearranged ALL cell lines.37 These data demonstrate that Ever increasing evidence suggests that activated kinases
ectopic H3K79me2 is associated with gene expression play a central role in the pathogenesis of leukemias and
in human leukemias as is the case in mouse leukemias. myeloproliferative syndrome.43 The most dramatic evi-
Identification of histone methyltransferases as impor- dence for such a role is activation of the ABL tyrosine
tant for MLL-fusion mediated leukemias provides hope kinase by the BCR-ABL fusion produced by the t(9;22),
that therapeutics targeting these enzymes may be par- and its inhibition by imantinib (Gleevec).44. Other
ticularly useful in these leukemias. mutant kinases frequently identified in AML are the
receptor tyrosine kinases FLT3 and c-KIT.43 Recent
Multi-step pathogenesis of mixed lineage leukemia- mouse experiments support the hypothesis that DNA-
rearranged leukemias? binding fusion proteins generated by leukemia associat-
Mouse models of leukemia predict multiple genetic ed translocations perform different functions than acti-
events are necessary for the development of acute vated tyrosine kinases.45 DNA-binding fusions either
leukemias. “Knock-in” models of MLL-rearranged AML induce a block in differentiation or activate self-renewal
have been developed where the fusion genes generated in developing hematopoietic progenitors, while activat-
by translocations found in human leukemias are under ed kinases may provide a survival or proliferation signal.

Figure 2. H3K79 Methylation in Human MLL-rearranged ALL. Identically scaled average tracks from ChIP-chip analysis
of H3K79me2 modifications associated with HOXA cluster loci in human CD34/19+ cells (n=5) or MLL-rearranged ALL
(MLL-r ALL ) (n=5). Reprinted with permission from reference 36.

| 10 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

This has prompted the hypothesis that at least two dif- gene. Cell 2004;6:437-43.
16. Jude CD, Climer L, Xu D, Artinger E, Fisher JK, Ernst P. Cell
ferent classes of mutations are necessary for leukemoge- Unique and independent roles for MLL in adult hematopoiet-
nesis.46 ic stem cells and progenitors. Stem Cell 2007;1:324-37.
Identification of FLT3 mutations in MLL-rearranged 17. Borrow J, Shearman AM, Stanton VP, Becher R, Collins T,
ALLs suggest that a multi-step process may also be nec- Williams AJ, et al. The t(7;11)(p15;p15) translocation in acute
myeloid leukaemia fuses the genes for nucleoporin NUP98
essary for MLL-rearranged leukemias ALL.47 However, and class I homeoprotein HOXA9. Nat Genet 1996;12:159-67.
given the epigenetic dysregulation described above in 18. Nakamura T, Largaespada DA, Lee MP, Johnson LA,
MLL-rearranged ALLs, the question arises as to whether Ohyashiki K, Toyama K, et al. Fusion of the nucleoporin gene
NUP98 to HOXA9 by the chromosome translocation
these leukemias might not require further genetic t(7;11)(p15;p15) in human myeloid leukaemia. Nat Genet
changes and that the majority of oncogenic “progres- 1996;12:154-8.
sion” in MLL-rearranged ALL is a result of epigenetic 19. Armstrong SA, Staunton JE, Silverman LB, Pieters R, den Boer
ML, Minden MD, et al. MLL translocations specify a distinct
changes that are driven by the MLL-fusion. This is sup- gene expression profile that distinguishes a unique leukemia.
ported by recent genome wide studies that have Nat Genet 2002;30:41-7.
demonstrated very few gains or losses of chromosomal 20. Yeoh EJ, Ross ME, Shurtleff SA, Williams WK, Patel D,
regions in MLL-rearranged ALLs.48 If MLL-rearranged Mahfouz R, et al. Classification, subtype discovery, and pre-
diction of outcome in pediatric acute lymphoblastic leukemia
ALLs are largely driven by epigenetic changes, then by gene expression profiling. Cancer Cell 2002;1:133-43.
therapeutics targeting chromatin structure might be par- 21. Ross ME, Mahfouz R, Onciu M, Liu HC, Zhou X, Song G, et
ticularly useful in this dreadful disease. Further genetic, al. Gene expression profiling of pediatric acute myelogenous
leukemia. Blood 2004;104:3679-87.
genomic and biochemical studies should help us deter- 22. Ferrando AA, Armstrong SA, Neuberg DS, Sallan SE,
mine how best to exploit this knowledge for therapeu- Silverman LB, Korsmeyer SJ, et al. Gene expression signatures
tic benefit. in MLL-rearranged T-lineage and B-precursor acute leukemias:
dominance of HOX dysregulation. Blood 2003;102:262-8.
23. Faber J, Krivtsov AV, Stubbs MC, Wright R, Davis TN, van den
Heuvel-Eibrink M, et al. HOXA9 is required for survival in
References human MLL-rearranged acute leukemias. Blood 2009;113:
2375-85.
1. Biondi A, Cimino G, Pieters R, Pu, CH. Biological and thera- 24. Kumar AR, Li Q, Hudson WA, Chen W, Sam T, Yao Q, et al.
peutic aspects of infant leukemia. Blood 2000;96:24-33. A role for MEIS1 in MLL-fusion gene leukemia. Blood 2009;
2. Felix CA, Hosler MR, Winick NJ, Masterson M, Wilson AE, 113:1756-8.
Lange BJ. ALL-1 gene rearrangements in DNA topoisomerase 25. Ross ME, Zhou X, Song G, Shurtleff SA, Girtman K, Williams
II inhibitor-related leukemia in children. Blood 1995;85:3250- WK, et al. Classification of pediatric acute lymphoblastic
6. leukemia by gene expression profiling. Blood 2003;102:2951-
3. Domer PH, Fakharzadeh SS, Chen CS, Jockel J, Johansen L, 9.
Silverman GA, et al. Acute mixed-lineage leukemia 26. Erfurth F, Hemenway CS, de Erkenez AC, Domer PH. MLL
t(4;11)(q21;q23) generates an MLL-AF4 fusion product. Proc fusion partners AF4 and AF9 interact at subnuclear foci.
Natl Acad Sci USA 1993;90:7884-8. Leukemia 2004;18:92-102.
4. Gu Y, Nakamura T, Alder H, Prasad R, Canaani O, Cimino G, 27. Krivtsov AV, Armstrong SA. MLL translocations, histone mod-
et al. The t(4;11) chromosome translocation of human acute ifications and leukaemia stem-cell development. Nat Rev
leukemias fuses the ALL-1 gene, related to Drosophila tritho- Cancer 2007;7:823-33.
rax, to the AF-4 gene. Cell 1992;71:701-8. 28. Cheung N, Chan LC, Thompson A, Cleary ML, So CW.
5. Tkachuk DC, Kohler S, Cleary ML. Involvement of a homolog Protein arginine-methyltransferase-dependent oncogenesis.
of Drosophila trithorax by 11q23 chromosomal translocations Nat Cell Biol 2007;9:1208-15.
in acute leukemias. Cell 1992;71:691-700. 29. Okada Y, Feng Q, Lin Y, Jiang Q, Li Y, Coffield VM, et al.
6. Ziemin-van der Poel S, McCabe NR, Gill HJ, Espinosa R, Patel hDOT1L links histone methylation to leukemogenesis. Cell
Y, Harden A, et al. Identification of a gene, MLL, that spans the 2005;121:167-78.
breakpoint in 11q23 translocations associated with human 30. Milne TA, Martin ME, Brock HW, Slany RK, Hess JL.
leukemias. Proc Natl Acad Sci USA 1991;88:10735-9. Leukemogenic MLL fusion proteins bind across a broad region
7. Ayton PM, Cleary ML. Molecular mechanisms of leukemoge- of the Hox a9 locus, promoting transcription and multiple his-
nesis mediated by MLL fusion proteins. Oncogene 2001;20: tone modifications. Cancer Res 2005;65:11367-74.
5695-707. 31. Mueller D, Bach C, Zeisig D, Garcia-Cuellar MP, Monroe S,
8. Gaussmann A, Wenger T, Eberle I, Bursen A, Bracharz S, Herr Sreekumar A, et al. A role for the MLL fusion partner ENL in
I, et al. Combined effects of the two reciprocal t(4;11) fusion transcriptional elongation and chromatin modification. Blood
proteins MLL.AF4 and AF4.MLL confer resistance to apopto- 2007;110:4445-54.
sis, cell cycling capacity and growth transformation. 32. Guenther MG, Levine SS, Boyer LA, Jaenisch R, Young RA.
Oncogene 2007;26:3352-63. Cell A chromatin landmark and transcription initiation at most
9. Milne TA, Briggs SD, Brock HW, Martin ME, Gibbs D, Allis promoters in human cells. 2007;130:77-88.
CD, et al. MLL targets SET domain methyltransferase activity 33. Lachner M, O'Sullivan RJ, Jenuwein T.An epigenetic road map
to Hox gene promoters. Mol Cell 2002;10:1107-17. for histone lysine methylation. J Cell Sci 2003;116:2117-24.
10. Nakamura T, Mori T, Tada S, Krajewski W, Rozovskaia T, 34. Shilatifard A. Chromatin modifications by methylation and
Wassell R, et al. ALL-1 is a histone methyltransferase that ubiquitination: implications in the regulation of gene expres-
assembles a supercomplex of proteins involved in transcrip- sion. Annu Rev Biochem 2006;75:243-269.
tional regulation. Mol Cell 2002;10:1119-28. 35. Bitoun E, Oliver PL, Davies K E. The mixed-lineage leukemia
11. Hughes CM, Rozenblatt-Rosen O, Milne TA, Copeland TD, fusion partner AF4 stimulates RNA polymerase II transcrip-
Levine SS, Lee JC, et al. Menin associates with a trithorax fam- tional elongation and mediates coordinated chromatin remod-
ily histone methyltransferase complex and with the hoxc8 eling. Hum Mol Genet 2007;16:92-106.
locus. Mol Cell 2004;13:587-97. 36. Krivtsov AV, Feng Z, Lemieux ME, Faber J, Vempati S, Sinha
12. Milne TA, Dou Y, Martin ME, Brock HW, Roeder RG, Hess JL. AU, et al. H3K79 methylation profiles define murine and
MLL associates specifically with a subset of transcriptionally human MLL-AF4 leukemias. Cancer Cell 2008;14:355-68.
active target genes. Proc Natl Acad Sci USA 2005;102:14765- 37. Guenther MG, Lawton LN, Rozovskaia T, Frampton GM,
70. Levine SS, Volkert TL, et al. Aberrant chromatin at genes
13. Hess JL, Yu BD, Li B, Hanson R, Korsmeyer SJ. Defects in yolk encoding stem cell regulators in human mixed-lineage
sac hematopoiesis in Mll-null embryos. Blood 1997;90:1799- leukemia. Genes Dev 2008;22:3403-8.
806. 38. Corral J, Lavenir I, Impey H, Warren AJ, Forster A, Larson TA,
14. Yu BD, Hess JL, Horning SE, Brown GA, Korsmeyer SJ. et al. An Mll-AF9 fusion gene made by homologous recombi-
Altered Hox expression and segmental identity in Mll-mutant nation causes acute leukemia in chimeric mice: a method to
mice. Nature 1995;378:505-8. create fusion oncogenes. Cell 1996;85:853-61.
15. Ernst P, Fisher JK, Avery W, WadeS, Foy D, Korsmeyer SJ. Dev 39. Higuchi M, O'Brien D, Kumaravelu P, Lenny N, Yeoh EJ,
Definitive hematopoiesis requires the mixed-lineage leukemia Downing JR. Expression of a conditional AML1-ETO onco-

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 11 |
14th Congress of the European Hematology Association

gene bypasses embryonic lethality and establishes a murine sine kinase in the blast crisis of chronic myeloid leukemia and
model of human t(8;21) acute myeloid leukemia. Cancer Cell acute lymphoblastic leukemia with the Philadelphia chromo-
2002;1:63-74. some. N Engl J Med 2001;344:1038-42.
40. Wang J, Iwasaki H, Krivtsov A, Febbo PG, Thorner AR, Ernst 45. Huntly BJ, Shigematsu H, Deguchi K, Lee BH, Mizuno S,
P, et al. Conditional MLL-CBP targets GMP and models thera- Duclos N, et al. MOZ-TIF2, but not BCR-ABL, confers proper-
py-related myeloproliferative disease. Embo J 2005;24:368-81. ties of leukemic stem cells to committed murine hematopoiet-
41. Wiemels JL, Cazzaniga G, Daniotti M, Eden OB, Addison GM, ic progenitors. Cancer Cell 2004;6:587-96.
Masera G, et al. Prenatal origin of acute lymphoblastic 46. Kelly LM, Gilliland DG. Genetics of myeloid leukemias. Annu
leukaemia in children. Lancet 1999;354:1499-503. Rev Genomics Hum Genet 2002;3:179-98.
42. Ford AM, Ridge SA, Cabrera ME, Mahmoud H, Steel CM, 47. Armstrong SA, Kung AL, Mabon ME, Silverman LB, Stam RW,
Chan LC, et al. In utero rearrangements in the trithorax-relat- Den Boer ML, et al. Inhibition of FLT3 in MLL. Validation of a
ed oncogene in infant leukaemias. Nature 1993;363:358-60. therapeutic target identified by gene expression based classifi-
43. Scheijen B, Griffin JD. Tyrosine kinase oncogenes in normal cation. Cancer Cell 2003;3:173-83.
hematopoiesis and hematological disease. Oncogene 2002;21: 48. Mullighan CG, Goorha S, Radtke I, Miller CB. Coustan-Smith
3314-33. E, Dalton JD, et al. Genome-wide analysis of genetic alter-
44. Druker BJ, Sawyers CL, Kantarjian H, Resta DJ, Reese SF, Ford ations in acute lymphoblastic leukaemia. Nature 2007;446:
JM, et al. Activity of a specific inhibitor of the BCR-ABL tyro- 758-64.

| 12 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Acute lymphoblastic leukemia

The acute lymphoblastic leukemias of Down syndrome

S. Izraeli A B S T R A C T

Children with Down syndrome have a markedly increased risk for acute lymphoblastic leukemia (DS-
Sheba Medical Center and Tel Aviv ALL). These leukemias are exclusively of the B cell precursor phenotype and occur in a similar age to
University Medical School, Israel
“common” sporadic ALLs. Recent studies reveal that DS-ALLs are heterogeneous and differ from spo-
radic ALLs with acquired trisomy or tetrasomy 21. Acquired mutations in Arginine 683 of JAK2, result-
ing in a constitutive activation of JAK/STAT signaling, characterize about 20% of DS-ALLs and suggest
Hematology Education: the potential therapeutic utility of JAK2 inhibitors. In general, the prognosis of DS-ALL is inferior to
the education program for the sporadic ALL, mainly because of increased treatment toxicity and possibly because of the lack of prog-
annual congress of the European
nostically favorable genetic abnormalities. Recent data of improved prognosis on more intensified reg-
Hematology Association
imens implies that excessive chemotherapy dose reductions may not be appropriate for these patients.
2009;3:13-16 Current international surveys aimed at detailed characterization of patients’ clinical outcome and on-
going cooperative biological studies could lead to improved therapy for this high-risk disease.

hildren with Down syndrome (DS) with DS do not develop leukemia, such

C have a markedly enhanced incidence


of myeloid (ML-DS) and lymphoid
(DS-ALL) leukemias. The higher risk of
progression events are necessary for
leukemogenesis. Almost all the ML-DS
have an acquired mutation in the
leukemias in DS is striking in light of the megakaryocytic transcription factor
reduced risk of most solid tumors.1,2 This sug- GATA1.11,12 Does a similar cooperative
gests a leukemogenic role of constitutional genetic event, unique to DS, exist in DS-
trisomy 21. The ML-DS is a defined entity, ALL?
unique to DS, with a clear clinical presenta- There is high interest in the pathogenesis
tion and course, excellent response to of DS-ALL also because trisomy 21 (or
chemotherapy and a relatively well deci- sometimes tetrasomy 21) is the most com-
phered molecular pathogenesis.3,4 Much less mon acquired somatic chromosomal abnor-
is known about DS-ALL, and here the cur- malities in sporadic ALL.13 It is mostly found
rent understanding of its pathogenesis and in high hyperdiploid ALL (HHD-ALL), a sub-
clinical management is reviewed. type of ALL characterized by more than 50
The risk of DS-ALL has been estimated to chromosomes, always involving chromo-
be 10-20 times higher than sporadic ALL some 21. Hence, it is tempting to speculate
and it is the most common leukemia in chil- that constitutional and somatic trisomy 21
dren with DS.1,5 In most published multi- may facilitate leukemogenesis in a similar
institutional ALL protocols, DS-ALL com- fashion and, therefore, the study of DS-ALL
prises about 1 to 3% of total patients.6-8 The may have direct implications for sporadic
peak age is about 5 years, slightly older than childhood ALL.
the peak age of sporadic ALLs, possibly However, there are fundamental differ-
related to absence of DS-ALL below the age ences between constitutional and somatic tri-
of 1 year. The immunophenotype is typical somies that could explain the uniqueness of
for B cell precursor ALL5,9,10 with the notable DS leukemias. The former exists in all body
absence of T and pro-B ALLs. cells, whereas the latter is acquired and only
exists in transformed cells. Thus, constitu-
Pathogenesis tional trisomy can predispose to cancer in a
The excess of ALLs in DS raises several variety of ways. It may exert a direct onco-
general questions: genic activity in a cell autonomous manner,
a) Are these leukemias unique to DS (like similar to the somatic trisomy. Alternatively,
the ML-DS) or do children with DS have the trisomy could promote leukemia
a general increased risk for childhood because of aberrant effects on immediate
“common” B cell precursor ALL? micro-environment; for example, on the bone
b) What is the role of trisomy 21? Which marrow’s or fetal liver’s stroma cells that reg-
are the genes on trisomy 21 that confers ulate proliferation and differentiation of
increased risk for ALL? hematopoietic stem cells. More complex
c) What is the nature of the acquired may be the influence of the trisomy on the
somatic genetic events that cooperate macro-environment. For example, viral
with constitutional trisomy 21 in the infections and the immunological response
evolution to ALL? Since most children have been suggested to have a role in the

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 13 |
14th Congress of the European Hematology Association

pathogenesis of childhood common ALL.14,15 The


markedly increased risk of ALL in DS could also be
caused by the immunodeficiency, altered immunologi-
cal environment and the increased infection rate that
characterize DS.
Another fundamental difference between constitu-
tional and somatic trisomies is the timing of their occur-
rence during ontogeny. Constitutional trisomy 21 exists
since conception. Thus, it is possible that constitutional
trisomy 21 could lead to an expansion of “primitive”
fetal hematopoietic cells that are later transformed by
acquired somatic oncogenic events. Thus, the fetal ori-
gin of leukemia associated with constitutional trisomy
21 may differ from the sporadic leukemias. This is prob-
ably the case with ML-DS where expansion of primitive
megakaryoblastic precursors have been shown in-vivo Figure 1. Cooperating genetic events in DS leukemias. The
in human livers from DS fetuses.4,16,17 These abnormal myeloid leukemias of DS (ML-DS) are universally character-
ized by an acquired mutation in GATA1 (Additional somatic
precursors may be especially sensitive to the mutation genetic aberrations are also required). The lymphoid
in GATA1, universally observed in ML-DS.18 Similarly, leukemias (DS-ALL) are more heterogeneous. About 15% of
DS-ALL may be caused by an expansion of a “special” DS-ALLs carry similar aberrations of sporadic ALLs, namely
TEL/AML1 translocation or hyperdiploidy (HHD). Most of DS-
fetal lymphoid precursor by constitutional trisomy 21. ALLs have cooperating events that are relatively unique to
Recent studies demonstrate that unlike ML-DS, DS- DS. About 20% have an acquired JAK2 mutation, another
ALL is a heterogeneous disease suggesting complex 40% and additional X chromosome. Translocations into the
pathogenesis (Figure 1). Overall, there is a significantly IgH locus at 14q32 are also more common in DS-ALL.
lower prevalence of the common genetic subtypes of B
cell precursor ALL (BCR/ABL, TEL/AML1 and HHD) in
DS.10,19-24 This was confirmed in the recent largest cyto-
genetic study of 215 DS children with ALL.25 The preva- ies identified that acquired mutations in the JAK2 kinase
lence of TEL/AML1 and hyperdiploid ALLs was lower characterize about a fifth of all DS-ALLs.26,32,33 The
in DS patients compared to sporadic ALLs. However, if largest study from the iBFM group, involving 88 chil-
one takes into account the 20-fold increased risk of ALL dren with DS-ALL, revealed that children with JAK2
in DS,1 then there may be an absolute increase in the mutations were significantly younger (mean 4.5 vs. 8.6
incidence of these common subtypes of childhood yrs, p<0.001) at diagnosis. As this was not confirmed in
leukemias in DS. Similarly, genomic studies by CGH or a smaller study from the Children’s Oncology Group,33
SNP arrays have revealed similar acquired copy number it is presently unclear if the patients with JAK2 mutation
variations in DS and TEL/AML1 ALL.26,27 constitute a defined clinical subgroup. All mutant alleles
However, it emerges that the majority of the DS-ALLs are centered around a highly conserved arginine residue
differ from the sporadic ALLs containing excess chro- (R683) within the JAK2 pseudokinase domain. The
mosome 21. The most common cytogenetic abnormal- mutations immortalize primary mouse hematopoietic
ity in DS-ALL is an extra chromosome X, observed in progenitors, and cause constitutive JAK/STAT activation
close to half the patients.25 An additional copy of chro- and cytokine independent growth hematopoietic cells
mosome X is usually present in HHD sporadic ALL, dis- that are sensitive to pharmacological inhibition of
playing additional copies of multiple chromosomes JAK/STAT signaling. Similar to the GATA1s mutations
including chromosomes 21, X, 6 and occasionally chro- in DS myeloid malignancies, the novel mutations in R683
mosomes 10, 14, 17 and 18.13 However, the combina- of JAK2 are unique to DS-ALL and are not detected nei-
tion of trisomy 21 and extra chromosome X as a single ther in myeloproliferative neoplasms (MPNs) nor in
cytogenetic abnormality seems unique to DS-ALL, and sug- sporadic childhood ALL. Modeling of JAK2 pseudoki-
gests a yet unknown collaborating event between gene nase domain revealed that R683 is situated in an
(s) on chromosome 21 and X. exposed conserved region separated from the one
Another recurrent cytogenetic abnormality t(8;14) involved in MPNs.
(q11;q32) is associated with DS-ALL.25,28 This rare The association between a specific mutation in JAK2
translocation displaces the C/EBPδ gene into the IGH and DS-ALL is fascinating from several aspects, not only
locus. The C/EBP transcription factors induce myeloid relevant for DS but for the general understanding of
differentiation. Presumably their expression in B cell leukemogenesis and the biology of JAK signaling.
precursor ALL blocks lymphoid differentiation. Of 44 Acquired somatic point mutations in two different sites
patients reported with this translocation, 27% had DS- (V617 and R683) in the same domain of JAK2 leading to
ALL.28 apparently the same biochemical outcome (constitutive
The JAK2 kinase participates in signal transduction of activation of JAK2) are strictly associated with hemato-
growth promoting signals induced by several cytokines. logical malignancies arising in different lineages
Activating mutations in JAK2 are commonly found in (myeloid vs. B lymphoid, respectively). Importantly, the
myeloproliferative neoplasms.29,30 All are missense V617F mutation occurs in an early hematopoietic pro-
mutations in the JAK2 pseudokinase domain, causing its genitor and is also detected in lymphocytes in patients
constitutive activation leading to cytokine independent with MPNs,34-37 yet it causes an expansion only of the
growth. Following one case report,31 several large stud- myeloid compartment. One potential mechanism could

| 14 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

be the presence of lineage specific regulators of JAK2, JAK2 inhibitors are required for this specific group of
whose activity is disrupted by the specific point muta- leukemia prone children.
tions. The second fascinating observation is the tight
association between constitutional trisomy 21 and the
R683 mutated JAK2. This association does not simply References
depend on the presence of extra chromosome 21 since
it is not observed in HHD sporadic ALL. Rather it has to 1. Hasle H. Pattern of malignant disorders in individuals with
do with (most likely) some lymphoid developmental Down's syndrome. Lancet Oncol 2001;2:429-36.
2. Hasle H, Clemmensen IH, Mikkelsen M. Risks of leukaemia
effects of constitutional trisomy 21 or with, as yet and solid tumours in individuals with Down's syndrome.
unknown, a specific type of genomic instability con- Lancet 2000;355:165-9.
ferred by constitutional trisomy 21. Finally, these results 3. Malinge S, Izraeli S, Crispino JD. Insights into the manifesta-
tions, outcomes and mechanisms of leukemogenesis in Down
not only provide the first molecular specific lesion of syndrome. Blood 2009 Jan 12.
DS-ALL but also suggest that these leukemias are candi- 4. Izraeli S, Rainis L, Hertzberg L, Smooha G, Birger Y. Trisomy
dates for therapy with the novel JAK2 inhibitors that are of chromosome 21 in leukemogenesis. Blood Cells Mol Dis
2007;39:156-9.
being explored in early clinical trials for myeloprolifera- 5. James R, Lightfoot T, Simpson J, Moorman AV, Roman E,
tive neoplasms. Kinsey S. Acute leukemia in children with Down's syndrome:
the importance of population based study. Haematologica
Clinical course and therapy 2008;93:1262-3.
6. Ravindranath Y. Down syndrome and leukemia: new insights
Unlike ML-DS that is uniquely sensitive to into the epidemiology, pathogenesis, and treatment. Pediatr
chemotherapy, in particular to ARA-C, the prognosis of Blood Cancer 2005;44:1-7.
DS-ALL is less favorable in most of the clinical tri- 7. Ross JA, Spector LG, Robison LL, Olshan AF. Epidemiology of
leukemia in children with Down syndrome. Pediatr Blood
als.9,10,21,22,24,38-41 Marked toxicity manifested by increased Cancer. 2005;44:8-12.
mucositis, infections and death during intensive periods 8. Zeller B, Gustafsson G, Forestier E, Abrahamsson J, Clausen
of chemotherapy is observed. DS patients may be espe- N, Heldrup J, et al. Acute leukaemia in children with Down
syndrome: a population-based Nordic study. Br J Haematol.
cially sensitive to the toxic effects of Methotrexate, a 2005;128:797-804.
drug that is not used in AML, due to the excess activity 9. Whitlock JA, Sather HN, Gaynon P, Robison LL, Wells RJ,
of the folate transporter coded by a gene on chromo- Trigg M, et al. Clinical characteristics and outcome of children
with Down syndrome and acute lymphoblastic leukemia: a
some 21.42 However, severe toxicity is also observed to Children's Cancer Group study. Blood 2005;106:4043-9.
anthracyclines and to the marked immunosuppressive 10. Arico M, Ziino O, Valsecchi MG, Cazzaniga G, Baronci C,
effect of ALL therapy. Messina C, et al. Acute lymphoblastic leukemia and Down
Importantly, marked reduction of chemotherapy may syndrome: presenting features and treatment outcome in the
experience of the Italian Association of Pediatric Hematology
be a mistake in DS-ALL. Unlike “common” ALL (HHD and Oncology (AIEOP). Cancer 2008;113:515-21.
and TEL/AML1 ALLs), many of the DS-ALLs fall by 11. Rainis L, Bercovich D, Strehl S, Teigler-Schlegel A, Stark B,
NCI clinical criteria into a higher risk group.24 A recent Trka J, et al. Mutations in exon 2 of GATA1 are early events in
megakaryocytic malignancies associated with trisomy 21.
Children’s Cancer Group study demonstrated a surpris- Blood 2003;102:981-6.
ingly good survival in children with DS-ALL treated by 12. Wechsler J, Greene M, McDevitt MA, Anastasi J, Karp JE, Le
risk adjusted intensive chemotherapy protocols. Event- Beau MM, et al. Acquired mutations in GATA1 in the mega-
karyoblastic leukemia of Down syndrome. Nat Genet 2002;
free (56% vs. 74%; p<0.001) and disease-free (55% vs. 32:148-52.
73%; p<0.001) survival at 10 years was significantly 13. Heerema NA, Sather HN, Sensel MG, Zhang T, Hutchinson
lower in the standard-risk DS-ALL population com- RJ, Nachman JB, et al. Prognostic impact of trisomies of chro-
mosomes 10, 17, and 5 among children with acute lym-
pared with ALL in non DS, but not in high-risk DS-ALL phoblastic leukemia and high hyperdiploidy (> 50 chromo-
population (event-free survival, 62% vs. 59%; p=0.9; somes). J Clin Oncol 2000;18:1876-87.
disease-free survival, 64% vs. 59%; p=0.09). In addition, 14. Einav U, Tabach Y, Getz G, Yitzhaky A, Ozbek U, Amariglio
these differences persisted, regardless of treatment era N, et al. Gene expression analysis reveals a strong signature of
an interferon-induced pathway in childhood lymphoblastic
(early era [1983-1989] vs. recent era [1989-1995]). leukemia as well as in breast and ovarian cancer. Oncogene
Multivariate analysis revealed that presence of DS 2005;24:6367-75.
demonstrated an independent significant adverse prog- 15. Greaves MF. Aetiology of acute leukaemia. Lancet 1997; 349:
344-9.
nostic effect for the standard-risk population, but not 16. Chou ST, Opalinska JB, Yao Y, Fernandes MA, Kalota A,
for the high-risk patients.9 These results suggest that Brooks JS, et al. Trisomy 21 enhances human fetal erythro-
intensification of therapy for patients with DS-ALL is megakaryocytic development. Blood 2008 Sep 23.
17. Tunstall-Pedoe O, Roy A, Karadimitris A, de la Fuente J, Fisk
needed to maintain outcome comparable with those of NM, Bennett P, et al. Abnormalities in the myeloid progenitor
ALL in non DS patients. Similarly, a recent survey of compartment in Down syndrome fetal liver precede acquisi-
eight children with DS-ALL who underwent bone mar- tion of GATA1 mutations. Blood 2008 Aug 8.
18. Li Z, Godinho FJ, Klusmann JH, Garriga-Canut M, Yu C, Orkin
row transplantation reported that relapse and not treat- SH. Developmental stage-selective effect of somatically
ment related toxicity were the major causes for treat- mutated leukemogenic transcription factor GATA1. Nat Genet
ment failure. Indeed, the only surviving patients were 2005;37:613-9.
those that were treated by myeloablative chemothera- 19. Pui CH, Raimondi SC, Borowitz MJ, Land VJ, Behm FG, Pullen
DJ, et al. Immunophenotypes and karyotypes of leukemic
py.39 cells in children with Down syndrome and acute lymphoblas-
To understand better the response to therapy, toxicity tic leukemia. J Clin Oncol 1993;11:1361-7.
and treatment outcome, an international retrospective 20. Savasan S, Taub JW, Ravindranath Y. Down syndrome and
leukemia-an overview of cytogenetic and molecular events.
survey of the fate of children with DS-ALL treated on Turk J Pediatr 1997;39:519-31.
major cooperative ALL protocols has been initiated. 21. Dordelmann M, Schrappe M, Reiter A, Zimmermann M, Graf
Meanwhile, clinicians should be careful in dose reduc- N, Schott G, et al. Down's syndrome in childhood acute lym-
phoblastic leukemia: clinical characteristics and treatment out-
tion of DS-ALL patients. Prospective clinical trials and come in four consecutive BFM trials. Berlin-Frankfurt-Munster
incorporation of potentially less toxic drugs such as Group. Leukemia 1998;12:645-51.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 15 |
14th Congress of the European Hematology Association

22. Chessells JM, Harrison G, Richards SM, Bailey CC, Hill FG, 372:1484-92.
Gibson BE, et al. Down's syndrome and acute lymphoblastic 33. Gaikwad A, Rye CL, Devidas M, Heerema NA, Carroll AJ,
leukaemia: clinical features and response to treatment. Arch Izraeli S, et al. Prevalence and clinical correlates of JAK2 muta-
Dis Child 2001;85:321-5. tions in Down syndrome acute lymphoblastic leukaemia. Br J
23. Bassal M, La MK, Whitlock JA, Sather HN, Heerema NA, Haematol 2008 Dec 20.
Gaynon PS, et al. Lymphoblast biology and outcome among 34. Campbell PJ, Baxter EJ, Beer PA, Scott LM, Bench AJ, Huntly
children with Down syndrome and ALL treated on CCG- BJ, et al. Mutation of JAK2 in the myeloproliferative disorders:
1952. Pediatr Blood Cancer 2005;44:21-8. timing, clonality studies, cytogenetic associations, and role in
24. Whitlock JA. Down syndrome and acute lymphoblastic leukemic transformation. Blood 2006;108:3548-55.
leukaemia. Br J Haematol 2006;135:595-602. 35. Bogani C, Guglielmelli P, Antonioli E, Pancrazzi A, Bosi A,
25. Forestier E, Izraeli S, Beverloo B, Haas O, Pession A, Vannucchi AM. B-, T-, and NK-cell lineage involvement in
Michalova K, et al. Cytogenetic features of acute lymphoblas- JAK2V617F-positive patients with idiopathic myelofibrosis.
tic and myeloid leukemias in pediatric patients with Down Haematologica 2007;92:258-9.
syndrome: an iBFM-SG study. Blood 2008;111:1575-83. 36. Delhommeau F, Dupont S, Tonetti C, Masse A, Godin I, Le
26. Kearney L, Gonzalez De Castro D, Yeung J, Procter J, Horsley Couedic JP, et al. Evidence that the JAK2 G1849T (V617F)
SW, Eguchi-Ishimae M, et al. Specific JAK2 mutation mutation occurs in a lymphomyeloid progenitor in poly-
(JAK2R683) and multiple gene deletions in Down syndrome cythemia vera and idiopathic myelofibrosis. Blood 2007;109:
acute lymphoblastic leukemia. Blood 2009;113:646-8. 71-7.
27. Lo KC, Chalker J, Strehl S, Neat M, Smith O, Dastugue N, et 37. Ishii T, Bruno E, Hoffman R, Xu M. Involvement of various
al. Array comparative genome hybridization analysis of acute hematopoietic-cell lineages by the JAK2V617F mutation in
lymphoblastic leukaemia and acute megakaryoblastic polycythemia vera. Blood 2006;108:3128-34.
leukaemia in patients with Down syndrome. Br J Haematol 38. Watson MS, Carroll AJ, Shuster JJ, Steuber CP, Borowitz MJ,
2008 Jun 13. Behm FG, et al. Trisomy 21 in childhood acute lymphoblastic
28. Lundin C, Heldrup J, Ahlgren T, Olofsson T, Johansson B. B- leukemia: a Pediatric Oncology Group study (8602). Blood
cell precursor t(8;14)(q11;q32)-positive acute lymphoblastic 1993;82:3098-102.
leukemia in children is strongly associated with Down syn- 39. Meissner B, Borkhardt A, Dilloo D, Fuchs D, Friedrich W,
drome or with a concomitant Philadelphia chromosome. Eur J Handgretinger R, et al. Relapse, not regimen-related toxicity,
Haematol 2009;82:46-53. was the major cause of treatment failure in 11 children with
29. James C, Ugo V, Le Couedic JP, Staerk J, Delhommeau F, Down syndrome undergoing haematopoietic stem cell trans-
Lacout C, et al. A unique clonal JAK2 mutation leading to con- plantation for acute leukaemia. Bone Marrow Transplant
stitutive signalling causes polycythaemia vera. Nature 2007;40:945-9.
2005;434:1144-8. 40. Bohnstedt C, Taskinen M, Zeller B, Bjorgvinsdottir H,
30. Scott LM, Tong W, Levine RL, Scott MA, Beer PA, Stratton Hafsteinsdottir S, Schmiegelow K. Poor treatment compliance
MR, et al. JAK2 exon 12 mutations in polycythemia vera and in children with down syndrome and acute lymphoblastic
idiopathic erythrocytosis. N Engl J Med 2007;356:459-68. leukemia. J Pediatr Hematol Oncol 2009;31:79-80.
31. Malinge S, Ben-Abdelali R, Settegrana C, Radford-Weiss I, 41. Shah N, Al-Ahmari A, Al-Yamani A, Dupuis L, Stephens D,
Debre M, Beldjord K, et al. Novel activating JAK2 mutation in Hitzler J. Outcome and toxicity of chemotherapy for acute
a patient with Down syndrome and B-cell precursor acute lymphoblastic leukemia in children with Down syndrome.
lymphoblastic leukemia. Blood 2007;109:2202-4. Pediatr Blood Cancer 2009;52:14-9.
32. Bercovich D, Ganmore I, Scott LM, Wainreb G, Birger Y, 42. Matherly LH, Taub JW. Methotrexate pharmacology and
Elimelech A, et al. Mutations of JAK2 in acute lymphoblastic resistance in childhood acute lymphoblastic leukemia. Leuk
leukaemias associated with Down's syndrome. Lancet 2008; Lymphoma 1996;21:359-68.

| 16 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Acute myeloid leukemia

Acute myeloid leukemia biology


and leukemia stem cells

E. Gudgin A B S T R A C T
B. Huntly Acute myeloid leukemia (AML) is an extremely heterogeneous malignancy with many distinct sub-
groups defined by specific morphological, cytogenetic, molecular and gene expression characteristics.
Department of Haematology, This heterogeneity is also mirrored in the variability of the prognosis of individual patients with AML;
Cambridge Institute for Medical however, the majority of patients still die from the disease. AML was the first malignancy in which a
Research, UK cancer stem or initiating cell was demonstrated, and it is likely that this cellular compartment is
responsible for disease relapse and secondary resistance. The AML leukemia stem cell (LSC) is, there-
fore, a critical target for disease eradication. Evidence also exists for biological and phenotypic het-
Hematology Education: erogeneity within this AML LSC compartment, and this in part, may relate to the molecular complex-
the education program for the ity of the disease. It may also reflect the contribution of the target cell transformed to initiate the
annual congress of the European leukemia. This review will address the cellular pathogenesis of AML and the current controversies sur-
Hematology Association rounding the LSC in AML, such as its identity and the initial target cell of transformation. We will then
discuss the molecular pathogenesis of AML, particularly focusing on recurrent aberrations, which alter
2009;3:17-23 self-renewal and differentiation, two defining characteristics of normal stem cells. A greater under-
standing of LSC biology may allow molecular targeting of these LSC specific pathways, or of the LSC
itself, and should facilitate improved outcomes in AML.

ancer is caused by the acquisition of questions and discuss the identity and ori-

C multiple mutations within a cell,


with their interaction and cumulative
effects resulting in the full malignant pheno-
gins of the leukemia stem cell (LSC) in AML.
We will then discuss critical molecular path-
ways that alter self-renewal and differentia-
type. Self-renewal, either inherent within tion and likely control LSC biology and the
the cell initially transformed, or acquired AML hierarchy. We will also discuss the
early through somatic mutation, is an potential for therapeutically modulating
absolute requirement in this process. The these pathways.
evolution of the phenotype requires long-
lived cells, which can accumulate the requi- The cellular biology of acute myeloid leukemia
site number of mutations. Other require- Stem cells were first demonstrated in the
ments, such as increased proliferative hematopoietic system and are, by defini-
potential, evasion of apoptosis and differen- tion, able to self-renew and differentiate
tiation arrest occur over time with the into multiple lineages. They sit at the apex
acquisition of further mutations.1 Acute of a hierarchy of cells and their progeny pass
myeloid leukemia (AML) is an excellent through lineage restricted progenitor popu-
example of this process, with numerous lations before differentiating into fully
cytogenetic and molecular abnormalities mature, functional effector cells. This
described which alter these critical cellular process of differentiation is accompanied by
functions. In addition, proof-of-principle for a rapidly decreasing self-renewal potential,
the cancer stem cell hypothesis was first with no self-renewal potential occurring
demonstrated in AML.2-4 This hypothesis outside of the stem cell compartment
proposed that the malignant clone is (Figure 1A). Stem cells themselves are
arranged as a hierarchy, with a small popu- organised in a hierarchy, with long-term
lation of cancer stem or initiating cells con- self-renewing hematopoietic stem cells
tinuously replenishing the bulk of the (HSC) giving rise to short-term self-renew-
tumour, which itself lacks self-renewal abil- ing HSC, and then multipotent progenitors.
ity. Subsequently, cancer stem cells (CSCs) By analogy, cancers have long been pro-
have been observed in other leukemias and posed to also organise themselves in disor-
solid-organ tumours.5 However, recent stud- dered hierarchies, and this cancer stem cell
ies are questioning some assumptions about hypothesis was first demonstrated in AML.
CSCs.6-8 Furthermore, the origins of self-
renewal, where this self-renewal resides The identity of the leukemia stem cell in acute
within the cellular hierarchy of AML, and myeloid leukemia
how these properties either relate to the In seminal experiments over a decade ago,
combination of transforming mutations or Dick and colleagues from Toronto identified
to the cell initially transformed are also cells which could transfer disease in xeno-
unclear. In this review, we address these transplant experiments into immunocom-

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 17 |
14th Congress of the European Hematology Association

promised (NOD/SCID) murine recipients.2,3 These cells


only comprised a fraction of the total tumour bulk and
demonstrated a surface phenotype, lineage negative
(Lin–), CD34+/CD38– similar to that of normal HSC,
which reconstitutes non-malignant hematopoiesis
within the same recipients.2 Moreover,
Lin–/CD34+/CD38+ cells did not transfer the disease.
However, refinement of the phenotype of LSC in AML
has also suggested some important differences
between LSC and normal HSCs. Antigens present on
normal HSC but not on LSC include Thy1 (CD90) and
c-KIT (CD117),9 while conversely, antigens present on
LSC, but not or only at low level on HSCs, include the
IL3 receptor chain alpha (CD123),10 the C-type lectin- Figure 1. The balance of self-renewal and differentiation in
like molecule 1(CLL-1),11 the cell adhesion molecule normal hematopoiesis and transformation. (A) This
demonstrates that during progression through the stem
CD44,12 and the immunoglobulin superfamily member, cell compartment and into the progenitor compartment,
CD96.13 Importantly, extending the proven clinical util- self-renewal is lost while differentiation program s are
ity of antibody therapy in hematological malignan- being initiated. (B) This process is finely balanced, with
cies,14 these antigens, and others that are present or self-renewal and differentiation inversely correlated and
possibly antagonistic to each other. This is demonstrated
upregulated in AML LSC by comparison with normal in CEBPA deficient mice, which not only demonstrate a
HSC, could serve as a specific means to therapeutically defect in myeloid differentiation but also show increased
target LSC.15 self-renewal in reconstitution assays.
However, recent work has introduced a note of cau-
tion in the interpretation of many of these experi-
ments.6 It had been previously accepted that the
immunocompromised mice used as recipients had little the CD34+/CD38+ compartment was shown to contain
or no residual immune function and that the antibodies the majority of the LSC activity in many AML
used to define surface phenotypes were neutral. Bonnet patients.6 In normal human hematopoietic ontogeny,
and colleagues have demonstrated that in AML, cells that are CD34+/CD38+ correspond to a more
NOD/SCID mice, the strain of mouse used in many of mature committed progenitor cell population.
the above functional experiments to initially define and Furthermore, studies prospectively conducted in
further refine the AML LSC surface phenotype, may murine leukemia models also suggest that usually LSCs
still be able to immunologically clear cells bound by can develop from progenitor cells. In these experi-
commercially available antibodies, specifically CD38. ments, populations of myeloid committed cells that
In contrast, this did not occur in the more immunocom- lacked self-renewal properties but expressed a variety
promised NOD/SCID/IL2rγnull strain.6 Furthermore, out- of leukemia-associated fusion genes or mutations
side of the hematopoietic system, and again using (MOZ-TIF2,17 MLL-ENL,18 MLL-AF919 and CEBPA20)
NOD-SCID-IL2rγnull recipient mice, up to 27% of unse- were purified by flow cytometry, and subsequently
lected human melanoma cells formed tumours, com- able to generate AML when transplanted into mice.
pared to less than 0.001% in NOD-SCID mice.8 Evidence for progenitor transformation also exists in
Therefore, the choice of immunocompromised recipi- other forms of leukemia, such as acute lymphoblastic
ent mouse strain, the differing choice of markers used leukemia (ALL), where the LSC may be positive for the
to define LSC populations, and perhaps even the com- progenitor maker CD1921,22 and in myeloid blast trans-
mercial antibodies to these markers have influenced formation of chronic myeloid leukemia (CML), where
both the perceived identity of the LSC in AML and the the LSC appears to have the phenotype of a granulo-
likely size of this population. However, although there cyte-monocyte precursor (GMP).23,24 Taken all together,
is likely to be variability between different malignan- these data suggest that there is a greater variability in
cies in the proportion of cancer-initiating cells, in AML, the immune-phenotype of LSCs than previously
the current evidence supports LSCs as being only a thought, and that the LSC may arise from progenitor as
small proportion of the malignant population. well as stem cells (Figure 2).
Transplantation experiments have shown that engraft-
ment is increased in NOD-SCID-IL2rγnull mice, but, The molecular biology of acute myeloid leukemia
where quantitated, still only represents only around The heterogeneity of AML is reflected in the spec-
1% of the total cells.16 trum of mutations with which it is recurrently associat-
ed.25 These include numerical chromosome abnormali-
The origins of the leukemia stem cell in acute myeloid ties, such as trisomies and monosomies (e.g. trisomy 8
leukemia and monosomy 7), chromosomal rearrangements gen-
There are theoretical arguments for both primitive erating translocations [e.g. t(8;21) and t(15;17)], intersti-
and more differentiated cells as being the origin of tial deletions (del 5q and 7q) and amplifications (MLL-
LSCs in AML.4,15 Early studies suggested that the LSC in PTD, FLT3-ITD), and point mutations of specific critical
AML has a surface phenotype, Lin–/CD34+/CD38–. genes (NPMc, CEBPA, FLT3-TKD).25,26 In addition to
However, in recent experiments, when the inhibitory these genomic aberrations, it is becoming increasingly
effects of the CD38+ antibody on engraftment were clear that epigenetic abnormalities are also important in
corrected for by use of the NOD/SCID/IL2rγnull strain, the transcriptional dysregulation characteristic of AML.

| 18 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

Alteration of self-renewal pathways


Important pathways in normal HSC self-renewal
include the clustered HOX genes, the WNT/β-
CATENIN pathway, the PTEN/AKT/FOXO axis, BMI1
and polycomb group proteins and the NOTCH and
HEDGEHOG pathways.4,33 These same pathways are
also important for LSC self-renewal, but recent reports
have described differences in some critical aspects that
may potentially be exploited therapeutically in the

Figure 2. A proposed roadmap for LSC induction. The right


side of the figure demonstrates current interpretations of
the roadmap of normal myeloid ontogeny, with critical tran-
scription factors whose transcriptional program s control
the process shown. The left side of the panel speculates on
where transforming events occur within the stem and pro-
genitor compartments to generate LSC in AML evolution.

Figure 3. Reductionist model of AML generation. This is a


simplified model of the induction of AML, where the collab-
These lesions alter critical cellular processes and collab- oration of type 1 mutations, which confer a proliferative
orate to generate the full malignant phenotype. A sim- and survival advantage on cells, and type 2 mutations,
which impair differentiation and/or increase self-renewal,
plified system has been proposed, where the mutations lead to the development of AML. Selected representative
fall into complementation groups, predominantly examples of type 1 and type 2 mutations are shown.
affecting growth and survival (type 1 mutations) and
differentiation and self-renewal (type 2 mutations).27
Examples of these mutations are given in Figure 3.
While this model has proven helpful in our understand-
ing of such a complicated disease and has provided us
with critical processes to focus on therapeutically, it is
widely accepted to be an oversimplification. Many
mutations may have both type 1 and type 2 functions
directly, such as FLT3 mutations, which provide growth
and survival signals,28 yet also impair differentiation via
phosphorylation of CEBPA.29 In addition, DNA damage
caused by oncogenic tyrosine kinases30 and some chro-
mosomal fusion products, such as AML-ETO and PML-
RARA,31,32 may indirectly generate random mutations
which fall into a different complementation group. We
have chosen to focus on lesions, which alter the two
defining properties of stem cells: unlimited self-renewal
and multi lineage differentiation.
In order to maintain hematopoiesis throughout a life-
time, self-renewal must maintain the pool of HSCs,
whereby one or both daughter cells remain undifferen-
tiated and able to continue self-renewing. All stem cells
must regulate the relative balance between stem cell
maintenance and differentiation to replenish the
mature cells of an organ. If a stem cell is triggered to
begin differentiating, genes that maintain self-renewal
are switched off, and genes that enforce differentiation
are switched on (Figure 1 B, 2). A large network of stim-
ulatory and inhibitory genes, which are mostly tran-
scription factors, then directs these cells to their ulti- Figure 4. Alterations of critical self-renewal and differenti-
ation pathways in AML. Alterations of self-renewal and dif-
mate fate. Disruption of this balance leads to the unreg- ferentiation cooperate to generate AML. (A) An illustration
ulated self-renewal and block in differentiation seen in of how some critical self-renewal pathways are dysregulat-
AML. It is likely that these two processes are inversely ed in AML, and their relationship to recurrent genetic
lesions. Potential therapeutics, which might abrogate
regulated (Figure 1 A, B), as loss of self-renewal occurs these pathways, are shown in red. (B) An illustration of how
around the same time that differentiation begins, and it recurrent genetic lesions in AML target the expression and
is possible that the transcriptional program s which function of the master-regulators of myeloid differentiation
PU.1 and CEBPA. Potential therapeutics are shown in red.
drive these processes may be antagonistic.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 19 |
14th Congress of the European Hematology Association

future (Figure 4 A). pathway is common in AML patients and is down-


HOX genes. The HOX family of homeobox genes in stream of AML-associated mutations, such as those
which mammals are divided into four genomic clusters affecting RAS and the FLT-3 and C-KIT oncogenic tyro-
(A-D), encode DNA-binding transcription factors. In sine kinases.50 Inhibition of this pathway causes in vitro
normal bone marrow, HOX genes are predominantly apoptosis but also reduced clonagenic potential in AML
expressed in HSCs, and downregulated during terminal blasts51 and reduced NOD-SCID engraftment in vivo,
differentiation.34 They play a central role in develop- suggesting a LSC specific effect.51,52 Furthermore, as a
mental hematopoiesis and HSC self-renewal.35 single agent, rapamycin induced clinically significant
However, HOX genes are also globally dysregulated in responses in 4 of 9 patients with poor risk AML in a
the majority of patients with AML,36-38 and substantial phase I trial.52 Taken together, these data have prompt-
evidence implicates them in the pathogenesis of AML.39 ed the inclusion of mTOR/PI3K inhibitors in ongoing
Dysregulation of HOX genes by AML-associated muta- combination chemotherapy studies.
tions may be a central mechanism underlying the WNT/β-catenin pathway. The WNT/β-catenin
abnormal self-renewal of LSCs. Alterations of the (CTNNB1) pathway has been demonstrated to be con-
mixed lineage leukemia gene MLL, either transloca- stitutively activated in the majority of cases of human
tions or partial tandem duplications, collectively occur AML.53 This activation has been experimentally
in 5-10% of cases of AML, and are all associated with demonstrated to occur downstream of AML-associated
HOX gene dysregulation.36,40 In addition, it has also mutations, such as FLT3-ITD,56 AML1-ETO, PML-
been shown that other rarer leukemia-associated fusion RARA and PML-PLZF.54,55 In addition, α, β and γ catenin
proteins, such as CALM-AF10 and MOZ-CBP (MYST3- have all been implicated in the malignant self-renewal
CREBBP) also upregulate HOX genes, and that individ- of AML and myeloid blast crisis CML.23,55,57 Moreover,
ual HOX genes, such as HOXA9 and A10 are them- retroviral transduction of the LEF1 transcription factor,
selves occasionally rearranged with the NUP98 gene in an effector of the WNT/β-catenin pathway, generates
AML patients.39 Gene expression profiling of AML AML in a mouse model.58 This suggests potential ther-
patients with mutated nucleo-phosmin (NPMc), the apeutic options and small molecule inhibitors exist,
commonest acquired molecular abnormality in AML, which target the interaction of β-catenin and LEF/TCF
are highly associated with a stem cell molecular signa- transcription factors necessary for induction of the
ture that includes activation of HOX genes and the WNT/β-catenin pathway.59 Initial concerns that these
HOX-cofactor MEIS1.41 Furthermore, members of the agents would have unacceptable side-effects on normal
Drosophila Caudal-like CDX family, CDX2 and CDX4, stem cell function may prove unfounded as mouse
which regulate HOX genes in murine development,42 models have recently questioned the requirement for
have recently been shown to be significantly upregulat- WNT/β-catenin signaling in homeostatic
ed in the majority of human AML samples and may hematopoiesis.60,61 Therefore, similar agents may prove
represent a master regulator of HOX gene therapeutically useful in some forms of AML.
induction.43-45 However, despite their apparent impor-
tance, little is known about the downstream targets of Alteration of normal differentiation
HOX dysregulation, and this remains an important area Differentiation block of varying degrees is a cardinal
for future study. Interestingly, a recent report showed feature of AML,62 with the degree of block the major
that MLL rearranged leukemic cells are susceptible to determinant of the AML phenotype. The process of lin-
inhibition by GSK3-B inhibitors through a mechanism eage commitment and differentiation is tightly coordi-
which destabilises p27Kip1.46 Whether this treatment is nated by distinct transcriptional program s, which are
MLL specific or may be applicable to all AML cases in turn controlled by master-regulators. In the process
with HOX dysregulation remains to be seen. of myeloid determination and differentiation, the
PI3-K pathway. In normal HSC, quiescence and cell major master-regulators are the CEBPA and PU.1 tran-
cycle entry are very tightly controlled. Central to this scription factors.63
appears to be the PI3K-AKT-FoxO pathway, mediated CEBPA. The CCAAT/enhancer-binding-protein-α
through effectors, such as p21.47-49 Constitutive activa- (CEBPA) is a basic region leucine zipper (bZIP) tran-
tion of the PI3K pathway through targeted deletion of scription factor critical for myelopoiesis and growth
PTEN, the major lipid phosphatase attenuating PI3K control.64 It regulates a program of genes critical to
signaling, results in both HSC depletion and in the establishing myeloid identity and differentiation, such
development of acute leukemia in mice.47 The mice as G-CSF and secondary granule protein genes,65 and
demonstrated an initial myeloproliferative phase that also limits proliferation via E2F mediated inhibition of
was quickly followed by an acute leukemia that was c-MYC.64 The protein is expressed in two isoforms,
transplantable to secondary recipients.47 Inhibition of with differing transcriptional start sites, a longer p42
this pathway with the mTOR inhibitor rapamycin protein and a shorter p30 protein. Both contain the pro-
restored normal function to PTEN-/- HSCs and prevent- tein-protein interaction and DNA binding domains, but
ed the development of leukemia. In addition, in trans- they differ in that only p42 contains all of the transac-
plant experiments, rapamycin also depleted the num- tivation domains.64 CEBPA expression is detectable first
ber of leukemia-initiating cells and, importantly, pro- in committed myeloid progenitors, and its expression is
longed survival, even when administered to mice with upregulated as cells commit to mature granulocytes65,66
established leukemia. This shows that mechanistic dif- (Figure 2). Mice deficient in CEBPA die perinatally due
ferences between LSCs and HSCs may provide a ther- to a defect in glucose homeostasis, but they also com-
apeutic target, without destruction of the HSC com- pletely lack mature neutrophilic and eosinophilic gran-
partment. Similarly, constitutive activation of the PI3K ulocytes.67 The inverse correlation of self-renewal and

| 20 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

differentiation previously alluded to is demonstrated quency in AML patients with a complex karyotype. In
by the fact that mice deficient in CEBPA have an the homozygous state, this polymorphism abolishes
increased self-renewal and repopulating capacity, binding of the chromatin remodelling agent SATB1,
although they do not develop leukemia.68 reduces the enhancer activity of the URE and decreases
CEBPA may be dysregulated by a number of mecha- the expression of PU.1.87 PU.1 is also targeted by
nisms in AML (Figure 4B). Mutations of the CEBPA leukemia-associated mutations, such as FLT3-ITD,
gene, both acquired and inherited, occur. They are AML1-ETO and PML-RARA. AML1-ETO physically
described sporadically in around 10% of AML cases,69,70 binds to PU.1 and displaces its co-activator JUN, there-
and are associated with a relatively good prognosis.71 by downregulating its transcriptional activity,88 where-
There are multiple mutations described which mainly as PU.1 expression is downregulated by FLT3-ITD,89
lead to the generation of only the p30 isoform or alter and PML-RARA.90
the DNA binding of the p42 protein. Their unifying The paradigm of differentiation therapy already
feature appears to be a down-modulation of, or domi- exists for AML, where ATRA has revolutionized the
nant negative effect on CEBPA function.64 Mouse mod- treatment of the PML-RARA-associated promyelocytic
els engineered to expressed only p30 CEBPA or mutant variant. As an extension to this paradigm, therapeutic
CEBPA developed AML with complete penetrance.72,73 upregulation of CEBPA and PU.1 should alter the AML
Expression of CEBPA is also downregulated through phenotype and Figure 4B shows the potential mecha-
hypermethylation of CpG islands at the genes promot- nisms to achieve this. Furthermore, in the future, it may
er in a significant proportion of patients with AML.74-76 be possible to design peptodomimetics of CEBPA and
In addition, leukemia associated mutations can also tar- PU.1, or to block the interference by leukemia-associat-
get the translation of CEBPA, as occurs in myeloid ed fusion genes by redirection therapy91 or by systemic
transformation of CML, where high levels of BCR-ABL delivery of shRNA.63
induce the RNA binding protein hnRNP E2, which
facilitates degradation of the CEBPA transcript.77
Recurrent mutations associated with AML also disrupt References
the function of CEBPA. These include the PML-RARA,
AML1-ETO (RUNX1-RUNX1T1) and AML1/EVI1 gene 1. Hanahan D,Weinberg R. The hallmarks of cancer. Cell 2000;
100:57-70.
rearrangements associated with the t(15;17), t(8;21) and 2. Lapidot T, Sirard C, Vormoor J, Murdoch B, Hoang T,
t(3;21) translocations. They physically interact with Caceres-Cortes J, et al. A cell initiating human acute myeloid
CEBPA and either recruit transcriptional repressors, leukaemia after transplantation into SCID mice. Nature 1994;
prevent its binding with co-factors or impair its DNA 367:645-8.
3. Bonnet D, Dick JE. Human acute myeloid leukemia is organ-
binding, thereby blocking its transcriptional activity at ized as a hierarchy that originates from a primitive hemato-
target loci.78,79 Finally, CEBPA function can also be poietic cell. Nat Med 1997;3:730-7.
blocked by post-translational modification, such as 4. Huntly BJ, Gilliland DG. Leukaemia stem cells and the evolu-
tion of cancer-stem-cell research. Nat Rev Cancer 2005;5:311-
occurs in FLT3 mutant AML cells.29 This block appears 21.
to be dependent upon ERK1/2 phosphorylation of 5. Kvinlaug BT, Huntly BJ. Targeting cancer stem cells. Expert
residue serine 21 and as can be reversed by FLT3 or Opin Ther Targets 2007;11:915-27.
MEK1 inhibitors,29 which may be a useful therapeutic 6. Taussig DC, Miraki-Moud F, Anjos-Afonso F, Pearce DJ, Allen
K, Ridler C, et al. Anti-CD38 antibody-mediated clearance of
strategy. human repopulating cells masks the heterogeneity of
PU.1. The PU.1 transcription factor is a hematopoiet- leukemia-initiating cells. Blood 2008;112:568-75.
ic specific ETS family member, involved in the develop- 7. Kelly PN, Dakic A, Adams JM, Nutt SL, Strasser, A. Tumor
growth need not be driven by rare cancer stem cells. Science
ment of all hematopoietic lineages.63 PU.1 is highly 2007;317:337.
expressed in myelomonocytic cells and GMPs, with 8. Quintana E, Shackleton M, Sabel MS, Fullen DR, Johnson TM,
more primitive cells such as common myeloid progen- Morrison SJ. Efficient tumour formation by single human
melanoma cells. Nature 2008;456:593-8.
itors(CMP), and HSC expressing PU.1 at lower levels66 9. Blair A, Hogge DE, Ailles LE, Lansdorp PM, Sutherland HJ.
(Figure 2). Many genes critical for myeloid differentia- Lack of expression of Thy-1 (CD90) on acute myeloid
tion are described as targets for PU.1 and include G- leukemia cells with long-term proliferative ability in vitro and
CSF, M-CSF and GM-CSF.62 Complete loss of PU.1 is in vivo. Blood 1997;89:3104-12.
10. Jordan CT, Upchurch D, Szilvassy SJ, Guzman ML, Howard
embryonically lethal, and evidence also suggests that DS, Pettigrew AL, et al. The interleukin-3 receptor alpha
PU.1 is required for HSC function.83,84 However, chain is a unique marker for human acute myelogenous
although conditional loss of PU.1 does not lead to leukemia stem cells. Leukemia 2000;14:1777-84.
11. van Rhenen A, van Dongen GA, Kelder A, Rombouts EJ,
leukemia, a mouse engineered to express a graded Feller N, Moshaver B et al. The novel AML stem cell associat-
reduction of PU.1 to around 20% does develop AML,85 ed antigen CLL-1 aids in discrimination between normal and
suggesting that at least some PU.1 function is required leukemic stem cells. Blood. 2007;110:2659-66.
12. Jin L, Hope KJ, Zhai Q, Smadja-Joffe F, Dick JE. Targeting of
to mandate myeloid fate, malignant self-renewal or CD44 eradicates human acute myeloid leukemic stem cells.
both. Nat Med 2006;12:1167-74.
Many different processes in AML (Figure 4 B) target 13. Hosen N, Park CY, Tatsumi N, Oji Y, Sugiyama H, Gramatzki
PU.1. These include direct mutation, which occurs in M, et al. CD96 is a leukemic stem cell-specific marker in
human acute myeloid leukemia. Proc Natl Acad Sci USA
around 7% of patients. In general, these mutations alter 2007;104:11008-13.
the interaction of PU.1 with its protein partners AML1 14. Abutalib SA, Tallman MS. Monoclonal antibodies for the
and JUN, leading to decreased transcriptional activity.86 treatment of acute myeloid leukemia. Curr Pharm Biotechnol
2006;7:343-69.
Although no germ line mutations have been described 15. Chan WI, Huntly BJ. Leukemia stem cells in acute myeloid
for PU.1, a polymorphism in its upstream regulatory leukemia. Semin Oncol 2008;35:326-35.
element (URE) has been demonstrated at a higher fre- 16. Kennedy JA, Barabe F, Poeppl AG, Wang JC, Dick JE.
Comment on "Tumor growth need not be driven by rare can-

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 21 |
14th Congress of the European Hematology Association

cer stem cells". Science 2007;318:1722. author reply. 41. Deschamps J, van Nes J. Developmental regulation of the Hox
17. Huntly BJ, Shigematsu H, Deguchi K, Lee BH, Mizuno S, genes during axial morphogenesis in the mouse. Develop-
Duclos N, et al. MOZ-TIF2, but not BCR-ABL, confers prop- ment 2005;132:2931-42.
erties of leukemic stem cells to committed murine 42. Bansal D, Scholl C, Fröhling S, McDowell E, Lee BH, Döhner
hematopoietic progenitors. Cancer Cell 2004;6:587-96. K et al. Cdx4 dysregulates Hox gene expression and generates
18. Cozzio A, Passegué E, Ayton PM, Karsunky H, Cleary ML, acute myeloid leukemia alone and in cooperation with
Weissman IL. Similar MLL-associated leukemias arising from Meis1a in a murine model. Proc Natl Acad Sci USA 2006; 103:
self-renewing stem cells and short-lived myeloid progenitors. 16924-9.
Genes Dev 2003;17:3029-35. 43. Scholl C, Bansal D, Döhner K, Eiwen K, Huntly BJ, Lee BH et
19. Krivtsov AV, Twomey D, Feng Z, Stubbs MC, Wang Y, Faber al. The homeobox gene CDX2 is aberrantly expressed in
J et al. Transformation from committed progenitor to leu- most cases of acute myeloid leukemia and promotes leuke-
kaemia stem cell initiated by MLL-AF9. Nature 2006;442: mogenesis. J Clin Invest 2007;117:1037-48.
818-22. 44. Rawat VP, Thoene S, Naidu VM, Arseni N, Heilmeier B,
20. Kirstetter P, Anderson K, Porse BT, Jacobsen SE, Nerlov C. Metzeler K et al. Overexpression of CDX2 perturbs HOX
Activation of the canonical Wnt pathway leads to loss of gene expression in murine progenitors depending on its N-
hematopoietic stem cell repopulation and multilineage differ- terminal domain and is closely correlated with deregulated
entiation block. Nat Immunol 2006;7:1048-56. HOX gene expression in human acute myeloid leukemia.
21. Castor A, Nilsson L, Astrand-Grundström I, Buitenhuis M, Blood 2007.
Ramirez C, Anderson K, et al. Distinct patterns of hematopoi- 45. Wang Z, Smith KS, Murphy M, Piloto O, Somervaille TC,
etic stem cell involvement in acute lymphoblastic leukemia. Cleary ML et al. Glycogen synthase kinase 3 in MLL
Nat Med 2005;11:630-7. leukaemia maintenance and targeted therapy. Nature 2008;
22. le Viseur C, Hotfilder M, Bomken S, Wilson K, Röttgers S, 455:1205-9.
Schrauder A, et al. In childhood acute lymphoblastic leukemia, 46. Yilmaz OH, Valdez R, Theisen BK, Guo W, Ferguson DO, Wu
blasts at different stages of immunophenotypic maturation H, Morrison SJ. Pten dependence distinguishes haematopoiet-
have stem cell properties. Cancer Cell 2008;14:47-58. ic stem cells from leukaemia-initiating cells. Nature 2006;
23. Jamieson CH, Ailles LE, Dylla SJ, Muijtjens M, Jones C, 441:475-82.
Zehnder JL et al. Granulocyte-macrophage progenitors as 47. Tothova Z, Kollipara R, Huntly BJ, Lee BH, Castrillon DH,
candidate leukemic stem cells in blast-crisis CML. N Engl J Cullen DE et al. FoxOs Are Critical Mediators of
Med 2004;351:657-67. Hematopoietic Stem Cell Resistance to Physiologic Oxidative
24. Jamieson C, et al. Bioluminescent imaging of human Stress. Cell 2007;128:325-339.
Leukemic stem cell engraftment. ASH Annual Meeting 48. Cheng T, Rodrigues N, Shen H, Yang Y, Dombkowski D,
Abstracts 2005;106:696. Sykes M, Scadden DT. Hematopoietic stem cell quiescence
25. Estey E, Dohner H. Acute myeloid leukaemia. Lancet 2006; maintained by p21cip1/waf1. Science 2000;287:1804-8.
368:1894-907. 49. Martelli A M, Nyåkern M, Tabellini G, Bortul R, Tazzari PL,
26. Kelly L, Gilliland D. Genetics of myeloid leukemias. Ann Rev Evangelisti C, Cocco L. Phosphoinositide 3-kinase/Akt signal-
Genomics Hum Genetics 2002;3:179-98. ing pathway and its therapeutical implications for human
27. Gilliland DG, Griffin JD. The roles of FLT3 in hematopoiesis acute myeloid leukemia. Leukemia 2006;20:911-28.
and leukemia. Blood 2002;100:1532-42. 50. Xu Q, Thompson JE, Carroll M. mTOR regulates cell survival
28. Radomska HS, Bassères DS, Zheng R, Zhang P, Dayaram T, after etoposide treatment in primary AML cells. Blood 2005;
Yamamoto Y, et al. Block of C/EBP alpha function by phos- 106:4261-8.
phorylation in acute myeloid leukemia with FLT3 activating 51. Recher C, Beyne-Rauzy O, Demur C, Chicanne G, Dos
mutations. J Exp Med 2006;203:371-81. Santos C, Mas VM et al. Antileukemic activity of rapamycin
29. Skorski T. Oncogenic tyrosine kinases and the DNA-damage in acute myeloid leukemia. Blood 2005;105:527-34.
response. Nat Rev Cancer 2002;2:351-60. 52. Simon M, Grandage VL, Linch DC, Khwaja A. Constitutive
30. Alcalay M, Meani N, Gelmetti V, Fantozzi A, Fagioli M, activation of the Wnt/beta-catenin signalling pathway in
Orleth A, et al. Acute myeloid leukemia fusion proteins acute myeloid leukaemia. Oncogene 2005;24:2410-20.
deregulate genes involved in stem cell maintenance and DNA 53. Muller-Tidow C, Steffen B, Cauvet T, Tickenbrock L, Ji P,
repair. J Clin Invest 2003;112:1751-61. Diederichs S et al. Translocation products in acute myeloid
31. Viale A, De Franco F, Orleth A, Cambiaghi V, Giuliani V, Bossi leukemia activate the Wnt signalling pathway in hematopoi-
D, et al. Cell-cycle restriction limits DNA damage and main- etic cells. Mol Cell Biol 2004;24:2890-903.
tains self-renewal of leukaemia stem cells. Nature 2009;457: 54. Zheng X, Beissert T, Kukoc-Zivojnov N, Puccetti E,
51-6. Altschmied J, Strolz C et al. Gamma-catenin contributes to
32. Reya T, Morrison S, Clarke M, Weissman I. Stem cells, can- leukemogenesis induced by AML-associated translocation
cer, and cancer stem cells. Nature 2001;414:105-111. products by increasing the self-renewal of very primitive pro-
33. Pineault N, Helgason CD, Lawrence HJ, Humphries RK. genitor cells. Blood 2004;103:3535-43.
Differential expression of Hox, Meis1, and Pbx1 genes in 55. Tickenbrock L, Schwäble J, Wiedehage M, Steffen B, Sargin B,
primitive cells throughout murine hematopoietic ontogeny. Choudhary C et al. Flt3 tandem duplication mutations coop-
Exp Hematol 2002;30:49-57. erate with Wnt signaling in leukemic signal transduction.
34. Owens BM, Hawley RG. HOX and non-HOX homeobox Blood 2005;105:3699-706.
genes in leukemic hematopoiesis. Stem Cells 2002;20:364-79. 56. Liu TX, Becker MW, Jelinek J, Wu WS, Deng M, Mikhal-
35. Armstrong SA, Staunton JE, Silverman LB, Pieters R, den Boer kevich N et al. Chromosome 5q deletion and epigenetic sup-
ML, Minden MD et al. MLL translocations specify a distinct pression of the gene encoding alpha-catenin (CTNNA1) in
gene expression profile that distinguishes a unique leukemia. myeloid cell transformation. Nat Med 2007;13:78-83.
Nat Genet 2002;30:41-7. 57. Petropoulos, K Arseni N, Schessl C, Stadler CR, Rawat VP,
36. Bullinger L, Döhner K, Bair E, Fröhling S, Schlenk RF, Tib- Deshpande AJ et al. A novel role for Lef-1, a central transcrip-
shirani R et al. Use of gene-expression profiling to identify tion mediator of Wnt signaling, in leukemogenesis. J Exp Med
prognostic subclasses in adult acute myeloid leukemia. N Engl 2008;205:515-22.
J Med 2004;350:1605-16. 58. Sukhdeo K, Mani M, Zhang Y, Dutta J, Yasui H, Rooney MD
37. Valk PJ, Verhaak RG, Beijen MA, Erpelinck CA, Barjesteh van et al. Targeting the beta-catenin/TCF transcriptional complex
Waalwijk van Doorn-Khosrovani S, Boer JM et al. Pro- in the treatment of multiple myeloma. Proc Natl Acad Sci
gnostically useful gene-expression profiles in acute myeloid USA 2007;104:7516-21.
leukemia. N Engl J Med 2004;350:1617-28. 59. Koch U, et al. Simultaneous loss of {beta}- and {gamma}-
38. Frohling S, Scholl C, Bansal D, Huntly BJ. HOX Gene catenin does not perturb hematopoiesis or lymphopoiesis.
Regulation in Acute Myeloid Leukemia: CDX Marks the Blood 2007.
Spot? Cell Cycle 2007;6. 60. Scheller M, Huelsken J, Rosenbauer F, Taketo MM,
39. Dorrance AM, Liu S, Yuan W, Becknell B, Arnoczky KJ, Birchmeier W, Tenen DG et al. Hematopoietic stem cell and
Guimond M et al. Mll partial tandem duplication induces multilineage defects generated by constitutive beta-catenin
aberrant Hox expression in vivo via specific epigenetic alter- activation. Nat Immunol 2006;7:1037-47.
ations. J Clin Invest 2006;116:2707-16. 61. Tenen DG. Disruption of differentiation in human cancer:
40. Alcalay M, Tiacci E, Bergomas R, Bigerna B, Venturini E, AML shows the way. Nat Rev Cancer 2003;3:89-101.
Minardi SP et al. Acute myeloid leukemia bearing cytoplas- 62. Rosenbauer F, Tenen DG. Transcription factors in myeloid
mic nucleophosmin (NPMc+ AML) shows a distinct gene development: balancing differentiation with transformation.
expression profile characterized by up-regulation of genes Nat Rev Immunol 2007;7;105-17.
involved in stem-cell maintenance. Blood 2005;106:899-902. 63. Nerlov CC. EBPalpha mutations in acute myeloid leukaemias.

| 22 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

Nat Rev Cancer 2004;4:394-400. inhibitory action of hnRNP E2. Nat Genet 2002;30:48-58.
64. Radomska HS, Huettner CS, Zhang P, Cheng T, Scadden DT, 77. Westendorf JJ, Yamamoto CM, Lenny N, Downing JR, Selsted
Tenen DG et al. CCAAT/enhancer binding protein alpha is a ME, Hiebert SW et al. The t(8;21) fusion product, AML-1-
regulatory switch sufficient for induction of granulocytic ETO, associates with C/EBP-alpha, inhibits C/EBP-alpha-
development from bipotential myeloid progenitors. Mol Cell dependent transcription and blocks granulocytic differentia-
Biol 1998;18:4301-14. tion. Mol Cell Biol 1998;18:322-33.
65. Akashi K, Traver D, Miyamoto T, Weissman IL. A clonogenic 78. Tokita K, Maki K, Mitani K. RUNX1/EVI1, which blocks
common myeloid progenitor that gives rise to all myeloid lin- myeloid differentiation, inhibits CCAAT-enhancer binding
eages. Nature 2000;404:193-7. protein alpha function. Cancer Sci 2007;98:1752-7.
66. Zhang DE, Zhang P, Wang ND, Hetherington CJ, Darlington 79. Scott EW, Simon MC, Anastasi J, Singh H. Requirement of
GJ, Tenen DG et al. Absence of granulocyte colony-stimulat- transcription factor PU.1 in the development of multiple
ing factor signaling and neutrophil development in CCAAT hematopoietic lineages. Science 1994;265:1573-7.
enhancer binding protein alpha-deficient mice. Proc Natl 80. McKercher SR, Torbett BE, Anderson KL, Henkel GW, Vestal
Acad Sci USA 1997;94:569-74. DJ, Baribault H et al. Targeted disruption of the PU.1 gene
67. Zhang P, Iwasaki-Arai J, Iwasaki H, Fenyus ML, Dayaram T, results in multiple hematopoietic abnormalities. EMBO J
Owens BM et al. Enhancement of hematopoietic stem cell 1996;5:5647-58.
repopulating capacity and self-renewal in the absence of the 81. Rosenbauer F, Owens BM, Yu L, Tumang JR, Steidl U, Kutok
transcription factor C/EBP alpha. Immunity 2004;21:853-63. JL et al. Lymphoid cell growth and transformation are sup-
68. Pabst T, Mueller BU, Zhang P, Radomska HS, Narravula S, pressed by a key regulatory element of the gene encoding
Schnittger S et al. Dominant-negative mutations of CEBPA, PU.1. Nat Genet 2006;38:27-37.
encoding CCAAT/enhancer binding protein-alpha 82. Kim HG, de Guzman CG, Swindle CS, Cotta CV, Gartland L,
(C/EBPalpha), in acute myeloid leukemia. Nat Genet 2001; Scott EW, Klug CA et al. The ETS family transcription factor
27:263-70. PU.1 is necessary for the maintenance of fetal liver
69. Smith ML, Cavenagh JD, Lister TA, Fitzgibbon J. Mutation of hematopoietic stem cells. Blood 2004;104:3894-900.
CEBPA in familial acute myeloid leukemia. N Engl J Med 83. Iwasaki H, Somoza C, Shigematsu H, Duprez EA, Iwasaki-
2004;351:2403-7. Arai J, Mizuno S et al. Distinctive and indispensable roles of
70. Frohling S, Schlenk RF, Stolze I, Bihlmayr J, Benner A, PU.1 in maintenance of hematopoietic stem cells and their
Kreitmeier S et al. CEBPA mutations in younger adults with differentiation. Blood 2005;106:1590-600.
acute myeloid leukemia and normal cytogenetics: prognostic 84. Rosenbauer F, Wagner K, Kutok JL, Iwasaki H, Le Beau MM,
relevance and analysis of cooperating mutations. J Clin Oncol Okuno Y et al. Acute myeloid leukemia induced by graded
2004;22:624-33. reduction of a lineage-specific transcription factor, PU.1. Nat
71. Kirstetter P, Schuster MB, Bereshchenko O, Moore S, Dvinge Genet 2004;36:624-30.
H, Kurz E et al. Modeling of C/EBPalpha mutant acute 85. Mueller BU, Pabst T, Osato M, Asou N, Johansen LM,
myeloid leukemia reveals a common expression signature of Minden MD et al. Heterozygous PU.1 mutations are associat-
committed myeloid leukemia-initiating cells. Cancer Cell ed with acute myeloid leukemia. Blood 2003;101:2074.
2008;13:299-310. 86. Steidl U, Steidl C, Ebralidze A, Chapuy B, Han HJ, Will B et
72. Porse BT, Bryder D, Theilgaard-Mönch K, Hasemann MS, al. A distal single nucleotide polymorphism alters long-range
Anderson K, Damgaard I et al. Loss of C/EBP alpha cell cycle regulation of the PU.1 gene in acute myeloid leukemia. J Clin
control increases myeloid progenitor proliferation and trans- Invest 2007;117:2611-20.
forms the neutrophil granulocyte lineage. J Exp Med 2005; 87. Vangala RK, Heiss-Neumann MS, Rangatia JS, Singh SM,
202:85-96. Schoch C, Tenen DG et al. The myeloid master regulator
73. Hackanson B, Bennett KL, Brena RM, Jiang J, Claus R, Chen transcription factor PU.1 is inactivated by AML1-ETO in
SSet al. Epigenetic modification of CCAAT/enhancer binding t(8;21) myeloid leukemia. Blood 2003;101:270-7.
protein alpha expression in acute myeloid leukemia. Cancer 88. Mizuki M, Schwable J, Steur C, Choudhary C, Agrawal S,
Res 2008;68:3142-51. Sargin B et al. Suppression of myeloid transcription factors
74. Figueroa ME, Wouters BJ, Skrabanek L, Glass J, Li Y, and induction of STAT response genes by AML-specific Flt3
Erpelinck-Verschueren CA et al. Genome wide epigenetic mutations. Blood 2003;101:3164-73.
analysis delineates a biologically distinct immature acute 89. Mueller BU, Pabst T, Fos J, Petkovic V, Fey MF, Asou N et al.
leukemia with myeloid/T-lymphoid features. Blood 2009. ATRA resolves the differentiation block in t(15;17) acute
75. Wouters BJ, Jordà MA, Keeshan K, Louwers I, Erpelinck- myeloid leukemia by restoring PU.1 expression. Blood 2006;
Verschueren CA, Tielemans D et al. Distinct gene expression 107:3330-8.
profiles of acute myeloid/T-lymphoid leukemia with silenced 90. Steffen B, Serve H, Berdel WE, Agrawal S, Linggi B, Büchner
CEBPA and mutations in NOTCH1. Blood 2007;110:3706-14. T et al. Specific protein redirection as a transcriptional thera-
76. Perrotti D, Cesi V, Trotta R, Guerzoni C, Santilli G, Campbell py approach for t(8;21) leukemia. Proc Natl Acad Sci USA
K et al. BCR-ABL suppresses C/EBPalpha expression through 2003;100:8448-53.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 23 |
Acute myeloid leukemia

Minimal residual disease detection


in acute myeloid leukemias

G. Saglio A B S T R A C T
E. Messa
D. Cilloni Monitoring of residual disease (MRD) in patients with acute myeloid leukemia (AML) is now recog-
nized as an important diagnostic tool for assessing the response to treatment and the individual risk
of relapse. The study of minimal residual disease has been greatly promoted by the technologic advent
Department of Clinical and of multiparameter flow cytometry (MFC), polymerase chain reaction (PCR), and by the identification
Biological Sciences, University of of the genetic lesions involved in AML pathogenesis. Several studies have demonstrated that quanti-
Turin, Italy tative assessment of MRD by MFC is feasible in most AML cases, and may be of prognostic signifi-
cance. In parallel, using available molecular markers, including the recently recognized NPM1 muta-
tions in addition to fusion genes, MRD assessment by real-time quantitative PCR is now possible in
Hematology Education: more than half the AML patients with a sensitivity of at least 10–4. Finally, over-expression of the
the education program for the Wilms’ tumor gene (WT1) occurs in most AML patients, and represents a reliable marker for MRD
annual congress of the European assessment and quantification in 80-85% of AML patients. Recently, a large European Leukaemia
Hematology Association Netstudy has shown incorporating early assessment of MRD through a standardized WT1 assay may
help to enhance the risk stratification of AML patients.
2009;3:24-29

urrent treatment protocols for acute titative real-time quantitative PCR (RQ-

C myeloid leukemia (AML) are partly


based on prognostic factors, including
age, white blood cell (WBC) count,
PCR).17-18
In most AMLs, very sensitive, end-point
qualitative RT-PCR techniques are used to
immunophenotypic profiles, specific chro- reveal the presence and persistence of the
mosomal aberrations and other molecularly residual disease shortly after the end of
detectable abnormalities, such as FLT3 and induction and consolidation therapy. A
NMP1 gene defects.1-8 Individual patient out- notable exception includes acute promyelo-
come, however, cannot always be reliably cytic leukemia (APL), for which RT-PCR still
predicted just on the basis of these classical represents the gold standard for MRD deter-
parameters, and it is well known that mination by exploiting the presence of the
response to initial therapy may provide a PML-RARa fusion transcripts associated
further prognostic marker. Monitoring of with the presence of the t(15;17) transloca-
minimal residual disease (MRD) in AML tion. However, positive results from these
patients is now recognised as an important assays do not necessarily imply an increased
diagnostic tool that can be used to assess the risk of relapse.19-22 RQ-PCR, which intro-
response to treatment and to establish the duced an easy and rapid way to rank the
risk of relapse in the individual patient.9 PCR-positive patients into different sub-cat-
During the last 20 years, different tech- egories according to the amount of their
niques have enabled the detection of MRD, was able to fill this gap. Standardised
leukemic cells beyond the threshold of mor- RQ-PCR procedures for the most common
phology or conventional cytogenetics, types of fusion transcripts present in AML
which has generally had a detection of have also been developed, allowing the
around 5%. The newer techniques, which application of this innovative technology to
are principally based on immunophenotypic large-scale MRD studies within multi-centre
or polymerase chain reaction (PCR) analy- therapeutic trials.18
sis,10,11 have, in most instances, the sensitivi- The most common fusion genes (FG) tran-
ty to detect at least one leukemic cell out of scripts are present in only about 30% of all
103 background cells. The advent of multipa- AML cases, and are generally associated
rameter flow cytometry (MFC) greatly with corresponding well-known cytogenetic
improved immunophenotypic analysis. abnormalities. However, approximately
MFC allows the identification of leukemia- 50% of AMLs display a normal karyotype
associated aberrant immunophenotypes (NK).23 This problem has been partly over-
(LAIPs) at diagnosis.12-16 Similarly, the study come by the discovery of new frequent
of MRD was originally fuelled by the pro- molecular defects preferentially found in
gressive identification of the recurrent normal karyotype AML, such as FLT3 inter-
molecular lesions that are associated with nal tandem duplication (ITD) and NPM1
AML and by the advent of PCR technology mutations.7,24 Apart from serving as impor-
that includes either qualitative reverse-tran- tant prognostic markers, mutations in the
scriptase-PCR (RT-PCR) or, especially, quan- NMP1 gene have been demonstrated to rep-

| 24 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

resent reliable and robust markers for MRD testing and types, AMLs may simultaneously show more than a
quantification, and efforts to standardize these proce- single LAIP. The choice of LAIPs for MRD detection,
dures are currently ongoing.25 therefore, depends on several factors, including i) speci-
Given that approximately 30-40% of all AML cases ficity, such that the percentage of LAIP expression on
may still lack a specific and stable genetic target, such as normal BM cells should ideally be less than 0.1%; ii)
those described previously, genes that are highly and sensitivity, such that the percentage of LAIP expression
preferentially over-expressed in AML have been investi- on the leukemic blast population at diagnosis should be
gated as suitable markers for MRD analysis. In particu- at least 10%; iii) stability, such that LAIPs should not
lar, in approximately 85% of AML cases,26 the amount undergo phenotypic shifts during the time course of the
of Wilms’ tumor gene (WT1) gene transcript in AML cells disease.12-16 Although infrequent, in some cases, the last
is at least three logs higher than in normal bone marrow is known to occur between diagnosis and relapse.31-34
(BM). Since RQ-PCR may clearly distinguish between This aspect, along with the fact that it is not as specific
normal and abnormal levels of WT1 expression, WT1 as PCR, represents one of the major limitations of this
has been proposed as a type of universal marker for method. Conversely, this method has several advan-
MRD assessment in AML, particularly in those cases tages, such as being relatively easy, cheap, rapid and
that lack another suitable genetic marker.27 Recent data applicable for most cases (>80%).29,30 Although studies
from our laboratory, as well as data obtained from a of MRD detection by MFC in AML are limited and may
large European standardization study, recently promot- show some discrepancies when compared with ALL,
ed by the European Leukaemia Net, support the notion they indicate that MFC may represent a useful method
that assessment of WT1 transcript levels in both periph- for predicting relapse. In particular, most studies per-
eral blood (PB) and BM specimens collected post-consol- formed on AML samples from specific clinical trials
idation may bear important and independent prognostic show that the amount of MRD present after consolida-
significance in terms of the risk of relapse.28 tion and, in some studies, after induction, strongly cor-
relates with relapse-free survival (RFS).35-46 At the
Multiparameter flow cytometry moment, the lack of a sufficient degree of standardisa-
Flow cytometry may be the most accessible method tion among different laboratories hampers a precise
for MRD detection, because of its wide availability in identification of the best and most common LAIPs to
most hematological centres. Using an accurate combi- use. In addition, another major limitation of this tech-
nation of monoclonal antibodies (MoAbs) that are con- nology, if it is to be widely used in common clinical
jugated to different fluorochromes, immunophenotypic practice outside specialised centres and controlled trials,
detection of MRD in AML can be performed by identi- is the lack of precise thresholds at specific time-points
fying abnormal combinations or expressions of markers during follow-up, which define the significant MRD
on malignant cells at diagnosis.12,14 These markers would amounts for the risk of relapse.
either be not present or very infrequently present on
normal blood or BM cells.13-15 These are referred to as Reverse-transcriptase polymerase chain reaction
leukemia-associated aberrant immunophenotypes Several clinical studies have shown that chromosomal
(LAIPs), and can be subsequently used during follow-up aberrations and the corresponding fusion genes that are
to track the amount and behavior of the residual present in AML can be used for risk group classification
leukemic clone(s).29,30 LAIPS mainly derive from asyn- (Figure 1).2-47 In addition to their potential use for the
chronous antigen expression, antigen over-expression, cytogenetic/molecular classification of acute leukemia
lineage infidelity or the absence of lineage-specific anti- at diagnosis, chromosomal aberrations with leukemia-
gens. With respect to acute lymphoblastic leukemia specific fusion-gene regions can also be used as PCR tar-
(ALL) cells, which show more homogeneous pheno- gets for the detection of MRD during follow-up, with

Figure 1. Comparison of the sen-


sitivity of methods currently used
to detect and to quantify mini-
mal residual disease in AML.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 25 |
14th Congress of the European Hematology Association

sensitivities that generally reach 10–4 to 10–5.17 PCR constant expression level in all cell types at any stage of
analysis of fusion genes is based upon the design of maturation in BM and peripheral blood. A large
oligonucleotide primers located on opposite sides of the European cooperative study demonstrated that ABL,
breakpoint fusion regions, so that the PCR product con- GUS and β2 microglobulin might represent optimal con-
tains the tumour-specific fusion sequences. In most trols.18 This method avoids a number of false negative
types of chromosomal aberrations with fusion genes, results that can occur due to RNA degradation, one of
the breakpoints in different patients are spread over 10 the major pitfalls of RT-PCR. RQ-PCR targets may
kb or more, which represent distances that are difficult include fusion-gene transcripts, specific gene defects,
to span on a routine basis by PCR on DNA. However, specific mutations and aberrantly expressed genes.
in many acute leukemias, the fusion gene is transcribed
into a fusion mRNA, which can serve as the PCR target Real-time quantitative-PCR for quantitative assessment of
after reverse transcription into cDNA.48-49 fusion genes
The persistence of leukemia-specific hybrid tran- AML1-ETO and CBFB-MYH11 fusion transcripts,
scripts in the bone marrow and peripheral blood of which are associated respectively with the t(8;21)
patients with a t(8;21) or inv(16) AML at the end of the translocation and the inv(16), are the most common
conventional therapy is normal, and apparently compat- fusion genes present in AML. These two fusion tran-
ible with a durable hematological remission.50-52 This has scripts are also generally associated with favourable
favoured the shift towards RQ-PCR for monitoring outcomes. Up to 40% of these patients, however, may
most cases of AML.53,54 Despite this observation, RT- experience relapse, and so early identification of
PCR still remains the method of choice, not only for patients at high risk of relapse is extremely important to
confirmation of APL at diagnosis, but also to monitor determine clinical and therapeutic decisions. Several
MRD in this type of leukemia.19,55 Standardised condi- RQ-PCR studies have clearly shown that a log reduction
tions for RT-PCR analysis of PML-RARa FG transcripts decrease in the fusion transcripts, present at diagnosis,
have been developed by the BIOMED-1 Concerted at distinct time points of the treatment are prognostical-
Action, and different primer sets designed that allow ly important. In some studies,62-64 the prognostic value of
the detection of the various PML-RARa FG transcripts.17 post-induction AML1-ETO levels has been shown in
In the last decade, the availability of differentiation both childhood and adult AML, but other studies sug-
therapy with all-trans retinoic acid (ATRA) has pro- gest that the post-consolidation level may be more rele-
duced a remarkable improvement in the outcome of vant than the one achieved post-induction.65,66 Specific
patients with APL. The challenge is how to identify the thresholds of FG amounts after post-induction and post-
relatively small subgroup of patients, who have a partic- consolidation therapy have also been suggested to cor-
ular risk of relapse, and cannot be reliably distinguished relate with risk relapse. In contrast to AML1-ETO posi-
based on pre-treatment characteristics, and could poten- tive AML, positive PCR results that occur, even after
tially benefit from more intensive or alternative treat- several years of continuous complete remission (CCR),
ments during the first remission. During the last few have been described in CBFB-MYH11-positive AMLs,67
years, there has been general agreement from a number with only those patients who finally achieve a negative
of studies that a positive PML-RARa test after consolida- PCR result being able to persist with CCR. Long-term
tion is a strong predictor of subsequent hematological monitoring studies have suggested that a consistent
relapse, whereas repeatedly negative results are associ- increase in the FG amounts at any time during follow-
ated with long-term survival in the majority of these up is indicative of an impending relapse.68-70
patients.55-59 The Italian GIMEMA group reported that
recurrence of positive PCR results, which were detected Real-time quantitative-PCR for quantification of aberrant
by three-month BM surveillance performed after the FLIT3 and NPM1 transcripts
completion of therapy, was highly predictive of relapse. FLT3-ITD is the most frequent genetic lesion in AML,
The use of this strategy successfully predicted approxi- accounting for 25-30% and reaching a 40% frequency
mately 70% of relapses.60 Although the benefit of early in AMLs that have a normal karyotype, and is associat-
treatment at the time of molecular relapse has still to be ed with a bad prognosis.71 Investigational protocols with
definitely proven, preliminary evidence supports such a FLT3 tyrosine-kinase inhibitors are currently ongoing. A
strategy. In addition, considering that there have been large study demonstrated that FLT3-ITD transcripts
relatively few studies that report the use of RQ-PCR in could be used as markers for monitoring treatment
APL patients,61 the molecular monitoring of APL by tra- response by PCR.72 However, the necessity to use
ditional RT-PCR must be regarded as one of the most patient specific primers in order to achieve a sufficient
relevant examples of the impact of molecular genetics in sensitivity, as well as the instability of this marker at
clinical hematology. relapse observed in some cases, are both pitfalls pre-
venting widespread use of this analysis.
Real-time quantitative polymerase chain reaction NPM1 mutations are the most frequent genetic
RQ-PCR is now the method of choice in PCR-based changes in AMLs that have a normal karyotype.6 Many
quantification in AML patients. Although RQ-PCR has different NPM1 mutation variants have been described,
been reported to be less sensitive than nested RT-PCR, but the most common variant (MutA) represents 75%
it allows for the exact determination of the amount of of all cases, with the inclusion of two other mutations
residual disease and provides the sensitivity to detect accounting for more than 90% of the total. Therefore,
the residual disease within a given sample. This can be the use of three different primer-specific assays provides
established by measuring the amount of a so-called a reliable and highly sensitive RQ-PCR test for MRD
“control gene,” which is a gene that has a stable and monitoring, which can be performed in most patients

| 26 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

with NPM1 mutations.26 Recently, some studies have


speculated on the prognostic significance of this analy- References
sis, so we are still waiting confirmation from large 1. Cheson BD, Bennett JM, Kopecky KJ, Büchner T, Willman CL,
prospective clinical studies that use a standardized Estey EH, et al. Revised recommendations of the International
approach, such as the one currently planned within the Working Group for Diagnosis, Standardization of Response
Criteria, Treatment Outcomes, and Reporting Standards for
context of the European Leukaemia Net. Therapeutic Trials in Acute Myeloid Leukemia. J Clin Oncol
2003;21:4642-9.
Real-time quantitative-PCR for Wilms’ tumor gene overex- 2. Estey E, Döhner H. Acute myeloid leukemia. Lancet 2006;368:
1894-907.
pression 3. Grimwade D. The clinical significance of cytogenetic abnor-
The presence of AML patients who present with malities in acute myeloid leukaemia. Best Pract Res Clin
detectable genetic aberrations prompted several investi- Haematol 2001;14:497-529.
4. Mrózek K, Heerema NA, Bloomfield CD. Cytogenetics in
gators to validate the use of alternative markers for acute leukemia. Blood Rev 2004;18:115-36.
MRD detection that are suitable for use in the vast 5. Kottaridis PD, Gale RE, Linch DC. Flt3 mutations and
majority of AML patients. The Wilms’s tumor gene codes leukaemia. Br J Haematol 2003;122:523-38.
6. Falini B, Mecucci C, Tiacci E, Alcalay M, Rosati R, Pasqualucci
for a transcription factor has been shown to be highly L, et al. Cytoplasmic nucleophosmin in acute myelogenous
expressed in several hematopoietic tumours, including leukemia with a normal karyotype. N Engl J Med 2005; 352:
AMLs.27-28,73-76 Given that WT1 is expressed in normal BM 254-66.
7. Mrózek K, Bloomfield CD. Chromosome aberrations, gene
and peripheral blood, studies that use qualitative RT- mutations and expression changes, and prognosis in adult
PCR have provided conflicting results on the clinical acute myeloid leukemia. Hematology Am Soc Hematol Educ
value of this marker. However, the most recent investi- Program 2006;169-77.
8. Schlenk RF, Döhner K, Krauter J, Fröhling S, Corbacioglu A,
gations by RQ-PCR have been able to clearly distin- Bullinger L, et al. Mutations and treatment outcome in cytoge-
guish WT1 transcript levels found in AML cells, normal netically normal acute myeloid leukemia. N Engl J Med 2008;
hematopoietic and post-chemotherapeutic cells, and 358:1909-18.
9. Freeman SD, Jovanovic JV, Grimwade D. Development of
regenerating normal BM cells.28 Therefore, longitudinal minimal residual disease-directed therapy in acute myeloid
RQ-PCR analysis of WT1 transcript levels may prove to leukemia. Semin Oncol 2008;4:388-400.
be clinically relevant for AML monitoring. Since WT1 10. Campana D, Pui CH. Detection of minimal residual disease in
acute leukemia: methodologic advances and clinical signifi-
expression in normal PB is about one log lower than in cance. Blood 1995;85:1416-34.
normal BM, with the majority of normal PB samples 11. Campana D. Determination of minimal residual disease in
scoring negative, a sequential RQ-PCR study of WT1 leukemia patients. Br J Haematol 2003;121:823-38.
12. Macedo A, Orfao A, Vidriales MB, López-Berges MC,
expression in PB may further improve the sensitivity of Valverde B, González M, et al. Characterization of aberrant
MRD evaluation in AML, and also favour compliance phenotypes in acute myeloblastic leukemia. Ann Hematol
and sample availability. A study, which analyzed WT1 1995;70:189-94.
expression levels at diagnosis and during follow-up in 13. Macedo A, Orfao A, Gonzalez M, Vidriales MB, López-Berges
MC, Martínez A, et al. Immunological detection of blast cell
82 AML patients who were treated with standard subpopulations in acute myeloblastic leukemia at diagnosis:
chemotherapy protocols, provided important prognos- implications for minimal residual disease studies. Leukemia
tic information in AML by earlier identification of 1995;9:993-8.
14. Reading CL, Estey EH, Huh YO, Claxton DF, Sanchez G,
patients who were at a higher risk of relapse early after Terstappen LW, et al. Expression of unusual immunopheno-
induction chemotherapy.77 A recent, large European type combinations in acute myelogenous leukemia. Blood
cooperative study clearly demonstrated that WT1 is 1993;81:3083-90.
15. Kern W, Danhauser-Riedl S, Ratei R, Schnittger S, Schoch C,
overexpressed when compared to controls in more than Kolb HJ, et al. Detection of minimal residual disease in unse-
85% of AML cases. In cases with high-level expression, lected patients with acute myeloid leukemia using multipara-
MRD assessment following induction showed that a meter flow cytometry for definition of leukemia-associated
immunophenotypes and determination of their frequencies in
greater response in terms of WT1 transcript reduction normal bone marrow. Haematologica 2003;88:646-53.
was associated with a reduced risk of relapse, and that 16. Al-Mawali A, Gillis D, Hissaria P, Lewis I. Incidence, sensitiv-
this correlation remained significant after adjusting for ity, and specificity of leukemia-associated phenotypes in acute
myeloid leukemia using specific five-color multiparameter
age, WBC and cytogenetics. In addition, the failure to flow cytometry. Am J Clin Pathol 2008;129:934-45.
normalise WT1 transcript levels by the post-consolida- 17. van Dongen JJ, Macintyre EA, Gabert JA, Delabesse E, Rossi V,
tion time-point predicted a significantly increased risk Saglio G, et al. Standardized RT-PCR analysis of fusion gene
transcripts from chromosome aberrations in acute leukemia
of relapse with a highly significant prognostic value. for detection of minimal residual disease. Report of the BIO-
MED-1 Concerted Action: investigation of minimal residual
Conclusions disease in acute leukemia. Leukemia 1999;13:1901-28.
18. Gabert J, Beillard E, van der Velden VH, Bi W, Grimwade D,
Quantification of MRD provides the opportunity to Pallisgaard N, et al. Standardization and quality control stud-
improve prognostication significantly in AML and plays ies of ‘real-time’ quantitative reverse transcriptase polymerase
a major role as a stratification parameter in guiding risk- chain reaction of fusion gene transcripts for residual disease
detection in leukemia a Europe Against Cancer program.
adapted therapy. MFC may already be applied to most Leukemia 2003;17:2318-57.
patients with AML with a clinically relevant sensitivity, 19. Lo Coco F, Diverio D, Falini B, Biondi A, Nervi C, Pelicci PG.
but RQ-PCR is more sensitive, and the identification of Genetic diagnosis and molecular monitoring in the manage-
ment of acute promyelocytic leukemia. Blood 1999;94:12-22.
new suitable molecular markers is leading to increase 20. Jurlander J, Caligiuri MA, Ruutu T, Baer MR, Strout MP,
further its future applicability. Prospective studies, how- Oberkircher AR, et al. Persistence of the AML1/ETO fusion
ever, are needed to better define which checkpoints in transcript in patients treated with allogeneic bone marrow
transplantation for t(8;21) leukemia. Blood 1996;88:2183-91.
specific therapeutic protocols are the most appropriate 21. Miyamoto T, Nagafuji K, Akashi K, Harada M, Kyo T, Akashi
for the risk stratification of AML patients in response to T, et al. Persistence of multipotent progenitors expressing
treatment. AML1/ETO transcripts in long-term remission patients with
t(8;21) acute myelogenous leukemia. Blood 1996;87:4789-96.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 27 |
14th Congress of the European Hematology Association

22. Kern W, Haferlach C, Haferlach T, Schnittger S. Monitoring of 41. Kern W, Voskova D, Schoch C, Hiddemann W, Schnittger S,
Minimal Residual Disease in Acute Myeloid Leukemia. Haferlach T. Determination of relapse risk based on assess-
Cancer 2008;112:4-16. ment of minimal residual disease during complete remission
23. Grimwade D, Walker H, Oliver F, Wheatley K, Harrison C, by multiparameter flow cytometry in unselected patients with
Harrison G, et al. The importance of diagnostic cytogenetics acute myeloid leukemia. Blood 2004;104:3078-85.
on outcome in AML: analysis of 1,612 patients entered into 42. Kern W, Voskova D, Schoch C, Schnittger S, Hiddemann W,
the MRC AML 10 trial. The Medical Research Council Adult Haferlach T. Prognostic impact of early response to induction
and Children’s Leukaemia Working Parties. Blood 1998;92: therapy as assessed by multiparameter flow cytometry in
2322-33. acute myeloid leukemia. Haematologica 2004;89:528-40.
24. Nakao M, Yokota S, Iwai T, Kaneko H, Horiike S, Kashima K, 43. Venditti A, Maurillo L, Buccisano F, Del Poeta G, Mazzone C,
et al. Internal tandem duplication of the flt3 gene found in Tamburini A, et al. Pretransplant minimal residual disease
acute myeloid leukemia. Leukemia 1996;10:1911-18. level predicts clinical outcome in patients with acute myeloid
25. Gorello P, Cazzaniga G, Alberti F, Dell'Oro MG, Gottardi E, leukemia receiving high-dose chemotherapy and autologous
Specchia G, et al. Quantitative assessment of minimal residual stem cell transplantation. Leukemia 2003;17:2178-82.
disease in acute myeloid leukemia carrying nucleophosmin 44. Gianfaldoni G, Mannelli F, Baccini M, Antonioli E, Leoni F,
(NPM1) gene mutations. Leukemia 2006;20:1103-8. Bosi A. Clearance of leukaemic blasts from peripheral blood
26. Inoue K, Ogawa H, Sonoda Y, Kimura T, Sakabe H, Oka Y, et during standard induction treatment predicts the bone mar-
al. Aberrant overexpression of the Wilms’ tumour gene (WT1) row response in acute myeloid leukaemia: a pilot study. Br J
in human leukemia. Blood 1997;89:1405-12. Haematol 2006;134:54-7.
27. Cilloni D, Gottardi E, De Micheli D, Serra A, Volpe G, Messa 45. Buccisano F, Maurillo L, Gattei V, Del Poeta G, Del Principe
F, et al. Quantitative assessment of WT1 expression by real MI, Cox MC, et al. The kinetics of reduction of minimal resid-
time quantitative PCR may be a useful tool for monitoring ual disease impacts on duration of response and survival of
minimal residual disease in acute leukemia patients. patients with acute myeloid leukemia. Leukemia
Leukemia. 2002;16:2115-21. 2006;20:1783-9.
28. Cilloni D, Messa F, Arruga F, Defilippi I, Gottardi E, Fava M, et 46. Laane E, Derolf AR, Bjorklund E, Mazur J, Everaus H,
al. Early prediction of treatment outcome in acute myeloid Söderhäll S, et al. The effect of allogeneic stem cell transplan-
leukemia by measurement of WT1 transcript levels in periph- tation on outcome in younger acute myeloid leukemia
eral blood samples collected after chemotherapy. Haema- patients with minimal residual disease detected by flow
tologica 2008;93:921-4. cytometry at the end of post-remission chemotherapy.
29. Kern W, Schnittger S. Monitoring of acute myeloid leukemia Haematologica 2006;91:833-6.
by flow cytometry. Curr Oncol Rep 2003;5:405-12. 47. Szczepanski T, Orfao A, van der Velden V, San Miguel JF, van
30. Al-Mawali A, Gillis D, Lewis I. The Role of Multiparameter Dongen JJ. Minimal residual disease in leukaemia patients.
Flow Cytometry for Detection of Minimal Residual Disease in Lancet Oncol 2001;2:409-17.
Acute Myeloid Leukemia. Am J Clin Pathol 2009;131:16-26. 48. Claxton DF, Liu P, Hsu HB, Marlton P, Hester J, Collins F, et al.
31. Kern W, Haferlach T, Schnittger S, Ludwig WD, Hiddemann Detection of fusion transcripts generated by the inversion 16
W, Schoch C. Karyotype instability between diagnosis chromosome in acute myelogenous leukemia. Blood 1994;
andrelapse in 117 patients with acute myeloid leukemia: 83:1750-6.
implications for resistance against therapy. Leukemia 2002; 16: 49. Perego RA, Marenco P, Bianchi C, Cairoli R, Urbano M, Nosari
2084-91. AM, et al. PML/RAR alpha transcripts monitored by poly-
32. Oelschlagel U, Nowak R, Schaub A, Köppel C, Herbst R, merase chain reaction in acute promyelocytic leukemia during
Mohr B, et al. Shift of aberrant antigen expression at relapse or complete remission, relapse and after bone marrow transplan-
at treatment failure in acute leukemia. Cytometry 2000;42: tation. Leukemia 1996;10:207-12.
247-53. 50. Nucifora G, Larson RA, Rowley JD. Persistence of the 8;21
33. Macedo A, San Miguel JF, Vidriales MB, López-Berges MC, translocation in patients with acute myeloid leukemia type
García-Marcos MA, Gonzalez M, et al. Phenotypic changes in M2 in long-term remission. Blood 1993;82:712-5.
acute myeloid leukaemia: implications in the detection of min- 51. Tobal K, Johnson PR, Saunders MJ, Harrison CJ, Liu Yin JA.
imal residual disease. J Clin Pathol 1996;49:15-8. Detection of CBFB/MYH11 transcripts in patients with inver-
34. Voskova D, Schoch C, Schnittger S, Hiddemann W, Haferlach sion and other abnormalities of chromosome 16 at presenta-
T, Kern W. Stability of leukemia-associated aberrant immuno- tion and remission. Br J Haematol 1995;91:104.
phenotypes in patients with acute myeloid leukemia between 52. Costello R, Sainty D, Blaise D, Gastaut JA, Gabert J, Poirel H,
diagnosis and relapse: comparison with cytomorphologic, et al. Prognosis value of residual disease monitoring by poly-
cytogenetic, and molecular genetic findings. Cytometry B Clin merase chain reaction in patients with CBF/MYH11-positive
Cytom 2004;62:25-38. acute myeloblastic leukemia. Blood 1997;89:2222.
35. San Miguel JF, Martinez A, Macedo A, Vidriales MB, López- 53. Schnittger S, Weisser M, Schoch C, Hiddemann W, Haferlach
Berges C, González M, et al. Immunophenotyping investiga- T, Kern W. New score predicting for prognosis in PML-
tion of minimal residual disease is a useful approach for pre- RARA+, AML1-ETO+, or CBFBMYH11+ acute myeloid
dicting relapse in acute myeloid leukemia patients. Blood leukemia based on quantification of fusion transcripts. Blood
1997;90:2465-70. 2003;102:2746-55.
36. Venditti A, Buccisano F, Del Poeta G, Maurillo L, Tamburini A, 54. Tobal K, Liu Yin JA. Monitoring of minimal residual disease by
Cox C, et al. Level of minimal residual disease after consolida- quantitative reverse transcriptase-polymerase chain reaction
tion therapy predicts outcome in acute myeloid leukemia. for AML1-MTG8 transcripts in AML-M2 with t(8;21). Blood
Blood 2000;96:3948-52. 1996;88:3704-9.
37. San Miguel JF, Vidriales MB, Lopez-Berges C, Díaz-Mediavilla 55. Perego RA, Marenco P, Bianchi C, Cairoli R, Urbano M, Nosari
J, Gutiérrez N, Cañizo C, et al. Early immunophenotypical AM, et al. PML/RAR alpha transcripts monitored by poly-
evaluation of minimal residual disease in acute myeloid merase chain reaction in acute promyelocytic leukemia during
leukemia identifies different patient risk groups and may con- complete remission, relapse and after bone marrow transplan-
tribute to postinduction treatment stratification. Blood tation. Leukemia 1996;10:207-12.
2001;98:1746-51. 56. Miller Jr WH, Levine K, DeBlasio A, Frankel SR, Dmitrovsky
38. Kern W, Haferlach T, Schoch C, Loffler H, Gassmann W, E, Warrell Jr RP. Detection of minimal residual disease in acute
Heinecke A, et al. Early blast clearance by remission induction promyelocytic leukemia by a reverse transcription polymerase
therapy is a major independent prognostic factor for both chain reaction assay for the PML/RAR-alpha fusion mRNA.
achievement of complete remission and long-term outcome in Blood 1993;82:1689-94.
acute myeloid leukemia: data from the German AML 57. Koller E, Karlic H, Krieger O, Mistrik M, Michlmayr G, Gadner
Cooperative Group (AMLCG) 1992 Trial Blood 2003;101:64- H, et al. Early detection of minimal residual disease by reverse
70. transcriptase polymerase chain reaction predicts relapse in
39. Coustan-Smith E, Ribeiro RC, Rubnitz JE, Razzouk BI, Pui acute promyelocytic leukemia. Ann Hematol 1995;70:75-8.
CH, Pounds S, et al. Clinical significance of residual disease 58. Korninger L, Knobl P, Laczika K, Mustafa S, Quehenberger P,
during treatment in childhood acute myeloid leukaemia. Br J Schwarzinger I,et al. PML-RAR alpha PCR positivity in the
Haematol 2003;123:243-52. bone marrow of patients with APL precedes haematological
40. Sievers EL, Lange BJ, Alonzo TA, Gerbing RB, Bernstein ID, relapse by 2-3 months. Br J Haematol 994;88:427-31.
Smith FO, et al. Immunophenotypic evidence of leukemia 59. Diverio D, Pandolfi PP, Biondi A, Avvisati G, Petti MC,
after induction therapy predicts relapse: results from a Mandelli F, et al. Absence of reverse transcription-polymerase
prospective Children’s Cancer Group study of 252 patients chain reaction detectable residual disease in patients with
with acute myeloid leukemia. Blood 2003;101:3398-406. acute promyelocytic leukemia in long-term remission. Blood

| 28 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

199;82:3556-9. state. Blood 2002;99:443-9.


60. Diverio D, Rossi V, Avvisati G, De Santis S, Pistilli A, Pane F, 69. Guerrasio A, Pilatrino C, De Micheli D, Cilloni D, Serra A,
et al. Early detection of relapse by prospective reverse tran- Gottardi E et al. Assessment of minimal residual disease
scriptase-polymerase chain reaction analysis of the (MRD) in CBFbeta/MYH11-positive acute myeloid leukemias
PML/RARalpha fusion gene in patients with acute promyelo- by qualitative and quantitative RT-PCR amplification of fusion
cytic leukemia enrolled in the GIMEMA-AIEOP multicenter transcripts. Leukemia 2002;16:1176-1.
‘‘AIDA’’ trial. GIMEMA-AIEOP Multicenter ‘‘AIDA’’ Trial 70. Marcucci G, Caligiuri MA, Dohner H, Archer KJ, Schlenk RF,
Blood 1998;92:784-9. Döhner K, et al. Quantification of CBFbeta/MYH11 fusion
61. Gallagher RE, Yeap BY, Bi W, Livak KJ, Beaubier N, Rao S, et transcript by real time RT-PCR in patients with INV(16) acute
al. Quantitative real-time RT-PCR analysis of PML-RAR alpha myeloid leukemia. Leukemia 2001;15:1072-80.
mRNA in acute promyelocytic leukemia: assessment of prog- 71. Schlenk RF, Döhner K, Krauter J, Fröhling S, Corbacioglu A,
nostic significance in adult patients from intergroup protocol Bullinger L, et al. Mutations and treatment outcome in cytoge-
0129. Blood 2003;101:2521-8. netically normal acute myeloid leukemia. N Engl J Med 2008;
62. Sugimoto T, Hiranmoy D, Shion I, et al. Quantitation of min- 358:1909-18.
imal residual disease in t(8;21)-positive acute myelogenous 72. Schnittger S, Schoch C, Dugas M, Kern W, Staib P, Wuchter C.
leukemia patients using real-time quantitative RT-PCR. Am J Analysis of FLT3 length mutations in 1003 patients with acute
Hematol 2000;64:101-6. myeloid leukemia: correlation to cytogenetics, FAB subtype,
63. Fujimaki S, Funato T, Harigae H, et al. A quantitative reverse and prognosis in the AMLCG study and usefulness as a mark-
transcriptase polymerase chain reaction method for the detec- er for the detection of minimal residual disease. Blood 2002;
tion of leukaemic cells with t(8;21) in peripheral blood. Eur J 100:59-66.
Haematol 2000;64:252-8. 73. Ostergaard M, Olesen LH, Hasle H, et al. WT1 gene expres-
64. Viehmann S, Teigler-Schlegel A, Bruch J, et al. Monitoring of sion: an excellent tool for monitoring minimal residual disease
minimal residual disease (MRD) by real-time quantitative in 70% of acute myeloid leukaemia patients-results from a sin-
reverse transcription PCR (RQ-RT-PCR) in childhood acute gle-centre study. Br J Haematol 2004;125:590-600.
myeloid leukemia with AML1/ETO rearrangement. Leukemia 74. Cilloni D, Gottardi E, Messa F, Fava M, Scaravaglio P, Bertini
2003;17:1130-6. M,, et al. Significant correlation between the degree of WT1
65. Perea G, Lasa A, Aventin A, et al. Prognostic value of minimal expression and the International Prognostic Scoring System
residual disease (MRD) in acute myeloid leucemia (AML) with Score in patients with myelodysplastic syndromes. J Clin
favorable cytogenetics [t(8;21) and inv(16)]. Leukemia 2006; Oncol 2003;21:1988-95.
20:87-94. 75. Cilloni D, Saglio G. Usefulness of quantitative assessment of
66. Schnittger S, Schoch C. Quantitative PCR based minimal Wilms tumor suppressor gene expression in chronic myeloid
residual disease detection in core binding factors leukemias: leukemia patients undergoing imatinib therapy. Semin
Prognostication and guiding of therapy. Leukemia Res 2006; Hematol 2003:40;37-41.
30:657-8. 76. Cilloni D, Messa F, Martinelli G, Gottardi E, Arruga F, Defilippi
67. Marcucci G, Caligiuri MA, Bloomfield CD. Defining the I, et al. WT1 transcript amount discriminates secondary or
‘absence’ of CBFbeta/MYH11 fusion transcript in patients reactive eosinophilia from idiopathic hypereosinophilic syn-
with acute myeloid leukemia and inversion of chromosome drome or chronic eosinophilic leukemia. Leukemia 2007;21:
16 to predict long term complete remission: a call for defini- 1442-50.
tion. Blood 1997;90:5022-4. 77. Cilloni D, Messa F, Arruga F, Defilippi I, Gottardi E, Fava M, et
68. Buonamici S, Ottaviani E, Testoni N, Montefusco V, Visani G, al. Early prediction of treatment outcome in acute myeloid
Bonifazi F et al. Real-time quantitation of minimal residual dis- leukemia by measurement of WT1 transcript levels in periph-
ease in inv(16)-positive acute myeloid leukaemia may indicate eral blood samples collected after chemotherapy. Haemato-
risk for clinical relapse and may identify patients in a curable logica 2008;93:921-4.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 29 |
Acute myeloid leukemia

Risk-adapted treatment for adult acute myeloid


leukemia

J. Sierra A B S T R A C T
S. Brunet Advances in the understanding of the pathophysiology of acute myeloid leukemia (AML) and the
recognition of new prognostic categories underline the need for risk-adapted treatment. Cumulative
Hematology Department, Hospital de experience with cytotoxic agents has been helpful in defining who may benefit from intensive
la Santa Creu i Sant Pau, chemotherapy. Elderly AML patients and those with severe comorbidities are normally candidates for
Autonomous University of Barcelona, conservative approaches or investigational agents with low extra-hematological toxicity. Intensior
Spain chemotherapy is indicated in the remainder. In the absence of favourable genetic markers, most stud-
ies support performing HLA-identical sibling transplantation in the first remission of AML. If an HLA-
compatible relative is not available, unrelated donors or cryopreserved umbilical cord blood are alter-
Hematology Education: natives when the risk of AML recurrence is high. Of note, reduced intensity conditioning extends
the education program for the hematopoietic transplantation to elderly and debilitated patients. Efforts are being made for further
annual congress of the European characterizing AML cases by novel molecular techniques. These techniques could also be applied to
Hematology Association predict sensitivity to treatment and to anticipate individual toxicities. Therapy against antigen, genet-
ic, epigenetic and vascular targets is also under investigation. Given the complexity and interactions
2009;3:30-37
of molecular pathways involved in AML, progress with this approach could be slower than other hema-
tologic disorders. However, this is an exciting challenge for the next years.

cute myeloid leukemia (AML) is now novel agents is actively being investigated

A recognised as a disease with a wide


clinical and biological diversity.1
Discovery of this heterogeneity is the con-
in other AML categories. The goal is to cure
most cases of AML or transform the disease
into a chronic disorder.
sequence of substantial progress in the
understanding of its pathophysiology. In Initial risk-adapted decision
the recently published World Health It is generally accepted that reaching
Organization classification of AML, the complete remission (CR) is the requisite to
variety of genetic findings in AML has been prolong survival in AML patients.4 Conse-
taken into account.2 Many of the included quently, intensive chemotherapy is usually
entities constitute new prognostic cate- recommended, except in patients of very
gories. With the introduction of antigen advanced age and/or those with severe
and/or molecular targeted therapies, the comorbidities. In practice, patients over 80
relevance of the precise characterization of years old usually only receive palliative
the disease is further emphasised.3 After measures and their life expectancy is not
initial treatment or during follow-up, it is longer than a few weeks. Decisions con-
now time to adjust AML therapy, not only cerning the intensity of treatment for AML
to classical factors, such as age and preced- patients between 65 and 80 years of age
ing hematologic disorders, but to individual depend on the anticipated probability of
co-morbidities, genetics of leukemic cells death due to therapy complications. The
and the presence of minimal residual dis- results expected from combination
ease (MRD). After many years of homoge- chemotherapy in this age interval have not
neous treatment with a modest improve- been clearly defined, as published trials fre-
ment in outcome, it is now becoming quently have the selection bias of including
apparent that an individualised therapeutic the healthiest patients. In such trials, CR
approach for AML could be the most rates range from 40-60%.5,6 The poor toler-
appropriate strategy. ance to intensive treatment and the com-
This review summarizes current knowl- mon development of chemoresistance in
edge useful for tailoring treatment in adult this age group justify the investigation of
AML. Pediatric AML and acute promyelo- innovative approaches with low extra-
cytic leukemia (APL) are excluded, even hematological toxicity, particularly in
though the latter is the paradigm for how patients with low leukemic burden. Within
targeted treatment may overcome the clinical trials, demethylating drugs such as
pathogenetic events leading to leukemia. 5-azacytidine or decitabine with/without
Based on the success in APL, the strategy of histone deacetylase inhibitors (valproic
combining classic cytotoxic drugs with acid, vorinostat), and antiangiogenic agents,

| 30 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

such as lenalidomide are being investigated.5,7,8,9


Hematologic improvement is observed in approxi-
mately 30-40% of AML patients treated with these
agents. Preliminary experiences suggest that the
response to demethylating drugs may not be affected
by the cytogenetics of leukemic cells. These agents
may prolong survival, even in the absence of CR crite-
ria, and pose fewer life-threatening complications than
combinations of cytotoxic drugs; furthermore, they
often allow outpatient management. Clofarabine, a
new purine analog, has remarkably low extra-hemato-
logical toxicity. It is presently being tested as a single
drug, and in combination with classic and novel agents
in adult patients with adverse prognosis AML.10
Gemtuzumab ozogamycin (GO) is another option to
consider in patients ineligible for intensive combina-
tion chemotherapy, although prolonged myelosup- Figure 1. Survival of patients with primary acute myeloid
pression is constant, and liver toxicity is to be expect- leukemia treated with intensive chemotherapy according
ed in a number of cases.11,12 In Europe, access to this to the Spanish CETLAM protocols: effect of age at diagno-
agent is restricted to a compassionate basis. A more sis.
classical alternative without significant extra-hemato-
logical toxicity is to administer subcutaneous low dose
cytarabine. In patients unfit for intensive chemothera-
py, this drug prolongs survival when compared to be of particular interest in determining treatment of
hydroxyurea.13 elder patients.22,23 Specific scores predicting death due
AML patients up to the age of 60-65 years are candi- to tumour lysis syndrome have also been reported.24
dates for intensive induction treatment, preferably Patients at high-risk for this complication should be
within cooperative clinical trials. The proportion of CR carefully monitored during CT, and supportive meas-
in these younger adults ranges between 65% and 85%, ures are of special relevance.
with regimens including an anthracycline or anthra- Chemoresistance is more frequent in elderly AML
cenodione for 3 days, and conventional-dose cytara- patients, mainly because they have a higher propor-
bine for 7-10 days.3,4 Twenty to thirty-five percent of tion of adverse cytogenetics.4,5,6 Currently, the cytoge-
patients need two courses of induction treatment to netics of AML cells are the most determinant factor on
achieve CR, and this group has an increased risk of short- and long-term sensitivity to chemotherapy in
leukemia recurrence. In most cases, a higher dose of any age group.25,26,27 Several classifications of cytogenet-
cytarabine or the addition of a third drug has no ic findings based on prognostic categories in AML
impact on the CR rate. In the HOVON experience, G- have been published by cooperative groups in Europe
CSF priming has been shown to be of benefit in the and the United States. These include the MRC,26,27
intermediate-risk cytogenetic group by improving the EORTC-GIMEMA,28 HOVON-SAKK,29 SWOG-
quality of remission, thereby, leading to a decrease in ECOG,30 and CALGB.31,32 Patients with adverse cytoge-
relapse incidence.14 This favourable effect has not been netics have a probability of refractoriness to
observed in other studies.15 By adding GO to induction chemotherapy as high as 40-50%. The poor outcome
chemotherapy, the Medical Research Council (MRC) of a complex karyotype harbouring three or more
of the United Kingdom observed a positive impact, not abnormalities could be further related to the presence
on CR rates, but on leukemia-free and overall survival of monosomies (two monosomies or one monosomy
of intermediate-risk AML patients.16 Within clinical tri- together with a structural abnormality) than to the
als, intensified CT, in case of slow blast clearance is number of chromosomal alterations.33 Other factors
another option that may improve the CR propor- predicting refractoriness have been identified in some
tion.17,18 With the same objective, the addition of mol- studies. Single nucleotide polymorphisms (SNPs)
ecularly-targeted therapy to induction CT is being affecting DNA-repair genes, such as xeroderma pig-
investigated in several trials.19 mentosa A are associated with decreased sensitivity to
The causes for not achieving a CR (induction failure) induction chemotherapy.34 Overexpression or
are toxic death and refractoriness of leukemic cells. As rearrangements involving the EV1 gene also favour
mentioned, advanced age has an adverse impact on refractoriness.35 In contrast, recently communicated
induction mortality and overall survival when consid- data indicate that this group of AML could be sensitive
ered as a continuous variable and at different age cut- to demetilating agents.36 Slow clearance of leukemic
offs (Figure 1). During induction, patients older than 70 cells from peripheral blood and/or marrow by mor-
years have a probability of dying in the order of 30- phologic, or flow cytometry examination is also pre-
50%.5,6,20 Recently published scoring systems that dictive of decreased sensitivity to CT.17,18,37,38,39,40 In con-
include preceding diseases and concurrent morbidities trast, few patients with favourable cytogenetic and
have been developed to predict mortality after molecular features experience resistance to induction
hematopoietic transplantation.21 These and other pre- treatment. Core binding factors (CBF) AML in non-eld-
dictive scores are starting to be applied to AML erly adults are associated with more than 85% proba-
patients receiving induction chemotherapy, and may bility of CR achievement, with less than 5% of cases

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 31 |
14th Congress of the European Hematology Association

Figure 2. Results of induc-


tion chemotherapy in the
different genetic categories
of acute myeloid leukemia.
Experience of the Spanish
CETLAM group.
adv: adverse; mk: monoso-
mal karyoptype; mut: muta-
tion; pos: positive; wt: wild
type.

being refractory (Figure 2).3,4,5 The results in this Postremission chemotherapy


favourable category may improve even further if clas- After achieving CR, the standard approach is to
sic chemotherapy regimens are intensified with novel administer consolidation CT. In practice, the excep-
agents. The results of a recent report from the MRC tions are patients who required three or more courses
combining chemotherapy with GO are encouraging, of induction CT to achieve CR with an HLA-compati-
although confirmation is needed.16 Other AML with ble donor. In this circumstance, consolidation treat-
high sensitivity to initial chemotherapy are the recent- ment is frequently omitted in order to proceed to
ly identified good molecular abnormalitites, that is, hematopoietic transplantation as soon as possible. The
NPM1 and CEBPA mutations without associated aim of this strategy is to avoid the early recurrence of
leukemia, which is particularly frequent in AML
adverse genetic features (Figure 2).40
patients with adverse cytogenetics, and to take advan-
Since so many factors must be considered, the ques-
tage of the conditioning regimen and graft versus
tion is whether it is necessary to wait for the results of leukemia effects. All other patients are candidates for
a full biologic evaluation of the disease before starting consolidation CT, which, in patients of up to 60 years,
CT. The available data indicate that delaying treat- usually includes high-dose cytarabine. In contrast, in
ment adversely affects patients up to 60 years old but patients above this age, it is safer to administer con-
not above that age.41 Therefore, young adults with ventional or intermediate-doses of this drug to avoid
AML should be treated shortly after diagnosis. Since an excess of neurologic and gastrointestinal toxicities.44
some trials combining chemotherapy and targeted The number of consolidation courses to be adminis-
treatment require molecular typing before inclusion, tered has not been clearly defined. In the case of non-
substantial efforts must be made here to obtain short- CBF leukemia, most patients receive one to three
term results of laboratory studies. In patients between courses. Patients with CBF leukemia have a decreased
60 and 80 years of age, it seems reasonable to delay the relapse rate if they receive three or four courses of
decision until the cytogenetics of AML is known. The high-dose cytarabine, rather than one or two.45,46 These
therapeutic approach in this age group should also take repeated consolidation courses lead to 70-80% of CBF
into consideration whether the patient is a potential leukemia patients becoming long-term survivors in
candidate for allogeneic hematopoietic transplantation remission. There are, however, certain patients with
after reduced intensity conditioning. Therefore, CBF leukemia who relapse, despite high-dose cytara-
bine; those with high WBC counts at diagnosis and/or
despite the presence of AML with poor-risk features, it
c-KIT mutations.47,48 Tyrosine kinase inhibitors, such as
makes sense to administer intensive CT if the patient
dasatinib in c-KIT mutated cases and hematopoietic
may benefit from a subsequent transplantation while transplantation are being investigated to improve the
in complete or partial remission. It is important to note results in these cases. As in CBF leukemias, it has been
that if a significant reduction in blasts is not achieved suggested that AML from the intermediate cytogenet-
after a first course of CT, recent experiences support ics group with a favourable molecular profile may also
proceeding directly to sequential regimens, combining benefit from repeated high-dose cytarabine consolida-
intensive rescue CT with reduced intensity condition- tion cycles.40 This group includes AML patients with
ing transplants.42,43 With such an approach, 40% of mutated nucleophosmin and without the internal tan-
refractory or relapsed patients may become long-term dem duplication of the FLT3 gene (NPM1+/FLT3–), and
survivors in remission. those with CEBPA mutations.49-52 In these cases, the

| 32 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

possible favourable impact from high-dose cytarabine


should be further investigated. Also in NPM1 positive
AML, recent studies indicate that the addition of
ATRA to CT should be explored, since this agent sig-
nificantly decreased the relapse risk in one study.53
In contrast to these favourable molecular markers,
others confer an adverse outcome to intermediate-risk
cytogenetics AML. These include the internal tandem
duplication of the FLT3 gene (FLT3/ITD),54-58 rearrange-
ments and the partial tandem duplication of the MLL
gene,59-62 mutations of the Wilms’ tumor 1 (WT1)63 and
over-expression of the BAALC64-66 and/or ERG
genes.67,68
Different combinations of postremission CT have
been administered in these unfavourable cases belong-
ing to the intermediate-risk cytogenetics category, and
in AML with adverse karyotype. So far, none of them
has shown that high-doses of cytarabine improve the
results compared to intermediate or conventional
doses. Figure 3. Relapse-free survival in the donor (black line)
To further exploit the antileukemic effect of CT, and no donor (red line) groups in AML patients with normal
some groups administered maintenance treatment. cytogenetics and mutations of NPM1 without FLT3-ITD.
This approach was effective in occasional studies;69 Adapted from Schlenk R et al. N Engl J Med 2008;358:
1909-1918; with permission.
therefore, novel maintenance strategies deserve inves-
tigation. In this sense, the CALGB recently reported
the use low-dose subcutaneous interleukin-2.70
Unfortunately, a protective effect on the development
of leukemia relapse was not observed. Other alterna-
tives, such as the administration of maintenance,
including GO or demethylating agents, are currently
being explored.71

Autologous transplantation
The role of autologous hematopoietic cell transplan-
tation (auto-HCT) in the first CR of AML remains con-
troversial.72 In many centers in the United States, and
in some European countries, this treatment has been
abandoned for this disease, whereas for other institu-
tions and cooperative groups, it is the treatment of
choice for patients without an HLA compatible donor.
Large meta-analyses are ongoing to define whether
autologous transplantation offers an advantage com-
pared with further consolidation CT or abstention. Figure 4. Leukemia-free survival in the donor (green line)
Reported series comparing this treatment versus and no donor (blue line) groups in AML patients from the
chemotherapy have the caveat of the relatively high intermediate and poor risk cytogenetics categories and
transplant-related mortality reported (10%) that does without favorable molecular markers. Experience of the
Spanish CETLAM group.
not reflect the improvement observed nowadays.73
The relapse rate after autografting for adverse cytoge-
netics AML is very high, and for this reason, this treat-
ment is being abandoned in this group of patients. In
contrast, the role of autologous transplantation, if any, availability or not of an HLA-identical sibling are need-
in favourable categories, such as CBF leukemia, ed to draw conclusions. Some studies have recently
NPM1+/FLT3– and CEBPA+ AML, particularly if high been published and others are currently ongoing.29,75,76
WBC counts or other adverse indicators are present, Knowing the strong impact of age and cytogenetics on
deserves investigation within clinical trials. the prognosis of AML, comparisons between treat-
ments have to take these factors into account.74,77
Allogeneic transplantation from HLA-identical siblings Ideally, molecular information with impact on progno-
It is clear that allogeneic hematopoietic cell trans- sis should also be available, but this is the case in only
plantation (allo-HCT) decreases the relapse rate com- a few reports.78,79 In summary, information from
pared to CT or auto-HCT.74 In many studies, this does prospective trials and published meta-analyses indi-
not translate into significantly improved leukemia-free cate that patients with CBF leukemia and
survival and/or overall survival as there is a 15-30% NPM1+/FLT3– AML have equal survival in donor and
transplant related mortality. As in the case of auto- no donor groups (Figure 3).29,75-77,79 In contrast, in most
HCT versus CT, large meta-analyses based on the studies, patients in the intermediate cytogenetics

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 33 |
14th Congress of the European Hematology Association

group without a favourable molecular profile, and essary. Improved methods for quantitative evaluation
those in the adverse karyotype category benefit from of MRD after treatment in AML, and their role in spe-
allogeneic transplantation (Figure 4). Some experiences cific molecular subgroups are being explored.94,95 Based
state that the advantage is limited to young patients, on the results, additional targeted treatment or intensi-
up to the age of 35 years, since in older patients, trans- fication with hematopoietic transplantation may be
plant related mortality counterbalances the decrease in decided. Finally, a large variety of new agents with
relapse rate observed after allograft.29 In patients aged potential activity in AML is under investigation in clin-
above this cut-off, an individualized decision concern- ical trials.96,97 Many drugs to treat AML are in at differ-
ing the administration of reduced intensity condition- ent stages of development in pharmaceutical compa-
ing may be an option when comorbidity scores are nies. All these mentioned advances indicate that
high. Reduced intensity conditioning has allowed research in biology, prognosis and risk-adapted treat-
extending the benefit of the graft versus leukemia ment in AML will continue to be a challenging and
effect to patients up to the age of 75.80-82 To define the evolving area in the coming years.
impact of this treatment on survival of elderly adults
with AML in first CR, a prospective randomized com- This work was supported in part by grants from the
parison versus no transplantation is ongoing in Europe. Instituto de Salud Carlos III, Spain (07/EC90065 and
06/0020/0101RD).
Alternative donor and sources for hematopoietic trans-
plantation
Since the majority of AML patients who are poten- References
tial candidates for allo-HCT lack an HLA-identical sib-
ling, the use of alternative donors and stem cell sources 1. Löwenberg B. Acute myeloid leukemia: the challenge of
capturing disease variety. Hematology Am Soc Hematol
has increased greatly over the last two decades.83,84 Educ Program 2008;2008:1-11.
Regarding unrelated transplants from adult donors 2. Swerdlow SH, Campo E, Harris NL, et al., editors WHO
(URD), results have improved due to the better selec- classification of tumours of haematopoietic and lymphoid
tissues. Lyon: IARC Press; 2008. p. 109-38.
tion of donor-recipient pairs based on high-resolution 3. Ravandi F, Burnett AK, Agura ED, Kantarjian HM. Progress
molecular HLA-typing.84 URD transplants were initial- in the treatment of acute myeloid leukemia. Cancer
ly restricted to patients with an extremely high-risk of 2007;110: 1900-10.
4. Estey E, Döhner H. Acute myeloid leukemia. Lancet 2006;
relapse due to increased transplant related mortality 368:1894-07.
(TRM), as compared to those from HLA-identical sib- 5. Estey E. Acute myeloid leukemia and myelodysplastic syn-
lings. Currently, the indications of the two procedures dromes in older patients. J Clin Oncol 2007;25:1908-15.
are similar since the risk of death due to complications 6. Appelbaum FR, Gundacker H, Head DR, Slovak ML,
Willman CL, Godwin JE et al. Age and acute myeloid leu-
is the same in experienced centres. Transplantation of kemia. Blood 2006;107:3481-85.
hematopoietic progenitors from unrelated cord blood 7. Issa JP, Garcia-Manero G, Giles FJ, Mannari R, Thomas D,
is also an option for adult patients with AML, although Faderl S et al. Phase 1 study of low-dose prolonged exposure
schedules of the hypomethylating agent 5-aza-2’-deoxycyti-
cell dose remains a limitation for this procedure.85,86 dine (decitabine) in hematopoietic malignancies. Blood
Due to the risk of graft failure or delayed engraftment, 2004;103:1635-40.
this type of transplant is usually restricted to patients 8. Garcia-Manero G, Kantarjian HM, Sanchez-Gonzalez B,
Yang H, Rosner G, Verstovsek S et al. Phase 1/2 study of the
with a high risk of leukemia relapse and who need an combination of 5-aza-2’-deoxycytidine with valproic acid in
allogeneic procedure on a short-term basis. In this cir- patients with leukemia. Blood 2006;108:3271-79.
cumstance and within clinical trials, haploidentical 9. Blum W, Klisovic RB, Hackanson B, Liu Z, Liu S, Devine H
family transplantation is another possibility.87 et al. Phase I study of decitabine alone or in combination
with valproic acid in acute myeloid leukemia. J Clin Oncol
Different approaches to improve immune recovery 2007;25:3884-91.
after the procedure, based on ex vivo or in vivo depletion 10. Musto P, Ferrara F. Clofarabine: In search of combinations
of alloreactive cells, are being explored. for the treatment of patients with high-risk acute myeloid
leukemia. Cancer 2008;113:1995-98.
11. Amadori S, Suciu S, Stasi R, Willemze R, Mandelli F, Selles-
New approaches for tailoring treatment lag D et al. Gemtuzumab ozogamicin (Mylotarg) as single-
Gene expression profiling (GEP) is a recently intro- agent treatment for frail patients 61 years of age and older
with acute myeloid leukemia: final results of AML-15B, a
duced method to identify molecular pathways phase 2 study of the European Organisation for Research
involved in AML and to select new therapeutic tar- and Treatment of Cancer and Gruppo Italiano Malattie
gets.88-91 GEP studies have shown to be useful by recog- Ematologiche dell’Adulto Leukemia Groups. Leukemia
2005;19:1768-73
nizing known AML subtypes, detecting new varieties, 12. Rajvanshi P, Shulman HM, Sievers EL, McDonald GB.
and separating otherwise not identifiable prognostic Hepatic sinusoidal obstruction after gemtuzumab ozogam-
groups. This method may be useful in the near future icin (Mylotarg) therapy. Blood 2002;99:2310-14.
to adjust treatment based on patterns of expression 13. Burnett AK, Milligan D, Prentice AG, Goldstone AH,
McMullin MF, Hills RK, Wheatley K et al. A comparison of
and genes involved. Methylation status of a large num- low dose cytarabine and hydroxyurea with or without all-
ber of genes may also be studied using this technique.92 trans retinoic acid for acute myeloid leukemia and high-risk
This may identify those patients who are more suscep- myelodysplastic syndrome in patients not considered fit for
intensive treatment. Cancer 2007;109:1114-24.
tible to responding to demethylating agents. The 14. Lowenberg B, van Putten W, Theobald M, Gmür J, Verdonck
recent discovery of the impact of certain micro RNAs L, Sonneveld P et al. Effect of priming with granulocyte
on the development and prognosis of AML justifies colony-stimulating factor on the outcome of chemotherapy
for acute myeloid leukemia. N Engl J Med 2003;349:743-52.
further investigation on this subject.93 For a better dis- 15. Amadori S, Suciu S, Jehn U, Stasi R, Thomas X, Marie JP et
crimination of susceptibility to treatment, additional al. Use of glycosylated recombinant human G-CSF (leno-
studies on individual genetic polymorphisms are nec- grastim) during and/or after induction chemotherapy in
patients 61 years of age and older with acute myeloid

| 34 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

leukemia: final results of AML-13, a randomized phase-3 32. Bloomfield CD, Lawrence D, Byrd JC, Carroll A, Pettenati
study. Blood 2005;106:27-34 MJ, Tantravahi R et al. Frequency of prolonged remission
16. Burnett AK, Kell WJ, Goldstone AH, et al. The addition of duration after high-dose cytarabine intensification in acute
gemtuzumab ozogamicin to induction chemotherapy for myeloid leukemia varies by cytogenetic subtype. Cancer
AML improves disease free survival without extra toxicity: Res 1998;58: 4173-4179.
preliminary analysis of 1115 patients in the MRC AML15 33. Breems DA, Van Putten WL, De Greef GE, Van Zelderen-
trial. Blood 2006;108:8A-8A. Bhola SL, Gerssen-Schoorl KB, Mellink CH et al. Mono-
17. Kern W, Voskova D, Schoch C, Schnittger S, Hiddemann W, somal karyotype in acute myeloid leukemia: a better indica-
Haferlach T et al. Prognostic impact of early response to tor of poor prognosis than a complex karyotype. J Clin
induction therapy as assessed by multiparameter flow Oncol 2008;26:4791-97.
cytometry in acute myeloid leukemia. Haematologica 2004; 34. Monzo M, Brunet S, Urbano-Ispizua A, Navarro A, Perea G,
89:528-40. Esteve J et al. Genomic polymorphisms provide prognostic
18. Lacombe F, Arnoulet C, Maynadié M, Lippert E, Luquet I, information in intermediate-risk acute myeloblastic
Pigneux A et al. Early clearance of peripheral blasts meas- leukemia. Blood 2006:4871-79.
ured by flow cytometry during the first week of AML induc- 35. Lugthart S, Drunen EV, Norden YV, van Hoven A, Erpelinck
tion therapy as new independent prognostic factor : A GOE- CA, Valk PJ et al. High EVI1 levels predict adverse outcome
LAMS study. Leukemia. 2008; Nov 6. [Epub ahead of print] in acute myeloid leukemia: prevalence of EVI1 overexpres-
19. Amadori S, Stasi R. New agents for the treatment of acute sion and chromosome 3q26 abnormalities underestimated.
myeloid leukemia. Hematology Education: the education Blood 2008.
program for the Annual Congress of the European 36. Lugthart S, Figueroa ME, Valk PJM, et al. Two different EVI1
Hematology Association 2008;2:33-8. expressing poor-risk AML subgroups with distinct epigenet-
20. Erba HP. Prognostic factors in elderly patients with AML and ic signatures uncovered by genome wide DNA methylation
the implications for treatment. Hematology Am Soc profiling. Blood. (ASH Annual Meeting Abstracts), Nov
Hematol Educ Program 2007;2007:420-27. 2008;112:757.
21. Sorror ML, Sandmaier BM, Storer BE, Maris MB, Baron F, 37. Kern W, Haferlach T, Schoch C, Loffler H, Gassmann W,
Maloney DG et al. Comorbidity and disease status-based Heinecke A et al. Early blast clearance by remission induc-
risk stratification of outcomes among patients with acute tion therapy is a major independent prognostic factor for
myeloid leukemia or myelodysplasia receiving allogeneic both achievement of complete remission and long-term out-
hematopoietic cell transplantation. J Clin Oncol 2007;25: come in acute myeloid leukemia: data from the German
4246-54. AML Cooperative Group (AMLCG) 1992 Trial. Blood 2003;
22. Giles FJ, Borthakur G, Ravandi F, Faderl S, Verstovsek S, 101:64-70.
Thomas D et al. The haematopoietic cell transplantation 38. San Miguel JF, Vidriales MB, Lopez-Berges C, Díaz-
comorbidity score is predictive of early death and survival in Mediavilla J, Gutiérrez N, Cañizo C et al. Early immunophe-
patients over 60 years of age receiving induction therapy for notypical evaluation of minimal residual disease in acute
acute myeloid leukaemia. Br J Haematol 2007;136:624-27. myeloid leukemia identifies different patient risk groups and
23. Etienne A, Esterni B, Charbonnier A, Mozziconacci MJ, may contribute to postinduction treatment stratification.
Arnoulet C, Coso D et al. Comorbidity is an independent Blood 2001;98:1746-51.
predictor of complete remission in elderly patients receiving 39. Buccisano F, Maurillo L, Gattei V, Del Poeta G, Del Principe
induction chemotherapy for acute myeloid leukemia. MI, Cox MC et al. The kinetics of reduction of minimal
Cancer 2007;109:1376-83. residual disease impacts on duration of response and sur-
24. Montesinos P, Lorenzo I, Martin G, Sanz J, Pérez-Sirvent vival of patients with acute myeloid leukemia. Leukemia
ML, Martínez D et al. Tumor lysis syndrome in patients 2006;20:1783-89.
with acute myeloid leukemia: identification of risk factors 40. Estey EH. Treatment of acute myeloid leukemia. Haemato-
and development of a predictive model. Haematologica logica 2009;94:10-16.
2008;93:67-74. 41. Estey E, Wang X-M, Thall P, et al. Plausibility of delaying
25. Gupta V, Chun K, Yi QL, Minden M, Schuh A, Wells R, induction therapy in untreated AML. Blood 2004;104:879a
Brandwein J. Disease biology rather than age is the most (abstr).
important determinant of survival of patients > or =60 years 42. Schmid C, Schleuning M, Ledderose G, Tischer J, Kolb HJ.
with acute myeloid leukemia treated with uniform intensive Sequential regimen of chemotherapy, reduced-intensity con-
therapy. Cancer 2005;103:2082-90. ditioning for allogeneic stem-cell transplantation, and pro-
26. Grimwade D, Walker H, Oliver F, Wheatley K, Harrison C, phylactic donor lymphocyte transfusion in high-risk acute
Harrison G et al. The importance of diagnostic cytogenetics myeloid leukemia and myelodysplastic syndrome. J Clin
on outcome in AML: analysis of 1,612 patients entered into Oncol 2005;23:5675-5687.
the MRC AML 10 trial. The Medical Research Council Adult 43. Schmid C, Schleuning M, Schwerdtfeger R, Hertenstein B,
and Children's Leukaemia Working Parties. Blood 1998;92: Mischak-Weissinger E, Bunjes D et al. Long term survival in
2322-33. refractory acute myeloid leukemia after sequential treat-
27. Grimwade D, Walker H, Harrison G, Oliver F, Chatters S, ment with chemotherapy and reduced intensity condition-
Harrison CJ et al. The predictive value of hierarchical cyto- ing for allogeneic stem cell transplantation. Blood 2006;
genetic classification in older adults with acute myeloid 108:1092-99.
leukemia (AML): analysis of 1065 patients entered into the 44. Morra E, Barosi G, Bosi A, et al. Clinical magament of pri-
United Kingdom Medical Research Council AML11 trial. mary non-acute promyelocytic leukemia acute myeloid
Blood 2001;98:1312-20. leukemia: practice guidelines by the Italian Society of
28. Suciu S, Mandelli F, de Witte T, Zittoun R, Gallo E, Labar B Hematology, the Italian Society of Experimental
et al. Allogeneic compared with autologous stem cell trans- Hematology and the Italian Group for Bone Marrow
plantation in the treatment of patients younger than 46 Transplantation. Haematologica 2009;94:102-12.
years with acute myeloid leukemia (AML) in first complete 45. Byrd JC, Dodge RK, Carroll A, Ferrara F, Locatelli F,
remission (CR1): an intention-to-treat analysis of the Marchetti M et al. Patients with t(8;21)(q22;q22) and acute
EORTC/GIMEMAAML-10 trial. Blood 2003;102:1232-40. myeloid leukemia have superior failure-free and overall sur-
29. Cornelissen JJ, van Putten WL, Verdonck LF, Theobald M, vival when repetitive cycles of high-dose cytarabine are
Jacky E, Daenen SM et al. Results of a HOVON/SAKK donor administered. J Clin Oncol 1999;17:3767-75.
versus no-donor analysis of myeloablative HLA-identical 46. Byrd JC, Ruppert AS, Mrozek K, Carroll AJ, Edwards CG,
sibling stem cell transplantation in first remission acute Arthur DC et al. Repetitive cycles of high-dose cytarabine
myeloid leukemia in young and middle-aged adults: benefits benefit patients with acute myeloid leukemia and
for whom? Blood 2007;109:3658-66. inv(16)(p13q22) or t(16;16)(p13;q22): results from CALGB
30. Slovak ML, Kopecky KJ, Cassileth PA, Harrington DH, Theil 8461. J Clin Oncol 2004;22:1087-94.
KS, Mohamed A et al. Karyotypic analysis predicts outcome 47. Nguyen S, Leblanc T, Fenaux P, Witz F, Blaise D, Pigneux A
of preremission and postremission therapy in adult acute et al. A white blood cell index as the main prognostic factor
myeloid leukemia: a Southwest Oncology Group/Eastern in t(8;21) acute myeloid leukemia (AML): a survey of 161
Cooperative Oncology Group Study. Blood 2000;96:4075- cases from the French AML Intergroup. Blood 2002;99:3517-
4083. 23.
31. Mayer RJ, Davis RB, Schiffer CA, Berg DT, Powell BL, 48. Paschka P, Marcucci G, Ruppert AS, Mrózek K, Chen H,
Schulman P et al. Intensive postremission chemotherapy in Kittles RA et al. Adverse prognostic significance of KIT
adults with acute myeloid leukemia. Cancer and Leukemia mutations in adult acute myeloid leukemia with inv(16) and
Group B. N Engl J Med 1994;331:896-903. t(8;21): a Cancer and Leukemia Group B Study. J Clin Oncol

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 35 |
14th Congress of the European Hematology Association

2006;24:3904-11. Blood 2003;102:1613-8.


49. Falini B, Mecucci C, Tiacci E, Alcalay M, Rosati R, 65. Baldus CD, Thiede C, Soucek S, Bloomfield CD, Thiel E,
Pasqualucci L et al. Cytoplasmic nucleophosmin in acute Ehninger G et al. BAALC expression and FLT3 internal tan-
myelogenous leukemia with a normal karyotype. N Engl J dem duplication mutations in acute myeloid leukemia
Med 2005;352:254-66. patients with normal cytogenetics: prognostic implications.
50. Dohner K, Schlenk RF, Habdank M, Scholl C, Rücker FG, J Clin Oncol 2006;24:790-97.
Corbacioglu A et al. Mutant nucleophosmin (NPM1) pre- 66. Baldus CD, Mrozek K, Marcucci G, Bloomfield CD. Clinical
dicts favorable prognosis in younger adults with acute outcome of de novo acute myeloid leukaemia patients with
myeloid leukemia and normal cytogenetics: interaction with normal cytogenetics is affected by molecular genetic alter-
other gene mutations. Blood 2005;106:3740-46. ations: a concise review. Br J Haematol 2007;137:387-400.
51. Preudhomme C, Sagot C, Boissel N, Cayuela JM, Tigaud I, 67. Marcucci G, Baldus CD, Ruppert AS, Radmacher MD,
de Botton S et al. Favorable prognostic significance of Mrózek K, Whitman SP et al. Overexpression of the ETS-
CEBPA mutations in patients with de novo acute myeloid related gene, ERG, predicts a worse outcome in acute
leukemia: a study from the Acute Leukemia French myeloid leukemia with normal karyotype: a Cancer and
Association (ALFA). Blood 2002;100:2717-23. Leukemia Group B study. J Clin Oncol 2005;23:9234-42.
52. Wouters BJ, Lowenberg B, Erpelinck-Verschueren CAJ, et al. 68. Marcucci G, Maharry K, Whitman SP, Vukosavljevic T,
Double CEBPA mutations, but not single CEBPA mutations, Paschka P, Langer C et al. High expression levels of the ETS-
define a subgroup of acute myeloid leukemia with a distinc- related gene, ERG, predict adverse outcome and improve
tive gene expression profile that is uniquely associated with molecular risk-based classification of cytogenetically normal
a favorable outcome. Blood 2009 Jan 26. [Epub ahead of acute myeloid leukemia: a Cancer and Leukemia Group B
print] Study. J Clin Oncol 2007;25:3337-43.
53. Schlenk RF, Döhner K, Kneba M, Götze K, Hartmann F, Del 69. Buchner T, Krug U, Berdel WE, Heinecke A, Sauerland MC,
Valle F et al. Gene mutations and response to treatment with Wörmann B, Hiddemann W. Maintenance for acute myeloid
all-trans retinoic acid in elderly patients with acute myeloid leukemia revisited. Curr Treat Options Oncol 2007;8:296-
leukemia. Results from the AMLSG trial AML HD98B. 04.
Haematologica 2009;94:54-60. 70. Baer MR, George SL, Caligiuri MA, Sanford BL, Bothun SM,
54. Yokota S, Kiyoi H, Nakao M, Iwai T, Misawa S, Okuda T et Mrózek K et al. Low-dose interleukin-2 immunotherapy
al. Internal tandem duplication of the FLT3 gene is preferen- does not improve outcome of patients age 60 years and
tially seen in acute myeloid leukemia and myelodysplastic older with acute myeloid leukemia in first complete remis-
syndrome among various hematological malignancies. A sion: Cancer and Leukemia Group B Study 9720. J Clin
study on a large series of patients and cell lines. Leukemia Oncol 2008;26:4934-9.
1997;11:1605-09. 71. Burnett AK, Knapper S. Targeting treatment in AML.
55. Kottaridis PD, Gale RE, Frew ME, Harrison G, Langabeer SE, Hematology Am Soc Hematol Educ Program 2007;2007:429-
Belton AA et al. The presence of a FLT3 internal tandem 34.
duplication in patients with acute myeloid leukemia (AML) 72. Breems DA, Lowenberg B. Acute myeloid leukemia and the
adds important prognostic information to cytogenetic risk position of autologous stem cell transplantation. Semin
group and response to the first cycle of chemotherapy: Hematol 2007;44:259-66.
analysis of 854 patients from the United Kingdom Medical 73. Burnett AK, Goldstone AH, Stevens RM, Hann IM, Rees JK,
Research Council AML 10 and 12 trials. Blood. 2001;98: Gray RG, Wheatley K et al. Randomized comparison of
1752-59. addition of autologous bone-marrow transplantation to
56. Thiede C, Steudel C, Mohr B, Schaich M, Schäkel U, intensive chemotherapy for acute myeloid leukaemia in first
Platzbecker U et al. Analysis of FLT3-activating mutations in remission: results of MRC AML 10 trial. UK Medical
979 patients with acute myelogenous leukemia: association Research Council Adult and Children’s Leukaemia Working
with FAB subtypes and identification of subgroups with Parties. Lancet 1998;351: 700-08.
poor prognosis. Blood 2002;99:4326-35. 74. Lodewyck T, Cornelissen JJ. Allogeneic stem cell transplan-
57. Schnittger S, Schoch C, Dugas M, Kern W, Staib P, Wuchter tation in acute myeloid leukemia: a risk-adapted approach.
C et al. Analysis of FLT3 length mutations in 1003 patients Blood Rev. 2008;22:293-02.
with acute myeloid leukemia: correlation to cytogenetics, 75. Yanada M, Matsuo K, Emi N, Naoe T. Efficacy of allogeneic
FAB subtype, and prognosis in the AMLCG study and use- hematopoietic stem cell transplantation depends on cytoge-
fulness as a marker for the detection of minimal residual dis- netic risk for acute myeloid leukemia in first disease remis-
ease. Blood 2002;100:59-66. sion: a metaanalysis. Cancer 2005;103:1652-58.
58. Frohling S, Schlenk RF, Breitruck J, Benner A, Kreitmeier S, 76. Oliansky DM, Appelbaum F, Cassileth PA, Keating A, Kerr J,
Tobis K et al. Prognostic significance of activating FLT3 Nieto Y et al. The role of cytotoxic therapy with hematopoi-
mutations in younger adults (16 to 60 years) with acute etic stem cell transplantation in the therapy of acute myel-
myeloid leukemia and normal cytogenetics: a study of the ogenous leukemia in adults: an evidence-based review. Biol
AML Study Group Ulm. Blood 2002;100:4372-80. Blood Marrow Transplant. 2008;14:137-80.
59. Caligiuri MA, Strout MP, Lawrence D, Arthur DC, Baer MR, 77. Burnett AK, Wheatley K, Goldstone AH, Stevens RF, Hann
Yu F et al. Rearrangement of ALL1 (MLL) in acute myeloid IM, Rees JH et al. The value of allogeneic bone marrow
leukemia with normal cytogenetics. Cancer Res 1998;58:55- transplant in patients with acute myeloid leukaemia at dif-
59. fering risk of relapse: results of the UK MRC AML 10 trial.
60. Schnittger S, Kinkelin U, Schoch C, Heinecke A, Haase D, Br J Haematol 2002;118:385-400.
Haferlach T et al. Screening for MLL tandem duplication in 78. Gale RE, Hills R, Kottaridis PD, Srirangan S, Wheatley K,
387 unselected patients with AML identify a prognostically Burnett AK et al. No evidence that FLT3 status should be
unfavorable subset of AML. Leukemia 2000;14:796-04. considered as an indicator for transplantation in acute
61. Dohner K, Tobis K, Ulrich R, Fröhling S, Benner A, Schlenk myeloid leukemia (AML): an analysis of 1135 patients,
RF, Döhner H. Prognostic significance of partial tandem excluding acute promyelocytic leukemia, from the UK MRC
duplications of the MLL gene in adult patients 16 to 60 years AML10 and 12 trials. Blood. 2005;106:3658-65.
old with acute myeloid leukemia and normal cytogenetics: a 79. Schlenk RF, Döhner K, Krauter J, Fröhling S, Corbacioglu A,
study of the Acute Myeloid Leukemia Study Group Ulm. J Bullinger L et al. Mutations and treatment outcome in cyto-
Clin Oncol 2002;20:3254-61 genetically normal acute myeloid leukemia. N Engl J Med
62. Munoz L, Nomdedeu JF, Villamor N, Guardia R, Colomer D, 2008;358:1909-18.
Ribera JM et al. Acute myeloid leukemia with MLL rear- 80. Hegenbart U, Niederwieser D, Sandmaier BM, Maris MB,
rangements: clinicobiological features, prognostic impact Shizuru JA, Greinix H et al. Treatment for acute myeloge-
and value of flow cytometry in the detection of residual nous leukemia by low-dose, total-body, irradiation-based
leukemic cells. Leukemia 2003;17:76-82. conditioning and hematopoietic cell transplantation from
63. Paschka P, Marcucci G, Ruppert AS, Whitman SP, Mrózek K, related and unrelated donors. J Clin Oncol 2006;24:444-53.
Maharry K et al. Wilms’ tumor 1 gene mutations independ- 81. Martino R, Caballero MD, Simon JA, Canals C, Solano C,
ently predict poor outcome in adults with cytogenetically Urbano-Ispízua A et al. Evidence for a graft-versus-leukemia
normal acute myeloid leukemia: a cancer and leukemia effect after allogeneic peripheral blood stem cell transplanta-
group B study. J Clin Oncol 2008;26:4595-602. tion with reduced-intensity conditioning in acute myeloge-
64. Baldus CD, Tanner SM, Ruppert AS, Whitman SP, Archer KJ, nous leukemia and myelodysplastic syndromes. Blood
Marcucci G et al. BAALC expression predicts clinical out- 2002;100:2243-5.
come of de novo acute myeloid leukemia patients with nor- 82. Valcarcel D, Martino R, Caballero D, Martin J, Ferra C, Nieto
mal cytogenetics: a Cancer and Leukemia Group B Study. JB et al. Sustained remissions of high-risk acute myeloid

| 36 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

leukemia and myelodysplastic syndrome after reduced- shirani R et al. Use of gene-expression profiling to identify
intensity conditioning allogeneic hematopoietic transplanta- prognostic subclasses in adult acute myeloid leukemia. N
tion: chronic graft-versus-host disease is the strongest factor Engl J Med 2004;350:1605-16.
improving survival. J Clin Oncol 2008;26:577-84. 90. Valk PJ, Verhaak RG, Beijen MA, Erpelinck CA, Barjesteh
83. Appelbaum FR. Hematopoietic cell transplantation from van Waalwijk van Doorn-Khosrovani S et al. Prognostically
unrelated donors for treatment of patients with acute useful gene-expression profiles in acute myeloid leukemia.
myeloid leukemia in first complete remission. Best Pract Res N Engl J Med 2004;350:1617-28.
Clin Haematol 2007;20:67-75. 91. Haferlach T, Kohlmann A, Schnittger S, Dugas M, Hidde-
84. Sierra J, Martino R, Sanchez B, Piñana JL, Valcárcel D, Brunet mann W, Kern W et al. Global approach to the diagnosis of
S et al. Hematopoietic transplantation from adult unrelated leukemia using gene expression profiling. Blood 2005;106:
donors as treatment for acute myeloid leukemia. Bone 1189-98.
Marrow Transplant 2008;41:425-37. 92. Gebhard C, Schwarzfischer L, Pham TH, Schilling E, Klug
85. Rocha V, Labopin M, Sanz G, Arcese W, Schwerdtfeger R, M, Andreesen R, Rehli M. Genome-wide profiling of CpG
Bosi A et al. Transplants of umbilical-cord blood or bone methylation identifies novel targets of aberrant hyperme-
marrow from unrelated donors in adults with acute thylation in myeloid leukemia. Cancer Res 2006;66:6118-28.
leukemia. N Engl J Med 2004;351:2276-85. 93. Marcucci G, Radmacher MD, Maharry K, Mrózek K,
86. Laughlin MJ, Eapen M, Rubinstein P, Wagner JE, Zhang MJ, Ruppert AS, Paschka P et al. MicroRNA expression in cyto-
Champlin RE et al. Outcomes after transplantation of cord genetically normal acute myeloid leukemia. N Engl J Med
blood or bone marrow from unrelated donors in adults with 2008;358:1919-28.
leukemia. N Engl J Med 2004;351:2265-75. 94. Kern MRD. Kern W, Haferlach C, Haferlach T, Schnittger S.
87. Aversa F, Terenzi A, Tabilio A, Falzetti F, Carotti A, Ballanti Monitoring of minimal residual disease in acute myeloid
S et al. Full haplotype-mismatched hematopoietic stem-cell leukemia. Cancer 2008;112:4-16.
transplantation: a phase II study in patients with acute 95. Hess CJ, Feller N, Denker F, Kelder A, Merle PA, Heinrich
leukemia at high risk of relapse. J Clin Oncol 2005;23:3447- MC et al. Correlation of minimal residual disease cell fre-
54. quency with molecular genotype in patients with acute
88. Schoch C, Kohlmann A, Schnittger S, Brors B, Dugas M, myeloid leukemia. Haematologica 2009;94:46-53.
Mergenthaler S et al. Acute myeloid leukemias with recipro- 96. Grant S. Is the focus moving toward a combination of tar-
cal rearrangements can be distinguished by specific gene geted drugs? Best Pract Res Clin Haematol 2008;21:629-37.
expression profiles. Proc Natl Acad Sci USA 2002;99:10008- 97. Haferlach T. Molecular genetic pathways as therapeutic tar-
13. gets in acute myeloid leukemia. Hematology Am Soc
89. Bullinger L, Dohner K, Bair E, Fröhling S, Schlenk RF, Tib- Hematol Educ Program 2008;2008:400-11.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 37 |
Bleeding disorders

von Willebrand factor: its role in hemostasis and


thrombosis

S.F. De Meyer A B S T R A C T
K. Vanhoorelbeke
A key player in thrombosis and hemostasis is von Willebrand factor (VWF), a multimeric glycopro-
tein that is essential for the recruitment of circulating platelets to damaged vessels and for protect-
Laboratory for Thrombosis Research,
IRC, KULeuven Campus Kortrijk, ing the anti-hemophilic coagulation factor VIII from rapid degradation. As a result, quantitative or
Belgium qualitative defects in VWF lead to the most common inherited bleeding disorder – von Willebrand dis-
ease. On the other hand, the presence of over-reactive VWF multimers may result in severe thrombot-
ic complications. While our knowledge on this intriguingly large hemostatic factor is increasing, its
pivotal role in maintaining the fine balance between bleeding and thrombosis is being revealed. In this
Hematology Education:
article, we will discuss the role of VWF in hemostasis and give a concise overview of how our grow-
the education program for the
annual congress of the European ing understanding of VWF biology can be used in the design of treatment modalities for both bleed-
Hematology Association ing and thrombotic complications.

2009;3:38-43

Structure of von Willebrand factor gen.10,11 Finally, VWF interacts with the
von Willebrand factor (VWF) is a large platelet receptor αIIbβ3 via its C1 domain.12
adhesive, multimeric glycoprotein that ful-
fils two crucial roles in primary hemostasis. von Willebrand factor size regulation
First, VWF functions as a bridge between Endothelial VWF is either constitutively
subendothelial structures in the injured ves- secreted into the blood or is stored in
sel wall and circulating blood platelets, Weibel-Palade bodies,13 from which it is
thereby supporting the formation of a released upon stimulation or, as recent evi-
platelet-rich plug that prevents excessive dence suggests, via an unstimulated basal
bleeding and promotes wound healing. pathway.14 VWF present in platelets is stored
Second, by serving as a carrier protein for in α-granules and is only released after
factor VIII (FVIII), it protects rapid degrada- platelet activation.15 Whereas VWF storage
tion of this clotting factor. VWF is synthe- granules contain ultra-large (UL) VWF multi-
sized exclusively in endothelial cells and mers (>10.000 kDa) for acute and local
megakaryocytes as pre-proVWF subunits.1 release where needed, circulating VWF is
Each subunit consists of four types of repeat- composed of smaller multimers ranging
ed domains that are arranged in the from 2 (500 kDa) to up to 40 monomers (up
sequence D1-D2-D’-D3-A1-A2-A3-D4-B1- to 10.000 kDa).16 The size of the VWF multi-
B2-B3-C1-C2-CK2,3 (Figure 1). After removal mers is an important regulator of their reac-
of the signal peptide, pro-VWF becomes gly- tivity since the largest multimers are the
cosylated and dimerizes through the forma- most active ones. Hence, since the circula-
tion of disulfide bonds between C-terminal tion of UL (and thus hyper-reactive) VWF
CK domains in the rough endoplasmic retic- multimers can cause spontaneous intravas-
ulum.4 Upon transportation through the cular thrombosis, as occurs in the devastat-
Golgi complex and the post-Golgi compart- ing disease thrombotic thrombocytopenic
ments, pro-VWF is sulphatated; carbohy- purpura, their size must be tightly regulated
drates are further processed and multimers upon release from endothelial cells and
are formed through head-to-head disulfide platelets. Multimer size is controlled by the
bonds between D3 domains.5,6 Concomitant metalloprotease ADAMTS13 (a disintegrin
with the multimerization, the propeptide and metalloprotease with thrombospondin
(D1D2 domains, Figure 1) is proteolytically type 1 repeats, 13),17,18 which digests the
removed from mature VWF. The modular released UL-VWF into the smaller multimers
structure of VWF forms the basis of its mul- found in plasma. Interestingly, the proteolyt-
tifunctional properties (Figure 1). The N-ter- ic site of VWF (Y1605-M160619) is buried in
minal D’ and D3 domains contain binding the A2 domain, and becomes exposed after
sites for FVIII7 and heparin.8 The A1 domain conformational changes induced by, for
is the essential binding site for the platelet example, shear or denaturating agents.20-22
glycoprotein (GP) Ib/IX/V-complex9 and has Once in the bloodstream, VWF multimers
multiple binding sites for a variety of other circulate with a half-life of 8 to 12 hours.
molecules while the A3 domain comprises The exact clearance mechanism of VWF is
the major binding sites for fibrillar colla- not yet fully understood, but recent evidence

| 38 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

Figure 1. Structure of VWF. VWF is synthesized as a pre-pro-VWF that comprises a 22-amino acid (aa) residue signal pep-
tide (SP), a 741 aa-residue pro-peptide and the 2050 aa-residue mature subunit. The pro-peptide and the mature sub-
unit compose pro-VWF (2791 residues) consisting of four types of repeated domains which are indicated. After removal
of the signal peptide from pre-pro-VWF, pro-VWF translocates into the endoplasmic reticulum, where pro-VWF subunits
associate to ‘tail-to-tail’-dimers by the formation of disulfide-bonds between the cysteine-rich carboxyl-terminal CK
domains, which in turn, are transported to the Golgi. There, the ‘tail-to-tail’-dimers multimerize by forming ‘head-to-head’
disulfide bonds between the aminoterminal cysteine-rich D3 domains. The main binding sites that are important for the
hemostatic function of VWF are indicated together with the ADAMTS13 cleavage site. Major regions in which mutations
have been found that are associated with VWD type 1 and 2 are also shown.

suggests a role for macrophages in the removal of the


VWF/FVIII complex in both liver and spleen.23

von Willebrand factor and its role in platelet adhesion and


aggregation
Under (venous) low shear conditions (<1000 s–1), the
first steps of platelet adhesion at sites of vascular injury
are established through direct interactions of the
platelet collagen receptors α2β1 and GPVI with the
exposed collagen. However, under higher shear condi-
tions (>1000 s–1) initial platelet adhesion becomes more
dependent on VWF to capture fast-flowing platelets.
Upon exposure of subendothelial structures at vascu-
lar lesions, VWF binds to fibrillar collagens through its Figure 2. Platelet adhesion. Under high shear conditions,
A3 domain (Figure 2).10,11 Elegant studies revealed that a platelet adhesion is initiated by the transient tethering of
the platelets to VWF, which is anchored to the damaged
single high affinity binding site in collagen type III24 vessel wall. This reversible interaction between the
interacts with the front face of the VWF A3 domain.25,26 platelets and collagen-bound VWF slows down the
Fluid dynamic forces on collagen-bound VWF induce a platelets and allows the formation of firm interactions
between the collagen receptors α2β1 and GPVI and the
conformational alteration, stretching the globular form exposed collagen. The concomitant platelet activation
of VWF to a more elongated structure.27,28 This results in results in granular release and activation of integrin
the abolition of the shielding of the A1 domain by the αIIbβ3.
A2 and/or D’D3 domains;29-31 thereby exposing its
GPIbα binding sites, which are situated in two separate
areas in the A1 domain.32,33 The interaction between the
A1 domain and the platelet receptor GPIbα is character- occur in the absence of platelet activation at pathologi-
ized by fast on- and off-rates, allowing transient tether- cal shear rates that exceed 10,000 s–1 found, for example,
ing and translocation of platelets on the damaged vessel in stenotic coronary arteries. Above this threshold,
wall.34-37 This deceleration mechanism permits platelets active A1 domains become exposed in soluble VWF
to establish more firm interactions between their colla- multimers, promoting additional platelet recruitment
gen receptors and the exposed subendothelium, eventu- via GPIbα binding.38 Apparently, plasma VWF does not
ally leading to final platelet arrest at the site of injury only bind to exposed collagen, but also to the first layer
(Figure 2). Platelet adhesion is followed by platelet acti- of collagen-bound VWF multimers.39,40 This process of
vation and aggregation involving interaction of activat- reversible self-association between immobilized VWF
ed αIIbβ3 with RGD sequences in fibrinogen and VWF. and plasma VWF modulates the adhesive properties of
Recently, it was suggested that platelet aggregation can immobilized VWF.39

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 39 |
14th Congress of the European Hematology Association

Physiological importance of von Willebrand factor Type 3 Von Willebrand disease


von Willebrand disease Type 3 VWD is the most severe VWD form, charac-
VWD is a bleeding disorder caused by inherited VWF terized by recessive inheritance with a prevalence rang-
abnormalities. The prevalence of VWD patients that ing from 0.5 to 5 per million, with virtually completely
need treatment is estimated to be 0.002-0.01%, absent VWF and in addition low levels of FVIII. Some 90
although low VWF-levels may be found in up to 1% of associated mutations51 are scattered over the entire gene
the population.41-43 VWD is commonly characterized by comprising partial/total deletions, small insertions,
mucosal hemorrhages, such as epistaxis, menorrhagia, exon/intron boundary splice site mutations which result
gastro-intestinal and gingival bleeding, and often pro- in exon skipping or aberrant splicing, missense and
longed bleeding after trauma to skin or mucous mem- frameshift mutations.
branes, especially during or after surgery. The most
severely affected patients, who also have low FVIII lev- Treatment of von Willebrand disease
els, additionally suffer from spontaneous soft-tissue Current treatments
bleeding. Although the more than 250 mutations found The main goal in treatment of VWD is the correction
in VWD (database: www.sheffield.ac.uk/vwf), provide of hemostatic deficits caused by VWF and FVIII defi-
useful information on the cause of the bleeding pheno- ciency. At present, two different treatment modalities
type, current VWD classification44,45 is based on the make up the mainstay of VWD management: adminis-
underlying pathophysiological defects, indicative for tration of desmopressin, which induces the secretion of
optimal clinical treatment. Partial or complete quantita- endogenous VWF from endothelial cells, and direct
tive deficiency of VWF results in type 1 and 3 VWD administration of exogenous VWF contained in plasma
respectively, and qualitative defects in type 2 VWD. derived concentrates.49,54-56

Type 1 von Willebrand disease Future treatments


This is found in 60-80% of all VWD cases, and is due Recently, new strategies to treat VWD are being
to mild/severe quantitative defects of VWF and FVIII. explored, including the development of a recombinant
Although low levels can be due to various causes, such VWF preparation, the use of interleukin (IL)-11 and
as ABO blood group, recent large studies indicated that attempts to provide a long-term cure for VWD by VWF
most are associated with mutations within VWF.46-48 gene transfer. Use of a standardized preparation of
Highly penetrant dominant mutations, mostly single recombinant VWF (rVWF) produced in CHO cells under
amino acid mutations in the D3-domain, either affect plasma-free conditions57 would eradicate the risk of
production following disturbed intracellular transport of blood-borne pathogen transmission in patients and
dimeric proVWF or provoke rapid clearance from the would bypass the high degree of variability present
circulation.49 among the FVIII/VWF concentrates. Recent observa-
tions that IL-11 increases VWF and FVIII levels in mice,58
Type 2 von Willebrand disease dogs59 and humans60 (by a not yet identified mechanism)
Subtype 2A VWD is the most common qualitative vari- stimulate further research on the use of recombinant IL-
ant with generally an autosomal dominant inheritance. 11 to treat VWD patients.58,60,61 Since VWD is a mono-
The large and intermediate multimers are lost due to genic disease, it is an appealing candidate for gene ther-
mutations in either the propeptide or the C-terminal cys- apy.62-66 Indeed, instead of repetitive on-demand replace-
tein-knot-domain, resulting in defective assembly and/or ment of the defective protein, a permanent correction of
in increased susceptibility to ADAMTS13.50 As a conse- the underlying genetic defect would provide a life-long
quence, both platelet- and collagen-binding are dis- cure that would dramatically improve the quality of life
turbed. Isolated defects within the A1-domain, causing of severe bleeders. Original concerns on the incorpora-
an impaired binding to platelet GPIbα51 give rise to sub- tion of the large VWF cDNA (8,4kb) into integrating
type 2M VWD, an autosomal dominant variant with viral vectors have been eliminated by the recent con-
(near) normal multimeric distribution. One family has struction of a lentiviral vector accommodating full
been identified with a mutation in the A3-domain, length VWF.65 Furthermore, using this vector, the pheno-
resulting in defective binding to collagen,52 also possibly type of blood outgrowth endothelial cells isolated from
causing type 2M VWD. In contrast, in subtype 2B VWD, dogs suffering from VWD type 3 could be completely
usually inherited as an autosomal dominant trait, various corrected. Since lentiviral vectors would predominantly
mutations within the A1-domain cause an increased and target the liver upon systemic administration, transgene
frequent spontaneous binding of VWF to GPIbα.51 The encoded VWF would be synthesized in liver cells; how-
larger multimers are lost from plasma as binding to ever, this is an ectopic site for VWF expression.
platelets facilitates proteolysis by ADAMTS13 and/or Interestingly, using the technique of hydrodynamic
results in faster clearance of platelet-VWF complexes, gene transfer in a murine model of severe VWD, we and
which often gives rise to thrombocytopenia.53 Together, others demonstrated that the liver is capable of produc-
they explain the apparently paradoxical bleeding diathe- ing VWF containing the full range of multimers.63,66,67
sis in these “gain-of-function” phenotypes. Subtype 2N Transgene encoded, liver expressed VWF restored FVIII
VWD, an autosomal recessive disorder, due to a levels, and most importantly, reversed the prolongation
decreased affinity of VWF for FVIII, is caused by some of the tail bleeding time,63,66,67 and in a FeCl3-induced
20 homozygous or compound heterozygous missense thrombosis model, restored the platelet plug forming
mutations in the D’ or D3 FVIII binding-domain. In case capacity, still one of the main functions of VWF in
of severe FVIII reduction, type 2N VWD resembles mild hemostasis.66,68 It is evident that these first steps towards
or moderate hemophilia A. gene therapy for VWD are only the beginning. More

| 40 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

studies in the future will undoubtedly reveal the possi- ic efficacy has been shown in two experimental throm-
bilities and limitations of VWF gene therapy to cure bosis models in dogs.83,84 ALX-0081 is a cameloid anti-
severe VWD. VWF A1-domain bivalent nanobody that reduces
thrombus formation in a modified Folts model in the
von Willebrand factor as antithrombotic target baboon femoral artery and ex vivo ristocetin induced
Because of the pivotal role of VWF in hemostasis and platelet aggregation.85 ARC1779 is a synthetically man-
thrombosis, there is an increasing interest in targeting ufactured, modified DNA/RNA aptamer, which by
VWF as antithrombotic strategy. Indeed, recently, many binding to the VWF A1 domain, inhibits its interaction
compounds have been developed that inhibit the bio- with GPIbα. This compound prevents the formation of
logical activity of VWF. As outlined above, one of the occlusive thrombi in cynomolgus macaques during con-
major roles of VWF is mediating platelet adhesion at tinuous electrical injury for six hours with only modest
sites of vascular injury. Recruitment of platelets is prolongation of the bleeding time at the optimal
accomplished by the binding of VWF to both platelets antithrombotic dose.86 In a placebo-controlled study in
(GPIbα) and exposed subendothelial collagen. 47 healthy volunteers, ARC1779 was well tolerated,
Consequently, interfering with the binding of VWF to and maintained its ex vivo activity, without bleeding
collagen, as well as inhibiting the binding of VWF to complications.87
GPIbα are appealing strategies for antithrombotic ther- It is clear that a potent antithrombotic effect is
apy. Since these interactions are first steps in platelet obtained at inhibitor doses that do not markedly affect
adhesion without any known redundancy, inhibition is bleeding times when the collagen-VWF-GPIbα axis is
expected to have a good antithrombotic potency. blocked. However, it is not yet fully clear whether
Moreover, it is anticipated that inhibition of platelet reduced bleeding times will also translate into a lower
adhesion might induce less clinical bleeding problems bleeding risk in patients. Nevertheless, all data clearly
compared to the antiplatelet drugs currently used in the indicate that the therapeutic window with these kind of
clinic. Indeed, since VWF has a dominant role in platelet compounds is significantly larger than with the current-
adhesion, essentially at high shear stress, interfering ly available antiplatelet agents. In analogy with this, we
with VWF-mediated platelet adhesion would be more and others recently demonstrated that, in a murine
specific for high-shear pathological (stenosed) arterial ischemic stroke model, deficiency of VWF or blocking
systems, leaving the hemostatic situation in venous sys- GPIbα protected the mice from brain ischemia without
tems rather intact.69-72 inducing excessive bleeding.88,89 If this can be confirmed
During the late nineties, two antithrombotic anti- in man, this class of compounds might even become the
human VWF antibodies were developed in two differ- treatment of choice for ischemic stroke.
ent groups; one targeting the VWF A1, thus inhibiting
its binding to GPIb and the other one targeting the VWF S.F.D.M. and K.V. are postdoctoral fellows of the FWO
A3 domain, thereby inhibiting VWF binding to collagen. (Fonds voor Wetenschappelijk Onderzoek, Belgium).
The first, monoclonal antibody AJvW2, was developed
by Yoshimoto and colleagues and was found to inhibit
the VWF-GPIbα interaction in various in vitro assays.73 References
When tested in experimental thrombosis models in ani-
mals, AJvW2 demonstrated a potent antithrombotic 1. Wagner DD. Cell Biology of Von Willebrand Factor. Ann Rev
Cell Biol 1990;6:217-46.
effect with no indication for severe bleeding complica- 2. Sadler JE, Shelton-Inloes BB, Sorace JM, Harlan JM, Titani K,
tions.73-75 The second antibody, murine monoclonal anti- Davie EW et al. Cloning and characterization of two cDNAs
body 82D6A3, was developed in our group and has an coding for human von Willebrand factor. Proc Natl Acad Sci
USA 1985;82:6394-8.
epitope that overlaps with the collagen binding region 3. Verweij CL, Diergaarde PJ, Hart M, Pannekoek H. Full-length
in the VWF A3-domain;76,77 thereby, inhibiting the bind- von Willebrand factor (vWF) cDNA encodes a highly repeti-
ing of VWF to fibrillar collagen type I and III under stat- tive protein considerably larger than the mature vWF subunit.
EMBO J 1986;5:1839-47.
ic and flow conditions.76,78 The in vivo antithrombotic 4. Katsumi A, Tuley EA, Bodo I, Sadler JE. Localization of disul-
efficacy of 82D6A3 has been demonstrated in baboons fide bonds in the cystine knot domain of human von
using a modified Folts model,79 where a strong reduction Willebrand factor. J Biol Chem 2000;275:25585-94.
5. Mayadas TN, Wagner DD. Vicinal cysteines in the prose-
of thrombus formation was observed after administra- quence play a role in von Willebrand factor multimer assem-
tion of 300 µg/kg 82D6A3, without significantly pro- bly. Proc Natl Acad Sci USA 1992;89:3531-5.
longing the bleeding time. In the mean time, both 6. Purvis AR, Gross J, Dang LT, Huang RH, Kapadia M, Townsend
AJvW2 and 82D6A3 have been humanized80,81 for test- RR, Sadler JE. Two Cys residues essential for von Willebrand
factor multimer assembly in the Golgi. Proc Natl Acad Sci USA
ing in humans. Humanized AJvW2 (named AJW200) 2007;104:15647-52.
was well tolerated when administered to 24 male 7. Foster PA, Fulcher CA, Marti T, Titani K, Zimmerman TS. A
healthy humans and ristocetin cofactor activity was sig- major factor VIII binding domain resides within the amino-
terminal 272 amino acid residues of von Willebrand factor. J
nificantly reduced until 12 hours after infusion of 0.01- Biol Chem 1987;262:8443-6.
0.05 mg/kg AJW200, without changing template bleed- 8. Fretto LJ, Fowler WE, McCaslin DR, Erickson HP, McKee PA.
ing times.82 In pursuit of these first studies, several other Substructure of human von Willebrand factor. Proteolysis by
V8 and characterization of two functional domains. J Biol
components have been developed, all targeting the Chem 1986;261:15679-89.
VWF A1 domain. GPG-290 is a recombinant fragment 9. Sixma JJ, Schiphorst ME, Verweij CL, Pannekoek H. Effect of
of human GPIbα with two gain-of-function mutations deletion of the A1 domain of von Willebrand factor on its
binding to heparin, collagen and platelets in the presence of
(G233V/ M239V) grafted on the Fc part of a human ristocetin. Eur J Biochem 1991;196:369-75.
IgG1, and expressed as a homodimer linked by the two- 10. Cruz MA, Yuan H, Lee JR, Wise RJ, Handin RI. Interaction of
disulfide bonds in the Fc hinge region. Its antithrombot- the von Willebrand factor (vWF) with collagen. Localization of
the primary collagen-binding site by analysis of recombinant

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 41 |
14th Congress of the European Hematology Association

vWF A domain polypeptides. J Biol Chem 1995;270:19668. 33. Huizinga EG, Tsuji S, Romijn RA, Schiphorst ME, de Groot
11. Lankhof H, van Hoeij M, Schiphorst ME, Bracke M, Wu YP, PG, Sixma JJ, Gros P. Structures of glycoprotein Ibalpha and its
Ijsseldijk MJ et al. A3 domain is essential for interaction of von complex with von Willebrand factor A1 domain. Science 2002;
Willebrand factor with collagen type III. Thromb Haemost 297:1176-9.
1996;75:950-8. 34. Savage B, Saldivar E, Ruggeri ZM. Initiation of platelet adhe-
12. Beacham DA, Wise RJ, Turci SM, Handin RI. Selective inacti- sion by arrest onto fibrinogen or translocation on von
vation of the Arg-Gly-Asp-Ser (RGDS) binding site in von Willebrand factor. Cell 1996;84:289-97.
Willebrand factor by site-directed mutagenesis. J Biol Chem. 35. Doggett TA, Girdhar G, Lawshe A, Schmidtke DW, Laurenzi
1992;267:3409-15. IJ, Diamond SL, Diacovo TG. Selectin-like kinetics and biome-
13. Weibel ER, Palade GE. New cytoplasmatic components in chanics promote rapid platelet adhesion in flow: the
arterial endothelia. J Cell Biol 1964;23:101-12. GPIb(alpha)-vWF tether bond. Biophys J 2002;83:194-205.
14. Giblin JP, Hewlett LJ, Hannah MJ. Basal secretion of von 36. Mody NA, Lomakin O, Doggett TA, Diacovo TG, King MR.
Willebrand factor from human endothelial cells. Blood 2008. Mechanics of transient platelet adhesion to von Willebrand
15. Cramer EM, Bretongorius J, Beesley JE, Martin JF. Ultra- factor under flow. Biophys J 2005;88:1432-43.
structural demonstration of tubular inclusions coinciding with 37. Yago T, Lou J, Wu T, Yang J, Miner JJ, Coburn L et al. Platelet
von Willebrand factor in pig megakaryocytes. Blood 1988;71: glycoprotein Ibalpha forms catch bonds with human WT vWF
1533-8. but not with type 2B von Willebrand disease vWF. J Clin Invest
16. Fowler WE, Fretto LJ, Hamilton KK, Erickson HP, McKee PA. 2008;118:3195-207.
Substructure of human von Willebrand factor. J Clin Invest. 38. Ruggeri ZM, Orje JN, Habermann R, Federici AB, Reininger
1985;76:1491-1500. AJ. Activation-independent platelet adhesion and aggregation
17. Fujikawa K, Suzuki H, McMullen B, Chung D. Purification of under elevated shear stress. Blood 2006;108:1903-10.
human von Willebrand factor-cleaving protease and its identi- 39. Savage B, Sixma JJ, Ruggeri ZM. Functional self-association of
fication as a new member of the metalloproteinase family. von Willebrand factor during platelet adhesion under flow.
Blood 2001;98:1662-6. Proc Natl Acad Sci USA 2002;99:425-30.
18. Gerritsen HE, Robles R, Lammle B, Furlan M. Partial amino 40. Ulrichts H, Vanhoorelbeke K, Girma JP, Lenting PJ, Vauterin S,
acid sequence of purified von Willebrand factor-cleaving pro- Deckmyn H et al. The von Willebrand factor self-association
tease. Blood 2001;98:1654-61. is modulated by a multiple domain interaction. J Thromb
19. Dent JA, Berkowitz SD, Ware J, Kasper CK, Ruggeri ZM. Haemost 2005;3:552-61.
Identification of a cleavage site directing the immunochemical 41. Rodeghiero F, Castaman G, Dini E. Epidemiological investiga-
detection of molecular abnormalities in type IIA von tion of the prevalence of von Willebrand's disease. Blood 1987;
Willebrand factor. Proc Natl Acad Sci USA 1990;87:6306-10. 69:454-9.
20. Furlan M, Robles R, Lämmle B. Partial purification and charac- 42. Werner EJ, Broxson EH, Tucker EL, Giroux DS, Shults J,
terization of a protease from human plasma cleaving von Abshire TC et al. Prevalence of von Willebrand disease in chil-
Willebrand factor to fragments produced by in vivo proteoly- dren: a multiethnic study. J Pediatr1993;123:893-8.
sis. Blood 1996;87:4223-34. 43. Sadler JE, Mannucci PM, Berntorp E, Bochkov N, Boulyjenkov
21. Tsai HM, Sussman II, Nagel RL. Shear stress enhances the pro- V, Ginsburg D et al. Impact, diagnosis and treatment of von
teolysis of von Willebrand factor in normal plasma. Blood Willebrand disease. Thromb Haemost 2000;84:160-74.
1994;83:2171-9. 44. Sadler JE. A revised classification of von Willebrand disease.
22. Tsai HM. Physiologic cleavage of von Willebrand factor by a For the Subcommittee on von Willebrand Factor of the
plasma protease is dependent on its conformation and Scientific and Standardization Committee of the International
requires calcium ion. Blood 1996;87:4235-44. Society on Thrombosis and Haemostasis. Thromb Haemost
23. van Schooten CJ, Shahbazi S, Groot E, Oortwijn BD, van den 1994;71:520-5.
Berg HM, Denis CV et al. Macrophages contribute to the cel- 45. Sadler JE, Budde U, Eikenboom JC, avaloro EJ, Hill FG,
lular uptake of von Willebrand factor and factor VIII in vivo. Holmberg L et al. Update on the pathophysiology and classifi-
Blood 2008;112:1704-12. cation of von Willebrand disease: a report of the
24. Lisman T, Raynal N, Groeneveld D, Maddox B, Peachey AR, Subcommittee on von Willebrand Factor. J Thromb Haemost
Huizinga EG et al. A single high-affinity binding site for von 2006;4:2103-14.
Willebrand Factor in collagen III, identified using synthetic 46. Eikenboom J, VAN M, V, Putter H, Goodeve A, Rodeghiero F,
triple-helical peptides. Blood 2006; 108:1704-12. Castaman G et al. Linkage analysis in families diagnosed with
25. Nishida N, Sumikawa H, Sakakura M, Shimba N, Takahashi type 1 von Willebrand disease in the European study, molecu-
H, Terasawa H et al. Collagen-binding mode of vWF-A3 lar and clinical markers for the diagnosis and management of
domain determined by a transferred cross-saturation experi- type 1 VWD. J Thromb Haemost 2006;4:774-82.
ment. Nat Struct Biol 2003;10:53-8. 47. James PD, Notley C, Hegadorn C, et al. The mutational spec-
26. Romijn RA, Bouma B, Wuyster W, Gros P, Kroon J, Sixma JJ, trum of type 1 von Willebrand disease: results from a
Huizinga EG. Identification of the collagen-binding site of the Canadian cohort study. Blood 2007;109:145-54.
von Willebrand factor A3-domain. J Biol Chem 2001;276: 48. Goodeve A. Genetics of type 1 von Willebrand disease. Curr
9985-91. Opin Hematol 2007;14:444-9.
27. Schneider SW, Nuschele S, Wixforth A, Gorzelanny C, 49. Nichols WL, Hultin MB, James AH, Leggo J, Tuttle A, Tinlin S
Alexander-Katz A, Netz RR et al. Shear-induced unfolding trig- et al. von Willebrand disease (VWD): evidence-based diagno-
gers adhesion of von Willebrand factor fibers. Proc Natl Acad sis and management guidelines, the National Heart, Lung, and
Sci USA 2007;104:7899-03. Blood Institute (NHLBI) Expert Panel report (USA).
28. Siedlecki CA, Lestini BJ, Kottke-Marchant KK, Eppell SJ, Haemophilia 2008;14:171-232.
Wilson DL, Marchant RE et al. Shear-dependent changes in the 50. O'Brien LA, Sutherland JJ, Weaver DF, Lillicrap D. Theoretical
three-dimensional structure of human von Willebrand factor. structural explanation for Group I and Group II, type 2A von
Blood 1996;88:2939-50. Willebrand disease mutations. J Thromb Haemost 2005;3:796-
29. Martin C, Morales LD, Cruz MA. Purified A2 domain of von 7.
Willebrand factor binds to the active conformation of von 51. Lillicrap D. Von Willebrand disease - phenotype versus geno-
Willebrand factor and blocks the interaction with platelet gly- type: deficiency versus disease. Thromb Res 2007;120 Suppl
coprotein Ibalpha. J Thromb Haemost 2007;5:1363-70. 1:S11-S16.
30. Nishio K, Anderson PJ, Zheng XL, Sadler JE. Binding of platelet 52. Ribba AS, Loisel I, Lavergne JM, Juhan-Vague I, Obert B,
glycoprotein Ib alpha to von Willebrand factor domain A1 Cherel G et al. Ser968Thr mutation within the A3 domain of
stimulates the cleavage of the adjacent domain A2 by von Willebrand factor (VWF) in two related patients leads to a
ADAMTS13. Proc Natl Acad Sci USA 2004;101:10578-83. defective binding of VWF to collagen. Thromb Haemost
31. Ulrichts H, Udvardy M, Lenting PJ, Pareyn I, Vandeputte N, 2001;86:848-54.
Vanhoorelbeke K et al. Shielding of the A1 Domain by the 53. Federici AB, Mannucci PM, Castaman G, Baronciani L,
D'D3 Domains of von Willebrand Factor Modulates Its Bucciarelli P, Canciani MT et al. Clinical and molecular predic-
Interaction with Platelet Glycoprotein Ib-IX-V. J Biol Chem tors of thrombocytopenia and risk of bleeding in patients with
2006;281:4699-707. von Willebrand disease type 2B: A cohort study of 67 patients.
32. Dumas JJ, Kumar R, McDonagh T, Sullivan F, Stahl ML, Blood 2008.
Somers WS et al. Crystal structure of the wild-type von 54. De Meyer S, De Maeyer B, Deckmyn H, Vanhoorelbeke K.
Willebrand factor A1-glycoprotein Ibalpha complex reveals Von Willebrand factor: drug and drug target. Curr Drug
conformation differences with a complex bearing von Targets Cardiovasc Haematol Disord 2009;9:9-20.
Willebrand disease mutations. J Biol Chem 2004;279:23327- 55. De Meyer S, Deckmyn H, Vanhoorelbeke K. Von Willebrand
34. factor to the rescue. Blood 2009;Mar 24 [Epub ahead of print]

| 42 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

56. Mannucci PM. How I treat patients with von Willebrand dis- 75. Kageyama S, Yamamoto H, Nakazawa H, Yoshimoto R. Anti-
ease. Blood 2001;97:1915-19. human vWF monoclonal antibody, AJvW-2 Fab, inhibits repet-
57. Plaimauer B, Schlokat U, Turecek PL, Mitterer A, Mundt W, itive coronary artery thrombosis without bleeding time pro-
Auer W et al. Recombinant von Willebrand factor: preclinical longation in dogs. Thromb Res 2001;101:395-404.
development. Semin Thromb Hemost 2001;27:395-403. 76. Vanhoorelbeke K, Depraetere H, Romijn RAP, Huizinga EG,
58. Denis CV, Kwack K, Saffaripour S, Maganti S, André P, Schaub De Maeyer M, Deckmyn H et al. A consensus tetrapeptide
RG et al. Interleukin 11 significantly increases plasma von selected by phage display adopts the conformation of a dom-
Willebrand factor and factor VIII in wild type and von inant discontinuous epitope of a monoclonal Anti-VWF anti-
Willebrand disease mouse models. Blood 2001;97:465-72. body that inhibits the von Willebrand factor-collagen interac-
59. Olsen EH, McCain AS, Merricks EP, Fischer TH, Dillon IM,
Raymer RA et al. Comparative response of plasma VWF in tion. J Biol Chem 2003;278:37815-21.
dogs to up-regulation of VWF mRNA by interleukin-11 versus 77. Staelens S, Hadders MA, Vauterin S, Platteau C, De Maeyer
Weibel-Palade body release by desmopressin (DDAVP). Blood M, Vanhoorelbeke K et al. Paratope determination of the
2003;102:436-41. antithrombotic antibody 82D6A3 based on the crystal struc-
60. Ragni MV, Jankowitz RC, Chapman HL, Merricks EP, Kloos ture of its complex with the von Willebrand factor A3-domain.
MT, Dillow AM et al. A phase II prospective open-label esca- J Biol Chem 2006;281:2225-31.
lating dose trial of recombinant interleukin-11 in mild von 78. Hoylaerts MF, Yamamoto H, Nuyts K, Vreys I, Deckmyn H,
Willebrand disease. Haemophilia 2008;14:968-77. Vermylen J et al. von Willebrand factor binds to native colla-
61. Jankowitz RC, Ragni MV, Chapman HL, et al. Recombinant gen VI primarily via its A1 domain. Biochem J 1997;324(Pt
Interleukin-11 (rhIL-11) in Women with Refractory 1):185-91.
Menorrhagia and Von Willebrand Disease. ASH Annual 79. Wu D, Vanhoorelbeke K, Cauwenberghs N, Meiring M,
Meeting Abstracts 2008;112:1210. Depraetere H, Kotze HF, Deckmyn H. Inhibition of the von
62. De Meyer SF, Pareyn I, Baert J, Deckmyn H, Vanhoorelbeke K. Willebrand (VWF)-collagen interaction by an antihuman VWF
False Positive Results in Chimeraplasty for Von Willebrand monoclonal antibody results in abolition of in vivo arterial
Disease. Thromb Res 2007;119:93-104. platelet thrombus formation in baboons. Blood 2002;99:3623-
63. Lenting PJ, de Groot PG, De Meyer SF, Vanhoorelbeke K, Pruss 28.
C, Lillicrap D et al. Correction of the bleeding time in von 80. Kageyama S, Yamamoto H, Nakazawa H, Matsushita J,
Willebrand factor (VWF)-deficient mice using murine VWF. Kouyama T, Gonsho A et al. Pharmacokinetics and pharmaco-
Blood 2007;109:2267-8.
64. Pergolizzi RG, Jin GC, Chan D, Pierre L, Bussel J, Ferris B et al. dynamics of AJW200, a humanized monoclonal antibody to
Correction of a murine model of von Willebrand disease by von Willebrand factor, in monkeys. Arterioscler Thromb Vasc
gene transfer. Blood 2006;108:862-9. Biol 2002;22:187-92.
65. De Meyer SF, Vanhoorelbeke K, Chuah MK, Pareyn I, Gillijns 81. Staelens S, Desmet J, Ngo TH, Vauterin S, Pareyn I, Barbeaux
V, Hebbel RP et al. Phenotypic correction of von Willebrand P et al. Humanization by variable domain resurfacing and
disease type 3 blood-derived endothelial cells with lentiviral grafting on a human IgG(4), using a new approach for deter-
vectors expressing von Willebrand factor. Blood 2006;107: mination of non-human like surface accessible framework
4728-36. residues based on homology modelling of variable domains.
66. De Meyer SF, Vandeputte N, Pareyn I, Petrus I, Lenting PJ, Mol Immunol 2005;43:1243-57.
Chuah MK et al. Restoration of plasma von Willebrand factor 82. Machin SJ, Clarke C, Ikemura O, et al. A humanized mono-
deficiency is sufficient to correct thrombus formation after clonal antibody against VWF A1 domain inhibits VWF:RiCof
gene therapy for severe von Willebrand disease. Arterioscler activity and platelet adhesion in human volunteers. J Thromb
Thromb Vasc Biol 2008;28: 1621-6. Haemost 2003;1 Supplement 1 July:OC328.
67. Marx I, Lenting PJ, Adler T, Pendu R, Christophe OD, Denis 83. Hennan JK, Swillo RE, Morgan GA, Leik CE, Brooks JM, Shaw
CV et al. Correction of bleeding symptoms in von Willebrand GD et al. Pharmacologic inhibition of platelet vWF-GPIb alpha
factor-deficient mice by liver-expressed von Willebrand factor interaction prevents coronary artery thrombosis. Thromb
mutants. Arterioscler Thromb Vasc Biol 2008;28:419-24. Haemost 2006;95:469-75.
68. Marx I, Christophe OD, Lenting PJ, Rupin A, Vallez MO,
Verbeuren TJ et al. Altered thrombus formation in von 84. Wadanoli M, Sako D, Shaw GD, Schaub RG, Wang Q,
Willebrand factor-deficient mice expressing von Willebrand Tchernychev B et al. The von Willebrand factor antagonist
factor variants with defective binding to collagen or GPIIbIIIa. (GPG-290) prevents coronary thrombosis without prolonga-
Blood 2008;112:603-9. tion of bleeding time. Thromb Haemost 2007;98:397-405.
69. De Meyer SF, Vanhoorelbeke K, Ulrichts H, Staelens S, Feys 85. Silence K, Jonckheere H, Casteels P, Roodt J, Meiring M,
HB, Salles I et al. Development of monoclonal antibodies that Lamprecht S, et al. ALX-0081 nanobody , an engineered biva-
inhibit platelet adhesion or aggregation as potential anti- lent anti-thrombotic drug candidate with improved efficacy
thrombotic drugs. Cardiovasc Hematol Disord Drug Targets and safety as compared to the marketed drugs. Blood 2008;
2006;6:191-207. 108.
70. Ulrichts H, Vanhoorelbeke K, Van de walle G, Katsutani S, De 86. Rottman JB, Gilbert M, Marsh H, Boomer R, Fraone J, Makim
Meyer S, Staelens S, et al. New approaches for antithrombot- A, et al. An anti-von Willebrand's factor A1 domain aptamer
ic antiplatelet therapies. Current Med Chem 11, 2261-3. 2004. inhibits arterial thrombosis induced by electrical injury in
71. Vanhoorelbeke K, Ulrichts H, Schoolmeester A, Deckmyn H. cynomolgus macaques. J Thromb Haemost 2007;5 Suppl 2, P-
Inhibition of platelet adhesion to collagen as a new target for S-664.
antithrombotic drugs. Curr Drug Targets Cardiovasc 87. Gilbert JC, DeFeo-Fraulini T, Hutabarat RM, Horvath CJ,
Haematol Disord 2003;3:125-40. Merlino PG, Marsh HN et al. First-in-human evaluation of anti
72. Vanhoorelbeke K, Ulrichts H, Van De Walle G, Fontayne A, von Willebrand factor therapeutic aptamer ARC1779 in
Deckmyn H. Inhibition of platelet glycoprotein Ib and its healthy volunteers. Circulation 2007;116:2678-86.
antithrombotic potential. Current Pharm Des 2007;13:2684-
97. 88. Kleinschnitz C, Pozgajova M, Pham M, Bendszus M,
73. Kageyama S, Yamamoto H, Nagano M Arisaka H, Kayahara T, Nieswandt B, Stoll G et al. Targeting platelets in acute experi-
Yoshimoto R et al. Anti-thrombotic effects and bleeding risk mental stroke: impact of glycoprotein Ib, VI, and IIb/IIIa
of AJvW-2, a monoclonal antibody against human von blockade on infarct size, functional outcome, and intracranial
Willebrand factor. Br J Pharmacol 1997; 122:165-71. bleeding. Circulation 2007;115:2323-30.
74. Yamamoto H, Vreys I, Stassen JM, Yoshimoto R, Vermylen J, 89. Kleinschnitz C, De Meyer SF, Schwarz T, Austinat M,
Hoylaerts MF et al. Antagonism of vWF inhibits both injury Vanhoorelbeke K, Nieswandt B et al. Deficiency of von
induced arterial and venous thrombosis in the hamster. Willebrand Factor Protects Mice from Ischemic Stroke. Blood
Thromb Haemost 1998;79:202-10. 2009;113:3600-3.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 43 |
Bleeding disorders

Phenotype, genotype and classification


in von Willebrand disease

A. Goodeve A B S T R A C T

The common autosomally-inherited, mucocutaneous bleeding disorder von Willebrand disease


Academic Unit of Haematology, (VWD) displays a range of different phenotypes and patterns of inheritance, which can be categorised
University of Sheffield School of
Medicine and Biomedical Science into three different types. Types 1 and 3 describe partial and virtually complete quantitative deficien-
and Sheffield Diagnostic Genetics cies of plasma von Willebrand factor (VWF), whereas type 2 comprises four subtypes differentiated by
Service, Sheffield Children’s NHS the specific VWF functions that they perturb. Mutation type and location responsible for each VWD
Foundation Trust, Sheffield, UK type are explored, along with a look at the mechanisms by which mutations result in disease.

Hematology Education:
the education program for the
annual congress of the European
Hematology Association

2009;3:44-49

on Willebrand factor (VWF) is a large the normal complement of high molecular

v multimeric plasma glycoprotein essen-


tial for primary hemostasis. When dam-
age to blood vessels occurs, it binds to colla-
weight (HMW) forms are present.5 Upon
secretion from endothelial cells or platelets,
VWF is cleaved by its protease, a disintegrin
gen in exposed sub-endothelium, and fol- and metalloproteinase with throm-
lowing a conformational change, to the bospondin motifs (ADAMTS13) between
platelet Glycoprotein 1b (Gp1b). This initi- residues p.Tyr1605_Met1606 to reduce its
ates platelet thrombus formation at the site size and thrombogenic potential.
of vascular damage. Additionally, VWF
binds to the coagulation factor VIII (FVIII), von Willebrand disease phenotype
protecting it from premature proteolytic Bleeding assessment
degradation in the circulation and delivering Assessment of whether an individual
it to injury sites. von Willebrand disease bleeds more than is normal can be difficult
(VWD), a common autosomally-inherited to assess and a number of clinical tools for
bleeding disorder results from perturbation taking a detailed bleeding history have been
of these functions through quantitative developed. Tosetto and colleagues devised
(types 1 and 3) or qualitative (type 2) defects. an extensive bleeding score, for which utili-
The disorder clinically affects approximately ty was demonstrated in two European stud-
1 in 10,000 individuals,1 with symptoms ies on type 1 VWD.6,7 Scores are given to
ranging from nose bleeds and easy bruising each of several different bleeding symptoms,
through to bleeding into joints and gastroin- including epistaxis, bruising and bleeding
testinal bleeding, with menorrhagia and after tooth extraction and surgery. Each
bleeding associated with childbirth being symptom is given increasingly positive
common symptoms in affected females. scores where intervention is required for
The VWF protein is composed of repeated cessation of bleeding. In the Molecular and
domains in the order D1-D2-D’-D3-A1-A2- Clinical Markers for the Diagnosis and
A3-D4-B1-B2-B3-C1-C2-CK. This large pro- Management of Type 1 VWD (MCMDM-
tein, comprising 2,813 amino acids,2 under- 1VWD) study, scores ranged from -1 to +4
goes carboxyl terminal dimerisation through for each symptom. Possible scores ranged
disulphide bonds joining CK domains, fol- from -3 (no bleeding following tooth extrac-
lowed by multimerisation involving inter- tion and two surgeries) to +45. Median
molecular disulphide bonding of adjacent scores were -1 in healthy controls and 9 in
D3 domains, prior to cleavage of the propep- type 1 VWD index cases.7 The majority of
tide (D1-D2 domains).3 Multimers, compris- index cases had significantly higher scores
ing up to 40 dimeric subunits4 are present in than healthy controls did. The recently pub-
the circulation and are required for full lished modified MCMDM-1VWD bleeding
hemostatic function. SDS-agarose gels can score8 uses symptoms shown to best dis-
be used to assess the extent of multimerisa- criminate bleeders from non-bleeders and
tion of VWF present in plasma, and whether can be completed in 5 to 10 minutes. Such a

| 44 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

score may become part of an initial standardised assess- This is because they frequently detect heterozygous sin-
ment of possible VWD patients in the future. gle nucleotide polymorphisms (SNPs), as well as the
mutation sought, but do not indicate the nature of the
Laboratory assessment variant identified.12 Therefore, DNA sequencing is the
Determination of VWF quantity and function in indi- preferred genetic analysis method. Determination of
viduals investigated for possible VWD requires a num- whether a sequence variant is pathogenic can be chal-
ber of tests: (i) amount of protein can be quantified lenging; in vitro expression has a useful role in demon-
using immunological analysis (VWF:Ag); (ii) ability to strating the likely pathogenicity of sequence variants.
aggregate platelets assessed following conformational Types 2 and 3 VWD have been examined for muta-
change mimicking binding to subendothelium stimulat- tions for nearly 20 years. As mutations in type 2 VWD
ed by the antibiotic ristocetin (VWF:RCo); (iii) quantity affect specific functional domains, and an isolated sec-
of FVIII through its activity in the coagulation cascade tion of VWF can be targeted for analysis, they have been
(FVIII:C). The normal range for each of these parame- more accessible to examination, and many mutations
ters is approximately 50 to 200 IU/dL. Specialist tests to have been reported, helping to define the extent of func-
differentiate specific subtypes are described in a subse- tional domains. A web-based database, compiled on
quent section. behalf of the International Society Thrombosis and
Haemostasis Scientific and Standardisation Committee
Genotype (ISTH-SSC) on VWF (VWFdb) is a repository for infor-
The VWF gene (VWF) is located near the tip of chro- mation on VWF sequence variation.11
mosome 12 at 12p13.3. The gene covers 178 kb of
genomic DNA organised into 52 exons. A partial von Willebrand disease
pseudogene VWFP on chromosome 22 (22q11.22- Type 1 von Willebrand disease
11.23), which is 97% similar in sequence to VWF,9 com- This most common form of VWD (up to 70% of
plicates genetic analysis and promotes gene conversion cases) comprises patients with a partial quantitative
mutations.10 Due to the very polymorphic nature of VWF deficiency, where the proportion of HMW multi-
VWF,11 mutation scanning techniques, including dena- mers is not significantly decreased. Plasma levels of
turing high performance liquid chromatography VWF range from approximately 5 to 50 IU/dL, with
(dHPLC), conformation sensitive gel electrophoresis VWF:Ag and VWF:RCo being similarly reduced, and
(CSGE) and single strand conformation polymorphism FVIII:C levels typically 1.5 times higher than those of
(SSCP) analysis do not work well in mutation detection. VWF (Table 1). Bleeding symptoms are often moderate

Table 1. Phenotype, genotype and mechanisms involved in von Willebrand disease.

VWD Type Description Phenotype* Genotypes Inheritance† & Mechanism


responsible penetrance

1 Partial quantitative VWF VWF:RCo & VWF:Ag Missense Generally AD Under investigation, but include
deficiency, HMW multimers reduced in parallel Other in-frame Reduced penetrance increased clearance
not significantly decreased VWF:Ag >5 IU/dL alterations common Enhanced ADAMTS13
Null alleles Small proportion AR susceptibility
Interaction with ABO
blood group O
Contribution of other
inherited/environmental factors
2A Reduced VWF-dependent VWF:RCo disproportionately Missense 2A (IIA & IIE) AD Enhanced ADAMTS13
platelet adhesion plus selective reduced to VWF:Ag Other in-frame Fully penetrant susceptibility
HMW multimer deficiency alterations 2A (IID) mostly AD Disrupted dimerisation/
Null alleles 2A (IIC) AR multimerisation
2B Increased affinity for platelet GpIb Enhanced RIPA sensitivity Missense AD Spontaneous GP1b binding
Variable VWF levels Other in-frame Fully penetrant Enhanced ADAMTS13 susceptibility
Platelet count often reduced alterations
2M Decreased VWF-dependent platelet VWF:RCo or VWF: Missense AD Reduced GP1b or collagen binding
adhesion without CB very disproportionately Other in-frame Fully penetrant
selective HMW multimer deficiency reduced to VWF:Ag alterations
2N Markedly decreased VWF:RCo & VWF: Missense AR Reduced FVIII binding
FVIII binding affinity Ag normal/reduced Null alleles
FVIII:C <30IU/dL
3 Virtually complete VWF VWF:RCo & VWF: Null alleles AR Lack of secretion
deficiency Ag <5IU/dL Missense
FVIII:C <10IU/dL
* levels;† AD: Autosomal dominant; AR: autosomal recessive.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 45 |
14th Congress of the European Hematology Association

to mild, but their severity is not closely linked to VWF than normal. Determination of the steady-state ratio of
level. Three recent systematic studies of type 1 VWD, VWF propeptide (VWFpp) to VWF:Ag or of dynamics of
which together have analysed about 300 patients, pro- VWF response following desmopressin infusion21 pro-
vided insight into this VWD type.13-15 Sixty to seventy vide information on VWF clearance. Ultra-large VWF
percent of cases have a mutation identified. Candidate multimers can sometimes highlight individuals with
mutations have been reported throughout VWF, from clearance mutations, as ADAMTS13-VWF cleavage is
the 5’ untranslated region (UTR), (c.-2731) through to reduced by rapid VWF clearance from the plasma.22
exon 52 (p.C2804). Figure 1 shows mutation location. p.Y1584C, present in approximately 1% of normal
The majority are missense changes (70%),16 with 5’ Caucasian populations,23 but very enriched in type 1
UTR, splice (leading to either exon skipping or to a non- VWD (8-25% of IC are heterozygous for the variant,
expressed (null) allele), small deletion, nonsense and homozygotes are rare) has been demonstrated to have
small insertion/duplication mutations, respectively slightly enhanced ADAMTS13 cleavage,24 not leading to
comprise the remainder, in order of frequency of occur- multimer abnormality. The p.C1584 variant is cleared
rence. Pathogenicity of 5’ UTR variants has yet to be more rapidly than p.Y1584, but the increase in clearance
demonstrated. Large deletions have recently been rate is much less overt than for the above mutations.
recognised to contribute to the spectrum of mutations.17 ABO blood group O also results in slightly enhanced
Two of thirty-four British patients were heterozygous clearance of VWF from plasma, and co-inheritance of
for an exon 4-5 deletion, first identified in British type 3 both blood group O and p.C1584 leads to more rapid
VWD patients. Mutations vary from fully penetrant, clearance of VWF from plasma (up to 1.8 times higher
dominantly inherited variations, where all individuals than for group A p.Y1584 homozygotes).25 This results
who inherit a particular sequence variant are affected by in a greater likelihood of low VWF levels and diagnosis
disease, for example the “Vicenza” p.R1205H mutation, of type 1 VWD. In the MCMDM-1VWD study, 85% of
to incompletely penetrant alterations, such as family members heterozygous for p.Y1584C were
p.Y1584C, where an interaction with blood group O is classed as affected, but index cases had been selected
required for disease.16 due to their bleeding symptoms. This variant, therefore,
After the coding region of VWF plus 3kb of 5’ UTR shows incomplete penetrance, a long-recognised feature
had been analysed, a significant proportion (30-40%) of of type 1 VWD for which explanations are beginning to
type 1 VWD patients had no candidate mutations iden- be made.
tified.13-15 The majority of these cases had VWF levels
greater than 20 IU/dL. Bleeding in these patients was Type 2 von Willebrand disease
not significantly different from that in patients with a Twenty to thirty percent of all VWD patients can be
candidate VWF mutation, who generally had lower classified as having type 2 disease, where VWF function
VWF levels.14 It is, therefore, likely that other hemostat- is affected largely as a result of missense alterations.
ic defects also contribute to bleeding in patients diag-
nosed with “type 1 VWD.” Type 2A von Willebrand disease
Type 1 is not subdivided into different categories. Decreased VWF-dependent platelet adhesion and
However, different mutation mechanisms resulting in selective deficiency of HMW VWF multimers charac-
the disease are beginning to be identified. The most terise this VWD type.4 Levels of VWF:Ag and VWF:RCo
recognisable is enhanced clearance rate of VWF from are reduced, whereas FVIII may be normal (Table 1).
the plasma. Mutations in the D3 domain, notably those The deficiency of large multimers can result from
affecting p.C1130, p.C1149 and p.R1205,18,19 plus defects in multimer assembly4 or from enhanced sensi-
residues elsewhere in VWF (e.g., p.S2179F),20 have a tivity to ADAMTS13 cleavage.26 An earlier classification
plasma residence time that is up to ten times shorter based on multimer abnormalities can be useful to subdi-

Figure 1. von
Willebrand factor pro-
tein showing func-
tional domains and
exons encoding
them, binding sites
and location of muta-
tions in each VWD
type.

| 46 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

vide type 2A,4 and correlates to an extent with function- gene, VWFP on chromosome 22 invades and replaces a
al impairment. Previous IIA comprises two groups of short section of the gene sequence10,32 may show a dif-
mutations, located in and close to the A2 domain, ferent phenotype. Thrombocytopenia and loss of HMW
where they cluster around the p.Tyr1605_Met1606 multimers may not feature, and enhanced RIPA at low
ADAMTS13 cleavage site. Group I mutations impair concentration may be the only clue as to VWF involve-
multimer assembly and enhance proteolysis, whereas ment in their symptoms.33,34
group II mutations do not affect large multimer secre- A phenocopy of 2B VWD, platelet type VWD,35
tion but enhance proteolysis. However, mutation mech- results from a reciprocal defect in platelet Glycoprotein
anism cannot be predicted by location. A small number 1b. Mutations in exon 1 of the GP1BA gene encode mis-
of common missense mutations make up about 50% of sense or in-frame alterations leading to enhanced affini-
all VWFdb reports,11 with mutations affecting p.S1506 ty for VWF.36,37 The two disorders, which are both dom-
and p.R1597 being the most common. These mutations inantly inherited and fully penetrant, can be most read-
are generally dominantly inherited and fully penetrant. ily discriminated by genetic analysis, or alternatively by
Figure 1 shows mutation location. plasma-platelet mixing studies.
VWF must dimerise and then multimerise prior to
secretion for full hemostatic activity. Other type 2A Type 2M von Willebrand disease
mutations specifically interfere with this process. The This subtype is comprised of qualitative variants with
CK domain at the carboxyl terminal of VWF is involved decreased VWF-dependent platelet adhesion but with-
in the formation of “tail-to-tail” dimers through disul- out a selective deficiency of HMW multimers.
phide bond formation. Mutations at, or affecting these Mutations disrupt VWF binding to either platelets or
cysteines, result in reduced or failed dimerisation (IID). subendothelium.4 This VWD type can be difficult to dis-
Multimer profiles show a characteristic pattern with criminate from both type 1 and 2A VWD. Good multi-
intermediate sized bands truncated by a monomer.27 mer analysis and the VWF-collagen binding assay
Mutations are mostly dominantly inherited, but some (VWF:CB) can be helpful. Very reduced VWF:RCo com-
homozygous recessive cases have been reported.11,28 pared to VWF:Ag indicates defective binding to platelet
Mutations in the D3 domain result in a characteristic Gp1b, whereas a low VWF:CB to VWF:Ag ratio indi-
multimer pattern with a smeary appearance, lacking cates that binding to collagen is disrupted. Due to diffi-
obvious multimer satellite “triplet” bands. This IIE phe- culties in identifying this subtype, relatively few muta-
notype29 often results from mutations affecting cysteine tions are reported on the VWFdb, only p.V1279I and
residues, which may be involved in inter-molecular p.I1425F having been reported more than once. The
disulphide bonds between adjacent D3 domains. majority of mutations reduce Gp1b binding and are
Mutations in the D1 and D2 domains may also inter- located in exon 28 (p.1266_1467). A single mutation
fere with disulphide bonding between D3 domains, resulting in 2M VWD due to reduced collagen binding
catalysed by two CGLC disulphide isomerase sites has been reported.38 Mutations are dominantly inherited
(p.159_162 and 521_524), thus affecting multimer for- and fully penetrant.
mation. Recessively inherited homozygous/compound
heterozygous missense mutations in exons 12-16 result Type 2N von Willebrand disease
in a characteristic 2A (IIC) multimer pattern, devoid of FVIII level is decreased disproportionately to VWF:Ag
satellite “triplet” bands.29 in this VWD type and multimers are normal, with
minor exceptions (outlined below). It is often challeng-
Type 2B von Willebrand disease ing to discriminate the disorder from mild hemophilia A
Qualitative variants with increased affinity for in males, or from carriership for hemophilia A in
platelet GPIb are classified as type 2B. Increased platelet females. Unless the pedigree is sufficiently large, deter-
agglutination at low ristocetin concentrations (deter- mining clear X-linked or autosomal recessive inheri-
mined in the ristocetin induced platelet agglutination tance may not be possible. Diagnosis is ideally achieved
(RIPA) assay) identifies such patients. Most patients through measuring the ability of patient plasma VWF to
have a reduction in HMW multimers, and the multimer bind to FVIII using an immunoassay (VWF:FVIIIB).
profile can demonstrate proteolysis of the VWF sub- However, this test is not widely available and genetic
units, resulting from increased ADAMTS13 susceptibil- analysis of the VWF:FVIIIB region of VWF or of the F8
ity.26 Following secretion, VWF may spontaneously bind gene may be required to discriminate the disorders and
to platelets facilitating ADAMTS13 proteolysis. The facilitate relevant genetic counselling. Two mutations
characteristic platelet binding, seen in many 2B patients, are necessary for 2N VWD, patients may be homozy-
can result in thrombocytopenia, which may be exacer- gous for a missense change, compound heterozygous
bated by pregnancy, stress and use of desmopressin. for two different VWF:FVIIIB missense mutations or
Dominantly inherited missense mutations lie within or more commonly compound heterozygous for one
close to the A1 domain, with mutations affecting only VWF:FVIIIB mutation, with a mutation resulting in a
15 different amino acid residues having been reported lack of VWF expression on the second allele. Relative
on the VWFdb (p.1266_1461).11 Mutations generally lie levels of VWF:Ag and FVIII can give clues as to which
within the amino terminal of the A1 domain at the situation is more likely; VWF levels are lower where a
“base” of the globular structure, in contrast to 2M muta- null mutation is present. More than 80% of the muta-
tions, which cluster at the “top” of the domain.30 tions reported are located in exons 18-20 (D’) domain,
Patients with p.P1266L mutations (previously referred with p.R854Q, p.R816W and p.T791M being particular-
to as type IB New York/Malmo31), which result from ly common. p.R854Q is present at polymorphic fre-
gene conversion where a section of the VWF pseudo- quencies in the Caucasian population.39 A much smaller

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 47 |
14th Congress of the European Hematology Association

proportion of mutations are found in exons 17 plus 24- Annu Rev Biochem 1998;67:395-424.
4. Sadler JE, Budde U, Eikenboom JC, Favaloro EJ, Hill FG,
27 (p.1053_1225).11 They may indirectly affect Holmberg L et al. Update on the pathophysiology and classifi-
VWF:FVIIIB through altering the confirmation of the cation of von Willebrand disease: a report of the Sub-
FVIII binding site. A small proportion of mutations, committee on von Willebrand Factor. J Thromb Haemost
including those affecting cysteine residues also have an 2006;4:2103-14.
5. Budde U, Schneppenheim R, Plendl H, Dent J, Ruggeri ZM,
effect on VWF multimer profile. Examples include Zimmerman TS. Luminographic detection of von Willebrand
p.C788R and p.C1225G.40,41 A patient’s symptoms most- factor multimers in agarose gels and on nitrocellulose mem-
ly result from their reduced FVIII level (typically 10-30 branes. Thromb Haemost 1990;63:312-15.
6. Rodeghiero F, Castaman G, Tosetto A, Batlle J, Baudo F,
IU/dL), thus resembling mild hemophilia A. Individuals Cappelletti A et al. The discriminant power of bleeding histo-
heterozygous for a single VWF:FVIIIB missense muta- ry for the diagnosis of type 1 von Willebrand disease: an inter-
tion are rarely symptomatic. national, multicenter study. J Thromb Haemost 2005;3:2619-
26.
7. Tosetto A, Rodeghiero F, Castaman G, Goodeve A, Federici
Type 3 von Willebrand disease AB, Batlle J et al. A quantitative analysis of bleeding symptoms
This virtually complete deficiency of VWF, where in type 1 von Willebrand disease: results from a multicenter
European study (MCMDM-1 VWD). J Thromb Haemost
VWF:RCo and VWF:Ag are less than 5 IU/dL and 2006;4:766-73.
FVIII:C is less than 10 IU/dL, is recessively inherited. 8. Bowman M, Mundell G, Grabell J, Hopman WM, Rapson D,
When the FVIII:C level is particularly low, patients can Lillicrap D, James P. Generation and validation of the
experience joint bleeding, in addition to a range of Condensed MCMDM-1VWD Bleeding Questionnaire for von
Willebrand disease. J Thromb Haemost 2008;6:2062-6.
mucocutaneous symptoms. Most heterozygous rela- 9. Mancuso DJ, Tuley EA, Westfield LA, Lester-Mancuso TL, Le
tives are unaffected by bleeding. Disease prevalence is Beau MM, Sorace JM, Sadler JE. Human von Willebrand factor
greatly increased in the offspring of consanguineous gene and pseudogene: structural analysis and differentiation
by polymerase chain reaction. Biochemistry 1991;30:253-69.
partnerships, rising from about 0.5/million to 6/million 10. Chen JM, Cooper DN, Chuzhanova N, Ferec C, Patrinos GP.
in these circumstances.42 Mutations result in lack of Gene conversion: mechanisms, evolution and human disease.
VWF expression, either through affecting its production Nat Rev Genet 2007;8:762-75.
11. ISTH-VWF-SSC. International Society on Thrombosis and
(nonsense, small deletion, splice, large deletion and Haemostasis Scientific and Standardization Committee VWF
small insertion mutations are responsible, in decreasing Information Homepage www.vwf.group.shef.ac.uk
order of frequency) or by missense mutations affecting 12. Soteh MH, Peake IR, Marsden L, Anson J, Batlle J, Meyer D et
VWF secretion. Most of the latter affect dimerisation al. Mutational analysis of the von Willebrand factor gene in
type 1 von Willebrand disease using conformation sensitive
(CK domain) or the propeptide and VWF multimerisa- gel electrophoresis: a comparison of fluorescent and manual
tion (D1-D3 domains). Other mutations are located techniques. Haematologica 2007;92:550-3.
throughout VWF, from p.47_2804. There are no partic- 13. Cumming A, Grundy P, Keeney S, Lester W, Enayat S, Guilliatt
A et al. An investigation of the von Willebrand factor genotype
ularly common mutations, but p.P812fs, p.R1659X and in UK patients diagnosed to have type 1 von Willebrand dis-
p.R2535X have each been reported in several patients. ease. Thromb Haemost 2006;96:630-41.
Founder effect, where the same mutant allele causes 14. Goodeve A, Eikenboom J, Castaman G, Rodeghiero F, Federici
AB, Batlle J et al. Phenotype and genotype of a cohort of fam-
disease in a population, is evident for a small number of ilies historically diagnosed with type 1 von Willebrand disease
type 3 VWD mutations, and is likely to be under-recog- in the European study, Molecular and Clinical Markers for the
nized as haplotype of mutated alleles is not commonly Diagnosis and Management of Type 1 von Willebrand Disease
(MCMDM-1VWD). Blood 2007;109:112-21.
reported. A recently described large deletion of exons 1- 15. James PD, Notley C, Hegadorn C, Leggo J, Tuttle A, Tinlin S et
3 is responsible for about half of type 3 VWD cases in al. The mutational spectrum of type 1 von Willebrand disease:
Hungary,43 and a complete 253kb VWF deletion has Results from a Canadian cohort study. Blood 2007;109:145-54.
been identified in both German and Italian type 3 16. Collins PW, Cumming AM, Goodeve AC, Lillicrap D. Type 1
von Willebrand disease: application of emerging data to clini-
patients.44 Deletion of exons 4-5 contributes to both cal practice. Haemophilia 2008;14:685-96.
type 3 and type 1 VWD in England,17 and the p.P812fs 17. Sutherland MS, Cumming AM, Bolton-Maggs PHB, et al. A
mutation is found in type 3 VWD in Scandinavia and novel recurrent deletion mutation associated with type 1 and
type 3 von Willebrand disease. Br J Haematol 2008;141 Supp
northern Europe.45-47 1:A193.
18. Haberichter SL, Castaman G, Budde U, Peake I, Goodeve A,
Summary Rodeghiero F et al. Identification of type 1 von Willebrand dis-
ease patients with reduced von Willebrand factor survival by
The above disease types allow the majority of assay of the VWF propeptide in the European study: molecu-
patients to be categorised by their VWD phenotype and lar and clinical markers for the diagnosis and management of
this informs management and genetic counselling of type 1 VWD (MCMDM-1VWD). Blood 2008;111:4979-85.
19. Schooten CJ, Tjernberg P, Westein E, Terraube V, Castaman G,
families. However, co-inheritance of different types of Mourik JA et al. Cysteine-mutations in von Willebrand factor
mutations may lead to complex phenotypes that are associated with increased clearance. J Thromb Haemost 2005;
challenging to classify, and novel mutations that do not 3:2228-37.
appear to readily fit into the classification continue to be 20. Haberichter SL, Balistreri M, Christopherson P, Morateck P,
Gavazova S, Bellissimo DB et al. Assay of the von Willebrand
identified.48 Investigation of such instances enhances factor (VWF) propeptide to identify patients with type 1 von
understanding of the complex biology of VWF. Willebrand disease with decreased VWF survival. Blood 2006;
108:3344-51.
21. Castaman G, Lethagen S, Federici AB, Tosetto A, Goodeve A,
Budde U et al. Response to desmopressin is influenced by the
References genotype and phenotype in type 1 Von Willebrand Disease
(VWD): results from the European study MCMDM-1VWD.
1. Bloom AL. von Willebrand factor: clinical features of inherited Blood 2008;111:3531-9.
and acquired disorders. Mayo Clin Proc 1991;66:743-51. 22. Budde U, Federici AB, Goodeve A, Eikenboom J, Schneppen-
2. Mancuso DJ, Tuley EA, Westfield LA, Worrall NK, Shelton- heim R. Detailed von Willebrand factor multimer analysis in
Inloes BB, Sorace JM et al. Structure of the gene for human von patients with von Willebrand disease in the European study,
Willebrand factor. J Biol Chem 1989;264:19514-27. molecular and clinical markers for the diagnosis and manage-
3. Sadler JE. Biochemistry and genetics of von Willebrand factor. ment of type 1 von Willebrand disease (MCMDM-1VWD):

| 48 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

reply to a rebuttal. J Thromb Haemost 2008. ular-weight multimers. N Engl J Med 1982;306:326-33.
23. Davies JA, Collins PW, Hathaway LS, Bowen DJ. Effect of von 36. Othman M, Notley C, Lavender FL, White H, Byrne CD,
Willebrand factor Y/C1584 on in vivo protein level and func- Lillicrap D et al. Identification and functional characterization
tion and interaction with ABO blood group. Blood 2007; of a novel 27-bp deletion in the macroglycopeptide-coding
109:2840-6. region of the GPIBA gene resulting in platelet-type von
24. Bowen DJ, Collins PW. An amino acid polymorphism in von Willebrand disease. Blood 2005;105:4330-6.
Willebrand factor correlates with increased susceptibility to 37. PT-VWD Ro. Registry on platelet type von Willebrand disease
proteolysis by ADAMTS13. Blood 2004;103:941-7. www.pt-vwd.org
25. Davies JA, Collins PW, Hathaway LS, Bowen DJ. von 38. Ribba AS, Loisel I, Lavergne JM, Juhan-Vague I, Obert B,
Willebrand factor: evidence for variable clearance in vivo Cherel G et al. Ser968Thr mutation within the A3 domain of
according to Y/C1584 phenotype and ABO blood group. J von Willebrand factor (VWF) in two related patients leads to a
Thromb Haemost 2008;6:97-103. defective binding of VWF to collagen. Thromb Haemost
26. Rayes J, Hommais A, Legendre P, Tout H, Veyradier A, Obert 2001;86:848-54.
B et al. Effect of von Willebrand disease type 2B and type 2M 39. Eikenboom JC, Castaman G, Vos HL, Bertina RM, Rodeghiero
mutations on the susceptibility of von Willebrand factor to
ADAMTS-13. J Thromb Haemost 2007;5:321-8. F. Characterization of the genetic defects in recessive type 1
27. Enayat MS, Guilliatt AM, Surdhar GK, Jenkins PV, Pasi KJ, Toh and type 3 von Willebrand disease patients of Italian origin.
CH et al. Aberrant dimerization of von Willebrand factor as Thromb Haemost 1998;79:709-17.
the result of mutations in the carboxy-terminal region: identi- 40. Allen S, Abuzenadah AM, Blagg JL, Hinks J, Nesbitt IM,
fication of 3 mutations in members of 3 different families with Goodeve AC et al. Two novel type 2N von Willebrand dis-
type 2A (phenotype IID) von Willebrand disease. Blood 2001; ease-causing mutations that result in defective factor VIII
98:674-80. binding, multimerization, and secretion of von Willebrand fac-
28. Tjernberg P, Vos HL, Spaargaren-van Riel CC, Luken BM, tor. Blood 2000;95:2000-7.
Voorberg J, Bertina RM et al. Differential effects of the loss of 41. Jorieux S, Gaucher C, Goudemand J, Mazurier C. A novel
intrachain- versus interchain-disulfide bonds in the cystine- mutation in the D3 domain of von Willebrand factor marked-
knot domain of von Willebrand factor on the clinical pheno- ly decreases its ability to bind factor VIII and affects its multi-
type of von Willebrand disease. Thromb Haemost 2006;96: merization. Blood 1998;92:4663-70.
717-24. 42. Rodeghiero F. von Willebrand disease: still an intriguing disor-
29. Schneppenheim R, Budde U. Phenotypic and genotypic diag- der in the era of molecular medicine. Haemophilia 2002;8:292-
nosis of von Willebrand disease: a 2004 update. Semin 300.
Hematol 2005;42:15-28. 43. Mohl A, Marschalek R, Masszi T, Nagy E, Obser T, Oyen F et
30. Hillery CA, Mancuso DJ, Evan Sadler J, Ponder JW, Jozwiak al. An Alu-mediated novel large deletion is the most frequent
MA, Christopherson PA et al. Type 2M von Willebrand dis- cause of type 3 von Willebrand disease in Hungary. J Thromb
ease: F606I and I662F mutations in the glycoprotein Ib binding Haemost 2008;6:1729-35.
domain selectively impair ristocetin- but not botrocetin-medi- 44. Schneppenheim R, Castaman G, Federici AB, Kreuz W,
ated binding of von Willebrand factor to platelets. Blood Marschalek R, Oldenburg J et al. A common 253-kb deletion
1998;91:1572-81. involving VWF and TMEM16B in German and Italian patients
31. Weiss HJ, Sussman, II. A new von Willebrand variant (type I, with severe von Willebrand disease type 3. J Thromb Haemost
New York): increased ristocetin-induced platelet aggregation 2007;5:722-8.
and plasma von Willebrand factor containing the full range of 45. Zhang ZP, Falk G, Blomback M, Egberg N, Anvret M. A single
multimers. Blood 1986;68:149-56. cytosine deletion in exon 18 of the von Willebrand factor gene
32. Gupta PK, Adamtziki E, Budde U, Jaiprakash M, Kumar H,
Harbeck-Seu A et al. Gene conversions are a common cause of is the most common mutation in Swedish vWD type III
von Willebrand disease. Br J Haematol 2005;130:752-8. patients. Hum Mol Genet 1992;1:767-8.
33. Holmberg L, Dent JA, Schneppenheim R, Budde U, Ware J, 46. Zhang ZP, Blomback M, Nyman D, Anvret M. Mutations of
Ruggeri ZM. von Willebrand factor mutation enhancing inter- von Willebrand factor gene in families with von Willebrand
action with platelets in patients with normal multimeric struc- disease in the Aland Islands. Proc Natl Acad Sci USA 1993;90:
ture. J Clin Invest 1993;91:2169-77. 7937-40.
34. Baronciani L, Federici AB, Castaman G, Punzo M, Mannucci 47. Schneppenheim R, Krey S, Bergmann F, Bock D, Budde U,
PM. Prevalence of type 2b 'Malmo/New York' von Willebrand Lange M et al. Genetic heterogeneity of severe von Willebrand
disease in Italy: the role of von Willebrand factor gene conver- disease type III in the German population. Hum Genet
sion. J Thromb Haemost 2008;6:887-90. 1994;94:640-52.
35. Weiss HJ, Meyer D, Rabinowitz R, Pietu G, Girma JP, Vicic WJ 48. Lanke E, Kristoffersson AC, Philips M, Holmberg L, Lethagen
et al. Pseudo-von Willebrand's disease. An intrinsic platelet S. Characterization of a novel mutation in the von Willebrand
defect with aggregation by unmodified human factor VIII/von factor propeptide in a distinct subtype of recessive von
Willebrand factor and enhanced adsorption of its high-molec- Willebrand disease. Thromb Haemost 2008;100:211-6.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 49 |
Bleeding disorders

Treatment options in inherited von Willebrand disease

A.B. Federici A B S T R A C T

von Willebrand disease (VWD) is the most common inherited bleeding disorder and is due to quan-
Angelo Bianchi Bonomi Haemophilia titative or qualitative defects of the von Willebrand factor (VWF). VWD is inherited by autosomal dom-
and Thrombosis Centre, Department
of Medicine and Medical inant or recessive patterns, but women with mild forms are symptomatic. There are six types of VWD
Specialities, University of Milan, with peculiar phenotype and genotype: VWD1, VWD2A, VWD2B, VWD2M, VWD2N and VWD3. The ris-
Milan, Italy tocetin cofactor activity (VWF:RCo) is the most useful test for diagnosis because it can mimic the
interactions between the VWF and platelets. Two therapeutic approaches are available to manage
patients with VWD: the release of endogenous VWF from endothelial compartments induced by
desmopressin (DDAVP); and the transfusion of exogenous VWF contained in VWF/FVIII plasma-derived
Hematology Education:
the education program for the concentrates. DDAVP is the treatment of choice for VWD1 because it can induce the release of nor-
annual congress of the European mal VWF from endothelial cells: it can also be useful in mild-moderate VWD2A, VWD2M and VWD2N.
Hematology Association In VWD3, and in severe forms of VWD1 and VWD2, DDAVP is not effective, and plasma-derived
VWF/FVIII concentrates should be used in bleedings, surgery and secondary-long term prophylaxis.
2009;3:50-54 Only the VWF/FVIII concentrates that have been evaluated in pharmacokinetic (PK) trials, as well as in
large retrospective or prospective efficacy studies, must be recommended to treat VWD patients.

Definition, epidemiology and classification Treatment and prevention of bleeding


von Willebrand disease (VWD) is the The goal of treatment is to correct the dual
most common inherited bleeding disorder defects of hemostasis, that is, abnormal
and is due to quantitative or qualitative platelet adhesion due to low or defective
defects of the von Willebrand factor (VWF). VWF, and abnormal intrinsic coagulation
VWD is inherited by autosomal dominant due to low FVIII.7 Two main therapeutic
or recessive patterns, but women with mild approaches are available: (i) desmopressin
forms are more symptomatic.1 In popula- (DDAVP) that releases endogenous VWF
tion-based studies, the prevalence of VWD from endothelial cells; (ii) exogenous VWF
is very high (0.81%),2 but the clinical rele- contained in VWF/FVIII concentrates. Table 1
vance of many of these cases is uncertain. If summarizes the choice of these two
we consider only patients referred for clini- approaches according to VWD types.
cal manifestations of bleeding, the actual
prevalence is 66-100 cases per million of the Desmopressin: definitions of biological
general population. response versus clinical efficacy
The most updated classification of VWD Desmopressin
has proposed six different types: VWD1, Desmopressin (1-deamino-8-D-arginine
VWD3, VWD2A, VWD2B, VWD2M, vasopressin, DDAVP) is a synthetic analogue
VWD2N.3 In the past, VWD1 was reported of vasopressin that it is relatively inexpensive
to be the most frequent form, accounting and carries no risk of transmitting blood-
for approximately 70% of cases. In a recent borne infectious agents. DDAVP, infused
study based on the reappraisal of VWD intravenously at a dose of 0.3 µg/kg diluted
diagnoses after 10 years (1998-2008), in 50 mL saline over 30 minutes, usually
VWD1 cases were only 671/1234 (55%).4 increases plasma VWF and FVIII three to five
This is because many cases previously diag- times above baseline levels within 30 to 60
nosed as VWD1 were re-diagnosed as minutes and, in general, high VWF and FVIII
VWD2 due to discrepant VWF measure- levels last for 6-8 hours.8 The drug is also
ments [ratio of ristocetin cofactor activity available in concentrated forms for subcuta-
(VWF:RCo) to VWF:Ag <0.6)], as previous- neous and intranasal administration, which
ly recommended.5 Age and VWD type dis- can be convenient for home treatment.9 At
tribution of the 1,234 patients enrolled in the time of diagnosis, a test dose of DDAVP
the Italian registry on VWD are shown in is recommended in VWD patients to estab-
Figure 1 A-B. The presence of qualitative lish the individual patterns of biological
defects of VWF in previously diagnosed response, and to predict clinical efficacy dur-
VWD1 has also been reported in 154 fami- ing bleeding, since the responses in a given
lies evaluated prospectively by a European patient are consistent on different occasions.5
Study entitled Molecular and Clinical Ideally, this infusion trial should be always
Markers for Diagnosis and Management of organized in non bleeding patients to avoid
VWD1.6 any interference of VWF consumption due to

| 50 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

bleeding or surgery. For this infusion trial, DDAVP must approach as for VWF/FVIII concentrates, and according
be administered at 0.3 µg/kg body weight, either by sub- to the following criteria: hemostasis clinically not differ-
cutaneous or intravenous route (in this case the com- ent from normal (excellent); mildly abnormal hemostasis,
pound is diluted in 50-100 mL of isotonic saline and partial or delayed control of spontaneous bleeding or
infused over 30 minutes). The time of sampling is sched- slight transient oozing from surgical wounds (good);
uled by taking the time of subcutaneous injection, or the moderately abnormal hemostasis, that is, bleeding not
beginning of intravenous infusion, as time 0. Plasma fully controlled but no need of additional therapy (mod-
samples should be obtained prior to infusion and at 60, erate); and no improvement at all with continuation of
120 and 240 minutes after administration of DDAVP bleeding and need of additional or alternative therapies
from at least one affected member of each family, prefer- (poor).12 The list and description of the published
ably the index case. In pediatric patients, excessive with- prospective or retrospective studies on biological
drawal of blood should be avoided and the evaluation response and clinical efficacy of DDAVP in inherited
should be limited at baseline and 240 minutes. The pro- VWD have been recently reported.13 Despite the wide-
tocol of the DDAVP infusion test with laboratory param- spread use of DDAVP in VWD, there are only a few
eters to assess the biological response has been described prospective clinical trials in large number of cases on
previously10,11 and is summarized in Table 2. DDAVP efficacy and safety aimed at determining bene-
In clinical practice, DDAVP infusions can be repeated fits and limits of this therapeutic approach. For these
every 12-24 hours, depending on the type and severity reasons, an investigator driven observational prospec-
of the bleeding episode. The efficacy of DDAVP in tive study on clinical efficacy of DDAVP in 200 patients
achieving hemostasis should be assessed using the same with VWD1 and VWD2 has been organized recently:

Table 1. Treatment of different types of von Willebrand dis-


ease.

Treatment of choice Alternative Adjunctive therapy


VWD1 Desmopressin Antifibrinolytics, estrogens
VWD2A VWF/FVIII concentrates Desmopressin for mild bleeding
VWD2B VWF/FVIII concentrates
VWD2M Desmopressin VWF/FVIII concentrates
VWD2N* Desmopressin VWF/FVIII concentrates
VWD3 VWF/FVIII concentrates Desmopressin, platelet concentrates
VWD3 with Recombinant factor VIII Recombinant activated factor VII
alloantibodies
* Not all VWD2N patients can respond to Desmopressin

Table 2. Recommendations for the infusion test with DDAVP.

Infusion test with desmopressin

Infusion protocol Administer in 30 minutes 0.3 µg/kg of


Desmopressin in 50 mL of saline.
The same dosage can also be
administered subcutaneously
Clinical and laboratory parameters VWF and FVIII activities must be measured
before and 0.5, 1, 2 and 4 hours after
Figure 1. Age distribution of the 1,234 patients enrolled in the starting Desmopressin infusion;
Italian Registry on VWD. (A) Each decade reports the frequen- check platelet count before and at least 2
cy of VWD1, VWD2 and VWD3. (B) VWD type distribution of
the same population, as classified in 2008 according to the hours after infusion.
recommendations reported in references (2). Note that the Patients with VWD2B should be excluded
percentage of VWD1 is lower than that found ten years before
by the Italian Association of Hemophilia Centers. The reason Definition of responsiveness VWD patients should be considered
of this lower percentage is related to the demonstration of responsive to Desmopressin if after 2
abnormal ratio (<0.6) between VWF:RCo and VWF:Ag. All the hours they show increases of baseline
patients were characterized by decreased RIPA (risto- levels of VWF:RCo and FVIII by at least
cetin>1.6 mg/ml) and by only partial loss of high molecular
weight multimers in plasma (VWD2M); however, mutations three times, with levels of at least 30 U/dL 10
could be identified only in 60% of cases. For this reason we
cannot exclude in this group of patients some rare cases of An updated definition of biological response for VWD1 has been proposed:
VWD2A (subgroup IIC, IIE, IID, IIF) and we have identified these Complete responders: Both VWF:RCo and FVIII:C ≥50 IU/dL after DDAVP;
patients as VWD2M/2A. Partial responders: VWF:RCo or FVIII:C <50 IU/dL but increased at least 3-fold;
Non responders: None of the above.11

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 51 |
14th Congress of the European Hematology Association

the effectiveness and safety of DDAVP will be evaluat- of factors by stabilizing the already formed fibrin clot. A
ed prospectively for 24 months during bleeding typical example is dental surgey, in which these drugs
episodes and minor or major surgeries in VWD patients can also be used locally as mouthwashes. We recom-
who were exposed to an infusion trial at enrollment. mend the use of AAA together with DDAVP for the pre-
Side effects of DDAVP are usually mild tachycardia, viously mentioned reasons. Even though DDAVP
headache and flushing. These are attributed to the vaso- induces a short-term increase of t-PA, there is no evi-
motor effects of the drug and can often be attenuated by dence that this effect affects hemostasis in treated
slowing the rate of infusion. Hyponatremia and volume patients.7
overload due to the anti-diuretic effects of DDAVP are
relatively rare and usually occur in young children.14 von Willebrand factor/FVIII concentrates
Though no thrombotic episodes have been reported in VWF/FVIII concentrates are indicated in VWD3 and in
VWD patients treated with DDAVP, this drug should be VWD2B because DDAVP can induce transient thrombo-
used with caution in elderly patients with atherosclerot- cytopenia, and in all VWD1 or VWD2 patients who are
ic disease and hypertension, as reported in patients with not responsive to DDAVP or who may have contraindi-
mild hemophilia A.15 In the past, DDAVP was used to cations to its use (Table 1). Minimal requirements for
manage bleeding in VWD2B patients. Even though plasma-derived VWF/FVIII concentrates in VWD manage-
there are no reported cases of thrombosis, DDAVP ment are: (i) they must contain biologically active VWF
should be avoided in VWD2B because of the transient that could correct the primary hemostasis defect and
thrombocytopenia due to the enhanced interaction of stabilize the endogenous FVIII molecule (the latter
increased levels of VWF2B with platelet glycoprotein Ib objective can be achieved independently of the content
(GpIb). More importantly, we cannot exclude in princi- in exogenous FVIII); (ii) they should be treated by viru-
ple that such enhanced VWF2B-GpIb interactions caus- cidal methods; (iii) before clinical use, they should be
ing circulating platelet aggregates might predispose rel- tested for pharmacokinetics (PK) and efficacy in retro-
atively old patients with VWD2B to thrombosis.13 spective or prospective clinical trials in relatively large
numbers of VWD patients.7 Among the many
Adjuvant treatments VWF/FVIII concentrates available in the market, only a
Antifibrinolytic amino acids (AAA) are synthetic drugs few meet these requirements (Table 3). VWF/FVIII con-
that interfere with the lysis of newly formed clots by centrates can be given to stop bleeding episodes when
saturating the binding sites on plasminogen, thereby they occur (treatment on demand), to prevent bleeding
preventing its attachment to fibrin and making plas- during surgery (prophylaxis for surgery), or to prevent
minogen unavailable within the forming clot. Epsilon recurrent bleeding at specific sites (secondary long-term
aminocaproic acid (50 mg/kg, four times a day) and prophylaxis). The PK and clinical efficacy results of the
tranexamic acid (15-25 mg/kg, three times a day) are the first prospective study in VWD were published in
most frequently used antifibrinolytic amino acids. Both 2002.16 This study included 53 patients receiving treat-
can be administered orally, intravenously or topically, ment with a doubly virus-inactivated VWF/FVIII concen-
and are useful alone or as adjuncts in the management trate (Alphanate®) for 87 bleeding episodes and in 39
of oral cavity bleeding, epistaxis, gastrointestinal bleed- patients receiving treatment for 71 surgical or invasive
ing and menorrhagia. They should be avoided in the diagnostic procedures. A good clinical response with
management of urinary tract bleeding. AAA are given as this VWF/FVIII concentrate was observed in 86% of the
adjuvants, because they help to reduce the total amount spontaneous bleeding episodes, and in 71% of surgical

Table 3. Plasma-derived concentrates containing von Willebrand factor.

A. Concentrates with published activity in von Willebrand disease subjects.

Concentrate Purification procedures Virucidal Rx VWF:RCo/Ag VWFRCo/FVIII Available in Manufacturer

Alphanate Heparin ligand CT SD; Dry heat 0.6 1.2 GE, IT, UK, US Grifols (US)
Biostate Precipitation/Heparin ligand CT SD:Dry heat 0.8 2.0 AU, ASIA CslBehring
Fanhdi Precipitation, heparin ligand CT SD; Dry heat 0.6 1.6 SP-IT Grifols (Sp)
Haemate-P† Polyelectrolyte precipitations Pasteurization 0.8 2.5 ASIA, EUR, US CslBehring
Wilate Affinity CT, size exclusion SD; Dry heat 0.7 0.8 GE Octapharma
Wilfactin Ion-exchange, affinity CTs SD; NF; Dry heat 0.7 60 FR LFB (Lille)
B. Concentrates with limited activity or no published studies in von Willebrand disease subjects.

Concentrate Purification procedures Virucidal Rx VWF:RCoAg* VWF:RCo/FVIII* Available in Manufacturer

Emoclot Ion-exchange CT SD; Dry heat 0.5 1.2 BR, IT Kedrion


Immunate Ion-exchange CT SD; Vapor heat 0.2 0.2 EUR Baxter
Koate DVI Precipitations, size exclusion SD; Dry heat 0.5 1.2 US Talecris
8Y Heparin/Glycine precipitations Dry heat 0.3 0.8 UK BioProducts
CT: chromatography; SD: solvent-detergent (t-N-butyl-PO4 with polysorbate, Tween, or otoxynol, Triton); nf: nanofiltration; na: not applicable (multiple countries).
*Ratio of ristocetin cofactor activity (VWF:RCo) to VWF:Ag or FVIII activity expressed as IU/mL (or % of a normal pool). †Humate P in US.

| 52 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

or invasive procedures.16 Two retrospective and one view of the very short half-life of FVIII without its VWF
prospective study have also been performed using carrier, recombinant FVIII had to be administered by
Fandhi®, a concentrate manufactured using a process continuous intravenous infusion at very large doses, to
very similar to that of Alphanate®17,18 (A. Federici et al., keep FVIII levels above 50 U/dL for 10 days after sur-
unpublished data, 2009). gery.32 Another possible therapeutic approach is the
Haemate-P®/Humate-P®, an intermediate-purity recombinant activated factor VII (rFVIIa) that can be
VWF/FVIII concentrate, has been widely used in VWD. used in VWD with allo-antibodies according to the
This product was introduced into clinical practice in same dosage and regimens as for hemophilia A patients
Europe (Haemate-P®) in 1984 and in the United States with inhibitors.33,34
(Humate-P®) in 1999. The first PK study of Haemate-P®,
published in 1998, was a single-centre evaluation Secondary long-term prophylaxis
involving six VWD3 patients.20 Results of a large retro- Patients with severe forms of VWD may have fre-
spective study organized by the Canadian Hemophilia quent hemarthroses, especially when FVIII levels are
Centers were published in 2002.21 Other published stud- below 10 U/dL, so that some of them develop target
ies include two retrospective analyses of Haemate joints similar to patients with moderate hemophilia A.
P®/Humate-P® efficacy and safety in Italian VWD Some patients have recurrent gastrointestinal (GI)
patients,12,22 as well as two prospective, multicenter, bleeding, often without lesions in the GI tract and need
open-label, non-randomised studies conducted in the treatment every day or every other day. Finally, there
USA on Haemate-P®/Humate-P® used in urgent bleeding are children who frequently have epistaxis and are
and urgent surgical events.23,24 The results of another severe enough to cause anemia. In these bleeders, the
prospective study in elective surgery with optimal therapy may be regular prophylaxis with VWF
HaemateP®/Humate-P® with dosing based on PK have concentrates rather than on demand treatment during
been recently published.25 bleeding episodes. The largest experience on secondary
The use of Wilate® in VWD management has been prophylaxis in VWD has been collected in Sweden in 35
reported in Germany since 200526 and the results of effi- patients with severe forms of VWD.35 Secondary pro-
cacy and safety in acute bleeding episodes, in surgical phylaxis was also implemented in a cohort of Italian
interventions and in secondary long-term prophylaxis patients with VWD.36 These two retrospective studies
will be published within a few months. Data have been suggest that cost-effectiveness of these prophylaxis reg-
also reported on the PK and clinical efficacy of Biostate®, imens versus on demand therapy should be further eval-
a VWF/FVIII concentrate available in Australia and uated in larger prospective studies.
Asia.27,28 A peculiar plasma-derived VWF concentrate
with low FVIII levels was introduced in France in 1992 Future perspectives
and the first PK study in VWD3 was published in 1996.26 Overall, the treatments currently available for VWD
An improved version of this concentrate (Wilfactin®), are quite satisfactory. For patients unresponsive to
which is almost devoid of FVIII, was evaluated in two DDAVP, VWF/FVIII concentrates are the only form of
large French and European studies and data on PK have available treatment. The fact that they are fractionated
been already published.30 Results in VWD3 show no from plasma is of concern for some, even if more than
major differences in VWF:RCo and VWF:Ag for the con- one viral inactivation method is used for most concen-
centrates that did or did not contain FVIII; the only dif- trates in the manufacturing process. Haemate P/Humate
ference was an approximate 6-hour delay in FVIII P is the only concentrate that uses only one viral inacti-
increase with the concentrate devoid of FVIII. vation method (pasteurization), but the safety record of
Therefore, administration of exogenous FVIII is recom- this product is impeccable. This favorable situation
mended in VWD3 cases with acute life-threatening notwithstanding, there are advanced plans to develop a
bleeding episodes or emergency surgeries.30 Clinical effi- therapeutic preparation of recombinant VWF. This
cacy results of the French and European studies have product containing only VWF will require the concomi-
been recently reported.31 tant administration of FVIII for the control of acute
On the whole, there is no evidence from retrospective bleeding episodes and for the prevention of excessive
or prospective clinical studies that the six VWF/FVIII bleeding at the time of major surgery. Attempts to cor-
concentrates (Alphanate®, Biostate®, Fandhi®, Haemate- rect VWD partially through gene replacement therapy
P®/Humate-P®, Wilate®, Wilfactin®) reported in Table 3 are in progress.37
differ with regards to efficacy, because no head-to-head
clinical study has been carried out. Therefore, all these The author would like to thank the work of Dr. Luigi
VWF/FVIII concentrates can be effective to manage or Flaminio Ghilardini, who prepared the figures reported in this
prevent bleeding in patients with VWD. manuscript.
Treatment of patients with alloantibodies to von Willebrand
factor References
For the rare patients with VWD3 who develop anti-
VWF allo-antibodies after multiple transfusions, the use 1. Federici AB, Mannucci PM. Management of inherited von
of VWF/FVIII concentrates is ineffective, but may even Willebrand disease in 2007. Ann Med 2007;39:346-58.
2. Rodeghiero F, Castaman G, Dini E. Epidemiological investiga-
cause post-infusion anaph.32 These reactions may be life tion of the prevalece of von Willebrand’s disease. Blood 1987;
threatening. To overcome this drawback, a patient 69:454-59.
undergoing emergency abdominal surgery was treated 3. Sadler JE, Budde U, Eikenboom JCJ, Favaloro EJ, Hill FG,
Holmberg L et al. Update on the pathophysiology and classifi-
with recombinant FVIII, because this product contain- cation of von Willebrand disease. A report of the subcommittee
ing no VWF, could not cause anaphylactic reactions. In on von Willebrand factor. J Thromb Haemost 2006;4:2103-14.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 53 |
14th Congress of the European Hematology Association

4. Federici AB, Bucciarelli P, Castaman G, et al. Prevalence and Factor concentrate, Haemate-P in preventing bleeding during
Determinants of bleeding in different types of von Willebrand surgery or invasive procedures in patients with von Wille-
disease: results of the first prospective multicenter study on brand disease. Haematologica 2003;88:1279-83.
814 Italian patients. Blood 2007, Abstract 713. 23. Gill JC, Ewenstein BM, Thompson AR, Mueller-Velten G,
5. Federici AB, Castaman G, Mannucci PM. Guidelines for the Schwartz BA. Successful treatment of urgent bleeding in von
diagnosis and management of VWD in Italy. Haemophilia Willebrand Disease with factor VIII/von Willebrand Factor
2002;8:607-21. concentrate (Humate-P): use of the ristocetin cofactor assay
6. Goodeve A, Eikenboom J, Castaman G, Rodeghiero F, Federici (VWF:RCo) to measure potency and to guide therapy. Haemo-
AB, Batlle J et al. Phenotype and genotyoe of a cohort of fam- philia 2003;9:688-95.
ilies historically diagnosed with type 1 von Willebrand disease 24. Thompson AR, Gill JC, Ewenstein BM, Mueller-Velten G,
in the European study, Molecular and Clinical Markers for the Schwartz BA. Successful treatment for patients with von
Diagnosis and Management of Type 1 von Willebrand disease. Willebrand Disease undergoing urgent surgery using factor
Blood 2007;109:112-21. VIII/von Willebrand Factor concentrate (Humate-P). Haemo-
7. Mannucci PM. Treatment of von Willebrand disease. N Engl J philia 2004;10:42-51.
Med 2004;351:683-94. 25. Lethagen S, Kyrle PA, Castaman G, Haertel S, Mannucci PM et
8. Mannucci PM, Ruggeri ZM, Pareti FI, Capitanio A. A new al. von Willebrand factor/factor VIII concentrate (Haemate P)
pharmacological approach to the management of hemophilia dosing based on pharmacokinetics: a prospective multicenter
and von Willebrand disease. Lancet 1977;1:869-72. trial in elective surgery. J Thromb Haemost 2007;5:1420-30.
9. Rodeghiero F, Castaman G, Mannucci PM. Prospective multi-
center study on subcutaneous concentrated desmopressin for 26. Stadler M, Gruber G, Kannicht C, Biesert L, Radomski KU,
home treatment of patients with von Willebrand disease and Suhartono H et al. Characterisation of a novel high-purity,
mild or moderate hemophilia A. Thromb Haemost 1996;76: double virus inactivated von Willebrand factor and factor VIII
692-6. concentrate (Wilate®). Biologicals 2006;34:281-8.
10. Federici AB, Mazurier C, Berntorp E, Lee CA, Scharrer I, 27. Favaloro EJ, Llyod J, Rowell J, Baker R, Rickard K, Kershaw G
Goudemand J et al. Biologic response to desmopressin in et al. Comparison of the pharmacokinetics of two von
patients with severe type 1 and type 2 von Willebrand disease: Willebrand factor concentrates [Biostate and AHF (High
results of a multicenter European study. Blood 2004;103:2032- Purity)] in people with vpn Willebrand disorder. Thromb
8. Haemost 2007;97:922-30.
11. Castaman G, Lethagen S, Federici AB, Tosetto A, Goodeve A, 28. Shortt J, Dunkley S, Rickard K, Baker R, Street A et al. Efficacy
Budde U et al. Response to desmopressin is influenced by the and safety of a high purity, double virus inactivated factor
genotype and phenotype in type 1 von Willebrand disease VIII/von Willebrand factor concentrate (Biostate) in patients
(VWD): results from the European Study MCMDM-1VWD. with von Willebrand disorder requiring invasive or surgical
Blood 2008;11:3531-9. procedures. Haemophilia 2007;13:144-8.
12. Federici AB, Castaman G, Franchini M, Morfini M, Zanon E, 29. Menache D, Aronson DL, Darr F, Montgomery RR, Gill JC,
Coppola A et al, Clinical use of Haemate P in inherited von Kessler CM et al. Pharmacokinetics of von Willebrand factor
Willebrand's disease: a cohort study on 100 Italian patients. and factor VIIIC in patients with severe von Willebrand dis-
Haematologica 2007;92:944-51. ease (type 3 VWD): estimation of the rate of factor VIIIC syn-
13. Federici AB. The use of desmopressin in von Willebrand dis- thesis. Br J Haematol 1996;94:740-5.
ease: the experience of the first 30 years (1977-2007). Haemo- 30. Goudemand J, Scharrer I, Berntorp E, Lee CA, Borel-Derlon A,
philia 2008 Jan;14 Suppl 1:5-14. Stieltjes N et al. Pharmacokinetic studies on Wilfactin, a von
14. Smith TJ, Gill JC, Ambruso DR, Hathaway WE. Hypo- Willebrand factor concentrate with a low factor VIII content
natremia and seizures in young children given DDAVP. Am J treated with three virus-inactivation/removal methods. J
Hematol 1989;31:199-202. Thromb Haemost 2005;3:2219-27.
15. Bond L, Bevin D. Myocardial infarction in a patient with 31. Borel-Derlon A, Federici AB, Rouseel-Robert V, et al
hemophilia A treated with DDAVP. N Engl J Med 1988;318: Treatment of severe von Willebrand disease with a high-puri-
121. ty von Willebrand factor concentrate (Wilfactin): a prospective
16. Mannucci PM, Chediack J, Hanna W, Byrnes J, Ledford M, study of 50 patients. J Thromb Haemost 2007;5:1115-24.
Ewenstein BM et al. for the Alphanate Study Group. 32. Bergamaschini L, Mannucci PM., Federici AB, Coppola R,
Treatment of von Willebrand disease with a high-purity factor Guzzoni S, Agostoni A. Posttransfusion anaphylactic reaction
VIII/von Willebrand factor concentrate: a prospective, multi- in a patient with severe von Willebrand disease: role of com-
center study. Blood 2002;99:450-56. plement and alloantibodies to von Willebrand factor. J Lab
17. Federici AB, Baudo F, Caracciolo C, Mancuso G, Mazzucconi Clin Med 1995;125:348-55.
MG, Musso R et al. Clinical efficacy of highly purified, doubly 33. Ciavarella N, Schiavoni M, Valenzano E, Mangini F, Inchingolo
virus-inactivated factor VIII/von Willebrand factor concentrate F. Use of recombinant factor VIIa (Novoseven) in the treat-
(Fanhdi) in the treatment of von Willebrand disease: a retro-
spective clinical study. Haemophilia 2002;8:761-7. ment of two patients with type III von Willebrand disease and
18. Bello IF, Yuste VJ, Molina MQ, Navarro FH. Fanhdi, efficacy an inhibitor against von Willebrand factor. Haemostasis 1996;
and safety in von Willebrand’s disease: prospective interna- 26:150-4.
tional study results. Haemophilia 2007;13:25-32. 34. Boyer-Neumann C, Dreyfus M, Wolf M, Veyradier A, Meyer
19. Federici AB, Barillari G, Zanon E et al. Efficacy and safety of D. Multi-therapeutic approach to manage delivery in an
highly-purified, doubly virus-inactivated VWF/FVIII concen- alloimmunized patient with type 3 von Willebrand disease. J
trates in patients with von Willebrand disease: an Italian retro- Thromb Haemost 2003;1:190-2.
spective study on 120 cases. Haemophilia 2009; Submitted. 35. Berntorp E, Petrini P. Long-term prophylaxis in von Willebrand
20. Dobrkovska A, Krzensk U, Chediack JR. Pharmacokinetics, disease. Blood Coagul Fibrinolysis 2005;16:S23-S6.
efficacy and safety of Humate-P® in von Willebrand disease. 36. Federici AB, Gianniello F, Canciani MT, Mannucci PM.
Haemophilia 1998;4 Suppl 3:S33-9. Secondary long-term prophylaxis in severe patients with von
21. Lillicrap D, Poon M-C, Walkers S, Xie F, Schwartz BA. Efficacy Willebrand disease: an Italian cohort study. Blood 2005;106:
and safety of the factor VIII/von Willebrand Factor concen- 507a, abstract 1782.
trate, Haemate-P/Humate-P: Ristocetin cofactor unit dosing in 37. De Meyer SF, Vanhoorelbeke K, Chuah MK, Pareyn I, Gillijns
patients with von Willebrand disease. Thromb Haemost V, Hebbel RP et al. Phenotypic correction of von Willebrand
2002;87:224-30. disease type 3 blood-derived endothelial cells with lentiviral
22. Franchini M, Rossetti G, Tagliaferri A, Pattacini C, Pozzoli D, vectors expressing von Willebrand factor. Blood 2006;107:
Lippi Get al. Efficacy and safety of factor VIII/von Willebrand 4728-36.

| 54 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Chronic lymphocytic leukemia

Cellular origin of chronic lymphocytic leukemia

U. Klein A B S T R A C T

Chronic lymphocytic leukemia (CLL) has long been recognized as a morphologically homogeneous
Institute for Cancer Genetics and disease of mature B-lymphocytes. However, the unique phenotypic and genotypic features of this
Herbert Irving Comprehensive
Cancer Center, Columbia University, tumor precluded a conclusive assignment of the CLL tumor precursor cell to a specific normal B-cell.
New York, USA First, the immunophenotype of CLL cells characterized by the expression of the cell surface antigens
CD5, CD23, CD27 and low level of surface immunoglobulin (Ig) are different from that of any normal
B-cell.1,2 Second, CLL cases are heterogeneous at the genetic level, since the rearranged Ig variable
region (IgV) genes of CLL cases can be either somatically hypermutated or unmutated,3-6 suggesting
Hematology Education:
that the corresponding tumor precursor cells may originate from distinct B-cell developmental stages.
the education program for the
annual congress of the European Over the past several years, results obtained from global gene expression profiling (GEP) analyses, as
Hematology Association well as insights into the expressed antibody repertoire of CLL from large scale IgV gene sequencing
studies, have led to the accepted view that all CLL cases, independent of their IgV gene hypermuta-
2009;3:55-60 tion status, originate from the oncogenic transformation of a common cellular precursor, which
appears to be an antigen-experienced B-lymphocyte.

B-cell development and cellular origin of (IgV) somatic hypermutation, which intro-
B-cell malignancies duces mainly point mutations into the
A major interest in the study of B-cell rearranged antibody gene with the aim of
malignancies is the identification of the nor- generating high affinity antibodies. GC B-
mal cellular counterpart from which a partic- cells with improved antigen binding are pos-
ular tumor type originates. Knowing the itively selected and differentiate into memo-
developmental stage a B-cell malignancy ry B-cells or plasma cells. GC B-cells also
corresponds to has obvious implications for undergo Ig class switching from the expres-
understanding its pathogenesis. It may sion of IgM to IgG, A, or E by a somatic
become possible to dissociate the compo- DNA recombination mechanism. In the
nents of the malignancy that reflect the nor- human, memory B-cells can be distinguished
mal differentiation stage from which the from naïve B-cells by the expression of the
tumor originated from truly disease-associ- cell surface antigen CD27. While in the
ated genes, thus potentially providing mouse, CD5+ B-cells comprise a functionally
insights into the mechanism of transforma- well-defined B-cell subset (B1 cells) that pro-
tion. Equally important, identified tumor- duces natural antibodies, CD5+ B-cells in
specific gene products may be developed human adults do not seem to exert the same
into valuable new diagnostic markers or role.
therapeutic targets. The descendents of the GC response –
The investigation of the peripheral B-cell memory and plasma cells – have somatically
differentiation at the phenotypic and molec- mutated IgV genes and frequently express
ular levels led to the establishment of a B-cell class switched isotypes. When the various
developmental “scheme” that was useful in subtypes of mature B-cell lymphomas were
assigning the various lymphoma subtypes to analyzed for their level of IgV gene somatic
specific differentiation stages.7,8 The hall- hypermutation during the 1990s, it was
mark of antibody-mediated immunity is the intriguing to find that almost all lymphoma
generation of memory and plasma cells – the subtypes expressed hypermutated antibody
cell types that form the basis of the immuno- genes (Figure 1).8,11 Most notably, this catego-
logical memory against pathogens – in the ry included follicular lymphoma, Burkitt
germinal center (GC) microenvironment of lymphoma, and diffuse large B-cell lym-
the peripheral lymph nodes.9,10 The precur- phoma (DLBCL), and virtually every case
sors of the GC response are bone marrow- displayed IgV hypermutations. These find-
derived, antigen-inexperienced (naïve) B- ings suggested that the precursor cells of
cells, which are activated by foreign antigens most lymphoma subtypes represent either a
with the help of T-cells and other immune GC B-cell that actively undergoes somatic
cells. Those cells move to the GC, where hypermutation, or a cell that has passed
they begin to divide and proliferate. In the through the GC-microenvironment. From
course of the GC-reaction, the antibody this finding, the concept developed that the
genes of the B-cells are modified by the GC-reaction plays a critical role in the gene-
process of immunoglobulin variable region sis of B-cell lymphomas,8,11 and that an acci-

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 55 |
14th Congress of the European Hematology Association

dental “misfiring” of the somatic hypermutation and level of IgV somatic hypermutation18-20 or of the expres-
class switch processes may cause potentially transform- sion of CD38,21 a cell surface antigen that is differential-
ing genomic mutations.12,13 Only few B-cell tumor sub- ly expressed on CLL cases. With more sensitive analysis
types showed both somatically mutated, as well unmu- methods, one could extract subtle differences in the
tated IgV genes, namely mantle cell lymphoma, with genotypically defined subsets (as described in the fol-
approximately 80% of the cases expressing unmutated lowing paragraph). These observations strongly suggest
IgV genes,14,15 and chronic lymphocytic leukemia (CLL), that CLL represents a phenotypically homogeneous dis-
which expresses both somatically mutated and unmu- ease. From this, it follows that CLL is markedly differ-
tated IgV genes at about equal percentages.3 ent from a disease entity, such as DLBCL,22 which is
Evidently, in the light of peripheral B-cell develop- characterized by an extensive phenotypic heterogeneity
ment (Figure 1), the fact that CLL can express IgV and whose individual subtypes originate from develop-
mutated or unmutated IgV genes suggests that this mentally distinct B-cell subpopulations (e.g., GC-type
tumor subtype may have precursors of different devel- from GC B-cells and activated B-cell (ABC)-type DLBCL
opmental stages, namely the antigen-inexperienced from plasmablasts). Instead, the available evidence sug-
(naïve) and antigen-experienced (memory/marginal gest that all CLL originate from a common cellular pre-
zone) stages. Further, pointing towards a biological dif- cursor as a result of a common pathogenetic mecha-
ference between the two genetically defined subtypes nis.18,19
was the observation that IgV mutated CLL cases gener- By applying more stringent criteria in the biostatisti-
ally show a more benign clinical course than unmutated cal (supervised) analysis of the GEP data, it is possible to
CLL.16,17 However, as will be discussed in the following identify a small number of transcripts differentially
paragraphs, the evidence accumulating over the last sev- expressed between somatically mutated versus unmu-
eral years was inconsistent with the notion that the tated CLLs.18,19 Recently, a group of differentially
major CLL subtypes originate from different cellular expressed microRNAs was added to this list.23 This spe-
precursors. cific set of genes (molecular signature) allowed the clas-
sification of an independent panel of CLL cases into the
Chronic lymphocytic leukemia shows a homogeneous gene two subgroups, demonstrating the existence of a subtle
expression profile, suggesting a common cellular precursor but consistent phenotypic difference. One of those
GEP analyses revealed that all CLL displayed a com- analyses revealed that the IgV unmutated CLL subgroup
mon gene expression profile that is independent of the seems to express high mRNA levels of genes that are

Figure 1. Germinal center reaction and lymphomagenesis. (A) B-cell development and Ig remodeling processes. Antigen-
inexperienced naïve B-cells are generated throughout life in the bone marrow where they rearrange their antigen recep-
tor genes by somatic DNA recombination of the variable (V), diversity (D), and joining (J) gene segments. Upon antigen
activation, and with the help of T-cells and other immune cells, naïve B-cells are driven into the germinal center (GC)
response, where they modify their rearranged IgV genes at the centroblast stage by somatic hypermutation, and at the
centrocyte stage by Ig class switch recombination. Apotosis eliminates GC B-cells that have acquired unfavorable somat-
ic hypermutations; antibody mutants with a higher affinity against the antigen are selected into the post-GC repertoire
and differentiate into memory B-cells or, via a plasmablast stage, into plasma cells. In an antigen recall response, mem-
ory B-cells quickly differentiate into plasma cells. (B) B-cell malignancies and the level of somatic hyper-mutation in their
rearranged IgV genes. Virtually all cases of diffuse large B-cell, Burkitt, follicular, Hodgkin and AIDS-associated lym-
phomas and multiple myeloma carry somatically mutated IgV genes, indicating that the corresponding tumor precursor
cells represent GC B-cells or have passed through the GC. However, mantle cell lymphoma is mainly characterized by the
expression of unmutated IgV genes, and CLL express mutated and unmutated IgV genes at almost equal percentages.
Acute lymphoblastic leukemia is a tumor of developing B-cells and expresses unmutated IgV genes.

| 56 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

known to be activated as a result of in vitro B-cell recep- BCR with its specific antigen may have an impact on
tor (BCR)-mediated stimulation;19 therefore, suggesting the clinical prognosis of CLL. A recently proposed
that CLL belonging to this subgroup are subjected to model attempts to integrate the findings that all CLL
signaling through the BCR in vivo. However, the situa- seem to be subjected to BCR stimulation on the one
tion is more complex. While the GEP pattern that sepa- hand, and that CLL subtypes have different clinical
rates somatically mutated and unmutated CLL allows prognoses on the other. It suggests that some CLL may
the classification of CLL cases into either subgroup at a be continuously stimulated by antigen which renders
high confidence level, it is not absolute. In agreement them “activated” (likely representing bad-prognosis
with this notion, CLL cases with a VH3-21 gene CLLs), whereas others may have acquired an anergic,
rearrangement show a low overall poor survival irre- “quite” state.36 Taken together, the results of the large-
spective of the level of IgV hypermutation,24 and also scale IgV gene repertoire analyses provide strong evi-
display a GEP, which is distinct from that of VH3-21- dence that antigen plays a critical role in the develop-
negative cases.25 These observations suggest a crucial ment of CLL.
role for a common antigen in the pathogenesis of VH3-
21-expressing CLL cases. In conclusion, the small but The gene expression profiling of chronic lymphocytic
measurable expression differences between IgV mutat- leukemia is related to that of antigen-experienced B-cells
ed and unmutated, or VH3-21-expressing and non- The immunophenotype of the CLL tumor cells, CD5+
expressing CLL cases, seem to reflect the physiological CD23+ CD27+ and sIglow, does not resemble that of any
consequences of external stimuli, such as the interaction normal B-cell.1,2 In addition, CLL tumor cells have a
of BCR with specific antigen rather than a particular characteristic cell surface phenotype resembling that of
developmental stage of the tumor precursor cell. (antigen)-activated B-cells.37 Thus, although it was rec-
ognized early on that CLL is a tumor of mature B-cells,
Somatically mutated and unmutated chronic lymphocytic the particular immunophenotype of the tumor cells pro-
leukemia express stereotype antigen-receptors with vided no direct clues about the normal cellular counter-
evidence of antigen-experience part of this disease. Global GEP, which simultaneously
The realization that a subgroup of CLL carry somati- measures the expression of thousands of marker genes
cally mutated antibody genes was a first indication that of a cell population, turned out to be extremely useful in
a sizable fraction of CLL may have undergone the GC- dissecting the cellular origin of B-cell malignancies, and
reaction and express selected BCRs;4 thus, hinting at the indeed provided new information on the cellular origin
possibility that antigenic selection may play a role in the of CLL. As discussed in a previous section, the results of
development of CLL. Analyses of larger panels of cases the GEP analyses suggest that all CLL originate from a
then found that in both somatically mutated and unmu- common cellular precursor. A more specific GEP-based
tated cases, certain IgV gene family members are approach using supervised analysis methods for a com-
expressed much more frequently in CLL than would be parison of GEP data from CLL with those of the various
expected from their expression in the antibody reper- normal B-cell subsets provides further insights into the
toire of normal B-cells6 [for review, see (3)]. This obser- identity of the putative normal counterpart of CLL. The
vation suggested that CLL express restricted sets of anti- results suggest that all CLL are mostly related in their
gen receptors irrespective of the level of IgV gene hyper- GEP to that derived from a specific B-cell subset, the
mutation, and led to the conclusion that many CLL CD27+ B-cells,18 which comprises a heterogeneous sub-
seem to be derived from previously stimulated B-cells.6 set of antigen-experienced B-cells. This includes classi-
Further studies that compared the IgV gene repertoires cal memory B-cells, as well as marginal zone B-cells (see
expressed by CLL with those of elderly normal adults below). Noticeably, the CLL-profile did not show any
provided conclusive evidence that the CLL-characteris- similarity to those derived from CD5+ cord blood,
tic repertoire indeed seems to be the result of antigen- CD27– (naïve), or GC B-cells.18,19
selection and does not reflect the aging process.26,27 CD27+ B-cells comprise up to 40% of B-cells in the
Intriguingly, the general picture emerged that CLL cases peripheral blood (PB) of adults, and are found in the lym-
from unrelated patients can have almost identical phoid tissues, predominantly in the marginal zone and
BCRs.24,28-32 This BCR ‘stereotypy’ occurs at the levels of the tonsillar subepithelium, which both represent major
IgV gene usage, V-D-J junctional regions (heavy chain sites of antigen-entry. The majority of CD27+ B-cells (80-
CDR3s), and combination of particular heavy chain 90%) expresses somatically mutated IgV genes, while
CDR3s with light chain CDR3s [summarized in (33)]. CD27– B-cells are almost exclusively unmutated.38,39 In
Thus, CLL represents a tumor characterized by the vitro, CD27+ cells, in contrast to CD27– cells, and irrespec-
expression of stereotyped BCRs. This skewed BCR tive of their Ig isotype, respond quickly by differentiat-
repertoire seems to be a result of an antigen-mediated ing into antibody-secreting cells.40-42 CD27+ B-cells can be
selection. In support of this notion, it recently became class switched, IgM-only, or IgM+IgD+.38,43 IgM-express-
possible to identify specific antigens that are recognized ing CD27+ cells respond to T-independent antigens,42,44
by BCRs expressed by CLL; these antigens represent which is consistent with the notion that the marginal
autoantigens derived from “body” cells undergoing zone comprises B-cells that are rapidly activated to
apoptosis, and some of the recognized epitopes show secret Ig against invading microorganisms.45 Thus, a
similarities to microbial antigens.34,35 common feature of CD27+ memory/marginal zone B-
As eluded in the previous section, results from GEP cells is that they have been specifically generated to react
analysis suggest the existence of active BCR-stimulation against exogenous antigens. Together with the observa-
in the IgV gene unmutated CLL subset. Thus, the phys- tion that the BCRs of CLL show evidence of antigenic
iological consequences of the binding of a particular selection, the unique functional characteristics of CD27+

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 57 |
14th Congress of the European Hematology Association

B-cells provide additional evidence for a phenotypic rela- While the morphological and phenotypic characteristics
tion of the CLL precursor cell to this cell type. of HCL are clearly distinct from that of any other B-cell
malignancy, including CLL,49 HCL resembles CLL in that
Relation of chronic lymphocytic leukemia to other B-cell its GEP is most closely related to that of CD27+ B-cells.48
lymphomas Moreover, both tumor entities characteristically lack
The notion that CLL is a tumor of antigen-experi- chromosomal translocations.50,51 These findings suggest
enced B-cells related to memory/marginal zone B-cells that the tumor precursor cells of both CLL and HCL
suggests that the multistep process of leukemogenesis may be related to CD27+ B-cells. Thus, CLL and HCL
begins in these cells.18 Additional evidence for this may belong to a distinct group of B-cell malignancies
hypothesis stems from the particular pattern of cytoge- originating from the oncogenic transformation of anti-
netic abnormalities in CLL, which differs markedly gen-experienced B-cells.
from that found in most types of mature B-cell malig-
nancies. The major subtypes of B-cell lymphoma, that A model for the cellular derivation of chronic lymphocytic
is, Burkitt lymphoma, follicular lymphoma, and DLBCL leukemia
characteristically show recurrent balanced chromoso- This review discusses the evidence suggesting that
mal translocations, typically involving the translocation CLL is a tumor of a functionally characterized subgroup
of a proto-oncogene into the Ig loci (e.g., BCL2, c-MYC, of (CD5–) B-lymphocytes, namely the antigen-experi-
BCL6).12 These translocations are thought to be a direct enced B-cells, which in the human comprise a sizable
result of errors during the processes of VDJ recombina- fraction in the lymphoid organs and the PB (~40% in
tion, class switch recombination, or somatic hypermu- the PB). As suggested by the restricted BCR repertoire
tation.11,12 However, CLL does not typically show recur- expressed by CLL cases, among this huge reservoir of
rent chromosomal translocations, but is instead associ- antibody specificities only few cells would represent
ated with chromosomal deletions and amplifications, actual targets for oncogenic transformation. Since virtu-
namely trisomy of chromosome 12 (16%), and dele- ally all CD27+ cells are CD5–, in this scenario the CLL-
tions of chromosomal regions 17p (p53; 7%), 11q associated expression of CD5 would be a consequence
(ATM; 18%), and 13q14 (55%).46 This is similar to the of the activation-phenotype of the tumor cell, as has
pattern of genomic aberrations found in solid tumors previously been suggested.52 Alternative scenarios are
(although the occurrence of non-recurrent, imbalanced based on the assumption that the putative CLL precur-
chromosomal translocations in a subgroup of CLL has sor cell corresponds to a minor, yet uncharacterized, B-
been reported.47 The general lack of reciprocal balanced cell subset. Thus, a unique CD5+ B-cell population
translocations involving the Ig loci in CLL indicates that occurring at low frequency in the lymphoid organs has
the causative Ig loci-remodeling mechanisms are not been identified that shows some features of the CLL
active in the tumor precursor cell. Instead, genomic tumor cell and has hence been invoked as a putative
aberrations eventually leading to clonal expansion of a normal counterpart of CLL.53 Also, the causal relation-
CLL precursor cell may occur in the developmental ship between CLL and monoclonal B-cell lymphocyto-
stage following the completion of these processes, pre- sis, which represents a benign clonal proliferation of
sumably during the antigen-driven proliferation of the CD5+ B-cells in the PB of adults with phenotypic resem-
corresponding B-cell. Hierarchical cluster analyses of blance to the CLL tumor cells, as well as a CLL-associ-
GEP data representing the various types of B-cell lym- ated pattern of genetic aberrations, remains to be deter-
phomas revealed that CLL clusters separately from the mined.54-56 Figure 2 depicts a model for CLL pathogene-
lymphomas of putative GC origin.18,19,22 These biostatis- sis that reflects the possible cellular origin of the tumor
tical analyses somewhat unexpectedly revealed that cell precursor based on its phenotypic relationship to
CLL cluster along with hairy cell leukemia (HCL).48 CD27+ B-cells. A naïve B-cell is driven by antigen stim-

Figure 2. A model for the cellular der-


ivation of CLL. Naïve B-cells (antigen-
inexperienced) may be driven either
into a T cell-dependent or T-inde-
pendent immune response (top two
branches). A separate pathway (bot-
tom branch) has been suggested in
which somatically mutated IgM-
expressing B-cells are generated in
an antigen-independent fashion.
Upon completion of either the GC-
response or the T-independent
response (a low level of hypermuta-
tion in T-independent responses has
been observed in an animal model)61
the cells now represent antigen-
experienced B-cells. These B-cells
may be continuously activated by
chronic antigen stimulation and
acquire genetic alterations that may
eventually cause oncogenic transfor-
mation and development of CLL.

| 58 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

ulation either into a GC-response where it undergoes iology and malignancy. Nat Rev Immunol 2008;8:22-33.
14. Thorselius M, Walsh S, Eriksson I, Thunberg U, Johnson A,
IgV hypermutation, or into a T-independent response Backlin C, et al. Somatic hypermutation and V(H) gene usage
without undergoing hypermutation. Both developmen- in mantle cell lymphoma. Eur J Haematol 2002;68:217-24.
tal pathways are generally thought to give rise to mem- 15. Camacho FI, Algara P, Rodriguez A, Ruiz-Ballesteros E,
ory/marginal zone B-cells.9,57 While IgM-expressing, Mollejo M, Martinez N, et al. Molecular heterogeneity in MCL
defined by the use of specific VH genes and the frequency of
somatically mutated B-cells are considered to represent somatic mutations. Blood 2003;101:4042-6.
memory/marginal zone B-cells that have undergone the 16. Damle RN, Wasil T, Fais F, Ghiotto F, Valetto A, Allen SL, et al.
GC without isotype switching,38,39 a recently proposed Ig V gene mutation status and CD38 expression as novel prog-
nostic indicators in chronic lymphocytic leukemia. Blood
alternative scenario invokes generation of these cells in 1999;94:1840-7.
an antigen-independent fashion by an extrafollicular 17. Hamblin TJ, Davis Z, Gardiner A, Oscier DG, Stevenson FK.
developmental pathway58,59 (bottom part of Figure 2). Unmutated Ig V(H) genes are associated with a more aggres-
sive form of chronic lymphocytic leukemia. Blood 1999;94:
Regardless of the generation of those IgM+ cells, it is 1848-54.
thought that chronic antigen stimulation may keep the 18. Klein U, Tu Y, Stolovitzky GA, Mattioli M, Cattoretti G,
CLL precursor cell in a constant state of activation, and Husson H, et al. Gene expression profiling of B cell chronic
lymphocytic leukemia reveals a homogeneous phenotype
over time, mutations may accumulate in the genome of related to memory B cells. J Exp Med 2001;194:1625-38.
those cells that eventually lead to oncogenic transforma- 19. Rosenwald A, Alizadeh AA, Widhopf G, Simon R, Davis RE,
tion. Taken together, it emerges that memory/marginal Yu X, et al. Relation of gene expression phenotype to immu-
noglobulin mutation genotype in B cell chronic lymphocytic
zone B-cells display features consistent with a putative leukemia. J Exp Med 2001;194:1639-47.
CLL precursor cell. All CLL express CD27 on the cell 20. Jelinek DF, Tschumper RC, Stolovitzky GA, Iturria SJ, Tu Y,
surface,60 and the pattern of Ig isotype distribution Lepre J, et al. Identification of a global gene expression signa-
among CLL cases (class switched, IgM-only, IgM+IgD+) ture of B-chronic lymphocytic leukemia. Mol Cancer Res
2003;1:346-61.
resembles that of CD27+ cells. Moreover, the IgV genes 21. Dürig J, Nückel H, Hüttmann A, Kruse E, Hölter T, Halfmeyer
of both mutated and unmutated CLL display evidence K, et al. Expression of ribosomal and translation-associated
of antigen-experience. Thus, the available evidence is genes is correlated with a favorable clinical course in chronic
lymphocytic leukemia. Blood 2003;101:2748-55.
compatible with the notion that CLL develops through 22. Alizadeh AA, Eisen MB, Davis RE, Ma C, Lossos IS, Rosen-
the oncogenic transformation of antigen-experienced B- wald A, et al. Distinct types of diffuse large B-cell lymphoma
cells. identified by gene expression profiling. Nature 2000;403:503-
11.
23. Fulci V, Chiaretti S, Goldoni M, Azzalin G, Carucci N,
The author would like thank Riccardo Dalla-Favera, Tavolaro S, et al. Quantitative technologies establish a novel
Andrea Califano, Gustavo Stolovitzky, Yuhai Tu, and Katia microRNA profile of chronic lymphocytic leukemia. Blood.
2007;109:4944-51.
Basso for their essential involvement in some of the GEP exper- 24. Tobin G, Thunberg U, Johnson A, Eriksson I, Soderberg O,
iments described here. Karlsson K, et al. Chronic lymphocytic leukemias utilizing the
VH3-21 gene display highly restricted Vlambda2-14 gene use
and homologous CDR3s: implicating recognition of a com-
mon antigen epitope. Blood 2003;101:4952-7.
References 25. Falt S, Merup M, Tobin G, Thunberg U, Gahrton G, Rosen-
quist R, et al. Distinctive gene expression pattern in VH3-21
1. Kipps TJ. Chronic lymphocytic leukemia. Curr Opin Hematol utilizing B-cell chronic lymphocytic leukemia. Blood 2005;
1998;5:244-53. 106:681-9.
2. Caligaris-Cappio F, Hamblin TJ. B-cell chronic lymphocytic 26. Potter KN, Orchard J, Critchley E, Mockridge CI, Jose A,
leukemia: a bird of a different feather. J Clin Oncol 1999; Stevenson FK. Features of the overexpressed V1-69 genes in
17:399-408. the unmutated subset of chronic lymphocytic leukemia are
3. Chiorazzi N, Ferrarini MB. Cell chronic lymphocytic distinct from those in the healthy elderly repertoire. Blood
leukemia: lessons learned from studies of the B cell antigen 2003;101:3082-4.
receptor. Ann Rev Immunol 2003;21:841-94. 27. Widhopf GF, 2nd, Kipps TJ. Normal B cells express 51p1-
4. Schroeder HW, Dighiero G. The pathogenesis of chronic lym- encoded Ig heavy chains that are distinct from those expressed
phocytic leukemia: analysis of the antibody repertoire. by chronic lymphocytic leukemia B cells. J Immunol 2001;166:
Immunol Today 1994;15:288-94. 95-102.
5. Oscier DG, Thompsett A, Zhu D, Stevenson FK. Differential 28. Widhopf GF, 2nd, Rassenti LZ, Toy TL, Gribben JG, Wierda
rates of somatic hypermutation in V(H) genes among subsets WG, Kipps TJ. Chronic lymphocytic leukemia B cells of more
of chronic lymphocytic leukemia defined by chromosomal than 1% of patients express virtually identical immunoglobu-
abnormalities. Blood 1997;89:4153-60. lins. Blood 2004;104:2499-504.
6. Fais F, Ghiotto F, Hashimoto S, Sellars B, Valetto A, Allen SL, 29. Messmer BT, Albesiano E, Efremov DG, Ghiotto F, Allen SL,
et al. Chronic lymphocytic leukemia B cells express restricted Kolitz J, et al. Multiple distinct sets of stereotyped antigen
sets of mutated and unmutated antigen receptors. J Clin Invest receptors indicate a role for antigen in promoting chronic lym-
1998;102:1515-25. phocytic leukemia. J Exp Med 2004;200:519-25.
7. Klein U, Goossens T, Fischer M, Kanzler H, Braeuninger A, 30. Tobin G, Thunberg U, Karlsson K, Murray F, Laurell A,
Rajewsky K, et al. Somatic hypermutation in normal and Willander K, et al. Subsets with restricted immunoglobulin
transformed human B cells. Immunol Rev 1998;162:261-80. gene rearrangement features indicate a role for antigen selec-
8. Stevenson F, Sahota S, Zhu D, Ottensmeier C, Chapman C, tion in the development of chronic lymphocytic leukemia.
Oscier D, et al. Insight into the origin and clonal history of B- Blood 2004;104:2879-85.
cell tumors as revealed by analysis of immunoglobulin vari- 31. Stamatopoulos K, Belessi C, Moreno C, Boudjograh M, Guida
able region genes. Immunol Rev 1998;162:247-59. G, Smilevska T, et al. Over 20% of patients with chronic lym-
9. Rajewsky K. Clonal selection and learning in the antibody sys- phocytic leukemia carry stereotyped receptors: Pathogenetic
tem. Nature 1996;381:751-8. implications and clinical correlations. Blood 2007;109:259-70.
10. MacLennan IC. Germinal centers. Ann Rev Immunol 1994;12: 32. Murray F, Darzentas N, Hadzidimitriou A, Tobin G, Boudjogra
117-39. M, Scielzo C, et al. Stereotyped patterns of somatic hypermu-
11. Küppers R, Klein U, Hansmann ML, Rajewsky K. Cellular ori- tation in subsets of patients with chronic lymphocytic
gin of human B-cell lymphomas. N Engl J Med 1999;341:1520- leukemia: implications for the role of antigen selection in
9. leukemogenesis. Blood 2008;111:1524-33.
12. Küppers R, Dalla-Favera R. Mechanisms of chromosomal 33. Ghia P, Chiorazzi N, Stamatopoulos K. Microenvironmental
translocations in B cell lymphomas. Oncogene 2001;20:5580- influences in chronic lymphocytic leukaemia: the role of anti-
94. gen stimulation. J Intern Med 2008;264:549-62.
13. Klein U, Dalla-Favera R. Germinal centres: role in B-cell phys- 34. Chu CC, Catera R, Hatzi K, Yan XJ, Zhang L, Wang XB, et al.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 59 |
14th Congress of the European Hematology Association

Chronic lymphocytic leukemia antibodies with a common Gene expression profiling of hairy cell leukemia reveals a phe-
stereotypic rearrangement recognize nonmuscle myosin notype related to memory B cells with altered expression of
heavy chain IIA. Blood 2008;112:5122-9. chemokine and adhesion receptors. J Exp Med 2004;199:59-
35. Catera R, Silverman GJ, Hatzi K, Seiler T, Didier S, Zhang L, et 68.
al. Chronic lymphocytic leukemia cells recognize conserved 49. Harris NL, Jaffe ES, Stein H, Banks PM, Chan JK, Cleary ML,
epitopes associated with apoptosis and oxidation. Mol Med et al. A revised European-American classification of lymphoid
2008;14:665-74. neoplasms: a proposal from the International Lymphoma
36. Muzio M, Apollonio B, Scielzo C, Frenquelli M, Vandoni I, Study Group Blood 1994;84:1361-92.
Boussiotis V, et al. Constitutive activation of distinct BCR-sig- 50. Haglund U, Juliusson G, Stellan B, Gahrton G. Hairy cell
naling pathways in a subset of CLL patients: a molecular sig- leukemia is characterized by clonal chromosome abnormali-
nature of anergy. Blood 2008;112:188-95. ties clustered to specific regions. Blood. 1994;83(9):2637-45.
37. Damle RN, Ghiotto F, Valetto A, Albesiano E, Fais F, Yan XJ, et 51. Sambani C, Trafalis DT, Mitsoulis-Mentzikoff C, Poulakidas
al. B-cell chronic lymphocytic leukemia cells express a surface E, Makropoulos V, Pantelias GE, et al. Clonal chromosome
membrane phenotype of activated, antigen-experienced B rearrangements in hairy cell leukemia: personal experience
lymphocytes. Blood 2002;99:4087-93.
38. Klein U, Rajewsky K, Küppers R. Human immunoglobulin and review of literature. Cancer Genet Cytogenet 2001;
(Ig)M+IgD+ peripheral blood B cells expressing the CD27 cell 129:138-44.
surface antigen carry somatically mutated variable region 52. Wortis HH, Teutsch M, Higer M, Zheng J, Parker DC. B-cell
genes: CD27 as a general marker for somatically mutated activation by crosslinking of surface IgM or ligation of CD40
(memory) B cells. J Exp Med 1998;188:1679-89. involves alternative signal pathways and results in different B-
39. Tangye SG, Liu YJ, Aversa G, Phillips JH, de Vries JE. cell phenotypes. Proc Natl Acad Sci USA 1995;92:3348-52.
Identification of functional human splenic memory B cells by 53. Dono M, Burgio VL, Colombo M, Sciacchitano S, Reverberi D,
expression of CD148 and CD27. J Exp Med 1998;188:1691- Tarantino V, et al. CD5+ B cells with the features of subepithe-
703. lial B cells found in human tonsils. Eur J Immunol 2007;37:
40. Agematsu K, Nagumo H, Yang FC, Nakazawa T, Fukushima 2138-47.
K, Ito S, et al. B cell subpopulations separated by CD27 and 54. Rawstron AC, Bennett FL, O'Connor SJ, Kwok M, Fenton JA,
crucial collaboration of CD27+ B cells and helper T cells in Plummer M, et al. Monoclonal B-cell lymphocytosis and
immunoglobulin production. Eur J Immunol 1997;27:2073-9. chronic lymphocytic leukemia. N Engl J Med 2008;359:575-83.
41. Kindler V, Zubler RH. Memory, but not naive, peripheral 55. Dagklis A, Fazi C, Sala C, Cantarelli V, Scielzo C, Massacane
blood B lymphocytes differentiate into Ig-secreting cells after R, et al. The immunoglobulin gene repertoire of low-count
CD40 ligation and costimulation with IL-4 and the differenti- CLL-like MBL is different from CLL: diagnostic implications
ation factors IL-2, IL-10, and IL-3. J Immunol 1997;159:2085- for clinical monitoring. Blood 2009 [Epub ahead of print].
90. 56. Landgren O, Albitar M, Ma W, Abbasi F, Hayes RB, Ghia P, et
42. Werner-Favre C, Bovia F, Schneider P, Holler N, Barnet M, al. B-cell clones as early markers for chronic lymphocytic
Kindler V, et al. IgG subclass switch capacity is low in leukemia. N Engl J Med 2009;360:659-67.
switched and in IgM-only, but high in IgD+IgM+, post-germi- 57. Obukhanych TV, Nussenzweig MC. T-independent type II
nal center (CD27+) human B cells. Eur J Immunol 2001;31: immune responses generate memory B cells. J Exp Med 2006;
243-9. 203:305-10.
43. Agematsu K, Hokibara S, Nagumo H, Komiyama A. CD27: a 58. Weller S, Braun MC, Tan BK, Rosenwald A, Cordier C, Conley
memory B-cell marker. Immunol Today 2000;21:204-6. ME, et al. Human blood IgM "memory" B cells are circulating
44. Dono M, Zupo S, Massara R, Ferrini S, Melagrana A, splenic marginal zone B cells harboring a prediversified
Chiorazzi N, et al. In vitro stimulation of human tonsillar
subepithelial B cells: requirement for interaction with activat- immunoglobulin repertoire. Blood 2004;104:3647-54.
ed T cells. Eur J Immunol 2001;31:752-6. 59. Kruetzmann S, Rosado MM, Weber H, Germing U, Tournilhac
45. Martin F, Kearney JF. Marginal-zone B cells. Nat Rev Immunol O, Peter HH, et al. Human immunoglobulin M memory B cells
2002;2:323-35. controlling Streptococcus pneumoniae infections are generat-
46. Döhner H, Stilgenbauer S, Döhner K, Bentz M, Lichter P. ed in the spleen. J Exp Med 2003;197:939-45.
Chromosome aberrations in B-cell chronic lymphocytic 60. van Oers MH, Pals ST, Evers LM, van der Schoot CE,
leukemia: reassessment based on molecular cytogenetic analy- Koopman G, Bonfrer JM, et al. Expression and release of CD27
sis. J Mol Med 1999;77:266-81. in human B-cell malignancies. Blood 1993;82:3430-6.
47. Mayr C, Speicher MR, Kofler DM, Buhmann R, Strehl J, Busch 61. Toellner KM, Jenkinson WE, Taylor DR, Khan M, Sze DM,
R, et al. Chromosomal translocations are associated with poor Sansom DM, et al. Low-level hypermutation in T cell-inde-
prognosis in chronic lymphocytic leukemia. Blood 2006;107: pendent germinal centers compared with high mutation rates
742-51. associated with T cell-dependent germinal centers. J Exp Med
48. Basso K, Liso A, Tiacci E, Benedetti R, Pulsoni A, Foa R, et al. 2002;195:383-9.

| 60 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Chronic lymphocytic leukemia

Clinical implications of novel biological markers


in chronic lymphocytic leukemia

R. Rosenquist A B S T R A C T

Considering that chronic lymphocytic leukemia (CLL) is an inherently heterogeneous disease with
Department of Genetics and varying clinical outcome, there has been great interest throughout the last 10 years in establishing
Pathology, Uppsala University,
Uppsala, Sweden reliable prognostic markers to predict its outcome. The immunoglobulin heavy-chain variable (IGHV)
gene mutation status and FISH analysis of recurrent genomic aberrations represents strong and robust
genetic markers that currently can be applied in CLL prognostication. A plethora of other biological
markers has been proposed at the protein (CD38, ZAP-70), DNA (TP53 mutations, telomere length) and
Hematology Education: RNA (CLLU1, LPL) level, where many of these are promising for outcome prediction. To be applied in
the education program for the
the clinical setting, however, it is very important to not only select biomarkers that have been repeat-
annual congress of the European
Hematology Association edly confirmed in independent cohorts, preferably in prospective trials, but also ones that can be ana-
lyzed by standardized assays. In this overview, the pros and cons of a number of biological, prognos-
2009;3:61-67 tic markers will be discussed. Many of these markers will require further evaluation, particularly in
relation to new treatment protocols. Ultimately, we may be able to create a ‘molecular prognostic
index’ in CLL comprised of the most informative and robust markers. By studying the mechanisms
behind these various markers, further insights into CLL pathogenesis will also be unraveled.

major breakthrough in chronic lym- unmutated B-cells, yet it was not until the

A phocytic leukemia (CLL) prognostics


was the observation that the somatic
hypermutation status of the immunoglobu-
mid-nineties that Schroeder and Dighiero
reported that CLL cells may display a more
diverse somatic hypermutation status than
lin heavy-chain variable (IGHV) genes could anticipated.11 In 1998, Fais et al. demonstrat-
group CLL into two main categories;1,2 IGHV ed that roughly 50% of their CLL patients
mutated CLL with an expected long overall had mutated IGHV genes,12 and this was-
survival and IGHV unmutated CLL, having closely followed by two simultaneous publi-
an inferior outcome. Moreover, the presence cations verifying the existence of two sub-
of certain genomic aberrations was also sets of CLL, i.e., IGHV mutated and unmu-
demonstrated to have great impact on CLL tated,1,2 both similar in size. Importantly,
prognosis,3,4 where poor-risk markers such these latter reports illustrated that CLL cells
as deletion of 11q and 17p are strongly asso- carrying unmutated IGHV genes (>98%
ciated with short survival. Following these identity to the corresponding germline
discoveries, there has been a tremendous IGHV gene) had a significantly shorter over-
interest in prognostic indicators in CLL, all survival compared to those cases show-
which is reflected by the numerous biologi- ing somatically hypermutated IGHV genes
cal markers suggested throughout the last (<98% identity). Furthermore, in line with
decade. These different markers could be the poor survival, IGHV unmutated CLL dis-
proteins, both surface-bound and intracellu- played a more progressive disease, showed
lar (CD38, ZAP-70, CD49d), RNA-based poor-risk genomic aberrations more fre-
(LPL, CLLU1, micro-RNA) or DNA-based quently and required more chemotherapy
[TP53 mutations, single nucleotide polymor- than mutated CLL.1,2 Many groups have sub-
phisms (SNPs), telomere length].1,5-10 It is sequently verified the strong predictive
clear that we need prognostic markers in capacity of the IGHV mutational status in
CLL to subdivide this heterogeneous dis- multivariate analysis, both in more “aggres-
ease; however, the essential question is sive” and in more “benign” cohorts (Figure
which markers/combination of markers 1).13-15
should be applied in clinical trials and in clin- The cutoff for dividing mutated and
ical routine. In this overview, the clinical and unmutated CLL cases was initially set at
biological role of many of these new mark- 98% identity in order to avoid counting
ers will be discussed, with a focus on biolog- polymorphisms as true mutations.
ical markers that have repeatedly been However, since most polymorphisms are
shown to be good predictors of prognosis in currently known, this issue is not as relevant
CLL. today. Although the 98% cutoff has repeat-
edly been shown to be the best statistical
Immunoglobulin gene analysis level for clinical discrimination of unmutat-
For many years, it was assumed that CLL ed and mutated CLL and is widely accepted,
cells would originate from CD5+, naïve, this is probably not as straightforward at a

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 61 |
14th Congress of the European Hematology Association

biological level. Firstly, the IGHV mutational status is


not absolute at the individual level. For instance, CLL
patients with borderline identity (97-98%) may have a
heterogeneous disease course, where some patients will
experience a more aggressive clinical course despite
being classified as “mutated”.15,16 Furthermore, excep-
tions to the mutated/unmutated classification have
been shown, for instance IGHV3-21 gene usage (see
below).17
The IG gene analysis is very stable and this test can be
performed at any time point during the course of the
disease since the IGHV mutational status remains con-
stant. That notwithstanding, there are many technical
aspects to consider when performing IG gene analysis
in CLL. In order to standardize this procedure, eight
European laboratories recently published recommenda-
tions for successful analysis18 and several educational
workshops focused on this topic. Despite the time-con-
suming nature of IGHV mutation analysis, it has now
become standard in many laboratories worldwide and
currently represents one of the most reliable prognostic
markers in CLL.

Immunoglobulin heavy-chain variable gene usage and


B-cell receptor “stereotypy”
In 2002, our group reported an exception to the
mutated and unmutated subcategorization. Examina-
tion of our cohort revealed a novel subset of CLL cases
utilizing the IGHV3-21 gene at a frequency of approxi-
mately 10%.17 The majority of these IGHV3-21 cases
carried mutated IGHV genes but still followed a disease
course similar to unmutated CLL cases. We verified this
finding in a follow-up study, where two-thirds of cases
had mutated IGHV genes and showed poor outcome.19
Interestingly, this subset had peculiar characteristics,
such as predominant λ expression and restricted IG
Figure 1. (A) Time to treatment and (B) overall survival
gene features; an extremely short and almost identical data in a subset of cases (n=202) from a population-based
complementarity determining region 3 (CDR3) together Scandinavian CLL cohort. The high proportion of IGHV
with usage of one lambda light-chain gene, IGLV3-21, in mutated (63%) versus unmutated (37%) cases reflects its
unselected nature.
greater than 50% of patients.19 The finding of a “stereo-
typed” B-cell receptor in non-related IGHV3-21 CLL
patients has been confirmed worldwide,20-22 and indi-
cates that antigens may be involved in CLL develop- stereotyped cases.27 Stereotyped IGHV4-34 cases also
ment. displayed unique characteristics, such as a low median
Thereafter, we and others have identified several age at diagnosis and IgG switched receptors.27
additional subsets with stereotyped heavy-chain Moreover, a subgroup expressing various IGHV1/5/7
CDR3s in combination with restricted light-chain gene genes but with similar CDR3 sequences (subset 1, the
usage, such as the IGHV1-69/IGKV3-20 and the IGHV4- “mixed” subset) had a very poor survival compared to
39/IGKV2-30 subset,23-26 further supporting the concept non-stereotyped cases.27 This apparent relationship
of antigen-driven leukemogenesis A major advance- between stereotypy and clinical outcome may therefore
ment in this area was made in 2007, when Stama- be useful and applicable in the foreseeable future in clin-
topoulos and colleagues investigated a large Mediter- ical reporting, in conjunction with the IGHV mutation-
ranean CLL cohort and demonstrated that at least 48 al status. We now have to continue performing large
different subsets existed, with more than 20% of their multi-center studies to fully elucidate the clinical rele-
CLL cases belonging to one of these ‘stereotyped’ sub- vance of various stereotyped subsets, which today com-
sets.27 Interestingly, they showed that stereotypy, inde- prises of more than 100 subsets.29
pendent of mutation status, may influence clinical out-
come in CLL, a finding which has lately been verified by The search for surrogate markers
others.28 For instance, stereotyped IGHV3-21 cases (des- CD38
ignated subset 2) had an inferior prognosis than non- Since the discovery of the clinical impact of the IGHV
stereotyped IGHV3-21 cases, with shorter time to pro- mutational status in CLL, many attempts have been
gression, as well as the presence of poor-risk mark- made to find surrogate markers to substitute the IG
ers.21,27-28 Conversely, stereotyped IGHV4-34 cases (sub- gene analysis, preferably markers suitable for flow cyto-
set 4) had a superior prognosis when compared to non- metric analysis. In 1999, Damle and colleagues showed

| 62 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

that expression of CD38 correlated well with the IGHV Lipoprotein lipase
mutation status and could be suitable as a surrogate Gene expression profiling also revealed that several
marker.1 Using a 30% cutoff, they demonstrated that all other genes were differently expressed in mutated and
CD38+ cases had unmutated IGHV genes, whereas most unmutated CLL.9 One such gene was lipoprotein lipase
of the CD38– cases carried mutated IGHV genes. Several (LPL) which was highly expressed in unmutated CLL.
reports have reproduced this initial finding;30,31 however, Using real-time PCR, several independent groups have
we and others have shown a less stringent correlation confirmed this finding and shown a strong correlation
between the markers in up to one-third of patients.13,32,33 with poor outcome in CLL.8,43,44 We recently investigat-
Furthermore, despite being easily measured by flow ed the LPL levels in 140 patients, both at the RNA and
cytometry, a consensus cutoff level has never been protein level, and confirmed high levels in unmutated
reached, and various cutoffs have been described (rang- CLL (Figure 2),45 where multivariate analysis revealed
ing from any positivity to 30%)1,13,31,34 In addition, CD38 LPL as a strong independent marker for survival predic-
levels have been reported to vary over time in up to a tion.
quarter of CLL cases,35 although this has not been
demonstrated by others.34 In a recent combined analysis
of CD38, ZAP-70 and IGHV mutation status, CD38 did
not improve the ability to predict time to treatment
compared to the other two markers,36 which is in line
with our own observation.15

ZAP-70
In 2001, Rosenwald and colleagues performed large-
scale gene expression profiling of unmutated and mutat-
ed CLL and surprisingly found rather few differently
expressed genes between the groups.9 One very inter-
esting finding was the upregulation of Zeta-Associated
Protein 70 (ZAP-70) in unmutated CLL. ZAP-70 was,
until then, well-studied in T-cells but had not been ana-
lyzed in B-cells. Several follow-up papers verified the
upregulation of ZAP-70 in unmutated CLL, and ZAP-70
analysis using flow-cytometry was proposed as a surro-
gate assay for IG gene analysis.37-39 For instance, Crespo
et al. showed that all cases with unmutated IGHV genes
were ZAP-70+, whereas most IGHV mutated cases were
ZAP-70–.37 In contrast, Rassenti et al. reported that up to Figure 2. Prognostic impact of low versus high LPL expres-
a quarter of cases had discordant mutation status and sion in 136 Swedish CLL patients (based on the threshold
value 0.0001 as defined by ROC curve analysis).45
ZAP-70 expression.40 These inconsistencies could in part
be attributed to the observation that patients with 11q
or 17p deletion could be ZAP-70– despite being IGHV
unmutated, whereas mutated IGHV3-21 CLL patients
could show high ZAP-70 expression.41
Although ZAP-70 may not be an ideal surrogate mark- Table 1. Established and potential biological, prognostic
er for the IGHV mutation status at the cohort or individ- markers in chronic lymphocytic leukemia.
ual level, it has repeatedly been shown to be an inde- Immunoglobulin gene analysis IGHV gene mutation status
pendent prognostic marker for time to progression and IGHV gene usage
overall survival in CLL.37,38,40 Some studies have also B-cell receptor ‘stereotypy’
shown that ZAP-70 may even be the superior marker for
estimating prognosis in CLL.36,40 However, since its dis- Flow cytometry CD38
covery there has been an intense debate how to perform ZAP-70
ZAP-70 analysis in a standardized way. This relates to CD49d
the fact that ZAP-70 (i) is an intracellular protein; (ii) is FCRL2
expressed not only by T-cells but also by normal B- Realtime-PCR LPL
cells:42 (iii) that different protocols have been employed CLLU1
using various antibodies and cutoffs. A multi-center Protein- or RNA-based MCL1
effort initiated by the European Research Initiative on TCL1
CLL (ERIC) has aimed at finding a standardized proto- HS1
col, which probably will be reported during 2009.
Genetic aberrations CLL-FISH
Newly proposed markers (various techniques) TP53 mutations
In addition to CD38 and ZAP-70, several other inter- ATM mutations
esting markers have recently been proposed to predict Clonal evolution
Genomic complexity
outcome in CLL. Brief presentations of four selected
Telomere length
markers follow, chosen from the rather extensive litera-
micro-RNAs 13-miR signature, miR-29c, miR-223
ture that has emerged lasting recent years (Table 1).

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 63 |
14th Congress of the European Hematology Association

CLLU1 tions, whilst trisomy 12 patients had a more intermedi-


Using differential display technique, Buhl and col- ate risk-profile.3 Since conventional cytogenetics tradi-
leagues identified a new gene expressed in unmutated tionally had a low success rate, fluorescence in situ
CLL.5 This gene was cloned and found not to be hybridization (FISH) analysis rapidly became the gold
expressed in any other tissue type or in other hemato- standard in CLL using a commercial panel of FISH probes
logical malignancies, and could thus be described as a to screen for 11q-, 13q-, +12 and 17p-. Patients with 11q
CLL-specific gene, CLL up-regulated gene 1 (CLLU1). or 17p deletion usually carry unmutated IGHV genes and
Furthermore, high CLLU1 expression correlated with they generally respond poorly to current treatment pro-
short time to treatment and overall survival, and com- tocols, in particular the 17p- group.13 For the latter group,
plemented several other prognostic markers, such as treatment regimes using alemtuzumab and prednisolone
stage, mutation status and ZAP-70 expression.5,46 To appear especially promising.52 FISH analysis is now con-
date, no “CLLU1” protein has been identified; however, sidered mandatory as a predictive marker, not only in
such a protein could make an interesting new therapeu- clinical trials but also in routine practice,53 particularly
tic target. when treatment is planned with the aim of long-term
remission. Since clonal evolution is a known phenome-
TCL-1 non in CLL,54 where poor-risk aberrations can emerge
TCL-1 is an anti-apoptotic protein which was first over time, it is also important to perform FISH analysis
described in T-cell leukemia47 and more recently high when there is a change in the patient’s clinical course or
levels of TCL-1 have been demonstrated in unmutated if the patient responds poorly to therapy.
CLL and in patients with 11q deletion.48 By applying
realtime-PCR we confirmed high levels of TCL-1 in TP53 mutations
unmutated CLL, however TCL-1 did not remain inde- Recently, the significant role of TP53 mutations was
pendent following multivariate analysis,49 possibly due reinforced in CLL. By screening the TP53 gene, Zentz et
to its close association to IGHV mutation status and 11q al. demonstrated that most cases with 17p deletion also
deletion. Interestingly, we observed that the poor-prog- displayed an accompanying TP53 mutation on the other
nostic IGHV3-21 group displayed high levels similar to allele.10 Importantly, they also reported that a proportion
the high ZAP-70 levels also observed it this group.41 of cases (4-5%) displayed a TP53 mutation in the
absence of a 17p deletion, and such cases had a poor
Micro-RNA prognosis similar to cases with 17p deletion. This find-
Micro-RNAs (miR) are short non-coding RNA mole- ing has now been confirmed by others.55 Different tech-
cules where each miR is believed to silence the expres- niques are available for detection of TP53 mutations,
sion of other target genes. Recently, genome-wide miR such as direct sequencing and denaturing high-pressure
analysis revealed a CLL-specific profile of 13 different liquid chromatography (dHPLC). dHPLC gives a higher
miRs. Interestingly, a combination of these miRs corre- sensitivity, although direct sequencing is more readily
lated with other prognostic markers and clinical out- available in most laboratories. In addition, it has been
come.50 Down-regulation of two individual miRs, shown that assessment of p53 dysfunction, using for
miR29c and miR223, was also recently associated with example, flow cytometry, predicts poor prognosis in
inferior outcome in CLL.51 In the coming years, we will CLL.56 Considering the very poor prognosis in the TP53
hopefully learn much about the role of specific micro- mutated group, it is reasonable to assume that mutation
RNAs, which will help us to unravel important aspects screening will soon be added to the FISH panel,
of CLL pathogenesis. although the most appropriate technique to apply has
yet to be agreed upon.
Genetics in chronic lymphocytic leukemia
As with many other B-cell lymphoma entities, there is Genomic complexity
no genetic aberration that is common to all CLL Difficulties exist culturing CLL cells whilst attempting
patients. Several recurrent aberrations have been to obtain proper metaphases for karyotyping. In recent
described, the most frequent being deletion of 13q14, years, however, new culturing protocols have been
followed by deletion of 11q22-23, trisomy 12 and dele- applied that have improved the success rate significant-
tion of 17p13.3,4 The 13q deletion has been intensively ly.57,58 By adding IL2 and CpG oligonucleotides, Haferlach
analyzed during the last 10 to 15 years without the suc- and colleagues achieved successful cultures in most of
cessful identification of any candidate genes. However, their 500 cases and detected clonal aberrations in more
it has lately been shown that miR15 and miR16, which than 80%.59 Interestingly, several reports have now
are encoded in the deleted region, are involved in the shown that an increasing number of aberrations and the
pathogenesis of CLL.50 In addition, the 11q22-23 dele- presence of translocations, especially unbalanced, will
tion spans the ATM gene, a very important regulator of predict poor outcome in CLL.57-59 Hence, we may consid-
cell division and DNA repair, whereas the 17p13 dele- er applying these new culturing protocols when analyz-
tion covers the TP53 gene, a tumor suppressor gene cru- ing for genomic aberrations in CLL.
cial in cell cycle regulation. Recent studies using high-resolution array-based tech-
nology have detected few novel recurrent aberrations in
CLL-FISH CLL.60,61 However, considering the recent advances with
In 2000, Döhner et al. proposed the hierarchical model oligonucleotide arrays and SNP-arrays, it is still possible
for classification of CLL, where patients with 13q dele- that further important aberrations will be detected. In
tion, as a single aberration, were shown to have superior line with recent cytogenetic studies, the finding of higher
outcome to patients with 11q and in particular 17p dele- genomic complexity (≥3 aberrations) using genomic

| 64 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

arrays was also predictive of poor outcome in CLL.62 We


recently investigated part of a population-based
Scandinavian cohort (203 of ~600 cases) using Affymetrix
250K SNP-arrays, where we confirmed the prognostic
impact of the recurrent aberrations and increasing com-
plexity (Figure 3).63 That notwithstanding, one has to
keep in mind that most of the cases with higher complex-
ity are cases with 11q/17p deletion.59 Although of great
interest, the application of whole-genome arrays is
expensive and the procedure remains to be standardized
before it can be used in the clinical setting.

SNP analysis
During the last seven years, numerous new correla-
tions between single SNPs and clinical outcome have
been proffered. Notably, genetic variants within the
P2X7, BCL-2, BAX, MCL1, and GNAS1 genes have been
reported,64-67 although none of these SNPs have subse-
quently been confirmed in independent materials.68-71 For
instance, the MDM2 SNP309 was shown to correlate
with poor prognosis in CLL,72 whereas a separate study
and our own unpublished data did not reveal any corre-
lation.73 The lesson learnt from all these studies is that a
SNP, which appears to give prognostic information in
CLL, requires re-confirmation in a second, independent
cohort before proposing it as a clinically applicable
prognostic marker.

Telomeres
Telomeres, the ends of the chromosomes, are com-
posed of repetitive DNA sequences (TTAGGG). During
each cell division, the telomere shortens, and when the
telomeres eventually become too short, the cell under-
goes apoptosis. However, in certain cells types, such as
stem cells and germ cells, and in cancer cells, the Figure 3. Genomic data achieved by applying high-resolu-
enzyme telomerase restores telomere length, thereby tion SNP-arrays (Affymetrix, 250K) in 203 CLL cases from
a Scandinavian population-based cohort. (A) Survival
allowing these cells an extended replicative capacity. In analysis according to the hierarchical classification (13q-,
CLL, we and others have shown that short telomeres no aberration, +12, 11q- and 17p-). (B) Survival analysis in
correlate strongly to unmutated IGHV genes and poor relation to increasing genomic complexity, when including
copy-number aberrations larger than 5 Mb. CNA, copy-num-
outcome, whereas longer telomeres are associated with ber aberration.
good prognosis and mutated IGHV genes.7,74-77 In unmu-
tated CLL, the short telomeres may reflect a higher pro-
liferative activity of the progenitor cells, although the a clinical setting. Firstly, for all markers, it is important
telomeres will remain intact due to high telomerase to perform extensive analysis, not only retrospectively
activity.77 Recently, we demonstrated that CLL patients but also prospectively, in independent, large cohorts and
with complex karyotype and/or presence of 11q-/17p- to evaluate them against already established markers.
also demonstrated significantly shorter telomeres.76 Hopefully, this will render a “molecular prognostic
Initially, the assay used to determine the length of index” in CLL, where the most suitable and strongest
telomeres was the time-consuming and cumbersome markers are combined. Furthermore, it is very essential
Southern blot technique.74 However, a simple PCR- to define markers that can predict therapy response in
based assay is now available, although standardization CLL, especially considering ongoing and new upcoming
between laboratories has to be performed if this test is strategies for therapy. To date, a few studies have exten-
to be routinely applied.7 sively compared predictive capacity of prognostic mark-
ers in large materials. In one of them, as mentioned,
Conclusions ZAP-70 was shown to be the strongest predictive mark-
During the last decade, several prognostic markers er of time to treatment, followed by the IGHV mutation
have been demonstrated to be solid and reproducible status, whereas CD38 did not add to the other two
markers to predict outcome in CLL, such as the IGHV markers.36 Similar prospective studies are now necessary
mutation status and FISH analysis. In addition to this, a to guide us through this “jungle” of prognostic markers.
very extensive list of markers has been proposed to be As a final note, one may say that we currently have
of prognostic significance in CLL, some of which have too many proposed markers in CLL. Conversely, while
been detailed in this paper and which appear promising. many of these will never be applied in the clinics, hope-
The next step is to determine which markers hold most fully we will learn more about CLL biology by trying to
prognostic value, but are reasonably easy to perform in understand the reasoning behind their deregulation.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 65 |
14th Congress of the European Hematology Association

leukemia is associated with high-risk disease and reflects anti-


References gen-driven, post-germinal center leukemogenic selection.
Blood 2008;111:5101-8.
1. Damle RN, Wasil T, Fais F, Ghiotto F, Valetto A, Allen SL, et al. 21. Ghia P, Stamatopoulos K, Belessi C, Moreno C, Stella S, Guida
Ig V gene mutation status and CD38 expression as novel prog- G, et al. Geographic patterns and pathogenetic implications of
nostic indicators in chronic lymphocytic leukemia. Blood 1999; IGHV gene usage in chronic lymphocytic leukemia: the lesson
94:1840-7. of the IGHV3-21 gene. Blood 2005;105:1678-85.
2. Hamblin TJ, Davis Z, Gardiner A, Oscier DG, Stevenson FK. 22. Thorselius M, Krober A, Murray F, Thunberg U, Tobin G,
Unmutated Ig V(H) genes are associated with a more aggres- Buhler A, et al. Strikingly homologous immunoglobulin gene
sive form of chronic lymphocytic leukemia. Blood 1999;94: rearrangements and poor outcome in VH3-21-using chronic
1848-54. lymphocytic leukemia patients independent of geographic ori-
3. Döhner H, Stilgenbauer S, Benner A, Leupolt E, Kröber A, gin and mutational status. Blood 2006;107:2889-94.
Bullinger L, et al. Genomic aberrations and survival in chronic 23. Ghiotto F, Fais F, Valetto A, Albesiano E, Hashimoto S, Dono
lymphocytic leukemia. TNew Engl J Med 2000;343:1910-6. M, et al. Remarkably similar antigen receptors among a subset
4. Juliusson G, Oscier DG, Fitchett M, Ross FM, Stockdill G, of patients with chronic lymphocytic leukemia. The Journal of
Mackie MJ, et al. Prognostic subgroups in B-cell chronic lym- clinical investigation 2004;113:1008-16.
phocytic leukemia defined by specific chromosomal abnormal- 24. Messmer BT, Albesiano E, Efremov DG, Ghiotto F, Allen SL,
ities. New Engl J Med 1990 S;323:720-4. Kolitz J, et al. Multiple distinct sets of stereotyped antigen
5. Buhl AM, Jurlander J, Jorgensen FS, Ottesen AM, Cowland JB, receptors indicate a role for antigen in promoting chronic lym-
Gjerdrum LM, et al. Identification of a gene on chromosome phocytic leukemia. J Exp Med 2004;200:519-25.
12q22 uniquely overexpressed in chronic lymphocytic 25. Tobin G, Thunberg U, Karlsson K, Murray F, Laurell A,
leukemia. Blood 2006;107:2904-11. Willander K, et al. Subsets with restricted immunoglobulin
6. Gattei V, Bulian P, Del Principe MI, Zucchetto A, Maurillo L, gene rearrangement features indicate a role for antigen selec-
Buccisano F, et al. Relevance of CD49d protein expression as tion in the development of chronic lymphocytic leukemia.
overall survival and progressive disease prognosticator in Blood 2004;104:2879-85.
chronic lymphocytic leukemia. Blood 2008;111:865-73. 26. Widhopf GF, 2nd, Rassenti LZ, Toy TL, Gribben JG, Wierda
7. Grabowski P, Hultdin M, Karlsson K, Tobin G, Aleskog A, WG, Kipps TJ. Chronic lymphocytic leukemia B cells of more
Thunberg U, et al. Telomere length as a prognostic parameter than 1% of patients express virtually identical immunoglobu-
in chronic lymphocytic leukemia with special reference to VH lins. Blood 2004;104:2499-504.
gene mutation status. Blood 2005;105:4807-12. 27. Stamatopoulos K, Belessi C, Moreno C, Boudjograh M, Guida
8. Oppezzo P, Vasconcelos Y, Settegrana C, Jeannel D, Vuillier F, G, Smilevska T, et al. Over 20% of patients with chronic lym-
Legarff-Tavernier M, et al. The LPL/ADAM29 expression ratio phocytic leukemia carry stereotyped receptors: Pathogenetic
is a novel prognosis indicator in chronic lymphocytic leukemia. implications and clinical correlations. Blood 2007;109:259-70.
Blood 2005;106:650-7. 28. Bomben R, Dal Bo M, Capello D, Forconi F, Maffei R, Laurenti
9. Rosenwald A, Alizadeh AA, Widhopf G, Simon R, Davis RE, L, et al. Molecular and clinical features of chronic lymphocytic
Yu X, et al. Relation of gene expression phenotype to immuno- leukaemia with stereotyped B cell receptors: results from an
globulin mutation genotype in B cell chronic lymphocytic Italian multicentre study. Br J Haematol 2009;144:492-506
leukemia. J Exp Med 2001;194:1639-47. 29. Murray F, Darzentas N, Hadzidimitriou A, Tobin G, Boudjogra
10. Zenz T, Krober A, Scherer K, Habe S, Buhler A, Benner A, et al. M, Scielzo C, et al. Stereotyped patterns of somatic hypermu-
Monoallelic TP53 inactivation is associated with poor progno- tation in subsets of patients with chronic lymphocytic
sis in chronic lymphocytic leukemia: results from a detailed leukemia: implications for the role of antigen selection in
genetic characterization with long-term follow-up. Blood leukemogenesis. Blood 2008;111:1524-33.
2008;112:3322-9. 30. Del Poeta G, Maurillo L, Venditti A, Buccisano F, Epiceno AM,
11. Schroeder HW, Jr., Dighiero G. The pathogenesis of chronic Capelli G, et al. Clinical significance of CD38 expression in
lymphocytic leukemia: analysis of the antibody repertoire. chronic lymphocytic leukemia. Blood. 2001;98(9):2633-9.
Immunol Today. 1994;15(6):288-94. 31. Ibrahim S, Keating M, Do KA, O'Brien S, Huh YO, Jilani I, et
12. Fais F, Ghiotto F, Hashimoto S, Sellars B, Valetto A, Allen SL, et al. CD38 expression as an important prognostic factor in B-cell
al. Chronic lymphocytic leukemia B cells express restricted sets chronic lymphocytic leukemia. Blood. 2001;98(1):181-6.
of mutated and unmutated antigen receptors. J Clin Invest 32. Hamblin TJ, Orchard JA, Gardiner A, Oscier DG, Davis Z,
1998;102:1515-25. Stevenson FK. Immunoglobulin V genes and CD38 expression
13. Krober A, Seiler T, Benner A, Bullinger L, Bruckle E, Lichter P, in CLL. Blood 2000;95:2455-7.
et al. V(H) mutation status, CD38 expression level, genomic 33. Thunberg U, Johnson A, Roos G, Thörn I, Tobin G, Sällström
aberrations, and survival in chronic lymphocytic leukemia. J, et al. CD38 expression is a poor predictor for VH gene muta-
Blood 2002;100:1410-6. tional status and prognosis in chronic lymphocytic leukemia.
14. Oscier DG, Gardiner AC, Mould SJ, Glide S, Davis ZA, Blood 2001;97:1892-4.
Ibbotson RE, et al. Multivariate analysis of prognostic factors in 34. Ghia P, Guida G, Stella S, Gottardi D, Geuna M, Strola G, et al.
CLL: clinical stage, IGVH gene mutational status, and loss or The pattern of CD38 expression defines a distinct subset of
mutation of the p53 gene are independent prognostic factors. chronic lymphocytic leukemia (CLL) patients at risk of disease
Blood 2002;100:1177-84. progression. Blood 2003;101:1262-9.
15. Tobin G, Thunberg U, Laurell A, Karlsson K, Åleskog A, 35. Hamblin TJ, Orchard JA, Ibbotson RE, Davis Z, Thomas PW,
Willander K, et al. Patients with chronic lymphocytic leukemia Stevenson FK, et al. CD38 expression and immunoglobulin
with mutated VH genes presenting with Binet stage B or C variable region mutations are independent prognostic variables
form a subgroup with a poor outcome. Haematologica 2005; in chronic lymphocytic leukemia, but CD38 expression may
90:465-9. vary during the course of the disease. Blood 2002;99: 1023-9.
16. Hamblin TJ, Davis ZA, Oscier DG. Determination of how 36. Rassenti LZ, Jain S, Keating MJ, Wierda WG, Grever MR, Byrd
many immunoglobulin variable region heavy chain mutations JC, et al. Relative value of ZAP-70, CD38, and immunoglobu-
are allowable in unmutated chronic lymphocytic leukaemia - lin mutation status in predicting aggressive disease in chronic
long-term follow up of patients with different percentages of lymphocytic leukemia. Blood 2008;112:1923-30.
mutations. Br J Haematol 2008;140:320-3. 37. Crespo M, Bosch F, Villamor N, Bellosillo B, Colomer D,
17. Tobin G, Thunberg U, Johnson A, Thorn I, Söderberg O, Rozman M, et al. ZAP-70 expression as a surrogate for
Hultdin M, et al. Somatically mutated Ig V(H)3-21 genes char- immunoglobulin-variable-region mutations in chronic lympho-
acterize a new subset of chronic lymphocytic leukemia. Blood cytic leukemia. New Engl J Med 2003;348:1764-75.
2002;99:2262-4. 38. Orchard JA, Ibbotson RE, Davis Z, Wiestner A, Rosenwald A,
18. Ghia P, Stamatopoulos K, Belessi C, Moreno C, Stilgenbauer S, Thomas PW, et al. ZAP-70 expression and prognosis in chron-
Stevenson F, et al. ERIC recommendations on IGHV gene ic lymphocytic leukaemia. Lancet 2004;363:105-11.
mutational status analysis in chronic lymphocytic leukemia. 39. Wiestner A, Rosenwald A, Barry TS, Wright G, Davis RE,
Leukemia 2007;21:1-3. Henrickson SE, et al. ZAP-70 expression identifies a chronic
19. Tobin G, Thunberg U, Johnson A, Eriksson I, Söderberg O, lymphocytic leukemia subtype with unmutated immunoglob-
Karlsson K, et al. Chronic lymphocytic leukemias utilizing the ulin genes, inferior clinical outcome, and distinct gene expres-
VH3-21 gene display highly restricted Vlambda2-14 gene use sion profile. Blood 2003;101:4944-51.
and homologous CDR3s: implicating recognition of a common 40. Rassenti LZ, Huynh L, Toy TL, Chen L, Keating MJ, Gribben
antigen epitope. Blood 2003;101:4952-7. JG, et al. ZAP-70 compared with immunoglobulin heavy-chain
20. Ghia EM, Jain S, Widhopf GF, 2nd, Rassenti LZ, Keating MJ, gene mutation status as a predictor of disease progression in
Wierda WG, et al. Use of IGHV3-21 in chronic lymphocytic chronic lymphocytic leukemia. New Engl J Med 2004;351:893-

| 66 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

901. 506 cases analysed with chromosome banding analysis, inter-


41. Kröber A, Bloehdorn J, Hafner S, Buhler A, Seiler T, Kienle D, phase FISH, IgV(H) status and immunophenotyping. Leukemia
et al. Additional genetic high-risk features such as 11q deletion, 2007;21:2442-51.
17p deletion, and V3-21 usage characterize discordance of 60. Gunn SR, Bolla AR, Barron LL, Gorre ME, Mohammed MS,
ZAP-70 and VH mutation status in chronic lymphocytic Bahler DW, et al. Array CGH analysis of chronic lymphocytic
leukemia. J Clin Oncol 2006;24:969-75. leukemia reveals frequent cryptic monoallelic and biallelic dele-
42. Scielzo C, Camporeale A, Geuna M, Alessio M, Poggi A, tions of chromosome 22q11 that include the PRAME gene.
Zocchi MR, et al. ZAP-70 is expressed by normal and malig- Leuk Res 2008.
nant human B-cell subsets of different maturational stage. 61. Lehmann S, Ogawa S, Raynaud SD, Sanada M, Nannya Y,
Leukemia 2006;20:689-95. Ticchioni M, et al. Molecular allelokaryotyping of early-stage,
43. Heintel D, Kienle D, Shehata M, Krober A, Kroemer E, untreated chronic lymphocytic leukemia. Cancer 2008;112:
Schwarzinger I, et al. High expression of lipoprotein lipase in 1296-305.
poor risk B-cell chronic lymphocytic leukemia. Leukemia 62. Kujawski L, Ouillette P, Erba H, Saddler C, Jakubowiak A,
2005;19:1216-23. Kaminski M, et al. Genomic complexity identifies patients
44. van't Veer MB, Brooijmans AM, Langerak AW, Verhaaf B, with aggressive chronic lymphocytic leukemia. Blood 2008;
Goudswaard CS, Graveland WJ, et al. The predictive value of 112:1993-2003.
lipoprotein lipase for survival in chronic lymphocytic leukemia. 63. Gunnarsson R, Isaksson A, Göransson A, Mansouri M, Jansson
Haematologica 2006;91:56-63. M, Cahill N, et al High Resolution Screening of Copy-Number
45. Mansouri M, Sevov M, Fahlgren E, Tobin G, Roos G, Alterations in Chronic Lymphocytic Leukemia using
Olivecrona G, et al. Differential RNA and protein levels of Affymetrix 250K SNP-Arrays Reveals a Higher Complexity of
lipoprotein lipase in chronic lymphocytic leukemia patients Genomic Alterations in Patients with Unmutated IGHV Genes.
with mutated and unmutated IGHV genes. Leuk Lymphoma Blood 2008;112:3133.
2007;48:S47. 64. Moshynska O, Sankaran K, Pahwa P, Saxena A. Prognostic sig-
46. Josefsson P, Geisler CH, Leffers H, Petersen JH, Andersen MK, nificance of a short sequence insertion in the MCL-1 promoter
Jurlander J, et al. CLLU1 expression analysis adds prognostic in chronic lymphocytic leukemia. J Nat Cancer Inst 2004;96:
information to risk prediction in chronic lymphocytic 673-82.
leukemia. Blood 2007;109:4973-9. 65. Nuckel H, Frey UH, Bau M, Sellmann L, Stanelle J, Durig J, et
47. Pekarsky Y, Palamarchuk A, Maximov V, Efanov A, Nazaryan al. Association of a novel regulatory polymorphism (-938C>A)
N, Santanam U, et al. Tcl1 functions as a transcriptional regula- in the BCL2 gene promoter with disease progression and sur-
tor and is directly involved in the pathogenesis of CLL. Proc vival in chronic lymphocytic leukemia. Blood 2007;109:290-7.
Nat Acad Sci USA 2008;105:19643-8. 66. Saxena A, Moshynska O, Sankaran K, Viswanathan S,
48. Herling M, Patel KA, Khalili J, Schlette E, Kobayashi R, Sheridan DP. Association of a novel single nucleotide polymor-
Medeiros LJ, et al. TCL1 shows a regulated expression pattern phism, G(-248)A, in the 5'-UTR of BAX gene in chronic lym-
in chronic lymphocytic leukemia that correlates with molecu- phocytic leukemia with disease progression and treatment
lar subtypes and proliferative state. Leukemia 2006;20:280-5. resistance. Cancer Lett 2002;187:199-205.
49. Mansouri M, Åleskog A, Sevov M, Tobin G, Rosenquist R. 67. Thunberg U, Tobin G, Johnson A, Soderberg O, Padyukov L,
TCL1 expression in CLL patients with IGHV3-21 usage resem- Hultdin M, et al. Polymorphism in the P2X7 receptor gene and
bles the expression pattern of unmutated CLL patients. Leuk survival in chronic lymphocytic leukaemia. Lancet 2002;360:
Lymphoma 2007;48:S48. 1935-9.
50. Calin GA, Ferracin M, Cimmino A, Di Leva G, Shimizu M, 68. Kaderi MA, Murray F, Jansson M, Merup M, Karlsson K, Roos
Wojcik SE, et al. A MicroRNA signature associated with prog- G, et al. The GNAS1 T393C polymorphism and lack of clinical
nosis and progression in chronic lymphocytic leukemia. N Engl prognostic value in chronic lymphocytic leukemia. Leuk Res
J Med 2005;353:1793-801. 2008;32:984-87.
51. Stamatopoulos B, Meuleman N, Haibe-Kains B, Saussoy P, Van 69. Kaderi MA, Norberg M, Murray F, Merup M, Sundstrom C,
den Neste E, Michaux L, et al. MicroRNA-29c and microRNA- Roos G, et al. The BCL2 promoter (-938C>A) polymorphism
223 downregulation has in vivo significance in chronic lym- does not predict clinical outcome in chronic lymphocytic
phocytic leukemia and improves disease risk stratification. leukemia. Leukemia 2008;22:339-43.
Blood 2009. 70. Skogsberg Å, Tobin G, Thunberg U, Åleskog A, Karlsson K,
52. Boyd K, Dearden CE. Alemtuzumab in the treatment of chron- Laurell A, et al. The G(-248)A polymorphism in the promoter
ic lymphocytic lymphoma. Expert Rev Anticancer Ther 2008; region of the Bax gene does not correlate with prognostic
8:525-33. markers or overall survival in chronic lymphocytic leukaemia.
53. Hallek M, Cheson BD, Catovsky D, Caligaris-Cappio F, Leukemia 2006;20:77-81.
Dighiero G, Dohner H, et al. Guidelines for the diagnosis and 71. Tobin G, Skogsberg Å, Thunberg U, Laurell A, Åleskog A,
treatment of chronic lymphocytic leukemia: a report from the Merup M, et al. Mcl-1 gene promoter insertions do not corre-
International Workshop on Chronic Lymphocytic Leukemia late with disease outcome, stage or VH gene mutation status in
updating the National Cancer Institute-Working Group 1996 chronic lymphocytic leukemia. Leukemia 2005;19:871-873.
guidelines. Blood 2008;111:5446-56. 72. Gryshchenko I, Hofbauer S, Stoecher M, Daniel PT, Steurer M,
54. Stilgenbauer S, Sander S, Bullinger L, Benner A, Leupolt E, Gaiger A, et al. MDM2 SNP309 is associated with poor out-
Winkler D, et al. Clonal evolution in chronic lymphocytic come in B-cell chronic lymphocytic leukemia. J Clin Oncol
leukemia: acquisition of high-risk genomic aberrations associ- 2008;26:2252-7.
ated with unmutated VH, resistance to therapy, and short sur- 73. Zenz T, Habe S, Benner A, Kienle D, Dohner H, Stilgenbauer S.
vival. Haematologica 2007;92:1242-5. The MDM2 -309 T/G promoter single nucleotide polymor-
55. Dicker F, Herholz H, Schnittger S, Nakao A, Patten N, Wu L, et phism does not alter disease characteristics in chronic lympho-
al. The detection of TP53 mutations in chronic lymphocytic cytic leukemia. Haematologica 2008;93:1111-3.
leukemia independently predicts rapid disease progression and 74. Hultdin M, Rosenquist R, Thunberg U, Tobin G, Norrback KF,
is highly correlated with a complex aberrant karyotype. Johnson A, et al. Association between telomere length and
Leukemia 2009;23:117-24. V(H) gene mutation status in chronic lymphocytic leukaemia:
56. Carter A, Lin K, Sherrington PD, Pettitt AR. Detection of p53 clinical and biological implications. Br J Cancer 2003;88:593-8.
dysfunction by flow cytometry in chronic lymphocytic 75. Ricca I, Rocci A, Drandi D, Francese R, Compagno M, Lobetti
leukaemia. Br J Haematol 2004;127:425-8. Bodoni C, et al. Telomere length identifies two different prog-
57. Dicker F, Schnittger S, Haferlach T, Kern W, Schoch C. nostic subgroups among VH-unmutated B-cell chronic lym-
Immunostimulatory oligonucleotide-induced metaphase cyto- phocytic leukemia patients. Leukemia 2007;21:697-705.
genetics detect chromosomal aberrations in 80% of CLL 76. Roos G, Krober A, Grabowski P, Kienle D, Buhler A, Dohner H,
patients: A study of 132 CLL cases with correlation to FISH, et al. Short telomeres are associated with genetic complexity,
IgVH status, and CD38 expression. Blood 2006;108:3152-60. high-risk genomic aberrations, and short survival in chronic
58. Mayr C, Speicher MR, Kofler DM, Buhmann R, Strehl J, Busch lymphocytic leukemia. Blood 2008;111:2246-52.
R, et al. Chromosomal translocations are associated with poor 77. Damle RN, Batliwalla FM, Ghiotto F, Valetto A, Albesiano E,
prognosis in chronic lymphocytic leukemia. Blood 2006;107: Sison C, et al. Telomere length and telomerase activity delin-
742-51. eate distinctive replicative features of the B-CLL subgroups
59. Haferlach C, Dicker F, Schnittger S, Kern W, Haferlach T. defined by immunoglobulin V gene mutations. Blood 2004;
Comprehensive genetic characterization of CLL: a study on 103:375-82.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 67 |
Chronic lymphocytic leukemia

Risk adapted chronic lymphocytic


leukemia management

P. Hillmen A B S T R A C T

The understanding of chronic lymphocytic leukemia (CLL) and the treatment of patients with this
Leeds General Infirmary, Leeds, disease has changed dramatically over the last 10 years. There are now more effective therapies result-
United Kingdom
ing in higher remission rates with prolonged progression free survival. In addition, the use of reduced
intensity conditioning allogeneic stem cell transplantation as a potentially curative procedure is being
more widely applied for selected patients. This has resulted in the development of risk-adapted ther-
Hematology Education: apy in CLL. We are beginning to predict which patients will respond to which therapies (for example,
the education program for the patients with p53 deletion will not respond well to purine analogues or conventional chemotherapy
annual congress of the European
but appear to respond well to alemtuzumab), and to tailor therapies to an individual patient’s disease
Hematology Association
characteristics, as well as according to their other comorbidities and their response to therapy.
2009;3:68-71 Intensive potentially curative allogeneic stem cell transplantation is being used in patients with poor
prognostic CLL at an earlier time-point in their disease. In this article, the current approaches to
patient directed therapies will be reviewed.

ver the last decade, our understand- Epidemiology and End Results) Registry is

O ing of chronic lymphocytic


leukemia (CLL) has changed dra-
matically. We now have a clearer under-
72 years (http://seer.cancer.gov/statfacts/
html/clyl.html). At this age, many individu-
als have comorbidities (mean of 3.6 comor-
standing of the pathophysiology of the dis- bid conditions between 65 and 74 years
ease including the dynamics of the CLL old), and these will impact the patient’s
clone,1 its molecular biology,2 the impor- ability to tolerate more intensive
tance of the interaction between the CLL chemotherapeutic regimes. In addition, cer-
cell, and its micro-environment and critical tain drugs, such as fludarabine, are cleared
role of antigen stimulation in the develop- by the kidneys, and therefore, specific
ment of CLL in some patients.3 In addition, comorbidities, such as renal dysfunction
the therapeutic options in CLL have devel- will directly contraindicate the use of flu-
oped considerably over the last decade darabine. Therefore, therapy in CLL needs
with the advent of combination immuno- to be adapted to the individual patient’s fit-
chemotherapy.4-6 Previously, when single ness. Thus, there is no single therapy that is
agent alkylating agents or purine analogues currently the “gold standard” standard in
were used but this is now being superseded CLL, as it varies depending on the patient’s
by the use of monoclonal antibodies alone fitness.
(alemtuzumab7) or in combination with
chemotherapy (rituximab8) and with an Pathophysiology of chronic lymphocytic
increasing and selective use of reduced leukemia
intensity conditioning allogeneic stem cell It appears that there are at least two bio-
transplant.9,10 We are now witnessing the logical sub-types of CLL, which can be dif-
introduction of novel agents, such as ferentiated by the presence or absence of
lenalidomide.11,12 Largely due to the avail- somatic mutations in the immunoglobulin
ability of more effective therapies, assays gene of the CLL clone.15,16 A proportion of
that can detect extremely low levels of CLL patients with mutated CLL presenting with
have been developed in order to assess the Binet Stage A disease are destined to
presence of detectable minimal residual dis- remain stable and never to require therapy.
ease (MRD) to as low as a single CLL cell in In contrast, almost all patients with unmu-
10,000 leucocytes.13,14 This rapid increase in tated CLL will need treatment at presenta-
our understanding of the disease and in the tion or a short time after presentation. It
therapeutic agents and end-points alters our could be considered that patients with “VH
approach to the treatment of patients with mutated” CLL have genetically “stable” dis-
CLL and the era of risk-adapted therapy is ease, whereas those with “VH unmutated”
now with us. CLL have a genetically “unstable” disease,
as patients with a genetic evolution, such as
Factors influencing risk-adapted therapy deletion of the p53 oncogene, usually have
Patient comorbidity unmutated CLL (Figure 1). However,
The median age at diagnosis of CLL in although the outcomes for patients with
North America on the SEER (Surveillance unmutated CLL after their initial therapy

| 68 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

are inferior to those with mutated CLL, there is no evi-


dence that stratifying patients to different therapies
according to their mutational status is beneficial. A key
unanswered question is whether patients with poor
risk CLL should receive treatment when they present
with Stage A disease before their CLL progresses. The
data that supports a “watch and wait” strategy in CLL
is based on large randomised trials in unselected early
stage patients,17-20 some of whom would never have
progressed, and using relatively ineffective therapies,
such as monotherapy with alkylating agents. At pres-
ent, the German and French CLL Study Groups are
participating in a large randomised trial comparing
early treatment with FCR against “watch and wait”
(the CLL7 trial) in patients with biologically poor-risk
Stage A CLL who would not conventionally receive
therapy. This trial, which is recruiting well, will prob-
ably provide an answer to this important question in Figure 1. The prognostic markers in CLL indicate that there
the near future. An alternative approach maybe to use are at least two sub-types of the disease. These are simi-
a therapy that does not have the potential to damage lar at an immunophenotypic level, which is used to estab-
DNA, and thus, result in genetic evolution of a “genet- lish the diagnosis. A minority of patients requiring therapy
have what might be considered “genetically stable” CLL,
ically unstable” CLL clone. Such therapies may include whereas the majority have “genetically unstable” CLL. This
monoclonal antibodies as single agents or agents, such difference may be defined by the mutational status of the
as lenalidomide. Trials with these agents in early stage immunoglobulin genes and the presence or absence of
other poor prognostic features. Both groups have high
poor risk CLL are being considered. response rates to initial combination chemotherapy, but
VH mutational status is an integral part of the CLL patients with “genetically stable” CLL rarely develop p53
clone that is a marker of the transformed original cell pathway abnormalities, or resistant disease at relapse in
contrast to “genetically unstable” CLL, in which evolution to
and will not alter throughout the duration of a resistant disease is common at relapse. A minority of
patient’s disease. In contrast, patients can acquire patients present with p53 pathway abnormalities and are
genetic abnormalities as part of the evolution of their inherently resistant to conventional chemotherapy.
disease. The best-characterized abnormality that can
evolve is the loss or mutation of the p53 oncogene.
This is usually identified by the loss of the p53 locus or
deletion of the short arm of chromosome 17 (17p dele-
tion).21 In most patients with 17p deletion, the p53 However, in patients with very large lymph nodes
gene on the other chromosome 17 has a mutation, and (individual nodes greater than 5 cm in diameter), the
therefore, there is no active normal p53 in the CLL cell. response rates to alemtuzumab are low,7 with very few
The chemotherapeutic agents conventionally used in patients achieving a complete remission. In these
CLL, either alkylating agents or purine analogues, patients, it is worth considering the use of therapies to
damage DNA (or prevent its repair), leading to apopto- reduce the size of the lymph nodes, such as high dose
sis or cell cycle arrest via the p53 pathway. Thus, CLL methyl prednisolone,22 followed by alemtuzumab.
cells with a dysfunctional p53 pathway cannot The deletion of 17p is the worst biological prognos-
respond in a “normal” way to chemotherapy in that tic marker but is relatively uncommon in patients
they are unable to undergo apoptosis or cell cycle when they initially require therapy. In contrast, the
arrest. This means that the CLL cell suffers DNA dam- deletion of the long arm of chromosome 11 (11q del)
age but is unable to die – clearly an inherently danger- occurs in as many as 20% of patients prior to their ini-
ous situation. It is entirely plausible that patients with tial therapy and is also a poor prognostic marker.4,23 It
p53 dysfunction are not only resistant to conventional is thought that the loss of the ataxia telangiectasia
chemotherapy, but that when they are exposed to mutated (ATM) gene from chromosome 11 results in
DNA damaging chemotherapy, their CLL cells develop the poor prognosis for these patients, who frequently
mutations in critical oncogenes and tumour suppressor present with bulky lymphadenopathy. The initial
genes. If true, this would result in the chemotherapeu- reports of the use of FCR from the front-line German
tic insult, leading to the development of more prolifer- CLL8 Trial suggest that the beneficial effects of the
ative or resistant sub-clones, and thus, a poorer prog- addition of rituximab to FC are particularly marked for
nosis. Therefore, it is sensible that conventional patients with 11q del and that these patients may even
chemotherapy should be avoided in patients with CLL show a survival advantage with FCR.24,25
with a deletion p53 and agents that do not depend on
the p53 pathway for their function should be Response to adapt therapy
employed. These agents include monoclonal antibod- It is becoming increasingly evident that remission
ies, particularly alemtuzumab, and steroids. Trials are duration, treatment-free survival and overall survival is
ongoing to study the combination of monoclonal anti- dependent on the depth of remission. Patients who
bodies and steroids in p53 deleted CLL. Now, the stan- achieve a complete remission by IWCLL Criteria26
dard therapy for these patients who cannot be recruit- have a better survival than those who do not. Even
ed into clinical trials should be alemtuzumab. more evident is that patients who have no detectable

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 69 |
14th Congress of the European Hematology Association

CLL at the end of therapy, those who are minimal porting the use of rituximab in addition to any other
residual disease (MRD) negative, have a far superior chemotherapeutic combination except for FC.
survival independent of the therapy used to achieve
MRD negativity.23 This means that the response to Patients with deletion of chromosome 17
therapy is an important factor affecting further treat- It is clear that fludarabine-based combination chemo-
ment. For example, there are now many ongoing stud- therapy is ineffective, and potentially deleterious, for
ies testing the use of alemtuzumab and other therapies patients with p53 dysfunction. In these patients, alem-
as consolidation following conventional immuno- tuzumab, either alone29,30 or in combination.31 In addi-
chemotherapy in order to eradicate detectable MRD. tion, patients with 17p deletion should be considered
Also, many allogeneic stem cell regimes include the for an allogeneic stem cell transplant; preferably, when
use of donor lymphocyte infusions in patients who they attain a remission.9 This is because even if patients
have detectable MRD post-transplant. Therefore, we reach a good remission, these tend to be short-lived
are now risk-adapting therapy according to MRD. and then the re-induction of a further remission may
not be possible. In addition, there is good evidence that
Optimal therapy for chronic lymphocytic leukemia the graft-versus-leukemia is as effective against 17p-
Immunochemotherapy deleted CLL compared to other cases, and the outcome
Three large randomised Phase III trials have previ- from allogeneic stem cell transplantation appears to be
ously demonstrated that the combination of fludara- equally good.
bine plus cyclophosphamide (FC) is superior to single
agent therapy with fludarabine or chlorambucil.4,5,6 MRD status
However, the patients in these trials were either young Patients who are MRD negative at the end of thera-
and/or were considered fit enough to tolerate fludara- py have a better survival than those who have
bine-based combination therapy. Thus, the logical detectable disease.7,13 This raises the question whether
backbone chemotherapy with which to combine other consolidation therapy with the aim of eradicating
therapies, such as monoclonal antibodies, is FC. MRD post-chemotherapy is a reasonable approach. At
Rituximab was added to FC (FCR) by the group at the present, there are a number of trials looking at the use
MD Anderson Cancer Center.8,27 When compared to of a variety of agents, such as alemtuzumab or lenalido-
historical controls from the same institution, it mide as consolidation, but to date an effective and safe
appeared that the addition of rituximab almost dou- schedule has not been established.
bled the complete remission rate and improved overall
survival. Recently, the results of two randomised Phase Conclusion
III trials comparing FC to FCR have been presented. Over the last decade, there have been huge strides in
One of these trials, the German CLL Study Group our understanding and treatment of CLL. We are now
CLL8 Trial,26 involved over 800 patients with CLL who just entering the era where biological features of a
required therapy by conventional criteria but were patient’s CLL are being used to define the most appro-
previously untreated, and the trial randomly assigned priate therapy. In addition, we are now exploring the
them to receive either FC or FCR. In the GMCLLSG tailoring of therapy to individual patients based on
CLL8 trial, the addition of rituximab to FC doubled the their comorbidity and/or response to previous therapy.
complete response rate (27% for FC compared to 52% It is likely that over the next 5-10 years, the risk adap-
for FCR) and improved the median progression-free tion of therapy in CLL will become the norm.
survival by approximately 10 months, thus meeting
the trial’s primary end-point. In addition, the REACH
Study28 also compared FCR with FC but in patients References
who had previously received one line of therapy.
Again, the REACH study showed a significant advan- 1. Messmer BT, Messmer D, Allen SL, Kolitz JE, Kudalkar P,
Cesar D, et al. In vivo measurements document the dynamic
tage in terms of complete response rate and progres- cellular kinetics of chronic lymphocytic leukemia B cells. J
sion free survival for the addition of rituximab. Clin Invest 2005;115:755-64.
Therefore, we can now consider FCR as the standard 2. Gribben J. Molecular Profiling in CLL. Hematology Am Soc
Hematol Educ Program 2008;444-9.
therapy for younger fitter patients. However, there are 3. Ghia P, Chiorazzi N, Stamatopoulos K. Microenvironmental
situations where FCR is not appropriate: influences in chronic lymphocytic leukaemia: the role of anti-
gen stimulation. J Intern Med 2008;264:549-62.
Elderly patient or those with significant comorbidity 4. Catovsky D, Richards S, Matutes E, Oscier D, Dyer MJ,
Bezares RF, et al. Assessment of fludarabine plus cyclophos-
In many patients, fludarabine and FC are contraindi- phamide for patients with chronic lymphocytic leukaemia
cated due to the intensity of the treatment, renal dys- (the LRF CLL4 Trial): a randomised controlled trial. Lancet
function and/or previous fludarabine-induced autoim- 2007;370:230-9.
5. Eichhorst BF, Busch R, Hopfinger G, Pasold R, Hensel M,
mune hemolysis. In these patients, there is no “gold Steinbrecher C, et al. Fludarabine plus cyclophosphamide ver-
standard” therapy but chlorambucil as a single agent sus fludarabine alone in first-line therapy of younger patients
remains the most frequently used treatment. with chronic lymphocytic leukemia. Blood 2006;107:885-91.
6. Flinn IW, Neuberg DS, Grever MR, Dewald GW, Bennett JM,
Theoretically, the addition of rituximab is likely to be Paietta EM, et al. Phase III trial of fludarabine plus cyclophos-
synergistic with chlorambucil, as appears to be the case phamide compared with fludarabine for patients with previ-
with most chemotherapies in other B-cell malignancies, ously untreated chronic lymphocytic leukemia: US Intergroup
Trial E2997. J Clin Oncol 2007;25:793-8.
such as follicular lymphoma or diffuse large B-cell lym- 7. Moreton P, Kennedy B, Lucas G, Leach M, Rassam SM,
phoma. However, there are no studies to date that Haynes A, et al. Eradication of minimal residual disease in B-
report this combination and no randomised trials sup- cell chronic lymphocytic leukemia after alemtuzumab thera-
py is associated with prolonged survival. J Clin Oncol 2005;

| 70 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

23: 2971-9. 21. Zenz T, Benner A, Döhner H, Stilgenbauer S. Chronic lym-


8. Tam CS, O’Brien S, Wierda W, Kantarjian H, Wen S, Do K, et phocytic leukemia and treatment resistance in cancer: the role
al. Long-term results of the fludarabine, cyclophosphamide, of the p53 pathway. Cell Cycle 2008;7:3810-4.
and rituximab regimen as initial therapy of chronic lympho- 22. Thornton PD, Matutes E, Bosanquet Ag, Lakhani AK, Grech
cytic leukemia. Blood 2008;112:975-980. H, Ropner JE, et al. High dose methyl prednisolone can
9. Dreger P, Corradini P, Kimby E, Michallet M, Milligan D, induce remissions in CLL patients with p53 abnormalities.
Schetelig J, et al. Indications for allogeneic stem cell transplan- Ann Hematol 2003;82:759-65.
tation in chronic lymphocytic leukemia: the EBMT transplant 23. Döhner H, Stilgenbauer S, Benner A, Leupolt E, Kröber A,
consensus. Leukemia 2007;21:12-7. Bullinger L, et al. Genomic aberrations and survival in chron-
10. Schetelig J, van Biezen A, Brand R, Caballero D, Martino R, ic lymphocytic leukemia. N Engl J Med 2000;343:1910-6.
Itala M, et al. Allogeneic hematopoietic stem-cell transplanta- 24. Hallek M, Fingerle-Rowson G, Fink AM, Busch R, Mayer J,
tion for chronic lymphocytic leukemia with 17p deletion: a Hensel M, et al. Immunochemotherapy with Fludarabine (F),
retrospective European Group for Blood and Marrow Cyclophosphamide (C), and Rituximab (R) (FCR) Versus
Transplantation analysis. J Clin Oncol 2008;26:5094-100. Fludarabine and Cyclophosphamide (FC) Improves Response
11. Chanan-Khan A, Miller KC, Musial L, Lawrence D, Padma- Rates and Progression-Free Survival (PFS) of Previously
nabhan S, Takeshita K, et al. Clinical efficacy of lenalidomide Untreated Patients (pts) with Advanced Chronic Lymphocytic
in patients with relapsed or refractory chronic lymphocytic Leukemia (CLL). Blood 2008;112:325.
leukemia: results of a phase II study. J Clin Oncol 2006;24: 25. Stilgenbauer S, Döhner H. Campath-1H-induced complete
5343-9. remission of chronic lymphocytic leukemia despite p53 gene
12. Ferrajoli A, Lee BN, Schlette EJ, O’Brien SM, Gao H, Wen S, mutation and resistance to chemotherapy. N Engl J Med
et al. Lenalidomide induces complete and partial remissions 2002; 347:452-3.
in patients with relapsed and refractory chronic lymphocytic 26. Hallek M, Cheson BD, Catovsky D, Caligaris-Cappio F,
leukemia. Blood 2008;111:5291-7. Dighiero G, Döhner H, et al. Guidelines for the diagnosis and
13. Rawstron AC, Kennedy B, Evans PA, Davies FE, Richards SJ, treatment of chronic lymphocytic leukemia: a report from the
Haynes AP, et al. Quantitation of minimal disease levels in International Workshop on Chronic Lymphocytic Leukemia
chronic lymphocytic leukemia using a sensitive flow cyto- updating the National Cancer Institute–Working Group 1996
metric assay improves the prediction of outcome and can be guidelines. Blood 2008;111:5446-56.
used to optimize therapy. Blood 2001;98:29-35. 27. Keating MJ, O'Brien S, Albitar M, Lerner S, Plunkett W, Giles
14. Rawstron AC, Villamor N, Ritgen M, Böttcher S, Ghia P, F, et al. Early results of a chemoimmunotherapy regimen of
Zehnder JL, et al. International standardized approach for fludarabine, cyclophosphamide, and rituximab as initial ther-
flow cytometric residual disease monitoring in chronic lym- apy for chronic lymphocytic leukemia. J Clin Oncol 2005;23:
phocytic leukaemia. Leukemia 2007;21:956-64. 4079-88.
14. Damle RN, Wasil T, Fais F, Ghiotto F, Valetto A, Allen SL, et 28. Robak T, Moiseev SI, Dmoszynska A, Solal-Céligny P,
al. Ig V Gene Mutation Status and CD38 Expression As Novel Warzocha K, Loscertales J, et al. Rituximab, Fludarabine, and
Prognostic Indicators in Chronic Lymphocytic Leukemia. Cyclophosphamide (R-FC) Prolongs Progression Free Survival
Blood 1999;94:1840-7. in Relapsed or Refractory Chronic Lymphocytic Leukemia
16. Unmutated Ig VH Genes Are Associated With a More (CLL) Compared with FC Alone: Final Results from the
Aggressive Form of Chronic Lymphocytic Leukemia. Blood International Randomized Phase III REACH Trial. Blood
1999;94:1848-54. 2008; 112: lba-1.
17. Dighiero G, Maloum K, Desablens B, Cazin B, Navarro M, 29. Stilgenbauer S, Zenz T, Winkler D, Bühler A, Busch R, Fin-
Leblay R, et al. Chlorambucil in indolent chronic lymphocyt- gerle-Rowson G, et al. Genomic aberrations, VH mutation
ic leukemia. N Engl J Med. 1998;338:1506-14. status and outcome after fludarabine and cyclophosphamide
18. Shustik C, Mick R, Silver R, Sawitsky A, Rai K, Shapiro L. (FC) or FC plus rituximab (FCR) in the CLL8 trial. Blood 2008;
Treatment of early chronic lymphocytic leukemia: intermit- 112:781.
tent chlorambucil versus observation. Hematol Oncol 30. Lozanski G, Heerema NA, Flinn IW, Smith L, Harbison J,
1988;6: 7-12. Webb J, et al. Alemtuzumab is an effective therapy for chron-
19. Montserrat E, Fontanillas M, Estape J, for the Spanish PETHE- ic lymphocytic leukemia with p53 mutations and deletions.
MA Group. Chronic lymphocytic leukemia treatment: an Blood 2004;103:3278-81.
interim report of PETHEMA trials. Leuk Lymphoma 1991; 31. Pettitt AR, Matutes E, Oscier D. Alemtuzumab in combina-
5:89-92. tion with high-dose methylprednisolone is a logical, feasible
20. CLL Trialists’ Collaborative Group. Chemotherapeutic and highly active therapeutic regimen in chronic lymphocyt-
options in chronic lymphocytic leukemia: a meta-analysis of ic leukaemia patients with p53 defects. Leukemia 2006;20:
the randomized trials. J Natl Cancer Inst 1999;91:861-8. 1441-5.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 71 |
Chronic myeloid leukemia

Stem cell persistence in chronic myeloid leukemia

M.W. Deininger Clinical and molecular features of chronic


myeloid leukemia positive cells, referred to as clonal evolution,
are rare in newly diagnosed chronic phase
Oregon Health & Science University,
Portland, OR, USA Chronic myeloid leukemia (CML) is char- patients.5 Moreover, polyclonal hemato-
acterized by a 2-phase clinical course. In the poiesis is restored in most patients with a
developed world, most patients are diag- complete cytogenetic response (CCyR) to
nosed in the chronic phase, which is charac- imatinib.6 Lastly, bone marrow infected with
Hematology Education: terized by grossly increased output of BCR-ABL retrovirus causes a polyclonal
the education program for the
annual congress of the European myeloid cells, whose differentiation and CML-like myeloproliferative disease upon
Hematology Association function is largely maintained.1 Historical transplantation into lethally irradiated syn-
data suggest that the median duration of the geneic recipients, consistent with a single hit
2009;3:72-77 chronic phase, in the absence of any effec- pathogenesis.7 What these data cannot
tive therapy, is 2-3 years, after which there is exclude is that in human CML, a mutation
a progressive loss of cellular differentiation may be acquired before BCR-ABL, but with-
that culminates in the blastic phase, an acute out causing clonal expansion. This notion
leukemia of myeloid or lymphoid pheno- has attracted renewed interest from observa-
type. While complications in the chronic tions in Ph-negative JAK2V617F-positive MPD,
phase are rare, blast crisis is a rapidly fatal where acute leukemia may arise from
disease. Thus, the foremost aim of CML JAK2WT cells, consistent with a JAK2V617F-neg-
therapy is to prevent the progression from ative abnormal cell at increased risk of trans-
the chronic to the blastic phase. formation to acute leukemia.8 Moreover, the
The cytogenetic hallmark of CML is the clinical course of newly diagnosed patients
Philadelphia chromosome (Ph), the conse- with seemingly exchangeable disease pre-
quence of the t(9;22)(q34;q11). At the molec- sentations can be very different, indicating a
ular level, the translocation leads to the level of disease heterogeneity that is not
fusion of BCR genetic sequences upstream appreciated by the resolution of morpholo-
of the ABL gene on chromosome 9. The gy and cytogenetics.9
resulting BCR-ABL fusion protein is that
activates multiple signaling pathways, Cell of origin
including mitogen activated protein kinases Studies using infection of immunopheno-
(MAPK), phosphotidyl inositol 3’ kinase typically and functionally defined murine
(PI3K) and Janus kinase/signal transduction hematopoietic progenitor and stem cells have
and activation of transcription (JAK/STAT) revealed that BCR-ABL does not confer self
signaling, amongst many others. renewal capacity to progenitor cells, unlike
Collectively, these pathways increase prolif- AML-related fusion genes, such as MOZ-
eration, inhibit apoptosis, induce genetic TIF2.10 This implies that the BCR-ABL translo-
instability and perturb the interaction cation must be acquired by a hematopoietic
between the CML cells and their microenvi- stem cell that is able to self-renew. In support
ronment. The identification of non-redun- of this, BCR-ABL has been demonstrated in all
dant components in this complex signaling hematopoietic lineages and in lineage-nega-
network has proved difficult, consistent tive CD34+/CD38– cells.11-12 For mechanisms
with a high degree of redundancy. However, that are not well understood, cellular expan-
it is clear that the tyrosine kinase activity of sion is, however, limited to the myeloid
BCR-ABL is conditio sine qua non of leuke- compartment, particularly granulocytes and
mogenesis.2 their precursors. Interestingly, the degree to
It has been a matter of debate whether which the “stem cell” compartment is pene-
BCR-ABL is sufficient to induce the chronic trated by the BCR-ABL-positive cell clone is
phase of CML. Early studies based on glu- extremely variable.12 Although precise data
cose 6 phosphate dehydrogenase (G6PD) are unavailable, it is believed that a more
isoenzyme expression in EBV-transformed complete replacement of the stem cell com-
Ph-negative B cell lines from CML patients partment with leukemic cells is a feature of
had suggested that a clonal state predates long-standing disease. From a clinical view,
the acquisition of Ph.3-4 Over the years, sub- the paucity of residual normal cells present
stantial evidence has accumulated in support in the marrow of patients with CML of
of the notion that chronic phase CML does many years’ duration may be responsible for
not require mutations in addition to Ph. the frequent cytopenias of such patients on
Thus, chromosomal abnormalities in Ph- imatinib therapy. From the fact that CML

| 72 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

originates in a hematopoietic stem cell, we may deduce term culture initiating cell (LTC-IC) assays on irradiated
that the organization of the leukemia follows the same human or murine stromal cells. While these cultures are
hierarchical structure as normal hematopoiesis. This relatively easy to standardize, they favor the outgrowth
would imply that acquisition of Ph by a cell further of normal over CML progenitor and stem cells; a fact
down in the hierarchy, and without self-renewal capac- that formed the basis for in vitro purging strategies.20-21
ity, will not produce disease. This would explain, why This is in obvious contrast to the human disease, and
BCR-ABL transcripts are detectable at very low level in clearly indicates that the assay system impacts on the
many healthy individuals, most of whom will never parameter it intends to measure, much in the sense of
develop CML.13 In contrast to the chronic phase, there is Heisenberg’s paradigm in physics. Thus, for the time
evidence that myeloid blast crisis may originate in a being, the study of CML stem cells is limited by the
granulocyte macrophage progenitor (GMP) that availability of accurate assays that do not affect func-
acquires self-renewal capacity through activation of β- tional properties, and all available data need to be inter-
catenin.14-15 This implies that there may be many more preted with caution.
leukemia stem cells at the time of blastic transformation
compared to chronic phase, which may explain the Residual disease in patients treated with BCR-ABL
treatment-refractory nature of blastic phase. inhibitors
Between 80-90% of patients with newly diagnosed
The difficulty of eradicating the Ph+ cell clone with chronic phase CML will achieve CCyR on therapy with
chemotherapy imatinib, with considerably lower rates in patients with
The notion that BCR-ABL is acquired by a hemato- advanced CML.22-23 However, residual disease remains
poietic stem cell fits very well to the old clinical obser- detectable in the majority of patients with a CCyR.24
vation that it is virtually impossible to eradicate CML Since recurrence is almost inevitable in these individu-
with chemotherapy. Thus, even with intensive multia- als, the current recommendation is to continue therapy
gent regimens, Ph-negativity (as assessed by cytogenet- indefinitely. This clinical observation corroborates labo-
ics) is only transient; suggesting that killing the Ph-pos- ratory data showing persistence of BCR-ABL expressing
itive cell clone would require killing the entire clonogenic cells.25-26 Only a minority of patients become
hematopoietic system. The only way to accomplish this “PCR undetectable” on imatinib treatment, a state
is myeloablative regimens used in allogeneic stem cell referred to as a complete molecular response (CMR). In
transplantation. While there is no doubt that a T cell anecdotal reports, a subset of these patients has main-
dependent graft versus leukemia effect mediates the tained responses even after discontinuation of ima-
potent graft versus leukemia effect, the contribution of tinib,27 and several clinical trials are currently underway
the conditioning regimen should not be underestimat- to investigate this question in a systematic fashion.
ed.16 Otherwise, it would be difficult to explain the sus- Interestingly, most recurrences occurred rather early
tained responses in some recipients of identical twin after discontinuation of imatinib, while the recurrence
allografts.17 On the other hand, a “PCR undetectable” free survival curves seem to stabilize over time. While
state is not equivalent to the absence of residual very preliminary, these data are intriguing and suggest
leukemia and even years post allograft, there continues that a subset of patients may be ‘curable’ with imatinib.
to be a small but not negligible risk of relapse.18 These What we currently do not know is whether the disease
data are in contrast to patients with acute leukemia, burden in these patients is zero, or whether they contin-
where a substantial subset of patients is curable with ue to harbor residual leukemia that is below the detec-
non-myeloablative chemotherapy regimens based on tion limit of the clinical BCR-ABL assays. Reassuringly,
cytarabine and anthracyclins. This suggests that AML in complete molecular responses were readily achieved
long-term responders may have originated at the level upon re-challenge with imatinib.
of a hematopoietic progenitor rather than a stem cell. Mathematical modeling has been applied to explain
CML cells, on the other hand, are likely to command the dynamics of BCR-ABL mRNA in patients on ima-
the vast armentarium of defenses that allow normal tinib, and to predict the drug’s ability or inability to
stem cells to weather the toxic stress of a lifetime. eliminate the disease. One study found a biphasic
course, where a rapid reduction of leukemia burden is
Chronic myeloid leukemia stem cells followed by a slow decline.28 The first phase is thought
The major obstacle to a better understanding of CML to reflect the killing of differentiating cells, while the
stem cells is the lack of distinct phenotypic and precise second is thought to mirror the reduction of leukemic
functional markers. Studies by the Vancouver group progenitor cells. This model has concluded that eradica-
have identified CML stem cells in the lineage- tion of the CML clone is impossible during the lifetime
/CD34+/CD38– cell population of CML cells, based on of the host. In contrast, another mathematical model
their ability to engraft immunodeficient mice. came to a different conclusion, suggesting that imatinib
Importantly, engraftment and serial transplantation has the general potential to eradicate the Ph-positive cell
capacity includes the quiescent (non-cycling) cells with- clone and that agents stimulating cell cycle entry may
in this population.19 Despite these stem cell features, it accelerate this process.29
should be noticed that enraftment is low, and the mice
do not develop leukemia. Yet, no surface marker has Mechanisms of chronic myeloid leukemia stem cell
been identified that would distinguish CML stem cell resistance to tyrosine kinase inhibitors
from their normal counterparts, preventing the isolation The fact that almost all patients achieve a complete
of such cells for prospective studies. The other widely hematologic response and most a complete cytogenetic
used system to study stem and progenitor cells are long response indicates that differentiating CML cells are

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 73 |
14th Congress of the European Hematology Association

dependent on BCR-ABL kinase activity, an observation BCR-ABL is active in CML stem cells treated with tyro-
that is well supported by in vitro data. Conceptually, sine kinase inhibitors. Studies from different laborato-
another possible explanation is that the kinase inactive ries have come to discordant conclusions, probably
BCR-ABL generated by imatinib is toxic to these cells. reflecting the difficulty of acutely measuring BCR-ABL
In contrast, CML stem cells are apparently much more activity in the relevant cells.31-33 In our own experience,
resistant to imatinib. A number of mechanisms have FACS using antibodies directed against phosphorylated
been proposed the ability of these cells to withstand CrkL (pCrkL) or pan-phosphotyrosine requires
imatinib (Figure 1). In vitro studies by the Glasgow group extremely stringent controls to rule out non-specific
have suggested that quiescent CML cells are resistant to binding, and immunoblot analysis is difficult due to the
imatinib and also the more potent BCR-ABL inhibitors usually small cell numbers. However, from a therapeu-
nilotinib and dasatinib, suggesting that neither of these tic perspective, there is no more important question to
agents has activity against the most critical cell popula- answer. If BCR-ABL is not inhibited, then efforts need
tion.30-32 One has to concede, however, that quiescence to be directed at improving BCR-ABL inhibition, by
is a functional state rather than a mechanistic explana- developing yet more potent inhibitors or by overcoming
tion for resistance. Furthermore, we cannot be certain drug transport mechanisms that prevent achieving a
that the quiescent lineage-/CD34+/CD38– cells isolated critical threshold of BCR-ABL inhibition in the relevant
from newly diagnosed patients are identical to the cells cells. Conversely, if imatinib (and the more potent sec-
that survive in vivo in patients treated with imatinib, ond-line inhibitors) suppress BCR-ABL in the most
and proof is lacking that they exhibit the functional primitive CML cells, and these cells still survive in cul-
characteristics of stem cells; that is, the ability to engraft ture despite BCR-ABL inhibition, this would indicate
immunodeficient mice and transplantability in serial that CML stem cells are not addicted to BCR-ABL and
transplantation assays. Other explanations for the TKI that disease persistence is not a BCR-ABL dependent
resistance of CML stem cells are their much higher lev- process. If so, then biochemically targeting BCR-ABL
els of BCR-ABL in comparison to more differentiated will not suffice to annihilate the Ph+ cell clone, despite
progenitor cells.33 This would be similar to overt resist- the profound suppression of CML progenitor cells, and
ance caused by BCR-ABL gene amplification or disease eradication will require a fundamentally differ-
increased mRNA expression. Furthermore, one study ent approach. At this point, it seems that more evidence
found a high frequency of kinase domain mutations in is accumulating to support the second possibility.39
patients with CCyR, a finding that was, however, not Perhaps this should not come as a surprise. If we consid-
confirmed in subsequent studies and may be due to a er how close chronic phase CML cells are to their nor-
particularly poor risk population with a high risk of mal counterparts in terms of function – there are just too
relapse.34-35 Lastly, drug transport proteins may decrease many of them. Shutting off BCR-ABL may be expected
intracellular imatinib levels below a critical threshold. to reverse them to normality rather than kill them, and
This could involve drug resistance proteins, such as Pgp, one may speculate that this may be particularly true for
but also hOCT-1, a cation pump that transports ima- the most primitive cells. Thus, CML stem cells, when
tinib into the cells. Several studies have shown correla- confronted with BCR-ABL inhibition, may be able to
tions between high levels of hOCT-1 expression or utilize the same physiological growth and survival path-
function and cytogenetic response.36-38 However, infor- ways as normal cells. On the other hand, we could hope
mation on hOCT-1 expression in the most primitive lin- that BCR-ABL-inhibited CML stem cells will never be
eage-/CD34+/CD38– cells is currently lacking. quite like normal cells, since they harbor a large, kinase-
Despite all these data, it seems that the critical ques- dead protein that is lacking from their normal equiva-
tion has not yet been resolved, namely whether or not lents and may interfere with physiological signaling.

Figure 1. Mechanisms
potentially involved in
the resistance of CML
stem cell to BCR-ABL
kinase inhibitors. FN -
fibronectin.

| 74 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

Some evidence for this comes from studies in cell lines al pharmaceutical companies.
transformed to cytokine-independence because of BCR- Another molecule implicated in CML stem cell sur-
ABL expression. While many of these lines can be res- vival is PML, the fusion partner of the retinoic acid
cued from imatinib-induced apoptosis by exogenous receptor α (RARA) in patients with acute promyelocyt-
growth factors, the rescue is incomplete in others, such ic leukemia and the t(15;17).45 Interestingly, targeting
as BCR-ABL expressing Mo7e cells.40 This raises two PML may be possible using Arsenic trioxide, an agent
questions. First, can we intercept such exogenous sur- with a long history in the treatment of CML and an
vival factors to target CML stem cells over normal stem approved drug. As a note of caution though, trials com-
cells? Secondly, are there other, cell-intrinsic pathways bining imatinib with As2O3 in patients who had failed
that are uniquely relevant to the survival or self-renew- imatinib showed rather limited success, and this
al of CML stem cells, but not to normal cells? approach is not being pursued further at the current
time. Whether targeting any of these pathways will
Pathways relevant to chronic myeloid leukemia stem cell eradicate the Ph+ stem cells remains to be seen in clini-
survival cal studies, but they clearly provide a therapeutic ration-
Studies into the pathways critical to CML stem cell ale. What frequently remains unaddressed in the murine
have mainly relied on murine models, either transgenic studies is the question to which extent the activation of
models or the standard transduction/transplantation these pathways in CML cells is dependent on BCR-ABL
model. The implicated pathways or molecules include or not. This will obviously decide whether their inhibi-
Wnt/β-catenin, Hedgehog and PML. tion will add to the effects of BCR-ABL inhibitors, and
β-catenin is a multifunctional protein with distinct whether they should be used simultaneously or sequen-
roles in cell adhesion and as a transcriptional co-activa- tially.
tor. β-catenin transcriptional activity is negatively regu-
lated through proteasomal degradation that requires Empirical approaches to target chronic myeloid leukemia
phosphorylation by GSK3β. Binding of Wnt proteins to stem cells
their cognate surface receptors inhibits GSK3β and The Glasgow group has tested a number of drugs and
induces β-catenin stabilization, nuclear translocation compounds for their ability to reduce the quiescent lin-
and activation of target genes. Many solid tumors eage-/CD34+/CD38– fraction of cells that survive the in
exhibit constitutive β-catenin activity because of muta- vitro exposure to imatinib.46 The only agent with this
tions in negative regulators, such as APC and axin, or in ability was farnesyl transferase inhibitors (FTI), which
β-catenin itself. In hematopoietic stem cells, β-catenin is on its own, and in combination with imatinib, had
required for self-renewal.41 Thus, there is more rapid modest clinical efficacy in patients with active CML.
exhaustion of hematopoietic stem cells in serial trans- Building on these data, they then tested BMS214662, an
plantation assays of mice lacking β-catenin. As noted FTI with previously described activity against quiescent
above, active (nuclear) β-catenin was shown in granulo- cells, for its ability to inhibit primitive quiescent CML
cyte-macrophage progenitor cells from CML patients in cells alone or in combination with imatinib. These stud-
myeloid blastic phase, but not chronic phase, suggesting ies showed a significant reduction of BCR-ABL-positive
that this may underlie the acquisition of self-renewal LTC-IC compared to imatinib alone in short-term liquid
capacity by this cell population.15 Conditional knockout culture assays.47 Interestingly, the effects of BMS214662
of β-catenin in transgenic mice attenuated CML-like do not seem to depend on its FTI activity, but involve
MPD.41 The mechanism of β-catenin activation in another yet unknown pathway. Unfortunately, for rea-
human CML has not been defined conclusively, but sons that are unclear, the manufacturer has decided not
may involve direct phosphorylation by BCR-ABL that to develop this compound any further.
prevents binding to axin, thereby stabilizing the pro- A number of other drugs have been tested in the lab-
tein.42 Altogether, this data suggests that targeting β- oratory, including proteasome inhibitors, histone
catenin may be useful for the treatment of CML. deacetylase (HDAC) inhibitors and DNA methytrans-
Unfortunately, despite efforts by many groups and com- ferase inhibitors, for their ability to target CML stem
panies, until now no clinical β-catenin inhibitors have cells alone or in conjunction with imatinib. A clinical
been identified. trial is planning to test LBH589, a potent HDAC
Hedgehog signaling has also been implicated in the inhibitor in combination with imatinib, in patients with
self-renewal of CML stem cells.43-44 The Hedgehog fam- molecular disease persistence.
ily of proteins has three members, Sonic, Desert and
Indian Hedgehog. Binding to their receptor, patched Targeting stem cells rather than their pathways
activates a signaling cascade the leads to stabilization of The unprecedented ability of imatinib to reduce dis-
the transcription factor Smoothen (SMO). Mice trans- ease burden in CML to very low levels has greatly stim-
planted with BCR-ABL transduced cells lacking SMO ulated interest in vaccination strategies. Non-specific
had significantly prolonged survival compared with approaches are mainly based on Interferon-α, the drug
SMO+/+ cells. Furthermore, expression of an activated therapy standard of care before the arrival of imatinib.
form of SMO accelerated the disease in the same model. Patients with CCyR to IFN-α, but to imatinib were
Conversely, cyclopamine, which inhibits Hedghog sig- shown to harbor cytotoxic T-cell directed against PR-3,
naling by stabilizing an inactive form of SMO, pro- a peptide derived from myeloblastin, a protein that is
longed survival of leukemic mice and reduced colony overexpressed on CML cells.48 This suggests that
formation by human CML cells, suggesting that target- responses to IFN-α are immunologically based. The
ing the Hedgehog pathway may be therapeutically combination of imatinib and interferon-α was tested in
effective. SMO inhibitors are in development by sever- several phase 1/2 studies, without clear benefit but sig-

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 75 |
14th Congress of the European Hematology Association

nificant toxicity. In contrast, a recent interim analysis 4. Raskind WH, Ferraris AM, Najfeld V, Jacobson RJ, Moohr JW,
Fialkow PJ et al. Further evidence for the existence of a clonal
from the French multicenter trial showed a significantly Ph-negative stage in some cases of Ph-positive chronic myelo-
higher rate of major molecular responses for the inter- cytic leukemia. Leukemia. 1993;7:1163-7.
feron/imatinib combination compared to imatinib and 5. Deininger MWN. Cytogenetics in patients on imatinib. Semin
imatinib/cytarabine.49 A second approach is vaccines Hematol 2002: In press.
6. Bumm T, Muller C, Al Ali HK, Krohn K, Shepherd P, Schmidt
based on peptides derived from the BCR-ABL junction. E et al. Emergence of clonal cytogenetic abnormalities in Ph-
Various types of vaccines have been tested in several cells in some CML patients in cytogenetic remission to ima-
clinical trials, with a suggestion of activity in patients tinib but restoration of polyclonal hematopoiesis in the major-
ity. Blood 2003;101:1941-9.
with low residual disease burden.50-51 Unfortunately, as 7. Daley GQ, Van Etten RA, Baltimore D. Induction of chronic
all trials were single-armed, these data await confirma- myelogenous leukemia in mice by the P210bcr/abl gene of the
tion in a prospective controlled study. Yet another Philadelphia chromosome. Science 1990;247:824-30.
8. Theocharides A, Boissinot M, Girodon F, Garand R, Teo SS,
approach is vaccines based on a leukemia cell line Lippert E et al. Leukemic blasts in transformed JAK2-V617F-
(K562) expressing GM-CSF, with promising preliminary positive myeloproliferative disorders are frequently negative
but as yet unconfirmed results.52 for the JAK2-V617F mutation. Blood 2007;110:375-9.
9. Yong AS, Szydlo RM, Goldman JM, Apperley JF, Melo JV.
Molecular profiling of CD34+ cells identifies low expression
Conclusion of CD7, along with high expression of proteinase 3 or elastase,
Imatinib has changed the face of CML from a deadly as predictors of longer survival in patients with CML. Blood
2006;107:205-12.
disease to a chronic ailment, manageable with drug 10. Huntly BJ, Shigematsu H, Deguchi K, Lee BH, Mizuno S,
therapy, and in many patients compatible with good or Duclos N et al. MOZ-TIF2, but not BCR-ABL, confers proper-
at least acceptable quality of life. As a result, CML ties of leukemic stem cells to committed murine hematopoiet-
prevalence has increased considerably and is set to ic progenitors. Cancer Cell 2004;6:587-96.
11. Takahashi N, Miura I, Saitoh K, Miura AB. Lineage involve-
increase even more over the next several decades. ment of stem cells bearing the Philedelphia chromosome in
Estimates are that in 2040 there will be 250,000 CML chronic myeloid leukemia in the chronic phase as shown by a
patients in the US alone. One might argue that we do combination of fluorescence-activated cell sorting and fluores-
cence in situ hybridization. Blood 1998;92:4758-63.
not need to worry, given the low risk of relapse in 12. Petzer AL, Eaves CJ, Lansdorp PM, et al. Characterization of
patients with a CCyR and the apparent lack of late tox- primitive subpopulation of normal and leukemic cells present
icities. However, despite generally good tolerability, in the blood of patients with newly diagnosed as well as
established chronic myeloid leukemia. Blood 1996;88:2162-71.
many patients do experience side effects that impair 13. Bose S, Deininger M, Gora-Tybor J, Goldman JM, Melo JV.
their quality of life. For women of childbearing age, the The presence of BCR-ABL fusion genes in leukocytes of nor-
inability to have children is a significant burden. Lastly, mal individuals: implications for the assessment of minimal
residual disease. Blood 1998;92:3362-7.
the health-economic burden of lifelong therapy is con- 14. Marin D, Bua M, Marktel S, et al. The combination of cytoge-
siderable. Resistance to imatinib, while still a problem, netic response after 6 months treatment with STI571 and the
affects only a minority of patients, so the focus has presencec of cytopenias in patients with CML in chronic
rightly shifted to the question of disease eradication and phase resistant to or intolerant of interferon-alfa defines four
different prognostic groups [abstract]. Blood 2001;98:846a.
the ability to stop medication. At this point, the most 15. Jamieson CH, Ailles LE, Dylla SJ, Jones C, Zehnder JL, Gotlib
promising finding is that some patients in CMR have J et al. Granulocyte-macrophage progenitors as candidate
stopped the drug and maintained PCR negativity, sug- leukemic stem cells in blast-crisis CML. N Engl J Med 2004;
351:657-67.
gesting that the drug may cure a few patients, although 16. Kolb HJ, Mittermuller J, Clemm C, Holler E, Ledderose G,
more follow-up is still warranted. How this is accom- Brehm G et al. Donor leukocyte transfusions for treatment of
plished is unknown, and what sets the disease in these recurrent chronic myelogenous leukemia in marrow trans-
plant patients. Blood 1990;76:2462-5.
patients apart from others remains to be determined. 17. Gale RP, Horowitz MM, Ash RC, Champlin RE, Goldman JM,
Does one simply have to treat long enough to deplete Rimm AA et al. Identical-twin bone marrow transplants for
the Ph+ cell clone? This would be supported by the fact leukemia. Ann Intern Med 1994;120:646-52.
18. Fang Y, Gratwohl A, van Houwelingen HC. Relapse risk
that the median levels of BCR-ABL transcripts in the assessment of transplantation for patients with chronic
IRIS study continue to decline.53 On the other hand, myeloid leukaemia. Chin Med J (Engl.) 2003;116:305-8.
there are many patients in whom the levels of residual 19. Holyoake T, Jiang X, Eaves C, Eaves A. Isolation of a highly
quiescent subpopulation of primitive leukemic cells in chronic
disease stabilize at a low level, without achieving PCR myeloid leukemia. Blood 1999;94:2056-64.
negativity, and who would have a rapid disease recur- 20. Udomsakdi C, Eaves CJ, Swolin B, Reid DS, Barnett MJ, Eaves
rence after stopping imatinib. Thus, much work AC et al. Rapid decline of chronic myeloid leukemic cells in
remains to be done to understand better the hetero- long-term culture due to a defect at the leukemic stem cell
level. Proc Natl Acad Sci USA 1992;89:6192-6.
geneity of the disease and its response to BCR-ABL-tar- 21. Barnett MJ, Eaves CJ, Phillips GL, Hogge DE, Klingemann HG,
geted therapy. At this point, it seems that imatinib may Lansdorp PM et al. Autografting in chronic myeloid leukemia
be curative for a minority of patients, but that a funda- with cultured marrow: update of the Vancouver Study. Stem
Cells 1993;11:64-6.
mentally different approach will be required to eradi- 22. Druker BJ, Guilhot F, O'Brien SG, Gathmann I, Kantarjian H,
cate the disease in the remaining patients. Gattermann N et al. Five-year follow-up of patients receiving
imatinib for chronic myeloid leukemia. N Engl J Med 2006;
355:2408-17.
23. de Lavallade H, Apperley JF, Khorashad JS, Milojkovic D, Reid
Reference AG, Bua M et al. Imatinib for newly diagnosed patients with
chronic myeloid leukemia: incidence of sustained responses in
1. Sawyers CL. Chronic myeloid leukemia. N Engl J Med 1999; an intention-to-treat analysis. J Clin Oncol 2008;26:3358-63.
340:1330-40. 24. Hughes TP, Kaeda J, Branford S, Rudzki Z, Hochhaus A,
2. Deininger MW, Goldman JM, Melo JV. The molecular biology Hensley ML et al. Frequency of major molecular responses to
of chronic myeloid leukemia. Blood 2000;96:3343-56. imatinib or interferon alfa plus cytarabine in newly diagnosed
3. Fialkow PJ, Martin PJ, Najfeld V, Penfold GK, Jacobson RJ, chronic myeloid leukemia. N Engl J Med 2003;349:1423-32.
Hansen JA. Evidence for a multistep pathogenesis of chronic 25. Bhatia R, Holtz M, Niu N, Gray R, Snyder DS, Sawyers CL et
myelogenous leukemia. Blood 1981;58:158-63. al. Persistence of malignant hematopoietic progenitors in

| 76 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

chronic myelogenous leukemia patients in complete cytoge- Fanning S et al. Effects of a selective inhibitor of the Abl tyro-
netic remission following imatinib mesylate treatment. Blood sine kinase on the growth of Bcr-Abl positive cells. Nat Med
2003;101:4701-7. 1996;2:561-6.
26. Chu S, Holtz M, Gupta M, Bhatia R. BCR/ABL kinase inhibi- 41. Zhao C, Blum J, Chen A, Kwon HY, Jung SH, Cook JM et al.
tion by imatinib mesylate enhances MAP kinase activity in Loss of β-catenin impairs the renewal of normal and CML
chronic myelogenous leukemia CD34+ cells. Blood 2004;103: stem cells in vivo. Cancer Cell 2007;12:528-41.
3167-74. 42. Coluccia AM, Vacca A, Dunach M, Mologni L, Redaelli S,
27. Rousselot P, Huguet F, Rea D, Legros L, Cayuela JM, Maarek O Bustos VH et al. Bcr-Abl stabilizes beta-catenin in chronic
et al. Imatinib mesylate discontinuation in patients with myeloid leukemia through its tyrosine phosphorylation.
chronic myelogenous leukemia in complete molecular remis- EMBO J 2007;26:1456-66.
sion for more than 2 years. Blood 2007;109:58-60. 43. Dierks C, Beigi R, Guo GR, Zirlik K, Stegert MR, Manley P et
28. Michor F, Hughes TP, Iwasa Y, Branford S, Shah NP, Sawyers al. Expansion of Bcr-Abl-positive leukemic stem cells is
CL, Nowak MA. Dynamics of chronic myeloid leukaemia. dependent on Hedgehog pathway activation. Cancer Cell
Nature 2005;435:1267-70. 2008; 14:238-49.
29. Roeder I, Horn M, Glauche I, Hochhaus A, Mueller MC, 44. Zhao C, Chen A, Jamieson CH, Fereshteh M, Abrahamsson A,
Loeffler M et al. Dynamic modeling of imatinib-treated chron- Blum J et al. Hedgehog signalling is essential for maintenance
ic myeloid leukemia: functional insights and clinical implica- of cancer stem cells in myeloid leukaemia. Nature 2009
tions. Nat Med 2006;12:1181-4. 45. Ito K, Bernardi R, Morotti A, Matsuoka S, Saglio G, Ikeda Y et
30. Graham SM, Jorgensen HG, Allan E, Pearson C, Alcorn MJ, al. PML targeting eradicates quiescent leukaemia-initiating
Richmond L, Holyoake TL. Primitive, quiescent, Philadelphia- cells. Nature 2008;453:1072-8.
positive stem cells from patients with chronic myeloid 46. Jorgensen HG, Allan EK, Graham SM, Godden JL, Richmond
leukemia are insensitive to STI571 in vitro. Blood 2002;99:319- L, Elliott MA et al. Lonafarnib reduces the resistance of primi-
25. tive quiescent CML cells to imatinib mesylate in vitro.
31. Copland M, Hamilton A, Elrick LJ, Baird JW, Allan EK, Jordan- Leukemia 2005;19:1184-91.
ides N et al. Dasatinib (BMS-354825) targets an earlier progen- 47. Copland M, Pellicano F, Richmond L, Allan EK, Hamilton A,
itor population than imatinib in primary CML but does not Lee FY et al. BMS-214662 potently induces apoptosis of chron-
eliminate the quiescent fraction. Blood 2006;107:4532-9. ic myeloid leukemia stem and progenitor cells and synergises
32. Jorgensen HG, Allan EK, Jordanides NE, Mountford JC, with tyrosine kinase inhibitors. Blood 2007;
Holyoake TL. Nilotinib exerts equipotent anti-proliferative 48. Burchert A, Wolfl S, Schmidt M, Brendel C, Denecke B, Cai D
effects to imatinib and does not induce apoptosis in CD34+ et al. Interferon-α, but not the ABL-kinase inhibitor imatinib
CML cells. Blood 2007
33. Jiang X, Zhao Y, Smith C, Gasparetto M, Turhan A, Eaves A, (STI571), induces expression of myeloblastin and a specific T-
Eaves C. Chronic myeloid leukemia stem cells possess multi- cell response in chronic myeloid leukemia. Blood 2003;101:
ple unique features of resistance to BCR-ABL targeted thera- 259-4.
pies. Leukemia 2007;21:926-35. 49. Guilhot F, Mahon FX, Guilhot J, et al. Randomized compari-
34. Chu S, Xu H, Shah NP, et al. Detection of BCR-ABL kinase son of Imatinib versus Imatinib combination therapies in
mutations in CD34+ cells from chronic myelogenous newly diagnosed chronic myeloid leukaemia (CML) patients
leukemia patients in complete cytogenetic remission on ima- in chronic phase (CP): First Results of the Phase III (SPIRIT)
tinib mesylate treatment. Blood 2005;105:2093-8. Trial from the French CML Group (FI LMC). Blood 2008;112:
35. Sherbenou DW, Wong MJ, Humayun A, McGreevey LS, 183.
Harrell P, Yang R et al. Mutations of the BCR-ABL-kinase 50. Bocchia M, Gentili S, Abruzzese E, et al. Effect of a p210 mul-
domain occur in a minority of patients with stable complete tipeptide vaccine associated with imatinib or interferon in
cytogenetic response to imatinib. Leukemia 2007;21:489-93. patients with chronic myeloid leukaemia and persistent resid-
36. Crossman LC, Druker BJ, Deininger MW, Pirmohamed M, ual disease: a multicentre observational trial. Lancet 2005;365:
Wang L, Clark RE. hOCT 1 and resistance to imatinib. Blood 657-62.
2005;106:1133-4. 51. Cathcart K, Pinilla-Ibarz J, Korontsvit T, Schwartz J, Zakhaleva
37. Wang L, Giannoudis A, Lane S, Williamson P, Pirmohamed M, V, Papadopoulos EB, Scheinberg DA. A multivalent bcr-abl
Clark RE. Expression of the uptake drug transporter hOCT1 is fusion peptide vaccination trial in patients with chronic
an important clinical determinant of the response to imatinib myeloid leukemia. Blood 2004;103:1037-42.
in chronic myeloid leukemia. Clin Pharmacol Ther 2008;83: 52. Smith BD, Kasamon YL, Miller CB, et al. K562/GM-CSF
258-64. Vaccination Reduces Tumor Burden, Including Achieving
38. White DL, Saunders VA, Dang P, Engler J, Venables A, Zrim S, Molecular Remissions, in Chronic Myeloid Leukemia (CML)
et al. Most CML patients who have a suboptimal response to Patients with Residual Disease on Imatinib Mesylate (IM).
imatinib have low OCT-1 Activity. Higher doses of imatinib Blood 2005;106:2858.
may overcome the negative impact of low OCT-1 Activity. 53. Hughes TP, Hochhaus A, Branford S, et al. Reduction of BCR-
Blood 2007;110:4064-72 ABL Transcript Levels at 6, 12, and 18 Months (mo) Correlates
39. Holtz M, Forman SJ, Bhatia R. Growth factor stimulation with Long-Term Outcomes on Imatinib (IM) at 72 Mo: An
reduces residual quiescent chronic myelogenous leukemia Analysis from the International Randomized Study of
progenitors remaining after imatinib treatment. Cancer Res Interferon versus STI571 (IRIS) in Patients (pts) with Chronic
2007; 67:1113-20. Phase Chronic Myeloid Leukemia (CML-CP). Blood 2008;
40. Druker BJ, Tamura S, Buchdunger E, Ohno S, Segal GM, 112:334.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 77 |
Chronic myeloid leukemia

Dynamics of mutated clones in chronic myeloid


leukemia patients

A. Hochhaus A B S T R A C T

Point mutations of the BCR-ABL tyrosine kinase domain are considered the predominant cause of
III Medizinische Klinik, imatinib resistance in chronic myeloid leukemia patients. The expansion of mutant BCR-ABL positive
Universitätsmedizin Mannheim,
Universität Heidelberg, Mannheim, clones during treatment with tyrosine kinase inhibitors (TKI) is based on a clonal selection of less sen-
Germany sitive cells. Eventually, clonal selection leads to molecular, cytogenetic and hematologic resistance and
to advanced stages of the disease. Time to relapse after first appearance of the mutation on low level
is variable and depends on the degree of resistance and the enzymatic activity of the mutated BCR-
ABL protein. Data have been reported on the reversibility of clonal selection after cessation of thera-
Hematology Education:
py with TKI. Response to second generation TKI is closely related to the specific sensitivity of the indi-
the education program for the
annual congress of the European vidual mutation. Knowledge of the sensitivities of mutations to different inhibitors may allow selec-
Hematology Association tion of the second line therapy based on molecular data. The availability of highly sensitive methods
to detect mutation allows for modeling the dynamics of mutated clones. However, there is no prospec-
2009;3:78-82 tive data suggestion that all low level mutation will eventually cause relapse. In the case of newly
detected, previously unpublished BCR-ABL sequence abnormalities, polymorphisms of ABL should be
excluded for the individual patient.

esistance to imatinib and second ge- tage of the mutated clone over clones con-

R neration tyrosine kinase inhibitors


(TKI) during the treatment of chronic
myeloid leukemia (CML) is frequently asso-
taining unmutated BCR-ABL or normal
hematopoiesis with wild-type ABL; that is,
at least three different cell types should be
ciated with point mutations in the BCR-ABL considered in any model of TKI resistance.
gene encoding the ATP binding region likely
to cause disease relapse.1 Early diagnosis and Dynamics of resistant clones during imatinib
monitoring of these mutations may be therapy
important in order to prevent the expansion In 2001, Gorre et al. described 11 patients
of resistant clones. Mutations may be cate- treated with imatinib for CML blast crisis or
gorized into four groups, based upon the Ph+ acute lymphoblastic leukemia (ALL)
crystallographic structure of cABL: (i) those who relapsed on treatment. BCR-ABL gene
which directly impair imatinib, binding to amplification was detected in three patients.
the catalytic domain of the oncogenic pro- Sequencing of the ATP-binding pocket and
tein; (ii) those within the phosphate binding the activation loop of the kinase domain
site (“P-loop”); (iii) those within the activa- showed an identical cytosine to thymidine
tion loop, preventing the kinase from mutation at ABL nucleotide 944 in six of
achieving the inactive conformation nine assessable patients. This mutation
required for imatinib binding; (iv) those resulted in a single amino-acid change at
within the catalytic domain.2 position 315, designated T315I. Threonine
Mutation detection may aid in risk stratifi- 315 forms a crucial hydrogen bond with
cation and molecular-based treatment deci- imatinib and the absence of an oxygen atom
sions. Hence, in addition to conventional in the substituted isoleucine prevented bond
sequencing, various high sensitive methods formation.7,12
have been developed to screen for mutations However, T315I was not the only muta-
in case of lack of optimal response or se- tion seen in imatinib resistant CML
condary relapse. Examples are Denaturating patients.12-14 Up to now, more than 80
High Performance Liquid Chromatography mutants15 have been described, some at a
(D-HPLC),3-4 Ligation-PCR,5-6 sequencing of higher frequency than others: 15 amino-acid
clones,7-8 ARMS (amplification refractory substitutions account for more than 85% of
mutation system) assays9 and quantitative the mutations, and the mutations responsi-
mutation specific PCR assays.10 Sensitive ble for 66% of reported cases occur at seven
methods were used to determine BCR-ABL sites only (G250, Y253, E255, T315, M351,
mutations in subpopulations of cells harbo- F359, H396). Furthermore, different amino-
ring intrinsic resistance, for example, per- acid substitutions can occur at the same
sistent leukemic stem cells.11 residue – for example, F317C or – L or – V –
Dynamics of clones harboring mutated and can confer different imatinib sensitivi-
BCR-ABL depend on the selective pressure ties. Certain mutations seem to occur more
by the TKI used and the proliferative advan- often in different disease phases. For exam-

| 78 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

ple, substitutions at M244, L248, F317, H396, and S417 sion after cessation although decreasing in size due to a
are more likely to occur in patients with chronic phase gradually declining BCR-ABL ratio. The reduction of the
(CP) disease, whereas those at Q252, Y253, E255, T315, proportion of BCR-ABLT315I expression under the thre-
E459, and F486 are often associated with advanced- shold of detection might offer the therapeutic option to
phase disease.12 resume at least temporarily a TKI treatment.19
The systematic and regular analysis of mutations by Quantification of the mutated clone to evaluate treat-
conventional sequencing in a cohort of 319 patients ment efficacy should consider the total BCR-ABL load,
with CML-CP (171 in early and 148 in late CP) who that is, the proportion of mutated BCR-ABL in total
were treated with imatinib revealed a 5-year cumulative BCR-ABL should be multiplied by the BCR-ABL tran-
incidence of mutations of 6.6% and 17% for early and script level.
late CP, respectively. Of the 319 patients, 214 (67%) Omacetaxine (Homoharringtonine, HHT), a cephalo-
achieved complete cytogenetic responses (CCyR). The taxine ester that has demonstrated clinical activity in
identification of a mutation without other evidence of CML, inhibits protein synthesis and induces cell diffe-
imatinib resistance was highly predictive for loss of rentiation and apoptosis. The absence of cross-resist-
CCyR and for progression to advanced phase, though ance between HHT and imatinib in resistant leukemic
the intervals from first identification to loss of CCyR cell lines has been demonstrated, and a possible syner-
and disease progression were relatively long (median, gy between the two compounds has been observed in
21 and 16 months, respectively). Mutations in the P- vitro. HHT decreases the production of the oncogenic
loop were associated with a higher risk of progression kinase BCR-ABL, by inhibition of transcription and
than mutations elsewhere.16 translation in the same way in BCR-ABLT315I mutant and
To test whether kinase domain mutations are a com- in cell lines harboring native BCR-ABL. BCR-ABLT315I
mon mechanism of disease persistence, patients in sta- may completely disappear during treatment with HHT,
ble CCyR were studied. Mutations were demonstrated even after persistence of the clone during hydroxyurea
in 8 of 42 (19%) cases. Four patients with mutations had therapy.20
a concomitant rise of BCR-ABL transcript levels, two of
whom subsequently relapsed; the remaining four did In vitro models on the dynamics of mutations on second
not have an increase in transcript levels and follow-up generation inhibitors and combinations of inhibitors
samples, when amplifiable, showed native BCR-ABL. Dasatinib, nilotinib and bosutinib are potent alternate
Thus, BCR-ABL-kinase domain mutations in patients ABL inhibitors with activity against many imatinib-
with a stable CCyR are infrequent, and their detection resistant BCR-ABL kinase domain mutants, except
does not consistently predict relapse.17 T315I. Ethyl-N-nitrosourea (ENU)-exposed Ba/F3-
Prior to hematologic or cytogenetic relapse, dynamics p210BCR-ABL cells were used to compare incidence and
of mutated clones in CML patients were investigated in types of KD mutants emerging in the presence of ima-
95 patients by D-HPLC and direct sequencing. In tinib, dasatinib, and nilotinib, alone and in dual combi-
patients harboring mutations, hematologic relapse nations. Resistant clones were almost exclusively BCR-
occurred after a median of 12.9 months (range, 0.9- ABL KD mutant at relevant concentrations of nilotinib
44.2), and BCR-ABL mutations first became detectable and dasatinib, consistent with a central role of KD
at a median of 5.8 months (range, 0-30.5) after starting mutations for resistance to these drugs. Twenty diffe-
imatinib therapy. The median interval of the first rent mutations were identified with imatinib, ten with
detectability of mutations and hematologic relapse cor- nilotinib and nine with dasatinib. At intermediate drug
related with the degree of resistance of the mutated levels, the spectrum narrowed to F317V and T315I for
clone to imatinib, expressed as the increase of the half dasatinib and Y253H, E255V, and T315I for nilotinib.
maximal inhibitory concentration (IC50) of imatinib to Thus, cross resistance in vitro is limited to T315I, which
cell lines. Nine patients showed evidence of BCR-ABL is also the only mutant isolated at drug concentrations
mutations prior to imatinib therapy (T315I, n=4; equivalent to maximal achievable plasma trough levels.
M351T, n=3; M244V and Y253H, n=1 each). The sensi- With drug combinations, maximal suppression of resist-
tive detection of small numbers of mutated clones could ant clone outgrowth was achieved at lower concentra-
provide clinical benefit by triggering early therapeutic tions compared with single agents, suggesting that such
interventions.4 combinations may be equipotent to higher dose single
agents.21 Through saturation mutagenesis, ten BCR-ABL
Dynamics of mutations after imatinib cessation mutations resistant to dasatinib were identified, eight of
Prior to the advent of second-generation TKI patients which occurred at drug contact residues. Some mutants
with resistance to imatinib were treated with non-spe- were unique to dasatinib, whereas others also conferred
cific drugs, like hydroxyurea. After stopping imatinib imatinib resistance. The combination of imatinib plus
due to lack of efficacy in case of a BCR-ABL mutation, dasatinib greatly reduced the recovery of drug-resistant
absolute and relative reduction of the mutated clone has clones.22
been observed in the majority of patients.18-19 This is To identify mutations in BCR-ABL that could result in
explained by the proliferative advantage of clones har- resistance to nilotinib, a cDNA library of BCR-ABL
boring unmutated BCR-ABL in the absence of the selec- mutants was introduced into Ba/F3 cells followed by
tive pressure of TKI. Deselection of mutant BCR-ABL selection in nilotinib. Eighty-six individual, drug-re-
positive clones after cessation of tyrosine kinase sistant colonies were recovered, and the SH3, SH2, and
inhibitor therapy is a common and reproducible phe- kinase domains of BCR-ABL were sequenced. Forty-six
nomenon. However, BCR-ABLT315I clones tend to colonies had single point mutations in BCR-ABL, with
account for a persisting proportion of BCR-ABL expres- seventeen different mutations, all within the kinase

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 79 |
14th Congress of the European Hematology Association

domain. Each of the 17 single point mutants were advanced-phase CML (T315, E255, Y253).12 In the nilo-
reconstructed by site directed mutagenesis of native tinib phase II study after imatinib resistance, among
BCR-ABL and found to be approximately 2.5- to 800- imatinib-resistant patients, the frequency of mutations
fold more resistant to nilotinib than native BCR-ABL. at baseline was 55%. In patients without baseline muta-
The mutations included six known imatinib–resistant tions, MCyR was achieved in 60%, CCyR in 40%, and
mutations, including T315I, which showed complete major molecular response (MMR) in 29% of patients
resistance to nilotinib. Most nilotinib-resistant mutants after 12 months of therapy versus 49%, 32%, and 22%
were also resistant to imatinib.23 of patients with mutations. Responses in patients har-
Indeed, results from in vitro models predicted most of boring mutations with high in vitro sensitivity to nilo-
the resistance mutations that were later detected in clini- tinib (IC50<150 nM), or mutations with unknown nilo-
cal trials with different kinase inhibitors. A practical tinib sensitivity, were equivalent to those for patients
measure of the sensitivity of a given mutation to a TKI without mutations. Patients with mutations that were
is its IC50 for that specific inhibitor, determined either on less sensitive to nilotinib in vitro (IC50>150 nM; Y253H,
cellular or biochemical assays. There are two relevant E255V/K, F359V/C) had less favorable responses with
consequences of IC50 determination of ABL mutations. 13%, 43%, and 18%, respectively, achieving MCyR;
First, it is possible to modify the clinical strategy in none achieved CCyR.27 Similar to nilotinib, a correlation
treatment resistant patients with a kinase domain muta- of the in vitro IC50 (<vs>3 nM) and response to dasatinib
tion based on the IC50 of drugs for the mutation; that is, was observed in CP CML patients after failure to ima-
choosing whether to increase the dose of imatinib or tinib. Except resistance to T315I, response was poor in
replace it with a second generation tyrosine kinase patients with F317L mutations, as predicted in vitro.28
inhibitor. In the case of the pan-resistant T315I muta-
tion, allogeneic stem cell transplantation could be con- Resistance to second generation inhibitors
sidered. Second, when finding a mutation in a resistant Analysis of BCR-ABL genotypes in CML patients
patient, it is important to search for its IC50 in a muta- who relapsed after sequential treatment with the ABL
tion database to link it to the clinical resistance, since inhibitors imatinib and dasatinib revealed evolving
some mutations may be completely resistant to an resistant BCR-ABL kinase domain mutations in all
inhibitor whilst others are “innocent bystanders” that cases. Twelve patients relapsed with the pan-resistant
accompany other mechanisms of resistance. A compre- T315I mutation, whereas six patients developed novel
hensive list of the in vitro IC50 to imatinib, dasatinib, nilo- BCR-ABL mutations predicted to retain sensitivity to
tinib, and bosutinib was published recently.24 Such imatinib based on in vitro studies. Three of these patients
tables, however, do not mirror the clinical activity com- were retreated with imatinib or nilotinib and respon-
pletely, since data on the pharmacokinetics of the va- ded; however, selection for compound mutants (2 or 3
rious inhibitors and the cellular availability are not con- BCR-ABL mutations in the same molecule) can substan-
sidered. tially limit the potential effectiveness of retreating
The identification of a specific type of mutation in patients with inhibitors that have previously failed.
relapsed patients seems to be of prognostic relevance Furthermore, drug-resistant mutations, when com-
since the different types of mutation correlate with the pounded, can increase oncogenic potency relative to the
risk of evolution of relapsed patients.25 The presence component mutants in transformation assays. These
and the type of a mutation, should, therefore, help to re- findings demonstrate the potential hazards of sequen-
evaluate the treatment strategy, particularly now that tial kinase inhibitor therapy and suggest a role for a
second generation TKI are available. Mutations with combination of ABL kinase inhibitors to prevent the
limited sensitivities have been described in vitro and in outgrowth of cells harboring drug-resistant BCR-ABL
vivo for both dasatinib and nilotinib. Screening for muta- mutations.29 In vivo, however, the level of most muta-
tions should always be carried out in patients who fail tions is fluctuating individually suggesting two different
to respond or who have a suboptimal response to ima- clones instead of one compound clone containing two
tinib, as defined by the recommendations of the mutations.
European LeukemiaNet.26 In CP CML patients after dasatinib therapy, among
1,043 patients analyzed, 174 had a mutational assess-
Dynamics of mutations during treatment with second ment at the time of dasatinib discontinuation. In these
generation inhibitors 174 patients, 54 new mutations occurred in 47 patients
Among patients with CML-CP who developed ima- (7 patients developed more than one new mutation and
tinib resistance after prior treatment with interferon α, 42 patients had new mutations with an IC50 to dasatinib
a BCR-ABL mutation was reported in 31-42% of >3 nM). Mutations at the time of dasatinib resistance
patients. In patients with imatinib-resistant advanced- were more common in patients with versus without
phase CML or Ph+ ALL, a BCR-ABL mutation was mutations at the time of resistance to imatinib.28 The
detected in 30% to 83% of patients. Although more expansion of a mutant Ph-positive clone that responds
than 90 different BCR-ABL mutations have been repor- initially to a second generation TKI may be due either to
ted affecting more than 55 amino acids, 15 amino-acid late acquisition of a second mutation in the originally
substitutions account for more than 85% of reported mutated clone, such as the T315I, or to acquisition of a
mutations, and 7 amino acids (G250, Y253, E255, T315, completely new mutant clone, such as F317L.30
M351, F359, H396) are responsible for two-thirds.
Mutation types vary by disease phase; in patients with Gatekeeper mutation BCR-ABLT315I
CML-CP, the most commonly mutated amino acids The BCR-ABLT315I mutation represents a major me-
(M244, M351, G250) are different to those found in chanism of resistance to TKI. CML patients with a BCR-

| 80 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

ABLT315I mutation have been reported to have poor pro- have a higher sensitivity, which can reach 0.1%. They
gnosis. Out of 27 patients with T315I, 20 developed are, however, directed to the search for specific muta-
T315I after imatinib failure (representing 11% of 186 tions and do not screen the entire kinase domain region
patients with imatinib failure), and 7 of 23 who devel- of the BCR-ABL gene. Various groups use D-HPLC as a
oped new mutations after second TKI with a median routine approach to screen for the presence of kinase
follow-up from mutation detection of 18 months. At domain mutations, which will then be confirmed and
the time of T315I detection, 10 were CP, and 17 in semi-quantified by sequence analysis. This method has
advanced disease. Except for the lack of response to sec- been shown to be applicable to the routine monitoring
ond TKIs, there was no difference in patient characte- of CML patients and to have a sensitivity of 0.5-5% in
ristics and outcome between patients with T315I and mutation detection. Nevertheless, from a clinical stand-
those with other or no mutations. Patients in CP had a point, the significance of very small subclones carrying
2-year survival rate of 87%. Although the T315I muta- kinase domain mutations has yet to be demonstrated.
tion is resistant to currently available TKIs, survival of In contrast to CP CML, BCR-ABL mutations are com-
patients with T315I remains mostly dependent on the mon in Ph+ acute lymphoblastic leukemia (ALL) patients
stage of the disease, with many CP patients having an at diagnosis and prior to imatinib therapy. Using D-
indolent course.31 HPLC and allele specific PCR, Pfeifer et al. detected
The objectives of an observational study were to esti- BCR-ABL mutations in a minor subpopulation of
mate outcome for Ph+ ALL and CML patients with leukemic cells in 40% of newly diagnosed and imatinib-
T315I mutation. Median time between TKI treatment naïve Ph+ ALL patients. At relapse, the dominant cell
start and T315I mutation detection was 29 months for clone harbored an identical mutation in 90% of cases;
CP, 15 for AP, 6 for BP, and 9 for Ph+ ALL. Median OS the overall prevalence of mutations at relapse was 80%.
from T315I mutation detection was 22.4, 28.4, 4.0, and P-loop mutations predominated and were not associa-
4.9 months, and median PFS was 11.5, 22.2, 1.8, and 2.5 ted with an inferior hematologic or molecular remission
months, respectively, for CP, AP, BP, and Ph+ ALL rate or shorter remission duration compared with
patients. These results confirm that survival of patients unmutated BCR-ABL. BCR-ABL mutations conferring
harboring a T315I mutation is dependent on the disease high-level imatinib resistance are present in a substan-
phase at the time of T315I mutation detection.32 tial proportion of patients with de novo Ph+ ALL and
eventually give rise to relapse.33
ABL polymorphisms A recent study on consecutive unselected CML
The BCR-ABLK247R change is based on a rare single patients found correlations between the pre-treatment
nucleotide polymorphism (SNP) of ABL occurring like- presence of low-level mutations and both stage of di-
wise in healthy controls and non-hematologic cell sease and clonal cytogenetic evolution, but not with the
types. Despite its juxtaposition to the P-loop, functio- probability of response to imatinib. Even the complete-
nal analysis showed no alteration compared to non- ly resistant T315I mutant, when detected at a low level
mutated BCR-ABL. In order to investigate if other prior to treatment, did not prove to be selected during
changes in the BCR-ABL kinase domain should be con- treatment.34 These findings are confirmed by observa-
sidered as SNP rather than acquired mutations, 911 tions on the kinetics of the mutated clones in a small
CML patients after failure or suboptimal response to group of early and late chronic phase CML patients
imatinib were screened for BCR-ABL kinase domain showing a lack of correlation between the residual di-
mutations. SNP analysis was based on the search for sease and the presence of a predominant kinase domain
nucleotide changes in corresponding normal, non- mutation.35 As a practical consequence, screening with
translocated ABL alleles by ABL allele-specific PCR fol- high sensitivity methods for low-titer kinase domain
lowing mutation analysis. In addition to the K247R mutations in newly diagnosed patients should be
polymorphism, five new SNPs within the BCR-ABL reserved for clinical investigations, since the long term
kinase domain were uncovered; two of them led to significance of the pre-treatment low titer mutations
amino acid changes. SNPs could theoretically contribute that may constitute a biological marker of the clone’s
to primary but not to secondary resistance to TKI and instability still needs to be assessed in prospective clini-
must, therefore, be distinguished from acquired muta- cal trials. On the other hand, the issue of evaluating
tions. Novel point mutations should be confirmed by kinase domain mutations during follow-up deserves
analyzing the normal ABL alleles to exclude polymor- more attention.2
phisms.15
The author was supported by the German José-Carreras-
Sensitive methods for mutation detection Foundation (DJCLS H 03/01).
Several methods have been used to detect the pre-
sence of kinase domain mutations in CML patients.
Some of these give a quantitative or semi-quantitative References
estimation of the relative proportion of the mutated
clone, and the sensitivity in some cases is very high 1. Hochhaus A, Erben P, Ernst T, Mueller MC. Resistance to tar-
geted therapy in chronic myelogenous leukemia. Semin
(1/10,000). Direct sequencing, which is now often per- Hematol 2007;44:S15-S24.
formed using automated high-throughput instruments, 2. Baccarani M, Pane F, Saglio G. Monitoring treatment of chro-
is still the most widespread method applied in the rou- nic myeloid leukemia. Haematologica 2008;93:161-9.
3. Soverini S, Martinelli G, Amabile M, Poerio A, Bianchini M,
tine monitoring of patients. The main disadvantage of Rosti G, et al. Denaturing-HPLC-based assay for detection of
this technique is its low sensitivity (around 20%). ABL mutations in chronic myeloid leukemia patients resistant
Techniques based upon the use of allele-specific PCR to Imatinib. Clin Chem 2004;50:1205-13.
4. Ernst T, Erben P, Müller MC, Paschka P, Schenk T, Hoffmann

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 81 |
14th Congress of the European Hematology Association

J, et al. Dynamics of BCR-ABL mutated clones prior to hema- (ENU)-based mutagenesis screen: high efficacy of drug combi-
tologic or cytogenetic resistance to imatinib. Haematologica nations. Blood 2006;108:2332-8.
2008;93:186-92. 22. Burgess MR, Skaggs BJ, Shah NP, Lee FY, Sawyers CL.
5. Preuner S, Denk D, Frommlet F, Nesslboeck M, Lion T. Comparative analysis of two clinically active BCR-ABL kinase
Quantitative monitoring of cell clones carrying point muta- inhibitors reveals the role of conformation-specific binding in
tions in the BCR-ABL tyrosine kinase domain by ligation- resistance. Proc Natl Acad Sci USA 2005;102:3395-400.
dependent polymerase chain reaction (LD-PCR). Leukemia 23. Ray A, Cowan-Jacob SW, Manley PW, Mestan J, Griffin JD.
2008;22:1956-61.
6. Pelz-Ackermann O, Cross M, Pfeifer H, Deininger M, Wang Identification of BCR-ABL point mutations conferring resist-
SY, Al Ali HK, et al. Highly sensitive and quantitative detec- ance to the Abl kinase inhibitor AMN107 (nilotinib) by a ran-
tion of BCR-ABL kinase domain mutations by ligation PCR. dom mutagenesis study. Blood 2007;109:5011-5.
Leukemia 2008;22:2288-91. 24. Redaelli S, Piazza R, Rostagno R, Magistroni V, Perini P, Marega
7. Gorre ME, Mohammed M, Ellwood K, Hsu N, Paquette R, Rao M, et al. Activity of bosutinib, dasatinib, and nilotinib against
PN, et al. Clinical resistance to STI-571 cancer therapy caused 18 imatinib-resistant BCR/ABL mutants. J Clin Oncol 2009;
by BCR-ABL gene mutation or amplification. Science 2001; 27:469-71.
293:876-80. 25. Soverini S, Martinelli G, Rosti G, Bassi S, Amabile M, Poerio
8. Shah NP, Nicoll JM, Nagar B, Gorre ME, Paquette RL, Kuriyan A, et al. ABL mutations in late chronic phase chronic myeloid
J, et al. Multiple BCR-ABL kinase domain mutations confer leukemia patients with up-front cytogenetic resistance to ima-
polyclonal resistance to the tyrosine kinase inhibitor imatinib tinib are associated with a greater likelihood of progression to
(STI571) in chronic phase and blast crisis chronic myeloid blast crisis and shorter survival: a study by the GIMEMA
leukemia. Cancer Cell 2002;2:117-25. Working Party on Chronic Myeloid Leukemia. J Clin Oncol
9. Roche-Lestienne C, Lai JL, Darre S, Facon T, Preudhomme C.
A mutation conferring resistance to imatinib at the time of 2005;23:4100-9.
diagnosis of chronic myelogenous leukemia. N Engl J Med 26. Baccarani M, Saglio G, Goldman J, Hochhaus A, Simonsson B,
2003;348:2265-6. Appelbaum F, et al. Evolving concepts in the management of
10. Gruber FX, Lamark T, Anonli A, Sovershaev MA, Olsen M, chronic myeloid leukemia: recommendations from an expert
Gedde-Dahl T, et al. Selecting and deselecting imatinib-re- panel on behalf of the European LeukemiaNet. Blood 2006;
sistant clones: observations made by longitudinal, quantitative 108:1809-20.
monitoring of mutated BCR-ABL. Leukemia 2005;19:2159-65. 27. Hochhaus A, Kim DW, Martinelli G, Hughes TP, Soverini S,
11. Jiang X, Zhao Y, Smith C, Gasparetto M, Turhan A, Eaves A, Branford S, et al. Nilotinib efficacy according to baseline BCR-
et al. Chronic myeloid leukemia stem cells possess multiple ABl mutations in patients with imatinib-resistant chronic
unique features of resistance to BCR-ABL targeted therapies. myeloid leukemia in chronic phase (CML-CP). Blood 2008;
Leukemia 2007;21:926-35. 112:1103-4.
12. Apperley JF. Part I: mechanisms of resistance to imatinib in 28. Müller MC, Cortes J, Kim DW, Druker BJ, Erben P, Pasquini R,
chronic myeloid leukaemia. Lancet Oncol 2007;8:1018-29. et al. Dasatinib efficacy in patients with chronic myeloid
13. Hochhaus A, Kreil S, Corbin AS, La Rosée P, Müller MC,
Lahaye T, et al. Molecular and chromosomal mechanisms of leukemia in chronic phase (CML-CP) and pre-existing BCR-
resistance to imatinib (STI571) therapy. Leukemia 2002;16: ABL mutations. Blood 112:171-2.
2190-6. 29. Shah NP, Skaggs BJ, Branford S, Hughes TP, Nicoll JM, Pa-
14. Hochhaus A, Kreil S, Corbin A, La Rosée P, Lahaye T, Berger quette RL, et al. Sequential ABL kinase inhibitor therapy
U, et al. Roots of clinical resistance to STI-571 cancer therapy. selects for compound drug-resistant BCR-ABL mutations with
Science 2001;293:2163. altered oncogenic potency. J Clin Invest 2007;117:2562-9.
15. Ernst T, Hoffmann J, Erben P, Hanfstein B, Leitner A, 30. Khorashad JS, Milojkovic D, Mehta P, Anand M, Ghorashian
Hehlmann R, et al. ABL single nucleotide polymorphisms may S, Reid AG, et al. In vivo kinetics of kinase domain mutations
masquerade as BCR-ABL mutations associated with resistance in CML patients treated with dasatinib after failing imatinib.
to tyrosine kinase inhibitors in patients with chronic myeloid Blood 2008;111:2378-81.
leukemia. Haematologica 2008;93:1389-93. 31. Jabbour E, Kantarjian H, Jones D, Breeden M, Garcia-Manero
16. Khorashad JS, de Lavallade H, Apperley JF, Milojkovic D, Reid G, O'Brien S, et al. Characteristics and outcomes of patients
AG, Bua M, et al. Finding of kinase domain mutations in with chronic myeloid leukemia and T315I mutation following
patients with chronic phase chronic myeloid leukemia
responding to imatinib may identify those at high risk of di- failure of imatinib mesylate therapy. Blood 2008;112:53-5.
sease progression. J Clin Oncol 2008;26:4806-13. 32. Nicolini F, Martinelli G, Kim DW, Hochhaus A, Mauro MJ,
17. Sherbenou DW, Wong MJ, Humayun A, McGreevey LS, Cortes J, et al. Epidemiological study on survival of chronic
Harrell P, Yang R, et al. Mutations of the BCR-ABL-kinase myeloid leukemia (CML) and Ph+ acute lymphoblastic
domain occur in a minority of patients with stable complete leukemia (ALL) patients with T315I mutation. Final analysis.
cytogenetic response to imatinib. Leukemia 2007;21:489-93. Blood 2008;112:76-7.
18. Müller MC, Lahaye T, Hochhaus A. [Resistance to tumor spe- 33. Pfeifer H, Wassmann B, Pavlova A, Wunderle L, Oldenburg J,
cific therapy with imatinib by clonal selection of mutated Binckebanck, A et al. Kinase domain mutations of BCR-ABL
cells]. Dtsch Med Wochenschr 2002;127:2205-7. frequently precede imatinib-based therapy and give rise to
19. Hanfstein B, Mueller MC, Kreil S, Schenk T, Lorentz C, relapse in patients with de novo Philadelphia-positive acute
Schwindel U, et al. Dynamics of mutant BCR-ABL positive lymphoblastic leukemia (Ph+ALL). Blood 2007;110:727-34.
clones after cessation of imatinib treatment. Haematologica 34. Willis SG, Lange T, Demehri S, Otto S, Crossman L,
2008;93:43. Niederwieser D, et al. High-sensitivity detection of BCR-ABL
20. Legros L, Hayette S, Nicolini FE, Raynaud S, Chabane K,
Magaud JP, et al. BCR-ABL(T315I) transcript disappearance in kinase domain mutations in imatinib-naive patients: correla-
an imatinib-resistant CML patient treated with homoharring- tion with clonal cytogenetic evolution but not response to
tonine: a new therapeutic challenge? Leukemia 2007;21:2204- therapy. Blood 2005;106:2128-37.
6. 35. Khorashad JS, Anand M, Marin D, Saunders S, Al Jabary T,
21. Bradeen HA, Eide CA, O'Hare T, Johnson KJ, Willis SG, Lee Iqbal A, et al. The presence of a BCR-ABL mutant allele in
FY, et al. Comparison of imatinib mesylate, dasatinib (BMS- CML does not always explain clinical resistance to imatinib.
354825), and nilotinib (AMN107) in an N-ethyl-N-nitrosourea Leukemia 2006;20:658-63.

| 82 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Chronic myeloid leukemia

Second generation tyrosine kinase inhibitors:


which and when?

H. Kantarjian A B S T R A C T
J. Cortes
Patients with newly diagnosed chronic myeloid leukemia (CML) treated with imatinib have an esti-
mated 7-year survival of 86% and event-free survival of 81%. The annual failure rate on imatinib is
Department of Leukemia, The
University of Texas, MD Anderson 2-4%. New treatment options are available for these patients. Second generation tyrosine kinase
Cancer Center, Houston, TX, USA inhibitors (dasatinib, nilotinib, bosutinib) have shown significant efficacy in CML after imatinib fail-
ure, and responses appear to be durable. Although all three agents are tyrosine kinase inhibitors, they
have different biochemical structures, and spectra and mechanisms of action. The clinical significance
Hematology Education: of these differences is not clear. As patients receive sequential therapies, some have developed failure
the education program for the
to two or more tyrosine kinase inhibitors or have the multi-resistant T315I mutation. Therapy in these
annual congress of the European
Hematology Association instances is mostly ineffective. New agents are being developed for these patients with promising
results.
2009;3:83-88

matinib has revolutionized the manage- as poor as that with imatinib failure.5 The

I ment of chronic myeloid leukemia


(CML). In the IRIS trial, 82% of patients
achieved a complete cytogenetic remission
“suboptimal response” population is more
heterogenous and the management recom-
mendations are less well defined. There
(CCyR). The estimated 7-year survival is have been no prospective trials comparing
86% and event-free survival (EFS) 81%.1 increasing doses of imatinib versus changing
This outcome compares favorably to the his- therapy. Here, we focus on patients with
torical median survival of 5 years. A subset imatinib failure.
of patients do not have an optimal response
with imatinib. The minimum acceptable Treatment approaches post imatinib failure
response on imatinib therapy should be a Imatinib dose escalation
CCyR. In the IRIS trial, 18% of patients did Before the availability of second genera-
not achieve a CCyR, 11% later lost CCyR, tion tyrosine kinase inhibitors (TKI), ima-
and 8% discontinued therapy for safety rea- tinib dose escalation was a preferred option
sons. Thus, approximately 30% of patients for patients with imatinib failure. Imatinib
need alternative therapies. The criteria for dose escalation to 800 mg in patients treated
diagnosing failure on imatinib therapy must with 400 mg resulted in a CCyR rate of 50%
be clear and precise. The European in patients with cytogenetic failure to ima-
LeukemiaNet have provided definitions of tinib; patients with hematologic failure have
what constitutes failure to imatinib therapy only a 5% probability of obtaining such a
(Table 1).2 These definitions identify patients response.6 Responses are durable and the 2-
with a significantly inferior outcome (medi- year EFS rate is 65%. Dose escalation is par-
an survival of 5 years).3 In this setting, a ticularly beneficial for patients who had a
change of therapy is clearly indicated. Early prior cytogenetic response to standard dose
intervention may improve outcome: imatinib. In these instances, the CCyR prob-
patients treated after loss of a major cytoge- ability was 73% and 2-year EFS rate 74%.
netic response (MCyR) have a significantly The introduction of second generation TKIs
better probability of achieving a CCyR has changed the role of imatinib dose escala-
(72%) compared to those in whom alterna- tion, which is now most frequently consid-
tive treatment is not initiated until loss of a ered for patients with suboptimal response
complete hematologic response (CHR) to standard dose imatinib.
(CCyR 42%). This translates into a signifi-
cantly better 24-month EFS with earlier Second generation tyrosine kinase inhibitors
treatment (EFS 89% vs. 29%).4 The Two of these TKIs, dasatinib and nilotinib
European LeukemiaNet definitions of sub- have already been approved by regulatory
optimal response (less than CHR at 3 agencies around the world for this indica-
months; less than partial cytogenetic tion, and others are under development.
response at 6 months; less than CCyR at 12
months; less than major molecular response Dasatinib
[MMR] at ≥18 months) also identify inferior Dasatinib is a potent Bcr-Abl tyrosine
outcomes compared to those with optimal kinase inhibitor with an IC50 of less than 1
response, although the outcome may not be nM. Dasatinib also inhibits other kinases,

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 83 |
14th Congress of the European Hematology Association

notably the Src family of kinases (SFKs, IC50 0.2-1.1 nM), Table 1. Failure on imatinib therapy according to the
as well as PDGFRβ (IC50 28 nM) and c-KIT (IC50 13 nM).7 European Leukemia Net.
Dasatinib inhibits most BCR-ABL kinase domain
mutants with the exception of T315I. Different mutants Months of treatment Failure response
have different levels of sensitivity to the inhibitor.8-9 A 3 No hematologic response
phase I study selected a dasatinib dose of 70 mg twice 6 Ph-positive 100%
daily (BID) for phase II studies based on the short half- 12 Ph-positive>35%
life of dasatinib, and recovery of kinase inhibition 8 to 18 Any Ph-positivity
Any time post ≥18 months Loss of hematologic reponse or complete
12 hours after administration. Phase II studies demon-
cytogenetic response
strated the efficacy of dasatinib in CML in all phases
post imatinib resistance or intolerance (Table 2). In a
study of 387 patients with CML in chronic phase who
were intolerant (n=99) or resistant (n=288) to imatinib,
a MCyR was achieved in 52% of patients resistant to decreased probability of adverse events. Grade 3 to 4
imatinib; it was complete in 40%. Corresponding rates thrombocytopenia occurred in 22% of patients treated
for patients with imatinib intolerance were 80% and with 100 mg QD compared to 32% with 50 mg BID and
75%, respectively.10 The probability of MCyR was high- 37 to 40% with 140 mg either in a single or divided dose
er with a prior cytogenetic response to imatinib com- schedule. The incidence of pleural effusions was 7%
pared with no prior cytogenetic response (72% vs. with the QD schedule. This resulted in a change in the
42%). The progression-free survival rate was 90% after standard dose of dasatinib in CML chronic phase to 100
a median follow-up of 15.2 months. A study compared mg QD. Dasatinib was effective in the advanced CML
dasatinib to imatinib dose escalation among patients phases, with high rates of hematologic and cytogenetic
who had failed prior therapy with imatinib. Patients responses.14-15 However, responses were less durable. In
were randomized to dasatinib 70 gm BID (N=101) or advanced phase disease, a study testing 70 mg BID ver-
imatinib 400 mg BID (N=49). With a median duration of sus 140 mg QD showed an improved toxicity profile
treatment of 14 months for the dasatinib arm and 3 with equivalent response rates, but the long-term effica-
months for the imatinib arm (crossover allowed for lack cy has not been established.
of MCyR at 12 weeks, intolerance, or progression), the
rates of CCyR were 30% with dasatinib and 7% with Nilotinib
imatinib.11 Treatment with dasatinib at this dose was Nilotinib is a phenylamino-pyrimidine derivative
well tolerated. Common adverse events were grade 3 to designed based on the crystal structure of imatinib in
4 neutropenia (49%) and thrombocytopenia (48%). complex with the ABL kinase.16 Nilotinib has a 20 to 30-
Pleural effusions were noted in 27% but were grade 3 to fold increase selectivity and improved affinity against
4 in only 6%.10 Increased risk of pleural effusions is unmutated BCR-ABL and activity against most clinical-
noted with hypertension, prior cardiac conditions, and a ly relevant BCR-ABL mutants, with the exception of
twice daily dose schedule.12 T315I.16 Based on its relatively long half life, nilotinib
The phase I study suggested that single daily dose was first used with a single daily dose schedule. Dose
schedules and doses of 100 mg daily might be as effec- proportionality was observed at doses up to 400 mg
tive at BID schedules and higher doses. A randomized QD, but reached a plateau beyond that dose. With BID
trial of four dose schedules of dasatinib was conducted: administration, the area under the curve increased pro-
100 mg QD, 50 mg BID, 140 mg QD, and 70 mg BID.13 portional to doses up to 600 mg BID.17 The maximum
This study showed that dasatinib 100 mg single daily tolerated dose was 600 mg BID; 400 mg BID was select-
dose resulted in equivalent response rates and showed a ed for phase II studies.

Table 2. Response to dasatinib, nilotinib and bosutinib after imatinib resistance.

Percent response
Response Dasatinib Nilotinib Bosutinib

Chronic Accelerated Blastic Chronic Accelerated Blastic ChronicAccelerated Blastic


N=387 n=174 n=157 n=321 N=137 N=136 N=146 N=51 N=38

Median follow-up (mo) 15 14 12+ 19 9 3 7 6 3


% Resistant 74 93 88-91 70 80 82 69 NR NR
Hematologic 79 40-50 94 56 19-22 85 54 36
CHR 91 45 27-29 76 31 11-13 81 54 36
NEL - 19 7 - 12 1 - 0 0
Cytogenetic NR 44 36-52 NR NR NR NR NR
Complete 49 32 26-46 44 20 29-32 34 27 35
Partial 11 7 7 15 12 10-16 13 20 18
% Survival (at x months) 96 (15) 82 (12) 50 (12) 88 (24) 67 (24) 42 (12) 98 (12) 60 (12) 50 (10)
NR: not reported; +Minimum follow-up. Data extracted from references 10, 14, 15, 18, 20, 22, 23, 38. Response ranges are for myeloid and lymphoid blastic phase subsets.

| 84 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

Table 3. Characteristics of second generation tyrosine kinase inhibitors.

Parameter Dasatinib Nilotinib Bosutinib

Potency (vs IM) 325 30 20-50


Target Dual Src & Abl Abl Dual Src & ABL
BCR-ABL binding Active + Inactive Inactive Intermediate
Resistant mutations T315I, T315I, E255V T315I
Mutations with intermediate sensitivity E255K/V, V299L, F317L E255K/V, Y253F/H, Q252H, F359V F317L, E255V/K
Standard dose (CP) 100 mg QD 400 mg BID 500 mg QD
Grade 3-4 neutropenia & thrombocytopenia 33%/22% 31%/33% 12%/21%
Other notable toxicities Pleural effusion, bleeding Bilirubin, lipase elevation Diarrhea, rash
C-kit inhibition (vs imatinib) Increased Similar None
PDGFR inhibition (vs imatinib) Increased Similar None
Clinical activity Highly active Highly active Highly active

In a phase II trial of nilotinib, 321 patients with CP


Table 4. In vitro sensitivity of different BCR-ABL mutants to
CML resistant or intolerant to imatinib were treated different tyrosine kinase inhibitors.
with 400 mg BID (Table 2). A CCyR and partial cytoge-
netic response (PCyR) occurred in 44% of patients and IC50-fold increase (WT=1)
15%, respectively.18 MCyR was sustained in 84% of
Imatinib Bosutinib Dasatinib Nilotinib
patients after 12 months. The 24-month survival rate
was 88%. Nilotinib was well tolerated. Common WT 1 1 1 1
adverse events were neutropenia and thrombocytope- L248V 3.54 2.97 5.11 2.80
nia, which were grade 3 or 4 in 31% and 33%, respec- G250E 6.86 4.31 4.45 4.56
tively. Other adverse events were mostly transient bio- Q252H 1.39 0.31 3.05 2.64
chemical abnormalities including hypophosphatemia, Y253F 3.58 0.96 1.58 3.23
hyperglycemia, indirect hyperbilirubinemia, and elevat- E255K 6.02 9.47 5.61 6.69
ed lipase. Only three patients developed pancreatitis. E255V 16.99 5.53 3.44 10.31
Hyperbilirubinemia occurred mostly in patients with D276G 2.18 0.60 1.44 2.00
the genotype associated with Gilbert’s syndrome.17 E279K 3.55 0.95 1.64 2.05
Nilotinib has the potential to prolong QTc, but signifi- V299L 1.54 26.10 8.65 1.34
T315I 17.50 45.42 75.03 39.41
cant prolongations (>500 msec) only occurred in three
F317L 2.60 2.42 4.46 2.22
patients.19
M351T 1.76 0.70 0.88 0.44
Nilotinib has also demonstrated activity in patients F359V 2.86 0.93 1.49 5.16
with accelerated phase20 and blastic phase (Table 2). L384M 1.28 0.47 2.21 2.33
Nilotinib is approved in CML for chronic and accelerat- H396P 2.43 0.43 1.07 2.41
ed phases after imatinib failure. H396R 3.91 0.81 1.63 3.10
G398R 0.35 1.16 0.69 0.49
Bosutinib F486S 8.10 2.31 3.04 1.85
Bosutinib (SKI-6060) is an orally bioavailable inhibitor
of Bcr-Abl and SFK. In contrast to imatinib, nilotinib and Mutations can be classified as sensitive (IC50 fold increase ≤2), resistant (between
2.01 and 10 or highly resistant (>10); T315I mutation). Data from reference 39.
dasatinib, it has little if any inhibitory activity against c-
KIT and PDGFR.21 A phase II study of bosutinib in CML
CP post imatinib resistance or intolerance is ongoing.
Among 146 patients with resistance (N=101) or intoler- differences is uncertain, (e.g., ABL configuration to
ance (N=45) to imatinib treated with bosutinib for a which the agent binds, or the inhibition of Src family of
median of only 7 months, the CCyR was 32% and kinases). Although these inhibitors are active in vitro
40%, respectively, with similar response rates for against most ABL mutants, the relative potency against
patients with and without mutations.22 Over 80% of particular mutants varies (Table 4). These differences
patients are projected to maintain their response after may be useful in selecting therapy.
24 months. Diarrhea occurred frequently but was grade Changing therapy should be considered for patients
3 to 4 in only 8%. Other toxicities included nausea, who have CML post imatinib failure as defined above.
abdominal discomfort, rash and vomiting; the only sig- An important recent question arising from the success
nificant grade 3 to 4 toxicity was a rash in 7%. of new generation TKIs is whether allogeneic SCT
Neutropenia and thrombocytopenia grade 3 to 4 should be always considered as the optimal second-line
occurred in 12% and 21% of patients, respectively. therapy post imatinib failure or whether one can rely on
Bosutinib has shown activity in advanced CML phase new generation TKI as a definitive second-line therapy,
(Table 2).23 before considering allogeneic SCT. The decision should
rely on several factors: (ii) the CML phase at the time of
Selecting second generation tyrosine kinase inhibitors imatinib failure; (ii) the patient age and source of SCT
Table 3 shows some of the differences between the (matched related, unrelated, mismatched); (iii) the pres-
different TKIs. The clinical significance of some of the ence of particular mutations; (iv) the initial response to

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 85 |
14th Congress of the European Hematology Association

the new generation TKIs. In general, patients who


progress on imatinib with accelerated or blastic phase Table 5. Choice of a second generation tyrosine kinase
inhibitors.
should consider allogeneic SCT immediately, regardless
of the risk, and use new-generation TKIs as a temporary Disease characteristics
approach to debulk CML disease, hoping to improve - AP/BP: favor dasatinib (?) and combinations
outcome post allogeneic SCT; these patients should be - chronic: see below
considered for TKI maintenance post allogeneic SCT. Mutations
Patients with imatinib failure who remain in chronic - T315I → none
phase may consider allogeneic SCT as second line ther- - nilotinib IC50 > 150 nM → avoid
apy if its risks are acceptable (younger age, fully - dasatinib IC > 3 nM → avoid
matched sibling donor). Otherwise, they may consider Patient Hx
new generation TKIs as a more definitive therapy and - Hypertension, CHF, lung problems, COPD → avoid dasatinib
evaluate this choice based on the particular identified - Severe diabetes, pancreatitis Hx → avoid nilotinib
mutations, early response to the new TKIs, and the gen- - QTc problems → be cautious with all (?)
eral condition and age of the patient. Older patients (age
≥65-70 years) and those with significant commodities,
may elect to continue on the new generation TKIs and
forgo the potential of a cure with allogeneic SCT in responses may not be very durable, with the median
favor of a durable control of the disease (functional cure) time to failure of 2-4 months for patients in advanced
and a better quality of life. Failure to achieve a cytoge- stages and 20 months for patients treated in chronic
netic response and particular mutations with high IC50s phase.27 New treatment options are needed for these
are associated with shorter durations of disease control. patients. Obviously patients who have failed two TKIs
Patients with T315I mutations (10-20% of patients post should be considered for allogeneic SCT if they have a
imatinib failure; 1-3% of all patients) should consider suitable donor
immediate allogeneic SCT.
Within the choice of the second-generation TKIs, the XL-228
decision is based first on the mutations detected, and XL228 is a potent, multi-kinase inhibitor with activi-
second on the patient’s history (Table 5). Patients with a ty against BCR-ABL, SFK FGFR1-3, the aurora kinases
mutation with a high IC50 to one TKIs should be treated (A, B and C), and insulin-like growth factor receptor 1
with other alternatives (Table 5). In patients with F317L, (IGF-1R). In vitro XL-228 inhibits T315I-mutated BCR-
dasatinib and probably bosutinib are less likely to be ABL at nanomolar concentrations and inhibits prolifera-
effective than nilotinib. In patients with Y253F muta- tion of Ph+ CML and ALL cell lines with IC50s of less
tions, dasatinib and bosutinib may be better choices. than 100 nM. A phase I study is in progress. Preliminary
Patients with a history of pleural effusions or chronic results indicate that XL228 is well tolerated with no
lung disease may not be good candidates for dasatinib. MTD reported to date at doses up to 10.8 mg/kg.
Patients with a history of pancreatits, severe diabetes, Importantly, hematologic and cytogenetic responses
and QTc prolongation may not be good candidates for have been reported in 7 of 35 (20%) patients who have
nilotinib. Still, for the majority of patients there are no failed multiple prior therapies, (2 CCyR, 2 PCyR and 2
clear features that can help with the decision. Evidence minor cytogenetic responses, and 1 return to chronic
of good clinical activity is available for all agents, mak- phase). Three of the nine patients with T315I respond-
ing them reasonable choices for most patients. ed (1 CCyR, 1 MCyR, and 1 minor cytogenetic
response).29
Third line therapy
With second generation TKI, nearly half of the PHA-739538
patients do not achieve a CCyR; others eventually lose PHA-739538 is an aurora kinase inhibitor with signif-
their response. Patients who do not achieve a MCyR by icant in vitro activity against BCR-ABL, even in the pres-
12 months from the start of therapy have a probability ence of mutations, including T315I.30 There is synergy
of progression of 17% over the next 12 months com- between PHA-739358 and imatinib in cells that have
pared to only 3% for those with MCyR by 12 months.24 some remaining sensitivity to imatinib. Preliminary
Prognosis can be established even at earlier timepoints. reports of a phase II study in 7 patients treated (6 with
Among patients with no cytogenetic response at 3 T315I), showed 2 cytogenetic response (1 CCyR, 1
months, only 7% achieved a MCyR by 12 months, minor).31
compared to 67% for those with at least a minor cyto-
genetic response. These landmarks can be used to deter- AP24534
mine when a patient can be considered to have primary AP24534 is a potent multikinase inhibitor with in vitro
resistance to second generation TKI therapy. Treatment activity against unmutated BCR-ABL (IC50 0.5 nM) as
choices for patients who fail two TKI are limited. Some well as several mutated isoforms (IC50 0.5-35.7 nM),
reports have suggested that using one more second gen- including T315I (IC50 11.4 nM). It also has significant
eration TKI can be effective in some patients after fail- activity against Flt3 and c-kit among others. AP24534
ure to imatinib and one other TKI. Dasatinib used after has shown significant activity in animal models of CML
imatinib and nilotinib failure induced a response in 57% and AML. In a mutagenesis assay, single-agent AP24534
among patients in all phases, although MCyR was at concentration of 40 nM was able to suppress the
achieved in only 26%.25 Nilotinib after imatinib and growth of colonies with mutant BCR-ABL.32 This is in
dasatinib failure resulted in responses.26 Unfortunately, contrast with similar experiments with dasatinib or

| 86 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

nilotinib, where colony growth with T315I occurred et al. Discovery of N-(2-chloro-6-methyl- phenyl)-2-(6-(4-(2-
hydroxyethyl)- piperazin-1-yl)-2-methylpyrimidin-4-yl-
even at high concentrations. AP24534 may thus prevent amino)thiazole-5-carboxamide (BMS-354825), a dual Src/Abl
the development of resistance. A phase I study of kinase inhibitor with potent antitumor activity in preclinical
AP24534 in patients with CML who have failed at least assays. J Med Chem. 2004;47:6658-61.
2 TKI or with T315I mutations, as well as other hema- 8. Shah NP, Tran C, Lee FY, Chen P, Norris D, Sawyers CL. Over-
riding imatinib resistance with a novel ABL kinase inhibitor.
tologic malignancies, is ongoing. Science 2004;305:399-401.
9. O'Hare T, Walters DK, Stoffregen EP, Jia T, Manley PW,
DCC-2036 Mestan J et al. In vitro activity of Bcr-Abl inhibitors AMN107
and BMS-354825 against clinically relevant imatinib-resistant
DCC-2036 is a multikinase inhibitor that binds to dis- Abl kinase domain mutants. Cancer Res 2005;65:4500-5.
tinct structural pockets that the ABL kinase uses to 10. Hochhaus A, Baccarani M, Deininger M, Apperley JF, Lipton
switch between the inactive and active confirmations JH, Goldberg SL et al. Dasatinib induces durable cytogenetic
responses in patients with chronic myelogenous leukemia in
(i.e., “switch pockets”). DCC-2036 is selective for ABL, chronic phase with resistance or intolerance to imatinib.
Flt3, TIE2 and SFK. It inhibits phosphorylation of ABL Leukemia 2008;22:1200-6.
via a non-ATP competitive mechanisms, and impairs 11. Kantarjian H, Pasquini R, Hamerschlak N, Rousselot P,
proliferation and induces apoptosis of cells transfected Holowiecki J, Jootar S et al. Dasatinib or high-dose imatinib
for chronic-phase chronic myeloid leukemia after failure of
with unmutated BCR-ABL, or with a wide variety of first-line imatinib: a randomized phase 2 trial. Blood 2007;109:
mutants, including T315I.33 In mice models, DCC-2036 5143-50.
prolonged survival of animals with BCR-ABL-induced 12. Quintas-Cardama A, Kantarjian H, O’Brien S, Borthakur G,
Bruzzi J, Munden R, Cortes J et al. Pleural effusion in patients
disease, both myeloid and lymphoid. First-in-man, dose with chronic myelogenous leukemia treated with dasatinib
finding, phase I studies with DCC-2036 are starting. after imatinib failure. J Clin Oncol 2007;25:3908-14.
13. Shah NP, Kantarjian HM, Kim DW, Réa D, Dorlhiac-Llacer PE,
Milone JH et al. Intermittent target inhibition with dasatinib
Omacetaxine (Homoharringtonine) 100 mg once daily preserves efficacy and improves tolerabili-
Omacetaxine is a cephalotaxine ester with activity in ty in imatinib-resistant and -intolerant chronic-phase chronic
CML.34 The mechanism of action is the inhibition of myeloid leukemia. J Clin Oncol 2008;26:3204-12.
14. Cortes J, Kim DW, Raffoux E, Martinelli G, Ritchie E, Roy L et
protein synthesis. In BCR-ABL cell lines, HHT reduced al. Efficacy and safety of dasatinib in imatinib-resistant or -
the Bcr-Abl protein level and the sequential administra- intolerant patients with chronic myeloid leukemia in blast
tion of omacetaxine and imatinib was synergistic in this phase. Leukemia. 2008;22:2176-83.
model.35 The effect was seen in cells with imatinib- 15. Guilhot F, Apperley J, Kim DW, Bullorsky EO, Baccarani M,
Roboz GJ et al. Dasatinib induces significant hematologic and
resistant BCR-ABL mutants, including T315I.35 cytogenetic responses in patients with imatinib-resistant or -
Omacetaxine has clinical activity in CML post imatinib intolerant chronic myeloid leukemia in accelerated phase.
failure.36 Omacetaxine has been used to treat patients Blood 2007;109:4143-50.
16. Weisberg E, Manley PW, Breitenstein W, Brüggen J, Cowan-
who have failed therapy with at least two TKIs or who Jacob SW, Ray A et al. Characterization of AMN107, a selec-
have developed T315I mutations. In an ongoing study tive inhibitor of native and mutant Bcr-Abl. Cancer Cell 2005;
for patients with T315I mutations, 7 of 25 (28%) 7:129-41.
17. Kantarjian H, Giles F, Wunderle L, Bhalla K, O'Brien S,
patients with CML in chronic phase had a cytogenetic Wassmann B et al. Nilotinib in imatinib-resistant CML and
response to omacetaxine. The mutated clone became Philadelphia chromosome-positive ALL. N Engl J Med 2006;
undetectable in 64% of patients.37 354:2542-51.
18. Kantarjian H, Giles F, Bhalla K, et al. Nilotinib in Chronic
Myeloid Leukemia Patients in Chronic Phase (CML-CP) with
Imatinib Resistance or Intolerance: 2-Year Follow-up Results
References of a Phase 2 Study. Blood 2008;112:3238.
19. Kantarjian HM, Giles F, Gattermann N, Bhalla K, Alimena G,
1. O’Brien SG, Guilhot F, Goldman J, et al. International Palandri F et al. Nilotinib (formerly AMN107), a highly selec-
Randomized Study of Interferon Versus STI571 (IRIS) 7-Year tive BCR-ABL tyrosine kinase inhibitor, is effective in patients
Follow-up: Sustained Survival, Low Rate of Transformation with Philadelphia chromosome-positive chronic myelogenous
and Increased Rate of Major Molecular Response (MMR) in leukemia in chronic phase following imatinib resistance and
Patients (pts) with Newly Diagnosed Chronic Myeloid intolerance. Blood 2007;110:3540-6.
Leukemia in Chronic Phase (CML-CP) Treated with Imatinib 20. Le Coutre P, Giles F, Hochhaus A, et al. Nilotinib in Chronic
(IM). Blood 2008;112:86. Myeloid Leukemia Patients in Accelerated Phase (CML-AP)
2. Baccarani M, Saglio G, Goldman J, Hochhaus A, Simonsson B, with Imatinib Resistance or Intolerance: 2-Year Follow-up
Appelbaum F et al. Evolving concepts in the management of Results of a Phase 2 Study. Blood 2008;112:3229.
chronic myeloid leukemia: recommendations from an expert 21. Puttini M, Coluccia AM, Boschelli F, Cleris L, Marchesi E,
panel on behalf of the European LeukemiaNet. Blood 2006; Donella-Deana A et al. In vitro and in vivo activity of SKI-606,
108:1809-20. a novel Src-Abl inhibitor, against imatinib-resistant Bcr-Abl+
3. Kantarjian H, O'Brien S, Talpaz M, Borthakur G, Ravandi F, neoplastic cells. Cancer Res 2006;66:11314-22.
Faderl S et al. Outcome of patients with Philadelphia chromo- 22. Cortes J, Kantarjian H, Kim D-W, et al. Efficacy and Safety of
some-positive chronic myelogenous leukemia post-imatinib Bosutinib (SKI-606) in Patients with Chronic Phase (CP) Ph+
mesylate failure. Cancer 2007;109:1556-60. Chronic Myelogenous Leukemia (CML) with Resistance or
4. Quintas-Cardama A, Cortes J, O'Brien S, et al. Dasatinib Early Intolerance to Imatinib Blood 2008;112:1098.
Intervention Following Cytogenetic or Hematologic 23. Gambacorti-Passerini C, Pogliani E, Baccarani M, et al. Bosu-
Resistance to Imatinib in Patients with Chronic Myeloid tinib (SKI-606) Demonstrates Clinical Activity and Is Well
Leukemia. Cancer 2009; [Epub ahead of print] Tolerated in Patients with AP and BP CML and Ph+ ALL. Blood
5. Marin D, Milojkovic D, Olavarria E, et al. European 2008;112:1101.
LeukemiaNet criteria for failure or suboptimal response reli- 24. Tam CS, Kantarjian H, Garcia-Manero G, Borthakur G,
ably identify patients with CML in early chronic phase treat- O'Brien S, Ravandi F et al. Failure to achieve a major cytoge-
ed with imatinib whose eventual outcome is poor. Blood netic response by 12 months defines inadequate response in
2008;112:4437-44. patients receiving nilotinib or dasatinib as second or subse-
6. Jabbour E, Kantarjian HM, Jones D, Shan J, O'Brien S, Reddy quent line therapy for chronic myeloid leukemia. Blood 2008;
N et al. Imatinib mesylate dose escalation is associated with 112:516-8.
durable responses in patients with chronic myeloid leukemia 25. Quintas-Cardama A, Kantarjian H, Jones D, Nicaise C,
post cytogenetic failure on standard-dose imatinib therapy. O'Brien S, Giles F et al. Dasatinib (BMS-354825) is active in
Blood 2008. Philadelphia chromosome-positive chronic myelogenous
7. Lombardo LJ, Lee FY, Chen P, Norris D, Barrish JC, Behnia K leukemia after imatinib and nilotinib (AMN107) therapy fail-

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 87 |
14th Congress of the European Hematology Association

ure. Blood 2007;109:497-9. in Vitro Resistance by AP24534, a Pan-BCR-ABL Inhibitor.


26. Giles F, Le Coutre P, Bhalla K, et al. Efficacy and Tolerability of Blood 2008;112:726.
Nilotinib in Chronic Myeloid Leukemia Patients in Chronic 33. Van Etten RA, Chan WW, Zaleskas VM, et al. Switch Pocket
Phase (CML-CP) Who Failed Prior Imatinib and Dasatinib Inhibitors of the ABL Tyrosine Kinase: Distinct Kinome
Therapy: Updated Results of a Phase 2 Study. Blood 2008; Inhibition Profiles and in Vivo Efficacy in Mouse Models of
112:3234. CML and B-Lymphoblastic Leukemia Induced by BCR-ABL
27. Garg RJ, Kantarjian H, O'Brien SM, et al. The Use of 2nd gen- T315I. Blood 2008;112:576.
eration Tyrosine Kinase Inhibitors (TKI) after Failure to 2 Prior 34. Kantarjian HM, Talpaz M, Santini V, Murgo A, Cheson B,
TKI: Long-Term Follow-up. Blood 2008;112:2119. O'Brien SM. Homoharringtonine: history, current research,
28. Velev N, Cortes J, Champlin R, et al. Stem Cell Transplant and future direction. Cancer 2001;92:1591-605.
(SCT) for Patients (pts) with Chronic Myeloid Leukemia 35. Chen R, Gandhi V, Plunkett W. A sequential blockade strategy
(CML) Resistant to Tyrosine Kinase Inhibitors (TKI) with for the design of combination therapies to overcome oncogene
BCR-ABL Kinase Domain (KD) Mutation T315I. Blood 2008; addiction in chronic myelogenous leukemia. Cancer Res 2006;
112:2120. 66:10959-66.
29. Cortes J, Paquette R, Talpaz M, et al. Preliminary Clinical 36. Quintas-Cardama A, Kantarjian H, Garcia-Manero G, O'Brien
Activity in a Phase I Trial of the BCR-ABL/IGF-1R/Aurora S, Faderl S, Estrov Z et al. Phase I/II study of subcutaneous
Kinase Inhibitor XL228 in Patients with Ph+ Leukemias with homoharringtonine in patients with chronic myeloid leukemia
Either Failure to Multiple TKI Therapies or with T315I who have failed prior therapy. Cancer. 2007;109:248-55.
Mutation. Blood 2008;112:3232. 37. Cortes J, Khoury HJ, Corm S, et al. Safety and Efficacy of
30. Gontarewicz A, Balabanov S, Keller G, Colombo R, Graziano Subcutaneous (SC) Omacetaxine Mepesuccinate in
A, Pesenti E et al. Simultaneous targeting of Aurora kinases Imatinib(IM)-Resistant Chronic Myeloid Leukemia (CML)
and Bcr-Abl kinase by the small molecule inhibitor PHA- Patients (pts) with the T315I Mutation – Results of An
739358 is effective against imatinib-resistant BCR-ABL muta- Ongoing Multicenter Phase II Study. Blood 2008;112:3239.
tions including T315I. Blood 2008;111:4355-64. 38. Giles F, Larson R, Kantarjian H, et al. Nilotinib (Tasigna) in pts
31. Paquette R, Shah N, Sawyers C, et al. PHA-739358, an Aurora with Ph+ CML-BC Who Are Resistant or Intolerant to
Kinase Inhibitor, Induces Clinical Responses in Chronic Imatinib. Blood 2007;110:1025.
Myeloid Leukemia Harboring T315I Mutations of BCR-ABL. 39. Redaelli S, Piazza R, Rostagno R, et al. Determination of the
Blood 2007;110:1030. Activity Profile of Bosutinib, Dasatinib and Nilotinib against
32. O’Hare T, Eide CA, Adrian LT, et al. Complete Suppression of 18 Imatinib Resistant Bcr/Abl Mutants. Blood 2008;112:322.

| 88 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Clinical trial design

Designing and interpreting quality-of-life and other


patient-reported outcomes in clinical trials

F. Efficace1 A B S T R A C T
M.A. Sprangers2
Health-related quality of life and other types of patient-reported outcomes (PRO) are being increas-
1 ingly reported as important outcome measures in cancer clinical trials. While these types of outcomes
Health Outcomes Research Unit,
Italian Group for Adult Hematologic have been mainly used in studies involving patients with solid tumours, there is now increased empha-
Disease, GIMEMA Data Center, sis in also using PRO in trials of patients with hematologic diseases. A number of potentially less toxic
Rome, Italy drugs are now available and newer treatments can potentially offer patients with hematologic dis-
2
Department of Medical Psychology, eases the possibility to be treated less aggressively, making PRO more critical in evaluating treatment
Academic Medical Center, University effectiveness. In addition to traditional clinical endpoints, PRO can provide valuable information to
of Amsterdam, The Netherlands make clinical decision-making better. However, assessing PRO in clinical trials requires careful consid-
eration of a number of methodological issues already at the protocol design stage. Robust methodol-
ogy and accurate reporting of results are crucial when evaluating PRO. This is in order to provide the
Hematology Education: scientific community and health care providers with a clear and transparent message about the
the education program for the impact of a given drug on a patient’s health status. This paper provides basic guidance to all those
annual congress of the European involved in hematologic clinical trials, and gives real-world examples on the added value of including
Hematology Association PRO for a better evaluation of overall treatment effectiveness.

2009;3:89-95

n 1996, the American Society of Clinical PRO as it applies to a wider set of outcomes

I Oncology (ASCO) pointed to the key


role health-related quality-of-life
(HRQOL) assessment plays in cancer
(including HRQOL). The US Food and Drug
Administration (FDA) define a PRO as a
“measurement of any aspect of a patient’s
research.1 Indeed, major research organiza- health status that comes directly from the
tions routinely consider inclusion of such patient (i.e., without the interpretation of
outcomes in randomized controlled trials the patient’s responses by a physician or
(RCTs).2 One of the National Cancer anyone else)”.8 PRO include a wide spec-
Institute Strategic Objectives is to ensure trum of measures, ranging from single item
the best outcome for all, including improv- instruments assessing a specific health
ing the “quality of life for cancer patients, domain (e.g., pain or fatigue) to broader
survivors and their families”.3 The World multidimensional constructs, such as
Health Organization (WHO) also points out HRQOL. It is important to make such a dis-
that the main goal of cancer control is “a tinction, as readers will find both terms in
reduction in the incidence of the disease the literature.
and of the associated morbidity and mortal- PRO are now highly valued by stakehold-
ity, as well as improved quality of life for ers. The FDA recently published a docu-
cancer patients and their families”.4 ment with the main goal of providing the
medical community its view on PRO
Health-related quality of life and patient instruments as effective endpoints in clini-
reported outcomes: what to measure? cal trials, and providing medical product
It is important to clarify what is meant by developer guidance on using PRO data used
HRQOL and Patient-Reported Outcomes to support claims in product labeling.8
(PRO), both of which are now often report- Using PRO as an outcome measure in a
ed in the medical literature. Defining clinical trial is, in essence, the only way of
HRQOL is challenging but there is now obtaining evidence-based data from the
general agreement that it refers to key patient’s view on the effect of a treatment.
areas, including minimally physical, psy- As stated by the FDA, some “treatment
chological and social functioning, as well as effects are known only to the patient,” and
symptoms induced by the disease and its such information can be lost when the
treatment.5-6 patient’s perspective “is filtered through a
More recently, the term “patient-reported clinician’s evaluation of the patient’s
outcome” (PRO) has been introduced in the response to clinical interview questions”.8
literature to describe a broader set of There is now robust evidence indicating
parameters self-reported by the patient.7 that PRO, for example, are independent
Here, we use the broader definition of prognostic factors of survival beyond and

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 89 |
14th Congress of the European Hematology Association

above clinical and laboratory data in patients with ic diseases (including acute leukemia, chronic lympho-
advanced diseases including those with hematological cytic leukemia, immune thrombocytopenic purpura
malignancies.9-11 and myelodysplastic syndromes) are paying greater
attention to HRQOL issues and are advocating more
What is the added-value of measuring PRO in hematologi- research into this area.18-22
cal clinical trials? Although little research has been conducted in
Several RCTs in patients with solid tumors have suc- hematology, and the number of studies has only start-
cessfully implemented PRO and have provided addi- ed to increase over the last few years, PRO assessment
tional robust data to understand better the overall has been shown to be feasible and of value in provid-
treatment effectiveness from the patients’ perspective. ing unique information. Table 1 provides a brief sum-
Some of these data have also served as a basis for drug mary of the key findings related to the added-value of
approval (or in support of) by the FDA.12-14 measuring PRO in selected RCTs of three major hema-
The number of trials conducted in patients with tological disease sites.
hematological diseases is substantially lower. There
are several reasons to explain this gap.15-17 Recently, Chronic lymphocytic leukemia
however, much emphasis is being placed on PRO Eichhorst and colleagues23 recently conducted a large
issues in hematological research. For example, recent RCT comparing Fludarabine (F) versus F plus
international recommendations for various hematolog- cyclophosphamide (FC) as first line therapy in younger

Table 1. Selected prospective RCTs, including PRO in patients with leukemia and myelodysplastic syndromes. (Adapted
from Efficace et al.17 and Caocci et al.53)

Authors Year of Overall no. of Disease Treatment PRO PRO measures Summary of traditional Summary of PROs
publication patients outline endpoint used clinical outcomes

Eichhorst et al.23,24 2007 375 CLL Fludarabine (F) Secondary EORTC QLQ-C30 F plus C is more effective Overall, HRQOL
with or without than F alone as first was comparable
Cyclophosphamide (C) line therapy for CLL patients between treatment arms
in terms of: complete remission;
overall response; PFS;
treatment-free survival time
Catovsky et al. 25 2007 777 CLL Fludarabine versus Secondary EORTC QLQ-C30 Better complete and Overall, HRQOL was
Fludarabine plus overall response rates, comparable amongst
Cyclophosphamide versus as well as progression-free treatment arms
Chlorambucil survival at 5 years for patients
with Fludarabine plus
Cyclophosphamide than Fludarabine
and better than with Chlorambucil
Hahn et al.27 2003 1,106 CML Imatinib (IM) versus Secondary FACT-BRM; IM significantly superior The Trial Outcome
and O’Brien26 interferon (IFN) -alfa plus Euro QoL-5D to IFN-alfa plus Ara-C Index of the FACT-BRM
low dose cytarabine in terms of CHR, MCR, was much higher in the IM
CCR, FFP, toxicity Group throughout
all study period
Kornblith et al. 29 2002 191 MDS Azacitidine (AZA) Secondary EORTC QLQ- C30; AZA yielded a higher Patients on the AZA
and Silverman et al.28 versus supportive care Mental health inventory; response rate, arm experienced
patient’s perception of reduced risk of leukemic significantly greater
improvement transformation and improvement in fatigue,
improved survival physical functioning,
positive affect, psychological
distress.Also patients
who crossed over from the
supportive care arm to the
azacitidine arm had
significantly improved QoL
Kantarijan et al.30 2006 170 MDS Decitabine plus Secondary EORTC QLQ-C30 Higher overall response Decitabine yielded
supportive care versus rate and longer median statistically better
supportive care alone time trend to AML in patients results for global health
treated with decitabine compared status, fatigue, and
to those on supportive care dyspnea in comparison
with supportive care
FACT-BRM: functional assessment of cancer therapy-biologic response modifiers; EORTC QLQ-C30: european organization for research and treatment of cancer quality
of life questionnaire; CML: chronic myeloid leukemia; CLL: chronic lymphocytic leukemia; MDS: myelodysplastic syndromes.

| 90 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

patients with CLL showing better response and pro- about the benefit of decitabine on patient’s HRQOL.
gression-free survival for the FC arm. Nevertheless, Overall, these studies provide good examples of
toxicity did not favor the FC arm, showing worse data, how PRO implementation in a RCT setting is feasible
mainly in terms of leukocytopenia and myelotoxicty. and can augment investigators’ knowledge of treat-
In this trial, HRQOL was used as a secondary end- ment effectiveness.
point, and the results obtained were of particular value
in understanding that worse toxicity did not translate Practical considerations in designing and reporting PRO
into worse HRQOL of patients treated in the FC arm. in clinical trials
The full analysis of HRQOL over time between the Information about side effects, symptoms and treat-
two treatment arms showed no significant differences, ment options are important to cancer patients as they
allowing the conclusion that the beneficial clinical enable them to make informed treatment decisions.
effects of FC was not obtained at the expense of detri- Cancer patients require information related to survival
mental effects on patient’s HRQOL.24 Such informa- estimates, as well as HRQOL issues.31 Providing
tion could have been lost if this outcome had not been patients with such evidence-based information is,
included in the original trial. Catovsky and colleagues25 therefore, of paramount importance. Thus, including
compared F versus FC versus Chlorambucil and includ- PRO as an effectiveness endpoint in a clinical trial set-
ed HRQOL as a secondary endpoint in the trial. While ting could potentially provide invaluable information
FC arm patients reported better clinical outcomes, a related to treatment side effects from the patients’ per-
preliminary analysis showed that HRQOL was gener- spectives.
ally comparable among treatment arms. The authors Measuring PRO should always follow the same rigor
explicitly mentioned that a full HRQOL report would as applied to measuring any other traditional clinical
follow, thus providing additional details on this analy- endpoint in clinical research. In phase I trials, PRO is
sis. However, preliminary HRQOL data of this trial not a relevant outcome measure but, in some phase II
lent additional support that FC treatment in CLL studies, measuring the patient’s perspective can be of
patients can be obtained with no major impact on value (e.g., in hypothesis generating for future phase III
patient’s HRQOL when compared to other standard studies). In phase III studies, however, inclusion of
treatments. PRO can always be of great value and should be con-
sidered as a possible study outcome. There is also gen-
Chronic myeloid leukemia eral agreement that PRO inclusion is relevant in virtu-
O’Brien and colleagues26 included HRQOL as a sec- ally all symptom management trials.32 The following
ondary endpoint in a trial comparing standard inter- paragraphs mainly address issues relevant in phase III
feron treatment with Imatinib (IRIS study), with the studies. If PRO are to fulfill their potential of allowing
main goal of providing evidence-based data of the health-care providers to make informed decisions
possible benefits in terms of HRQOL for the newer about the overall value and impact of a given treat-
treatment. When the full HRQOL analysis was pub- ment, investigators should pay careful attention to a
lished in a separate report by Hahn and colleagues,27 it number of methodological issues. Actually, evaluating
was clear that the use of Imatinib not only provided PRO in clinical trials requires making a number of chal-
superior traditional outcomes over the interferon- lenging decisions. It is fair to mention that previous
based treatment, but also had greater beneficial effects work investigating the quality of PRO assessment in
in a number of HRQOL areas. The effect size found oncology over the last 20 years have found a number
for the difference in HRQOL outcomes between treat- of methodological drawbacks that, in many cases,
ment arms was one of the largest ever shown in any have hampered a critical appraisal of results.33-36 An edi-
previous HRQOL trial-based report.27 Interestingly, torial which appeared in May 2002 in the official jour-
PRO data of this study were also used to support FDA nal of the ASCO37 stated that it is “disappointing” that,
approval of Imatinib for use in newly diagnosed, despite the fact that thousands of patients enrolled in
chronic phase CML.12 cancer clinical trials with a PRO component, “there are
relatively few examples of formal quality of life meas-
Myelodysplastic syndromes urement that have influenced individual patient deci-
Silverman and colleagues28 included HRQOL as a sion-making or treatment policies.” However, a signif-
secondary endpoint in a large trial comparing azaciti- icant learning curve, in terms of accuracy of PRO
dine (AZA) versus supportive care, demonstrating that reporting since 1990, has been recently demonstrated
treatment with AZA provided a delayed time to dis- in cancer clinical trials conducted in major solid
ease progression and death. A separate HRQOL paper tumors,38 thus supporting the hypothesis that results
was also published by Kornblith and colleagues,29 indi- from recent studies are more likely to support clinical
cating that treatment with AZA also provided benefi- decision-making than earlier published studies.
cial effects in terms of reduced fatigue, physical func- Some administrative and methodological decisions
tioning, positive effect and psychological distress. should be taken already at the time of protocol writ-
Another RCT compared decitabine plus supportive ing, while others are mainly relevant when reporting
care versus supportive care alone,30 led to the conclu- and disseminating PRO results.
sions that various HRQOL aspects can be improved by
using decitabine in addition to supportive care. At the stage of protocol writing
However, this trial reported few details on the design PRO can either be used as a secondary or primary
of HRQOL assessment and related outcomes, thus it is endpoint in a given trial but, in both instances, they
currently challenging to draw definitive conclusions should be an integral part of the research protocol,

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 91 |
14th Congress of the European Hematology Association

with a number of issues well addressed and planned at to patients (e.g., when and how);
this early stage. The first step is formulating a ration- - identify one responsible person for PRO data collec-
ale for PRO assessment in the particular trial and spec- tion in each participating institution to facilitate liai-
ifying what this type of outcome would add to the pri- son with the coordinating center;
mary endpoint of the study (in case PRO is planned as - provide regular feedback about study progress on
a secondary endpoint). Selecting the most appropriate compliance to study co-investigators.
instrument (e.g., questionnaire) for the particular trial It is worthy of note, that, on the basis of these rec-
deserves attention and must be justified in the proto- ommendations, recently conducted trials obtained a
col. Selecting the “right” questionnaire is a fundamen- good level of compliance, also in patients with
tal step in designing and conducting a PRO assessment advanced hematological malignancies.10
and needs careful evaluation of various aspects. Another issue to be addressed during protocol devel-
Questions need to be asked, such as: is the content of opment is the a priori definition of what constitutes a
the questionnaire appropriate to the research ques- “minimally important difference” (MID) in the PRO
tion? Does the questionnaire yield robust psychomet- measure of interest, and thus, to address clinical signif-
ric properties, in terms of validity, reliability and icance of outcomes. Using PRO poses unique problems
responsiveness? How interpretable are the scores? Is inherent to their subjective nature. For example, what
the questionnaire easy to administer? Besides these, is the meaning of a given statistically significant differ-
the cultural validity of the questionnaire has also to be ence in terms of HRQOL from a patient’s perspective?
verified, in case it has not been validated for the same Does a statistically significant difference in a HRQOL
population under study. The choice of instruments is domain between treatment arms necessarily reflect a
analogous to choosing laboratory tests, that is, it is subjectively meaningful difference perceived by
important to choose the most appropriate and sensi- patients? The challenging issue is how to evaluate “a
tive (laboratory) test for the purpose at hand.7 Detailed tangible benefit (improvement in health) with an
guidelines on how to make this choice have been pub- intangible construct (HRQOL)”.43 Since statistical sig-
lished and will assist investigators when designing nificance is dependent on sample size, it is possible
future studies.39 that a 2 to 3 unit change on a 0 to 100 PRO scale,
Nevertheless, the choice of instrument is only one results in a significant p-value if the results were based
aspect of the multi-faceted process of obtaining valid on a large sample. Most likely, this finding would not
and reliable data in a clinical trial. Another relevant be clinically meaningful. As a comprehensive discus-
issue is the problem of missing data. Difficulties with sion on assessing clinical significance is beyond the
data collection and compliance have historically been scope of this chapter, we would like to highlight that a
considered the major barriers to the successful imple- number of methods are available, which can be divid-
mentation of PRO in clinical trials.40 PRO data are col- ed into anchor-based and distribution-based approach-
lected at different time points during the course of the es.44-45 To illustrate, previous work has shown that the
study and missing data (i.e., questionnaires not com- MID for one of the most widely used cancer-specific
pleted and/or returned) from different scheduled questionnaires, the EORTC QLQ-C30, equals a 10-
assessments are unavoidable, mainly, due to patients’ point shift on its 0-100 response scale.46
health conditions and/or administrative failures. For further details on the main steps to be taken into
However, as lost data might not be missing at random, consideration at an early stage of protocol writing to
they cannot be ignored without introducing bias in ensure a successful PRO implementation, we refer the
outcome interpretation. If the pattern of missing data reader to other relevant documents in this area.42,47-48
is systematically different between treatment arms,
such information should alert investigators in further At the stage of reporting and disseminating PRO findings
exploring this issue and check, for example, if this Some issues need to be addressed, particularly at the
could be associated with the specific treatment (e.g., time of disclosing and disseminating PRO findings to
more toxic) of that trial arm. Given the importance of ensure consistency of reporting. First, when collecting
the representativeness of the data of the trial popula- PRO data in a trial, the amount of information would
tion, investigators are recommended to take actions to be better published also in a separate report from the
minimize the number of missing data during the main clinical findings. In this scenario of disclosing
study.40 A number of steps can be taken, including:40-42 PRO (when this has been used as a secondary endpoint
- the collection of PRO data should not be presented of a trial), the recommendation would be to include
as an “optional” part of the study rather as an inte- the major PRO results in the main original paper, and
gral and mandatory part of the trial; additionally publish a separate paper providing full
- avoid a PRO assessment including an unnecessary details of PRO analysis; thus, allowing the medical
large number of items. Clearly, the number of items community a critical appraisal of the results.
needs to be evaluated in light of the target popula- In Table 2, we report a number of practical basic
tion (e.g., patient’s age and disease), frequency of issues that investigators should consider when report-
assessments and resources available. For example, ing PRO assessment in clinical trials. We do not intend
older and more ill patients, a study design with to provide a comprehensive list of all detailed issues
many assessments, and a study with few resources, that need to be ideally reported, rather we provide a
would require a brief questionnaire; brief pragmatic guide on the main topics deserving
- provide brief guidelines to all investigators stressing attention when reporting PRO results. These issues are
the importance of PRO data collection and giving taken from the Minimum standard checklist for evaluating
practical and brief indication for PRO administration HRQOL outcomes in cancer clinical trials that has been

| 92 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

Table 2. Minimum standard criteria for PRO reporting in clinical trials. (Adapted from Efficace et al.49)

Items related to the conceptual design

A priori hypothesis stated


(Do the authors have a pre-defined PRO endpoint and/or stated expected changes due to the specific treatment?) Yes r No r N/A r†
Rationale for instrument reported
(Do the authors report a rationale for selecting a specific PRO measure?) Yes r No r

Items related to the Measurement

Psychometric properties reported*


(Assessed if a previously validated measure was used or psychometric properties were reported or referenced in the paper) Yes r No r
Cultural validity verified
(Assessed if the measure was validated for the specific study population) Yes r No r N/A r‡
Adequacy of domains covered
(Assessed if the measure covered, at least, the main PRO dimensions relevant for a generic cancer population and/or
according to the specific research question) Yes r No r

Items related to the methodology

Instrument administration reported


(Do the authors report who and/or in which clinical setting the PRO instrument was administered?) Yes r No r
Baseline compliance reported*
(Do the authors report the number of patients by treatment arms providing the initial PRO assessment?) Yes r No r
Timing of assessments documented
(Do the authors report the PRO timing of assessment during the trial?) Yes r No r
Missing data documented*
(Do the authors report the extent of PRO missing data during the trial between treatment arms?) Yes r No r

Items related to the Interpretation of outcomes

Clinical significance addressed


(Do the authors discuss PRO data being clinically significant from a patient’s perspective and not simply statistically significant?) Yes r No r
Presentation of results in general
(Do the authors discuss PRO findings giving any comment regardless of the results) Yes r No r
Each item can be scored as ‘yes’ (giving a score of 1) or ‘no’ (giving a score of 0), with higher sum scores indicating more robustness of the outcomes. * Although some
items might have different importance according to the specific study questions, these basic items should always be documented in the report; †if a study explicitly states an
exploratory PRO evaluation; ‡ if the PRO measure is validated in the same population as the one of the trial.

previously developed based on good practice in report- defined since baseline, for example, every second
ing HRQOL studies and was published in the Journal of week since baseline. Conversely, timing of assess-
Clinical Oncology in 2003.49 It was purposely devised to ments may be less critical in an “event-based design,”
help both investigators and readers in general. This where the assessments are defined in relationship to
checklist is primarily intended for reviewing and facil- events, for example, at the end of each chemotherapy
itating a critical appraisal and interpretation of PRO cycle, or following relapse and/or recurrence. Finally, it
reports and for guiding investigators when designing, is worth noting that the investigator should use this
and most importantly, writing PRO reports from a clin- checklist as a flexible and basic tool, putting more
ical trial. The items are devised to have a dichotomous emphasis on various items on a case-by-case basis.
answer (yes or no), and each can be scored as “yes”
(giving a score of 1) or “no” (giving a score of 0), with Conclusions
higher sum scores indicating more robustness of the In conclusion, robust methodology and accurate
outcomes. This tool is of pragmatic use, and has been reporting of data is crucial when evaluating PRO in
adopted in several studies to evaluate consistency and clinical trials in order to provide the scientific commu-
level of reporting. It has been shown to be sensitive in nity and health care providers with a clear and trans-
picking up differences in quality reporting over parent message about the impact of a given drug on
time.38,50-52 It is important to note that good reports may the patient’s health status. Caution is required when
have different emphases and some issues might have interpreting PRO data. Publications in this area stem-
different importance according to the specific study ming from a poor study design or simply reporting
questions. For example, reporting “timing of assess- inadequate information can potentially mislead read-
ment” and possibly time windows for PRO assessment ers when interpreting study outcomes; educational
is very important in studies adopting a “time-based efforts in this area are very important. By providing
design,” where the exact timing of assessments is basic guidance on the main issues deserving attention

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 93 |
14th Congress of the European Hematology Association

when interpreting the robustness of studies in this treatment of chronic lymphocytic leukemia: a report from the
International Workshop on Chronic Lymphocytic Leukemia
area, clinicians (and readers in general) will be able to updating the National Cancer Institute-Working Group 1996
evaluate PRO results critically. It is our hope that such guidelines. Blood 2008;111:5446-56.
practical guidelines will stimulate stringent and robust 21. Appelbaum FR, Rosenblum D, Arceci RJ, Carroll WL, Breitfeld
PRO-based RCTs in hematologic cancer. PP, Forman SJ et al. End points to establish the efficacy of new
agents in the treatment of acute leukemia. Blood 2007; 109:
1810-6.
22. Cheson BD, Greenberg PL, Bennett JM, Lowenberg B,
References Wijermans PW, Nimer SD et al. Clinical application and pro-
posal for modification of the International Working Group
1. ASCO. Outcomes of cancer treatment for technology assess- (IWG) response criteria in myelodysplasia. Blood 2006;108:
ment and cancer treatment guidelines. American Society of 419-25.
Clinical Oncology. J Clin Oncol 1996;14:671-9. 23. Eichhorst BF, Busch R, Hopfinger G, Pasold R, Hensel M,
2. Osoba D. The Quality of Life Committee of the Clinical Trials Steinbrecher C, et al. Fludarabine plus cyclophosphamide ver-
Group of the National Cancer Institute of Canada: organiza- sus fludarabine alone in first-line therapy of younger patients
tion and functions. Qual Life Res 1992;1:211-8. with chronic lymphocytic leukemia. Blood 2006;107:885-91.
3. National Cancer Institute. The NCI Strategic Plan for Leading 24. Eichhorst BF, Busch R, Obwandner T, Kuhn-Hallek I,
the Nation. To Eliminate the Suffering and Death due to Herschbach P, Hallek M, German CLL Study Group. Health-
Cancer National Institutes of Health Publication No. 06-5773 related quality of life in younger patients with chronic lym-
[document on the Internet]. Available from: http://strategic- phocytic leukemia treated with fludarabine plus cyclophos-
plan.nci.nih.gov/pdf/nci_2007_strategic_plan.pdf. phamide or fludarabine alone for first-line therapy: a study by
4. Stewart BW, Kleihues P, editors. World Health Organization. the German CLL Study Group. J Clin Oncol 2007;25:1722-31.
World Cancer Report. Lyon, IARC Press; 2003. 25. Catovsky D, Richards S, Matutes E, Oscier D, Dyer MJ,
5. Osoba D. Lessons learned from measuring health-related qual- Bezares RF, et al. Assessment of fludarabine plus cyclophos-
ity of life in oncology. J Clin Oncol 1994;12:608-16. phamide for patients with chronic lymphocytic leukaemia
6. Schumacher M, Olschewski M, Schulgen G. Assessment of (the LRF CLL4 Trial): a randomised controlled trial. Lancet
quality of life in clinical trials. Stat Med 1991;10:1915-30. 2007;370:230-9.
7. Osoba D. Translating the science of patient-reported out- 26. O’Brien SG, Guilhot F, Larson RA, Gathmann I, Baccarani M,
comes assessment into clinical practice. J Natl Cancer Inst Cervantes F, et al. Imatinib compared with interferon and low-
Monogr 2007;37:5-11. dose cytarabine for newly diagnosed chronic-phase chronic
8. US Food and Drug Administration: Guidance for Industry. myeloid leukemia. N Engl J Med 2003;348:994-1004.
Patient-reported outcome measures: Use in medical product 27. Hahn EA, Glendenning GA, Sorensen MV, Hudgens SA,
development to support labeling claims [document on the Druker BJ, Guilhot F, et al. Quality of life in patients with
Internet]. Available from: http://www.fda.gov/cder/guid- newly diagnosed chronic phase chronic myeloid leukemia on
ance/5460dft.pdf. imatinib versus interferon alfa plus low-dose cytarabine:
9. Efficace F, Innominato P, Bjarnason G, Coens C, Humblet Y, results from the IRIS Study. J Clin Oncol 2003;21:2138-46.
Tumolo S et al. Validation of patient’s self-reported social func- 28. Silverman LR, Demakos EP, Peterson BL, Kornblith AB,
tioning as an independent prognostic factor for survival in Holland JC, Odchimar-Reissig R et al. Randomized controlled
metastatic colorectal cancer patients. Results of an internation- trial of azacitidine in patients with the myelodysplastic syn-
al study by the Chronotherapy Group of the European drome: a study of the cancer and leukemia group B. J Clin
Organisation for Research and Treatment of Cancer. J Clin Oncol 2002;20:2429-40.
Oncol 2008;26:2020-6. 29. Kornblith AB, Herndon JE, Silverman LR Silverman LR,
10. Dubois D, Dhawan R, van de Velde H, Esseltine D, Gupta S, Demakos EP, Odchimar-Reissig R et al. Impact of Azacitidine
Viala M, et al. Descriptive and prognostic value of patient- on the Quality of Life of patients with myelodysplastic syn-
reported outcomes: the bortezomib experience in relapsed and drome treated in a randomized phase III trial: a Cancer and
refractory multiple myeloma. J Clin Oncol. 2006;24:976-82. Leukemia Group B study. J Clin Oncol 2002;20:2441-52.
11. Gotay CC, Kawamoto CT, Bottomley A, Efficace F. The prog- 30. Kantarjian H, Issa JP, Rosenfeld CS, Bennett JM, Albitar M,
nostic significance of patient-reported outcomes in cancer clin- DiPersio J et al. Decitabine Improves Patient Outcomes in
ical trials. J Clin Oncol 2008;26:1355-63. Myelodysplastic Syndromes. Results of a Phase III Randomiz-
12. Rock EP, Kennedy DL, Furness MH, Pierce WF, Pazdur R, ed Study. Cancer 2006;106:1794-803.
Burke LB. Patient-reported outcomes supporting anticancer 31. Hagerty RG, Butow PN, Ellis PA, Lobb EA, Pendlebury S,
product approvals. J Clin Oncol 2007;25:5094-9. Leighl N et al. Cancer patient preferences for communication
13. Osoba D, Tannock IF, Ernst DS, et al. Health-related quality of of prognosis in the metastatic setting. J Clin Oncol 2004;22:
life in men with metastatic prostate cancer treated with pred- 1721-30.
nisone alone or mitoxantrone and prednisone. J Clin Oncol 32. Lipscomb J, Reeve BB, Clauser SB, Abrams JS, Bruner DW,
1999;17:1654-63. Burke LB et al, Patient-reported outcomes assessment in can-
14. Ganz PA, Gotay CC. Use of patient-reported outcomes in cer trials: taking stock, moving forward. J Clin Oncol 2007;
phase III cancer treatment trials: lessons learned and future 25:5133-40.
directions. J Clin Oncol 2007;25:5063-9. 33. Bottomley A, Therasse P. Quality of life in patients undergo-
15. Efficace F, Novik A, Vignetti M, Mandelli F, Cleeland CS. ingsystemic therapy for advanced breast cancer: Lancet Oncol
Health-related quality of life and symptom assessment in clin- 2002;3: 620-8.
ical research of patients with haematological malignancies: 34. Bottomley A, Efficace F, Thomas R, Vanvoorden V, Ahmedzai
where are we now and where do we go from here? Haema- S. Health-related quality of life in non small-cell lung cancer:
tologica 2007;92:1596-8. methodologic issues in randomized controlled trials. J Clin
16. Molica S. Quality of life in chronic lymphocytic leukemia: a Oncol 2003;21:2982-92.
neglected issue. Leuk Lymphoma 2005;46:1709-14. 35. Efficace F, Bottomley A, Vanvoorden V, Blazeby JM.
17. Efficace F, Kemmler G, Vignetti M, Mandelli F, Molica S, Methodological issues in assessing health-related quality of
Holzner B. Health-related quality of life assessment and life of colorectal cancer patients in randomized controlled tri-
reported outcomes in leukaemia randomised controlled trials - als. Eur J Cancer. 2004;40:187-97.
a systematic review to evaluate the added value in supporting 36. Efficace F, Bottomley A, van Andel G. Health-related quality of
clinical decision making. Eur J Cancer 2008;44:1497-506. life in prostate carcinoma patients: a systematic review of ran-
18. Rodeghiero F, Stasi R, Gernsheimer T, Michel M, Provan D, domized controlled trials. Cancer 2003;97:377-88.
Arnold DM et al. Standardization of terminology, definitions 37. Levine MN, Ganz PA. Beyond the development of quality of
and outcome criteria in immune thrombocytopenic purpura life instruments: where do we go from here? J Clin Oncol
(ITP) of adults and children: Report from an International 2002; 20:2215-6.
Working Group. Blood 2008:Nov 12. [Epub ahead of print]. 38. Efficace F, Osoba D, Gotay C, Sprangers M, Coens C,
19. Tefferi A, Barosi G, Mesa RA, Cervantes F, Deeg HJ, Reilly JT Bottomley A. Has the quality of health-related quality of life
et al. International Working Group (IWG) consensus criteria reporting in cancer clinical trials improved over time? Towards
for treatment response in myelofibrosis with myeloid meta- bridging the gap with clinical decision making. Ann Oncol
plasia, for the IWG for Myelofibrosis Research and Treatment 2007; 18:775-81.
(IWG-MRT). Blood 2006;108:1497-503. 39. Fitzpatrick R, Davey C, Buxton MJ, Jones DR et al. Evaluating
20. Hallek M, Cheson BD, Catovsky D, Caligaris-Cappio F, patient-based outcome measures for use in clinical trials.
Dighiero G, Döhner H et al. Guidelines for the diagnosis and Health Technol Assess 1998;2:1-74.

| 94 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

40. Fayers P, Hays R. Assessing Quality of Life in Clinical trials. comes: the example of health related quality of life-a European
New York (NY): Oxford University Press 2005. guidance document for the improved integration of health
41. Hürny C, Bernhard J, Joss R, Willems Y, Cavalli F, Kiser J et al. related quality of life assessment in the drug regulatory
Feasibility of quality of life assessment in a randomized phase process. Drug Inf J 2002;36:209-38.
III trial of small cell lung cancer--a lesson from the real world- 49. Efficace F, Bottomley A, Osoba D Gotay C, Flechtner H,
-the Swiss Group for Clinical Cancer Research SAKK. Ann D'haese S, Zurlo A. Beyond the development of health-relat-
Oncol 1992;3:825-31. ed quality of life (HRQOL) measures. A checklist for evaluat-
42. Fayers PM, Hopwood P, Harvey A, Girling DJ, Machin D, ing HRQOL outcomes in cancer clinical trials-does HRQOL
Stephens R et al. Quality of life assessment in clinical trials-- evaluation in prostate cancer research inform clinical decision-
guidelines and a checklist for protocol writers: the U.K. making? J Clin Oncol 2003;21: 3502-11.
Medical Research Council experience. MRC Cancer Trials 50. Cocks K, King MT, Velikova G, Fayers PM, Brown JM.
Office. Eur J Cancer 1997;33:20-8. Quality, interpretation and presentation of European
43. Sloan J, Symonds T, Vargas-Chanes D, et al: Practical guide- Organisation for Research and Treatment of Cancer quality of
lines for assessing the clinical significance of health-related life questionnaire core 30 data in randomised controlled trials.
quality of life changes within clinical trials. Drug Inf J 2003;
Eur J Cancer 2008;44:1793-8.
37:23-32.
44. Sprangers MA, Moinpour CM, Moynihan TJ, Patrick DL, 51. van Meerbeeck JP, Gaafar R, Manegold C, Van Klaveren RJ,
Revicki DA. Assessing meaningful change in quality of life Van Marck EA, Vincent M et al. Randomized phase III study
over time: a users’ guide for clinicians. Mayo Clin Proc 2002; of cisplatin with or without raltitrexed in patients with malig-
77:561-71. nant pleural mesothelioma: an intergroup study of the
45. Lydick E, Epstein RS. Interpretation of quality of life changes. European Organisation for Research and Treatment of Cancer
Qual Life Res 1993;2:221-6. Lung Cancer Group and the National Cancer Institute of
46. Osoba D, Rodrigues G, Myles J, Zee B, Pater J. Interpreting the Canada. J Clin Oncol 2005;23,6881-9.
significance of changes in health-related quality-of-life scores. 52. Blazeby JM, Avery K, Sprangers M, Pikhart H, Fayers P,
J Clin Oncol 1998;16:139-44. Donovan J. Health-related quality of life measurement in ran-
47. Gotay CC, Korn EL, McCabe MS, Moore TD, Cheson BD. domized clinical trials in surgical oncology. J Clin Oncol
Quality-of-life assessment in cancer treatment protocols: 2006;24:3178-86.
research issues in protocol development. J Natl Cancer Inst 53. Caocci G, La Nasa F, Efficace F. Health-related quality of life
1992;84:575-9. and symptom assessment in patients with myelodysplastic
48. Chassany O, Sagnier P, Marquis P, et al. Patient-reported out- syndromes. Exp Rev Hematol 2009;2:69-80.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 95 |
Clinical trial design

The use of genomics in clinical trial design

R. Simon
Importance of prognostic and predictive have good outcome whether or not they
classifiers needed or benefited from tamoxifen.
National Cancer Institute
Bethesda, Maryland 20892-7434, Genomics offers great opportunities to Developing a prognostic classifier using a
USA improve cancer therapeutics by identifying group of patients that are homogeneous
key molecular targets and by selecting the with regard to both treatment and the prog-
right treatment for the right patient. In this nostic factors that are incorporated into cur-
paper, we shall focus on the latter objective. rent practice standards is very important for
Hematology Education: Current practice in oncology involves treat- ensuring that the classifier has potential
the education program for the ing many patients who do not benefit for medical utility to aid in treatment selection.
annual congress of the European each one who does. For example, patients We use the term classifier to indicate any
Hematology Association with an early stage solid tumor may have an categorization of tumors based one or more
80% or better long-term disease free survival biological measurements. If there are two
2009;3:96-100 with local therapy. There would often be categories, such as good prognosis or poor
substantial interest in identifying a prognosis, then the classifier is called a bina-
chemotherapy regimen that increased the ry classifier. A binary predictive classifier
chance from 80-85%. This would represent would categorize patients into those who
a 25% relative reduction in the hazard of are likely benefit from the specified thera-
recurrence. However, 80% of the patients peutic versus those less likely to benefit. A
do not require systemic therapy, and the prognostic or predictive index provides a grad-
absolute improvement of 5 percentage ed or continuous value based on one or more
points results in treating 20 patients for each biological measurements. For example, both
one who benefits. OncotypeDx and Mammaprint tests are
Because cancer is a life threatening dis- prognostic indices. For purposes of clinical
ease, treating 20 patients to benefit 1 is trial design or patient management, cut-
acceptable, if the benefit is sufficiently great. points are often introduced to convert a
However, it would be better for the patients prognostic index to a prognostic classifier.
if we could prospectively identify those Prognostic or predictive classifiers or
patients whose prognosis with local therapy indices may be based on single genes, single
alone is sufficiently good that they do not proteins, or on expression levels of many
require systemic therapy. The OncotypeDx genes. For example, OncotypeDx is based
and Mammaprint tests are examples of com- on expression of 21 genes. In order to have a
mercially available prognostic classifiers for classifier or index, however, it is essential
patients with early stage breast cancer.1-2 that the multiple components be combined
It would also be useful to be able to classi- in a completely defined manner. Many gene
fy patients accurately whose prognosis with expression indices are weighted averages of
local therapy is not as good into those who expression levels of multiple genes with the
are and those who are not likely to benefit weights explicitly defined. In order to use a
from a specific therapeutic regimen. Such a classifier for new patients, it is not sufficient
treatment specific test is generally called a just to have a set of genes. The patient can-
predictive classifier. This is not an accurate use not be classified unless the test defines how
of language, as prognostic classifiers are used the expression levels are to be combined and
for predicting outcome, but it is well estab- what cut-points to use for grouping the
lished to reserve the phrase predictive classi- index values into prognostic categories.
fier or predictive marker for those tests asso- Many publications reporting the develop-
ciated with predicting benefit for a specific ment of prognostic gene expression signa-
therapy. tures provide only a set of genes, not a well-
Useful prognostic classifiers are them- defined classifier that can be tested on inde-
selves treatment specific, but not in quite the pendent data.
same way as predictive classifiers. The Although gene expression profiling is
OncotypeDx prognostic classifier was devel- actively used to develop prognostic classi-
oped for patients with axillary node nega- fiers, much new drug development is based
tive, estrogen receptor positive breast cancer on single gene predictive biomarkers.
who received local therapy plus tamoxifen. Estrogen receptor expression is a classic pre-
It does not identify the patients whose dictive biomarker for effectiveness of
tumors respond to tamoxifen. It identifies tamoxifen in breast cancer. Her2 amplifica-
the node negative ER positive patients who tion is an important predictive biomarker for

| 96 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

Table 1. Definitions.

Term Definition

Biomarker Biological measurement


Predictive biomarker Biomarker measured before treatment to identify patients
likely to benefit from a specific treatment
Prognostic biomarker Biomarker measured before treatment to provide information about
long-term outcome; often incorporates tumor factors and treatment sensitivity
Prognostic or predictive classifier Algorithm that provides a category (e.g., high vs low)
prediction based on one or more biological measurements
Prognostic or predictive index Algorithm that provides a score based on several biological measurements
Gold standard Biological measurement of perfect accuracy
Analytical validation Establishing reproducibility and robustness of test.
If gold standard exists, establishing accuracy of test for measuring biological target
Clinical validation/correlation Establishing accuracy of test for predicting clinical outcome
Medical/Clinical utility Establishing that use of the test enables better treatment decisions to be made
that result in improved patient outcome
Enrichment design RCT comparing new treatment to control restricting entry to patients most likely (or
least unlikely) to benefit from the new treatment compared to the control

effectiveness of trastuzumab. In fact, over-expression of metastatic breast cancer restricted entry to patients
the Her2 protein was critical to the development of whose tumors over-expressed the Her2 protein.3
trastuzumab in patients with metastatic breast cancer. If Restricting entry in that way was important for several
the phase III clinical trial used for establishing the effec- reasons. It meant that the primary analysis was prospec-
tiveness of trastuzumab in metastatic breast cancer had tively defined as evaluation of trastuzumab plus
not used Her2 testing to focus the analysis on patients chemotherapy versus chemotherapy alone in patients
whose tumors over-expressed Her2, the effectiveness of whose tumors over-expressed Her2. Restricting entry
the antibody would almost surely not have been estab- based on Her2 made it clear that the analysis was not
lished.3 This is because only about 25% of patients have the result of post-hoc data dredging. It also meant that
tumors that over-express Her2 and the benefit of the trial was sized to have adequate statistical power for
trastuzumab in those patients would have been diluted the evaluation of trastuzumab versus control in Her2
and obscured by the lack of effectiveness of the anti- positive patients.
body in the remaining 75% of patients in an overall Today, there appears to be greater resistance by regu-
analysis. lators to clinical trials that restrict patient entry based on
Trastuzumab had a very low response rate in a putative predictive biomarker. This is based on con-
metastatic breast cancer. Had the phase II studies not cern that the therapeutic might also benefit test negative
separately evaluated patients whose tumors over-
patients. Sometimes the credentials of the predictive
expressed Her2, the phase III trial would probably never
biomarker are based on the understanding of the biolo-
have been initiated. But comparing response rates to
gy of the therapeutic target and data from pre-clinical
trastuzumab for patients whose tumors are Her2 posi-
tive versus those whose tumors are Her2 negative does models. Understanding mechanisms is often imperfect
not establish the medical utility of trastuzumab. Since or seriously flawed, and regulators would often prefer
partial tumor response is not an indication of patient to see human data that establish the credentials of the
benefit or drug approval in metastatic breast cancer, predictive classifier. If the pre-clinical evidence is not
comparing response rates between Her2 negative and compelling, it is often possible to include test negative
Her2 positive patients cannot establish the medical util- patients at least in phase II studies of the therapeutic. It
ity of the therapeutic. Comparing survivals between may be important to include such patients in phase II
Her2 positive patients who receive trastuzumab and studies to establish a cut-point for the test. Showing
Her2 negative patients who receive trastuzumab would that there is a gradation in response rate with test result
also not establish medical utility of trastuzumab. It in phase II studies may support exclusion of test nega-
would only establish that Her2 testing is prognostic for tive patients from the phase III trial. However, there
survival in patients treated with trastuzumab. That in may not be sufficient phase II data to justify excluding
itself would be of no real medical value. test negative patients from the phase III trial. In some
To establish the medical utility of trastuzumab cases, however, the pre-clinical data will be compelling
required a randomized clinical trial with a control group and it will not be satisfactory to include test negative
of patients not receiving trastuzumab. The randomized patients in phase III trials in order to demonstrate that
trial that led to the FDA approval of trastuzumab in the drug is ineffective for them.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 97 |
14th Congress of the European Hematology Association

Validation of prognostic and predictive tests enables physicians to make better treatment decisions.
The term validation has several meanings for prognos- Many classifiers are developed using convenience sam-
tic and predictive classifiers. ples of available specimens not selected for purposes of
addressing a question of medical decision-making. For
Analytical validation such classifiers there may be an insurmountable gap
First is the analytical validation of an assay for meas- between clinical correlation and medical utility.
uring the classifier. Analytical validation has traditional-
ly meant that the assay accurately measures what it Clinical trials for establishing the medical utility of prog-
claims to measure. This presumes, however, the exis- nostic classifiers
tence of a gold standard way of measuring the true A prognostic classifier can have medical utility if it
value. For gene expression based predictive classifiers, enables physicians to withhold a debilitating or expen-
there is usually no gold standard. Whereas an RT-PCR sive treatment from patients whose prognosis is suffi-
assay might be accepted as a gold standard for measuring ciently good with local therapy alone or a less intensive
gene expression of an RNA sample, this ignores ques- standard approach. To evaluate utility of such a classifi-
tions of the representativeness of the RNA sample for er one would screen eligible patients using the prognos-
the target tissue prior to biopsy. For assays in which tic classifier and register patients whose prognostic cat-
there is no gold standard, analytical validation generally egory was sufficiently favorable, using a prospectively
means reproducibility of the assay and robustness to tis- defined cutoff. The therapy being addressed would be
sue handling and laboratory variations. Careful develop- withheld from the registered patients. If the long-term
ment of an analytically validated assay is important for outcome of the registered patients is sufficiently good,
all later steps of validation. the prognostic classifier is considered to have medical
utility. This is the design being used in the TAILORx
Clinical validation/correlation clinical trial for prospective validation of OncotypeDx.8
Most reports describing the development of a predic- The MINDACT clinical trial randomizes patients pre-
tive classifier based on gene expression address clinical dicted to be good risk by Mammaprint but for whom
correlation, which is sometimes called clinical validation. the standard of care involves chemotherapy between
Clinical validation involves evaluating the degree of withholding or not-withholding chemotherapy.
accuracy of the classifier for predicting some clinical However, the primary analysis and sample size for
outcome. For classifiers developed from microarray MINDACT is not based on the randomized compari-
gene expression data, it is essential to separate the cases son. As in TAILORx, it is based on evaluating the long-
used for developing the classifier from the cases used for term disease-free survival of good prognosis category
evaluating the classifier. With traditional regression patients for whom chemotherapy has been withheld.9
modeling, the number of candidate variables is much The effect of chemotherapy is expected to be very small
less than the number of cases and separation of training and the primary analysis is one of evaluating the test as
and validation cases is often not practiced. Failure to a prognostic classifier in the absence of chemotherapy.
observe this key separation principle with microarray Some people consider the gold standard evaluation of
based classifiers, however, results in enormous bias in medical utility of a new test to involve randomizing
the resulting estimate of accuracy.4 patients to treatment determination based on practice
The most straightforward way of preserving the key standards or based on the test. The test is considered to
separation principle is the split-sample method of parti- have medical utility if treatment outcome is improved
tioning the set of samples into a training set and a test overall for the group randomized to test based treat-
set. Cross-validation is an alternative to the split sample ment assignment. The test would also be considered to
method of estimating prediction accuracy5 while pre- have medical utility if outcome is the same for the two
serving the cardinal separation principle. Molinaro et al.6 randomized groups but the patients randomized to test
describe and evaluate many variants of cross-validation determined treatment have reduced adverse events,
and bootstrap re-sampling for classification problems inconvenience or expense. This kind of prospective clin-
where the number of candidate predictors vastly ical trial design is very inefficient and rarely practical. It
exceeds the number of cases. The cross-validated pre- generally requires an enormous sample size because
diction error is an estimate of the prediction error asso- many or most patients in both randomization groups
ciated with application of the algorithm for model receive the same treatment.10-11 It is generally better to
building to the entire dataset. The model building screen all relevant patients using the test and to evaluate
process must be repeated from scratch for each loop of the implications of acting or not acting on the test in
the cross-validation and so the process must be com- subsets of patients defined by the test and current prac-
pletely algorithmic. Simon et al.4 showed that cross-val- tice standards.
idating the fitting of the prediction model after selection
of differentially expressed genes from the full data set Clinical trials for establishing the medical utility of predic-
results in a highly biased estimate of prediction accura- tive classifiers
cy. This is one of the most common and most serious Simon et al.12-14 discussed prospective clinical trial
errors made in using cross-validation.7 designs for the co-development of new drugs and com-
panion diagnostics. The objective of a phase III pivotal
Medical utility clinical trial is to evaluate whether a new drug, given in
The third level of validation of a predictive classifier is a defined manner, has medical utility for a defined set of
that of determining whether the classifier has medical patients. Pivotal trials test pre-specified hypotheses
utility. A classifier generally has medical utility only if it about treatment effectiveness in specified patient popu-

| 98 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

lation groups. The role of a predictive biomarker classi- line at http://linus.nci.nih.gov/brb/.


fier is to specify the population of patients. The process Jiang et al.20 reported on a “Biomarker Adaptive
of classifier development may be exploratory and sub- Threshold Design” for situations where a predictive
jective, but the use of the classifier in the pivotal trial index is available at the start of the trial, but a cut-point
must not be. for converting the index to a binary classifier is not
With an enrichment design, a diagnostic test is used established. Freidlin and Simon21 proposed an adaptive
to restrict eligibility for a randomized clinical trial of a design for a phase III trial that can be used when no clas-
regimen containing a new drug to a control regimen.15-17 sifier is available at the start of the trial. The design pro-
This approach was used for the development of vides for development of the classifier and evaluation of
trastuzumab in which patients with metastatic breast treatment effects in subsets determined by the classifier
cancer whose tumors expressed HER2 in an immuno- in a single trial. The analysis plan of the adaptive signa-
histochemistry test were eligible for randomization.3 ture design is structured to preserve the principal of sep-
Simon and Maitournam13,18-19 studied the efficiency of arating the data used for developing a classifier from the
this approach relative to the standard approach of ran- data used for evaluating treatment in subsets deter-
domizing all patients without measuring the diagnostic. mined by the classifier, although both processes are part
They found that the efficiency of the enrichment design of the same clinical trial.
depended on the prevalence of test positive patients and Establishing the clinical utility of a classifier for use of
on the effectiveness of the new treatment in test nega- an established treatment can be more difficult. Clinical
tive patients. When fewer than half of the patients are utility depends on a variety of factors including other
test positive and the new treatment is relatively ineffec- treatments available, availability of more easily meas-
tive in test negative patients, the number of randomized ured predictive factors and practice standards. It may be
patients required for an enrichment design is often dra- more difficult to conduct prospective trials that involve
matically smaller than the number of randomized withholding widely used treatments.
patients required for a standard design. Zhao and Simon
have made the methods of sample size planning for the Using archived tumor specimens
design of enrichment trials available on line at Archived specimens are routinely used to establish
http://linus.nci.nih.gov/brb/. analytical validity and clinical validity of tests. The
The enrichment design does not provide data on the more challenging issue is, however, under what condi-
effectiveness of the new treatment compared to control tions are they useful for establishing medical utility of
for test negative patients. Consequently, unless there is prognostic and predictive classifiers. In some cases, it
preliminary data or compelling biological evidence that may be possible to utilize archived tumor samples from
the new drug is not effective in test negative patients, patients treated in a randomized clinical trial to simulate
the enrichment design may not be adequate to support reasonably the analysis that would have been per-
approval of the test as a medical device, and designs that formed in a prospective trial. This is only a viable strat-
include both test positive and test negative patients egy when archived specimens are available for a large
should be considered. proportion of the patients in an appropriately designed
When both test positive and test negative patients are previously conducted randomized trial. The retrospec-
included in the clinical trial, it is essential that an analy- tive strategy is not credible unless the plan for the retro-
sis plan be pre-defined in the protocol for how the pre- spective analysis is completely specified in writing prior
dictive classifier will be used in the analysis. It is not suf- to performing assays on the archived specimens. The
ficient just to stratify the randomization with regard to classifier must be completely determined by data exter-
the classifier without specifying a complete analysis nal to the clinical trial used for retrospective analysis.
plan. The purpose of the pivotal trial is to evaluate the Because retrospective classification of archived speci-
new treatment in the subsets determined by the pre- mens does not accurately reflect the challenges of tissue
specified classifier. The purpose is not to modify or opti- handling and assay performance encountered prospec-
mize the classifier. If the classifier is a composite gene tively in a time frame that enables real-world treatment
expression based classifier, the purpose of the design is selection, it is important to separately establish the ana-
not to re-examine the contributions of each component.
lytical validity of the assay for use with archived tissue.
If one does any of this, then an additional phase III trial
may be needed to evaluate treatment benefit in subsets
determined by the new classifier. In moving from post-
hoc correlative science to reliable predictive medicine, it
References
is important to strictly separate the data used for devel- 1. Paik S, Shak S, Tang G, Kim C, Baker J, Cronin M, et al. A
oping classifiers from the data used for testing treatment multigene assay to predict recurrence of tamoxifen-treated,
effects in subsets determined by those classifiers. node-negative breast cancer. N Engl J Med 2004;351:2817-26.
2. van-de-Vijver MJ, He YD, Veer LJvt, Dai H, Hart AAM,
Reliable conclusions can only be achieved by strictly Voskuil DW, et al. A gene expression signature as a predictor
honoring this principle. The process of classifier devel- of survival in breast cancer. N Engl J Med 2002;347:1999-2009.
opment can be exploratory, but the process of evaluat- 3. Slamon DJ, Leyland-Jones B, Shak S, Fuchs H, Paton V, Baja-
monde A, et al. Use of chemotherapy plus a monoclonal anti-
ing treatments should not be; it should be based on test- body against HER2 for metastatic breast cancer that overex-
ing pre-specified hypotheses in pre-specified patient presses HER2. N Engl J Med 2001;344:783-92.
groups.11 Simon14 describes a variety of analysis strate- 4. Simon R, Radmacher MD, Dobbin K, McShane LM. Pitfalls in
the use of DNA microarray data for diagnostic and prognostic
gies and Zhao and Simon have made the methods of classification. J Nat Cancer Inst 2003;95:14-8.
sample size planning for designs involving treatment of 5. Radmacher MD, McShane LM, Simon R. A paradigm for class
both test positive and test negative patients available on prediction using gene expression profiles. J Comp Biol 2002;9:
505-12.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 99 |
14th Congress of the European Hematology Association

6. Molinaro AM, Simon R, Pfeiffer RM. Prediction error estima- 10:6759-63.


tion: A comparison of resampling methods. Bioinformatics 14. Simon R. Using genomics in clinical trial design. Clin Cancer
2005;21:3301-7. Res 2008;14:5984-93.
7. Dupuy A, Simon R. Critical review of published microarray 15. Temple RJ. Special study designs: Early escape, enrichment,
studies for cancer outcome and guidelines on statistical analy- studies in non-responders. Communications in Statistical
sis and reporting. J Nat Cancer Inst 2007;99:147-57. Theory and Methods 1994;23:499-531.
8. Sparano JA, Paik S. Development of the 21-gene assay and its 16. Temple RJ. Enrichment designs: Efficiency in development of
application in clinical practice and clinical trials. J Clin Oncol cancer treatments. J ClinOncol. 2005;23:4838-9.
2008;26:721-8. 17. Schilsky RL. Target practice: oncology drug development in
9. Bogaerts J, Cardoso F, Buyse M, Braga S, Loi S, Harrison JA, et the era of genomic medicine. Clin Trials 2007;4:163-6.
al. Gene signature evaluation as a prognostic tool: Challenges 18. Simon R, Maitournam A. Evaluating the efficiency of targeted
in the design of the MINDACT trial. Nat Clin Prac Oncol
designs for randomized clinical trials: Supplement and
2006;3:540-51.
10. Sargent DJ, Conley BA, Allegra C, Collette L. Clinical trial Correction. Clin Cancer Res 2006;12:3229.
designs for predictive marker validation in cancer treatment 19. Maitournam A, Simon R. On the efficiency of targeted clinical
trials. J Clin Oncol 2005;23:2020-7. trials. Stat Med 2005;24:329-39.
11. Pusztai L, Hess KR. Clinical trial design for microarray predic- 20. Jiang W, Freidlin B, Simon R. Biomarker adaptive threshold
tive marker discovery and assessment. Ann Oncol 2004;15: design: A procedure for evaluating treatment with possible
1731-7. biomarker-defined subset effect. J Nat Cancer Inst 2007;99:
12. Simon R. A roadmap for developing and validating therapeu- 1036-43.
tically relevant genomic classifiers. J ClinOncol 2005;23:7332- 21. Freidlin B, Simon R. Adaptive signature design: An adaptive
41. clinical trial design for generating and prospectively testing a
13. Simon R, Maitournam A. Evaluating the efficiency of targeted gene expression signature for sensitive patients. Clin Cancer
designs for randomized clinical trials. Clin Cancer Res 2005; Res 2005;11:7872-8.

| 100 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Clinical trial design

Do commonly-used clinical trial designs


reflect clinical reality?

E. Estey A B S T R A C T

This paper contends that commonly used clinical trial designs do not reflect clinical reality as viewed
Fred Hutchinson Cancer Research by patients or physicians. Specifically, randomized phase 3 designs focus on improvements that are
Center and University of Washington
School of Medicine, Seattle, USA more significant statistically than medically and put an emphasis on avoiding a false positive result
that is more appropriate for diseases that are curable, in contrast to acute leukemias. The resultant
large sample sizes needed for each treatment restrict the trial to one or two new treatments, although
historical reality suggests the difficulty in knowing, without clinical data, whether these are the best
Hematology Education: of several new treatments. The p-value-based statistics discourage use of data from previous patients
the education program for the
in the trial to inform treatment of subsequent patients, contravening patients’ assumptions. Standard
annual congress of the European
Hematology Association phase 2 trials focus on a single outcome, ignoring the complexity of medical practice, and ignore prog-
nostic heterogeneity. Finally, although patients are more interested in whether a new treatment is bet-
2009;3:101-105 ter than another is, rather than whether it is active, randomization between different treatments does
not begin until phase 2 trials have been completed. This paper proposes alternatives based on the
Bayesian statistical approach.
The thesis that I will develop here is that commonly used clinical trial designs are unrealistic in the
sense that they do not correspond well to patients’ views of medical practice and greatly over-simplify
such practice. By emphasizing Bayesian rather than p-value-based statistics and focusing on AML, I hope
to familiarize physicians with some of the many new published designs that address these problems.

The standard phase 3 trial no satisfactory treatment for most patients


These trials typically randomize approxi- with AML, the medical risk of a false posi-
mately 400 patients between two therapies.1-3 tive is much less. Indeed, the near universal
This relatively large number is required to choice of p=0.05 and power=80%, regard-
detect relatively small improvements with a less of the disease in question, ignores the
false positive rate less than 5% (p<0.05) and reality that diseases vary considerably in cur-
a false negative rate less than 20% (80% ability.
power). For example, the trials in references Consequently, phase 3 AML trials should
1-3 targeted increases in median EFS or sur- perhaps seek more clinically meaningful
vival of 6 to 12 months, in 2 year EFS or sur- improvements and permit higher p-values.
vival of from 10-20% and in CR rate from 50 Although this formulation would result in
-65%. Consider the relevance of a 6-month loss of power to detect relatively small
improvement in survival to an otherwise advances, I question whether leukemia ther-
healthy 65-year-old man with untreated apeutics advances in such small increments.
AML. Such a patient might expect to live In particular, it would appear that quantum
another 15 years if he did not have AML but therapeutic advances are not infrequent, as
only another one half-year if he is random- with all-trans retinoic acid (ATRA) and
ized to a standard treatment arm. In such a arsenic trioxide (ATO) for APL, 2 chloro-
case, he only retains 0.5/15 (3%) of his nor- deoxyadenosine for hairy cell leukemia,
mally remaining life expectancy. If he is ran- high-dose ara-C and likely gemtuzumab
domized to the investigational arm and it is ozogamycin for CBF AML, and imatinib for
“successful,” he gains another half-year and CML. Even if there were value in retaining
now retains 1/15 (7%) of his life expectancy. sufficient power to detect small advances,
While statistically significant, I doubt many the added value may not justify the neces-
patients would consider this result medically sary sample sizes, which prevent expedi-
significant. Hence, the targeted improve- tious completion of trials and simultaneous
ment does not reflect clinical reality. investigation of a large number of new ther-
The choice of a false positive rate of 0.05 apies.
but a false negative rate of 0.20 implies a
preference for more protection against a P-value-based versus bayesian approaches
false positive than a false negative result. (Table 1)
This is quite sensible when satisfactory Patients naturally prefer “adaptive”
treatment exists for the disease in question, designs, those that permit treatment deci-
and hence, replacement of this standard sions for subsequent patients in a trial to be
with a falsely positive new therapy is partic- based on results in previous patients.
ularly undesirable. However because there is However, p-value-based designs tend to dis-

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 101 |
14th Congress of the European Hematology Association

Table 1. Definitions.

p value: the probability of observing the data or more extreme data under the null
hypothesis; the latter states that there is truly no difference between two treat-
ments. For example, assume the null hypothesis is that, among 10 people, 5 would
prefer red and 5 blue. In fact, 8 preferred red and 2 blue. The p-value is calculat-
ed as the probability that, under the null hypothesis, 8/10 would prefer red + the
probability that 9/10 would prefer red + the probability that all 10 would prefer
red.
Bayes theorem states:

P(B|A) = [P (A|B)] [P(B)]

[P (A|B)] [P(B)] + [P (A|not B)] [P (not B)]

where P is probability, B is a hypothesis, and A are observed data. P(B) is known as


the prior probability of the hypothesis, while P(B|A) is the posterior probability of the Figure 1. Bayesian probability distributions using a trial of
a new therapy in relapsed AML as an example. The values
hypothesis. Thus, the p-value is based on the probability of data given a hypothesis on the horizontal axis are different probabilities of CR. The
while Bayesians compute the posterior probability of a hypothesis given data. values on the vertical axis represent the weight assigned
Physicians often mistakenly believe that a p-value is a Bayesian posterior probability to each CR probability. Prior to treatment, although the
average CR rate is thought to be 20%, some credence is
assigned to each probability of CR (prior probability distri-
bution, dotted line). After observing 5/10 CRS (first poste-
rior probability distribution, dashed line), the average CR
courage frequent examination of incoming data. This rate is close to 50% and no credence is given CR rates less
reflects the inextricable link between p-value and trial than 10% or greater than 90%, reflecting the impact of the
observed data on the prior. Thus, the posteriors become
design, such that the same data can produce different p- successively more informative as the data accumulate,
values depending on the particular design used (Table and shift to reflect the overall average behavior of the
1).4-7 For example, it is well-known that the probability data. After observing 7 CRs in the next 30 patients (total
12 CRs in 40 patients), the average CR rate is approxi-
of finding an association at p<0.05 increases purely by mately 30% and no credence is given a CR rate greater
chance as the number of tests of significance that are than 60% (2nd posterior probability distribution solid line).
performed increases.8-9 Computing the proportion of the area under the curve that
is to the right of a CR rate of 0.4 gives the current proba-
Accordingly, interim analyses of clinical trials are gen- bility that the CR rate is greater than 0.4. This probability
erally performed at p-values much less than 0.05 in can be used to make treatment decisions.
order to preserve an approximately 0.05 level of signifi-
cance at the final analysis. For example, the design pro-
posed by Fleming et al. stops a trial, declaring one arm
superior, only with p-values of 0.005, 0.006, 0.007, and bution of θ, which describes uncertainty about θ after
0.009 at the 1st, 2nd, 3rd, and 4th of 4 interim analyses, observing the data (Figure 1). In contrast to p-value-
respectively. This of course makes it difficult to stop 1:1 based methods, Bayesian inference is not affected by
randomization to an arm, even when the probability the experimental design since data only enter inferences
that that arm is inferior is greater than 90%, leading through the likelihood function. Consequently, when
most patients to prefer randomization to the better arm. making decisions or inferences based on accruing data,
The dependence of p-value on trial design is such Bayes’s theorem may be repeatedly applied, with the
that, in a case when the final planned analysis yields a posterior at each stage becoming the prior for the next
p-value of 0.051, but in which subsequently obtained stage. The probability distributions in this sequence
data strengthen the evidence in favor of a difference, become increasingly informative about θ as the data
these data cannot be used, since they were not obtained accumulate. This process, known as “Bayesian learn-
as part of the planned experiment. ing,” (Figure 1) is especially useful in sequential data
The Bayesian approach provides flexibility, and in monitoring during a clinical trial. The current posterior
particular, encourages interim analyses. The approach probability distribution may be used to modify doses,
begins with parameters, such as the probability of CR unbalance a randomization in favor of a treatment with
or, when comparing two treatments, the probability relatively superior performance, or terminate a trial
that the relative risk of survival is greater than 1.0. early due to either superiority of a treatment or futility.
These parameters (denoted here by θ) are random quan- The Bayesian approaches’ flexibility can be appreciated
tities, with probability distributions describing one’s by contrasting its ability to incorporate data obtained
uncertainty about them. One begins with a prior distri- subsequent to trial completion with the p-value
bution, p(θ), that characterizes the uncertainty about θ approach’s inability to do this, as noted above.
before observing any data. The second Bayesian quanti- A significant issue with the Bayesian approach is set-
ty is the likelihood, L(data | θ), which describes the ting prior probabilities. In Figure 1, we made the prior
probability of observing any specified data given any non-informative reflecting a lack of any information
value of θ; examples of likelihoods are the binomial dis- about response to the new drug. However, it might be
tribution for binary events and the normal(bell-shaped) contended that the prior should be more informative,
distribution for continuous variables. Bayes’ theorem incorporating knowledge of previous trials with other
multiplies the prior by the likelihood of observing the drugs in relapsed AML. The choice of prior obviously
data given the parameter to arrive at a “posterior” distri- influences computation of the posterior – the more

| 102 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

informative the prior, the more data needed to influence corresponding to a power of 80%. In contrast, if the true
the posterior. The designs described below generally CR rates were 30%, 40%, and 30% with TA, IA, and TI,
use non-informative priors. A more detailed presenta- respectively, the probability that the design would cor-
tion would describe how selection of different priors rectly select IA as superior was only 10%. Hence, in this
influences the posterior. case, the design provided much more protection against
a false negative than a false positive. The false positive
Adaptive randomization rate could have been decreased by eliminating the
Bayesian designs for adaptive randomization repeat- requirement that, with high probability, TA or TI be at
edly use interim data to compute the probability that least 10% worse than IA before either of these arms
one arm of a randomized trial is better than the other(s), would close. However, this would have also increased
unbalancing the randomization to favor the likely better the false negative rate contrary to the desire of the clin-
treatment.10-11 If this probability crosses a pre-specified ical investigators to maintain a low false negative rate.
boundary, the inferior arm is shut down before the As outlined above, adaptive randomization fails to
maximal sample size is reached. However, it may re- account for the possible imbalance in prognostic covari-
open if further analyses indicate that results with the ates between patients randomized on each arm. This
open arm(s) are deteriorating such that the probability issue has recently been addressed, together with how
that this arm(s) is superior has decreased. adaptive randomization may be used with censored
A trial adaptively randomizing patients over age 50 data as might arise when survival is the endpoint.13 In
with untreated AML among idarubicin + ara-C (IA, the any event, implementation of adaptive randomization
standard), troxacitabine + ara-C (TA), and troxacitabine requires that patients only infrequently present for ran-
+ idarubcin (TI) illustrates the process.12 The first 15 domization before there has been sufficient opportuni-
patients were randomized fairly among the three arms. ty to observe the outcome in previous patients.
As each patient after the 15th entered the trial, we com-
puted the posterior probability that the CR rate with IA Accounting for prognostic heterogeneity in single arm trials
was greater than or equal to 10% better than that with New drugs are typically tested in single-arm phase 2
TA or TI. If this probability was less than 0.15 accrual to trials before investigation in phase 3. The most com-
IA was suspended. If in contrast the posterior probabil- monly used design for single arm phase 2 trials is the
ity was greater than 0.85 that the CR rate with TA or TI Simon 2-stage (S2S) design.14-15 Rates of no interest
was greater than or equal to 10% worse with IA accru- (known as p0), typically corresponding to the historical
al to either TA or TI was suspended. Depending on rate, and of interest (p1) typically 0.15 to 0.20 higher
results in arms that remained open, a closed arm could than p0 are specified, together with maximum false pos-
re-open. A maximum of 75 patients were to be random- itive and false negative rates (typically 0.10). These
ized. The TI arm closed and remained closed after the parameters determine the number of patients treated in
first five patients failed to respond, while the TA arm the first stage and the minimum number of responses
closed and remained closed after the CR rate was 3/11, needed to proceed to a second stage of specified num-
at which time the CR rate in the IA arm was 10/18. If ber. After the latter is completed, a drug is accepted if
the 34 patients who had been entered on the trial when the number of responses is greater than a specified min-
both TA and TI arms were closed had been randomized imum.
fairly, 11 patients would have received each of TA, TI, The S2S unrealistically assumes that treated patients
and IA. With adaptive randomization, only 16, rather have homogeneous prognoses. Certainly, in AML this is
than 22 patients, received the inferior TA or TI arms, unlikely to be the case.16 Hence reliance on the S2S risks
probably corresponding with how patients visualize declaring drugs inactive when they might have been
clinical practice, found active had a better prognostic group been treated.
The possibility certainly exists that stopping arms so Conducting separate phase 2 trials in distinct prognostic
early might lead to a false negative conclusion. groups is time consuming and does not allow informa-
Beginning adaptive randomization only once 15 to 20 tion gained in one prognostic group to affect the trial in
patients have been randomized equally among the var- a second prognostic group.
ious treatment arms reduces the problem. At any rate, it A method that accounts for treatment-prognostic sub-
is critical to examine how the design performs under group interactions has been proposed, specifically using
various clinical scenarios, that is, what are its operating data from the trial to estimate the degree to which the
characteristics (OC). OC include the probabilities that results in the different subgroups can be combined.17
the design will correctly select a truly superior treatment There are two levels of prior probability distributions
or incorrectly select a truly inferior treatment, as well as (“hierarchical Bayes”). The first is the usual probability
the median number of patients treated on each arm. If of response to a drug in each of, for example, two prog-
clinicians feel the OC are unsatisfactory, the parameters nostic groups. The second quantifies prior belief that
above, such as the criterion probabilities of 0.15 or 0.85, the response in one prognostic group can inform the
or the number of patients to be fairly randomized are probability of response in the other. As usual, these pri-
changed until desirable OC are obtained. Table 2 illus- ors are updated as the trial proceeds.
trates 1000 computer simulations for two scenarios in Consider a hypothetical trial of a new drug in
the IA versus TA versus TI trial. In the first, the true CR relapsed AML. Actual historical data indicated a
rates with TA, IA, and TI are 50%, 40%, and 30%, response rate of 21% in 169 patients. This rate was 11%
respectively; hence, the correct conclusion is that TA is (118 patients) if initial CR duration was less than 1 year
superior. As parameterized above, the probability was but 43% (51 patients) if initial CR duration was greater
80% that the design would reach the correct conclusion, than 1 year. The goal was to increase response rate to

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 103 |
14th Congress of the European Hematology Association

31% (absolute increase of 0.2) in the worse prognostic Table 2. Operating characteristics for IA vs. TA vs. TI trial.
group and to 58% (absolute increase of 15%) in the bet-
ter prognostic group. Since the historical data suggest True CR Rates Correct Probability
that 69% of patients will be in the worse group, the conclusion correct conclu-
overall targeted improvement is [0.20×0.69}+[0.15× sion
0.31]=0.18. Thus, an S2S design would set 21% as p0
and 0.21+0.18=0.39 as p1. Setting the nominal false pos- TA IA TI
itive and false negative rates at 0.10, the S2S would treat 50% 40% 30% TA superior 80%
22 patients in a first stage, and the trial would stop if less
than five responses occurred. If greater than four
responses occurred, an additional 21 patients would be
enrolled and the drug declared a success if responses Tables 3A and B Comparative operating characteristics of
were seen in more than 12/43 patients. Thus, to make STI and Simon 2 stage (S2S) designs.
the proposed design (hereafter, STI because it examines
Probability Mean
subgroup-treatment interactions) comparable to the True (Reject) # Pts
STS, we specify that STI will also take its first look after
22 patients have been evaluated and will also set its Subgroup CR Rate S-TI S2S S-TI S2S
false negative rate at 0.10. Better 0.58 0.10 0.75 21 10
Table 2 compares the operating characteristics of the Worse 0.11 0.90 0.75 19 25
STI and S2S designs. In Table 3A, the new drug achieves
its goal in the better but not the worse group. Because Probability Mean
the S2S does not consider interactions between prog- True (Reject) # Pts
nostic subgroups and treatment, it has the same proba-
bility (0.75) of rejecting the drug in both groups. In con- Subgroup CR Rate S-TI S2S S-TI S2S
trast, the STI is less likely to reject the drug in the better Better 0.43 0.50 0.26 13 11
group and more likely to reject it in the worse group. Worse 0.31 0.10 0.26 27 30
Furthermore, 52% of the patients treated with STI will
be in the better group versus only 29% with S2S. Table
3B illustrates that, in the case where the desired
improvement occurs in the worse but not the better
group, STI is more likely to accept and reject the drug in to stop if it appeared likely that they were only relevant
the appropriate subgroups. Although conducting sepa- for a small subset of the eligible population. Designs
rate Simon 2-stage designed trials in better and worse that monitor multiple outcomes are readily available.22-23
subgroups corrects this problem, S2S’s inability to allow
results in one subgroup to affect the conduct of the trial
in the other subgroup continues to result in a smaller Testing more new therapies and allowing earlier compari-
proportion of patients belonging to the group where son of these
treatment seems more effective relative to historical Patients are more interested in whether one therapy is
better than another than whether either therapy is
Monitoring multiple outcomes “active.” Because comparison is best done through ran-
The great majority of clinical trials specify one “pri- domization, it has been proposed that randomization
mary” outcome, such as toxicity, response rate, or sur- begin earlier than is now the case. In particular, selection
vival. Stopping rules are based only on the primary out- designs have been proposed in which a relatively small
come. This formulation appears unrealistic, ignoring the number of patients are randomized among several new
complexities of medical practice and clinical research. therapies.22,24 The rationale is that, although many new
For example, because phase 1 trials are often quite small therapies are available that may be tested in different
and, unrealistically, fail to account for covariates other schedules and combinations, pre-clinical rationale is an
than dose associated with toxicity, knowledge of toxic- imperfect guide to selecting which new drug to com-
ity is often incomplete after phase 1.18-20 It follows that it pare with a standard. Thus, a compelling pre-clinical
is desirable in phase 2 to formally measure both rationale did not exist for arsenic trioxide in APL, flu-
response and toxicity and allow stopping based on darabine in CLL, and cladribine in hairy cell leukemia,
either outcome. Consider also a trial of a new therapy, while many drugs that failed clinically were accompa-
postulated to be less toxic than standard 3+7, in older nied by seemingly unassailable rationales. A Bayesian
patients with untreated AML. While the reduced toxic- selection design randomizes 45 to 80 patients among
ity might improve survival relative to 3+7, it might also three to four therapies. Each therapy begins with the
reduce CR rate, with long-term survival most likely in same prior probability distribution. As patients are
patients achieving CR.21 However, some decrease in CR treated, the priors are updated with these posteriors
rate would be accepted provide survival increased. used to shut down accrual to an arm if, for example, the
Thus, the trial would formally monitor both survival probability that its true response rate is greater than
and CR, stopping if the decrement in CR rate appeared 20% worse than a competing arm is high. At the end of
too great or the increase in survival insufficient. The the trial, the arm with the highest response rate among
proportion of eligible patients who actually enrol on a those not shut down is selected for further study, per-
trial is often relatively low due to selection bias. The haps in comparison to standard therapy.
consequences of such bias might be reduced were trials Such selection designs are often criticized as “under-

| 104 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

powered phase 3 trials.” Examination of selection 4. Berger J, Berry D. Statistical analysis and the illusion of objec-
tivity. Am Sci. 1988;76:159-165.
designs’ operating characteristics indicate that, in a sce- 5. Berry DA, Stangl DK, editors. Bayesian Biostatistics. New
nario where three drugs have the same true response York: Marcel Dekker; 1996.
rate and the fourth provides an absolute 20% improve- 6. Goodman SA, Toward evidence-based medical statistics,1: the
ment, the probability of correctly selecting the fourth p-value fallacy. Ann Intern Med 1999;130:996-1004.
7. Goodman SA, Toward evidence-based medical statistics, 2:
drug (that is the probability that it will not stop early the Bayes factor. Ann Intern Med 1999;130:1005-13.
plus the probability that it will have the highest 8. Hilsenbeck S, Clark G, McGuire W. Why do so many prognos-
response rate at the end of the trial) is only about 60%. tic factors fail to pan out? Breast Cancer Res Treat 1992;22:
197-206.
This of course contrasts with the aforementioned 80% 9. Altman D, Lausen B, Sauerbrei W, Schumacher M. Dangers of
power typical of randomized trials, involving, for exam- using “optimal” cutpoints in the evaluation of prognostic fac-
ple, a new drug versus a standard. However, the 80% tors. J Nat Cancer Inst 1994;86:829-35.
figure is purely nominal, ignoring the process used to 10. Berry D, Eick S. Adaptive assignment vs. balanced randomiza-
tion in clinical trials: a decision analysis. Stat Med 1995;14:
select the new drug. Assume that four new therapies 231-46.
were available for comparison with a standard, and that 11. Thall P, Wathan J. Practical Bayesian adaptive randomization
because pre-clinical rationale cannot substitute for clini- in clinical trials. Eur J Cancer 2007;43:859-66.
12. Giles F, Kantarjian H, Cortes J, Garcia-Manero G, Verstovsek
cal data in the selection process, each was equally like- S, Faderl S et al. Adaptive randomized study of idarubicin and
ly to be useful clinically. It follows that the probability cytarabine versus troxacitabine and cytarabine versus troxac-
of correctly selecting the best drug was 25%. This 25% itabine and idarubicin in untreated patients 50 years or older
with adverse karyotype acute myeloid leukemia. J Clin Oncol
is ignored in the computation of 80% power; if it were 2003;21:1722-7.
not, the power of the trial would be 25%×80%=20%. 13. Cheung YK, Inoue LY, Wathen JK, Thall PF. Continuous
Thus, the selection design’s 60% probability of correct Bayesian adaptive randomization based on event times with
selection should be viewed, not in relation to 80% covariates. Stat Med 2006;25:55-70.
14. Simon R. Optimal two-stage designs for phase 2 clinical trials.
power, but in relation to the 25% probability of correct Controlled Clin Trials 1989:10:1-10.
selection that would obtain in the absence of the selec- 15. Thall PF, Simon R. Incorporating historical control data in plan-
tion design. Recognizing these issues, the Medical ning phase 2 clinical trials. Stat Med 1990;9:215-28.
16. Estey E, Dohner H. Acute myeloid leukaemia. Lancet 2006;
Research Council-sponsored trials in AML in the United 368:1894-907.
Kingdom are employing selection designs rather than 17. Wathen J, Thall PF, Cook J, Estey E. Accounting for patient
more conventional phase 3 designs. heterogeneity in phase 2 clinical trials. Stat Med 2008;27:
2802-15.
18. Thall P, Lee S. Practical model-based dose-finding in phase I
clinical trials: methods based on toxicity. Int J Gynecol Cancer
References 2003;13:251-61.
19. Rogatko A, Babb JS, Wang H, Slifker MJ, Hudes GR. Patient
1. Baer M, George S, Dodge R, O'Loughlin KL, Minderman H, characteristics compete with dose as predictors of acute treat-
Caligiuri MA et al. Phase 3 study of the multidrug resistance ment toxicity in early phase clinical trials. Clin Cancer Res
modulator PSC-833 in previously untreated patients 60 years 2004;10:4645-51.
of age and older with acute myeloid leukemia: Cancer and 20. Thall PF, Nguyen HQ, Estey E. Patient-specific dose finding
Leukemia Group B Study 9720. Blood 2002;100:1224-32 . based on bivariate outcomes and covariates. Biometrics
2. Rowe J, Neuberg D, Friedenberg W, Bennett JM, Paietta E, 2008;64:1126-36.
Makary AZ et al. A phase 3 study of three induction regimens 21. Estey E, Garcia-Manero G, Giles F, et al. Clinical relevance of
and of priming with GM-CSF in older adults with acute CRp in untreated AML. Blood 2005;106:541.
myeloid leukemia: a trial by the Eastern Cooperative 22. Estey E, Thall PF. New designs for phase 2 trials. Blood
Oncology Group. Blood 2004;103:479-85. 2003;102:442-8.
3. van der Holt B, Lowenberg B, Burnett A, Knauf WU, Shepherd 23. Thall PF, Estey E, Sung H. A new statistical method for dose-
J, Piccaluga PP et al. The value of the MDR1 reversal agent finding based on efficacy and toxicity in early phase clinical
PSC-833 in addition to daunorubicin and cytarabine in the trials. Invest New Drugs 1999:17:155-67.
treatment of elderly patients with previously untreated acute 24. Thall PF, Estey EA. Bayesian strategy for screening cancer
myeloid leukemia, in relation to MDR1 status at diagnosis. treatments prior to phase 2 clinical evaluation. Stat Med 1993;
Blood 2005;106:2646-54. 12:1197-211.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 105 |
Diffuse large B cell lymphoma

Molecular basis of aggressive B-cell


non-Hodgkin lymphomas

E. Hartmann A B S T R A C T
A. Rosenwald
Diffuse large B-cell lymphomas (DLBCL) and Burkitt lymphomas (BL) represent major entities in the
World Health Organization (WHO) classification. Genetic alterations in DLBCL include translocations of
Institute of Pathology, University of
Würzburg, Germany BCL6 (30-40%), BCL2 (20 to 30%) and MYC (10%), and inactivation of the tumor suppressor TP53
(20%); none of which are specific for this malignancy. Aberrant somatic hypermutation may target var-
ious proto-oncogenes, including PIM1, MYC and PAX5. On the gene expression level, the germinal cen-
ter B-cell type (GCB DLBCL) and the activated B-cell type (ABC DLBCL) can be discerned and show dif-
Hematology Education: ferences in underlying genetic alterations, activation of oncogenic pathways and clinical behavior. The
the education program for the
more favorable clinical course for patients with GCB DLBCL treated with CHOP appears to remain sig-
annual congress of the European
Hematology Association nificant in the R-CHOP era. Microarray-based studies have suggested new potential therapeutic targets
in DLBCL, including PKC-β and SYK inhibitors. CARD11 shows oncogenic mutations in a subset of
2009;3:106-111 DLBCL, and could be targeted to block aberrant NF-κB activation, a feature of ABC DLBCL. BL is char-
acterized by the translocation of the MYC oncogene. Gene expression and molecular studies suggest a
gray zone between DLBCL and BL that led to the new provisional category of “B-cell lymphoma, unclas-
sifiable, with features intermediate between DLBCL and BL” in the new WHO classification.

mong aggressive B-cell non-Hodgkin ly expressed in GC B-cells,5 but is down-reg-

A lymphomas (B-NHL), diffuse large B-


cell lymphomas (DLBCL) and Burkitt
lymphomas (BL) represent major entities in
ulated upon terminal B-cell differentiation
into plasma cells. It acts as a transcriptional
repressor and targets genes involved in the
the World Health Organization (WHO) clas- DNA damage response and cell cycle control
sification scheme,1 and this review will focus (including TP53, CDKN1B/p27 and
on the molecular basis of these clinically CCND2/Cyclin D2), thereby allowing DNA
important malignancies. In DLBCL, the remodeling events (e.g., somatic hypermuta-
well-recognized widely varying molecular tion, SHM) to occur in the GC environ-
features do not come as a surprise, given the ment.6-7 BCL6 expression prevents terminal
large number of its clinical variants, morpho- B-cell differentiation by suppressing
logical and immunohistochemical subgroups PRDM1/BLIMP1, a “master regulator” of
and differences in overall survival. In BL, the plasma cell differentiation.6,8 Taken together,
molecular basis appears less heterogeneous deregulated and sustained expression of
compared to DLBCL, but recent investiga- BCL6 may promote lymphomagenesis
tions have highlighted the problematic his- through aberrant proliferation and survival
tomorphological, genetic and molecular gray signals and a B-cell differentiation blockade.
zone between BL and DLBCL that may bear Other frequently detected genetic alter-
important clinical implications. ations in DLBCL include the translocation
t(14;18)(q32;q21), the hallmark translocation
Diffuse large B-cell lymphoma of follicular lymphoma, that also occurs in
The diverse clinical course of DLBCL is 20-30% of DLBCL,1,9-10 and rearrangements
also reflected on the molecular level. In con- of the MYC gene locus in 8q24 detectable in
trast to other B-cell lymphomas, no unifying approximately 10% of cases.1,3,11-12 Moreover,
primary genetic alteration has been identi- the BCL2/18q21 and MYC/8q24 gene loci,
fied so far in DLBCL. Instead, a large spec- as well as the REL gene located in 2p13, are
trum of genetic alterations may contribute to amplified in around 20% of cases.3,13
the development and progression of this Inactivation of the tumor suppressor TP53
lymphoma subtype (Figure 1). located in 17p13.1 has been reported in
The most frequently observed genetic approximately 20% of DLBCL and is associ-
alterations in DLBCL lead to constitutive ated with a poor prognosis.3,14-15 A misdirec-
expression of the BCL6 oncogene, and tion of the somatic hypermutation machin-
include translocations of BCL6 in the chro- ery might be another important mechanism
mosomal band 3q27 (30-40% of DLBCL) as involved in the molecular pathogenesis of
well as mutations in the 5’ non-coding regu- DLBCL. While physiologically the somatic
latory sequences of the BCL6 locus.1-3 BCL6 hypermutation process affects predominant-
expression is necessary for the formation of ly immunoglobulin genes, aberrant somatic
germinal centers (GC)4 and is physiological- hypermutation has been observed in up to

| 106 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

50% of DLBCL targeting various proto-oncogenes


(including PIM1, MYC, RHOH/TTF and PAX5) leading
to potentially oncogenic mutations.16
Microarray-based gene expression profiling studies
also contributed to the elucidation of the molecular het-
erogeneity in DLBCL (Figure 2). Importantly, two bio-
logically and clinically distinct subgroups of DLBCL, the
germinal center B-cell like (GCB) DLBCL and the acti-
vated B-cell like (ABC) DLBCL, have been identified
based on differences in their gene expression pattern.17
In the treatment era of CHOP/CHOP-like chemothera-
py (Cyclophosphamide, Vincristine, Doxorubicin and
Prednisone), the clinical course of patients with GCB
and ABC DLBCL differed markedly, with a more favor-
able prognosis for patients with the GCB subtype.10 Figure 1. Molecular features of diffuse large B cell lym-
Additional gene expression signatures associated with phomas (DLBCL), Burkitt lymphomas (BL) and the gray
zone between the two entities. SHM, somatic hypermuta-
overall survival in the CHOP era include the “prolifera- tion; Ig, immunoglobulin genes.
tion signature,” representing tumor cell proliferation,
the “lymph node signature,” capturing molecular
aspects of the microenvironment and the “MHC class II
signature.” High expression of the proliferation signa- predict the clinical course with high accuracy.19
ture was associated with an unfavorable prognosis, The notion that the tumor microenvironment may
whereas increased expression of the lymph node and contribute importantly to the biological variability of
MHC class II signatures correlated with increased sur- DLBCL was also substantiated by another gene expres-
vival times.10 Loss of the MHC class II protein expres- sion profiling study.20 In this study, Monti and col-
sion on the tumor cells measured immunohistochemi- leagues defined three prominent gene expression clus-
cally predicts for an inferior clinical course, suggesting ters in DLBCL. One cluster reflected the host response
that a diminished tumor immunosurveillance may neg- as a defining feature of a subset of DLBCLs (“host
atively impact on clinical outcome.18 response cluster, HR”), and two additional gene clusters
The question whether the prognostic impact of the were found to be differentially expressed within their
molecular distinction of DLBCL into the GCB and ABC cohort of DLBCL tumors, termed “oxidative phospho-
DLBCL subgroups is still relevant in the R-CHOP treat- rylation (OxPhos)” and “B-cell receptor/proliferation
ment era (CHOP combined with Rituximab) was (BCR)”, the latter showing an increased expression of
addressed in a recent study by Lenz and co-workers.19 genes involved in the BCR signaling cascade. This group
This study demonstrated the predictive value of the also proposed a 13 gene-based survival predictor in
gene expression-based distinction of the GCB/ABC DLBCL, amongst which the genes PDE4B and PKC-β
DLBCL subgroups in a large series of DLBCL patients were found to be overexpressed in lymphoma speci-
treated with R-CHOP.19 Moreover, biological features of mens from patients that followed a fatal clinical
the microenvironment associated with the clinical course.21 The subdivision of DLBCL into the ABC and
course of the disease could be more precisely dissected. GCB subtypes based on gene expression profiling stud-
Specifically, a “stromal-1” signature (including genes ies is further corroborated by additional molecular fea-
that reflect extracellular matrix components and genes tures that are distinctive between the two groups. The
expressed by cells derived from the monocytic lineage), analysis of the immunoglobulin gene mutational status
and a “stromal-2” signature (including genes related to revealed an intraclonal variation in GCB DLBCL indicat-
angiogenesis and tumor blood vessel density), were ing ongoing somatic hypermutation as a marker of ger-
identified that appear to function synergistically and to minal center (GC) derivation, whereas the ABC DLBCL

Figure 2. Gene expression features


of diffuse large B cell lymphoma
(DLBCL). Neoplastic B-cells vary in
the expression of the proliferation
signature, the major histocompatibil-
ity complex (MHC) II signature and
the germinal center B-cell (GCB) sig-
nature.10 Two additional signatures
were described by Monti at al.,20
namely the ‘oxidative phosphoryla-
tion, OxPhos’ and the ‘B cell recep-
tor/proliferation, BCR’ signatures.
Molecular features of the microenvi-
ronment are captured by the stromal
score, consisting of the synergistic
stromal-1 and stromal-2 signatures,19
as well as by the host response sig-
nature.20

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 107 |
14th Congress of the European Hematology Association

subtype usually displayed mutated immunoglobulin may be the use of quantitative RT-PCR-based tech-
genes without evidence for ongoing somatic hypermu- niques. For example, a six-gene model including LMO2,
tation suggesting a post germinal center phenotype.22 BCL6, FN1, CCND2, SCYA3 and BCL2 has been sug-
On the chromosomal level, alterations that preferential- gested for routine application in DLBCL,34 and this test
ly occur in GCB DLBCL include the translocation also predicts survival if applied to formalin fixed and
t(14;18)(q32;q21), gains/amplifications in 2p including paraffin embedded specimens from patients treated
the REL gene locus, as well as gains/amplifications of with R-CHOP.35
12q.10,23 In contrast, ABC DLBCL show more frequently Results from gene expression profiling and other
losses of 6q, trisomy 3 or gains/amplifications of 3q, and molecular studies also suggest a number of new poten-
gains/amplifications of 18q.23 In addition, a recent study tial therapeutic targets in DLBCL. One pathway may be
using a high resolution array comparative genomic the NF-κB signaling cascade that is constitutively acti-
hybridization (CGH) platform identified deletions in vated in the prognostically unfavorable ABC DLBCL
9p21.3, the genomic locus of the INK4a/ARF tumor sup- subgroup, and molecules, such as CARD11,28 or the very
pressor, and a small gain in 19q, including the gene locus recently described casein kinase 1α36 may represent
of the transcription factor SPIB, to occur more frequent- attractive new targets in this pathway. The recent find-
ly in ABC DLBCL.24 The combined analysis of data from ing of an increased tumor blood vessel density in
a genome-wide copy number and gene expression DLBCL with an unfavorable clinical course suggests a
analysis revealed the transcription factor SPIB as a high- potential benefit of the addition of antiangiogenic
ly up-regulated gene in this small amplicon in 19q, and agents to current therapeutic regimens in these
knockdown of SPIB by RNA interference was found to patients,19 and this approach is currently being tested in
be toxic to ABC but not to GCB DLBCL cell lines.24 a large international trial. The finding by Margaret
ABC DLBCL are further characterized by constitutive Shipp’s group that a subset of DLBCL is characterized
activation of the antiapoptotic NF-κB signaling path- by enhanced BCR signaling20 has led to the evaluation of
way,25-27 and a loss-of-function RNA interference screen a SYK-inhibitor (R406) in DLBCL cell lines, with recent-
in ABC and GCB DLBCL cell lines revealed CARD11 as ly reported promising results.37 This group also evaluat-
a key molecule contributing to the enhanced NF-κB-sig- ed the PKC-β inhibitor enzastaurin in a phase II study
naling in ABC DLBCL.27 A subsequent study uncovered for patients with relapsed and refractory DLBCL,38
missense mutations in the coiled-coil domain of demonstrating the immediate generation of hypotheses
CARD11 as a mechanism of its oncogenic activation, for new treatments based on information that original-
and reported that these mutations occur more frequent- ly came from gene expression profiling studies.
ly in ABC DLBCL.28 Therefore, therapeutic targeting of
molecules involved in NF-κB signaling and, in particular, Burkitt lymphoma
inhibition of CARD11 might be promising approaches The diagnosis of BL relies on the combination of dis-
to improve treatment options for the prognostically tinct morphological, immunophenotypic and molecular
unfavorable ABC DLBCL subgroup. features. In Europe, the sporadic variant of BL prevails,
Following the publications from gene expression pro- accounting for 30-50% of childhood lymphomas. EBV
filing studies in DLBCL, various attempts have been infection has been recognized as an important co-factor
made to translate the prognostic information into sim- in the pathogenesis of BL, and is detectable in nearly
plified tests that may be more applicable for routine 100% of patients with the endemic variant and in 20-
clinical practice. Hans and colleagues studied 152 30% and 30-40% of the sporadic and immunodeficien-
DLBCL tumors from patients treated with anthracyclin- cy-associated cases, respectively.1
based chemotherapeutic regimens with a panel of On the molecular level, BL is characterized by chro-
immunohistochemical markers, and reported that stain- mosomal rearrangements involving the MYC gene in
ings for CD10, BCL6 and IRF4/MUM1 may be suffi- 8q24. In around 80% of the cases, the t(8;14)(q24;q32)
cient to subdivide DLBCL into GCB and non-GCB cases can be detected, whereas the remaining 20% show vari-
retaining the prognostic information.29 However, vari- ant translocations involving the light chain loci
ous subsequent studies using the same or slightly mod- (t(2;8)/t(8;22)), all resulting in transcriptional induction
ified immunohistochemical marker panels in other of MYC by juxtaposition to immunoglobulin gene reg-
DLBCL cohorts yielded highly inconsistent results ulatory elements.39
demonstrating that it may be difficult to capture the The t(8;14) translocation breakpoints can be classified
wealth of information provided by complex gene according to their location with respect to both the
expression signatures in just a few immunohistochemi- 8q24/MYC and the 14q32/immunoglobulin heavy
cal stains. On the other hand, different selection criteria chain gene (IgVH) loci. Three different breakpoint local-
and treatment protocols in the studied DLBCL cohorts, izations within the MYC gene locus have been recog-
as well as variations in the staining and scoring tech- nized: the class I breakpoints within the first exon or
niques of different laboratories and observers may have intron of MYC, class II breakpoints that occur immedi-
significantly contributed to the varying results.30-31 The ately 5’ to MYC and the more distant class III break-
incorporation of new, promising markers, such as points. In the IgVH locus, breakpoints located in the
GCET2/HGAL32 and LMO233 into existing immunohis- VDJ-region, as well as in the class switch region have
tochemical algorithms may, however, improve our abil- been detected. Interestingly, the locations of the break-
ity to discern the more favorable GCB DLBCL cases points differ between the epidemiologic variants of BL,
from the remainder. pointing to the formation of the translocation at differ-
An interesting alternative to translate complex gene ent stages of B-cell differentiation. In endemic BL, the
expression signatures into routinely applicable tests breakpoints in the MYC gene most frequently fall into

| 108 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

the class III, and the breakpoints in the immunoglobulin in BL compared to DLBCL included MYC and its target
locus usually occur in the VDJ region, indicating an ori- genes, which was not unexpected since a deregulation
gin of the translocation during erroneous VDJ-recombi- of MYC expression is a key oncogenic event in BL. Two
nation in early B-cell stages in the bone marrow. In com- signatures were expressed at lower levels in BL com-
parison, the sporadic and immunodeficiency-associated pared to DLBCL, one including MHC class I genes and
forms show more frequently class I MYC breakpoints another including NF-κB target genes.47 In another
and IgVH breakpoints located in the class switch region, study, Hummel and colleagues performed48 gene expres-
pointing to a rearrangement during immunoglobulin sion and genetic profiling in 220 tumor specimens from
class switching that occurs after activation of the B cell.39 patients diagnosed with BL, DLBCL and unclassifiable
Apart from MYC gene translocations, only few sec- mature B-NHL by using Affymetrix U133 A gene
ondary chromosomal abnormalities have been detected expression arrays, as well as comparative genomic
in BL using conventional CGH. Among the most com- hybridization. A statistical approach called “core group
monly reported alterations are gains in chromosomes 1 extension” defined an expression signature of 58 genes
(1q), 7 (7q) and 12, as well as losses in chromosome 6 that allowed the subdivision of the lymphomas into
and 17p.40-41 molecular BL (mBL; 22%), non-molecular BL (non-mBL;
MYC is a key transcription factor that controls impor- 58%) and a relatively large group of intermediate cases
tant cellular processes, such as growth control, differen- (20%). In accordance with the results from Dave et al.,47
tiation and apoptosis. In the absence of additional sur- several genes of the NF-κB signaling pathway were
vival signals, over-expression of MYC triggers apopto- found to be expressed at lower levels in the mBL group.
sis, either in a p53 dependent manner via the p14 On the genetic level, the mBL cases were characterized
ARF/MDM2/TP53 pathway or independently of p53, by relatively few chromosomal alterations apart from
for example, mediated through the pro-apoptotic BH3- the MYC translocation (low chromosomal complexity).
only protein BIM.42-43 In BL, several genetic and epigenet- Conversely, the non-mBL (DLBCL) and intermediate
ic alterations that may counteract MYC-induced apop- cases harbored significantly more chromosomal imbal-
totic signals have been reported, such as inactivating ances (high chromosomal complexity), and the correla-
mutations of TP53 in around 30% of the cases, but also tion with clinical parameters revealed that the presence
inactivation of the CDKN2a locus, overexpression of of a MYC translocation in these groups was associated
MDM2 or hypermethylation of p73.44 Furthermore, in a with poor survival.48 Taken together, both studies iden-
substantial proportion of cases missense mutations in tified a characteristic transcriptional profile of BL that
the translocated MYC allele have been detected, that are differed from that of DLBCL. In addition, however, they
particularly located in the conserved MYC box1 ele- described a subset of lymphomas that – according to
ment,45 and, more recently, specific mutations in this current WHO criteria – were classified as DLBCL, but
area of the MYC gene locus have been shown to showed a gene expression profile characteristic of BL
increase tumorigenicity by failure to induce BIM-medi- “discrepant BL”.40,47-48 A subsequent study that combined
ated apoptosis.46 gene expression profiling and conventional CGH detect-
By analyzing morphologic, immunophenotypic and ed a high number of chromosomal alterations among
cytogenetic features, the clinically important distinction “discrepant BL”, which is in contrast to the low chromo-
between DLBCL and classical BL that fulfill all diagnos- somal complexity observed in typical BL. Since the
tic criteria of the WHO classification is usually unprob- chromosomal alterations in the “discrepant BL’ sub-
lematic. However, a subset of aggressive B-NHL can group also differed somewhat from the alterations
show morphologic and immunophenotypic features observed in typical DLBCL, these cases might constitute
intermediate between BL and DLBCL, thereby prevent- a separate molecular subgroup of aggressive B-NHL.
ing a straightforward classification into one diagnostic The group of “discrepant BL” appears to include so-
category. Importantly, the presence of the called dual translocation cases, that is, aggressive B-NHL
t(8;14)(q24;q32) involving MYC cannot be viewed as a carrying both the MYC and a BCL2 (and/or BCL6)
robust cytogenetic marker for this discrimination, since translocation.40,47-48 Uniformly, these lymphomas were
MYC translocations also occur in a subset of bona fide reported to have a devastating clinical course.49-51
DLBCL. In summary, recent studies have provided new
In 2006, two microarray-based studies47-48 analyzed insights into the molecular basis of BL that sharpened
the gene expression profiles of large cohorts of patients the molecular delineation between BL and DLBCL.
with aggressive B-NHL in order to better characterize However, there remains a subset of cases that cannot be
BL on the molecular level and to identify transcriptional clearly assigned to the Burkitt or DLBCL category with
characteristics of BL that might help to distinguish BL currently available morphological, immunophenotypi-
from DLBCL. Dave and coworkers47 studied the gene cal and molecular techniques. To reflect this “molecular
expression profiles of 303 patients diagnosed with gray zone,” the latest WHO classification also suggests
aggressive B-NHL, including BL and DLBCL, and devel- a “diagnostic gray zone” category termed “B-cell lym-
oped a two-step gene expression-based molecular clas- phoma, unclassifiable, with features intermediate
sifier that allowed assignment of all but one case to the between DLBCL and BL.” Importantly, this category
diagnostic categories of BL, ABC DLBCL, GCB DLBCL should not be viewed as a new and separate diagnostic
and primary mediastinal B-cell lymphoma (PMBL). lymphoma entity; instead, it is realized that this group
Cluster analysis of the genes included in the BL classifi- of aggressive lymphomas not meeting the criteria for
er revealed several prominent gene expression signa- classical BL or DLBCL, is molecularly, genetically and
tures providing further insights into molecular features clinically heterogeneous and needs further molecular
of BL. One gene cluster that was more highly expressed and clinical evaluation.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 109 |
14th Congress of the European Hematology Association

EH and AR are supported by the Interdisciplinary Center for RC et al. Diffuse large B-cell lymphoma outcome prediction
by gene-expression profiling and supervised machine learning.
Clinical Research (IZKF), University of Würzburg, Germany. Nat Med 2002;8:68-74.
22. Lossos IS, Alizadeh AA, Eisen MB, Chan WC, Brown PO,
Botstein D et al. Ongoing immunoglobulin somatic mutation
References in germinal center B cell-like but not in activated B cell-like dif-
fuse large cell lymphomas. Proc Natl Acad Sci USA 2000;97:
1. Swerdlow SH, Campo E, Harris NL, Jaffe ES, Pileri SA, Stein 10209-13.
H, et al. World Health Organization Classification of Tumours 23. Bea S, Zettl A, Wright G, Salaverria I, Jehn P, Moreno V et al.
of Haematopoietic and Lymphoid Tissues. In: Bosman FT, Diffuse large B-cell lymphoma subgroups have distinct genet-
Lakhani SR, Ohgaki H, editors. World Health Organization ic profiles that influence tumor biology and improve gene-
Classification of Tumors. Lyon, France: IARC Press; 2008. expression-based survival prediction. Blood 2005;106:3183-90.
2. Lo Coco F, Ye BH, Lista F, Corradini P, Offit K, Knowles DM et 24. Lenz G, Wright GW, Emre NC, Kohlhammer H, Dave SS,
al. Rearrangements of the BCL6 gene in diffuse large cell non- Davis RE et al. Molecular subtypes of diffuse large B-cell lym-
Hodgkin's lymphoma. Blood 1994;83:1757-9. phoma arise by distinct genetic pathways. Proc Natl Acad Sci
3. Lossos IS. Molecular pathogenesis of diffuse large B-cell lym- USA 2008;105:13520-5.
phoma. J Clin Oncol 2005;23:6351-7. 25. Lam LT, Davis RE, Pierce J, Hepperle M, Xu Y, Hottelet M et
4. Dent AL, Shaffer AL, Yu X, Allman D, Staudt LM. Control of al. Small molecule inhibitors of IkappaB kinase are selectively
inflammation, cytokine expression, and germinal center for- toxic for subgroups of diffuse large B-cell lymphoma defined
mation by BCL-6. Science 1997;276:589-92. by gene expression profiling. Clin Cancer Res 2005;11:28-40.
5. Cattoretti G, Chang CC, Cechova K, Zhang J, Ye BH, Falini B 26. Davis RE, Brown KD, Siebenlist U, Staudt LM. Constitutive
et al. BCL-6 protein is expressed in germinal-center B cells. nuclear factor kappaB activity is required for survival of acti-
Blood 1995;86:45-53. vated B cell-like diffuse large B cell lymphoma cells. J Exp Med
6. Shaffer AL, Yu X, He Y, Boldrick J, Chan EP, Staudt LM. BCL- 2001;194:1861-74.
6 represses genes that function in lymphocyte differentiation, 27. Ngo VN, Davis RE, Lamy L, Yu X, Zhao H, Lenz G et al. A
inflammation, and cell cycle control. Immunity 2000;13:199- loss-of-function RNA interference screen for molecular targets
212. in cancer. Nature 2006;441:106-10.
7. Phan RT, Dalla-Favera R. The BCL6 proto-oncogene suppress- 28. Lenz G, Davis RE, Ngo VN, Lam L, George TC, Wright GW et
es p53 expression in germinal-centre B cells. Nature 2004; 432: al. Oncogenic CARD11 mutations in human diffuse large B
635-9. cell lymphoma. Science 2008;319:1676-9.
8. Shaffer AL, Lin KI, Kuo TC, Yu X, Hurt EM, Rosenwald A et 29. Hans CP, Weisenburger DD, Greiner TC, Gascoyne RD,
al. Blimp-1 orchestrates plasma cell differentiation by extin- Delabie J, Ott G et al. Confirmation of the molecular classifi-
guishing the mature B cell gene expression program. cation of diffuse large B-cell lymphoma by immunohisto-
Immunity 2002;17:51-62. chemistry using a tissue microarray. Blood 2004;103:275-82.
9. Weiss LM, Warnke RA, Sklar J, Cleary ML. Molecular analysis 30. de Jong D, Xie W, Rosenwald A, Chhanabhai M, Gaulard P,
of the t(14;18) chromosomal translocation in malignant lym- Klapper W et al. Immunohistochemical prognostic markers in
phomas. N Engl J Med 1987;317:1185-9. diffuse large B-cell lymphoma: validation of tissue microarray
10. Rosenwald A, Wright G, Chan WC, Connors JM, Campo E, as a prerequisite for broad clinical applications. A study from
Fisher RI et al. The use of molecular profiling to predict sur- the Lunenburg Lymphoma Biomarker Consortium (LLBC). J
vival after chemotherapy for diffuse large-B-cell lymphoma. N Clin Pathol 2008.
Engl J Med 2002;346:1937-47. 31. de Jong D, Rosenwald A, Chhanabhai M, Chhanabhai M,
11. Niitsu N, Okamoto M, Miura I, Hirano M. Clinical signifi- Gaulard P, Klapper W et al. Immunohistochemical prognostic
cance of 8q24/c-MYC translocation in diffuse large B-cell lym- markers in diffuse large B-cell lymphoma: validation of tissue
phoma. Cancer Sci 2008. microarray as a prerequisite for broad clinical applications - a
12. Akasaka T, Akasaka H, Ueda C, Yonetani N, Maesako Y, Shi- study from the Lunenburg Lymphoma Biomarker Consort-
mizu A et al. Molecular and clinical features of non-Burkitt's, ium. J Clin Oncol 2007;25:805-12.
diffuse large-cell lymphoma of B-cell type associated with the 32. Natkunam Y, Lossos IS, Taidi B, Zhao S, Lu X, Ding F et al.
c-MYC/immunoglobulin heavy-chain fusion gene. J Clin Expression of the human germinal center-associated lym-
Oncol 2000;18:510-18. phoma (HGAL) protein, a new marker of germinal center B-
13. Rao PH, Houldsworth J, Dyomina K, Parsa NZ, Cigudosa JC, cell derivation. Blood 2005;105:3979-86.
Louie DC et al. Chromosomal and gene amplification in dif- 33. Natkunam Y, Farinha P, Hsi ED, Hans CP, Tibshirani R, Sehn
fuse large B-cell lymphoma. Blood 1998;92:234-40. LH et al. LMO2 protein expression predicts survival in patients
14. Young KH, Weisenburger DD, Dave BJ, Smith L, Sanger W, with diffuse large B-cell lymphoma treated with anthracy-
Iqbal J et al. Mutations in the DNA-binding codons of TP53, cline-based chemotherapy with and without rituximab. J Clin
which are associated with decreased expression of Oncol 2008;26:447-54.
TRAILreceptor-2, predict for poor survival in diffuse large B- 34. Lossos IS, Czerwinski DK, Alizadeh AA, Wechser MA,
cell lymphoma. Blood 2007;110:4396-405. Tibshirani R, Botstein D, Levy R. Prediction of survival in dif-
15. Young KH, Leroy K, Moller MB, Colleoni GW, Sánchez-Beato fuse large-B-cell lymphoma based on the expression of six
M, Kerbauy FR et al. Structural profiles of TP53 gene muta- genes. N Engl J Med 2004;350:1828-37.
tions predict clinical outcome in diffuse large B-cell lym- 35. Malumbres R, Chen J, Tibshirani R, Johnson NA, Sehn LH,
phoma: an international collaborative study. Blood 2008;112: Natkunam Y et al. Paraffin-based 6-gene model predicts out-
3088-98. come in diffuse large B-cell lymphoma patients treated with R-
16. Pasqualucci L, Neumeister P, Goossens T, Nanjangud G, CHOP. Blood 2008;111:5509-14.
Chaganti RS, Küppers R, Dalla-Favera R. Hypermutation of 36. Bidere N, Ngo VN, Lee J, Collins C, Zheng L, Wan F et al.
multiple proto-oncogenes in B-cell diffuse large-cell lym- Casein kinase 1alpha governs antigen-receptor-induced NF-
phomas. Nature 2001;412:341-6. kappaB activation and human lymphoma cell survival. Nature
17. Alizadeh AA, Eisen MB, Davis RE, , Lossos IS, Rosenwald A et 2008.
al. Distinct types of diffuse large B-cell lymphoma identified 37. Chen L, Monti S, Juszczynski P, Daley J, Chen W, Witzig TE
by gene expression profiling. Nature 2000;403:503-11. et al. SYK-dependent tonic B-cell receptor signaling is a ration-
18. Rimsza LM, Roberts RA, Miller TP, Unger JM, LeBlanc M, al treatment target in diffuse large B-cell lymphoma. Blood
Braziel RM et al. Loss of MHC class II gene and protein 2008;111:2230-7.
expression in diffuse large B-cell lymphoma is related to 38. Robertson MJ, Kahl BS, Vose JM, de Vos S, Laughlin M, Flynn
decreased tumor immunosurveillance and poor patient sur- PJ et al. Phase II study of enzastaurin, a protein kinase C beta
vival regardless of other prognostic factors: a follow-up study inhibitor, in patients with relapsed or refractory diffuse large
from the Leukemia and Lymphoma Molecular Profiling B-cell lymphoma. J Clin Oncol 2007;25:1741-6.
Project. Blood 2004;103:4251-8. 39. Boxer LM, Dang CV. Translocations involving c-myc and c-
19. Lenz G, Wright G, Dave SS, Xiao W, Powell J, Zhao H et al. myc function. Oncogene 2001;20:5595-610.
Stromal gene signatures in large-B-cell lymphomas. N Engl J 40. Salaverria I, Zettl A, Bea S, Hartmann EM, Dave SS, Wright
Med 2008;359:2313-23. GW et al. Chromosomal alterations detected by comparative
20. Monti S, Savage KJ, Kutok JL, Feuerhake F, Kurtin P, Mihm M genomic hybridization in subgroups of gene expression-
et al. Molecular profiling of diffuse large B-cell lymphoma defined Burkitt's lymphoma. Haematolo-gica 2008;93:1327-
identifies robust subtypes including one characterized by host 34.
inflammatory response. Blood 2005;105:1851-61. 41. Garcia JL, Hernandez JM, Gutierrez NC, Flores T, González D,
21. Shipp MA, Ross KN, Tamayo P, Weng AP, Kutok JL, Aguiar Calasanz MJ et al. Abnormalities on 1q and 7q are associated

| 110 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

with poor outcome in sporadic Burkitt's lymphoma. A cytoge- 47. Dave SS, Fu K, Wright GW, Lam LT, Kluin P, Boerma EJ et al.
netic and comparative genomic hybridization study. Molecular diagnosis of Burkitt's lymphoma. N Engl J Med
Leukemia. 2003;17:2016-24. 2006;354:2431-42.
42. Dang CV, O'Donnell K A, Juopperi T. The great MYC escape 48. Hummel M, Bentink S, Berger H, Klapper W, Wessendorf S,
in tumorigenesis. Cancer Cell 2005;8:177-8. Barth TF et al. A biologic definition of Burkitt's lymphoma
43. Adhikary S, Eilers M. Transcriptional regulation and transfor- from transcriptional and genomic profiling. N Engl J Med
mation by Myc proteins. Nat Rev Mol Cell Biol 2005;6:635-45. 2006;354:2419-30.
44. Lindstrom MS, Wiman KG. Role of genetic and epigenetic 49. Le Gouill S, Talmant P, Touzeau C, Moreau A, Garand R, Juge-
changes in Burkitt lymphoma. Semin Cancer Biol 2002;12: Morineau N et al. The clinical presentation and prognosis of
381-7.
45. Hoang AT, Lutterbach B, Lewis BC, Yano T, Chou TY, Barrett diffuse large B-cell lymphoma with t(14;18) and 8q24/c-MYC
JF et al. A link between increased transforming activity of lym- rearrangement. Haematologica 2007;92:1335-42.
phoma-derived MYC mutant alleles, their defective regulation 50. Kanungo A, Medeiros LJ, Abruzzo LV, Lin P. Lymphoid neo-
by p107, and altered phosphorylation of the c-Myc transacti- plasms associated with concurrent t(14;18) and 8q24/c-MYC
vation domain. Mol Cell Biol 1995;15:4031-42. translocation generally have a poor prognosis. Mod Pathol
46. Hemann MT, Bric A, Teruya-Feldstein J, Herbst A, Nilsson JA, 2006;19:25-33.
Cordon-Cardo C et al. Evasion of the p53 tumour surveillance 51. Niitsu N, Okamoto M, Miura I, Hirano M. Clinical features
network by tumour-derived MYC mutants. Nature 2005;436: and prognosis of de novo diffuse large B-cell lymphoma with
807-11. t(14;18) and 8q24/c-MYC translocations. Leukemia 2009.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 111 |
Diffuse large B cell lymphoma

Using molecular signatures to identify rational


therapeutic targets in diffuse large B-cell lymphomas

B. Chapuy A B S T R A C T
M. Shipp
Diffuse large B-cell lymphomas (DLBCL) are heterogeneous disorders with significant differences in
clinical outcome, multiple genetic abnormalities and recognized pathological subtypes. Although clin-
Dana Farber Cancer Institute,
Harvard Medical School, ical prognostic classifications can be used to identify patients who are less likely to be cured with
Boston, MA, USA standard therapy, these algorithms do not identify alternative treatment strategies. Transcriptional
profiling and associated genetic and functional analyses have increased our understanding of recog-
nized pathological subtypes of DLBCL and additional groups of tumors with shared features and
reliance upon targetable survival pathways. This review will focus on rational therapeutic targets that
Hematology Education: have been identified by comprehensive molecular analyses of DLBCLs and translated into clinical ini-
the education program for the tiatives.
annual congress of the European
Hematology Association

2009;3:112-117

iffuse large B-cell lymphoma dard chemotherapy were previously evalu-

D (DLBCL) is the most common lym-


phoid malignancy in adults, account-
ing for nearly 30-40% of all non-Hodgkin’s
ated.9 Two of the genes and pathways asso-
ciated with poor response to standard regi-
mens in this initial study9 – the cyclic adeno-
lymphomas (NHLs) in the Western world.1-3 sine monophosphate (c-AMP)—specific
Although a substantial fraction of patients phosphodiesterase, PDE4B and protein
can be cured with the combination of anti- kinase Cβ – prompted further analysis and
CD20 and empiric antracycline-based clinical evaluation.22,23 The association of
chemotherapy, up to 40% of the patients PKCβ expression with poor outcome and
relapse and ultimately succumb to their dis- shortened survival was confirmed in an
ease.4-7 In addition to being clinically hetero- independent series of primary DLBCLs eval-
geneous disorders, DLBCLs exhibit multiple uated by immunohistochemistry.24
genetic lesions and include recognized PKCβ is a serine/threonine kinase that
pathologic subtypes.1,3 phosphorylates the scaffolding protein
Earlier analyses of biologic heterogeneity CARD11 (CARMA1), which brings the
in DLBCL focused on individual genes asso- kinases transforming growth factor-activat-
ciated with treatment outcome, known ed kinase 1 (TAK1) and Iκ-inhibitor kinase
function in other malignancies, and normal (IκK), into close proximity. This allows
lymphocyte development.1,8 With more TAK1 to phosphorylate IκK, the initial step
comprehensive whole-genome techniques, in activating the pro-survival NFκB path-
coordinate transcriptional signatures of spe- way.25-29 PKCβ is also a critical component of
cific groups of large B-cell lymphomas have the VEGF signaling pathway,30-32 which is of
been defined and used to characterize rela- note because VEGF-mediated tumor angio-
tionships between certain tumors and nor- genesis has been linked with poor prognosis
mal cells of origin, define clinically recog- in DLBCL.33,34 The expression of PKCβ in
nized and previously unidentified subtypes, high-risk DLBCLs and the role of the kinase
identify subtype-associated survival path- in critical signaling pathways including
ways and delineate signatures of outcome.9-19 NFκB and VEGF-mediated tumor angiogen-
esis suggested that PKCβ might be a rational
Signatures of outcome - PKCβ in poor progno- therapeutic target in DLBCL and prompted
sis diffuse large B-cell lymphoma analysis of candidate target inhibitors.
Clinical risk stratification models, such as Enzastaurin HCI is an adenosine triphos-
the international prognostic index (IPI) can phate-competitive, selective inhibitor of
be used to identify DLBCL patients who are PKCβ that induced apoptosis and inhibited
less likely to be cured with current the proliferation of DLBCL, glioblastoma,
therapy.20-21 However, these clinical models and colon carcinoma cell lines and
do not provide insights into molecular het- xenografts at low micromolar doses.35-36
erogeneity, pathogenetic mechanisms or After the doses and safety profile were
associated rational therapeutic targets. For determined in a phase I study,37 we conduct-
this reason, the transcriptional profiles of ed a phase II multicenter trial of oral
DLBCLs with different responses to stan- Enzastaurin in patients with relapsed/refrac-

| 112 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

tory DLBCL.23 Twelve of fifty-five patients with als, it will be of interest to determine whether such tar-
relapsed DLBCL (22%; 95% CI, 13-46%) experienced geted agents could replace radiation in MLBCL.
freedom from progression (FFP) for greater than or equal
to two cycles, and eight patients remained free from Developmental signatures – NFkB in ABC-type diffuse
progression for greater than or equal to four cycles large B-cell lymphomas
(15%; 95% CI, 6-27%).23 Of particular interest, four A transcriptional profile-based model has been
patients (7%; 95% CI, 2-18%) continue to experience described that relates subsets of DLBCLs to certain
FFP 20 plus to 50 plus months after study entry.23 These stages of normal B-cell development.12-14 “Germinal cen-
pilot data prompted the development of additional mul- ter (GC)-type” tumors share certain features with nor-
ticenter phase III trials of standard induction therapies mal GC B cells and “activated B-cell (ABC)-type”
(CHOP/rituxan) with or without Enzastaurin as initial DLBCLs have additional traits in common with in vitro-
therapy in patients with high intermediate/high risk activated post-germinal B cells.12-14 A third poorly
DLBCL. defined group of tumors, “others,” includes 17-40% of
DLBCLs in recent series.10,14 Of clinical interest, the
Signatures of large B-cell lymphoma subtypes – NFkB in GCB-type DLBCLs have a more favorable outcome in
MLBCL comparison to the ABC-type tumors.13,14,16 The outcome
In additional studies, investigators used transcrip- differences in DLBCLs defined by cell of origin (COO)
tional profiling to delineate the signature of primary have not been consistently captured with more limited
MLBCL, distinguish this clinically distinct entity from 3 or 4-parameter immunohistochemical signatures,
DLBCL and identify shared features of MLBCL and underscoring the importance of the more comprehen-
classical Hodgkin lymphoma (cHL).11,15 Like cHL, pri- sive transcriptional profiles.41,42 In previous studies,
mary MLBCL exhibited near-uniform nuclear localiza- developmentally defined “ABC-type” DLBCLs were
tion of cREL-containing NFκB heterodimers (Figure 1), found to be particularly dependent upon the NFκB sur-
increased expression of NFκB target genes and vival pathway and NFκB activation signals mediated by
increased NFκB activity in functional assays.11,38,39 the upstream pathway components, CARD11, BCL10
Although many patients with primary MLBCL and MALT1.43-45 These observations raise the interesting
respond well to current combined modality therapy, possibility that ABC-type DLBCLs may be more sensi-
CHOP-Rituxan and mediastinal radiation, the long- tive to targeted inhibitors of the NFkB pathway.
term sequelae of radiation remain a concern.40 As spe- Although the NFκB pathway (Figure 1) is a promising
cific NFκB inhibitors became available for clinical tri- rational treatment target, available candidate inhibitors

Figure 1. NFkB Pathway in DLBCL and MLBCL. (A) Canonical and alternative NFkB pathways. Multiple stimuli activate I-
kappa-B kinase 1 (IkK), a member of the serine/threonine protein kinase family. Activated IkK itself phosphorylates IkB
proteins in the canonical NFkB pathway. The phosphorylation of IkB-proteins results in their dissociation from the NFkB
heterodimers, c-REL/p50 or RELA/p50, within the cytosol and the subsequently polyubiquitination and proteasomal
degradation of IkkB proteins. The liberated NFkB heterodimers translocate to the nucleus and induce target gene expres-
sion. In the alternative NFkB pathway, activated IKK phosphorylates and targets NFkB2 for proteasomal degradation, lib-
erating RELB/p52 for translocation to the nucleus and target gene induction. (B) Nuclear cREL in primary MLBCL.
Primary mediastinal B-cell lymphomas exhibit constitutive activation of the canonical NFkB pathway and localization of
nuclear c-REL by immunohistochemistry.11,38-39

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 113 |
14th Congress of the European Hematology Association

have either not been potent enough for clinical develop-


ment or have multiple other likely mechanisms of
action. For example, the proteasomal inhibitor, borte-
zomib, blocks the proteasomal degradation of the
cytosolic inhibitor of kappaB, IκKB, but also affects
many other cellular proteins and pathways.46 It is also
important to note the complexity of NFκB activation.47
Whereas MLBCLs and Hodgkin lymphomas rely, at
least in part, on c-REL containing heterodimers and the
canonical NFκB activation pathway, ABC-type DLBCLs
do not exhibit a cREL heterodimer-dependent immuno-
histochemical pattern or activation signature.38,44 As tar-
geted inhibitors of the NFκB pathway are developed
and evaluated in specific lymphoid malignancies, the
composition of NFκB heterodimers and the relative con-
tributions of the canonical and alternative NFκB path-
ways (Figure 1) will need to be considered.

Signatures of comprehensive clusters


Figure 2. DLBCL subtypes have different sensitivities to
Given the striking genetic heterogeneity and recog- BCL6 inhibitors. BCR and OxPhos DLBCL cell lines exhibit
nized histologic variants of DLBCL, our group rea- differential sensitivity to the BCL6 peptide inhibitor BPI.51
soned that clinically relevant disease subtypes
remained to be defined. Transcriptional profiling, an
analytical approach that selects the most stable num-
ber of discrete tumor clusters (consensus clustering),48
and additional functional studies were used to identify for therapeutic trials of BCL6 inhibitors when such
three groups of DLBCLs: “Host Response (HR),” agents are available for clinical trials.
“Oxidative Phosphorylation (OxPhos)” and “B-cell
Receptor (BCR) tumors”.10,38,49-51 OxPhos tumors have Tonic B-cell signaling in BCR-type diffuse large B-cell
increased expression of genes regulating oxidative lymphomas
phosphorylation, mitochondrial function, electron As noted, BCR DLBCLs have a transcriptional profile
transport, and proteasomal degradation.10 BCR tumors notable for increased expression of multiple compo-
have increased expression of multiple components of nents of the BCR signaling cascade including the SYK
the B-cell receptor (BCR) signaling cascade and addi- tyrosine kinase.10,50 Recent studies highlight the role of
tional B-cell specific/B-cell essential transcription fac- B-cell receptor (BCR)-mediated survival signals in nor-
tors, such as BCL6.10 In contrast to the other two mal B cells and B-cell lymphomas.50,55-58 Engagement of
groups, Host Response tumors were largely character- the BCR induces phosphorylation of Igα and β
ized by their prominent immune/inflammatory immunoreceptor tyrosine-based activation motif
response, including a prominent T-cell/dendritic cell (ITAMS) and recruitment and activation of SYK and
infiltrate.10,38,49 downstream pathways (Figure 3). Although BCR signal-
Consistent with their morphological features, pro- ing is triggered by antigen binding, emerging data high-
file-defined HR tumors included a subset previously light the role of “tonic” BCR survival signals in the
identified as the DLBCL subtype, T-cell/histiocyte-rich absence of receptor engagement. In murine models,
BCL (T/HRBCL).3,10 HR tumors shared clinical features which illustrated the role and consequences of tonic
with morphologically defined T/HRBCLs, including BCR signaling, loss of the BCR or the excision of the Igα
increased involvement of liver, spleen and bone mar- ITAM triggered apoptosis of peripheral B cells.59-60
row and younger age at presentation.3,10,52 Like The protein tyrosine kinase SYK plays a critical role in
T/HRBCLs, HR tumors also had fewer known genetic tonic BCR signaling, transmitting downstream events
abnormalities. Of interest, primary HR tumors also and amplifying the original signal (Figure 3). The activi-
had a robust NFκB target gene signature, as did ty of SYK is tightly regulated by BCR-associated phos-
microdissected primary tumor cells from phorylation and protein tyrosine phosphatase-mediated
T/HRBCLs.17,38 inhibition.56,57,61,62 In recent studies, we found that over-
expression of the SYK phosphatase, PTPROt, decreased
BCL6 in BCR-type diffuse large B-cell lymphomas cellular proliferation and induced apoptosis of BCR-
BCR DLBCLs have increased expression of BCL6, a dependent lymphomas in the absence of BCR crosslink-
transcription repressor required for normal germinal ing.50,58 These studies indicate that these tumors depend
center B cell development;53 these tumors also have upon SYK-mediated tonic BCR survival signals.
more frequent translocations of the BCL6 locus.10 Of For these reasons, we recently assessed the role of
interest, these tumors also exhibit selective coordinate SYK-dependent tonic BCR survival signals in primary
regulation of BCL6 target genes.51 Consistent with DLBCLs and DLBCL cell lines and evaluated the in vitro
these observations, BCR tumors were significantly efficacy of a competitive SYK inhibitor, R406 (Figure
more sensitive to a highly selective BCL6 peptide 4).50 R406-mediated SYK inhibition has already been
inhibitor, BPI (Figure 2).51,54 These data suggest that evaluated in murine models of allergen-induced airways
patients with BCR DLBCLs may be good candidates hyper-responsiveness63 and rheumatoid arthritis.64 R406

| 114 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

induced apoptosis in the majority of DLBCL cell lines


and primary tumors and specifically inhibited tonic
BCR signaling (Figure 4). The BCR-dependent, R406-
sensitive DLBCL cell lines were independently found to
have the “BCR-type” transcriptional profiles, suggesting
that BCR-dependent DLBCLs might be identified by
their coordinate molecular signature.50
The in vitro studies indicated that SYK-dependent
tonic BCR signaling was a targetable survival pathway
in certain B-cell lymphomas, prompting further clinical
investigation. A phase I/II trial of an oral version of the
SYK inhibitor, R788/fostamatimib disodium (FOS D),
was recently completed.65 After confirming an appropri-
ate dose for phase II testing, the oral SYK inhibitor was
evaluated in relapsed/refractory DLBCL, follicular lym-
phoma and additional B-cell malignancies. In this clini-
cal trial, there was clear evidence of activity in DLBCL.65
Additional clinical trials are planned.

Future directions
Emerging data from the analyses of coordinate molec-
ular signatures of subsets of large B-cell lymphomas
suggest that groups of tumors rely on specific survival
pathways that may represent rational treatment targets.
Figure 3. B-cell-receptor signaling. B-cells and a subset of As clinical trials of targeted agents are implemented, it
DLBCLs rely on B-cell-receptor (BCR) mediated survival will be important to have practical methods of identify-
signals. BCR signaling induces phosphorylation of Igα and
β immunoreceptor tyrosine activation motifs (ITAMs) by ing tumors that rely upon specific functional pathways.
SRC family kinases, such as LYN. ITAM phosphorylation To date, immunohistochemical signatures based on
results in recruitment of the spleen tyrosine kinase SYK, components of coordinate transcriptional profiles have
which initiates downstream events and amplifies the orig-
inal signal. been a major focus.39,42

Figure 4. Inhibition of
SYK-mediated BCR
signaling in DLBCL cell
lines and primary
DLBCLs. SYK inhibi-
tion selectively
induces apoptosis in
BCR-type DLBCL cell
lines.50 (A) Single cell
phosphoflow cytome-
try can be used to
assess SYK-dependent
phosphorylation of the
target protein BLNK in
the absence (green) or
presence (red) of BCR
crosslinking in R406-
sensitive and resistant
DLBCL cell lines.50 (B)
Primary DLBCLs.50 (C)
Only DLBCLs that
exhibit SYK-dependent
BCR-signaling are sen-
sitive to R406 mediat-
ed SYK inhibition.50

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 115 |
14th Congress of the European Hematology Association

Recent studies suggest that it may be possible to nant Hodgkin lymphoma as revealed by global gene expres-
sion analysis. J Exp Med 2008;205:2251-68.
obtain transcriptional profiles from fixed paraffin- 18. Hummel M, Bentink S, Berger H, Klapper W, Wessendorf S,
embedded tumor specimens.66 Another alternative is to Barth TF et al. A Biologic Definition of Burkitt's Lymphoma
develop functionally relevant target signatures suggest- from Transcriptional and Genomic Profiling. N Engl J Med
ed by candidate survival pathways.50 Future directions 2006;354:2419-30.
19. Dave SS, Fu K, Wright GW, Lam LT, Kluin P, Boerma EJ et al.
will likely include the analysis of rational targeted Molecular diagnosis of Burkitt's lymphoma. N Engl J Med
agents in combination with standard empiric 2006;354:2431-42.
chemotherapy, antibody therapy, and in association 20. Shipp M HDCAJftIN-HsLPFP, et al. A Predictive Model for
Aggressive Non-Hodgkin's Lymphoma. N Engl J Med 1993;
with complementary specific target inhibitors. 329:987-94.
21. Sehn LH, Berry B, Chhanabhai M, Fitzgerald C, Gill K,
Hoskins P, et al. The revised International Prognostic Index (R-
IPI) is a better predictor of outcome than the standard IPI for
References patients with diffuse large B-cell lymphoma treated with R-
CHOP. Blood 2007;109:1857-61.
1. Abramson JS, Shipp MA. Advances in the biology and thera- 22. Smith PG, Wang F, Wilkinson KN, Savage KJ, Klein U, Neuberg
py of diffuse large B-cell lymphoma: moving toward a molec- DS et al. The phosphodiesterase PDE4B limits cAMP-associat-
ularly targeted approach. Blood 2005;106:1164-74. ed PI3K/AKT-dependent apoptosis in diffuse large B-cell lym-
2. Armitage JO, Weisenburger DD. New approach to classifying phoma. Blood 2005;105:308-16.
non-Hodgkin's lymphomas: clinical features of the major his- 23. Robertson MJ, Kahl BS, Vose JM, de Vos S, Laughlin M, Flynn
tologic subtypes. Non-Hodgkin's Lymphoma Classification PJ et al. Phase II Study of Enzastaurin, a Protein Kinase C Beta
Project. J Clin Oncol 1998;16:2780-95. Inhibitor, in Patients With Relapsed or Refractory Diffuse
3. Swerdlow SC, Harris E, Jaffe NL, Pileri ES, Stein SA, Thiele H, Large B-Cell Lymphoma. J Clin Oncol 2007;25:1741-6.
et al. WHO Classification of Tumours of Haematopoietic and 24. Hans CP, Weisenburger DD, Greiner TC, Chan WC, Aoun P,
Lymphoid Tissues. In: Bosman FJ, Lakhani ES, Ohgak SR, edi- Cochran GT et al. Expression of PKC-beta or cyclin D2 pre-
tors. World Health Organization Classification of Tumours dicts for inferior survival in diffuse large B-cell lymphoma.
2nd ed. Lyon: IARC; 2008. Mod Pathol 2005;18:1377-84.
4. Pfreundschuh M, Schubert J, Ziepert M, Schmits R, Mohren
M, Lengfelder E et al. Six versus eight cycles of bi-weekly 25. Shinohara H, Yasuda T, Aiba Y, Sanjo H, Hamadate M,
CHOP-14 with or without rituximab in elderly patients with Watarai H et al. PKC beta regulates BCR-mediated IKK activa-
aggressive CD20+ B-cell lymphomas: a randomised controlled tion by facilitating the interaction between TAK1 and
trial (RICOVER-60). Lancet Oncol 2008;9:105-16. CARMA1. J Exp Med 2005;202:1423-31.
5. Coiffier B, Lepage E, Briere J, Herbrecht R, Tilly H, Bou- 26. Sommer K, Guo B, Pomerantz JL, Bandaranayake AD,
abdallah R et al. CHOP chemotherapy plus rituximab com- Moreno-García ME, Ovechkina YL et al. Phosphorylation of
pared with CHOP alone in elderly patients with diffuse large- the CARMA1 linker controls NF-kappaB activation. Immunity
B-cell lymphoma. N Engl J Med 2002;346:235-42. 2005;23:561-74.
6. Habermann TM, Weller EA, Morrison VA, Gascoyne RD, 27. Rueda D, Thome M. Phosphorylation of CARMA1: the
Cassileth PA, Cohn JB et al. Rituximab-CHOP versus CHOP link(er) to NF-kappaB activation. Immunity 2005;23:551-3.
alone or with maintenance rituximab in older patients with 28. Su TT, Guo B, Kawakami Y, Sommer K, Chae K, Humphries
diffuse large B-cell lymphoma. J Clin Oncol 2006;24:3121-7. LA et al. PKC-beta controls I kappa B kinase lipid raft recruit-
7. Pfreundschuh M, Trumper L, Osterborg A, Pettengell R, ment and activation in response to BCR signaling. Nat
Trneny M, Imrie K et al. CHOP-like chemotherapy plus ritux- Immunol 2002;3:780-6.
imab versus CHOP-like chemotherapy alone in young 29. Saijo K, Mecklenbrauker I, Santana A, Leitger M, Schmedt C,
patients with good-prognosis diffuse large-B-cell lymphoma: a Tarakhovsky A. Protein kinase C beta controls nuclear factor
randomised controlled trial by the MabThera International kappaB activation in B cells through selective regulation of the
Trial (MInT) Group Lancet Oncol 2006;7:379-91. IkappaB kinase alpha. J Exp Med 2002;195:1647-52.
8. Gascoyne RD. Emerging prognostic factors in diffuse large B 30. Yoshiji H, Kuriyama S, Ways DK, Yoshii J, Miyamoto Y,
cell lymphoma. Curr Opin Oncol 2004;16:436-41. Kawata M et al. Protein kinase C lies on the signaling pathway
9. Shipp MA, Ross KN, Tamayo P, Weng AP, Kutok JL, Aguiar RC for vascular endothelial growth factor-mediated tumor devel-
et al. Diffuse large B-cell lymphoma outcome prediction by opment and angiogenesis. Cancer Res. 1999;59:4413-8.
gene-expression profiling and supervised machine learning. 31. Teicher BA, Menon K, Alvarez E, Galbreath E, Shih C, Faul
Nat Med 2002;8:68-74. MM. Antiangiogenic and antitumor effects of a protein kinase
10. Monti S, Savage KJ, Kutok JL, Feuerhake F, Kurtin P, Mihm M Cbeta inhibitor in murine lewis lung carcinoma and human
et al. Molecular profiling of diffuse large B-cell lymphoma Calu-6 non-small-cell lung carcinoma xenografts. Cancer
identifies robust subtypes including one characterized by host Chemother Pharmacol. 2001;48:473-80.
inflammatory response. Blood 2005;105:1851-61. 32. Suzuma K, Takahara N, Suzuma I, Isshiki K, Ueki K, Leitges
11. Savage KJ, Monti S, Kutok JL, Cattoretti G, Neuberg D, De M et al. Characterization of protein kinase C beta isoform's
Leval L et al. The molecular signature of mediastinal large B- action on retinoblastoma protein phosphorylation, vascular
cell lymphoma differs from that of other diffuse large B-cell endothelial growth factor-induced endothelial cell prolifera-
lymphomas and shares features with classical Hodgkin lym- tion, and retinal neovascularization. Proc Natl Acad Sci USA
phoma. Blood 2003;102:3871-9. 2002;99:721-6.
12. Alizadeh AA, Eisen MB, Davis RE, Ma C, Lossos IS, 33. Salven P, Teerenhovi L, Joensuu H. A high pretreatment serum
Rosenwald A et al. Distinct types of diffuse large B-cell lym- vascular endothelial growth factor concentration is associated
phoma identified by gene expression profiling. Nature 2000; with poor outcome in non-Hodgkin's lymphoma. Blood 1997;
403:503-11. 90:3167-72.
13. Rosenwald A, Wright G, Chan WC, Connors JM, Campo E, 34. Bertolini F, Paolucci M, Peccatori F, Cinieri S, Agazzi A,
Fisher RI et al. The Use of Molecular Profiling to Predict Ferrucci PF et al. Angiogenic growth factors and endostatin in
Survival after Chemotherapy for Diffuse Large-B-Cell non-Hodgkin's lymphoma. Br J Haematol 1999;106:504-9.
Lymphoma. N Engl J Med 2002;346:1937-47. 35. Sauma S, Yan Z, Ohno S, Friedman E. Protein kinase C beta 1
14. Wright G, Tan B, Rosenwald A, Hurt EH, Wiestner A, Staudt and protein kinase C beta 2 activate p57 mitogen-activated
LM. A gene expression-based method to diagnose clinically protein kinase and block differentiation in colon carcinoma
distinct subgroups of diffuse large B cell lymphoma. Proc Natl cells. Cell Growth Differ 1996;7:587-94.
Acad Sci USA 2003;100:9991-6. 36. Graff JR, McNulty AM, Hanna KR, Konicek BW, Lynch RL,
15. Rosenwald A, Wright G, Leroy K, Yu X, Gaulard P, Gascoyne Bailey SN et al. The protein kinase Cbeta-selective inhibitor,
RD et al. Molecular Diagnosis of Primary Mediastinal B Cell Enzastaurin (LY317615.HCl), suppresses signaling through the
Lymphoma Identifies a Clinically Favorable Subgroup of AKT pathway, induces apoptosis, and suppresses growth of
Diffuse Large B Cell Lymphoma Related to Hodgkin human colon cancer and glioblastoma xenografts. Cancer Res
Lymphoma. J Exp Med 2003;198:851-62. 2005;65:7462-9.
16. Lenz G, Wright G, Dave SS, Xiao W, Powell J, Zhao H et al. 37. Carducci MA, Musib L, Kies MS, Pili R, Truong M, Brahmer JR
Stromal gene signatures in large-B-cell lymphomas. N Engl J et al. Phase I dose escalation and pharmacokinetic study of
Med 2008;359:2313-23. enzastaurin, an oral protein kinase C beta inhibitor, in patients
17. Brune V, Tiacci E, Pfeil I, Döring C, Eckerle S, van Noesel CJ et with advanced cancer. J Clin Oncol 2006;24:4092-9.
al. Origin and pathogenesis of nodular lymphocyte-predomi- 38. Feuerhake F, Kutok JL, Monti S, Chen W, LaCasce AS,

| 116 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

Cattoretti G et al. NF{kappa}B activity, function, and target- fuse large B cell lymphomas. Proc Natl Acad Sci USA 2007;
gene signatures in primary mediastinal large B-cell lymphoma 104:3207-12.
and diffuse large B-cell lymphoma subtypes. Blood 2005; 52. Achten R, Verhoef G, Vanuytsel L, De Wolf-Peeters C. T-
106:1392-9. cell/histiocyte-rich large B-cell lymphoma: a distinct clinico-
39. Rodig SJ, Savage KJ, LaCasce AS, Weng AP, Harris NL, Shipp pathologic entity. J Clin Oncol 2002;20:1269-77.
MA et al. Expression of TRAF1 and nuclear c-Rel distinguish- 53. Klein U, Dalla-Favera R. Germinal centres: role in B-cell phys-
es primary mediastinal large cell lymphoma from other types iology and malignancy. Nat Rev Immunol 2008;8:22-33.
of diffuse large B-cell lymphoma. Am J Surg Pathol 2007; 31: 54. Polo JM, Dell'Oso T, Ranuncolo SM, Cerchietti L, Beck D, Da
106-12. Silva GF et al. Specific peptide interference reveals BCL6 tran-
40. Johnson PW, Davies AJ. Primary mediastinal B-cell lymphoma. scriptional and oncogenic mechanisms in B-cell lymphoma
Hematology Am Soc Hematol Educ Program 2008;2008:349- cells. Nat Med 2004;10:1329-35.
58. 55. Gauld SB, Dal Porto JM, Cambier JC. B cell antigen receptor
41. Nyman H, Adde M, Karjalainen-Lindsberg ML, et al. signaling: roles in cell development and disease. Science 2002;
Prognostic impact of immunohistochemically defined germi- 296:1641-2.
nal center phenotype in diffuse large B-cell lymphoma patients 56. Rolli V, Gallwitz M, Wossning T, Flemming A, Schamel WW,
treated with immunochemotherapy. Blood 2007;109:4930-5. Zürn C, Reth M. Amplification of B cell antigen receptor sig-
42. Hans CP, Weisenburger DD, Greiner TC, Gascoyne RD, naling by a Syk/ITAM positive feedback loop. Mol Cell 2002;
Delabie J, Ott G et al. Confirmation of the molecular classifi- 10:1057-69.
cation of diffuse large B-cell lymphoma by immunohisto- 57. Monroe JG. ITAM-mediated tonic signalling through pre-BCR
chemistry using a tissue microarray. Blood 2004;103:275-82. and BCR complexes. Nat Rev Immunol 2006;6:283-94.
43. Lenz G, Davis RE, Ngo VN, Lam L, George TC, Wright GW et 58. Chen L, Juszczynski P, Takeyama K, Aguiar RCT, Shipp MA.
al. Oncogenic CARD11 mutations in human diffuse large B Protein tyrosine phosphatase receptor-type O truncated
cell lymphoma. Science 2008;319:1676-9. (PTPROt) regulates SYK phosphorylation, proximal B-cell-
44. Davis RE, Brown KD, Siebenlist U, Staudt LM. Constitutive receptor signaling, and cellular proliferation. Blood 2006;108:
nuclear factor kappaB activity is required for survival of acti- 3428-33.
vated B cell-like diffuse large B cell lymphoma cells. J Exp Med 59. Kraus M, Alimzhanov MB, Rajewsky N, Rajewsky K. Survival
2001;194:1861-74. of resting mature B lymphocytes depends on BCR signaling
45. Ngo VN, Davis RE, Lamy L, Yu X, Zhao H, Lenz G et al. A
loss-of-function RNA interference screen for molecular targets via the Igalpha/beta heterodimer. Cell 2004;117:787-800.
in cancer. Nature 2006;441:106-10. 60. Lam KP, Kuhn R, Rajewsky K. In vivo ablation of surface
46. Leonard JP, Furman RR, Coleman M. Proteasome inhibition immunoglobulin on mature B cells by inducible gene targeting
with bortezomib: a new therapeutic strategy for non- results in rapid cell death. Cell 1997;90:1073-83.
Hodgkin's lymphoma. Int J Cancer 2006;119:971-9. 61. Reth M. Hydrogen peroxide as second messenger in lympho-
47. Panwalkar A, Verstovsek S, Giles F. Nuclear factor-kappaB cyte activation. Nat Immunol 2002;3:1129-34.
modulation as a therapeutic approach in hematologic malig- 62. Wienands J, Larbolette O, Reth M. Evidence for a preformed
nancies. Cancer 2004;100:1578-89. transducer complex organized by the B cell antigen receptor.
48. Monti S, Tamayo P, Mesirov J, Golub T. Consensus clustering: Proc Natl Acad Sci USA 1996;93:7865-70.
a resampling-based method for class discovery and visualiza- 63. Matsubara S, Koya T, Takeda K, Joetham A, Miyahara N, Pine
tion of gene expression microarray data. J Mach Learn Res P et al. Syk activation in dendritic cells is essential for airway
2003;52:91-118. hyperresponsiveness and inflammation. Am J Respir Cell Mol
49. Takahashi H, Feuerhake F, Kutok JL, Monti S, Dal Cin P, Biol 2006;34:426-33.
Neuberg D et al. FAS Death Domain Deletions and Cellular 64. Cha HS, Boyle DL, Inoue T, Schoot R, Tak PP, Pine P, Firestein
FADD-like Interleukin 1{beta} Converting Enzyme Inhibitory GS. A novel spleen tyrosine kinase inhibitor blocks c-Jun N-
Protein (Long) Overexpression: Alternative Mechanisms for terminal kinase-mediated gene expression in synoviocytes. J
Deregulating the Extrinsic Apoptotic Pathway in Diffuse Large Pharmacol Exp Ther 2006;317:571-8.
B-Cell Lymphoma Subtypes. Clin Cancer Res 2006;12:3265- 65. Friedberg JW, Sharman J, Schaefer-Cutillo J, et al. Fostamatinib
71. Disodium (FosD), An Oral Inhibitor of Syk, Is Well-Tolerated
50. Chen L, Monti S, Juszczynski P, Daley J, Chen W, Witzig TE and Has Significant Clinical Activity in Diffuse Large B Cell
et al. SYK-dependent tonic B-cell receptor signaling is a ration- Lymphoma (DLBCL) and Chronic Lymphocytic Leukemia
al treatment target in diffuse large B-cell lymphoma. Blood (SLL/CLL). Blood 2008;112.
2008;111:2230-7. 66. Rimsza LM, Leblanc ML, Unger JM, Miller TP, Grogan TM,
51. Polo JM, Juszczynski P, Monti S, Cerchietti L, Ye K, Greally JM Persky DO et al. Gene expression predicts overall survival in
et al. Transcriptional signature with differential expression of paraffin-embedded tissues of diffuse large B-cell lymphoma
BCL6 target genes accurately identifies BCL6-dependent dif- treated with R-CHOP. Blood 2008;112:3425-33.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 117 |
Diffuse large B cell lymphoma

Treatment options for relapsing patients with diffuse


large B cell lymphoma

C. Gisselbrecht A B S T R A C T

Salvage chemotherapy followed by high dose therapy and autologous stem cell transplantation
Institut d’Hématologie, (ASCT) is the standard of treatment for chemosensitive relapses in diffuse large B cell lymphoma.
Hopital Saint Louis, Paris, France
Selection of salvage regimen was never done by a randomized study. To answer this question, the
intergroup CORAL trial included lymphoma patients with DLBCL CD 20+ in first relapse or patients
refractory after first line therapy, who were randomized between R-DHAP and R-ICE. Responding
Hematology Education: patients received BEAM and ASCT. Intent to treat analysis was made on 396 randomized patients (R
the education program for the ICE:202; R DHAP:194). The overall response rate was 63%, with 38% complete remission. There was
annual congress of the European
no difference in response rate between R-ICE 63.5% and R-DHAP 62.8%. ASCT was performed for 206
Hematology Association
patients. There was no significant difference between R-ICE and R-DHAP for 3 year EFS 26% vs. 35%
2009;3:118-122 (p=0.6) and OS 47% versus 51% (p=0.5). EFS was affected by: prior treatment with rituximab; early
relapse less than 12 m; secondary IPI 2-3. On going studies are evaluating new conditioning regimens
with radioimmunotherapy, other combinations of chemotherapy and immunotherapy post transplant
as in CORAL study. Early relapses/ refractory patients to upfront rituximab-based chemotherapy have
a poor response rate and prognosis. New approaches are warranted. Chemotherapy has a poor
response rate and prognosis. A better biological understanding of these patients and new approaches
are required.

ggressive lymphoma patients who (p=0.076). In the overall cohort, patients

A relapse or fail to achieve a CR have a


poor outcome with a life expectancy
of 6 months. Since less than 10% of these
given salvage chemotherapy plus rituximab
achieved significantly longer survival at 2
years than those who received salvage
patients obtain long-term disease-free sur- chemotherapy without rituximab (2-year
vival1 with a salvage regimen alone, it has OS: 58% vs. 24%; p=0.00067).3
long been established that salvage chemo-
therapy should, whenever possible, be fol- Selecting a salvage regimen
lowed by consolidation with HDT and then Various old and new drugs are treatment
ASCT in a chemosensitive patient. options for DLBCL in the salvage setting.
All patients are now treated with front line The effectiveness of these agents has been
rituximab (R) and chemotherapy. The analy- evaluated mainly in non-randomized stud-
sis of registry data from patients treated ies, and the difficulty of obtaining a cure or
with R CHOP confirmed that a major a prolonged disease-free period with con-
improvement in the treatment of diffuse ventional salvage chemotherapy may
large B cell lymphoma is observed in the explain the large number of phase II studies
general population.2 that have been conducted in this setting.
Fewer relapses are seen among patients Consequently, salvage regimens outcomes
with 0-2 IPI factors (10-20%); however, 50% are generally expressed as response rates and
of relapses are still seen for patients with the possibility of collecting stem cells for
more than 2 IPI factors. ASCT. Survival data very often represent a
In the absence of transplantation, the out- mixture of transplanted patients and those
come of relapsing patients is still poor. In the not eligible for transplantation. No clear
long-term analysis of data from the LNH 98- superiority of one regimen over the other
5 trial, comparing CHOP and R CHOP3 has been demonstrated in the absence of
among patients who received first-line treat- randomized study.
ment with CHOP, those who received ritux- Advances in salvage therapy are needed
imab-containing salvage therapy at relapse for two reasons: first, to overcome resistance
achieved significantly longer survival than to chemotherapy, enabling more patients to
those who underwent salvage therapy with achieve a CR, thus allowing suitable candi-
chemotherapy alone (p=0.00043). In con- dates to proceed to transplantation and, sec-
trast, among those patients who received R- ond, to optimize transplantation procedures.
CHOP as first-line therapy, the survival of The addition of rituximab to CHOP
patients who underwent salvage chemother- chemotherapy has significantly improved
apy plus rituximab at relapse did not differ the CR rate, event-free and overall survival
significantly from the survival of those who rates compared to CHOP alone, as first-line
received salvage chemotherapy alone treatment of aggressive NHL, without

| 118 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

increasing toxicity.4 Thus, combining rituximab with nificant effect of rituximab treatment on FFS and overall
ICE (R-ICE), one of the most effective salvage regimens, survival when adjusted for time, since upfront treat-
has been given to patients with relapsed/refractory dis- ment, age, performance status and secondary age
ease. Results from the first 36 assessable patients who adjusted or secondary IPI. However, less than 5% of the
had relapsed (n=23) or refractory (n=13) disease follow- patients had been previously exposed to rituximab.7
ing a single standard anthracycline-based treatment for What is the optimal chemotherapy regimen to com-
diffuse large B-cell lymphoma have been reported.5-6 bine with rituximab as salvage therapy for DLBCL? The
The overall response rate was 78% and the CR rate CORAL intergroup trial compared the association of rit-
53%. The CR rate was significantly higher for patients uximab, Ifosfamide, etoposide, carboplatinum, R-ICE
receiving R-ICE than historical controls, with a similar and rituximab dexamethasone aracytine and cisplat-
second-line IPI given ICE (p=0.006). Patients with inum R-DHAP. DLBCL CD 20+ in first relapse or
relapsed disease had a significantly higher overall patients’ refractory after first line therapy were random-
response rate than those who had primary refractory ized between R-DHAP and R-ICE. Responding patients
disease (96% vs. 46%; p=0.01). Several phase II studies received BEAM and ASCT and were randomized
with various regimens have been reported going in the between observation and maintenance with rituximab
same direction (Table 1). for 1 year. Intent to treat analysis was made on the first
The clear demonstration is provided by a prospective 396 patients randomized in 11 countries (R ICE:202; R
randomized trial exploring the potential benefits associ- DHAP:194).8 The median age was 55 years. In 225
ated with the addition of rituximab to platinum-based patients, a relapse greater than 12 months was observed
salvage regimens. In the study conducted by the after initial complete remission. In 166 cases, patients
HOVON group, 239 patients with relapsed or refracto- did not achieve initial complete remission (refractory) or
ry DLBCL received a salvage regimen consisting of had an early relapse in less than 12 months. Two hun-
DHAP-VIM-DHAP, with or without rituximab, fol- dred and forty-four patients were treated with combi-
lowed by ASCT. Analysis of the 225 patients evaluable nation chemotherapy with prior exposure to rituximab.
showed that after two courses of chemotherapy, PR/CR At the time of inclusion in the study, there were 240
was obtained in 54% of the patients in the DHAP arm patients with Stage III to IV, 198 patients with elevated
and 75% in the R-DHAP arm (p≤01; intention-to-treat LDH. At relapse, 226 patients had a secondary IPI 0 to 1
analysis). Post-transplantation PR/CR was obtained in and 149 patients, sIPI 2 to 3. Patients with prior expo-
50% and 73% of the patients, respectively (p=0.003). A sure to rituximab had more refractory disease and
marked difference in favour of the R-DHAP arm was adverse prognostic factors. The overall response rate
observed at 24 months for failure-free survival, 50% was 63%, with 38% complete remission. There was no
versus 24% (p<0.001) but for OS 52% vs. 59% difference in response rate between R-ICE 63.5% (CI:
(p=0.15).7 Cox regression analysis demonstrated a sig- 56-70%) and R-DHAP 62.8% (CI: 55-69%), and in

Table 1. Rituximab in combination with platinum-containing regimens.

Regimen Disease status n ORR/CR ASCT performed Survival in ASCT patients

R-ICE5 DLBCL 36 78%/53% 70% 67% OS at 2 years


R-ICE26 Aggressive B NHL 8 75%/50% 100% Not given
R-various (mostly ICE)27 DLBCL 59 Not given 100% 81% OS at 2 years
R-ICE28 Aggressive B NHL 11 89%/67% 27% Not given
R-DHAP29 Aggressive B NHL 53 62%/32% 38% Median 20.4 months OS
DHAP/VIM/DHAP ± R7 Aggressive B NHL 225 75%/46% vs. 63% vs. At 2 years FFS 50% vs. 24% in
54%/35% 46% ± R, favour of R (p<0.001)
± R, respectively respectively
R-ESHAP30 DLBCL/MCL/HD 24 (15 = DLBCL) Not given 79% DFS=53%
R-ESHAP31 DLBCL/MCL 6 100%/67% Not done Not applicable
R-ESHAP32 Aggressive B NHL 26 92 %/46% 88% median OS and PFS not yet reached
R-ESHAP33 Aggressive B NHL 18 56%/28% 38% Not stated
R-ICE or R-DHAP34 DLBCL 10 60%/10% Not done Not applicable
R-ASHAP35 Aggressive B NHL 20 75%/45% 25% Not stated
R-DHAOX36 NHL 33 (10 were HIV+) 70%/27% Not stated Not stated
R-ICE or R-DHAP followed by Relapsed DLBCL 396 63%/38% 52% (n= 204) 49% OS at 3 years
HDT (BEAM) and ASCT ± R
maintenance8
*Planned interim analysis for the first 400 patients. ASCT: autologous stem cell transplant; ASHAP: doxorubicin, methylprednisolone, cytarabine, cisplatin; CR; com-
plete response, DFS: disease-free survival; FFS: failure-free survival; DHAOX: dexamethasone, oxaliplatin; DHAP: dexamethasone, cytarabine, cisplatin; DLBCL, diffuse
large B-cell lymphoma; ESHAP: etoposide, methylprednisolone, cytarabine, cisplatin; FU: follow-up; HD, Hodgkin’s disease; ICE: ifosfamide, carboplatin, etoposide;
HDT: high dose therapy; MCL, mantle-cell lymphoma; NHL, non-Hodgkin’s lymphoma; ORR: overall response rate, OS: overall survival; PFS: progression-free survival;
R: rituximab; VIM: etoposide, ifosfamide, methotrexate.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 119 |
14th Congress of the European Hematology Association

mobilization adjusted response rate 52% vs. 54%. Improving conditioning regimen
Factors significantly affecting response (p<0.0001) were Monoclonal antibodies, owing to their targeted effica-
refractory/relapse less than 12 months with a response cy, have now become established components of thera-
rate of 46% vs. 88%, secondary IPI greater than 1: 52% peutic regimens to combat hematological malignancies.
vs. 71% and prior exposure to rituximab: 51% vs. 83%. The development of radiolabelled immunotherapies,
There were fewer serious adverse events in the RICE such as 90Y-ibritumomab tiuxetan has capitalized on the
regimen when compared to R DHAP. targeting ability of antibodies to deliver therapeutic
From this first randomized study on relapses, there doses of radiation to disseminated tumour sites. In a
was obviously no difference in response rate and the number of independent studies in the transplant setting,
90
ability to mobilize stem cell between the two major reg- Y-ibritumomab tiuxetan has been shown to have a
imens used around the world in DLBCL. promising role as a component of conditioning regi-
mens, not only to augment standard chemotherapy reg-
Prognostic factors at relapse imens (immunotherapy-enhanced conditioning) but
Only 206 ASCT were performed in the CORAL also as the sole conditioning agent, extending the option
study, with an unexpected drop out of 50%, mainly due of transplantation to a wider range of patients.12-19 In
to tumor progression. The same drop out was observed ASCT, the use of 90Y-ibritumomab tiuxetan in combina-
in the PARMA study. There was not significant differ- tion with HDT has no effect on the speed of engraft-
ence between R-ICE and R-DHAP for 3-year event free ment and has a toxicity profile similar to conventional
survival (EFS 26% vs. 35% p=0.6) and overall survival conditioning regimens.13,17 Further phase II, and subse-
OS (47% vs. 51%, p=0.5), respectively. Three years EFS quent phase III, studies are now needed to evaluate the
was affected by prior treatment with rituximab, 21% vs. role of 90Y-ibritumomab tiuxetan (Zevalin) in combina-
none 47% (p<0.0001); early relapse less than 12 months tion with HDT (e.g., Z-BEAM vs BEAM) or alone prior
20% versus greater than 12 months 45% (p<0.0001); to ASCT. These studies should investigate different
secondary IPI 2 to 3: 18% vs. 0 to 1: 40% (p=0.0001). In patient subgroups and analyse outcomes according to
the Cox model, all these parameters were significant histology.18-19
(p<0.0001) for EFS, PFS and OS but not the treatment
Rituximab as post-transplantation maintenance/
arm. As early relapse less than 12 months was the main
consolidation
prognostic factor. We looked in the population with
relapse greater than 12 months if prior exposure to rit- In a study in which rituximab was used during stem
cell mobilization (for in vivo purging prior to stem cell
uximab will or will not affect the outcome. There was
harvest) and for consolidation of response at days 1 and
no difference in EFS, OS between these two subgroups
8 after transplantation in patients with relapsed aggres-
with or without rituximab exposure. Obviously, the
sive NHL, the 2-year OS rate was 80%, compared with
response rate is also affected by other parameters such
53% in historical controls who underwent ASCT with-
as age, time to progression, less than 1 year or not and
out rituximab (p=0.002). Similarly, the DFS rate after a
secondary international prognostic index IPI9,10 but now median follow-up of 20 months was 67%, compared
prior exposure to rituximab should be add. with 43% in a historical control group (p=0.004).20
Due to the introduction in the armamentarium of Rituximab can also consolidate the response to trans-
very efficient monoclonal antibody, we are now experi- plantation when given once weekly at weeks 4 to 8
encing patients with early relapse more refractory to after transplantation. Of 26 patients with relapsed NHL
any available treatment. who received post-transplantation rituximab on this
schedule, 7 showed measurable responses in sites of
PET scan prior to transplant
known disease involvement.21 Similarly, in a separate
There is a need to improve salvage regimen with new study, rituximab was given once weekly at weeks 4 to 8
drugs or experimental strategy. PET scans have been after salvage ASCT and repeated if needed over 4 weeks
incorporated in the definition of response. The quality at month 622 to 21 patients with relapsed or refractory
of response before transplant is highly predictive of the large-cell lymphoma. After a median follow-up of 30
outcome. FDG-PET can also be of prognostic value months, the EFS rate was 81% and the OS rate was
when performed prior to autologous stem cell therapy 85%.22 As most patients with DLBCL receive rituximab
(ASCT) in patients with NHL who had previously as a component of their first-line treatment, the role of
achieved remission and were receiving consolidation adjuvant immunotherapy after salvage therapy with or
ASCT. Results indicated that a PET-positive result prior without ASCT should be defined more precisely
to ASCT was associated with a poor duration of through randomized studies. In the CORAL trial,
response and a cumulatively higher risk of relapse, par- patients with relapsed DLBCL were randomised after
ticularly if the post-ASCT was still PET positive.11 ASCT to either rituximab maintenance for 1 year or
Conversely, a PET-negative result prior to ASCT would observation. The study is now completed and 240
predict a better duration of response and suggested that patients have been randomized in the second part. A
further PET-based investigations post-ASCT would not longer follow up is necessary before providing results.8
be mandatory.11 This method is more sensitive to pre-
dict a low rate of relapse in PET-negative patients. Rituximab and other chemotherapy regimens without
Standardization of the procedure is in progress to avoid transplantation
false positive results. Nevertheless, it still provides a bet- However, not all patients are candidates for transplan-
ter evaluation of response and who could be a candidate tation because of age, previous transplantation and/or
for a more experimental approach. comorbidity. Therefore, effective and well-tolerated sal-

| 120 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

vage therapies with minimal toxicities are still needed.


More recently, oxaliplatin and gemcitabine have also References
been shown to be active separately against relapsed or 1. Bosly A, Coiffier B, Gisselbrecht C, Tilly H, Auzanneau G,
refractory NHL. A study on 24 patients assessed the Andrien F, et al. Bone marrow transplantation prolongs sur-
efficacy and safety of oxaliplatin replacement of cis- vival after relapse in aggressive-lymphoma patients treated
with the LNH-84 regimen. J Clin Oncol 1992;10:1615-23.
platin in the DHAP regimen for patients with relapsed 2. Sehn LH, Berry B, Chhanabhai M, Fitzgerald C, Gill K,
or refractory NHL.23 Because the objective response rate Hoskins P, et al. The revised International Prognostic Index (R-
was 77% for patients treated at first relapse with no IPI) is a better predictor of outcome than the standard IPI for
patients with diffuse large B-cell lymphoma treated with R-
major renal and neurotoxicity, this regimen is particular- CHOP. Blood 2007;109:1857-61.
ly suitable in elderly patients.23 In hematologic malig- 3. Feugier P, Van Hoof A, Sebban C, Solal-Celigny P, Bouabdallah
nancies, gemcitabine alone had some degree of efficacy R, Ferme C, et al. Long-Term Results of the R-CHOP Study in
the Treatment of Elderly Patients With Diffuse Large B-Cell
against aggressive NHL. The combination of rituximab- Lymphoma: A Study by the Groupe d'Etude des Lymphomes
platinum and gemcitabine, GEMOX, seems promising de l'Adulte. J Clin Oncol 2005;23:4117-26.
and is currently being evaluated. In a single-arm study 4. Coiffier B, Lepage E, Briere J, Herbrecht R, Tilly H, Bou-
abdallah R, et al. CHOP chemotherapy plus rituximab com-
involving 46 patients with aggressive B-NHL who pared with CHOP alone in elderly patients with diffuse large-
received up to eight cycles of rituximab, gemcitabine B-cell lymphoma. N Engl J Med 2002;346:235-42.
and oxaliplatin in combination (R-GEMOX), 38 patients 5. Kewalramani T, Zelenetz AD, Nimer SD, Portlock C, Straus
(83%) responded after four cycles and 50% were in CR D, Noy A, et al. Rituximab and ICE as second-line therapy
before autologous stem cell transplantation for relapsed or pri-
or CRu by the end of treatment.24 With a median follow mary refractory diffuse large B-cell lymphoma. Blood 2004;
up of 27 months, the 2-year and overall survival rates 103:3684-8.
were 53% and 66%, respectively. The estimated medi- 6. Zelenetz AD, Hamlin P, Kewalramani T, Yahalom J, Nimer S,
Moskowitz CH. Ifosfamide, carboplatin, etoposide (ICE)-
an time to progression (TTP) was 20 months (range 1.6 based second-line chemotherapy for the management of
to >33.5 months) and median duration of response (DR) relapsed and refractory aggressive non-Hodgkin's lymphoma.
was 18 months (range 0.5 to >31.4 months). In addition, Ann Oncol 2003;14:i5-10.
7. Vellenga E, van Putten WL, van 't Veer MB, Zijlstra JM, Fibbe
this platform was chosen to investigate the combination WE, van Oers MH, et al. Rituximab improves the treatment
of chemotherapy and enzastaurin. Enzastaurin is a results of DHAP-VIM-DHAP and ASCT in relapsed/progres-
potent, selective inhibitor of PKC and (PI3K)/protein sive aggressive CD20+ NHL: a prospective randomized
HOVON trial. Blood 2008;111:537-43.
kinase 3 (AKT) pathways known to promote tumor- 8. Gisselbrecht C, Glass B, Mounier N, Gill D, Linch DC, Trneny
induced angiogenesis, as well as tumor-cell survival and M, et al. C. R-ICE versus R-DHAP in relapsed patients with
proliferation.25 A phase II trial with oral enzastaurin plus CD20 diffuse large B-cell lymphoma (DLBCL) followed by
autologous stem cell transplantation: CORAL study. ASCO
GEMOX is ongoing. Oral enzastaurin is also tested as 2009, J Clin Oncol 2009.
maintenance after the end of chemotherapy. The devel- 9. Guglielmi C, Gomez F, Philip T, Hagenbeek A, Martelli M,
opment of new monoclonal antibodies, new generation Sebban C, et al. Time to relapse has prognostic value in
of anti CD 20, anti CD 40, anti CD 22, or more sophis- patients with aggressive lymphoma enrolled onto the Parma
trial. J Clin Oncol 1998;16:3264-9.
ticated immunoconjugate anti CD22 with calichea- 10. Blay J, Gomez F, Sebban C, Bachelot T, Biron P, Guglielmi C,
mycin (CMC 544) are now evaluated in the clinic, as et al. The International Prognostic Index correlates to survival
well as lenalidomide which has show promising activi- in patients with aggressive lymphoma in relapse: analysis of
the PARMA trial. Parma Group. Blood 1998;92:3562-8.
ties in lymphoma. At present, no real “gold standard” 11. Filmont JE, Gisselbrecht C, Cuenca X, Deville L, Ertault M,
treatment exists for relapsed aggressive NHL. Because Brice P, et al. The impact of pre- and post-transplantation
the goal of salvage therapy for aggressive NHL is CR, positron emission tomography using 18-fluorodeoxyglucose
on poor-prognosis lymphoma patients undergoing autologous
and because the major breakthrough in the treatment of stem cell transplantation. Cancer 2007;110:1361-9.
B-cell malignancies was the addition of rituximab to 12. Gianni AM, Berinstein NL, Evans PA, Lopez-Guillermo A,
chemotherapy for front-line treatment, most physicians Solano C. Stem-cell transplantation in non-Hodgkin's lym-
phoma: improving outcome. Anticancer Drugs 2002;13:S35-
are in favor of including rituximab in salvage 42.
chemotherapy regimens. 13. Cilley J, Winter JN. Radioimmunotherapy and autologous
stem cell transplantation for the treatment of B-cell lym-
Conclusion phomas. Haematologica 2006;91:114-20.
14. Winter JN, Inwards D, Spies S, Wiseman G, Patton D, Erwin
Most regimens are giving the same response rate and W, et al. 90Y Ibritumomab Tiuxetan (Zevalin®; 90YZ) Doses
there is no difference between the two most widely Calculated To Deliver up to 1500 cGy to Critical Organs May
used salvage treatments, R ICE and R DHAP. According Be Safely Combined with High-Dose BEAM and
Autotransplant in NHL. Blood 2006;108:3807.
to the main prognostic factors, response rates range 15. Shimoni A, Zwass T, Oksman Y, Hardan I, Shem-Tov N,
from 80-50% and 3 years event free survival from 60- Yerushalmi R, et al. Ibritumomab Tiuxetan (Zevalin) Followed
30%. If the introduction of rituximab was the main by High-Dose Chemotherapy and Autologous Stem-Cell
Transplantation Results in Improved Survival of Patients with
progress in the treatment of B cell lymphoma, patients Chemo-Refractory Non-Hodgkin’s Lymphoma Expected To
failing to respond to rituximab combination chemother- Have Poor Outcome with Standard Pre-Transplant Condition-
apy have a very poor prognosis and can be candidates ing. Blood 2006;108:3052.
16. Shimoni A, Zwas ST, Oksman Y, Hardan I, Shem-Tov N,
for more experimental innovative treatments. Any Yerushalmi R, et al. Yttrium-90-ibritumomab tiuxetan
progress can be easily detected in a poor prognosis (Zevalin) combined with high-dose BEAM chemotherapy and
patient. Enrolment in large randomized study will be autologous stem cell transplantation for chemo-refractory
aggressive non-Hodgkin's lymphoma. Exp Hematol 2007;35:
more problematic in the absence of significant progress 534-40.
in response rate. From the CORAL study, we started, 17. Chiesa C, Botta F, Di Betta E, Coliva A, Maccauro M, Aliberti
with the help of pathobiologists a Bio-CORAL program, G, et al. Dosimetry in myeloablative (90)Y-labeled ibritu-
momab tiuxetan therapy: possibility of increasing adminis-
as we need to better understand poor prognosis patient tered activity on the base of biological effective dose evalua-
and to design new approaches. tion. Preliminary results. Cancer Biother Radiopharm 2007;22:

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 121 |
14th Congress of the European Hematology Association

113-20. phoma. Haematologica 2004;89:763.


18. Gisselbrecht C, Schmitz N, Mounier N, Ma D, Trneny M, 27. Obrlikova PO, Vackova B, Pylik R, Krejcova H, Klener P,
Hagberg H, et al. R-ICE Versus R-DHAP in Relapsed Patients Trneny M. Effects of pretransplantation treatment with
with CD20 Diffuse Large B-Cell Lymphoma (DLBCL) Rituximab on outcomes of autologous stem-cell transplanta-
Followed by Stem Cell Transplantation and Maintenance tion for diffuse large B-cell lymphoma. Haematologica 2006;
Treatment with Rituximab or Not: First Interim Analysis on 91:217.
200 Patients. CORAL Study. Blood 2007;110:159a. 28. Herishanu Y, Terstman S, Perri C, Gipstein L, Ben-Tal O,
19. Krishnan A, Nademanee A, Fung HC, Raubitschek AA, Polliack A, et al. The combination of Rituximab, Etoposide,
Molina A, Yamauchi D, et al. Phase II trial of a transplantation Ifosfamide, and Carboplatin (RICE) is safe and effective sal-
regimen of yttrium-90 ibritumomab tiuxetan and high-dose vage therapy for elderly patients with aggressive non-
chemotherapy in patients with non-Hodgkin's lymphoma. J Hodgkin's lymphoma. Haematologica 2005;90:999.
Clin Oncol 2008;26:90-5. 29. Mey UJ, Olivieri A, Orlopp KS, Rabe C, Strehl JW, Gor-
20. Khouri IF, Saliba RM, Hosing C, Okoroji GJ, Acholonu S, schlueter M, et al. DHAP in combination with rituximab vs
Anderlini P, et al. Concurrent administration of high-dose rit- DHAP alone as salvage treatment for patients with relapsed or
uximab before and after autologous stem-cell transplantation refractory diffuse large B-cell lymphoma: a matched-pair
for relapsed aggressive B-cell non-Hodgkin's lymphomas. J analysis. Leuk Lymphoma. 2006;47:2558-66.
Clin Oncol 2005;23:2240-7. 30. Shrestha S, Johnson C, Jain S, Patel DV, and Mehrotra B.
21. Rapoport AP, Meisenberg B, Sarkodee-Adoo C, Fassas A, ESHAP+/– Rituximab as Salvage Therapy for Relapsed
Frankel SR, Mookerjee B, et al. Autotransplantation for Lymphoma Prior to Stem Cell Transplant: Single Institution
advanced lymphoma and Hodgkin's disease followed by post- Experience. Blood 2004;104: 4601.
transplant rituxan/GM-CSF or radiotherapy and consolidation 31. Venugopal P, Gretory SA, Showel J, Shammo J, Enschede S,
chemotherapy. Bone Marrow Transplant 2002;29:303-12. Larson M, et al. Rituximab (Rituxan) Combined with ESHAP
22. Horwitz SM, Negrin RS, Blume KG, Breslin S, Stuart MJ, Chemotherapy Is Highly Active in Relapsed/ Refractory
Stockerl-Goldstein KE, et al. Rituximab as adjuvant to high- Aggressive Non-Hodgkin’s Lymphoma. Blood 2004;104:4636.
dose therapy and autologous hematopoietic cell transplanta- 32. Hicks L, Buckstein R, Mangel J, Piliotis E, Imrie K, Matthew C,
tion for aggressive non-Hodgkin lymphoma. Blood 2004;103: et al. Rituximab Increases Response to ESHAP in Relapsed,
777-83. Refractory, and Transformed Aggressive B-Cell Lymphoma.
23. Chau I, Webb A, Cunningham D, Hill M, Rao S, Ageli S, et al. Blood 2006;108:3067.
An oxaliplatin-based chemotherapy in patients with relapsed 33. Arnold C, Cuthbert R, Morris TCM, Kettle P, Jones F, Drake
or refractory intermediate and high-grade non-Hodgkin's lym- M. Rituximab-ESHAP as salvage therapy in relapsed aggres-
phoma. Br J Haematol 2001;115:786-92. sive B cell lymphoma. Haematologica 2005;90:1146.
24. El Gnaoui T, Dupuis J, Belhadj K, Jais JP, Rahmouni A, Copie- 34. Bieker R, Kessler T, Berdel WE, Mesters RM. Rituximab in
Bergman C, et al. Rituximab, gemcitabine and oxaliplatin: an combination with platinum-containing chemotherapy in
effective salvage regimen for patients with relapsed or refrac- patients with relapsed or primary refractory diffuse large B-cell
tory B-cell lymphoma not candidates for high-dose therapy. lymphoma. Oncol Rep 2003;10:1915-7.
Ann Oncol 2007;18:1363-8. 35. Aydin S, Duhrsen U, Nuckel H. Rituximab plus ASHAP for the
25. Robertson MJ, Kahl BS, Vose JM, de Vos S, Laughlin M, Flynn treatment of patients with relapsed or refractory aggressive
PJ, et al. Phase II study of enzastaurin, a protein kinase C beta non-Hodgkin's lymphoma: a single-centre study of 20
inhibitor, in patients with relapsed or refractory diffuse large patients. Ann Hematol 2007;86:271-6.
B-cell lymphoma. J Clin Oncol 2007;25:1741-6. 36. Rupolo M, Spina M, Michieli M, Berretta M, Abbruzzese L,
26. Panovska-Stavridis I, Georgievski B, Cevreska L, Stojanovic A, Mazzucato M, et al. R-DHAOX as Salvage Regimen in
Siljanovski N, Ivanovski M, et al. Rituximab plus ICE regimen Patients (pts) with Relapsed/Resistant Non-Hodgkin[spos]s
for relapsed and refractory aggressive non-Hodgkin lym- Lymphoma (NHL). Blood 2004;104:1323.

| 122 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Hematopoiesis

Anatomy of the hematopoietic stem cell niche


during development

E. Dzierzak A B S T R A C T

Stem cell microenvironments and their component elements, particularly the growth factors and
Erasmus Medical Center, Erasmus signalling pathways that play a role in the survival, expansion and/or physiologic function of stem cells
Stem Cell Institute, Dept of Cell
Biology are of fundamental interest in the field of stem cell biology. Within the adult blood system,
Rotterdam, The Netherlands hematopoietic stem cells (HSCs) are supported in the osteoblastic and vascular niches of the bone
marrow. Genetic manipulations within the mouse bone marrow microenvironment have led to the
identification of some of the molecules involved in HSC homing, maintenance and differentiation.
During ontogeny, in early stages of HSC development, several anatomically distinct tissues serve as the
hematopoietic-supportive microenvironment and include the yolk sac, aorta-gonad-mesonephros
Hematology Education:
the education program for the (AGM) region, placenta and fetal liver. While the first HSCs are known to emerge autonomously in the
annual congress of the European mouse AGM region at midgestation, little is known about the potent microenvironment that promotes
Hematology Association the generation of these stem cells. The current knowledge on the AGM microenvironment, its cellular
complexity and its relationship to the generation and maintenance of the first HSCs is presented here.
2009;3:123-127

ematopoietic stem cells (HSC) are the niches.11,12 Some of the key molecular regula-

H foundation of the adult hematopoiet-


ic system and sustain the life long
production of all blood lineages. HSCs are
tors within the bone marrow niches include
N-cadherin, CD150, BMPR1A, and the
SDF1/CXCR4, Notch, Wnt, Hedgehog,
maintained in specialized microenviron- Tie2/Angiopoietin, TGF and FGF signaling
ments throughout development. In the pathways.13,14 Stromal cell lines derived from
adult, the bone marrow harbors the majori- the bone marrow and fetal liver tissues have
ty of HSCs.1 Genetic studies first demon- been instrumental in further characterization
strated the importance of the HSC – niche of these hematopoietic supportive niches.
interaction [reviewed in (2)]: mouse strains Many stromal cell lines have been generated
deficient in the c-kit receptor tyrosine kinase and are characteristically heterogeneous. A
(W mice) and kit-ligand (KL; Steel mice) few stromal cell lines maintain HSCs ex vivo
showed that while bone marrow from W in co-cultures for long periods.6,15,16
mutant mice could not repopulate the Transcriptional profiling of HSC support-
hematopoietic system of wild type irradiat- ive/non-supportive stromal cell lines has
ed recipient mice, bone marrow from Steel revealed a complex genetic program involv-
mutant mice could.3 Yet, the Steel bone mar- ing a wide variety of known molecules and
row was defective for the support of wild molecules whose function in hematopoiesis
type hematopoietic cells. Thus, within the is unknown,17-20 suggesting the concerted and
bone marrow microenvironment, the c-kit- perhaps overlapping actions of stromal mol-
KL signalling axis is important for hemato- ecules/cells in supporting HSCs. These
poietic progenitor/stem cell maintenance results provide a basis for the study of the
and function. Together with the develop- microenvironment throughout develop-
ment of methods to culture this complex ment.
microenvironment ex vivo,4-6 further studies
dissecting the cellular and molecular aspects Microenvironments important for hematopoi-
of the bone marrow microenvironment etic development in the conceptus
were stimulated. Prior to the necessity for a supportive
microenvironment, developmental microen-
Adult hematopoietic supportive vironments promote the generation of
microenvironment hematopoietic progenitor and stem cells.
The bone marrow microenvironment is Several tissues early within the development
very complex, containing osteoblastic niches of the conceptus are able to promote the
and vascular niches within the trabecular generation of these cells from precursors
regions of the long bones.7-9 HSCs are sup- called hemangioblasts (the common meso-
ported and maintained in close association dermal precursor to both endothelial and
to the so-called “stromal cells” of the niches10 hematopoietic lineages) or hemogenic
and live-animal tracking of hematopoietic endothelium (endothelial cells that retain
progenitor/stem cells has shown the homing potency for the hematopoietic lineage)
ability of these cells to the bone marrow [reviewed in (21)]. Recent cell tracing exper-

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 123 |
14th Congress of the European Hematology Association

iments in the mouse have demonstrated that the


founder hematopoietic cells for the adult hematopoietic
system are generated during a specific window of devel-
opmental time – in the mouse, this is approximately
between embryonic day (E) 10 and E12.22 Both extraem-
bryonic tissues (yolk sac and placenta) and the intraem-
bryonic AGM are sites of hematopoietic cell genera-
tion,23-26 but it is as yet uncertain whether one or all of
these sites contribute cells to the adult. Following their
generation, the hematopoietic progenitor and stem cells
are thought to migrate to the fetal liver,27 where they are
maintained/expanded until the bone marrow microen-
vironment is established. As shown in Figure 1, HSCs
are found in all the hematopoietic sites of the concep-
tus.21 While the AGM has been shown to autonomous-
ly generate HSCs,25 it is as yet unclear whether the
extraembryonic sites can also generate HSCs.
Nonetheless, the fact that HSCs are found at in all the
Figure 1. Generation and migration of hematopoietic stem
hematopoietic sites of the conceptus, suggests migra- cells throughout development. Hematopoietic stem cells
tion between anatomically distinct tissues that provide (HSC) begin to be generated in the AGM region at embry-
HSC supportive microenvironments. As the site where onic day (E)10.5 in mouse gestation and between weeks 4
to 6 of gestation in the human embryo. HSCs generation
the first HSCs are generated, investigations have continues only during a short window of time – most like-
focused on the AGM midgestation microenvironment ly until E12/13 in the mouse conceptus. It is yet unknown
to provide molecular insights into the how HSCs are whether the extraembryonic tissues, yolk sac and placenta,
established. can generate HSCs. HSCs migrate and colonize the fetal
liver, and subsequently, the bone marrow at the indicated
times. Red dots indicate HSCs.
The aorta-gonad-mesonephros microenvironment
AGM HSCs are thought to be derived from aortic
endothelial cells, progressing through an endothelial cell
stage that expresses VE-cadherin.22 Cells expressing
hematopoietic markers28-30 are closely adherent to the
vascular endothelium of the aorta at midgestation and
are thought to bud from these endothelial cells into the
lumen as they take on HSC identity (Figure 2). Such
hematopoietic clusters appear at E10 in the mouse aorta
and the first adult-repopulating HSCs are autonomous-
ly generated in the aorta at E10.5 (>34 somite pairs).25,31,32
Although clusters are found both dorsally and ventrally,
HSC activity is localized exclusively to the ventral
aspect of the mouse midgestation aorta, suggesting a
strong positive ventral positional influence in the devel-
opment of AGM hematopoiesis.33 Indeed, hematopoiet-
ic transcription factors, such as Gata2 and Runx1
(required for HSC generation) are expressed in cells of Figure 2. Model for hematopoietic stem cell generation
from hemogenic endothelium. The first generation of
the ventral clusters and endothelium.29,33-35 Several devel- hematopoietic stem cells (HSC) has been localized to the
opmental factors and signaling molecules involved in ventral wall of the dorsal aorta. Based on endothelial and
these pathways are also expressed in the ventral aortic hematopoietic marker expression, HSC generation occurs
during the transition from an endothelial phenotype
region at midgestion, including BMP4.37,38 Within the (hemogenic endothelium) to a hematopoietic phenotype. It
normal physiology of the embryo, the AGM lies is thought that HSC emerge from the endothelial wall (blue
between the ventral tissue that includes the endoderm- cells) and bud into the lumen of the aorta, resulting in
derived gut and the dorsal tissue including the ecto- closely adherent clusters of hematopoietic cells (orange
cells). The inductive factors (blue and green arrows) driving
derm-derived notochord and neural tube. Using AGM the generation of HSCs are thought to come from the mes-
explant cultures, we have preliminary data suggesting enchymal cell layer (beige cells) underlying the aorta and
that dorsal tissues/signals repress AGM HSC activity from ventral endodermal tissues (not shown). These fac-
tors act in concert to drive the expression of transcription
and ventral tissues/signals enhance HSC emergence. factors, such as Gata2 and Runx1 to turn on an HSC genet-
Thus, morphogens and local signals emanating from the ic program. h:hematopoietic cell; e:endothelial cell;
ventral endodermal tissues may be responsible for m:mesenchymal cell.
induction of HSCs.

Mesenchymal stromal cells


What are the cells that make up the hematopoietic are the stromal cells of this microenvironment. It is
generative and/or supportive microenvironment of the known that the AGM region contains different types of
conceptus? In the adult bone marrow, mesenchymal mesenchymal stem/progenitor cells. As reported by
stem/progenitor cells and their differentiating progeny Minasi et al.39 in the quail embryo, a population of aorta-

| 124 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

associated stem cells contributes to cartilage, bone and be inhibited in their activity by gremlin, a BMP antago-
muscle tissues and also to blood,when transplanted into nist. Interestingly, a ventral localization of BMP4 expres-
chick recipients. Based on its differentiation potential, sion in the mesenchyme underlying HSC-containing
this cell was called a “meso-angioblast.” Cells with aortic clusters was found at E11 .37 This finding confirms
more restricted lineage differentiation potential have the ventral expression previously reported in the human
been localized to the mouse AGM region and to the AGM region,38 and proves that BMP4 plays a relatively
other major hematopoietic tissues during mouse late role in the regulation of HSCs as they emerge in the
ontogeny. These are characteristically mesenchymal midgestation AGM. BMP4 does not act as a survival fac-
stem/progenitor cells. Interestingly, mapping and fre- tor but may promote the differentiation and/or expan-
quency analysis of mesenchymal progenitors in the sion of AGM HSCs. Earlier acting endodermal factors
mouse conceptus show that mesenchymal progenitors, are suggested to effect the transition of mesoderm to
with the potential to differentiate into cells of the hematopoietic differentiation. The formation of primi-
osteogenic, adipogenic and/or chondrogenic lineages, tive erythroblasts in the chick yolk sac requires the
reside in most of the sites harboring hematopoietic cells. inductive influence of endodermal cells,54 and avian
Mesenchymal progenitors appear in the AGM region at somatopleural mesoderm is respecified to exhibit
the time of HSC emergence,40 suggesting a functional hematopoietic and vascular potential when briefly
coordination during development of the mesenchymal exposed to endoderm.55 Remarkably in mouse embryo
and hematopoietic lineages. Many stromal cell lines explant grafts, visceral endoderm can induce erythroid
have been established from the AGM region, placenta development in prospective neuroectoderm56 and
and fetal liver, and the yolk sac has yielded a few stro- hedgehog proteins were shown to mimic these effects.57
mal cell lines.16,41-47 We previously isolated and character- Studies in Zebrafish embryos show that hedgehog is
ized stromal cell lines from the AGM subregions (aorta involved at three distinct stages in dorsal aorta and
and urogenital ridges) and also from the embryonic liver hematopoietic development.58 Taken together, these
and ventrally located gut.44,45 Most of these clones were studies suggest that ventral positional information in
derived from transgenic mice expressing the thermola- the endoderm promotes hematopoietic induction. We
bile form of the SV40 Tag gene under the control of the have tested the hypothesis that tissues positioned dor-
β-actin or PGK (phosphoglycerate kinase) promoters. sal or ventral to the AGM affect HSC induction and test-
Phenotypic characterization places the AGM stromal ed whether Hedgehog proteins may play a role in HSC
lines in the vascular smooth muscle cell hierarchy induction. Our preliminary data show that ventral tis-
(VSMC) in between a mesenchymal stem cell and a sues have a positive influence on AGM HSC induction
VSMC.48 In vivo and in vitro assays show that some of the and implicate Hedgehog protein as a potential positive
AGM stromal clones are potent supporters of effector in the induction of AGM HSCs.
hematopoietic progenitors and HSCs as compared to
adult bone marrow and fetal liver cell lines.44 Hence, the Summary
AGM cell lines can provide important signals for the The hematopoietic system requires a supportive
maintenance of the first HSCs. Indeed, some of these microenvironment throughout development. In parallel
lines can support the hematopoietic differentiation of to the earliest stages of the hematopoietic cell generation
embryonic stem cells.49 Similar to the mesenchymal dif- in the conceptus, the related microenvironment consist-
ferentiation potential of bone marrow and fetal liver ing of mesenchymal stem/progenitor cells is undergoing
stromal cell lines, AGM cell lines were able to give rise coordinate growth. Such cells are found in all the major
to osteoblasts, adipocytes and endothelial cells.50 Even sites of hematpoietic cell generation – the yolk sac, AGM
after differentiation, osteoblastic stromal cells continue and placenta. Stromal cell lines isolated from these
to support the growth of hematopoietic cells.37,50 These embryonic tissues have provided insights into the sig-
results suggest that the AGM hematopoietic microenvi- nalling molecules and pathways involved in the many
ronment within the AGM region is, in some ways, very aspects of hematopoietic cell growth and maintenance.
similar to that in the bone marrow. Yet its ability in vivo Since the AGM region generates the first HSCs, study of
to provide HSC inductive signals suggests that it is more this tissue has revealed the importance and positive
complex. While one AGM cell line, AGM-S3, was influence of BMP4 and potentially other ventral and/or
reported previously to promote the generation of HSCs endodermal factors on AGM HSC growth. Further pro-
from early mesodermal precursors,51 this result has not filing of stromal cell lines, as well as novel complex ex
been repeated and no other AGM cell lines have been vivo cultures and mouse models in which such factors are
shown to yield HSC inductive effects. Insights into the dysregulated, will provide information on additional
molecular identity of the AGM microenvironment have developmental factors, their downstream effectors and
been provided by transcriptional profiling of closely- the interactions/convergence of multiple signalling net-
related AGM stromal clones that differentially support works to direct the generation, expansion and mainte-
HSCs.20,37 Expression analyses identified putative HSC nance of the founder HSCs for the adult hematopoietic
regulatory factors - two novel regulators, β-NGF (a neu- system. These data should influence strategies for the
rotrophic factor) and MIP-1γ (a C-C chemokine family generation and/or expansion of HSCs ex vivo for use in
member) and one known regulator, BMP4 (a TGF-β clinical cell replacement therapies.
family member). BMP4 was previously shown to act at
the mesodermal and primitive erythropoietic stages.52,53 This work is supported by the Landsteiner Society for Blood
When added to AGM explant cultures, these three fac- Research (0614), NIH R37 (DK51077), Dutch BSIK Stem
tors enhance the in vivo repopulating ability of AGM Cells in Development and Disease (03038), Dutch BSIK
HSCs.37 E11 AGM HSCs express BMP receptors and can Tissue Engineering.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 125 |
14th Congress of the European Hematology Association

26. Rhodes KE, Gekas C, Wang Y, Lux CT, Francis CS, Chan DN,
References et al. The emergence of hematopoietic stem cells is initiated in
the placental vasculature in the absence of circulation. Cell
1. Wilson A, Trumpp A. Bone-marrow haematopoietic-stem-cell Stem Cell 2008;2:252-63.
niches. Nat Rev Immunol 2006;6:93-106. 27. Johnson GR, Moore MA. Role of stem cell migration in initia-
2. Broudy VC. Stem cell factor and hematopoiesis. Blood 1997; tion of mouse foetal liver haemopoiesis. Nature 1975;258:726-
90:1345-64. 8.
3. Russell ES. Hereditary anemias of the mouse: a review for 28. de Bruijn M, Ma X, Robin C, Ottersbach K, Sanchez MJ,
geneticists. Adv Genet. 1979;20:357-459. Dzierzak E. HSCs localize to the endothelial layer in the
4. Ploemacher RE, van der Sluijs JP, Voerman JS, Brons NH. An in midgestation mouse aorta. Immunity 2002;16:673-83.
vitro limiting-dilution assay of long-term repopulating 29. North T, de Bruijn M, Stacy T, Talebian L, Lind E, Robin C, et
hematopoietic stem cells in the mouse. Blood 1989;74:2755- al. Runx1 Expression Marks Long-Term Repopulating HSCs in
63. the Midgestation Mouse Embryo. Immunity. 2002;16:661-72.
5. Spooncer E, Boettiger D, Dexter TM. Continuous in vitro gen- 30. Taoudi S, Morrison AM, Inoue H, Gribi R, Ure J, Medvinsky
eration of multipotential stem cell clones from src-infected cul- A. Progressive divergence of definitive haematopoietic stem
tures. Nature 1984;310:228-30. cells from the endothelial compartment does not depend on
6. Sutherland HJ, Eaves CJ, Lansdorp PM, Thacker JD, Hogge contact with the foetal liver. Development 2005;132:4179-91.
DE. Differential regulation of primitive human hematopoietic 31. de Bruijn M F, Speck NA, Peeters MC, Dzierzak E. Definitive
cells in long-term cultures maintained on genetically engi- hematopoietic stem cells first develop within the major arteri-
neered murine stromal cells. Blood 1991;78:666-72. al regions of the mouse embryo. Embo J 2000;19:2465-74.
7. Calvi LM, Adams GB, Weibrecht KW, Weber JM, Olson DP, 32. Muller AM, Medvinsky A, Strouboulis J, Grosveld F, Dzierzak
Knight MC, et al. Osteoblastic cells regulate the haematopoi- E. Development of hematopoietic stem cell activity in the
etic stem cell niche. Nature. 2003;425:841-6. mouse embryo. Immunity 1994;1:291-301.
8. Kiel MJ, Yilmaz OH, Iwashita T, Yilmaz OH, Terhorst C, 33. Taoudi S, Medvinsky A. Functional identification of the
Morrison SJ. SLAM family receptors distinguish hematopoiet- hematopoietic stem cell niche in the ventral domain of the
ic stem and progenitor cells and reveal endothelial niches for embryonic dorsal aorta. Proc Natl Acad Sci USA 2007;104:
stem cells. Cell2005;121:1109-21. 9399-403.
9. Zhang J, Niu C, Ye L, Huang H, He X, Tong WG, et al. 34. Cai ZL, de Bruijn M, Ma X, Dortland B, Luteijn T, Downing JR,
Identification of the haematopoietic stem cell niche and con- et al. Haploinsufficiency of AML1/CBFA2 affects the tempor-
trol of the niche size. Nature2003;425:836-41. tal and spatial generation of hematopoietic stem cells in the
10. Taichman RS, Reilly MJ, Emerson SG. The Hematopoietic mouse embryo 2000;13:423-31.
Microenvironment: Osteoblasts and The Hematopoietic 35. Ling KW, Ottersbach K, van Hamburg JP, Oziemlak A, Tsai FY,
Microenvironment. Hematology 2000;4:421-6. Orkin SH, et al. GATA-2 plays two functionally distinct roles
11. Lo Celso C, Fleming HE, Wu JW, Zhao CX, Miake-Lye S, during the ontogeny of hematopoietic stem cells. J Exp Med
Fujisaki J, et al. Live-animal tracking of individual haematopoi- 2004;200:871-82.
etic stem/progenitor cells in their niche. Nature 2009;457:92-6. 35. Minegishi N, Ohta J, Yamagiwa H, Suzuki N, Kawauchi S,
12. Xie Y, Yin T, Wiegraebe W, He XC, Miller D, Stark D, et al. Zhou Y, et al. The mouse GATA-2 gene is expressed in the
Detection of functional haematopoietic stem cell niche using para-aortic splanchnopleura and aorta-gonads and meso-
real-time imaging. Nature 2009;457:97-101. nephros region. Blood 1999;93:4196-207.
13. Arai F, Hirao A, Ohmura M, Sato H, Matsuoka S, Takubo K, et 37. Durand C, Robin C, Bollerot K, Baron MH, Ottersbach K,
al. Tie2/angiopoietin-1 signaling regulates hematopoietic stem Dzierzak E. Embryonic stromal clones reveal developmental
cell quiescence in the bone marrow niche. Cell 2004;118:149- regulators of definitive hematopoietic stem cells. Proc Natl
161. Acad Sci USA 2007;104:20838-43.
14. Li Z, Li L. Understanding hematopoietic stem-cell microenvi- 38. Marshall CJ, Kinnon C, Thrasher AJ. Polarized expression of
ronments. Trends Biochem Sci. 2006;31:589-95. bone morphogenetic protein-4 in the human aorta-gonad-
15. Bennaceur-Griscelli A, Pondarre C, Schiavon V, Vainchenker mesonephros region. Blood 2000;96:1591-3.
W, Coulombel L. Stromal cells retard the differentiation of 39. Minasi MG, Riminucci M, De Angelis L, Borello U, Berarducci
CD34(+)CD38(low/neg) human primitive progenitors B, Innocenzi A, et al. The meso-angioblast: a multipotent, self-
exposed to cytokines independent of their mitotic history. renewing cell that originates from the dorsal aorta and differ-
Blood 2001:97:435-41. entiates into most mesodermal tissues. Development 2002;
16. Moore KA, Ema H, Lemischka IR. In vitro maintenance of 129:2773-83.
highly purified, transplantable hematopoietic stem cells. Blood 40. Mendes S, Robin C. Dzierzak E. Mesenchymal progenitor
1997;89:4337-47. cells localize within hematopoietic sites throughout ontogeny.
17. Graf L, Iwata M, Torok-Storb B. Gene expression profiling of Development. 2005.
the functionally distinct human bone marrow stromal cell 41. Fennie C, Cheng J, Dowbenko D, Young P, Lasky LA. CD34+
lines HS-5 and HS-27a. Blood 2002;100:1509-11. endothelial cell lines derived from murine yolk sac induce the
18. Hackney JA, Charbord P, Brunk BP, Stoeckert CJ, Lemischka proliferation and differentiation of yolk sac CD34+
IR, Moore, KA. A molecular profile of a hematopoietic stem hematopoietic progenitors. Blood. 1995;86,4454-67.
cell niche. Proc Natl Acad Sci USA 2002;99:13061-6. 42. Klemmt PA, Carver JG, Kennedy SH, Koninckx PR, Mardon
19. Liu LQ, Sposato M, Liu HY, Vaudrain T, Shi MJ, Rider K, et al. HJ. Stromal cells from endometriotic lesions and endometrium
Functional cloning of IGFBP-3 from human microvascular from women with endometriosis have reduced decidualiza-
endothelial cells reveals its novel role in promoting prolifera- tion capacity. Fertil Steril. 2006;85:564-72.
tion of primitive CD34+CD38- hematopoietic cells in vitro. 43. Ohneda O, Fennie C, Zheng Z, Donahue C, La H, Villacorta R,
Oncol Res 2003;13:359-71. et al. Hematopoietic stem cell maintenance and differentiation
20. Oostendorp RA, Robin C, Steinhoff C, Marz S, Brauer R, are supported by embryonic aorta-gonad-mesonephros
Nuber UA, et al. Long-term maintenance of hematopoietic region-derived endothelium. Blood. 1998;92:908-19.
stem cells does not require contact with embryo-derived stro- 44. Oostendorp RA, Medvinsky AJ, Kusadasi N, Nakayama N,
mal cells in cocultures. Stem Cells 2005:23:842-51. Harvey K, Orelio C, et al. Embryonal subregion-derived stro-
21. Dzierzak E, Speck NA. Of lineage and legacy: the develop- mal cell lines from novel temperature- sensitive SV40 T anti-
ment of mammalian hematopoietic stem cells. Nat Immunol gen transgenic mice support hematopoiesis. J Cell Sci 2002;
2008;9:129-36. 115:2099-108.
22. Chen MJ, Yokomizo T, Zeigler BM, Dzierzak E, Speck NA. 45. Oostendorp RA, Harvey KN, Kusadasi N, de Bruijn MF, Saris
Runx1 is required for the endothelial to haematopoietic cell C, et al. Stromal cell lines from mouse aorta-gonads-meso-
transition but not thereafter. Nature 2009;457:887-91. nephros subregions are potent supporters of hematopoietic
23. Cumano A, Dieterlen-Lievre F, Godin I. Lymphoid potential, stem cell activity. Blood 2002; 99:1183-9.
probed before circulation in mouse, is restricted to caudal 46. Xu MJ, Tsuji K, Ueda T, Mukouyama YS, Hara T, Yang FC,
intraembryonic splanchnopleura. Cell 1996;86:907-16. Ebihara Y, et al. Stimulation of mouse and human primitive
24. Lux CT, Yoshimoto M, McGrath K, Conway SJ, Palis J, Yoder hematopoiesis by murine embryonic aorta-gonad-meso-
MC. All primitive and definitive hematopoietic progenitor nephros-derived stromal cell lines. Blood 1998;92:2032-40.
cells emerging prior to E10 in the mouse embryo are products 47. Yoder MC, Papaioannou VE, Breitfeld PP, Williams DA.
of the yolk sac. Blood 2007 Murine yolk sac endoderm- and mesoderm-derived cell lines
25. Medvinsky A, Dzierzak E. Definitive hematopoiesis is support in vitro growth and differentiation of hematopoietic
autonomously initiated by the AGM region. Cell 1996;86:897- cells. Blood 1994;83:2436-43.
906. 48. Charbord P, Oostendorp R, Pang W, Herault O, Noel F, Tsuji T,

| 126 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

et al. Comparative study of stromal cell lines derived from genetic protein-4 is required for mesoderm formation and pat-
embryonic, fetal, and postnatal mouse blood-forming tissues. terning in the mouse. Genes Dev 1995;9:2105-16.
Exp Hematol 2002;30:1202-10. 54. Wilt FH. Erythropoiesis in the chick embryo: The role of endo-
49. Ledran MH, Krassowska A, Armstrong L, Dimmick I, derm. Science 1965;147:1588-90.
Renstrom J, Lang R, et al. Efficient hematopoietic differentia- 55. Pardanaud L, Dieterlen-Lievre F. Manipulation of the angiopoi-
tion of human embryonic stem cells on stromal cells derived etic/hemangiopoietic commitment in the avian embryo.
from hematopoietic niches. Cell Stem Cell. 2008;3:85-98. Development 1999;126:617-27.
50. Durand C, Robin C, Dzierzak, E. Mesenchymal lineage poten- 56. Belaoussoff M, Farrington SM, Baron MH. Hematopoietic
tials of aorta-gonad-mesonephros stromal clones. Haematolo- induction and respecification of A-P identity by visceral endo-
gica 2006;91:1172-9.
51. Matsuoka S, Tsuji K, Hisakawa H, Xu M, Ebihara Y, Ishii T, et derm signaling in the mouse embryo. Development 1998; 125:
al. Generation of definitive hematopoietic stem cells from 5009-18.
murine early yolk sac and paraaortic splanchnopleures by 57. Dyer MA, Farrington SM, Mohn D, Munday JR, Baron MH.
aorta-gonad-mesonephros region-derived stromal cells. Blood Indian hedgehog activates hematopoiesis and vasculogenesis
2001;98:6-12. and can respecify prospective neurectodermal cell fate in the
52. Schmerer M, Evans T. Primitive erythropoiesis is regulated by mouse embryo. Development 2001;128:1717-30.
Smad-dependent signaling in postgastrulation mesoderm. 58. Gering M, Patient R. Hedgehog signaling is required for adult
Blood 2003;102:3196-205. blood stem cell formation in zebrafish embryos. Dev Cell
53. Winnier G, Blessing M, Labosky PA, Hogan BL. Bone morpho- 2005;8:389-400.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 127 |
Hematopoiesis

Population dynamics of hematopoietic stem cells

H. Takizawa1 A B S T R A C T
M.G. Manz1,2
Hematopoietic stem cells (HSCs) continuously provide all hemato-lymphoid cells throughout the
1 lifetime of an individual. Moreover, HSCs exist in “excess” as one individual can share its HSCs with
Institute for Research in
Biomedicine (IRB), Bellinzona, another, in which they homeostatically expand and equally form blood for the respective lifetime. This
Switzerland; is demonstrated over several decades by clinical hematopoietic cell, or bone marrow transplantation,
2
Department of Hematology, IOSI, the most advanced and broadly applied somatic stem cell therapy to date. However, as successful as
Bellinzona, Switzerland hematopoietic transplantation works, it is difficult to reveal HSC correlates and to study their biolo-
gy, especially in humans. Here, we here reflect on recent and mostly, in mice generated data on HSC
phenotype, numbers, homeostatic turn-over, and contribution to hematopoiesis, and try to translate
Hematology Education: how this knowledge might impact on our concepts of human HSCs and hematopoiesis in steady-state,
the education program for the hematopoietic challenges as severe bleeding, infections, or HSC transplantation.
annual congress of the European
Hematology Association

2009;3:128-132

ematopoietic stem cells (HSCs) are (FACS) to isolate minute populations or sin-

H defined by their capacity to self-


renew and to give rise to all mature
cell types of the hemato-lymphoid system
gle live cells from bulk cellular populations,
purification of candidate HSCs became tech-
nically possible.3 Although various kinds of
for the lifetime of an individual. HSC-con- culture assays have been established to
taining bone marrow or peripheral blood cell probe HSC activity in vitro, the gold-standard
transplantation from one individual to assay to functionally identify HSCs contin-
another, that is, allogeneic transplantation, ues to be in vivo transplantation followed by
represents the prime example of successful long-term follow-up. That is, lifelong recon-
somatic stem cell therapy with about 20,000 stitution of hematopoiesis in conditioned
transplants worldwide, and about 10,000 (mostly irradiated) mice or in some cases,
transplants in Europe per year.1,2 Although serial transplantation several times into
tremendous headway has been made in another recipient; a task that requires self-
basic understanding of HSC biology, several renewal and most likely homeostatic expan-
long-standing questions still await definitive sion of HSCs. This assay, thus, asks for max-
answers: (i) what identifies HSCs and allows imal engagement of HSCs, that is appropri-
prospective isolation to purity; (ii) how ate homing to a most supportive environ-
many HSCs are contained in a body, in ment and massive contribution to blood for-
which niches do they reside, what deter- mation, and represents a stress test similar to
mines expansion in growing organisms and human hematopoietic cell transplantation,
homeostatic maintenance at fairly constant an engagement not likely to occur often in
levels in adult organisms; (iii) how often and normal adult HSC physiology. Using this
how synchronous do HSCs divide in steady- approach, HSCs have been found to be
state; (iv) do all or only some HSCs con- included in adult bone marrow mature
tribute to hematopoiesis at any given time, hematopoietic lineage marker negative or
or do HSCs engage sequentially and repeti- low (Lin-/lo) cell fractions, express c-Kit (the
tively to blood formation. Touching on these receptor for stem cell factor, SCF) and stem
questions, we discuss recent data generated cell antigen 1 (Sca-1).4,5 This “LSK” popula-
mostly in mouse models and try to extrapo- tion accounts for about 0.08% of total bone
late these findings to human HSCs and marrow nucleated cells and contains all
hematopoiesis, an exercise that is intended HSCs; however, the HSC frequency within
to stimulate fruitful basic-science to medical- the LSK population was determined to be
practice discussions. only about 3.3%.6 Further refinement of
marker combinations determined HSC fre-
Phenotypic identification, numbers, and loca- quency to be about 20% in CD34– LSK,7 and
tion of HSCs in mice about 50% in CD150+CD244–CD48– LSK
With the development of monoclonal that account for 0.006% of total bone mar-
antibody technology in the seventies and row nucleated cells.8 Beyond surface marker
advances in fluorescent-activated cell sorting expression, HSCs can also be enriched by

| 128 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

their biologic property to eliminate drugs efficiently, as Hematopoietic stem cells turnover in mice
for example, DNA binding Hoechest33342, a property Evaluation of HSC turnover has been addressed by in
that defines them as a “side population” by FACS.9 vivo labeling with 5-bromo-2-deoxyuridine (BrdU) that
According to this and previous work, it is estimated that is incorporated in DNA replicating cells.11,26 Animals
the true HSC frequency in an adult mouse accounts for were administered with BrdU for 10-13 days and sub-
0.003% of 5×108 whole body total bone marrow nucle- jected to BrdU-free chase. Results demonstrated that
ated cells, which translates into a total HSC number of about 6-8% of HSCs per day enter cell cycle in a ran-
approximately 15000 per adult, 20-25 g body weight dom fashion and almost all HSCs divide every 8 weeks.
mouse.10-12 HSCs are newly generated during intra-uter- Mathematical simulation provides a powerful tool to
ine development until about mid-gestation, are expand- estimate HSC behavior based on available data and dif-
ed with growth of the organism until young adulthood, ferent assumptions more precisely (i.e., replication,
and are subsequently homeostatically maintained.13 Of apoptosis and differentiation). Based on the experimen-
interest, one mouse contains about 500 HSCs in fetal tal data with BrdU retention, it was estimated that the
liver at day 14.5, about 1000 HSCs at birth, and as dis- steady-state differentiation rate of HSCs to progenitors
cussed above, about 15000 HSCs in adulthood. As mice is 0.01-0.02/day, their transition rate from a quiescent
have a birth weight of about 1 g and gain weight to status to a proliferative phase is 0.02-0.05/day and the
adulthood to about 20-25 g, the expansion factor of rate of apoptosis from proliferative phase is 0.07-
HSC numbers correlates reasonably with the factor of 0.23/day.27 Assuming that all events related to HSCs
body weight gain.14,15 occur independently and randomly (i.e., a stochastic
Bone marrow (BM) has been known to be the primary model), the outcome compared closely to previous
organ of adult hematopoiesis, providing a place to main- studies using limiting-dilution, competitive repopula-
tain HSCs called the HSC “niche.” Recent studies using tion assays.28 However, direct functional evidence for
microscopic analysis revealed that osteoblastic cells lin- HSC turnover was lacking in these models, as the detec-
ing along the bone, form the so-called osteoblastic HSC tion of BrdU requires membrane permeabilization, lead-
niche.16-19 Also, it was suggested that vascular cells sur- ing to cell death. Furthermore, it has been demonstrated
rounding sinusoids within the BM and spleen might that BrdU retention is not HSC specific and that BrdU
form a HSC niche, the so-called vascular niche, especial- has mitogenic effects, that is, it induces HSC prolifera-
ly after gamma-irradiation or HSC mobilization.8,18 tion.11,29 Importantly, BrdU labeling experiments are
However, because of the close proximity of osteoblasts based on the premise that HSCs take up label equally,
to the perivascular region, a clear separation between and estimates would go wrong if there are different
distinct osteoblastic and vascular niches seems to be HSC populations that divide and contribute to
anatomically difficult.17 Of note, upon single cell i.v. hematopoiesis at different frequencies. Thus, the divi-
transplantation HSCs (Lin-/lo c-Kit+ Sca-1+CD34– within sional dynamics of HSC populations have to be evaluat-
the side population) can home to their niche with ed by assays possibly covering all HSCs and allowing,
almost 100% efficiency.20 This demonstrates that HSCs based on sensitive divisional history, functional analysis
are equipped with a very robust machinery that guides as a read out.
homing to the BM niche upon intravenous injection via Recently, HSC turnover has been studied with more
circulation, transmigration through BM endothelium sophisticated divisional tracking systems. One group
barrier, and lodgment in proper HSC niche.21 Given this developed a technique to label cell surface proteins by in
efficacy, it seems very likely that blood to bone marrow vivo biotin injection, and showed that in the hematopoi-
homing is a physiological process occurring on a regular etic compartment, slow dividing cells contain candidate
basis. Indeed, some HSCs (10-100) are found to leave HSCs.30 In disagreement with previous studies, howev-
bone marrow and circulate in blood at any given time er, the observation was that no HSC activity was pres-
and are rapidly cleared again, with some of them locat- ent within a dormant cell population retaining high
ing to the estimated about 1% free HSC niches in bone biotin label at 3 weeks of chase. Other groups used
marrow.22,23 This has been most conclusively demon- genetically engineered mouse strains expressing histon
strated by parabiosis experiments, where mice were sur- H2B-green fluorescent protein (H2B-GFP) controlled by
gically joined to temporarily share their blood circula- a tetracycline-responsive regulatory element.29,31,32 These
tion, which led to continuous both-sided engraftment of transgenic mice were crossed with other strains
hematopoiesis post separation of animals.23 expressing the tetracycline transactivator protein under
What could be the physiological relevance of blood- a CMV promoter, an endogenous scl promoter, or
born HSCs? It has not been demonstrated definitively ROSA26 promoter. Subsequently, doxycycline treat-
yet, but as bone marrow is an organ with multiple loca- ment was used to turn on and shut off H2B-GFP expres-
tions, HSC circulation may be important to keep HSC sion followed by chase. About 20% of cells with HSCs
populations constant in different active marrow spaces phenotype retained H2B-GFP expression after 24-28
throughout life. In addition, circulating HSCs might be weeks and approximately 5% of HSCs were labeled for
able to feed in the thymus to provide T cell progenitors more than 44 weeks. Mathematical simulation assum-
and play a role in innate immunesurveillance by traf- ing a uniform turnover rate of HSCs did not fit the
ficking through peripheral organs and giving rise to experimental data, while a two-population model
myeloid lineage cells upon demand.24,25 Finally, circulat- showed better fit. This indicated the presence of hetero-
ing HSC allow for re-activating extramedullary geneous HSC pools, a dormant pool composed of 15-
hematopoiesis, for example, in spleen and liver after 20% of the entire HSC population that divides every 55
massive blood loss or in the case of critical bone mar- to 145 days, which is equivalent to one fifth of mouse
row space reduction. lifetime, and an actively cycling pool composed of 80-

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 129 |
14th Congress of the European Hematology Association

85% of all HSCs that divides every 9-36 days. mouse or human to human transplants). In contrast,
Transplantation experiments with these populations although serial transplantation is possible, the number
showed that the majority of long-term HSC activity is of repopulating cells and chimerism declines over time
contained in the dormant HSC pool. Importantly, it was indicating that human HSCs are not maintained.34
shown that the dormant HSC pool can be activated to Given the observation that HSC numbers increase
cycle in response to myelosuppression by 5-FU or proportional to body weight in growing mice from
gamma-irradiation, and subsequently can reach a dor- birth to adulthood, it seems reasonable to make the
mant state again after homeostasis is reestablished, con- assumption that HSC numbers correlate with body
firming a reversible cycling phenotype of HSCs depend- weight, at least within the range of a normal body mass
ing on the environmental context. From these studies, index. An about 70 kg human adult contains about
the dormant HSC pool would play a role as reservoir for 1.5×1012 nucleated bone marrow cells as determined by
catastrophic situations of hematopoiesis, such as exces- 59
Fe distribution.37 Indeed human total bone marrow
sive bleeding or acute infections. Keeping some HSCs in cells to mouse total bone marrow cells reflect the same
an inactive state by reducing the metabolic machinery ratio as human body weight to mouse body weight (70
could be an evolved strategy to protect HSCs from kg-25 g). Given the assumption that human bone mar-
cycling and aging related changes33 and replication asso- row contains the same HSC frequency as observed in
ciated DNA mutations, potentially leading to cell death mice (0.003%, discussed above), an adult 70 kg human
or cancer. being would contain about 45×106, and a 3.5 kg new-
Some limitations to the above described new divi- born about 2.25×106 HSCs.
sional tracking systems remain. Although calculations Similarly, calculations for HSC turn-over can be made.
on HSC turnover rate were performed under the If mouse HSC cycle once every 21-145 days, they divide
assumption that more than five divisions are needed 5 to 35 times within a 2-year lifetime of a mouse, num-
until H2B-GFP expression becomes undetectable, it is bers that seem reasonable given the assumption of the
not feasible to evaluate the exact number of divisions. Hayflick limit. If human HSCs would divide with the
This is because uniform fluorescent intensity cannot be same frequency in a 75-year lifetime, they would divide
achieved at beginning of chase, and thus, numbers of 188-1300 times. Alternatively, cycling frequency could
divisions are difficult to assess and might lead to over- be kept constant and adapted to life expectancy. If this
estimation of the dormant pool size, given dormancy holds true, human HSCs would divide 5-35 times in 75
defines zero division. To address HSC cycling kinetics years, accounting for a HSC division every 2-15 years.
on a one-by-one divisional basis, we employ CFSE (car- Interestingly, measurement of telomere length in
boxyfluorescein diacetate succinimicyl ester) labeling of humans from the age of 0-90 years are compatible with
HSCs and subsequent transfer into non-conditioned 15-30 HSC divisions in early life, followed by less than
recipients, thus mimicking steady-state HSC traveling one division per year in the following years, accounting
and turn-over as described above. for about 70-100 divisions during a lifetime.38 Thus, the
Questions that have not been answered yet in the second assumption seems to get relatively close to real-
series of these studies are: (i) whether some actively life human HSC cycling frequency.
cycling HSCs might re-enter quiescence even in steady- Nevertheless, it would be reasonable and valuable to
state; (ii) whether all HSCs at any given time contribute investigate larger animals to estimate human HSC biol-
to blood formation and (iii) to what extend distinct HSC ogy. Surprisingly, evaluation of HSCs in cats revealed
pools are regulated by intrinsic and extrinsic, environ- that the total number (11.400±5.400) is very close to
mental programs in steady-state and upon challenge. that of mice, which was taken as evidence that the
number of HSCs is conserved in mammals.39,40 However,
Implications for hematopoietic stem cells biology in since the demand for blood production per day increas-
humans es with body size, HSCs in larger mammals need to pro-
Comparatively little is known about pool size, surface duce more blood per cell. Further studies on baboons
phenotype, cycling, and localization of human HSCs and macaques (weigh; 11-26 kg and 5-8 kg, life span; 30-
due to practical and ethical constrains, as well as due to 45 years and 25 years, respectively) by retrovius-medi-
lack of representative and predictive animal models. ated HSC gene marking experiments and telomere
Both in vitro and in vivo experiments have been utilized length analysis revealed that the turnover rate of
to identify human HSCs. Using the long-term culture- baboon HSCs was once per 23-36 weeks. Extrapolation
initiating cell (LTC-IC) assay, where cells are cultured of human HSC replication rate from this data resulted in
for several weeks on bone marrow feeder cells and the 192 times turn-over during a life-time, which is similar
immunodeficient mouse repopulating assay, human to the frequency observed by telomere-shortening in
HSCs were found in the Lin–CD34+CD38-/lo frac- human blood, and would be consistent with a con-
tion.3,34,35 Recently, it was shown that successful long- served HSC cycling number among species.38,41 While an
term, that is, at least 3 months engraftment was adult mouse contains about 2 mL, an adult human con-
observed in NOD/SCID/IL2Rγ-/- mice transplanted with tains about 5000 mL of blood, accounting for a factor of
as few as 10 human HSCs enriched cells defined by Lin- 2500 difference. If, however, the HSC and the HSC
CD34+CD38–CD90+CD45RA– from umbilical cord cycling numbers are conserved in mammals, there must
blood or bone marrow.36 However, it should be noted be other mechanisms ensuring sufficient blood produc-
that the NOD/SCID immunodeficient mouse repopu- tion. This could be an increased active HSC pool size
lating assay and its variants, which are currently used as contributing to hematopoiesis correlating with animal
best human HSC surrogate assay, do not allow homeo- size, a scaled number of committed progenitors produc-
static expansion of HSCs (as observed in mouse to ing differentiated cells, a longer time and burst size from

| 130 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

HSC to mature cells, and a reduced frequency of blood in the clinic, understanding HSC population dynamics
cell death. Concerning the first possibility, the dilemma in steady-state and upon transplantation has been
of different blood cell demand with the same numbers proven difficult and only made substantial progress
of HSCs was mathematically approached using “allo- recently in small animal models. The challenge for the
metric scaling” of the active hematopoietic stem cell future remains to translate this knowledge to clinical
pool. In this model, the active HSC pool increases across settings in humans in order to bridge the gap between
mammalian species (40, 385, 4690, and 9640 active basic research and clinical medicine.
HSCs for cats, humans, pilot whales, and Asian ele-
phants, respectively).42 Of note, the size of the active This work was in part supported by a Postdoctoral
HSC pool was consecutively used to explain the higher Fellowship of the Japanese Society for the Promotion of Science
frequency of acquired HSC disorders in humans versus for Research Abroad to H.T., the Swiss National Science
cats or mice.43 However, given the recent data on mouse Foundation (310000-116637), the Oncosuisse (OCS-02019-
HSC turn-over and active HSC pool size, other possible 02-2007), and the Bill and Melinda Gates Foundation to
explanations as replication time and size of each M.G.M.
hematopoietic compartment need to be addressed
experimentally and by computational analysis.44
Overall, we believe that, similar as scaling up gut crypt References
stem cells in longer intestines in large versus small ani-
mals, scaling up HSC number with size and weight of 1. Appelbaum FR. Hematopoietic-cell transplantation at 50. N
Engl J Med 2007;357:1472-5.
species would be the evolutionary most simple solution 2. Gratwohl A, Baldomero H, Frauendorfer K, Niederwieser D.
to the problem. Why are there regional differences in stem cell transplantation
activity? An EBMT analysis. Bone Marrow Transplant 2008;
Hematopoietic stem cell transplantation 42:S7-S10.
3. Kondo M, Wagers AJ, Manz MG, Prohaska SS, Scherer DC,
In syngeneic mice, 2×105 nucleated bone marrow cells Beilhack GF, et al. Biology of hematopoietic stem cells and
are sufficient to rescue an animal from irradiation that progenitors: implications for clinical application. Annu Rev
Immunol 2003;21:759-806.
otherwise would lead to death from bleeding and ane- 4. Spangrude GJ, Heimfeld S, Weissman IL. Purification and
mia within about 3 weeks45 and establish a lifelong characterization of mouse hematopoietic stem cells. Science
100% chimerism. In fully MHC mismatched mice, 1988;241:58-62.
5. Uchida N, Weissman IL. Searching for hematopoietic stem
about 10-fold more bone marrow cells are needed. This cells: evidence that Thy-1.1lo Lin- Sca-1+ cells are the only
correlates with 0.05% and 0.5% of total bone marrow stem cells in C57BL/Ka-Thy-1.1 bone marrow. J Exp Med
cells of a mouse, or 7 and 70 HSCs, respectively. These 1992;175:175-84.
HSCs re-establish by homeostatic expansion the HSC 6. Osawa M, Nakamura K, Nishi N, Takahasi N, Tokuomoto Y,
Inoue H, et al. In vivo self-renewal of c-Kit+ Sca-1+ Lin(low/)
pool within about 4 months,46 that is, the HSC pool hemopoietic stem cells. J Immunol 1996156:3207-14.
needs to expand 200 to 2000 fold. 7. Osawa M, Hanada K, Hamada H, Nakauchi H. Long-term
In HLA matched human hematopoietic cell transplan- lymphohematopoietic reconstitution by a single CD34-
low/negative hematopoietic stem cell. Science 1996;273:242-
tation, a marrow nucleated cell dose of 3.65×108/kg 5.
body weight was reported to be associated with a 8. Kiel MJ, Yilmaz OH, Iwashita T, Yilmaz OH, Terhorst C,
favorable outcome,47 and a median number of 2×108 Morrison SJ. SLAM family receptors distinguish hematopoiet-
ic stem and progenitor cells and reveal endothelial niches for
bone marrow nucleated cells/kg body weight was stem cells. Cell 2005;121:1109-21.
reported to be transplanted in the 2008 EBMT 9. Goodell MA, Brose K, Paradis G, Conner AS, Mulligan RC.
Handbook of Hematopoietic Stem Cell Transplantation. Isolation and functional properties of murine hematopoietic
In a 70 kg individual, this correlates with about 2% and stem cells that are replicating in vivo. J Exp Med 1996;183:
1797-806.
1% of total nucleated marrow cells, respectively. If 10. Boggs DR. The total marrow mass of the mouse: a simplified
humans, similar as mice have a HSC frequency of method of measurement. Am J Hematol 1984;16:277-86.
0.003%, transplants contain 0.9×106 and 0.45×106 true 11. Kiel MJ, He S, Ashkenazi R, Gentry SN, Teta M, Kushner JA,
et al. Haematopoietic stem cells do not asymmetrically segre-
HSCs. If, however, the HSC number is conserved in gate chromosomes or retain BrdU. Nature 2007;449:238-42.
mammals, transplants would contain about 300 and 150 12. Sudo K, Ema H, Morita Y, Nakauchi H. Age-associated charac-
HSCs. In any case, the transplanted HSC pool needs to teristics of murine hematopoietic stem cells. J Exp Med
2000;192:1273-80.
expand 50-100 fold to re-establish normal homeostatic 13. Cumano A, Godin I. Ontogeny of the hematopoietic system.
HSC levels. Annu Rev Immunol 2007;25:745-85.
Of note, while humans receive 2-40 fold more bone 14. Bowie MB, Kent DG, Dykstra B, McKnight KD, McCaffrey L,
Hoodless PA, et al. Identification of a new intrinsically timed
marrow cells compared to mice relative to their respec- developmental checkpoint that reprograms key hematopoiet-
tive total marrow cell numbers, mice need to receive no ic stem cell properties. Proc Natl Acad Sci USA 2007;104:5878-
further treatment, while humans frequently need to be 82.
substituted with red blood cells and thrombocytes. Thus, 15. Kim I, He S, Yilmaz OH, Kiel MJ, Morrison SJ. Enhanced
purification of fetal liver hematopoietic stem cells using SLAM
there seems to be substantial differences in the dynamics family receptors. Blood 2006;108:737-44.
of blood formation between species, an issue that needs 16. Calvi LM, Adams GB, Weibrecht KW, Weber JM, Olson DP,
to be accounted for in future comparative analysis. Knight MC, et al. Osteoblastic cells regulate the haematopoi-
etic stem cell niche. Nature 2003;425:841-6.
17. Lo Celso C, Fleming HE, Wu JW, Zhao CX, Miake-Lye S,
Conclusions Fujisaki J, et al. Live-animal tracking of individual haematopoi-
HSCs exist in “excess” as one individual can share its etic stem/progenitor cells in their niche. Nature 2009;457:92-6.
18. Xie Y, Yin T, Wiegraebe W, He XC, Miller D, Stark D, et al.
HSCs with another in which these homeostatically Detection of functional haematopoietic stem cell niche using
expand and form blood for the respective lifetime. real-time imaging. Nature 2009;457:97-101.
While HSC transplantation has been proven successful 19. Zhang J, Niu C, Ye L, Huang H, He X, Tong WG, et al.
Identification of the haematopoietic stem cell niche and con-

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 131 |
14th Congress of the European Hematology Association

trol of the niche size. Nature 2003;425:836-41. diploid cell strains. Exp Cell Res 1961;25:585-621.
20. Matsuzaki Y, Kinjo K, Mulligan RC, Okano H. Unexpectedly 34. Manz MG. Human-hemato-lymphoid-system mice: opportu-
efficient homing capacity of purified murine hematopoietic nities and challenges. Immunity 2007;26:537-41.
stem cells. Immunity 2004;20:87-93. 35. Shultz LD, Ishikawa F, Greiner DL. Humanized mice in trans-
21. Lapidot T, Dar A, Kollet O. How do stem cells find their way lational biomedical research. Nat Rev Immunol 2007;7:118-30.
home? Blood 2005;106:1901-10. 36. Majeti R, Park CY, Weissman IL. Identification of a hierarchy
22. Bhattacharya D, Rossi DJ, Bryder D, Weissman IL. Purified of multipotent hematopoietic progenitors in human cord
hematopoietic stem cell engraftment of rare niches corrects blood. Cell Stem Cell 2007;1:635-45.
severe lymphoid deficiencies without host conditioning. J Exp 37. Skarberg KO. Cellularity and cell proliferation rates in human
Med 2006;203:73-85. bone marrow. I. An in vivo method to estimate the total mar-
23. Wright DE, Wagers AJ, Gulati AP, Johnson FL, Weissman IL. row cellularity in man. Acta Med Scand 1974;195:291-9.
Physiological migration of hematopoietic stem and progenitor 38. Rufer N, Brummendorf TH, Kolvraa S, Bischoff C, Christensen
cells. Science 2001;294:1933-6. K, Wadsworth L, et al. Telomere fluorescence measurements
24. Massberg S, Schaerli P, Knezevic-Maramica I, Kollnberger M, in granulocytes and T lymphocyte subsets point to a high
Tubo N, Moseman EA, et al. Immunosurveillance by hemato- turnover of hematopoietic stem cells and memory T cells in
poietic progenitor cells trafficking through blood, lymph, and early childhood. J Exp Med 1999;190:157-67.
peripheral tissues. Cell 2007;131:994-1008. 39. Abkowitz JL, Catlin SN, McCallie MT, Guttorp P. Evidence
25. Nagai Y, Garrett KP, Ohta S, Bahrun U, Kouro T, Akira S, et al. that the number of hematopoietic stem cells per animal is con-
Toll-like receptors on hematopoietic progenitor cells stimulate served in mammals. Blood 2002;100:2665-7.
innate immune system replenishment. Immunity 2006;24: 40. Gordon MY, Lewis JL, Marley SB. Of mice and men... and ele-
801-12. phants. Blood 2002;100:4679-80.
26. Cheshier SH, Morrison SJ, Liao X, Weissman IL. In vivo prolif- 41. Shepherd BE, Kiem HP, Lansdorp PM, Dunbar CE, Aubert G,
eration and cell cycle kinetics of long-term self-renewing
hematopoietic stem cells. Proc Natl Acad Sci USA 1999;96: LaRochelle A, et al. Hematopoietic stem-cell behavior in non-
3120-5. human primates. Blood 2007;110:1806-13.
27. MacKey MC. Cell kinetic status of haematopoietic stem cells. 42. Dingli D, Pacheco JM. Allometric scaling of the active
Cell Prolif 2001;34:71-83. hematopoietic stem cell pool across mammals. PLoS ONE
28. Abkowitz JL, Golinelli D, Harrison DE, Guttorp P. In vivo 2006; 1:e2.
kinetics of murine hemopoietic stem cells. Blood 2000;96: 43. Lopes JV, Pacheco JM, Dingli D. Acquired hematopoietic
3399-405. stem-cell disorders and mammalian size. Blood 2007;110:
29. Wilson A, Laurenti E, Oser G, van der Wath RC, Blanco-Bose 4120-2.
W, Jaworski M, et al. Hematopoietic stem cells reversibly 44. Dingli D, Traulsen A, Pacheco JM. Compartmental architec-
switch from dormancy to self-renewal during homeostasis ture and dynamics of hematopoiesis. PLoS ONE 2007;2:e345.
and repair. Cell 2008;135:1118-29. 45. Na Nakorn T, Traver D, Weissman IL, Akashi K. Myelo-
30. Nygren JM, Bryder D. A novel assay to trace proliferation his- erythroid-restricted progenitors are sufficient to confer radio-
tory in vivo reveals that enhanced divisional kinetics accompa- protection and provide the majority of day 8 CFU-S. J Clin
ny loss of hematopoietic stem cell self-renewal. PLoS ONE Invest 2002;109:1579-85.
2008;3:e3710. 46. Allsopp RC, Cheshier S, Weissman IL. Telomere shortening
31. Challen GA, Goodell MA. Promiscuous expression of H2B- accompanies increased cell cycle activity during serial trans-
GFP transgene in hematopoietic stem cells. PLoS ONE 2008; plantation of hematopoietic stem cells. J Exp Med 2001;
3:e2357. 193:917-24.
32. Foudi A, Hochedlinger K, Van Buren D, Schindler JW, Jaenisch 47. Sierra J, Storer B, Hansen JA, Bjerke JW, Martin PJ, Petersdorf
R, Carey V, et al. Analysis of histone 2B-GFP retention reveals EW, et al. Transplantation of marrow cells from unrelated
slowly cycling hematopoietic stem cells. Nat Biotechnol donors for treatment of high-risk acute leukemia: the effect of
2009;27:84-90. leukemic burden, donor HLA-matching, and marrow cell
33. Hayflick L, Moorhead PS. The serial cultivation of human dose. Blood 1997;89:4226-35.

| 132 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Hematopoiesis

Dynamic interactions between the nervous and


immune systems with the microenvironment regulate
hematopoietic stem cell migration and development.
The essential roles of SDF-1 and CXCR4

K. Lapid A B S T R A C T
T. Lapidot
Hematopoietic stem cell (HSC) migration and development are tightly regulated processes in order
to produce and replenish the circulation with immature and maturing blood cells on demand.
Department of Immunology,
Weizmann Institute of Science, Physiological interactions between the nervous and immune systems, with local microenvironments in
Rehovot, Israel various organs throughout the body are needed to maintain homeostasis. These interactions include
dynamic regulation of bone remodeling, orchestrating the evolving stem cell niches, which impact
indirectly on rare, HSC populations and their progeny as part of host defense and repair mechanism.
Signals by the nervous system also directly regulate the motility and proliferation of hematopoietic
Hematology Education:
stem and progenitor cells (HSPC) that hierarchically express functional neurotransmitter receptors,
the education program for the
annual congress of the European which are also regulated by myeloid cytokines. Reciprocal effects of activated leukocytes, neuronal
Hematology Association and stromal cells via secretion of cytokines, chemokines, neurotransmitters and proteolytic enzymes
are part of the dynamic crosstalk between these overlapping systems, providing a stem cell regulato-
2009;3:133-139 ry brain bone blood triad. Homing, retention, egress, recruitment and clinical mobilization of HSPCs
will be discussed in the context of this crosstalk, focusing on the essential roles of the chemokine
Stromal Derived Factor-1 (SDF-1) and its major receptor CXCR4 in the functional preclinical NOD/SCID
mouse model.

ost blood forming stem and progen- one amino acid and are cross reactive.13

M itor cells are located in the bone


marrow (BM), anchored to special-
ized niches; however, there are very low lev-
Currently, this ligand is the only known
powerful chemotactic factor for human and
murine HSPCs14,15 In vivo migration of both
els of motile progenitors in the circulation as normal and leukemic human stem cells, and
part of homeostasis.1 These low levels are their engraftment of the BM of transplanted
dramatically amplified during alarm situa- immune deficient mice are dependent on
tions in response to stress signals due to SDF-1/CXCR4 interactions.8,14,16,17 Apart from
injury, bleeding, infections and inflamma- mediating hematopoietic stem cell motility,
tion, and as part of host defense and repair SDF-1 plays roles in survival and quiescence,
mechanism.2,3 Stress induced recruitment of and murine HSCs from inducible KO of
progenitor cells to the circulation is mim- CXCR4 demonstrate uncontrolled cycling
icked in clinical mobilization protocols by and impaired repopulation ability.7,18 This
chemotherapy and/or repeated granulocyte review will elaborate on the SDF-1/CXCR4
colony stimulating factor (G-CSF) stimula- dependent regulation of normal and
tions, in which immature cells with repopu- leukemic human CD34+ progenitors, dis-
lation potential are harvested from the blood cussing both engraftment and induced mobi-
for autologous and allogeneic transplanta- lization processes in chimeric NOD/SCID
tions of myeloablated patients.2,4,5 HSPCs are mice and also wild type murine HSPC.
identified and characterized in functional Furthermore, the emerging concept of the
transplantation assays based on their ability microenvironmetal regulation of HSCs
to migrate in the circulation, home to the mobilization and recruitment will be dis-
host BM and durably repopulate it with cussed, involving bone remodeling and sig-
immature and maturing leukocytes, which nals from the nervous system.
in turn are released back to the circulation on
demand. The chemokine SDF-1 (also termed Homing and engraftment of immature human
CXCL12), and its major receptor CXCR4, CD34+ cells in immune-deficient mice
regulate many types of stem cells and play Functional preclinical animal models for
pivotal roles during development. SDF-1 is engraftment of human HSPCs (derived
highly expressed in the BM by stromal cells, from human BM, cord blood or G-CSF
including bone-lining osteoblasts, endothe- mobilized peripheral blood) were devel-
lial, reticular and perivascular cells.6-8 Murine oped in pre-immune sheep and immune
embryos that lack SDF-1 or CXCR4 have deficient mice.19,20 Several immune deficient
multiple lethal defects, including lack of fetal strains have been developed, of which
BM seeding by hematopoietic progenitor Non-Obese Diabetic Immune Deficient
cells.9-11 In addition, cleavage of SDF-1 by (NOD/SCID) is the most common model,
CD26 reduces homing of murine HSPC to lacking mature T and B cells and having
the BM.12 Human and murine SDF-1 differ in weak innate immunity.21 Other strains that

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 133 |
14th Congress of the European Hematology Association

lack also NK cell activity, such as NOD/SCID/β2m22 tion of human progenitor cells, revealing a two way
and NOD/SCID/IL-2Rγ-/- (NOG)23 mice are routinely cross talk in which inhibition of adhesion molecules
used today. These models have identified primitive over-rules SDF-1 induced activation and migration,
human CD34+/CD38–/low cells with long term repopu- which could lead to their retention in the BM.40
lation in transplanted animal recipients.24,25 These pre- Altogether, it is concluded that by triggering chemo-
clinical immune deficient mouse models can predict taxis and adhesion machineries, SDF-1/CXCR4 guide
clinical outcome in leukemia patients and repopulation human HSPCs into the BM of recipient chimeric mice,
of normal cells in these mice reflects on their clinical enabling efficient homing and engraftment.
potential.26,27 Homing, engraftment and mobilization of Nevertheless, the SDF-1/CXCR4 story does not end in
human CD34+ enriched progenitors in chimeric cell-autonomous mechanisms, and in fact, harbors a
NOD/SCID mice are dependent on CXCR4 signal- broader concept. We have found that BM endothelial
ing,14,28,29 demonstrated by direct blockage of CXCR4 cells, which also express CXCR4 on their surface, can
by neutralizing antibodies. Transplantation of translocate functional SDF-1 from the circulation in a
enriched human CD34+ cells overexpressing CXCR430 CXCR4 dependent manner, controlling SDF-1 levels in
or in vivo pre-treatment with intravenously injected the BM, and thus actively guide transplanted HSPCs in
SDF-1,31 result in enhanced homing and repopulation their course of homing to the BM.42 These findings
capacity, further supporting the essential roles of SDF- open a door to the involvement of other cellular fac-
1/CXCR4 in the engraftment process. This significance tors in the environment, regulating availability of SDF-
is manifested in stress situations as well; for instance, 1 and affecting indirectly stem cell function (will be
when chemotherapy or irradiation are applied, it discussed later on).
results in increased SDF-1 expression in the BM of
recipient mice, thereby mediating increased repopula- The leukemic point of view
tion by transplanted cells.6 Of note, pre-conditioning Utilization of the immune deficient mice models
by irradiation or chemotherapy is essential for durable enabled identification of human acute myeloid
high level reconstitution by transplanted cells. In vitro leukemia (AML) and Pre-B acute lymphoblastic
SDF-1 induced migration capacity of mobilized CD34+ leukemia (ALL) initiating cells, and more recently, other
enriched human cells correlates with their in vivo types of human cancer stem cells.43,44 Human Pre-B ALL
repopulation potential in autologous transplanted cells home to the same SDF-1-rich vascular micro-
patients32 and immune deficient mice.14 The lipid medi- domains as murine HSPCs in the BM, and antagonizing
ator Sphingosine-1 Phosphate (S1P) also serves as a CXCR4 abolishes their homing.45 Moreover, leukemic
modest chemoattractant for progenitor cells. Using an stem cells were recently found to utilize the same spe-
agonist for its receptor (S1P1) augments SDF-1 induced cialized stem cell niches rich area in the BM, the endos-
migration of human CD34+ cells, resulting in increased teum region, which provides chemoprotective signals to
homing,33 while overexpression of S1P1 leads to reduc- the AML stem cells46 (also see Figure 1). Since leukemic
tion in human CD34+ cell migration and homing by cells are derived from hematopoietic progenitors and
downregulating CXCR4 expression.34 Various share overlapping features, it is plausible to assume that
cytokines, such as Hepatocyte Growth Factor (HGF), SDF-1/CXCR4 contribute to their malignant pheno-
interleukin-6, Stem Cell Factor (SCF) and FLT-3 lig- type. Indeed, blockage of CXCR4 reduces homing by
and,29,35,36 as well as other factors, such as human Pre-B ALL and AML cells to the BM of recipient
prostaglandins,37 can upregulate CXCR4 expression or NOD/SCID mice.16,27 Likewise, CXCR4 is important in
enhance CXCR4 signaling in immature human CD34+ chronic myeloid leukemia pathogenesis.47 AML cells
cells, and thus, improve their motility and repopula- were found to be more CXCR4 dependent than normal
tion potential. Taken together, these findings imply human repopulating progenitor cells, and exhibited
that not only SDF-1 expression is dynamically poor engraftment upon anti-CXCR4 treatment in pre-
expressed in the BM, but also CXCR4. Interestingly, established chimeras.16 A similar observation has been
CXCR4 can be functionally transferred from platelet shown for human AML with unique anti-CD44 Ab
microparticles to immature human CD34+ cells and treatment, which eradicated AML in chimeric mice,
augment their SDF-1 mediated homing and engraft- while only minimally affecting normal human
ment potential.38 Understanding the mechanisms gov- hematopoietic cells.48 It should be noted that CXCR4+
erning SDF-1 and CXCR4 activity may assist in plasma microparticles contribute to the homing process
improving current transplantation protocols. SDF-1 of human AML cells, in a similar fashion to normal
activates the major adhesion molecules LFA-1, VLA-4, human HSPCs, by transferring membranal CXCR4, and
VLA-5 and CD44, which participate in human CD34+ are, as a result, involved in the progression of the dis-
cell homing and repopulation in transplanted ease.38,49 Furthermore, high expression of CXCR4 corre-
NOD/SCID mice.39-41 These adhesion molecules inter- lates with poor prognosis in AML patients.50 These find-
act with Extra-Cellular Matrix (ECM) components, ings suggest that mechanisms governing homing and
such as fibronectin and hyaluronan, which are also engraftment of normal human HSPCs are also crucial
expressed in the BM niches, as well as with adhesion for understanding human leukemias.
molecules, such as VCAM-1 and ICAM-1, expressed
by BM endothelial cells, reticular cells and osteoblasts. Stem cell recruitment and mobilization
All of which are important for active homing to the Recruitment of HSPCs to the peripheral blood (PB)
BM and retention of HSPCs, which is pre-requisite for following treatment with chemotherapy, or cytokines,
subsequent successful repopulation.19 Of interest, neu- is a clinical process termed mobilization.2 This process
tralization of CD44 prevents SDF-1 mediated activa- mimics enhancement of the physiological release of

| 134 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

(A) An illustration of a femur with


its regions. (B) DiR labeled human
pre-B ALL G2 cells were injected
into a NOD/SCID mouse. Homing
was examined 16 hours post trans-
plantation. The obtained femur was
quickly frozen and cut smoothly in
a cryostat device. The image was
taken using a CCD controlled fluo-
rescent microscope. Leukemic
cells localize closely to the endos-
teum along the diaphysis (yellow
arrow) and in trabeculae-rich
regions in the metaphysis and epi-
physis (green arrow).

HSPCs from the BM reservoir in response to stress sig- enzymes, such as matrix metalloproteinases (MMPs),
nals during injury and inflammation as part of host which in turn deactivate SDF-1 by its cleavage. In fact,
defense and repair. Mobilization requires active naviga- proteolytic activity enables breakdown of adhesion
tion of stem cells across physical endothelial and ECM interactions in the BM and loss of retention in the
barriers to the circulation, which requires proteolytic course of recruitment and mobilization processes,4 in
enzymes activity and breakdown of adhesion interac- which SDF-1 induced MMP-9 plays a major role by
tions. The SDF-1/CXCR4 axis plays a pivotal role in this cleaving membrane bound SCF.61 The released soluble
active navigation, since concomitant blockage of SCF augments cell motility. G-CSF induced mobiliza-
CXCR4 or SDF-1 by neutralizing antibodies, during tion is associated with neutrophilia,62 and activated neu-
repetitive G-CSF stimulations, prevents both human trophils secrete serine proteases, such as neutrophil elas-
and murine progenitor cell mobilization.28 Lack of mobi- tase and cathespsin G, which have been shown to
lization upon G-CSF treatment in murine CXCR4-/- BM cleave SDF-1,28 CXCR4,63 c-Kit64 and VCAM-1.65
chimeras supports this observation.51 Interference with Therefore, inhibition of elastase results in reduced G-
CXCR4 signaling needed for HSPC retention in the BM, CSF induced mobilization.28 In spite of these observa-
also results in progenitor cell mobilization, as demon- tions, mice lacking these proteases still react normally to
strated by inhibition of PKCζ and RAC1 activities.37,52,53 G-CSF treatment and are capable of cleaving SDF-1,66
CXCR4 levels are increased on BM HSPCs following G- indicating redundancy among proteases activity.
CSF treatment, enhancing their migration potential Indeed, in addition to secreted proteases, membranal
towards SDF-1.28 On the other hand, SDF-1 levels are proteases were found to participate in the SDF-1 cleav-
reduced in the BM and are transiently increased in the age activity, including CD26 and MT1-MMP, which are
PB following G-CSF treatment, implying that by con- also expressed by the progenitor cells themselves.67,68
trolling SDF-1 levels, HSCs could be recruited to site of The latter has also been shown to mediate G-CSF
demand.28 For example, upon liver injury or limb induced mobilization of immature human CD34+ cells
ischemia, SDF-1 expression is increased, mediating spe- by CD44 cleavage. Our data reveal that RECK, which is
cific recruitment of progenitor cells to these organs as the endogenous inhibitor of MT1-MMP and MMP2/9,
part of host defense and repair mechnaisms.54,55 In a sim- is inversely regulated on human CD34+ HSPCs and
ilar fashion, high levels of S1P in the blood and lymph mouse BM cells.67 Upon G-CSF treatment, MT1-MMP
attract circulating murine HSPCs, which navigate their expression is increased, while RECK expression is
ways to peripheral organ, where they can undergo decreased, emphasizing the importance of balanced
induced activation and differentiation.56 Repetitive daily proteolytic activity in the BM. Since SDF-1 and CXCR4
administrations of SDF-1 or a single dose administration interactions are essential for HSC retention, as observed
of the Met-SDF-1 analog, which has a longer half life in by increased circulating HSCs in inducible CXCR4 KO
vivo, trigger preferential mobilization of murine progen- mice,18 it is logical to assume that disruption of this
itor cells.57-59 Notably, during G-CSF induced mobiliza- interaction would result in loss of retention. The bicy-
tion, SDF-1 levels in the BM are transiently increased, clam AMD3100 was found to cause rapid mobilization
followed by their eventual reduction at mRNA and pro- of mouse, human and non-human primate HSPCs.69-71
tein levels.28,60 SDF-1 stimulations activate proteolytic AMD3100 treatment also synergistically augments G-

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 135 |
14th Congress of the European Hematology Association

CSF induced mobilization. Moreover, AMD3100 mobi- and survival.82,83 In addition, G-CSF, SDF-1 or HGF
lized progenitors demonstrate increased SDF-1 induced administration induces both an increase in osteoclast
migration in vitro and in vivo repopulation potential,69 numbers and preferential mobilization of progenitors.57
synergized upon combination of AMD3100 and G-CSF Stress-induced situations, such as mild bleeding and LPS
treatments. Interestingly, preliminary results reveal that treatment, also trigger HSPCs mobilization concomi-
AMD3100 rapidly induces increased SDF-1 secretion tantly with osteoclast activity. Indeed, we have demon-
from the BM stromal cells to the circulation.72 Blocking strated that osteoclasts are directly involved in inducing
the SDF-1/CXCR4 axis, using neutralizing antibodies to progenitor cell egress by cathepsin K mediated degrada-
either SDF-1 or CXCR4, in steady state and in tion of major endosteal niche components, such as SDF-
AMD3100 treated mice, reduced mobilization of 1, osteopontin and SCF,57 linking bone remodeling with
HSPCs, but not mature WBCs,72 strengthening the mobilization of HSPCs. Osteoclast activation by
hypothesis that this pathway is relatively more selective RANKL preferentially increases mobilization of imma-
for immature cells. These results complicate the theory ture cells, whereas osteoclasts inhibition by the hor-
of passive disruption of SDF-1/CXCR4 interactions by mone clacitonin decreases G-CSF and LPS induced
AMD3100, suggesting that active SDF-1/CXCR4 is also mobilization, as well as homeostatic release of progeni-
necessary for steady state egress and AMD3100 induced tors. Of note, mobilization induced by repeated stimu-
progenitor cell mobilization. Our preliminary experi- lations with RANKL, HGF or SDF-1 was selective in
ments reveal that AMD3100 does not affect only terms of mobilizing HSPCs to the PB, but not mature
hematopoietic cells, but also BM stromal cells, which leukocytes.57 Utilization of PTPε deficient young female
also express CXCR4.42,72 In response to AMD3100, SDF- mice, which have transient and mild impairment of
1 secretion from BM endothelial cells and osteoblasts in osteoclast adhesion and resorption,84 also demonstrated
vitro and in vivo to the circulation is increased, further reduced numbers of circulating progenitors and capaci-
contributing to transient, local SDF-1 gradients towards ty to mobilize, even in response to repeated RANKL
the blood, after which, motile HSPCs follow. In conclu- stimulations, despite normal levels of immature cells in
sion, the involvement of other cellular players, such as the BM.57 Another example for the involvement of
neutrophils and BM stromal cells, in mobilization osteoclasts in the mobilization process is presented by
processes, strongly imply that regulation of cell- CD45 KO mice. CD45 is a pan leukocyte phosphatase
autonomous motility properties are only part of the pic- that is dynamically expressed only on leukocytes. CD45
ture that includes a complex regulation by the BM KO mice demonstrate defective osteoclast activity,
microenvironment. altered metaphysial trabecular bone structure, resem-
bling mild osteopetrosis, and reduced BM pool of prim-
A cross-talk between bone remodeling and stem cell itive HSPCs.85 RANKL and suboptimal G-CSF induced
egress, recruitment and mobilization mobilization are consequently impaired in these mice.
Bones are dynamic and continuously undergo forma- These results reveal that HSPCs are involved in regulat-
tion by bone lining osteoblasts and resorption by the ing their own levels and the dynamic BM microenviron-
stem cell derived fused-monocytes, termed osteoclasts, ment via their osteoclast progeny. Parathyroid hormone
throughout life. Osteoblasts are involved in maintaining (PTH) maintains calcium homeostasis by accelerating
quiescent HSCs at the endosteal niches and play a role bone remodeling, demonstrated by proliferation of
in supporting BM hematopoiesis.73 Genetic manipula- osteoblasts that express its receptor, which is followed
tions that cause an increase or a decrease in osteoblasts by osteoclast activation. The effects of PTH on
numbers have led to an increase or a decrease in HSCs osteoblasts has been shown to increase the HSC pool
numbers respectively.74-76 Following G-CSF administra- size via the Notch pathway;74 however, PTH adminis-
tion, BM SDF-1 mRNA levels are reduced, and it was tration also results in HSC mobilization.86,87 Moreover,
discovered that reduction in numbers and activity of pre-treatment of mice with PTH five weeks prior to
osteoblasts, which produce SDF-1, is one of the major administration of G-CSF, augments mobilization. It is
reasons.60 Decrease in BM SDF-1 levels is correlated yet to be determined whether osteoclasts are involved
with mobilization.28 The reduction in osteoblasts results or not in PTH induced mobilization. Altogether, bone
from stimulation of the sympathetic system during G- remodeling has a strong impact on stem cell function
CSF treatment77 (further discussed in next section). and the degree of both steady-state progenitor cell
Furthermore, osteoblasts isolated from cyclophos- egress and stress-induced mobilization by shaping the
phamide + G-CSF treated mice could expand HSPCs in dynamic BM microenvironment and the evolving stem
vitro at higher rates than osteoblasts from non-treated cell niches.
mice.78 One of the factors promoting HSC self-renewal
is interleukin-10, whose secretion by endosteal Direct and indirect regulation of stem cell egress, recruit-
osteoblasts is upregulated following irradiation induced ment and mobilization by the nervous system
stress.79 Thus, it is suggested that BM niches are under- Early in the 20th century, researchers found a marked
going dynamic changes, during stress induced mobiliza- increase in the number of circulating leukocytes follow-
tion, to allow proliferation and egress of HSPCs. ing injection of adrenaline (epinephrine), a neurotrans-
Following G-CSF administration, osteoclasts activity is mitter produced by the sympathetic system, into
upregulated, explaining why G-CSF mobilized PB healthy human volunteers.88 In rats, epinephrine admin-
donors show reduced bone mass.3,80,81 Osteoclasts istration stimulated the release of leukocytes from the
express both SDF-1 and CXCR4, and SDF-1 is involved spleen, BM and lymphatic organs, contributing signifi-
in recruitment of osteoclast precursors to the bone sur- cantly to leukocytosis.89 Recently, sympathetic system
face and promotes immature osteoclast development induced mobilization of more primitive cells was docu-

| 136 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

mented (reviewed in 90). The BM is highly innervated Concluding remarks


and sympathetic nerve endings localize, especially in The major aim of pre-clinical hematopoietic stem cell
proximity to endothelial cells and the endosteum in the research is to develop improved procedures to harvest
epiphysis and metaphysis, regions known to be and transplant HSPCs, together with improved reconsti-
enriched with stem cell niches, implying indirect control tution capacity for clinical protocols. Today, G-CSF
over HSC function.91 Indeed, immature and primitive administration is the most common clinical procedure
progenitor cells, among other leukocytes, express β2 to mobilize HSPCs intended for transplantation; how-
adrenergic receptors, as well as dopamine receptors.92 ever, the emergence of rapid mobilizing agents, such as
These receptors are upregulated in G-CSF mobilized AMD3100, promises a development of improved clini-
immature human CD34+ cells or upon in vitro treatment cal procedures. Furthermore, they exhibit synergistic
with myeloid cytokines, also suggesting a direct role for mobilizing effects in combination with G-CSF treat-
sympathetic system stimulation in inducing mobiliza- ment.69,98 Apart from developing improved mobilization
tion of HSPCs. β2 adrenergic stimulation by norepi- protocols, improving the capacity of transplanted
nephrine administration in mice resulted in increased HSPCs to engraft can be achieved by increasing cell
numbers of circulating HSPCs, while administration of homing, for example, ex vivo treatments that enhance
a β2 adrenergic antagonist, resulted in their reduced PB CXCR4 signaling.19 Importantly, the recent discoveries
numbers.92 The importance of the nervous system for of dynamic interactions between the microenvironment
the mobilization process was determined by establish- and the resident BM stem cells, which include the bone,
ing mice that display aberrant nerve conduction or that immune and nervous systems, provide a broader per-
underwent catecholaminergic ablation without affect- spective over stem cell regulation and function. Stromal
ing the BM resident HSCs pool during steady state dependent control over SDF-1 availability in the BM,
homeostasis. Strikingly, G-CSF administration in these osteoblasts and osteoclasts activity and both direct and
mice had no effect on HSPCs mobilization.77 Our pre- indirect adrenergic stimuli dynamically regulate steady
liminary data demonstrate that norepinephrine admin- state egress and recruitment of HSPCs to the circulation,
istration stimulates release of SDF-1 from BM stromal as well as maintenance in the BM. Thus, manipulation
cells to the PB, after which progenitors follow, conse- of the brain bone blood triad may assist in developing
quently increasing their numbers in the circulation.72 improved clinical protocols in the future.
Interestingly, this norepinephrine induced mobilization
was selective, since no increase in circulating mature
leukocytes was observed. Recently, it was revealed that References
neuronal stimulation plays a role in steady-state egress 1. Laird DJ, von Andrian UH, Wagers AJ Stem cell trafficking in
of HSPCs and not only during stress situations. Both tissue development, growth, and disease. Cell 2008;132:612-30.
human and murine circulating HSPCs exhibit robust 2. Lapidot T, Petit I. Current understanding of stem cell mobiliza-
tion: the roles of chemokines, proteolytic enzymes, adhesion
daily oscillations.93,94 In fact, β3 adrenergic stimulations molecules, cytokines, and stromal cells. Exp Hematol 2002;
were affected by circadian rhythmic control over norep- 30:973-81.
inephrine release, leading to downregulation of SDF-1 3. Kollet O, Dar A, Lapidot T. The multiple roles of osteoclasts in
host defense: bone remodeling and hematopoietic stem cell
transcription in BM stromal cells. As a result, SDF-1 syn- mobilization. Annu Rev Immunol 2007;25:51-69.
thesis fluctuates, triggering circadian rhythmic oscilla- 4. Velders GA, Fibbe WE. Involvement of proteases in cytokine-
tions of PB circulating progenitor cells.93,94 Furthermore, induced hematopoietic stem cell mobilization. Ann N Y Acad
Sci 2005;1044:60-9.
it seems that G-CSF-induced leukocytosis is also 5. Winkler IG, Levesque JP. Mechanisms of hematopoietic stem
dependent on the timing of G-CSF injection in both cell mobilization: when innate immunity assails the cells that
mice95 and human patients.96 Unexpectedly, adrenergic make blood and bone. Exp Hematol 2006;34:996-1009.
6. Ponomaryov T Peled A, Petit I, Taichman RS, Habler L, Sand-
stimulation suppressed bone-lining osteoblasts, causing bank J et al. Induction of the chemokine stromal-derived fac-
a significant reduction in SDF-1 production,60,77 thereby tor-1 following DNA damage improves human stem cell func-
enabling cell egress. It was found that G-CSF induced tion. J Clin Invest 2000;106:1331-9.
7. Sugiyama T, Kohara H, Noda M, Nagasawa T. Maintenance of
mobilization requires adrenergic signals, since G-CSF the hematopoietic stem cell pool by CXCL12-CXCR4
administration in mice lacking catecholaminergic activi- chemokine signaling in bone marrow stromal cell niches.
ty did not result in osteoblast suppression, SDF-1 down- Immunity 2006;25:977-88.
8. Dar A, Kollet O, Lapidot T. Mutual, reciprocal SDF-1/CXCR4
regulation in the BM or subsequent progenitor cell interactions between hematopoietic and bone marrow stro-
mobilization.77 In addition to suppression of osteoblasts, mal cells regulate human stem cell migration and development
adrenergic stimulation positively affects osteoclasts, in NOD/SCID chimeric mice. Exp Hematol 2006;34:967-75.
9. Zou Y R, Kottmann AH, Kuroda M, Taniuchi I, Littman DR.
leading to loss of bone mass.97 It is yet to be determined Function of the chemokine receptor CXCR4 in haemato-
whether adrenergic stimulation also exerts its effect on poiesis and in cerebellar development. Nature 1998;393:595-9.
HSCs mobilization via osteoclasts. Apart from showing 10. Nagasawa T, Hirota S, Tachibana K, Takakura N, Nishikawa S,
Kitamura Y et al. Defects of B-cell lymphopoiesis and bone-
a direct induction of mobilization by the sympathetic marrow myelopoiesis in mice lacking the CXC chemokine
system, evidence for indirect effects are accumulating, PBSF/SDF-1. Nature 1996;382:635-8.
including circadian control over SDF-1 synthesis and 11. Ara T, Tokoyoda K, Sugiyama T, Egawa T, Kawabata K, Naga-
sawa T et al. Long-term hematopoietic stem cells require stro-
regulation of osteoblasts and osteoclast activity. These mal cell-derived factor-1 for colonizing bone marrow during
new findings broaden the concept of complex microen- ontogeny. Immunity 2003;19:257-67.
vironmental regulation of progenitor cell egress, orches- 12. Christopherson KW, Hangoc G, Mantel CR, Broxmeyer HE.
trating bone remodeling processes, dynamic nervous Modulation of hematopoietic stem cell homing and engraft-
ment by CD26. Science 2004;305:1000-3.
system and immune system interactions with stromal 13. Shirozu M, Nakano T, Inazawa J, Tashiro K, Tada H, Shino-
cells, regulating stem cell function. hara T, Honjo T. Structure and chromosomal localization of
the human stromal cell-derived factor 1 (SDF1) gene.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 137 |
14th Congress of the European Hematology Association

Genomics 1995;28:495-500. ing. Mol Immunol 2008;46:166-71.


14. Peled A, Petit I, Kollet O, Magid M, Ponomaryov T, Byk T et 35. Kollet O, Petit I, Kahn J, Samira S, Dar A, Peled A et al. Human
al. Dependence of human stem cell engraftment and repopula- CD34(+)CXCR4(-) sorted cells harbor intracellular CXCR4,
tion of NOD/SCID mice on CXCR4. Science 1999;283:845-8. which can be functionally expressed and provide NOD/SCID
15. Wright DE, Bowman EP, Wagers AJ, Butcher EC, Weissman IL. repopulation. Blood 2002;100:2778-86.
Hematopoietic stem cells are uniquely selective in their migra- 36. Fukuda S, Broxmeyer HE, Pelus LM. Flt3 ligand and the Flt3
tory response to chemokines. J Exp Med 2002;195:1145-54. receptor regulate hematopoietic cell migration by modulating the
16. Tavor S, Petit I, Porozov S, Avigdor A, Dar A, Leider-Trejo L et SDF-1alpha(CXCL12)/CXCR4 axis. Blood 2005;105:3117-26.
al. CXCR4 regulates migration and development of human 37. Goichberg P, Kalinkovich A, Borodovsky N, Tesio M, Petit I,
acute myelogenous leukemia stem cells in transplanted Nagler A et al. cAMP-induced PKCzeta activation increases
NOD/SCID mice. Cancer Res 2004;64:2817-24. functional CXCR4 expression on human CD34+ hematopoi-
17. Kucia M, Reca R, Miekus K, Wanzeck J, Wojakowski W, etic progenitors. Blood 2006;107:870-9.
Janowska-Wieczorek A et al. Trafficking of normal stem cells 38. Janowska-Wieczorek A, Majka M, Kijowski J, Baj-Krzy-
and metastasis of cancer stem cells involve similar mecha- worzeka M, Reca R, Turner AR et al. Platelet-derived
nisms: pivotal role of the SDF-1-CXCR4 axis. Stem Cells 2005; microparticles bind to hematopoietic stem/progenitor cells
23:879-94. and enhance their engraftment. Blood 2001;98:3143-9.
18. Nie Y, Han YC, Zou YR. CXCR4 is required for the quiescence 39. Peled A, Kollet O, Ponomaryov T, Petit I, Franitza S,
of primitive hematopoietic cells. J Exp Med 2008;205:777-83. Grabovsky V et al. The chemokine SDF-1 activates the inte-
19. Lapidot T, Dar A, Kollet O. How do stem cells find their way grins LFA-1, VLA-4, and VLA-5 on immature human CD34(+)
home? Blood 2005;106:1901-10. cells: role in transendothelial/stromal migration and engraft-
20. Lapidot T, Kollet O. The essential roles of the chemokine SDF- ment of NOD/SCID mice. Blood 2000;95:3289-96.
1 and its receptor CXCR4 in human stem cell homing and 40. Avigdor A, et al. CD44 and hyaluronic acid cooperate with
repopulation of transplanted immune-deficient NOD/SCID SDF-1 in the trafficking of human CD34+ stem/progenitor cells
and NOD/SCID/B2m(null) mice. Leukemia 2002;16:1992- to bone marrow. Blood 2004;103:2981-9.
2003. 41. Peled A, Grabovsky V, Habler L, Sandbank J, Arenzana-
21. Meyerrose TE, Herrbrich P, Hess DA, Nolta JA. Immune-defi- Seisdedos F, Petit I et al. The chemokine SDF-1 stimulates inte-
cient mouse models for analysis of human stem cells. Bio- grin-mediated arrest of CD34+ cells on vascular endothelium
techniques 2003;35:1262-72. under shear flow. J Clin Invest 1999;104:1199-211.
22. Kollet O, Peled A, Byk T, Ben-Hur H, Greiner D, Shultz L, 42. Dar A, Goichberg P, Shinder V, Kalinkovich A, Kollet O,
Lapidot T. Beta2 microglobulin-deficient (B2m(null)) Netzer N et al. Chemokine receptor CXCR4-dependent inter-
NOD/SCID mice are excellent recipients for studying human nalization and resecretion of functional chemokine SDF-1 by
stem cell function. Blood 2000;95:3102-5. bone marrow endothelial and stromal cells. Nat Immunol
23. Ishikawa F, Yasukawa M, Lyons B, Yoshida S, Miyamoto T, 2005;6:1038-46.
Yoshimoto G et al. Development of functional human blood 43. Dick JE. Stem cell concepts renew cancer research. Blood
and immune systems in NOD/SCID/IL2 receptor {gamma} 2008;112:4793-807.
chain(null) mice. Blood 2005;106:1565-73. 44. Kong Y, Yoshida S, Saito Y, Doi T, Nagatoshi Y, Fukata M et al.
24. Larochelle A, Vormoor J, Hanenberg H, Wang JC, Bhatia M, CD34+CD38+CD19+ as well as CD34+CD38-CD19+ cells
Lapidot T et al. Identification of primitive human hematopoi- are leukemia-initiating cells with self-renewal capacity in
etic cells capable of repopulating NOD/SCID mouse bone human B-precursor ALL. Leukemia 2008;22:1207-13.
marrow: implications for gene therapy. Nat Med 1996;2:1329- 45. Sipkins DA, Wei X, Wu JW, Runnels JM, Côté D, Means TK et al.
37. In vivo imaging of specialized bone marrow endothelial
25. Zanjani ED, Flake AW, Almeida-Porada G, Tran N, Papa- microdomains for tumour engraftment. Nature 2005;435:969-73.
yannopoulou, T. Homing of human cells in the fetal sheep 46. Ishikawa F, Yoshida S, Saito Y, Hijikata A, Kitamura H, Tanaka
model: modulation by antibodies activating or inhibiting very S et al. Chemotherapy-resistant human AML stem cells home
late activation antigen-4-dependent function. Blood 1999;94: to and engraft within the bone-marrow endosteal region. Nat
2515-22. Biotechnol 2007;25:1315-21.
26. Angelopoulou MK, et al. A preclinical xenotransplantation ani- 47. Ptasznik A, Urbanowska E, Chinta S, Costa MA, Katz BA,
mal model to assess human hematopoietic stem cell engraft- Stanislaus MA et al. Crosstalk between BCR/ABL oncoprotein
ment. Transfusion. 2004;44:555-66. and CXCR4 signaling through a Src family kinase in human
27. Spiegel A, Kollet O, Peled A, Abel L, Nagler A, Bielorai B et al. leukemia cells. J Exp Med 2002;196;667-78.
Unique SDF-1-induced activation of human precursor-B ALL 48. Jin L, Hope KJ, Zhai Q, Smadja-Joffe F, Dick JE. Targeting of
cells as a result of altered CXCR4 expression and signaling. CD44 eradicates human acute myeloid leukemic stem cells.
Blood. 2004;103:2900-7. Nat Med 2006;12:1167-74.
28. Petit I, Szyper-Kravitz M, Nagler A, Lahav M, Peled A, Habler 49. Kalinkovich A, Tavor S, Avigdor A, Kahn J, Brill A, Petit I et al.
L et al. G-CSF induces stem cell mobilization by decreasing Functional CXCR4-expressing microparticles and SDF-1 corre-
bone marrow SDF-1 and up-regulating CXCR4. Nat Immunol late with circulating acute myelogenous leukemia cells.
2002;3:687-94. Cancer Res 2006;66:11013-20.
29. Kollet O, Spiegel A, Peled A, Petit I, Byk T, Hershkoviz R et al. 50. Spoo AC, Lubbert M, Wierda WG, Burger JA. CXCR4 is a
Rapid and efficient homing of human CD34-CD38(- prognostic marker in acute myelogenous leukemia. Blood
low)CXCR4(+) stem and progenitor cells to the bone marrow 2007;109:786-91.
and spleen of NOD/SCID and NOD/SCID/B2m(null) mice. 51. Christopher MJ, Liu F, Hilton M J, Long F, Link DC. Sup-
Blood. 2001;97:3283-91. pression of CXCL12 production by bone marrow osteoblasts
30. Kahn J, Byk T, Jansson-Sjostrand L, Petit I, Shivtiel S, Nagler A, is a common and critical pathway for cytokine-induced mobi-
Hardan I et al. Overexpression of CXCR4 on human CD34+ lization. Blood 2009.
progenitors increases their proliferation, migration, and 52. Petit I, Goichberg P, Spiegel A, Peled A, Brodie C, Seger R et al.
NOD/SCID repopulation. Blood 2004;103:2942-9. Atypical PKC-zeta regulates SDF-1-mediated migration and
31. Cashman J, Dykstra B, Clark-Lewis I, Eaves A, Eaves C. development of human CD34+ progenitor cells. J Clin Invest
Changes in the proliferative activity of human hematopoietic 2005;115:168-76.
stem cells in NOD/SCID mice and enhancement of their trans- 53. Cancelas JA, Lee AW, Prabhakar R, Stringer KF, Zheng Y,
plantability after in vivo treatment with cell cycle inhibitors. J Williams DA. Rac GTPases differentially integrate signals reg-
Exp Med 2002;196:1141-9. ulating hematopoietic stem cell localization. Nat Med
32. Voermans C, Kooi ML, Rodenhuis S, van der Lelie H, van der 2005;11:886-91.
Schoot CE, Gerritsen WR. In vitro migratory capacity of 54. Kollet O, Shivtiel S, Chen YQ, Suriawinata J, Thung SN,
CD34+ cells is related to hematopoietic recovery after autolo- Dabeva MD et al. HGF, SDF-1, and MMP-9 are involved in
gous stem cell transplantation. Blood 2001;97:799-804. stress-induced human CD34+ stem cell recruitment to the
33. Kimura T, Boehmler AM, Seitz G, Kuçi S, Wiesner T, liver. J Clin Invest 2003;112:160-9.
Brinkmann V et al. The sphingosine 1-phosphate receptor ago- 55. Jin DK, Shido K, Kopp HG, Petit I, Shmelkov SV, Young LM et
nist FTY720 supports CXCR4-dependent migration and bone al. Cytokine-mediated deployment of SDF-1 induces revascu-
marrow homing of human CD34+ progenitor cells. Blood larization through recruitment of CXCR4+ hemangiocytes.
2004;103:4478-86. Nat Med. 2006;12:557-67.
34. Ryser MF, Ugarte F, Lehmann R, Bornhauser M, Brenner S. 56. Massberg S, Schaerli P, Knezevic-Maramica I, Köllnberger M,
S1P(1) overexpression stimulates S1P-dependent chemotaxis Tubo N, Moseman EA et al. Immunosurveillance by
of human CD34+ hematopoietic progenitor cells but strongly hematopoietic progenitor cells trafficking through blood,
inhibits SDF-1/CXCR4-dependent migration and in vivo hom- lymph, and peripheral tissues. Cell 2007;131:994-1008.

| 138 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

57. Kollet O, Dar A, Shivtiel S, Kalinkovich A, Lapid K, Sztainberg 78. Mayack SR, Wagers AJ. Osteolineage niche cells initiate
Y et al. Osteoclasts degrade endosteal components and pro- hematopoietic stem cell mobilization. Blood 2008;112:519-31.
mote mobilization of hematopoietic progenitor cells. Nat Med 79. Kang YJ, Yang SJ, Park G, Cho B, Min CK, Kim TY et al. A
2006;12:657-64. novel function of interleukin-10 promoting self-renewal of
58. Shen H, Cheng T, Olszak I, Garcia-Zepeda E, Lu Z, Herrmann hematopoietic stem cells. Stem Cells 2007;25:1814-22.
S et al. CXCR-4 desensitization is associated with tissue local- 80. Watanabe T, Suzuya H, Onishi T, Kanai S, Kaneko M,
ization of hemopoietic progenitor cells. J Immunol 2001; Watanabe H et al. Effect of granulocyte colony-stimulating
166:5027-33. factor on bone metabolism during peripheral blood stem cell
59. Moore MA, Hattori K, Heissig B, Shieh JH, Dias S, Crystal RG, mobilization. Int J Hematol 2003;77:75-81.
Rafii S. Mobilization of endothelial and hematopoietic stem 81. Takamatsu Y, Simmons PJ, Moore RJ, Morris HA, To LB,
and progenitor cells by adenovector-mediated elevation of Lévesque JP. Osteoclast-mediated bone resorption is stimulat-
serum levels of SDF-1, VEGF, and angiopoietin-1. Ann N Y ed during short-term administration of granulocyte colony-
Acad Sci 2001:938:36-47. stimulating factor but is not responsible for hematopoietic
60. Semerad CL, Christopher MJ, Liu F, Short B, Simmons PJ, progenitor cell mobilization. Blood. 1998;92:3465-73.
Winkler I et al. G-CSF potently inhibits osteoblast activity and 82. Yu X, Huang Y, Collin-Osdoby P, Osdoby P. Stromal cell-
CXCL12 mRNA expression in the bone marrow. Blood 2005; derived factor-1 (SDF-1) recruits osteoclast precursors by
106:3020-7. inducing chemotaxis, matrix metalloproteinase-9 (MMP-9)
61. Heissig B, Hattori K, Dias S, Friedrich M, Ferris B, Hackett NR activity, and collagen transmigration. J Bone Miner Res 2003;
et al. Recruitment of stem and progenitor cells from the bone 18:1404-18.
marrow niche requires MMP-9 mediated release of kit-ligand. 83. Wright LM, Maloney W, Yu X, Kindle L, Collin-Osdoby P,
Cell 2002;109;625-37.
62. Falanga A, Marchetti M, Evangelista V, Manarini S, Oldani E, Osdoby P. Stromal cell-derived factor-1 binding to its che-
Giovanelli S et al. Neutrophil activation and hemostatic mokine receptor CXCR4 on precursor cells promotes the
changes in healthy donors receiving granulocyte colony-stim- chemotactic recruitment, development and survival of human
ulating factor. Blood. 1999;93:2506-14. osteoclasts. Bone. 2005;36:840-53.
63. Levesque JP, Hendy J, Takamatsu Y, Simmons PJ, Bendall LJ. 84. Chiusaroli R, Knobler H, Luxenburg C, Sanjay A, Granot-Attas
Disruption of the CXCR4/CXCL12 chemotactic interaction S, Tiran Z, Miyazaki T et al. Tyrosine phosphatase epsilon is
during hematopoietic stem cell mobilization induced by GCSF a positive regulator of osteoclast function in vitro and in vivo.
or cyclophosphamide. J Clin Invest 2003;111:187-96. Mol Biol Cell 2004;15:234-44.
64. Levesque JP, Hendy J, Winkler IG, Takamatsu Y, Simmons PJ. 85. Shivtiel S, Kollet O, Lapid K, Schajnovitz A, Goichberg P,
Granulocyte colony-stimulating factor induces the release in Kalinkovich A et al. CD45 regulates retention, motility, and
the bone marrow of proteases that cleave c-KIT receptor numbers of hematopoietic progenitors, and affects osteoclast
(CD117) from the surface of hematopoietic progenitor cells. remodeling of metaphyseal trabecules. J Exp Med 2008;205:
Exp Hematol 2003;31:109-17. 2381-95.
65. Levesque JP, Takamatsu Y, Nilsson SK, Haylock D N, Simmons 86. Brunner S, Zaruba MM, Huber B, David R, Vallaster M,
PJ. Vascular cell adhesion molecule-1 (CD106) is cleaved by Assmann G et al. Parathyroid hormone effectively induces
neutrophil proteases in the bone marrow following hemato- mobilization of progenitor cells without depletion of bone
poietic progenitor cell mobilization by granulocyte colony- marrow. Exp Hematol 2008;36:1157-66.
stimulating factor. Blood 2001;98:1289-97. 87. Adams GB, Martin RP, Alley IR, Chabner KT, Cohen KS, Calvi
66. Levesque JP, Liu F, Simmons PJ, Betsuyaku T, Senior RM, Pham LM et al. Therapeutic targeting of a stem cell niche. Nat
C, Link DC. Characterization of hematopoietic progenitor Biotechnol 2007;25:238-43.
mobilization in protease-deficient mice. Blood 2004;104:65-72. 88. Benschop RJ, Rodriguez-Feuerhahn M, Schedlowski M.
67. Vagima Y, Avigdor A, Goichberg P, Shivtiel S, Tesio M, Catecholamine-induced leukocytosis: early observations, cur-
Kalinkovich A et al. MT1-MMP and RECK are involved in rent research, and future directions. Brain Behav Immun 1996;
human CD34+ progenitor cell retention, egress, and mobiliza- 10:77-91.
tion. J Clin Invest 2009;119:492-503. 89. Iversen PO, Stokland A, Rolstad B, Benestad HB. Adrenaline-
68. Christopherson KW, Cooper S, Broxmeyer HE. Cell surface induced leucocytosis: recruitment of blood cells from rat
peptidase CD26/DPPIV mediates G-CSF mobilization of spleen, bone marrow and lymphatics. Eur J Appl Physiol
mouse progenitor cells. Blood 2003;101;4680-6. Occup Physiol 1994;68:219-27.
69. Broxmeyer HE, Orschell CM, Clapp DW, Hangoc G, Cooper 90. Spiegel A, Kalinkovich A, Shivtiel S, Kollet O, Lapidot T. Stem
S, Plett PA et al. Rapid mobilization of murine and human cell regulation via dynamic interactions of the nervous and
hematopoietic stem and progenitor cells with AMD3100, a immune systems with the microenvironment. Cell Stem Cell
CXCR4 antagonist. J Exp Med 2005;201:1307-18. 2008;3:484-92.
70. Larochelle A, Krouse A, Metzger M, Orlic D, Donahue RE, 91. Artico M, Bosco S, Cavallotti C, Agostinelli E, Giuliani-Piccari
Fricker S et al. AMD3100 mobilizes hematopoietic stem cells G, Sciorio S et al. Noradrenergic and cholinergic innervation of
with long-term repopulating capacity in nonhuman primates. the bone marrow. Int J Mol Med. 2002;10:77-80.
Blood 2006;107:3772-8. 92. Spiegel A, Shivtiel S, Kalinkovich A, Ludin A, Netzer N,
71. Liles WC, Rodger E, Broxmeyer HE, Dehner C, Badel K, Goichberg P et al. Catecholaminergic neurotransmitters regu-
Calandra G, Christensen J et al. Augmented mobilization and late migration and repopulation of immature human CD34+
collection of CD34+ hematopoietic cells from normal human cells through Wnt signaling. Nat Immunol 2007;8:1123-31.
volunteers stimulated with granulocyte-colony-stimulating 93. Mendez-Ferrer S, Lucas D, Battista M, Frenette PS. Haema-
factor by single-dose administration of AMD3100, a CXCR4
antagonist. Transfusion 2005;45:295-300. topoietic stem cell release is regulated by circadian oscilla-
72. Dar A, et al. AMD3100 Signals Via the Nervous System, tions. Nature 2008;452:442-7.
Inducing Release to the Circulation of Bone Marrow SDF-1, 94. Lucas D, Battista M, Shi PA, Isola L, Frenette PS. Mobilized
Which Is Crucial for Progenitor Cell Mobilization. Blood hematopoietic stem cell yield depends on species-specific cir-
2006;108:1315. cadian timing. Cell Stem Cell 2008;3:364-6.
73. Porter R L, Calvi LM. Communications between bone cells 95. Ohdo S, Arata N, Furukubo T, Yukawa E, Higuchi S, Nakano
and hematopoietic stem cells. Arch Biochem Biophys 2008; S, Ogawa N et al. Chronopharmacology of granulocyte
473:193-200. colony-stimulating factor in mice. J Pharmacol Exp Ther 1998;
74. Calvi LM, Adams GB, Weibrecht KW, Weber JM, Olson DP, 285:242-6.
Knight MC et al. Osteoblastic cells regulate the haematopoiet- 96. Sato S, Kigawa J, Irie T, Itamochi H, Kanamori Y, Kamazawa
ic stem cell niche. Nature 2003;425:841-6. S et al. Timing of G-CSF administration based on the circadi-
75. Zhang J, Niu C, Ye L, Huang H, He X, Tong WG et al. an rhythm in patients with ovarian cancer. Am J Clin Oncol
Identification of the haematopoietic stem cell niche and con- 2002;25:289-90.
trol of the niche size. Nature 2003;425:836-41. 97. Kondo H, Nifuji A, Takeda S, Ezura Y, Rittling SR, Denhardt
76. Visnjic D, Kalajzic Z, Rowe DW, Katavic V, Lorenzo J, Aguila DT, Nakashima K et al. Unloading induces osteoblastic cell
HL. Hematopoiesis is severely altered in mice with an induced suppression and osteoclastic cell activation to lead to bone loss
osteoblast deficiency. Blood 2004;103:3258-64. via sympathetic nervous system. J Biol Chem 2005;280:
77. Katayama Y, Battista M, Kao WM, Hidalgo A, Peired AJ, 30192-200.
Thomas SA, Frenette PS. Signals from the sympathetic nerv- 98. Pelus, L. M. Peripheral blood stem cell mobilization: new reg-
ous system regulate hematopoietic stem cell egress from bone imens, new cells, where do we stand. Curr Opin Hematol
marrow. Cell 2006;124:407-21. 2008;15:285-92.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 139 |
Hodgkin disease

Nodular lymphocyte-predominant Hodgkin’s lymphoma


in children and adults

J. Landman-Parker A B S T R A C T

Nodular lymphocyte-predominant Hodgkin’s lymphoma is a rare subtype of CD20+ Hodgkin’s lym-


Université Pierre et Marie Curie- phoma characterized by indolent course, young male predominance early stage and overall good prog-
Paris 6/Service d’hématologie et
d’oncologie pédiatrique Hôpital nostic. New understandings in the biology of the lymphocyte predominant cell biology are described.
Armand Trousseau APHP, Paris, The major clinical concerns are related to late effects of treatment described in patients and the
France occurence of high grade lymphoma. We report current strategies in this context and emphasize the
need for clinical studies.

Hematology Education:
the education program for the
annual congress of the European
Hematology Association

2009;3:140-143

odular lymphocyte-predominant ly all cases, and epithelial membrane antigen

N Hodgkin’s lymphoma (NLPHL) is a


rare sub-type of CD20+ Hodgkin’s
lymphoma (HL), which represents 5-10% of
(EMA) in more than 50%. The major differ-
ential diagnosis of NLPHL is LRCHD.3,4 In
contrast to Hodgkin Reed Sternberg cells
all HL cases. NLPHD is characterized by (HRS) of LRCHD, LP cells lack CD30, CD15
indolent course, young male predominance, and EBV genome expression. Surrounding
early stage and overall good prognostic, cells are predominantly small B cells and
although late occurrence of high-grade lym- numerous CD4+/CD57+ T cells. The study
phoma and late deaths related to treatment performed by the European Task Force on
are described in patients. Past two decades lymphocyte predominant Hodgkin lym-
have brought biological and clinical new phoma demonstrated that 20% of the past
insights in NLPHL that lead to a new cases diagnosed as NLPHL were LRCHD in
approach of this rare entity. a review of 521 cases.3 In this study, differen-
tiating between T-cell rich B cell lymphoma
Histopathology and diffuse form of LPHD was uncommon
In 1994, the REAL classification separated and only occurred in 2% of the cases.
NLPHD as a distinct clinicopathologic entity
from other subtypes of HD, which were Biology of lymphocyte predominant cells
subsumed under the term “classical HD” LP cells are clonally related B-cell–derived
(cHD). This new classification was followed malignant cells.5-7 Analysis of isolated L&H
by the World Health Classification (WHO) cells for the variable region of the heavy
classification in 2001.1 Subsequently, an chain of immunoglobulin (IGVH) genes
additional type of cHD with an abundance rearrangements revealed that these cells
of lymphocytes was recognized, named present clonal rearrangements and carry a
“lymphocyte-rich form of classical HD” high load of somatic mutations – a feature
(LRCHL). In the 2008 WHO classification of they share with normal germinal center B
tumors of hematopoietic and lymphoid tis- cells. The Ig VH rearrangements are usually
sues, NLPHL is defined as a monoclonal B functional with mRNA detectable in the LP
cell neoplasm, characterized by a nodular, or cells.7 L&H cells express CD-20 and multiple
a nodular and diffuse proliferation of lym- other B-lymphocyte markers, including ger-
phocytic and histiocytic (L&H) cells (or pop- minal centre B-cell differentiation antigens,
corn cells), now named lymphocyte pre- such as the transcription factors Bcl-6,8 acti-
dominant (LP) cells.2,3 From a morphologic vation-induced cytidine deaminase,9 and
point of view, the lymph node architecture centerine.10,11 Cytogenetic studies and molec-
is totally or partially replaced by a nodular ular analysis of microdissected HRS and LP
infiltrate predominantly consisting of small cells revealed frequent Bcl-6 gene rearrange-
lymphocytes, histiocytes and LP cells, which ments.12 Aberrant somatic hypermutation,
are few (less than 5% of the cells). identified as a mechanism for genomewide
Immunophenotyping of LP cells are positive instability in diffuse large B-cell lymphoma,
for CD 20, CD 79a, CD45 and BCL6 in near- is demonstrated in LP cells.13 Aberrant

| 140 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

somatic hypermutation targets various protooncogenes have often been treated with chemotherapy and radio-
in LP cells, such as PAX5 or Myc, and more recently therapy according to standard classical HL
SOCS1, which may alter their function, thereby con- protocols.16,26-28 Early stage patients treated with radio-
tributing to NLPHL pathogenesis.14 A recent gene therapy alone usually received extended field radiation,
expression study of isolated LP cells indicated that these involving field radiation therapy with 30-36 Gray,29
cells resemble an intermediate developmental stage chemotherapy,30 or more recently, monoclonal antibody
between germinal centre and memory B cells, and therapy using Rituximab or an anti-CD-20 radiolabeled
defines NLPHL as a distinct entity closely related to T antibody the 90Y-ibritumomab tiuxetan for advanced
cell – rich B cell lymphoma and cHL. This study reports stages of LPHL.31
a selection of 49 genes related to the phenotype of LP
cells, among them notably ubiquitin D, which may also Adenectomy, followed by watch-and-wait strategy
contribute to genome instability in NLPHL.15 Occasionally, patients with early stage LPHL have
been treated with surgery alone. In 1983, Miettinen et al.
Clinical presentation reported an overall survival of 93% at five years in 31
Patients typically present with early stage disease, adult LPHL cases of retrospectively diagnosed LPHL,
mainly IA, with peripheral lymph nodes involvement, who remained untreated except for lymph-node exci-
that is, cervical, axillary, and inguinal, rather than sion because the original histology was reported as
mediastinal involvement, which is rarely seen. There benign.32 In 1984, Hansmann et al. reported on 24 cases
is a striking male predominance. This particular clini- of LPHL that, for various reasons, had surgery only. Nine
cal presentation is described in the report of the of them achieved long-term remission.33 These reports
European Task Force on lymphocyte predominant suggested that surgery alone might be an option in early
Hodgkin lymphoma and confirmed by others.3,16,17 In stage LPHL. This strategy was applied in children in two
their data set on adults patients with NLPHL, median studies with limited number of cases.34,35 An retrospec-
age distribution is 35 years and 70% of NLPHL tive update of 58 cases has been recently published by
patients are male, which is three times more than in the European Network Euronet Pediatric Hodgkin lym-
cHD. In children, NLPHL is very rare. In a collaborative phoma.19 The decision to use surgery alone in these
European study, 85% of their 58 patients were male, early stage patients was made on an individual basis by
which is higher than in pediatric cHD, and median age the treating physicians in consultation with the parents
was 11 years.18,19 In the study of the European Project in an effort to limit treatment toxicity whilst maintain-
on 219 patients with confirmed NLPHL, early stage I ing a successful outcome. There were 50 boys out of 58
was common with 53%, 28% stage II, 13% bulky dis- patients, with a median age of 11 years (age range was
ease, 10% B symptoms, 7% mediastinal mass, and 8% 4-17); stage IA n=54, IIA n= 2, IIIA n=2. With a median
had splenic involvement. Of 394 NLPHL patients follow up of 43 months, overall survival is 100% and
report by the German Hodgkin study Group, 63% PFS 57%. Fifty-one out of fifty-eight patients achieved
were in early the favorable stage, 16% were in early complete remission (CR) after surgery, although precise
unfavorable stage, and 21% were in advanced stage of CR definition was not controlled in this retrospective
disease. LPHL patients often presented with normal study. In the CR group, overall PFS is 67% (95% CI
ESR (96%) and normal LDH (84%). A mediastinal 51%; 82%). All cases with residual disease after initial
bulky tumor or extranodal involvement (6%) were surgery relapsed (p=0.003) (7/7). From 18 stage IA
rarely seen.16 Staging procedures in cHD commonly patients with relapse, 11 had a local relapse and 7
used FDG-PET scanning. NLPHL, despite indolent evo- relapsed at stage IIA. One patient (IIIA) presented with
lution, has an avidity for FDG and, therefore, a signif- high-grade B cell lymphoma (NHL) at 10 years of fol-
icant place for staging in NLPHL.20,21 Few cases of low-up. These results suggest that a substantial propor-
NLPHL are reported associated with neurological dis- tion of patients stage IA with one or two involved nodes
eases, such as cerebral angeitis, polyradiculonevritis or might be cured after complete surgery. This approach is
cerebellar ataxia (personal observation).22,23 These obser- currently explored by the Pediatric Euronet on Hodgkin
vations emphasis the role of B lymphocytes in the Lymphoma and the Children Oncology Group in US in
pathogeny of the disease. prospective ongoing studies. In adults, the French GELA
group opened a registry on NLPHL in 1993, and a sus-
Prognosis and treatment tained CR was observed in 50% of the patients given a
Prognosis is favorable with an indolent course of dis- watch-and-wait strategy after surgical excision of the
ease and patients deaths are mostly related to secondary lymph node at NLPHL diagnosis (personal communication).
(treatment related) malignancies or transformation to Consider-ing the limited data published, long-term
aggressive B cell lymphoma. NLPHL patients have an observat ion that is needed and indolent course of the
overall survival range from 83-96%, and a relapse free disease, it is still difficult to recommend a strategy. At
survival dependent on treatment strategy, stage and fol- least two questions remains unsolved: is no further
low up, from 70-88%. Late and multiple relapses are treatment a safe option in patient with stage IA who
more frequent than in cHD. These previous results have have obtained CR after initial adenectomy, and what is
already been reviewed in 2006 and 2008.24,25 the risk of a significant upstaging at relapse and subse-
As clinical and pathological studies have suggested quent transformation into a more aggressive B-cell lym-
that LPHL and cHL differ in pathology and clinical phoma. Nevertheless, considering resection alone in a
behavior, treatment strategies for LPHL patients are very limited disease without associated risk factors and
needed. Clinical questions on the best treatment for complete excision controlled by modern imaging evalu-
these patients are still open. In the past, LPHL patients ation is a realistic option.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 141 |
14th Congress of the European Hematology Association

Low dose chemotherapy in NLPHL cacy of rituximab alone in relapse. The need of treat-
In order to limit morbidity related to treatment, alter- ment for patients in confirmed complete remission after
native chemotherapy to standard cHL treatment has initial surgery for stage I disease is an option, particular-
been proposed in children.36,37 With limited follow up, ly in children, in order to limit treatment related mor-
French and UK pediatricians have reported efficacy of bidity.
low dose chemotherapy alone with vinblastine, low
dose cyclofosphamide and prednisone.38
References
Rituximab in NLPHL
1. Jaffe ES, Harris NL, Stein H, Isaacson PG. Classification of
As almost 100% of LP cells express CD-20, Rituximab lymphoid neoplasms: the microscope as a tool for disease dis-
could be an effective approach, and treatment based on covery. Blood 2008;112:4384-99.
Rituximab alone on a classical schedule has been report- 2. Swerdlow SH, Harris NL, Jaffe ES, Pileri SA, Stein H, Thiele J,
ed in a phase II and retrospective study, but very limit- et al. WHO Classification of Tumors of Haematopoietic and
Lymphoid Tissues: IARC: Lyon 2008.
ed data are available. The German Hodgkin Study 3. Anagnostopoulos I, Hansmann ML, Franssila K, Harris M,
group reported long-term results in relapse NLPHL in a Harris NL, Jaffe ES et al. European Task Force on Lymphoma
phase II study in 2008.39 With 63 months follow-up on project on lymphocyte predominance Hodgkin disease: histo-
logic and immunohistologic analysis of submitted cases
15 confirmed NLPHL patients, PFS is 50% and median reveals 2 types of Hodgkin disease with a nodular growth pat-
time to relapse 33 months. Deaths are related to high- tern and abundant lymphocytes. Blood 2000;96:1889-99.
grade B cell lymphoma, stage IV and second malignan- 4. Shimabukuro-Vornhagen A, Haverkamp H, Engert A, Bal-
leisen L, Majunke P, Heil G et al. Lymphocyte-rich classical
cies. Seven other patients are reported from in a single Hodgkin's lymphoma: clinical presentation and treatment out-
institution, with 100% of CR and three relapses at a come in 100 patients treated within German Hodgkin's Study
median time of 27 months.40 Single cases have been Group trials. J Clin Oncol 2005;23:5739-45.
5. Braeuninger A, Kuppers R, Strickler JG, Wacker HH, Rajewsky
reported with successful results in literature but current- K, Hansmann ML. Hodgkin and Reed-Sternberg cells in lym-
ly no available studies on rituximab alone as first line phocyte predominant Hodgkin disease represent clonal popu-
therapy in LP have been published, although efficacy is lations of germinal center-derived tumor B cells. Proc Natl
probably high but seems limited in the time duration. Acad Sci USA 1997;94:9337-42.
6. Kuppers R, Rajewsky K, Braeuninger A, Hansmann ML. L&H
Rituximab combined with chemotherapy is reported in cells in lymphocyte-predominant Hodgkin's disease. N Engl J
relapsed or newly diagnosed cHL but available data are Med 1998;338:763-64; author reply: 764-5.
very limited.41,42 Combined treatment with ABVD, 7. Marafioti T, Hummel M, Anagnostopoulos I, Foss HD, Falini
B, Delsol G et al. Origin of nodular lymphocyte-predominant
DHAP or gemcitabine could, therefore, be an interesting Hodgkin's disease from a clonal expansion of highly mutated
option for NLPHL, particularly in rare advanced stage or germinal-center B cells. N Engl J Med 1997;337:453-8.
presence of adverse prognostic factors or relapse after 8. Falini B, Bigerna B, Pasqualucci L, Fizzotti M, Martelli MF,
Pileri S et al. Distinctive expression pattern of the BCL-6 pro-
radiation therapy in early stage, but controlled studies tein in nodular lymphocyte predominance Hodgkin's disease.
are recommended. Blood 1996;87:465-71.
9. Greiner A, Tobollik S, Buettner M, Jungnickel B, Herrmann K,
Kremmer E, Niedobitek G. Differential expression of activa-
Multiples relapses and high grade B cell lymphoma tion-induced cytidine deaminase (AID) in nodular lympho-
transformation cyte-predominant and classical Hodgkin lymphoma. J Pathol
The rate of secondary diffuse large B cell lymphoma 2005;205:541-7.
and specifically T-cell rich B cell lymphoma is reported 10. Montes-Moreno S, Roncador G, Maestre L, Martínez N,
Sanchez-Verde L, Camacho FI et al. Gcet1 (centerin), a highly
although, if considering the new lymphoma classifica- restricted marker for a subset of germinal center-derived lym-
tions and the long-term outcome, more informative phomas. Blood. 2008;111:351-8.
data are necessary.43-45 It varies from 3-10% at 10 years, 11. Kuppers R. The biology of Hodgkin's lymphoma. Nat Rev
Cancer 2009;9:15-27.
but may be higher if prolonged observation is docu- 12. Renne C, Martin-Subero JI, Hansmann ML, Siebert R. Molec-
mented. The pattern of relapses is rather local but stages ular cytogenetic analyses of immunoglobulin loci in nodular
or resistance may increase with time, leading to aggres- lymphocyte predominant Hodgkin's lymphoma reveal a
recurrent IGH-BCL6 juxtaposition. J Mol Diagn 2005; 7:352-6.
sive strategies, including high doses chemotherapy with 13. Liso A, Capello D, Marafioti T, Tiacci E, Cerri M, Distler V et
stem cells support or radio labeled CD-20 monoclonal al. Aberrant somatic hypermutation in tumor cells of nodular-
treatment. This pattern of relapses emphasizes the need lymphocyte-predominant and classic Hodgkin lymphoma.
Blood 2006;108:1013-20.
of repeated biopsy to reconsider histology, as mixed or 14. Mottok A, Renne C, Willenbrock K, Hansmann ML, Braun-
T-cell rich B cell lymphoma may occur, and be misdiag- inger A. Somatic hypermutation of SOCS1 in lymphocyte-
nosed. predominant Hodgkin lymphoma is accompanied by high
JAK2 expression and activation of STAT6. Blood 2007;110:
3387-90.
Conclusion 15. Brune V, Tiacci E, Pfeil I, Döring C, Eckerle S, van Noesel CJ et
NLPHL is a rare entity related to germinal centers al. Origin and pathogenesis of nodular lymphocyte-predomi-
transformation and classical Hodgkin disease. A con- nant Hodgkin lymphoma as revealed by global gene expres-
sion analysis. J Exp Med 2008;205:2251-68.
firmed diagnosis by hematopathologist in the case of 16. Nogova L, Reineke T, Brillant C, Sieniawski M, Rüdiger T,
CD-20 HL is strongly recommended considering the Josting A et al. Lymphocyte-predominant and classical
risk of misdiagnosis with LRCHD. Treatment strategy Hodgkin's lymphoma: a comprehensive analysis from the
German Hodgkin Study Group. J Clin Oncol 2008;26:434-9.
adapted to early stage of the disease, as well as indolent 17. Bodis S, Kraus MD, Pinkus G, Silver B, Kadin ME, Canellos GP
course and risk of subsequent malignancies is the major et al. Clinical presentation and outcome in lymphocyte-pre-
challenge. Treatment according to early stage cHL is dominant Hodgkin's disease J Clin Oncol 1997;15:3060-6.
18. Karayalcin G, Behm FG, Gieser PW, Kung F, Weiner M, Tebbi
associated with good results but the role of Rituximab CK et al. Lymphocyte predominant Hodgkin disease: clinico-
associated with chemotherapy with low rate of adverse pathologic features and results of treatment--the Pediatric
effects has to be investigated considering the high effi- Oncology Group experience. Med Pediatr Oncol 1997;29:519-
25.

| 142 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

19. Mauz-Korholz C, Gorde-Grosjean S, Hasenclever D, Shankar Y, Bertrand Y et al. Lympho-cyte-predominant Hodgkin's lym-
A, Dörffel W, Wallace WH et al. Resection alone in 58 children phoma in children: therapeutic abstention after initial lymph
with limited stage, lymphocyte-predominant Hodgkin lym- node resection a Study of the French Society of Pediatric
phoma-experience from the European network group on pedi- Oncology J Clin Oncol 2003;21:2948-52.
atric Hodgkin lymphoma. Cancer 2007;110:179-85. 35. Murphy SB, Morgan ER, Katzenstein HM, Kletzel M. Results
20. Holalkere N, Takvorian R, Blake M, Toomey C, Michaelson J, of little or no treatment for lymphocyte-predominant Hodgkin
Rahemtullah A, et al. Intensity of FDG Uptake on PET Scan disease in children and adolescents. J Pediatr Hematol Oncol
Varies by Histologic Subtype of Hodgkin Lymphoma. Blood 2003;25:684-7.
2007;110:4393. 36. Donaldson SS, Link MP, Weinstein HJ, Rai SN, Brain S, Billett
21. Ansquer C, Hervouet T, Devillers A, de Guibert S, Gastinne T, AL et al. Final results of a prospective clinical trial with VAMP
Le Gouill S et al. 18-F FDG-PET in the staging of lymphocyte- and low-dose involved-field radiation for children with low-
predominant Hodgkin's disease. Haematologica 2008;93:128- risk Hodgkin's disease. J Clin Oncol. 2007;25:332-7.
31. 37. Landman-Parker J, Pacquement H, Leblanc T, Habrand JL,
22. Roggerone S, Traverse-Glehen A, Derex L, Honnorat J, Berger Terrier-Lacombe MJ, Bertrand Y et al. Localized childhood
F, Salles G et al. Recurrent cerebral venous thrombosis reveal- Hodgkin's disease: response-adapted chemotherapy with
ing paraneoplastic angiitis in Hodgkin's lymphoma. J Neuro- etoposide, bleomycin, vinblastine, and prednisone before low-
oncol 2008;89:195-8.
23. Ingber S, Buckstein R. Paraneoplastic lumbosacral axonal dose radiation therapy-results of the French Society of
polyradiculopathy preceding the diagnosis of nodular lympho- Pediatric Oncology Study MDH90. J Clin Oncol 2000;18:1500-
cyte predominant Hodgkin lymphoma: a case report. Leuk 7.
Lymphoma 2008;49:2009-11. 38. Shankar A, Hall G, Gorde-Grosjean S, Mauz Koerholz C,
24. Nogova L, Rudiger T, Engert A. Biology, clinical course and Korholz D, Wallace WH, et al. Treatment of Children &
management of nodular lymphocyte-predominant hodgkin Adolescents with Early Stage Nodular Lymphocyte Pre-
lymphoma. Hematology Am Soc Hematol Educ Program dominant Hodgkin Lymphoma with a Low Intensity Short
2006; 266-72. Duration Chemotherapy Regimen [CVP] - on Behalf of the
25. Fanale MA, Younes A. Nodular lymphocyte predominant EuroNet-PHL Group. Blood 2006;108:2471.
Hodgkin's lymphoma. Cancer Treat Res 2008;142:367-81. 39. Schulz H, Rehwald U, Morschhauser F, Elter T, Driessen C,
26. Wilder RB, Schlembach PJ, Jones D, Chronowski GM, Ha CS, Rüdiger T et al. Rituximab in relapsed lymphocyte-predomi-
Younes A et al. European Organization for Research and nant Hodgkin lymphoma: long-term results of a phase 2 trial
Treatment of Cancer and Groupe d'Etude des Lymphomes de by the German Hodgkin Lymphoma Study Group (GHSG).
l'Adulte very favorable and favorable, lymphocyte-predomi- Blood 2008;111:109-11.
nant Hodgkin disease. Cancer 2002;94:1731-8. 40. Azim HA Jr, Pruneri G, Cocorocchio E, Cinieri S, Raviele PR,
27. Wirth A, Yuen K, Barton M, Roos D, Gogna K, Pratt G et al. Bassi S et al. Rituximab in lymphocyte-predominant Hodgkin
Long-term outcome after radiotherapy alone for lymphocyte- disease. Oncology 2009; 76:26-9.
predominant Hodgkin lymphoma: a retrospective multicenter 41. Wedgwood AR, Fayad LE, McLaughlin P, Hagemeister FB,
study of the Australasian Radiation Oncology Lymphoma Romaguera J, Rodriguez MA, Samaniego F, Younes A.
Group. Cancer 2005;104:1221-9. Rituximab + ABVD Improves Event-Free Survival (EFS) in
28. Schlembach PJ, Wilder RB, Jones D, Ha CS, Fayad LE, Younes Patients with Classical Hodgkin Lymphoma in All
A et al. Radiotherapy alone for lymphocyte-predominant International Prognostic Score (IPS) Groups and in Patients
Hodgkin's disease. Cancer J 2002;8:377-83. Who Have PET Positive Disease after 2-3 Cycles of Therapy.
29. Nogova L, Reineke T, Eich HT, Josting A, Müller-Hermelink Blood 2007;110:215.
HK, Wingbermühle K et al. Extended field radiotherapy, com- 42. Younes A, Pro B, Goy A, Romaguera JE, Hagemeister FB,
bined modality treatment or involved field radiotherapy for McLaughlin P. Gemcitabine Plus Rituximab Therapy of
patients with stage IA lymphocyte-predominant Hodgkin's Patients with Relapsed and Refractory Classical Hodgkin
lymphoma: a retrospective analysis from the German Lymphoma. Blood 2005;106:1498.
Hodgkin Study Group (GHSG). Ann Oncol 2005; 16:1683-7. 43. Achten R, Verhoef G, De Wolf-Peeters C. The importance of
30. Hall GW, Katzilakis N, Pinkerton CR, Nicolin G, Ashley S,
McCarthy K et al. Outcome of children with nodular lympho- follow-up biopsies of clinically suspicious lymphadenopathy
cyte predominant Hodgkin lymphoma - a Children's Cancer in nodular lymphocyte predominant Hodgkin's lymphoma.
and Leukaemia Group report. Br J Haematol 2007;138:761-8. Haematologica 2003;88:1431-2.
31. Zojer N, Mirzaei S, Ludwig H. Successful treatment of a 44. Feugier P, Labouyrie E, Djeridane M, Jenabian A, Dubruille V,
patient with lymphocyte-predominant Hodgkin's lymphoma Berthou C et al. Comparison of initial characteristics and long-
with yttrium-90-ibritumomab tiuxetan. Eur J Haematol 2008; term outcome of patients with lymphocyte-predominant
81:322-4. Hodgkin lymphoma and classical Hodgkin lymphoma at clin-
32. Miettinen M, Franssila KO, Saxen E. Hodgkin's disease, lym- ical stages IA and IIA prospectively treated by brief anthracy-
phocytic predominance nodular. Increased risk for subsequent cline-based chemotherapies plus extended high-dose irradia-
non-Hodgkin's lymphomas. Cancer 1983;51:2293-300. tion. Blood 2004;104:2675-81.
33. Hansmann ML, Zwingers T, Boske A, Loffler H, Lennert K. 45. Bennett MH, MacLennan KA, Vaughan Hudson G, Vaughan
Clinical features of nodular paragranuloma (Hodgkin's dis- Hudson B. Non-Hodgkin's lymphoma arising in patients treat-
ease, lymphocyte predominance type, nodular). J Cancer Res ed for Hodgkin's disease in the BNLI: a 20-year experience.
Clin Oncol 1984;108:321-30. British National Lymphoma Investigation. Ann Oncol 1991;(2
34. Pellegrino B, Terrier-Lacombe MJ, Oberlin O, Leblanc T, Perel Suppl 2):83-92.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 143 |
Hodgkin disease

The prognostic role of positron emission tomography


scan in Hodgkin’s lymphoma

A. Gallamini A B S T R A C T

Several prognostic factors have been proposed for Hodgkin lymphoma (HL); however, their predic-
Hematology Department and BMT tive value in defining treatment outcome has been questioned. Positron emission tomography using
Unit, Cuneo, Italy
[18F]-fluoro-2-deoxy-D-glucose (FDG-PET) is an important tool for staging, treatment response assess-
ment, post-treatment prognosis definition of residual mass, and prediction of autologous stem cell
transplantation outcomes in HL. Moreover, FDG-PET has shown high accuracy and overall predictive
Hematology Education: value in therapy response forecasts when performed very early during standard ABVD chemotherapy.
the education program for the The treatment of HL has shown an improving efficacy during the last 30 years, and durable remissions
annual congress of the European
are now achieved in more than 85% of the patients; however, some concerns still exist about the tox-
Hematology Association
icity and risk of over treatment. Therefore, new prognostic factors, such as FDG-PET, seem to play a
2009;3:144-150 role in HL in defining a risk-adapted therapy for the single patient. Many trials are currently under-
way, both in limited and advanced HL, and are aimed at exploring the role of a PET-driven, risk-adapt-
ed therapy. However, many questions remain unanswered, such as the standardization criteria for early
PET scanning and interpretation, the role of interim-PET during the course of therapies with different
intensity, and during follow-up.

The clinical usefulness of prognostic factors in but the cumulative 5-year risk of secondary
Hodgkin lymphoma acute leukemia or myelodysplastic syn-
The ABVD polychemotherapy regimen drome was 2.2% vs. 0.4% for the standard
has long been considered the golden stan- treatment. Moreover, nearly two-thirds of
dard for Hodgkin lymphoma (HL) therapy. the patients are exposed to the risks of over
The recommended schedule is three or four treatment.
courses, plus Involved-Field (IF) radiothera- In limited-stage HL, the long-term seque-
py in limited-stage, and six courses plus lae of radiotherapy, such as cardiovascular
“consolidation” radiotherapy for bulky or events or secondary neoplasm arising inside
residual disease in advanced stage patients.1 the fields of radiation, are responsible for
However, 20-30% of the advanced-stage most deaths in long-term survivors for HL.9
patients fail to achieve durable remissions, For these reasons, the search for new
and ultimately die of recurrent/resistant parameters able to predict treatment out-
lymphoma.2 come has been spurred on in the last few
In the nineties, more aggressive regimens years, and prognostic markers regained
were proposed, such as escalated BEACOPP. great popularity. They play now a central
Their efficacy has been shown superior to role in planning a new therapeutic strategy
ABVD in at least three randomized studies, aimed to single out patients with a very
both in terms of progression-free and overall poor prognosis, candidates for an aggres-
survival.3-5 Diehl and colleagues for the sive front-line therapy, and to spare toxici-
German Hodgkin Lymphoma Study Group ty to the majority of the patients.10 [18F]-flu-
demonstrated a superiority of escalated oro-2-deoxy-D-glucose positron emission
BEACOPP versus the COPP/ABVD regimen, tomography (FDG-PET) seems to play an
with an 8-year progression-free survival of ideal role in this perspective. The FDG
85% for the former, and 69% for the latter. uptake by the tumor is a consequence of an
At the beginning of the millennium, the intact metabolic activity and reflects the
same authors claimed that there was no viability of the neoplastic cells. Therefore,
longer any need for prognostic factors, since in principle, PET scanning could give a visu-
they had lost their predictive power, as treat- al update of the state of the lymphoma at
ment was successfully adapted to disease every time point during treatment, and pos-
burden.6 Consequently, some clinicians con- sibly identify patients with different
sider this regimen to be the standard therapy chemosensitivity and treatment response.
for advanced-stage HL. In this perspective, two major points are
These brilliant results, however, have currently being investigated in several cur-
been partially hampered by an increased rent clinical trials:
risk of secondary malignancies, with a 1. In the early-stages, could involved-field
higher risk benefit ratio for a non-trivial radiotherapy be delivered only to
subset of the patients.7 The most common- patients with a poor prognosis, as defined
ly reported side effects were short-term by a positive interim-PET after two
hematological toxicities and amenorrhoea,8 ABVD courses?

| 144 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

2. In advanced-stages, could an aggressive poly- definition on CR, PR, stable or progressive disease by
chemotherapy be reserved for patients with a very the International Workshop Criteria on Treatment
poor prognosis, as defined by a positive-interim-PET Response in Lymphomas.15 So far, two experiences
after two ABVD courses? have been published on the clinical consequences of
A possible answer to question 1 could arise from the the application of these new criteria in assessing tumor
H10 study of GELA-EORTC-IIL and from MRC response to treatment. The concept of CRU has been
RAPID study; for question 2, the RATHL study, the abolished, and patients defined in CR or CRU at the
GHSH HD18 and the GITIL HD0607 will try to give an end of treatment had an identical outcome; patients in
answer (see the paragraph on open issues). PR had a progression free survival similar to the ones
in stable or progressive disease.25,26
The role of FDG-PET in post-treatment evaluation Despite the good response to therapy, treatment of
FDG-PET was first introduced in the management of HL results in residual mass in up to 64-80% of the
lymphomas in the early 1990s. It is now recognised as patients, as shown by conventional restaging modali-
an important tool for staging and treatment response ties.22,27,28 After Jerusalem’s preliminary study,29 many
assessment in Hodgkin and non-Hodgkin lym- reports focused on the role of FDG-PET for post-treat-
phomas.14,15 At the end of therapy, the persistent FDG ment evaluation of a residual mass in lymphoma.
uptake is considered a proof of survival of viable neo- Quite recently, Terasawa systematically reviewed all
plastic cells.16,17 the studies published so far on this issue, and reported
Moving from these concepts, several reports in the a sensitivity for HL patients ranging between 43% and
literature have demonstrated the high sensitivity and 100%, and a specificity ranging between 67% and
specificity of FDG-PET in tumor response assessment. 100%30 (Figure 3).
In a recent accurate meta-analysis from 13 studies on With these premises, FDG-PET has been proposed
408 HL patients, upon exclusion of other studies not to drive the decision of delivering consolidation radio-
fulfilling the minimal requirements for review (full ring therapy in cases of single residual mass persistence at
of CT-PET, adequate follow-up, definition of the refer- the end of lymphoma treatment, and its role has been
ence test), Zijlstra and colleagues were able to demon- proven essential.31,32 In the German Cooperative
strate a pooled sensitivity and specificity of PET in Hodgkin Lymphoma Study Group’s experience, PET-
defining treatment outcomes of 84% and 90%, respec- driven decisions of delivering consolidation radiother-
tively18 (Figure 2). apy at the end of treatment for Hodgkin’s lymphoma
Thanks to these results, in 2007 FDG-PET was pro- has allowed a significant reduction of its use from 70%
posed as an essential tool in defining treatment of the patients in HD nine trials to a 12% in the HD in
response and it has been integrated in the previous fifteen trials.32

Figure 1. Sensitivity and specificity of FDG-PET for treatments response (adapted from Zijlstra, by permission)

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 145 |
14th Congress of the European Hematology Association

Figure 2. Sensitivity and specificity for treatment response in case of residual mass (adapted from Terasawa, by permission).

Early interim positron emission tomography during conven- Proposed Criteria for PET-2 interpretation after ABVD chemotherapy.
tional Hodgkin lymphoma treatment
More recently, PET has emerged as a useful prognos- Negative
tic tool, which may predict treatment outcome when 0 no uptake
performed very early during therapy (early PET). A 1 uptake ≤ mediastinum
number of studies have focused on its prognostic role 2 uptake ≥ mediastinum but ≤ liver
after two33-35 or three36 courses of standard ABVD
Positive
chemotherapy, and some studies have also suggested
3 uptake > liver in some sites even if uptake ≤ liver or mediastinum
that PET can predict treatment response as early as at other sites
after one course of chemotherapy.37-40 Overall, in HL 4 uptake > liver in over 90% of sites or development of new uptake
the positive and negative predictive values have consistent with progressive disease
ranged between 60-80% and 80-90%, respectively,
while in non-Hodgkin lymphoma (NHL) the positive
Figure 3. Proposed semi-quantitative criteria for interim-
predictive value was lower (40-60%). In all studies, the PET interpretation in HL (M Juweid and J Radford, person-
authors confirmed the prognostic role of early PET in al communication, 2007).
predicting treatment outcome and concluded that it is
useful and reliable for assessment of the treatment
response.
Nearly 80% of the HL patients show a complete nor- tive early PET, but only 60% of those patients eventual-
malization of the PET scan after two courses of ly experienced treatment failure.45 The same group ret-
ABVD.34,35 This phenomenon, that has been called rospectively re-evaluated the same cohort of patients
“metabolic CR”,41,42 can be explained by the peculiar using SUV analysis and reported that when using an
architecture and organization of the neoplastic tissue, SUV maximum reduction of 66% as the cut-off value,
where only a few, scattered neoplastic cells (accounting the positive predictive value shifted from 43-83%.46
for less than 1% of the total cellular population) are sur- In HL, after two courses of ABVD, early PET is posi-
rounded by a population of non-neoplastic mononu- tive and minimally positive in 20% and 9-10% of the
clear bystander cells. The latter are probably responsible patients, respectively.34-36,47 The patients with minimal
for the immortalisation of Hodgkin and Reed-Sternberg residual FDG uptake (MRU) on early PET are character-
(HRS) cells by stimulating cytokine production by CD ized by a very good prognosis36,47 and it has been pro-
4+ lymphoid cells recruited by TARC (thymus and acti- posed that a PET scan with a faint FDG uptake consis-
vation-related chemokines: paracrine loop), or by trig- tent with MRU be considered as negative.36,48 Since the
gering cytokine release by the HRS cells able to induce majority of patients displaying MRU on early PET still
survival of the same cells (autocrine loop).43 In cases pre- have an excellent prognosis, this phenomenon could be
senting with bulky lesions at diagnosis, a negative early related to non-specific activity in macrophages and
PET is often associated with a persisting bulky lesion of other inflammatory cells infiltrating the tumor as a
more or less unchanged size. The explanation might be response to the chemotherapy rather than to a chemo-
that the chemotherapy switches off the production of resistance.17 Hutchings and Mikhael first introduced the
chemokines by the activated lymphoid cells, as term of MRU as a low-grade uptake of FDG (just above
described for TARC. The latter can be measured in the background uptake) in a focus within an area of previ-
serum of HL patients and its level correlated to the qual- ous documented disease, reported by the nuclear medi-
ity of treatment response; for patients in CR, the levels cine physicians as unlikely to represent active malignan-
are much lower than in patients with stable or progress- cy.36,49 Later, in a joint Italian and Danish study,48 MRU
ing disease.44 The situation is different in NHL. In a was defined as a faint, non-focal FDG uptake in a site of
French study, a substantial fraction of diffuse, large B- involvement at diagnosis, with an SUV lower or equal
cell lymphoma (DLCL) patients (40%) showed a posi- to the one recorded in the mediastinal blood pool struc-

| 146 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

Table 1. Prognostic meaning of FDG-PET prior to ASCT.

Author N. Histology Indication Timing PPV PNV 2-y PFS for PET resp 2-y PFS for PET non-resp
(ref) patients PET

Jabbour64 68 HL Rel/Pro Pre ASCT 72% 76% 76% 27%


Schot 65 117 NHL/HL Rel/Pro Pre ASCT 73% 67% 73% 25%
Spaepen66 60 NHL/HL Rel/Pro PreASCT 87% 90% 100% 24%
Filmont67 20 NHL/HL Rel/Pro PreASCT 91% 87% 87% 7%
Svoboda68 50 NHL/HL Rel/Pro; 3 line PreASCT 94% 46% 50% 12%
Crocchiolo69 53 NHL/HL Rel/Pro PreASCT 43% 72% 90% 55%

tures (MBPS). A similar semi-quantitative approach has Early interim-positron emission tomography during BEA-
recently been proposed for interim PET interpretation in COPP or equivalent therapies
the context of an International Protocol for advanced- Very recently, during the Seventh International
stage HL (Table 1). Symposium on Hodgkin Lymphoma and the Fiftieth
The prognostic role of early chemotherapy sensitive- Annual ASH meeting, preliminary results were present-
ness assessment seems independent and more predic- ed on the prognostic role of early PET after two courses
tive of treatment outcome than IPS. In the afore men- of BEACOPPesc in advanced HL.55-57 All these studies are
tioned Italian-Danish study early PET proved to be the characterized by a relatively good outcome of patients
only statistically significant predictor of treatment out- with a positive early interim scan. In particular, in both
come, whatever the IPS score in the single patient. From studies, the number of false positive studies and the low
this point of view, interim FDG-PET seems to represent positive predictive value proved to be a problem; 10%,
a step forward in the direction of a single-patient, risk- 20% and 83%, and 60%, 46% and 17%, respectively.
adapted therapeutic strategy.48 It is noteworthy that, in Despite the relatively good outcome of the interim-PET
the same study patients with a negative early PET positive patients, possibly due to a late “salvage” effect
showed a 2-year progression-free survival (PFS) equal or of escalated BEACOPP; the higher proportion of posi-
even better than the one for the entire cohort of patients tive scans (30%), as compared to the one observed after
treated with more aggressive (and more toxic) regimens ABVD (20%) points to the low specificity of PET during
such as BEACOPPesc. BEACOPP. The mean time between the last administra-
tion of BEACOPPesc and early PET ranged between 11
The ideal time for interim positron emission tomography and 17 days.55-56 Currently, we do not know the optimal
during therapy timing for early PET after BEACOPPesc, but one could
Two questions concerning the ideal time for interim- speculate that the risk of false-positive studies due to a
PET scanning are still unsettled: non-specific inflammatory effect is greater early after
BEACOPPesc than after ABVD, due to the more effi-
1. What is the ideal time for interim-PET scan after
cient tumor kill. Other possible explanations include the
chemotherapy administration?
frequent use of growth factors (G-CSF) and the
2. What is the ideal number of chemotherapy cycles
increased rate of infections and use of antibiotics after
before interim-PET scan?
BEACOPPesc compared with after ABVD.
As far the point (1) is concerned, Spaepen has shown We do not know yet the prognostic role of early PET
in mice that FDG uptake by neoplastic cells and reactive after BEACOPPesc, since until now it has not been eval-
inflammatory macrophages was minimal 14 days after uated in prospective studies where no therapeutic
chemotherapy administration.17 In a review of the pub- changes were based on early PET results. The ongoing
lished experience of interim PET early during treatment, HD-18 study of the German Hodgkin Study Group will
Kasamon concluded that the optimal time for perform- specifically address this issue. Now, I do not recom-
ing interim PET during chemotherapy ranged between mend early PET in routine clinical practice as a basis for
7 and 14 days after chemotherapy.50 early therapeutic adaptation in patients treated with
As far as point (2) the answer could depend on the BEACOPPesc outside a clinical trial.
aggressiveness of the tumor and the efficacy of the
chemotherapy. In a preliminary experience Iagaru, in a Prognostic value of FDG-PET before high-dose chemother-
small cohort of 20 HL and NHL patients found, by ∆ apy/autologous stem-cell transplantation
SUV variation analysis, found that both 18F-FDG High-dose chemotherapy (HDT) followed by autolo-
PET/CT scans obtained at two and four cycles correlat- gous stem-cell transplantation (ASCT) has been success-
ed well (correlation coefficient 0.98 and 0.80, respective- fully used for relapsing or primary resistant HL.58-61 The
ly) with end-treatment response. Furthermore, scans only significant prognostic factors were the duration of
performed at two cycles did not differ significantly from response to first-line chemotherapy and the status of
scans performed at four cycles in terms of ∆ SUV from the disease at transplant or, in other words, the
baseline to post-treatment.51 In HL, most experiences chemosensitivity of the tumor. In a review of the pub-
have been performed after two courses of chemothera- lished literature, evidence points to the high overall pre-
py, although promising preliminary reports have stress- dictive value of pre-transplant FDG-PET. Some reports
es the high predictive value after one cycle.38-40) compose a mixture of NHL and HL, while others deal

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 147 |
14th Congress of the European Hematology Association

exclusively with HL. In general, the predictive value is uptake; (ii) a semi-quantitative score with reference
higher in HL in respect to NHL and the positive predic- organs, such as mediastinum and liver could be used to
tive value (PPV) is higher than the negative predictive better distinguish between unspecific residual uptake
value (NPV) (see Table 3). In particular the PPV ranges and persisting positive lesions; (iii) a baseline study is
between 91% and 43%, while the PNV between 90% strongly recommended, since early PET interpretation is
and 46%. These wide-range fluctuations are mainly due based on a site-to-site comparison of FDG uptake both
to the presence of a wide array of NHL subtypes that, as before and after chemotherapy; (iv) in the case of visual
already known, display different FDG avidity.62,63 assessment, the inter-observer concordance was no
higher than 50% in absence of defined rules for PET
The role of FDG-PET in follow-up scan interpretation; (v) the quantitative approach seems
FDG-PET has been proposed in the follow-up of HL superior in terms of predictive power on treatment out-
patients. In a preliminary experience of follow-up per- come in NHL; (vi) the quantitative approach needs a
formed with serial FDG-PET scans every 6 months in 36 more stringent standardization since the SUV value
HL patients in CR after ABVD therapy, Jerusalem found could be influenced by different variables; (vii) the rela-
six false-positive studies, and no false-negative studies tive SUVMAX reduction of an interim PET with respect to
out of 119 performed scans. In five other positive stud- the baseline scan was equivalent to the relative SUVMEAN
ies, FDG-PET preceded the relapse after a median of 3.5 reduction or to the absolute SUVMAX calculation, but less
(1-9) months.70 In a series of 57 HL patients who under- operator-dependent and less influenced by the different
went a systematic follow-up with FDG PET every 6 PET scanners.52-54
months for the first two years, and then once in a year
for the following three years, Zinzani et al found a rela- Open Issues: ongoing trials
tively high percentage of patients in which the relapse Limited-stage Hodgkin lymphoma
suspicion was based on PET results: (21/57: 37%). In all In the EORTC/GELA/IIL H10 trial, patients with lim-
cases, the positive scans were recorded at mediastinal ited stage IA-IIB are stratified in two risk classes and
site. All underwent histological confirmation by surgical evaluated for treatment response after two ABVD
biopsy. HL relapse was found in only 10/21 (48%) courses; thereafter, they are randomised to receive a
patients. More than three quarters of the positive scans PET-independent treatment, a PET-driven treatment
were found 6 and 12 months after treatment end.71 In with chemotherapy intensification, or de-escalation in
another experience where PET was performed every 6 PET positive and PET-negative patients, respectively.11 A
months during follow-up in a cohort of 45 HL patients similar project is ongoing in the United Kingdom; a ran-
in CR after six ABVD courses, Zuckerman found the domised trial (RAPID) comparing no further treatment
rate of false positive was 9/45 (20%).72 Based on these with involved field RT following three cycles ABVD
results the use of FDG-PET in the follow-up of HL and a “negative” (-ve) PET scan in limited-stage HL
patients cannot be routinely recommended and further patients.12
studies are warranted prior to any definite conclusion.73
Advanced-stage Hodgkin lymphoma
Controversial problems
Several trials are running worldwide aimed at assess-
Many issues remain controversial, and important ing the overall clinical impact of a strategy of early ther-
questions regarding PET scanning standardization and apy intensification (with BEACOPP or IGEV followed
interpretation are still unanswered: (i) it is still unclear by ASCT) in patients with an interim PET positive after
whether a qualitative approach using visual assessment two courses of ABVD (RATHL from BMC Nordic
only is preferable or whether a semi-quantitative evalu- Group and GISL), trial HD 18 from GHSG, Trial HD
ation by SUV analysis should be applied; (ii) the differ- 0607 from GITIL (Gruppo Italiano Terapie Innovative
ent FDG avidity and uptake patterns before and during nei Linfomi). Preliminary results have been presented
chemotherapy between Hodgkin and non-Hodgkin
from the Italian GITIL group; interim-PET scans per-
lymphomas (NHL) prevents the use of common criteria
formed after two ABVD courses in a cohort of 136
for PET interpretation for all lymphomas; (iii) it is still
patients with stage IIB-IVB HL – 19/136 (14%) were
uncertain if the prognostic value of early PET varies
found to be positive with a semi-quantitative score sys-
between different therapeutic regimens; (iv) it is still
tem. They were treated with BEACOPP (a escalated + 4
unknown whether a strategy based on early chemother-
baseline courses) and after a mean follow-up of 14
apy intensification only in interim-PET positive patients
months, 15/19 were in continuous CR.13
could turn out in improving the overall outcome of the
entire cohort of HL patients.
The role of early PET in HL has been extensively eval-
uated and a number of answers to the above-mentioned References
issues have been proposed. Recently a number of “posi- 1. Canellos GP, Niedzwiecki D. Long term follow-up of
tion papers” have been published on the issue of stan- Hodgkin’s disease trial. N Engl J Med 2002;346,1417-8.
dardization criteria for interim-PET interpretation, 2. Bonadonna G, Viviani S., Bonfante V, Gianni AM, Valagussa P
et al. Survival in Hodgkin’s disease patients – Report of 25
focusing on the dilemma of qualitative or quantitative years of experience at the Milan cancer Institute. Eur J Cancer
assessment. The authors concluded that: (i) there is no 2005;41:998-1006.
need for quantitative assessment in HL, and visual 3. Diehl V, Franklin J, Pfreundschuh M, Lathan B, Paulus U,
Hasenclever D et al. Standard and increased-dose BEACOPP
assessment gives the best results, provided that some chemotherapy compared with COPP-ABVD for advanced
“liberal” criteria for a negative scan interpretation are Hodgkin's disease. N Engl J Med 2003;348:2386-95.
accepted, including the cases of minimal residual 4. Federico M, Luminari S, Dell’Olio M, et al. ABVD vs. COPPE-
BVCAD (CEC) vs. BEACOPP for the initial treatment of

| 148 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

patients with advanced Hodgkin Lymphoma (HL). Preliminary of residual mediastinal Hodgkin disease. Blood 2001;98:2930-
results of HD2000 GISL trial. Haematologica 2007;92:34. 34
5. Gianni AM, Rambaldi A, Zinzani PL, et al. Comparable 3-year 24. Zinzani PL, Magagnoli M, Chierichetti F, Zompatori M,
outcome following ABVD or BEACOPP first-line chemothera- Garraffa G, Bendandi M, et al. The role of positron emission
py, plus pre-planned high-dose salvage, in advanced Hodgkin tomography (PET) in the management of lymphoma patients.
lymphoma (HL): A randomized trial of the Michelangelo, Ann Oncol 1999;10:1181-4.
GITIL and IIL cooperative groups. 2008 ASCO Annual 25. Brepoels L, Stroobants S, De Wever W, Spaepen K, Vanden-
Meeting Proceedings. J Clin Orthod 2008;26:8506. berghe P, Thomas J et al. Hodgkin lymphoma: Response
6. Hasenclever D. The disappearance of prognostic factors in assessment by Revised International Workshop Criteria. Leuk
Hodgkin’s disease. Ann Oncol 2002;13:75-8. Lymphoma 2007;48:1539-47.
7. Josting A, Wiedenmann S, Franklin J, May M, Sieber M, Wolf 26. Dupuis J, Itti E, Rahmouni A, Hemery F, Gisselbrecht C, Lin C
J et al. Secondary myeloid leukemia and myelodysplastic syn- et al. Response assessment after an inductive CHOP or CHOP-
dromes in patients treated for Hodgkin’s disease: Aa report like regimen with or without rituximab in 103 patients with
from the German Hodgkin’s Lymphoma Study Group. J Clin diffuse large B-cell lymphoma: integrating 18fluorodeoxyglu-
Oncol 2003;21:3440-6. cose positron emission tomography to the International
8. Behringer K, Breuer K, Reineke T, May M, Nogova L, Klimm Workshop Criteria. Ann Oncol [epub Dec 11, 2008].
B et al. Secondary amenorrhea after Hodgkin’s lymphoma is 27. Radford JA, Cowan RA, Flanagan M, Dunn G, Crowther D,
influenced by age at treatment, stage of disease, chemothera- Johnson RJ, Eddleston B. The significance of residual mediasti-
py regimen, and the use of oral contraceptives during therapy: nal abnormality on the chest radiograph following treatment
a report from the German Hodgkin’s Lymphoma Study for Hodgkin’s disease. J Clin Oncol 1988;6: 940-6.
Group. J Clin Oncol 2005;23: 7555-64. 28. De Vita VT, Mauch PM, Harris NL. Dodgkin’s disease. In
9. Brusamolino E, Baio A, Orlandi E. Long-term events in adult DeVita VT Jr, Helmann S, Rosemberg SA, editors. Cancer.
patients with Clinical Stage IA-IIA Nonbulky Hodgkin’s Principles & Practice of Oncology, 5th ed. Philadelphia (PA):
Lymphoma Treated with Four Cycles of Doxorubicin, Lippincott-Raven; 1997:2242-83.
Bleomycin, Vinblastine, and Dacarbazine and Adjuvant 29. Jerusalem G, Beguin Y, Fassotte MF, Najjar F, Paulus P, Rigo P,
Radiotherapy: A Single-Institution 15-Year Follow-up. Clin Fillet G. Whole-body positron emission tomography using
Cancer Res 2006;12:6487-93. 18F-Fluorodeoxyglucose for posttreatment evaluation in
10. Kasamon YL, Wahl RL. FDG PET and risk-adapted therapy in Hodgkin’s disease and non-Hodgkin’s lymphoma has higher
Hodgkin’s and non-Hodgkin’s lymphoma. Curr Opin Oncol diagnostic and prognostic value than classical computed
2008;20:206-19. tomography scan imaging. Blood 1999;94:429-33.
11. Fitoussi O, Brice P, Hirt S. EARLY Interim FDG-PET Scan in 30. Terasawa T, Nihashi T, Hotta T, Nagai H. 18F-FDG PET for
Localised Hodgkin Lymphoma: Evaluation of Practice in 5 posttherapy assessment of Hodgkin’s disease and aggressive
French Centers. Blood. 2008;112,4935. non-Hodgkin’s lymphoma: a systematic review. J Nucl Med
12. Radford J, O’Doherty M, Barrington S, et al. Results of the 2nd 2008;49:13-21.
Planned Interim Analysis of the RAPID Trial (involved field 31. Savage K, Connors JM, Wilson D, et al. FDG-PET Guided
radiotherapy versus no further treatment) in Patients with Consolidative Radiotherapy in Patients with Advanced Stage
Clinical Stages 1A and 2A Hodgkin Lymphoma and a ‘nega- Hodgkin Lymphoma with Residual Abnormalities on Post-
tive’ FDG-PET Scan after 3 Cycles ABVD. Blood 2008;112: Chemotherapy CT scans. Blood 2007;110:213.
369. 32. Kobe C, Dietlein M, Franklin J, Markova J, Lohri A, Amthauer
13. Gallamini A, Rambaldi A, Patti C, et al. BEACOPP Chemo- H et al. Positron emission tomography has a high negative pre-
therapy Is Able to Induce Durable Complete Remission in dictive value for progression or early relapse for patients with
Poor-Prognosis Hodgkin’s Lymphoma Patients With a Positive residual disease after first-line chemotherapy in advanced-
Interim PET After 2 ABVD Cycles. Blood 2008;112:2594. stage Hodgkin lymphoma. Blood. 2008;112:3989-94.
14. Juweid ME, Stroobants S, Hoekstra OS, Mottaghy FM, 33. Torizuka T, Nakamura F, Kanno T, Futatsubashi M, Yoshikawa
Dietlein M, Guermazi A et al. Use of positron emission E, Okada H et al. Early therapy monitoring with FDG-PET in
tomography for response assessment of lymphoma: consen- aggressive non-Hodgkin's lymphoma and Hodgkin’s lym-
sus of the Imaging Subcommittee of International Harmon- phoma. Eur J Nucl Med Mol Imaging. 2004;31:22-8.
ization Project in Lymphoma. J Clin Oncol 2007;25: 571-8. 34. Hutchings M, Loft A, Hansen M, Pedersen LM, Buhl T,
15. Cheson BD, Pfistner B, Juweid ME, Gascoyne RD, Specht L, Jurlander J et al. FDG-PET after two cycles of chemotherapy
Horning SJ et al. Revised response criteria for malignant lym- predicts treatment failure and progression-free survival in
phoma. J Clin Oncol. 2007;25:579-86. Hodgkin lymphoma. Blood 2006;107:52-9.
16. Pauwels EK, McCready VR, Stoot JH, van Deurzen DF. The 35. Gallamini A, Rigacci L, Merli F, Nassi L, Bosi A, Capodanno I
mechanism of accumulation of tumour-localising radiophar- et al. The predictive value of positron emission tomography
maceuticals. Eur J Nucl Med 1998;25:277-305. scanning performed after two courses of standard therapy on
17. Spaepen K, Stroobants S, Dupont P, Bormans G, Balzarini J, treatment outcome in advanced stage Hodgkin's disease.
Verhoef G et al. [18F]FDG PET monitoring of tumour response Haematologica 2006;91:475-81.
to chemotherapy: does [18F]FDG uptake correlate with the 36. Hutchings M, Mikhaeel NG, Fields PA, Nunan T, Timothy AR.
viable tumour cell fraction? Eur J Nucl Med Mol Imaging Prognostic value of interim FDG-PET after two or three cycles
2003;30:682-8. of chemotherapy in Hodgkin lymphoma. Ann Oncol 2005; 16:
18. Zijlstra JM, Lindauer-Van der werf G, Hoekstra O, Hooft L, 1160-8.
Riphagen II, Huijgens PC. 18F-fluoro-deoxyglucose positron 37. Romer W, Hanauske AR, Ziegler S, Thödtmann R, Weber W,
emission tomography for post-treatment evaluation of malig- Fuchs C et al. Positron emission tomography in non-Hodgkin’s
nant lymphoma: a systematic review. Haematologica 2006; lymphoma: assessment of chemotherapy with fluorode-
91:522-9. oxyglucose. Blood 1998;91:4464-71.
19. de Wit M, Bohuslavizki KH, Buchert, Bumann D, Clausen M, 38. Kostakoglu L, Coleman M, Leonard JP, Kuji I, Zoe H,
Hossfeld DK et al. 18FDG-PET following treatment as valid Goldsmith SJ. PET predicts prognosis after 1 cycle of chemo-
predictor for disease-free survival in Hodgkin’s lymphoma. therapy in aggressive lymphoma and Hodgkin’s disease. J Nucl
Ann Onco 2001;12:29-37. Med. 2002;43:1018-27.
20. Hueltenschmidt B, Sautter-Bihl ML, Lang O, Maul FD, Fischer 39. Kostakoglu L, Goldsmith SJ, Leonard JP, Christos P, Furman
J, Mergenthaler HG, Bihl H. Whole body positron emission RR, Atasever T et al. FDG-PET After 1 Cycle of Therapy
tomography in the treatment of Hodgkin disease. Cancer Predicts Outcome in Diffuse Large Cell Lymphoma and
2001;91:302-10. Classic Hodgkin Disease. Cancer. 2006;107,2678-87.
21. Mikhaeel NG, Timothy AR, O’Doherty MJ, Hain S, Maisey 40. Rigacci L, Castagnoli A, Carpaneto, A Carrai V, Vaggelli L,
MN. 18-FDG-PET as a prognostic indicator in the treatment of Matteini M et al. Can (18)F-FDG PET after first cycle
aggressive non-Hodgkin’s lymphoma-comparison with CT. chemotherapy predict the efficacy of therapy in Hodgkin’s
Leuk Lymphoma 2000;39:543-53. disease? Haematologica 2002;87:ELT24.
22. Naumann R, Vaic A, Beuthien-Baumann B, Bredow J, Kropp J, 41. MacManus MP, Seymour J, Hicks RJ. Overview of early
Kittner T et al. Prognostic value of positron emission tomogra- response assessment in lymphoma with FDG-PET. Cancer
phy in the evaluation of post-treatment residual mass in Imaging 2007;7:10-8.
patients with Hodgkin’s disease and non-Hodgkin’s lym- 42. Kostakoglu L. Early prediction of response to therapy: the clin-
phoma. Br J Haematol 2001;115: 793-800. ical implications in Hodgkin’s and non-Hodgkin’s lymphoma.
23. Weihrauch MR, Re D, Scheidhauer K, Ansen S, Ansén S, Eur J Nucl Med Mol Imaging 2008;35:1413-20.
Dietlein M, Bischoff S et al. Thoracic positron emission 43. Skinnider BF, Mak TW. The role of cytokines in classical
tomography using 18F-fluorodeoxyglucose for the evaluation Hodgkin lymphoma. Blood 2002;99:4283-97.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 149 |
14th Congress of the European Hematology Association

44. Weihrauch MR, Manzke O, Beyer M, Haverkamp H, Diehl V, grafted after a first relapse. Ann Oncol 2005;16:625-33.
Bohlen H et al. Elevated Serum Levels of CC Thymus and 60. Tarella C, Cuttica A, Vitolo U, Liberati M, Di Nicola M,
Activation-Related Chemokine (TARC) in Primary Hodgkin’s Cortelazzo S et al. High-Dose Sequential Chemotherapy and
Disease: Potential for a Prognostic Factor. Cancer Res 2005; Peripheral Blood Progenitor Cell Autografting in Patients with
65:5516-9. Refractory and/or Recurrent Hodgkin Lymphoma. Cancer
45. Haioun C, Itti E, Rahmouni A, Brice P, Rain JD, Belhadj K et al. 2003;97:748-59.
[18F]fluoro-2-deoxy-D-glucose positron emission tomography 61. Kuruvilla J, Nagy T, Pintilie M, Tsang R, Keating A, Crump M.
(FDG-PET) in aggressive lymphoma: an early prognostic tool Similar Response Rates and Superior Early Progression-Free
for predicting patient outcome. Blood 2005;106:1376-81. Survival with Gemcitabine, Dexamethasone, and Cisplatin
46. Lin C, Itti E, Haioun C, Petegnief Y, Luciani A, Dupuis J et al. Salvage Therapy Compared with Carmustine, Etoposide,
Early 18F-FDG PET for prediction of prognosis in patients Cytarabine, and Melphalan Salvage Therapy Prior to Auto-
with diffuse large B-cell lymphoma: SUV-based assessment logous Stem Cell Transplantation for Recurrent or Refractory
versus visual analysis. J Nucl Med 2007;48:1626-32. Hodgkin Lymphoma. Cancer 2006;106:353-60.
47. Zinzani PL, Tani M, Fanti S, Alinari L, Musuraca G, Marchi E 62. Elstrom R, Guan L, Baker G, Nakhoda K, Vergilio JA, Zhuang
et al. Early positron emission tomography (PET) restaging: a H et al. Utility of FDG-PET scanning in lymphoma byWHO
predictive final response in Hodgkin’s disease patients. Ann classification. Blood. 2003;101:3875-6.
Oncol 2006;17:1296-1300. 63. Tsukamoto N, Kojima M, Hasegawa M, Oriuchi N,
48. Gallamini A, Hutchings M, Rigacci L, Specht L, Merli F, Matsushima T, Yokohama A et al. The Usefulness of 18F-flu-
Hansen M et al. Early interim 2-[18F]fluoro-2-deoxy-D-glu- orodeoxyglucose Positron Emission Tomography (18F-FDG-
cose positron emission tomography is prognostically superior PET) and a Comparison of 18F-FDG-PET With 67Gallium
to international prognostic score in advanced-stage Hodgkin’s Scintigraphy in the Evaluation of Lymphoma. Cancer 2007;
lymphoma: a report from a joint Italian-Danish study. J Clin 110:652-9.
Oncol 2007;25:3746-52. 64. Jabbour E, Hosing C, Ayers G, Nunez R, Anderlini P, Pro B et
49. Mikhaeel NG, Hutchings M, Fields PA, O’Doherty MJ, Timo- al. Pretransplant positive positron emission tomography/galli-
thy AR. FDG-PET after two to three cycles of chemotherapy um scans predict poor outcome in patients with recur-
predicts progression-free and overall survival in high-grade rent/refractory Hodgkin lymphoma. Cancer 2007;109: 2481-9.
non-Hodgkin lymphoma. Ann Oncol 2005 65. Schot BW, Zijlstra JM, Sluiter WJ, van Imhoff GW, Pruim J,
50. Kasamon YL, Jones RJ, Wahl RL. Integrating PET and PET/CT Vaalburg W, Vellenga E. Early FDG-PET assessment in combi-
into the Risk-Adapted Therapy of Lymphoma. J Nucl Med nation with clinical risk scores determines prognosis in recur-
2007;48:19S–27S. ring lymphoma. Blood 2007;109:486-91.
51. Iagaru A, Wang Y, Carina Mari C, Quon A, Goris ML, Horning 66. Spaepen K, Stroobants S, Dupont P, Vandenberghe P, Maertens
S, Gambhir SS. 18F-FDG-PET/CT evaluation of response to J, Bormans G et al. Prognostic value of pretransplantation
treatment in lymphoma: when is the optimal time for the first positron emission tomography using fluorine 18-fluoro-
re-evaluation scan? Hell J Nucl Med 2008;11(3):153-6. deoxyglucose in patients with aggressive lymphoma treated
52. Weber WA. 18F-FDG PET in Non-Hodgkin’s Lymphoma: with high-dose chemotherapy and stem cell transplantation.
Qualitative or Quantitative? J Nucl Med 2007;48:1580-2. Blood 2003;102:53-9.
53. Gallamini A, Hutchings M, Avigdor A, Polliack A. Early inter- 67. Filmont JE, Czernin J, Yap C, Silverman DH, Quon A, Phelps
im PET scan in Hodgkin lymphoma: where do we stand? Leuk ME, Emmanouilides C. Value of F-18 Fluorodeoxyglucose
Lymphoma 2008;45:659-62. Positron Emission Tomography for Predicting the Clinical
54. Zijlstra JM, Comans EF, van Lingen A, Hoekstra OS, Gundy Outcome of Patients With Aggressive Lymphoma Prior to and
CM, Willem Coebergh J, Bongers V. FDG PET in lymphoma: After Autologous Stem-Cell Transplantation. Chest 2003;124;
The need for standardization of interpretation. An observer 608-13.
variation study. Nucl Med Commun 2007;28:798-803. 68. Svoboda J, Andreadis C, Elstrom R, Chong EA, Downs LH,
55. Gallamini A, Viviani S, Bonfante V, et al. Early interim FDG- Berkowitz A et al. Prognostic value of FDG-PET scan imaging
PET during intensified BEACOPP therapy shows a lower in lymphoma patients undergoing autologous stem cell trans-
predicitve value than during conventional ABVD chemothera- plantation. Bone Marrow Transplant 2006;38:211-6.
py. Haematologica 2007;92:71. 69. Crocchiolo R, Canevari C, Assanelli A, Lunghi F, Tassara M,
56. Avigdor A, Bulvik S, Dann EJ, et al. Combined escBEACOPP- Stanghellini ML et al. Pre-transplant 18FDG-PET predicts out-
ABVD therapy for advanced Hodgkin's lymphoma patients come in lymphoma patients treated with high-dose sequential
with high IPS score: An effective regimen and low positive chemotherapy followed by autologous stem cell transplanta-
predictive value of early FDG-PET/CT. Blood. 2007;110,2319. tion. Leuk Lymphoma 2008;49:727-33.
57. Dann E, Tamir A, Epelbaum R, et al. Not All Positive Interim 70. Jerusalem G, Beguin Y, Fassotte MF, Belhocine T, Hustinx R,
PET/CT Scans Are Reliably Predictive for Progression Free Rigo P, Fillet G et al. Early detection of relapse by whole-body
Survival in Hodgkin Lymphoma (HL) Patients. A Proposed positron emission tomography in the follow-up of patients
Model for a Functional Scoring Scale. Blood 2008;112,1452. with Hodgkin’s disease. Ann Oncol 2003;14:123-30.
58. Schmitz N, Pfistner B, Sextro M, Sieber M, Carella AM, 71. Zinzani PL, Tani M, Trisolini R, Fanti S, Stefoni V, Alifano M
Haenel M et al. Aggressive conventional chemotherapy com- et al. Histological verification of positive positron emission
pared with high-dose chemotherapy with autologous tomography findings in the follow-up of patients with medi-
haemopoietic stem-cell transplantation for relapsed chemo- astinal lymphoma. Haematologica 2007;92:771-7.
sensitive Hodgkin’s disease: a randomised trial. Lancet 2002; 72. Zuckerman D, Lacasce A, Jacobsen E, et al. High False Positive
359:2065-71. Rate with the Use of CT and FDG-PET in Post-Remission
59. Sureda A, Constans M, Iriondo A, Arranz R, Caballero MD, Surveillance for Hodgkin Lymphoma. Blood 2007;110:2327.
Vidal MJ et al. Prognostic factors affecting long-term outcome 73. Seam P, Juweid ME, Cheson BD. The role of FDG-PET scans
after stem cell transplantation in Hodgkin’s lymphoma auto- in patients with lymphoma. Blood 2007;110:3507-16.

| 150 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Hodgkin disease

Recent advances in the treatment


of early-stage Hodgkin’s lymphoma

D.A. Eichenauer A B S T R A C T
A. Engert
The standard of care for patients with early-stage Hodgkin lymphoma (HL) has changed from wide-
field radiotherapy alone to combined modality strategies consisting of a brief chemotherapy followed
First Department of Internal
Medicine, University Hospital by involved-field radiotherapy (IF-RT). Since the vast majority of patients treated with this approach
Cologne, Cologne, Germany, and achieve permanent remissions and long-term survival rates beyond 90%, cure rates can hardly be fur-
German Hodgkin Study Group ther improved. Therefore, the major goal of current clinical research is to minimize therapy-related
acute and late toxicities, such as secondary malignancies, infertility, heart failure and pulmonary dys-
function, without compromising treatment efficacy. Different strategies are being pursued. A possible
reduction of chemotherapy, as well as the minimization of radiation fields from IF to involved-node
Hematology Education:
the education program for the (IN) and the value of risk-adapted strategies based on PET imaging is currently being investigated in
annual congress of the European clinical trials worldwide.
Hematology Association

2009;3:151-154

odgkin lymphoma (HL) is a malig- bined modality strategies were introduced

H nant disease of the lymphatic system


with an incidence of 2-3/100.000/
year in developed countries. Generally, HL
and randomized trials demonstrated them to
be more effective than radiotherapy alone.5,6
Thus, there is currently no indication for the
occurs in all age groups but young adults are use of radiotherapy alone in HL, except for
most often affected.1 Two major subtypes the treatment of patients with stage IA
can be distinguished, classical Hodgkin lym- NLPHL, without risk factors. This small sub-
phoma (cHL), which accounts for about set of patients, analyzed by the German
95% of all cases, and nodular lymphocyte Hodgkin Study Group (GHSG) and the
predominant Hodgkin lymphoma (NLPHL), European Organisation for Research and
representing about 5% of cases.2 Treatment of Cancer (EORTC), revealed no
Due to substantial treatment improve- significant outcome differences between
ments over the past decades, HL has patients treated with 30 Gy involved-field
changed from an incurable disease to one of radiotherapy (IF-RT) and patients treated
the adult malignancies with the best cure with extended-field RT (EF-RT) or combined
rates; a fact resulting in a steadily growing modality strategies.7-8 Consequentially, the
number of long-term survivors. Since these least toxic approach, 30 Gy IF-RT, was
survivors often suffer from treatment-associ- adopted as the standard of care.
ated late toxicities, such as secondary malig-
nancies, infertility, heart failure and pul- Combined modality approaches for early favor-
monary dysfunction, reducing the frequency able-stage Hodgkin lymphoma
of long-term sequelae without compromis- Various randomized trials from different
ing treatment efficacy has become the major study groups showed the superiority of
challenge of today’s clinical research in HL.3 combined modality treatment over radio-
This text aims at giving an overview on the therapy alone in patients with early favor-
changes in the treatment of early-stage HL in able HL.
recent years and of current developments to The HD7 trial, conducted by the GHSG,
improve therapy further. compared 30 Gy EF-RT plus 10 Gy to the IF
alone with two cycles of ABVD chemother-
Radiotherapy alone apy followed by the same radiotherapy.
Early favorable and early unfavorable- Tumor control was superior in patients treat-
stage HL are distinguished by the presence ed with the combined modality approach,
or absence of certain risk factors (Table 1). resulting in a significantly better 7-year free-
For many years, wide-field radiotherapy dom from treatment failure (FFTF) (88% vs.
alone was considered the treatment of 67%, p<0.001).6 The ability to reduce radia-
choice for early favorable-stage HL. By this tion fields to IF without compromising treat-
approach, many patients achieved a com- ment efficacy has been shown in a number
plete remission (CR) but the relapse rate was of trials, so that combined modality treat-
high and overall survival (OS) was not satis- ment, including a brief chemotherapy, such
fying.4 To improve treatment results, com- as ABVD followed by IF-RT has become the

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 151 |
14th Congress of the European Hematology Association

accepted standard therapy in early favorable HL.9,10 patients with early unfavorable HL within the GHSG.14
In trials already closed but not fully analyzed, the pos- Another new approach in the treatment of early unfa-
sible reduction of either chemotherapy or radiotherapy vorable HL is the PVAG14 regimen. The protocol con-
was evaluated. The purpose of the GHSG HD10 trial sisting of prednisone, vinblastine, doxorubicin and gem-
was to investigate whether it is possible to reduce the citabine administered in a 14-day cycle is currently
dose of IF-RT from 30 Gy to 20 Gy after chemotherapy under investigation in a GHSG phase II trial.
consisting of two or four cycles of ABVD. The interim
analyses performed so far revealed no significant differ- Chemotherapy alone
ences between 20 Gy or 30 Gy IF-RT, and between two Since it is well known that a number of severe long-
or four cycles of ABVD chemotherapy.11 The final analy- term sequelae, such as secondary malignancies, pul-
sis of the trial is expected later in 2009. monary fibrosis or hypothyroidism can potentially be
The GHSG follow-up trial HD13 aimed at decreasing caused by radiotherapy, several groups initiated trials
toxicity from the ABVD backbone by reducing the evaluating the impact of radiotherapy after adequate
number of drugs given. Patients were randomized chemotherapy.
between two cycles of ABVD, ABV, AVD or AV In a trial conducted by the National Cancer Institute
chemotherapy followed by 30 Gy IF-RT. A safety analy- (NCI) of Canada and the Eastern Cooperative Oncology
sis performed in June 2006 detected a fourfold increase Group (ECOG), 399 patients with limited-stage HL
of events in the ABV and the AV arm, respectively, com- (nonbulky clinical stages IA and IIA) were enrolled.
pared to the ABVD standard arm. This increase could They were randomly assigned to either receive four to
not be explained by chance variation. Hence, the six cycles of ABVD chemotherapy alone or ABVD com-
ABV/AV arms were closed. The question whether AVD bined with STNI. With a median follow-up of 4.2 years,
is equivalent to ABVD will be answered in future analy- freedom from disease progression was superior in
ses of the trial. patients receiving combined modality treatment (93%
in the combined modality vs. 87% in the chemotherapy
Combined modality approaches for early unfavorable-stage alone arm), indicating a better tumor control by addi-
Hodgkin lymphoma tional radiation after chemotherapy. There were no sig-
Patients with early unfavorable HL are commonly nificant differences in terms of EFS and OS (88% vs.
treated with combined modality approaches. However, 86% and 94% vs. 96%, respectively).15 However, longer
the optimal chemotherapy regimen and the number of observation and documentation of patients included in
chemotherapy cycles needed was a matter of debate. this trial is required to draw final conclusions because
In the EORTC/Groupe d’etude des Lymphomes de follow-up is still too short to assess, particularly the
l’adulte (GELA) H8U study, 996 patients were treated radiotherapy-related long-term side effects.
with either six cycles of MOPP-ABV plus IF-RT, four Another study conducted at the Memorial Sloan-
cycles of MOPP-ABV plus IF-RT or four cycles of Kettering Cancer Center (MSKCC) led to similar results.
MOPP-ABV plus subtotal nodal irradiation (STNI). All A total of 152 patients with nonbulky clinical stages IA,
groups had similar 5-year event-free survival (EFS) (84% IB, IIA, IIB and IIIA were prospectively randomized to
vs. 88% vs. 87%) and 10-year OS estimates (88% vs. either six cycles of ABVD chemotherapy alone or six
85% vs. 84%). Thus, four cycles of chemotherapy fol- cycles of ABVD followed by radiotherapy. Although no
lowed by IF-RT was proposed as the standard treatment significant differences in terms of CR duration, freedom
for patients with early unfavorable HL.12 from progression (FFP) and OS could be detected, a ten-
In the EORTC/GELA follow-up H9U study, patients dency towards a superior outcome in patients receiving
were randomized into three treatment arms consisting combined modality treatment was observed. At 60
of four cycles of ABVD, six cycles of ABVD or four months, 91% of patients receiving combined modality
cycles of BEACOPPbaseline, each followed by 30 Gy IF- treatment and 87% of patients receiving chemotherapy
RT. An interim analysis at a median follow-up of four alone were still in CR (after 94% of patients in both
years showed no significant differences regarding EFS
and OS between the treatment arms while increased
toxicity was observed with BEACOPPbaseline.13
In the GHSG HD14 trial, patients were randomly Table 1. EORTC and GHSG risk factors defining treatment
groups clinical stage I/II Hodgkin lymphoma.
assigned to either two cycles of BEACOPPescalated fol-
lowed by two cycles of ABVD chemotherapy or to four Trial group Risk factors Treatment groups
cycles of ABVD chemotherapy. All patients received 30
Gy IF-RT. In 2008, a planned interim analysis of the EORTC/GELA 1) Large MM Early favourable:
trial, including data from 1010 patients at a median 2) Age ≥50 CS I or II without any RF
observation of three years was performed. This analysis 3) B symptoms and ESR ≥30 mm/h Early unfavourable:
revealed a significant superiority of the treatment with or no B symptoms and ESR ≥50 mm/h CS I or II without any RF
two cycles of BEACOPPescalated, followed by two 4) ≥4 involved sites
cycles of ABVD plus 30 Gy IF-RT compared to four
cycles of ABVD plus 30 Gy IF-RT in terms of FFTF (96% GHSG 1) Large MM Early favourable:
vs. 90%). Therefore, the study arm consisting of four 2) Extranodal disease CS I or II without any RF
cycles of ABVD followed by 30 Gy IF-RT was closed 3) B symptoms and ESR ≥30 mm/h Early unfavourable:
and the regimen consisting of two cycles of or no B symptoms and ESR ≥50 mm/h CS I and IIA with any RF;
4) ≥3 involved sites CS IIB with RF 3 and/or 4
BEACOPPescalated plus two cycles of ABVD followed
by 30 Gy IF-RT was adopted as new standard for MM: mediastinal mass; ESR: erythrocyte sedimentation rate; CS: clinical stage; RF: risk
factor.

| 152 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

arms had initially achieved CR). FFP and OS rates were


86% and 97%, respectively, for patients treated with
chemotherapy plus radiotherapy compared to 81% and
90%, respectively, for patients who received chemo-
therapy alone.16
In the EORTC/GELA H9F study for patients with
early favorable HL, all patients received six cycles of
EBVP chemotherapy. Patients with CR/CRu were then
randomized between 36 Gy IF-RT, 20 Gy IF-RT and no
RT. The trial could not be completed according to plan
since the no RT arm had to be closed prematurely due
to an excessive rate of events.13
The results of these trials indicate that omission of
radiotherapy after an appropriate chemotherapy might
only be possible in carefully selected patients. Currently, Figure 1. HD16 trial for early favorable-stage Hodgkin lym-
one of the most relevant questions is to identify and phoma.
possibly further reduce toxicity in these patients. Since
response assessment based on CT scans alone does not
seem adequate to distinguish between those patients
who may be sufficiently treated with chemotherapy (Figure 1). In a large British trial, which started recruit-
alone and those who require additional radiotherapy or ment in 2003, patients with a negative PET after three
even more intensive treatment, ongoing trials focus on cycles of ABVD chemotherapy are randomized
the predictive value and possible stratification based on between either no further treatment or IF-RT. Patients
the results of interim fluoro-deoxy-glucose positron with positive PET receive a fourth cycle of ABVD
emission tomography (FDG-PET). chemotherapy followed by IF-RT.21

Response adapted therapy based on FDG-PET Conclusions


As indicated by Danish and Italian groups, early inter- The introduction of combined modality approaches
im FDG-PET might be a good predictor for treatment has led to cure rates beyond 90% in patients with early-
failure in HL patients.17-18 Furthermore, the results of the stage HL. Since these cure rates can hardly be further
GHSG HD15 trial for patients with advanced HL indi- improved, current trials aim at evaluating whether a
cate that FDG-PET is a valuable tool for the decision response-adapted stratification of treatment based on
whether patients with residual lymphoma after the risk profile of the individual patient is possible and
chemotherapy should be irradiated or not. Patients with practicable. At the moment, FDG-PET is considered the
residual lymphoma larger than 2.5 cm had PET scans. most promising tool to distinguish between those
Patients with negative PET were not irradiated; patients patients who might receive a reduced treatment with-
with positive PET were irradiated. The negative prog- out worsening the prognosis and those patients who
nostic value (NPV), defined as the portion of patients might benefit from an intensified treatment. However,
without progression, relapse or radiotherapy within 12 results from large randomized trials addressing this
months, was 94%.19 Based on these findings, various issue are pending. Therefore, a brief chemotherapy fol-
ongoing trials evaluate whether treatment of patients lowed by IF-RT remains the standard treatment for
with early-stage HL might be stratified based on the patients with early-stage HL.
results of an interim PET scan. In the EORTC/GELA
H10 trial, the treatment in the standard arm consists of
three cycles of ABVD chemotherapy for patients with References
favorable risk profile and four cycles of ABVD
1. Engert A, Dreyling M. Hodgkin's lymphoma: ESMO clinical
chemotherapy for patients with unfavorable risk pro- recommendations for diagnosis, treatment and follow-up.
file. Chemotherapy is followed by involved-node RT Ann Oncol 2008;19:ii65-6.
(IN-RT), which means a reduction of the radiation fields 2. Diehl V, Sextro M, Franklin J, Hansmann ML, Harris N, Jaffe E,
compared to the standard IF-RT technique.20 In the et al. Clinical presentation, course, and prognostic factors in
lymphocyte-predominant Hodgkin's disease and lymphocyte-
experimental study arms, treatment is stratified based rich classical Hodgkin's disease: report from the European
on the results of a PET scan performed after the second Task Force on Lymphoma Project on Lymphocyte-Pre-
chemotherapy cycle. Patients with negative PET receive dominant Hodgkin's Disease. J Clin Oncol 1999;17:776-83.
3. Friedman DL, Constine LS. Late effects of treatment for
two or three additional cycles of ABVD depending on Hodgkin lymphoma. J Natl Compr Canc Netw 2006;4:249-57.
their risk profile. Patients with positive PET receive an 4. Horwich A, Specht L, Ashley S. Survival analysis of patients
intensified treatment consisting of two cycles of with clinical stages I or II Hodgkin's disease who have
relapsed after initial treatment with radiotherapy alone. Eur J
BEACOPPescalated followed by IN-RT. In the GHSG Cancer 1997;33:848-53.
HD16 trial for patients with early favorable HL, patients 5. Press OW, LeBlanc M, Lichter AS, Grogan TM, Unger JM,
receive two cycles of ABVD chemotherapy. Then, PET Wasserman TH, et al. Phase III randomized intergroup trial of
subtotal lymphoid irradiation versus doxorubicin, vinblastine,
scan is performed in all patients. In the experimental and subtotal lymphoid irradiation for stage IA to IIA
arm, those patients with positive PET additionally Hodgkin's disease. J Clin Oncol 2001;19:4238-44.
receive 30 Gy IF-RT, those with negative PET do not. In 6. Engert A, Franklin J, Eich HT, Brillant C, Sehlen S, Cartoni C,
et al. Two cycles of doxorubicin, bleomycin, vinblastine, and
the standard arm, all patients receive 30 Gy IF-RT sub- dacarbazine plus extended-field radiotherapy is superior to
sequent to chemotherapy irrespective of the PET result radiotherapy alone in early favorable Hodgkin's lymphoma:

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 153 |
14th Congress of the European Hematology Association

final results of the GHSG HD7 trial. J Clin Oncol 2007;25: Hodgkin lymphoma (HL): an analysis of the German Hodgkin
3495-502. Study Group (GHSG) Blood 2008;112:367.
7. Nogova L, Reineke T, Eich HT, Josting A, Muller-Hermelink 15. Meyer RM, Gospodarowicz MK, Connors JM, Pearcey RG,
HK, Wingbermuhle K, et al. Extended field radiotherapy, com- Bezjak A, Wells WA, et al. Randomized comparison of ABVD
bined modality treatment or involved field radiotherapy for chemotherapy with a strategy that includes radiation therapy
patients with stage IA lymphocyte-predominant Hodgkin's in patients with limited-stage Hodgkin's lymphoma: National
lymphoma: a retrospective analysis from the German Cancer Institute of Canada Clinical Trials Group and the
Hodgkin Study Group (GHSG). Ann Oncol 2005;16:1683-7. Eastern Cooperative Oncology Group. J Clin Oncol 2005;23:
8. Raemaekers J, Kluin-Nelemans H, Teodorovic I, Meerwaldt C, 4634-42.
Noordijk E, Thomas J, et al. The achievements of the EORTC 16. Straus DJ, Portlock CS, Qin J, Myers J, Zelenetz AD,
Lymphoma Group. European Organisation for Research and Moskowitz C, et al. Results of a prospective randomized clin-
Treatment of Cancer. Eur J Cancer 2002;38:S107-13. ical trial of doxorubicin, bleomycin, vinblastine, and dacar-
9. Bonadonna G, Bonfante V, Viviani S, Di Russo A, Villani F, bazine (ABVD) followed by radiation therapy (RT) versus
Valagussa P. ABVD plus subtotal nodal versus involved-field ABVD alone for stages I, II, and IIIA nonbulky Hodgkin dis-
radiotherapy in early-stage Hodgkin's disease: long-term ease. Blood 2004;104:3483-9.
results. J Clin Oncol 2004;22:2835-41. 17. Hutchings M, Loft A, Hansen M, Pedersen LM, Buhl T,
10. Noordijk EM, Carde P, Mandard AM, Mellink WA, Jurlander J, et al. FDG-PET after two cycles of chemotherapy
Monconduit M, Eghbali H, et al. Preliminary results of the predicts treatment failure and progression-free survival in
EORTC-GPMC controlled clinical trial H7 in early-stage Hodgkin lymphoma. Blood 2006;107:52-9.
Hodgkin's disease. EORTC Lymphoma Cooperative Group. 18. Gallamini A, Hutchings M, Rigacci L, Specht L, Merli F,
Groupe Pierre-et-Marie-Curie. Ann Oncol 1994;5:107-12.
11. Engert A, Pluetschow A, Eich HT, Herrmann R, Doerken B, Hansen M, et al. Early interim 2-[18F]fluoro-2-deoxy-D-glu-
Kanz L, et al. Combined modality treatment of two or four cose positron emission tomography is prognostically superior
cycles of ABVD followed by involved field radiotherapy in the to international prognostic score in advanced-stage Hodgkin's
treatment of patients with early stage Hodgkin’s lymphoma: lymphoma: a report from a joint Italian-Danish study. J Clin
Update interim analysis of the randomised HD10 study of the Oncol 2007;25:3746-52.
German Hodgkin Study Group (GHSG) Blood 2005;106:2673. 19. Kobe C, Dietlein M, Franklin J, Markova J, Lohri A, Amthauer
12. Ferme C, Eghbali H, Meerwaldt JH, Rieux C, Bosq J, Berger F, H, et al. Positron emission tomography has a high negative
et al. Chemotherapy plus involved-field radiation in early- predictive value for progression or early relapse for patients
stage Hodgkin's disease. N Engl J Med 2007;357:1916-27. with residual disease after first-line chemotherapy in
13. Noordijk EM, Thomas J, Ferme C, van’t Veer MB, Brice P, advanced-stage Hodgkin lymphoma. Blood 2008;112:3989-94.
Divine M, et al. First results of the EORTC-GELA H9 random- 20. Girinsky T, van der Maazen R, Specht L, Aleman B, Poortmans
ized trials: the H9-F trial (comparing 3 radiation dose levels) P, Lievens Y, et al. Involved-node radiotherapy (INRT) in
and H9-U trial (comparing 3 chemotherapy schemes) in patients with early Hodgkin lymphoma: concepts and guide-
patients with favorable and unfavorable early stage Hodgkin’s lines. Radiother Oncol 2006;79:270-7.
lymphoma. J Clin Oncol 2005;23:6505. 21. Radford J, O´Doherty M, Barrington S, Qian W, Patrick P,
14. Borchmann P, Engert A, Pluetschow A, Fuchs M, Markova J, Coltart S, et al. results of the 2nd planned interim analysis of
Lohri A, et al. Dose-intensified combined modality treatment the RAPID trial (involved field radiotherapy versus no further
with 2 cycles of BEACOPPescalated followed by 2 cycles of treatment) in patients with clinical stages 1A and 2A Hodgkin
ABVD and involved-field radiotherapy (IF-RT) is superior to 4 lymphoma with a “negative” FDG-PET scan after 3 cycles
cycles of ABVD and IF-RT in patients with early unfavourable ABVD. Blood 2008;112:abstract 369.

| 154 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Multiple myeloma

Pathobiology, genetics and clinical outcome


of myeloma

G. Morgan
ultiple myeloma (MM) is presently repressor BCL6, which results in the upregu-
K. Boyd

Brookes Lawley Building, Institute of


Cancer Research, Brookes Lawley
M an incurable malignancy that affects
over 3,000 new individuals each
year in the United Kingdom (UK). The inci-
lation of BLIMP1. BLIMP1 expression results
in repression of Pax5 and consequently
upregulation of XBP1. An important control
Building, 15 Cotswold Road, Sutton, dence increases with age, although approxi- point of these events relies on the presence
Surrey SM2 5NG, UK mately 40% of patients present under the of nascent immunoglobulin within the
age of 60 years. The clinical picture involves endoplasmic reticulum, which requires cor-
Hematology Education: a combination of bone destruction, immune rect folding mediated via the unfolded pro-
the education program for the deficiency, bone marrow, and renal failure tein response (UPR). The UPR ensures cor-
annual congress of the European due to the accumulation of malignant plas- rect folding, processing, export or degrada-
Hematology Association ma cells within the bone marrow. The out- tion of proteins emerging from the endoplas-
look for patients is poor, with a median sur- mic reticulum (ER). Activation of the UPR
2009;3:155-160 vival of approximately 5 years. Traditional results in a bias of translation towards the
staging systems used in MM separate synthesis of chaperone proteins involved in
patients into prognostic groups using simple protein folding, and an increase in disposal
laboratory parameters. The first reliable sys- of misfolded proteins. At the same time,
tem to be devised, the Durie-Salmon staging there is activation of the NF-κB pathway,
system, is a surrogate measurement of which acts as a survival signal. A key regula-
tumor bulk and combines a number of fac- tor of the UPR is the chaperone protein BiP,
tors including the extent of bone lesions, the which is associated with the luminal
presence of hypercalcaemia, and the level of domains of the serine/threonine kinases
paraprotein and hemoglobin.1 These basic PERK, IRE1 and the transcription factor
measurements enable clinicians to make a ATF6. In the presence of misfolded proteins,
rough estimate of prognosis, as patients with this interaction is destabilised, resulting in
stage IA disease have a median survival of 5 the release of PERK and IRE1, which simul-
years compared to patients with stage IIIB taneously results in their autocatalytic acti-
disease, who have a median survival of 14.7 vation. The most important molecule for
months. In the intervening years, a number normal plasma cell development is IRE1.
of other prognostic factors have been identi- IRE1 recruits TRAF2, which activates sig-
fied: beta 2 microglobulin (β2m), a polypep- nalling via JNK and p38 with associated acti-
tide that forms part of the extra cellular por- vation of NF-κB. For plasma cell develop-
tion of major histocompatibility complex ment, the most important function resides in
(MHC) class I molecules; the plasma cell the kinase and endoribonuclease domains.
labelling index (PCLI), a measure of tumour The endoribonuclease activity regulates the
cell proliferation; plasmablastic morphology; activation of the transcription factor XBP1.
and cytogenetics. Currently, the most easily Activation of XBP1 by the cleavage of XBP1
reproducible and most widely used staging mRNA to form XBP1s results in terminal dif-
system is the International Staging System ferentiation to a functional plasma cell. The
(ISS) which categorises patients into three activation of XBP1 also provides a survival
groups based on their presenting β2m and signal for cells passing this point, which is
albumin, with stage I patients having a mediated by IL6, as well as a positive feed-
median survival of 62 months, compared to back signal to the ER allowing it to handle
29 months in stage III patients.2 These stag- more unfolded protein. PERK phosphoryla-
ing systems do not reliably predict outcome tion inhibits the general translation initiation
in individual patients and have not been val- factor eIF2a, resulting in a general shutdown
idated in the era of novel agents. of non-chaperone protein synthesis. In the
Importantly, they are not based upon our presence of unfolded proteins, the precursor
enhanced understanding of the biological form of ATF6 is also released from BiP and
and molecular characterization of the MM translocates to the nucleus, where it medi-
cell and the role of the bone marrow ates the expression of key genes involved in
microenvironment in disease pathogenesis. the UPR response, including BiP and XBP1.
Following the recognition of misfolded pro-
Normal plasma cell differentiation teins, disposal is via the ubiquitin-protea-
In order for a B cell to exit the germinal some pathway. If there is failure to adapt to
centre and differentiate into a plasma cell, it the accumulation of unfolded protein in the
must downregulate the transcriptional ER, the cell is deleted via apoptosis mediat-

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 155 |
14th Congress of the European Hematology Association

ed by a number of pathways. CHOP/GADD153 is acti- progress per year to myeloma, with the risk of progres-
vated by activation of PERK and ATF4 with consequent sion remaining constant for a patient over a 25 year
suppression of bcl-2 transcription and the induction of period.3 The majority of patients, therefore, do not
other pro-apoptotic genes. Induction of CHOP is itself progress to malignant disease, and it is useful to consid-
repressed by NFkB, demonstrating the tight control er the genetic mechanisms leading to the development
within the UPR of survival and apoptotic mechanisms. of myeloma within this framework. Some genetic
lesions are likely to be initiating events, leading to
The environment and signalling pathways of plasma cells immortalization of the clone and producing the MGUS
Within the bone marrow, plasma cells exist in spe- phenotype, whilst secondary lesions are required to
cialised niches in close proximity to accessory cells, produce the proliferation and growth advantage
including stromal cells, osteoclasts, osteoblasts, and required to result in a malignant phenotype.
microvessels. This proximity triggers “adhesion mole- Two common genetic lesions in myeloma are IgH
cule” mediated interactions, which generate important translocations and hyperdiploidy. Normal plasma cells
autocrine and paracrine loops mediating long-term sur- undergo class switch recombination events centered on
vival and proliferation of the myeloma clone. Key the immunoglobulin heavy chain locus at 14q32 in
cytokines mediating these processes, secreted by stro- order to determine the class of immunoglobulin secret-
mal cells, include IL6, IGF1, VEGF, BAFF, FGF, SDF1 and ed by the mature cell. Illegitimate class switch events
TNFα. Important molecules secreted by the plasma cell result in translocations occurring between 14q, with the
include TNFα, VEGF and TGFβ, which further upregu- breakpoint at the IgH locus and a number of partner
late proliferative cytokines, such as IL6 secreted by the chromosomes. These molecular events juxtapose the
stromal elements. The CD40/CD40L interaction upreg- strong IgH enhancer next to an oncogene, resulting in
ulates adhesion molecules, and consequently, IL6 and up-regulated expression of the oncogene. Hyper-
VEGF secretion further. Other key interactions are with diploidy is gain of the odd numbered chromosomes,
osteoclasts (HGF, IL1β and MIP1α), osteoblasts (DKK, resulting in a complement of 48 to 74 chromosomes,
RANKL, OPG, sFRP2) and vascular elements (VEGF). and the mechanisms underlying this are not well under-
Either directly through cell adhesion or via growth fac- stood. Both these lesions have been found with similar
tor engagement, there is activation of a network of frequency in MGUS and myeloma, so probably consti-
downstream signalling molecules. Amongst the best tute initiating events producing clonal immortalization
understood of these pathways are the canonical and but are not enough in themselves to result in myeloma.4
non canonical NF-κB pathways, the JAK/STAT, Loss of heterozygosity (LOH) of an allele is a common
RAS/RAF/MAPK and PI3k/AKT pathways. Genetic mechanism for disease progression, conceptually lead-
alterations in the myeloma clone can clearly impact ing to under-expression of a tumor suppressor gene.
these plasma cell interactions, leading to significant LOH of 1p, for example, is found in 4.5% of MGUS
changes in cellular behaviour and, as such, characteriz- cases but 15% of myeloma cases, suggesting an associ-
ing these lesions can define the abnormal biology of the ation with disease progression.5 Homozygous deletions,
myeloma clone, giving insights into clinical behavior. resulting in biallelic silencing, is a progressive form of
They can also define therapeutic targets. this method of gene inactivation and has been found to
be prevalent in myeloma. Another potential molecular
Genetic mechanisms and molecular modelling of myeloma mechanism of disease progression is mutation, which
Myeloma is thought to result from the transformation results in the silencing of one allele, where perhaps the
of a proliferative “plasmablastic” cell located in the ger- other allele is already down-regulated by LOH or anoth-
minal centre. The progeny of this cell migrate to spe- er mechanism. Mutations of TP53 on 17p, for example,
cialised niches in the bone marrow, where they mature have never been described in MGUS, and are seen at
towards terminally differentiated antibody secreting rate of less than 1% in myeloma cases as a whole.
plasma cells. During this process of terminal differentia- However, in cases, which have LOH of 17p the muta-
tion, it is essential that the plasma cell should exit from tion rate is nearer 25%, which again demonstrates that
cell cycle, and close linkage of terminal differentiation co-operating events are necessary to produce the malig-
with cell cycle exit is crucial to prevent the continued nant phenotype. Another potential mechanism, where a
expansion of an immortalized pre-myelomatous clone. deleted region can lead to gene upregulation, is mediat-
Molecular lesions leading to either abnormal differenti- ed via loss of microRNAs (miRNA), which are small (17-
ation or abnormal cell cycle progression are, therefore, 25 bases) oligonucleotide sequences that regulate the
two potentially relevant pathways to myeloma. In addi- translation of other genes by complementary base pair-
tion to the requirement for generating antibody produc- ing to their mRNA. By this method, it has been estimat-
ing plasma cells, ensuring long term immunological ed that they regulate the expression of 30% of the
memory is another important part of normal plasma cell genome. Methylation of tumor suppressor genes is
biology, which if deregulated, could lead to clonal another epigenetic mechanism of gene silencing and has
immortalization. The pathological activation of survival been identified in myeloma where it has been shown to
pathways and a retained ability to proliferate in be of prognostic importance.6
response to antigen are two further potentially impor- Gain of genetic material manifest as gain of whole or
tant pathways to myeloma. interstitial chromosomal regions represents an alternate
Monoclonal gammopathy of undetermined signifi- means of tumor progression, which conceptually leads
cance (MGUS) is a clonal plasma cell dyscrasia charac- to overexpression of oncogenes. Gain of 1q has been
terized by a monoclonal paraprotein but no evidence of identified as being a region that is commonly gained in
end organ damage. One percent of patients with MGUS myeloma, and has prognostic significance.7

| 156 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

Classification of myeloma Table 1. Relationship of cyclin D expression and transloca-


Progress has been made in the classification of myelo- tion group.
ma based on its genetic make-up. Two main subgroups
of myeloma have been defined: hyperdiploid and non- Group Cyclin D Ig translocation (gene) %
hyperdiploid. Hyperdiploid cases are genetically and
clinically heterogeneous but overall, the group appears 1 3 6p21 (D3) 3.7
to have a slightly favorable prognosis. The non-hyper- 2 1 11q13 (D1) 17.3
diploid group is characterized by a high frequency of 3 2 16q23 (MAF) 8.6
4 2 4p16 (FGFR3/MMSET) 11.1
illegitimate class switch recombination events centred
5 2 - 21.0
on the IgH locus (14q32). Five main translocation part- 6 2 +1 (low) - 6.2
ners have been identified: 7 1 (low) - 32.1
1)t(4;14)(p16;q32). This translocation is found in 10% of 8 none - 1.2
myeloma patients and results in the dysregulation of
two genes.8 Firstly, on the derivative 14 (der(14))
fibroblast growth factor receptor 3 (FGFR3), a recep-
tor tyrosine kinase, is over-expressed and occasional- of newly diagnosed myeloma patients emphasizes the
ly mutated. In addition on the der(4) MMSET is over- complexity and heterogeneity of the myeloma genome.
expressed. MMSET has been shown to regulate cell In our data set, each patient had a median of 14 regions
cycle progression and cell adhesion.9-11 t(4;14) is posi- of loss of heterozygosity of which three per patient
tively associated with IgA myeloma and is an inde- were due to UPD, the remainder being due to deletion
pendent predictor of poor outcome. The majority of events.21 Most new information has arisen from this
t(4;14) cases also have del(13). technology platform, or from comparative genomic
2)t(6;14)(p21;q32). This rare translocation (4% of pri- hybridization arrays (CGH), which provide similar copy
mary myeloma samples) results in CCND3 overex- number information.
pression. Despite this apparent complexity, there are recurrent
3)t(11;14)(q13;32). This occurs in 15% of myeloma non-random structural chromosomal abnormalities and
patients and leads to overexpression of cyclin D1 it is possible to define some key regions and genes that
(CCND1), a cell cycle regulator. Outcome is neutral in are important in myeloma pathogenesis and have prog-
this group.8,12-14 nostic impact.
4)t(14;16)(q32;q23). This results in overexpression of Deletion of 1p is a frequent event in myeloma, occur-
the transcription factor c-MAF and is associated with ring in 30% of cases, and with four potential minimally
poor prognosis.15-17 Characterization of this group has deleted regions with deletion of some regions confer-
identified upregulation of Cyclin D2 (CCND2), a pro- ring an adverse prognosis.22,23 We have previously iden-
moter of cell cycle progression.18 tified a region at 1p32.3, which had LOH in 15% of
5)The t(14;20)(q32;q21) results in the overexpression of cases of newly diagnosed myeloma, and using a FISH
MAFB, a transcription factor that has similar down- probe for this region, hemizygous deletions were found
stream targets to c-maf including upregulation of in 4.5% of MGUS cases and 10.3% of SMM cases, sug-
CCND2, and is also associated with poor progno- gesting that deletion of this region is associated with
sis.19,20 disease progression.5 This region contains two genes,
A global molecular classification strategy building on whose expression is abrogated by deletion: CDKN2C
knowledge of the IgH translocation partners and based and FAF1. CDKN2C, also known as p18, is a negative
on the expression of D group cyclins has been pro- regulator of the G1/S cell cycle transition checkpoint,
posed. The rationale for the use of the D group cyclins and its deletion has been shown to induce cellular pro-
in this way is based on observations that the levels of liferation.24 FAF1 is involved in the ubiquitin pathway
cyclin D1, D2 and D3 are high, combined with the prior and inhibits the chaperone activity of the heat shock
observations that there are switch translocations, which proteins, Hsc70 and Hsp70,25,26 as well as being a nega-
dysregulate cyclin D1 or D3. It is possible to recognise tive regulator of the NF-κB pathway.27 Cases with hem-
eight distinct groups using these two basic features. izygous or homozygous deletion of CDKN2C/FAF1 had
a lower overall survival compared with cases with an
Genome mapping intact 1p32 region (20 months vs. 46 months).5
The completion of the Human Genome Project has Gain of 1q is common in primary myeloma samples,
led to the generation of a range of new technologies that being found in approximately 35% of our cases.
have transformed the way genomic abnormalities in Increased expression of genes mapping to chromosome
myeloma can be interrogated. Single nucleotide poly- 1q has been linked to shorter progression-free survival
morphism (SNP) micro-array chips focus on polymor- and overall survival post autologous stem cell trans-
phic nucleotides which are scattered throughout the plant.28 1q21 has previously been identified as a mini-
human genome, accounting for genetic variation mally amplified region, which contains the potential
between individuals in a population. The arrays allow candidate oncogenes CKS1B, PDZK1 and BCL9.7,29 We
detection of allelic loss or gain, both of which can be find a minimally deleted region between 1q21.1-q23.2,
important in tumor pathogenesis. Uniquely, they are a region containing 679 genes. Of these, only ANP32E
also able to identify uniparental disomy where both loci has significantly increased expression levels and is
are inherited from the same parent, the result of incom- linked to adverse overall survival.
plete chromosomal segregation during mitosis or A third of our cases had deletion of 6q, the majority
recombination between chromatids. SNP array analysis being interstitial deletions with the most commonly

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 157 |
14th Congress of the European Hematology Association

deleted region centred on 6q25.3. Deletion of this region in myeloma and the central role of the NF-κB pathway
was not associated with adverse prognosis in our in myeloma pathogenesis.
dataset, but there is the potential for genes located here Hemizygous deletion of 17p is detected in approxi-
to have prognostic importance. Twenty-nine percent of mately 10% of primary myeloma cases. 17p deletion is
cases have deletion of 8p, and most of these are large an independent adverse prognostic factor predicting sig-
deletions encompassing more than 75% of the short nificantly shorter overall survival in patients treated
arm with small interstitial events being rare. Deletion of with an autologous transplant regimen (15 vs. 48
this region was not linked to adverse outcome in this months),40 and we confirm this poor prognosis in our
data set. We have identified a high rate of homozygous dataset. Although the FISH probe targets the TP53 locus
deletion of a small region of 11q22.2, which contains at 17p13, mapping of samples with LOH of 17p demon-
BIRC1 and BIRC2, both of which play a role in NF-?B strates that the majority of samples have hemizygous
regulation and are, therefore, very likely to be relevant deletion of a large part of 17p, and it is difficult to define
to myeloma pathogenesis, but which do not carry prog- a minimally deleted region. Other groups have shown
nostic significance when deleted. that hemizygous deletion of 17p correlates with
Monoallelic loss of 13 is the commonest genetic decreased expression of TP53, and low expression of
abnormality of myeloma cells, found in 44% of cases TP53 to be associated with an inferior clinical out-
and is usually caused by monosomy of the whole chro- come.41 We do not find low TP53 expression in samples
mosome as opposed to interstitial deletion of 13q. with LOH in our dataset, but we have found a high
When detected by conventional cytogenetics it has been mutation rate in exons 5-9 of the remaining TP53 allele.
associated with poor prognosis.30 However, when The mutation rate for cases with LOH of 17p is 25%,
detected by FISH the prognostic significance lessens. In compared to a background rate of less than 1% in cases
our dataset, if cases which are strongly and independ- without 17p-, so it is likely that TP53 is the gene associ-
ently associated with poor outcome are removed, that ated with the poor outcome in this cytogenetic group.
is, 17p- and the poor risk IgH translocation [t(4;14),
t(14;16), t(14;20)], the effect of 13q deletion becomes Relevance of deletional mapping
neutral. It is difficult to define a minimally deleted Biology of myeloma
region, but 13q14.11-q14.3 contains all but three of our This type of data has allowed us to understand some
cases, and contains 16 significantly under-expressed of the important biological pathways, which include
genes, of which RB1 shows the largest fold change. This abnormalities of cell cycle progression, aberrant survival
region also contains the microRNA genes mir-16-1 and gene expression, abnormalities of terminal differentia-
mir-15a which have also been implicated in myeloma tion and downstream activation of cell signalling path-
pathogenesis.31 ways. Overexpression of a cyclin D family member is
NF-κB pathway activation has been linked to muta- recognized as being a recurrent abnormality and loss of
tions and deletions of several regulators of the canonical negative regulators of cell cycle progression have also
and non-canonical pathway.32,33 The net result of these been identified as being important. Later phases of dis-
abnormalities is inhibition of the central negative regu- ease often have increased proliferation and MYC abnor-
lator of the pathway, IKK, resulting in a lifting of its reg- malities. Survival abnormalities are encapsulated in
ulatory effect and, therefore, constitutive NF-κB activa- what we understand about the frequent events such as
tion. The single most common abnormality is inactiva- 17p- (TP53 mutation) and abnormalities of WWOX.
tion of TRAF3, which maps to 14q32. Biallelic loss of RAS mutations are frequent in late phase disease and
this locus has been demonstrated in 4% of myeloma lead to IL6 independent survival. In an analogous fash-
patients, whilst 12% were shown to have loss of het- ion, we also understand now that deregulation of the
erozygosity. Moreover, decreased expression of TRAF3 NF-κB pathway by both deletional events and mutation
was linked to clinical outcome based on retrospective is important in disease progression. Examples of genes
analysis of a trial of relapsed myeloma patients treated affected in the NF-κB pathway are the CYLD, TRAF and
with either dexamethasone or bortezomib. Patients BIRC genes, which will become increasingly important
with low TRAF3 expression had high response rates to in predicting response to specific therapeutic modalities.
bortezomib (89% vs. 40%) and low response rates to The overexpression of XBP1 and other abnormalities of
steroids (10% vs. 30%).32 this key regulator gene and pathway in terminal plasma
Deletion of 16q23 has been found in 20% of primary cell differentiation, points the way to the relevance of
myeloma samples. Overall survival at 3 years in cases abnormalities of plasma cell differentiation to myeloma
with del(16q) was 42% vs. 72% in those without dele- cell survival and proliferation.
tion, although longer follow-up data suggests a neutral
prognostic effect. Using SNP-based gene mapping com- Clinical outcome of myeloma
bined with gene expression data it is possible to identi- Taken together this high-resolution gene mapping has
fy two key genes which are dysregulated in these cases: identified regions and genes with relevance to myeloma
WWOX and CYLD.34 WWOX is a known tumor sup- pathogenesis, and some with prognostic significance.
pressor gene deleted in other malignancies, such as However, few of these abnormalities are associated
prostate and breast cancer.35,36 CYLD is a tumor suppres- with a sufficiently poor prognosis to consider changing
sor gene first identified in cylindroma tumors, a heredi- a patient from conventional treatment pathways. This
tary skin cancer37 and is known to function as a negative may change as the accrual of more mapping cases
regulator of the NF-κB pathway.38,39 It is possible to iden- changes levels of statistical significance, but we present-
tify mutations in CYLD in the cases with LOH, con- ly have much larger and more robust data related to
firming its role as an important tumor suppressor gene genetic lesions detected by FISH. 17p- and the poor risk

| 158 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

translocations (t(4;14), t(14;16), t(14;20)) have a much multiple myeloma. J Clin Oncol 2005;23:3412-20.
3. Kyle RA, Rajkumar SV, Offord JR, Larson DR, Plevak MF,
worse prognosis than cases without these lesions, and Melton LJ. A Long-term study of prognosis in monoclonal
would fit the criteria of high risk disease. gammopathy of undetermined significance. N Engl J Med
2002;346:564-9.
Gene expression signatures 4. Fonseca R, Bailey RJ, Ahmann GJ, Rajkumar SV, Hoyer JD,
Lust JA et al. Genomic abnormalities in monoclonal gam-
An alternative approach to looking at individual mopathy of undetermined significance. Blood 2002;100:1417-
lesions is to use patterns of gene expression, termed 24.
gene expression signatures. The first prognostic gene 5. Leone PE, Walker BA, Jenner MW, Chiecchio L, Dagrada G,
Protheroe RK et al. Deletions of CDKN2C in multiple myelo-
expression signature to be defined in this way was ma: biological and clinical implications. Clin Cancer Res 2008;
based on a patient set who received a tandem autotrans- 14:6033-41.
plantation-based regimen.28 A list of 70 genes was pro- 6. Takada S, Morita K, Hayashi K, Matsushima T, Sawamura M,
Murakami H, Nojima Y. Methylation status of fragile histidine
duced, the differential expression of which was able to triad (FHIT) gene and its clinical impact on prognosis of
identify 13% of presenting patients at risk of early dis- patients with multiple myeloma. Eur J Haematol 2005;75:505-
ease-related death, making it a more discriminating 10.
prognostic indicator than the ISS. More recently, a 15 7. Shaughnessy J. Amplification and overexpression of CKS1B at
chromosome band 1q21 is associated with reduced levels of
gene survival signature has been produced by a similar p27Kip1 and an aggressive clinical course in multiple myelo-
method based on an IFM series of patients treated with ma. Hematology 2005;10:117-26.
chemotherapy followed by high dose melphalan with 8. Avet-Loiseau H, Facon T, Facon T, Brigaudeau C, Morineau N,
Maloisel F et al. High incidence of translocations t(11;14)
stem cell rescue.42 High-risk patients had overexpression (q13;q32) and t(4;14)(p16;q32) in patients with plasma cell
of genes involved in cell cycle progression and a 3 year malignancies. Cancer Res 1998;58:5640-5.
OS of 47%. Low risk patients displayed hyperdiploid 9. Chesi M, Nardini E, Lim RS, Smith KD, Kuehl WM, Bergsagel
PL. The t(4;14) translocation in myeloma dysregulates both
signatures and had a 3 year OS of 90%. Again, this sig- FGFR3 and a novel gene, MMSET, resulting in IgH/MMSET
nature discriminates patients with the best and worst hybrid transcripts. Blood 1998;92:3025-34.
prognoses more effectively than the ISS. We have pro- 10. Richelda R, Baldini L, Baldini L, Cro L, Viggiano L, Marzella R,
duced a prognostic gene expression signature by a dif- Rocchi M et al. A novel chromosomal translocation t(4;14)
(p16.3;q32) in multiple myeloma involves the fibroblast
ferent method. We mapped regions with homozygous growth factor receptor 3 gene. Blood 1997;90:4062-70.
deletions and combined this with expression array data 11. Brito JL, Walker B, Jenner M, Dickens NJ, Brown NJ, Ross FM
to identify genes whose expression is abrogated by et al. MMSET deregulation affects cell cycle progression and
adhesion regulons in t(4;14) myeloma plasma cells. Haema-
deletion. We have used this approach to define a prog- tologica 2009;94:78-86.
nostic signature based on the expression of six genes. 12. Fonseca R, Witzig TE, Gertz MA, Kyle RA, Hoyer JD, Jalal SM,
Greipp PR. Multiple myeloma and the translocation
Summary of current status t(11;14)(q13;q32): a report on 13 cases. Br J Haematol 1998;
101:296-301.
Understanding the genetic abnormalities that are 13. Fonseca R, Blood EA, Oken MM, Kyle RA, Dewald GW,
associated with malignant behaviour in plasma cells is Bailey RJ et al. Myeloma and the t(11;14)(q13;q32); evidence
important as they ultimately define the way that for a biologically defined unique subset of patients. Blood
2002;99:3735-41.
myeloma behaves in an individual, and are, therefore, 14. Gertz MA, Lacy MQ, Dispenzieri A, Greipp PR, Litzow MR,
likely to be the best way of predicting prognosis in the Henderson KJ et al. Clinical implication of the t(11;14)
individual. Our current data suggests that few single (q13;q32), t(4;14)(p16.3;q32) and -17p13 in myeloma patients
treated with high dose therapy. Blood 2005;100:1417-24.
genetic lesions have sufficient prognostic power to jus- 15. Chesi M, Bergsagel PL, Shonukan OO, Martelli ML, Brents LA,
tify a change of treatment. The exceptions are 17p-, Chen T et al. Frequent dysregulation of the c-maf proto-onco-
where the median survival is dismal at approximately gene at 16q23 by translocation to an Ig locus in multiple
myeloma. Blood 1998;91:4457-63.
22 months, and the unfavourable IgH translocations, 16. Sawyed JR, Lukacs JL, Munshi N, Desikan KR, Singhal S,
where the median survival is 26 months, compared to Mehta J et al. Identification of new nonrandom translocations
50 months for those without the lesions. Several gene in multiple myeloma with multicolor spectral karyotyping.
expression signatures have recently been produced Blood 1998;1998:4269-78.
17. Sawyer JR, Lukacs JL, Munshi N, Desikan KR, Singhal S,
which may have sufficient power to identify the high- Mehta J et al. Identification of new nonrandom translocations
est risk patients. These may ultimately prove to be the in multiple myeloma with multicolor spectral karyotyping.
most powerful way of identifying high-risk patients. Blood 1998;92:4269-78.
18. Hurt EM, Wiestner A, Rosenwald A, Shaffer AL, Campo E,
However, they are relatively new in myeloma and are Grogan T et al. Overexpression of c-maf is a frequent onco-
derived assessments of the tumor as opposed to 17p- genic event in multiple myeloma that promotes proliferation
and IgH translocations, which are anchored in the and pathological interactions with bone marrow stroma.
Cancer Cell 2004;5:191-9.
tumor genome. They also rely on technology, which is 19. Boersma-Vreugdenhil GR, Kuipers J, Van Stralen E, Peeters T,
not widely available in the clinical setting so remain an Michaux L, Hagemeijer A et al, The recurrent translocation
experimental tool. FISH abnormalities, possibly in com- t(14;20)9q32;q12) in multiple myeloma results in aberrant
expression of MAFB:a molecular and genetic analysis of the
bination with independent risk factors such as β2m, are chromosomal breakpoint. Br J Haematol 2004;126:355-63.
currently the most accurate prognostic tool we have for 20. van Stralen E, van de Wetering M, Agnelli L, Neri A, Clevers
the newly presenting patient with myeloma. HC, Bast BJ. Identification of primary MAFB target genes in
multiple myeloma. Exp Hematol 2009;37:78-86.
21. Walker B, Jenner M, Cheng L, Gonzalez D, Johnson D, Ross F,
et al. Integration of global SNP-based mapping and expression
References arrays reveals key regions, mechanisms, and genes important
in the pathogenesis of multiple myeloma. Blood 2005;108:
1. Durie BG, Salmon SE. A clinical staging system for multiple 1733-43.
myeloma. Correlation of measured myeloma cell mass with 22. Hayami Y, Iida S, Nakazawa N, Hanamura I, Kato M,
presenting clinical features, response to treatment, and sur- Komatsu H et al. Inactivation of the E3/LAPTm5 gene by chro-
vival. Cancer 1975;36:842-54. mosomal rearrangement and DNA methylation in human
2. Griepp PR, Durie BG. et al, International staging system for multiple myeloma. Leukemia 2003;17:1650-7.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 159 |
14th Congress of the European Hematology Association

23. Schultheis B, Krämer A, Willer A, Hegenbart U, Goldschmidt 34. Jenner MW, Leone PE, Walker BA, Ross FM, Johnson DC,
H, Hehlmann R. Analysis of p73 and p53 gene deletions in Gonzalez D et al. Gene mapping and expression analysis of
multiple myeloma. Leukemia 1999;13:2099-103. 16q loss of heterozygosity identifies WWOX and CYLD as
24. Dib A, Peterson TR, Raducha-Grace L, Zingone A, Zhan F, being important in determining clinical outcome in multiple
Hanamura I et al. Paradoxical expression of INK4c in prolifer- myeloma. Blood 2007;110:3291-300.
ative multiple myeloma tumors: bi-allelic deletion vs increased 35. Qin HR, Iliopoulos D, Semba S, Fabbri M, Druck T, Volinia S
expression. Cell Div 2006;1:23. et al. A role for the WWOX gene in prostate cancer. Cancer
25. Song EJ, Yim SH, Kim E, Kim NS, Lee KJ. Human Fas-associat- Res 2006;66:6477-81.
ed factor 1, interacting with ubiquitinated proteins and val- 36. Iliopoulos D, Fabbri M, Druck T, Qin HR, Han SY, Huebner K.
osin-containing protein, is involved in the ubiquitin-protea- Inhibition of breast cancer cell growth in vitro and in vivo:
some pathway. Mol Cell Biol 2005;25:2511-24. effect of restoration of Wwox expression. Clin Cancer Res
26. Kim HJ, Song EJ, Lee YS, Kim E, Lee KJ. Human Fas-associat- 2007;13:268-74.
ed factor 1 interacts with heat shock protein 70 and negative- 37. Bignell GR, Warren W, Seal S, Takahashi M, Rapley E, Barfoot
ly regulates chaperone activity. J Biol Chem 2005;280:8125-33. R et al. Identification of the familial cylindromatosis tumour-
27. Park MY, Moon JH, Lee KS, Choi HI, Chung J, Hong HJ, Kim suppressor gene. Nat Genet 2000;25:160-5.
E. FAF1 suppresses IkappaB kinase (IKK) activation by disrupt- 38. Brummelkamp TR, Nijman SM, Dirac AM, Bernards R et al.
ing the IKK complex assembly. J Biol Chem 2007;282:27572-7. Loss of the cylindromatosis tumour suppressor inhibits apop-
28. Shaughnessy JD, Zhan F, Burington BE, Huang Y, Colla S, tosis by activating NF-kappaB. Nature 2003;424:797-801.
Hanamura I et al. A validated gene expression model of high- 39. Kovalenko A, Chable-Bessia C, Cantarella G, Israël A, Wallach
risk multiple myeloma is defined by deregulated expression of
genes mapping to chromosome 1. Blood 2007;109:2276-84. D, Courtois G. The tumour suppressor CYLD negatively reg-
29. Inoue J, Otsuki T, Hirasawa A, Imoto I, Matsuo Y, Shimizu S ulates NF-kappaB signalling by deubiquitination. Nature 2003;
et al. Overexpression of PDZK1 within the 1q12-q22 amplicon 424:801-5.
is likely to be associated with drug-resistance phenotype in 40. Chang H, Yi QL, Reece D, Stewart AK. p53 gene deletion
multiple myeloma. Am J Pathol 2004;165:71-81. detected by fluorescent in situ hybridization iks an adverse
30. Tricot G, Barlogie B, Jagannath S, Bracy D, Mattox S, Vesole prognostic factor for patients with multiple myeloma follow-
DH et al. Poor prognosis in multiple myeloma is associated ing autologous stem cell transplantation. Blood 2005;105:358-
only with partial or complete deletions of chromosome 13 or 60.
abnormalities involving 11q and not with other karyotype 41. Xiong W, Wu X, Starnes S, Johnson SK, Haessler J, Wang S et
abnormalities. Blood 1995;86:4250-6. al. An analysis of the clinical and biological significance of
31. Fonseca TH. MicroRNA 15a and 16-1 target identidication in TP53 loss and the identification of potential novel transcrip-
deletion 13 multiple myeloma. Blood 2008;112:631. tional targets of TP53 in multiple myeloma. Blood 2008.
32. Keats JJ, Fonseca R, Chesi M, Schop R, Baker A, Chng WJ et al. 42. Decaux OLL, Lodé L, Magrangeas F, Charbonnel C, Gouraud
Promiscuous mutations activate the noncanonical NF-kappaB W, Jézéquel P et al. Prediction of survival in multiple myeloma
pathway in multiple myeloma. Cancer Cell 2007;12:131-44. based on gene expression profiles reveals cell cycle and chro-
33. Annunziata CM, Davis RE, Demchenko Y, Bellamy W, Gabrea mosomal instability signatures in high-risk patients and hyper-
A, Zhan F et al. Frequent engagement of the classical and alter- diplod signatures in low risk patients: a study of the
native NF-kappaB pathways by diverse genetic abnormalities Intergroupe Francophone dy Myelome. J Clin Oncol 2008;26:
in multiple myeloma. Cancer Cell 2007;12:115-30. 4798-805.

| 160 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Multiple myeloma

Experimental agents knocking at the door of myeloma


treatment

N.C. Munshi
espite the advances in conventional from outcome-annotated clinical specimens
Boston VA Healthcare System,
Jerome Lipper Multiple Myeloma
Center, Dana-Farber Cancer
D and high dose chemotherapy, multi-
ple myeloma (MM) remains incur-
able. In order to overcome resistance to cur-
has allowed for further identification of
DNA based prognostic classification system.
It has defined discrete minimal common
Institute, Harvard Medical School, rent therapies and improve patient outcome, regions and generated a refined list of MM
Boston, USA novel biologically-based treatment gene candidates residing within these MCRs
approaches are being developed based upon as potential therapeutic targets.13,14
targeting the MM cell, as well as its BM Techniques, such as systematic RNA inter-
Hematology Education: microenvironment.1 Towards validating ference (RNAi) lethality screening of the
the education program for the novel targets and developing newer agents, “druggable genome” in human myeloma cell
annual congress of the European we and others have developed in vitro sys- lines (HMCL), is now providing a compre-
Hematology Association tems and in vivo animal models to study hensive map of molecular vulnerabilities in
growth, and survival mechanisms intrinsic human MM. This technique identifies addi-
2009;3:161-165 to MM cells and extrinsic factors (MM cell- tional targets with functional significance in
bone marrow stromal cell interactions, MM cell survival.
cytokines, angiogenesis) promoting MM cell Genomic and proteomic studies can also
growth, survival, drug resistance, and migra- provide the preclinical basis for clinical appli-
tion in the BM microenvironment.2-6 These cation of novel targeted therapies. For exam-
model systems have allowed for the devel- ple, our in vitro studies demonstrated that
opment of several promising biologically- exposure of MM cells to bortezomib down
based therapies, which can target the MM regulates DNA repair-related genes and
cells in their BM microenvironment, and induces cleavage of DNA repair kinases,
thereby, overcome classical drug resistance such as DNA PKcs in a dose and time
in vitro, including thalidomide (Thal) and its dependent manner.15 For the first time, this
more potent immunomodulatory analogs, observation suggests that bortezomib
lenalidomide, as well as proteasome inhibits DNA repair. Subsequent in vitro stud-
inhibitor bortezomib.7 ies demonstrated that coupling bortezomib
Once in vitro promise of these novel agents with DNA damaging agents (alkylating
is demonstrated, they are rapidly tested in agents and anthracyclines) can enhance sen-
murine models of human myeloma.8 Thal sitivity or even restore sensitivity to these
and lenalidomide9,10 and bortezomib11 all agents in resistant MM cells.16 Already clini-
inhibit human MM cell growth, decrease cal protocols coupling Velcade with Doxil
associated angiogenesis, and prolong host and with melphalan17,18 are demonstrating
survival in animal models. These agents very promising clinical results.
have already demonstrated marked clinical
anti-MM activity and have now been incor- Understanding of the molecular signalling
porated in the routine management of both pathways to identify novel targets
newly diagnosed, as well as relapsed multi- Understanding how the interaction and
ple myeloma. However, despite a high-level adhesion of MM cells to bone-marrow stro-
of success of these new agents, patients mal cells (BMSCs) leads to adhesion- and
relapse, and there is a need for novel target- cytokine-induced signaling, and delineating
ed therapies in myeloma. those pathways mediating tumor cell
growth, survival, and drug resistance in the
Oncogenomics to identify novel targets BM milieu has improved our understanding
Understanding the molecular pathogene- of myeloma cell pathobioogy and provided
sis of MM by gene expression profiling has series of potential therapeutic targets. (Figure
demonstrated sequential genetic changes 1).1 MM cell binding to BMSCs induces
from normal to malignant plasma cells and nuclear factor B (NF-κB) in BMSCs, which
highlighted specific pathways important in upregulates adhesion molecules and cyto-
the multistep transformation process of nor- kines secretion, including interleukin-6 (IL-
mal plasma cells to monoclonal gammopa- 6), insulin-like growth factor-1 (IGF1),
thy of undetermined significance to MM.12 tumor-necrosis factor-α (TNF-α), and vascu-
Moreover, a high-resolution array based lar endothelial growth factor (VEGF). Adhe-
comparative genomic hybridization (aCGH) sion and cytokines activate p42/44 mitogen-
analyzing recurrent copy number alterations activated protein kinase (MAPK), Janus
with integrated expression profiling data kinase (JAK)/signal transducer and activator

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 161 |
14th Congress of the European Hematology Association

Figure 1. Potential molecular targets in multiple myeloma cells and their interactions with the BM microenvironment.

of transcription 3 (STAT3), and/or phosphatidylinositol


Table 1. Cell surface targeting agents in clinical studies.
3-kinase (PI3K)/Akt, and their downstream pathways
that mediate MM cell growth, survival, and migration. Target Agent Phase
Specifically, the Ras/Raf/MAPK pathway mediates pro-
liferation of MM. JAK/STAT3, along with upregulation IL6 CNTO328 (mAb) I/II
of Bcl-XL and Mcl-1, mediates survival. PI3K/Akt, IL6R Altizumab (mAb) III
through downstream activation of BAD and NF-κB CD40 SGN-40 (mAb) I
and/or inactivation of caspase-9, mediates anti-apopto-
HCD122) (mAb) I
sis. NF-κB and forkhead in rhabdomyosarcoma (FKHR)
CD56 huN901-DM1 (mAb) I
modulate cyclin D and KIP1, thereby regulating cell-
cycle progression. Signalling through PI3K induces CD74 anti-CD74 mAb I/II
downstream protein kinase C (PKC) activity and MM CD138 BT-162 (mAb) I
cell migration (Hideshima et al. Nat Can Review 2007). CS1 HuLuc63 (mAb) I
RANKL AMG162 (mAb) I/II
Identification and validation of novel targeted multiple DKK-1 Anti-DKK-1 mAb I
myeloma therapies: cell surface targets MUC1 AR20.5 (BrevaRex)(mAb) I/II
Proteamic and genomic studies have identified a BAFFR (TACI) TACI-IG (mAb) I/II
number of cell surface molecules and have formed the CD52 Alemtuzumab (mAb) II
basis for therapeutic tareting. These targets are either TRAIL Ppo2L/TRAIL (Apo2 ligand) I
adhesion molecules or growth factor receptors on the Mapatumumab (HGS-ETR1) I/II
MM cells, or are cytokines present in the BM milieu IGF1, IGF1R CP-571 (mAb) I
with significant effect on myeloma cell behavior. Table EM164 (mAb) I
1 lists the molecular targets and agents directed at these VEGF, VEGFR Bevacizumab (mAb) II
cell surface targets19-21 in clinical studies. SU5416 (SMI) II
Majority of these molecules are targeted using mono- Vatalanib (PTK-787) (SMI) II
clonal antibodies (mAb), which are now established as Zactima (ZD6474) (SMI) II
an important therapeutic modality in various cancers, FGF, PDGF TKI258 (mAb) I
especially after the success of Rituximab targeting FGFR3 Dasatinib (SMI) II
CD20 in the treatment of B-cell non-Hodgkin’s lym-
phoma. Although there is still no mAb-based therapy Adapted from Hideshima et al. 2007 Nature Reviews Cancer .

| 162 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

approved for the treatment of MM, a number of agents Table 2. Intracellular molecule targeting agents in clinical
are at various stages of development. Many of these studies.
mAbs are now being tested as single agents but more
importantly, a number of them are being evaluated as a Target Agent Single agent/ Phase
conjugate molecule (CD56,22 CD38, CD13823) with the combination
ability to deliver potent chemotherapeutic agents
specifically to myeloma cells. Earlier studies have Proteasome NPI-0052 Single/Lenalidomide I/II
shown that Rituximab had minimal clinical activity due PR-171 Single II
Mitochondria GCS-100 Single I/II
to only few MM cells expressing CD20. The significant
CCI-779 Bortezomib II
increase in the number of antibodies in clinical trials are RAD001 Bortezomib II
now mainly attributed to better understanding of MM Akt Perifosine Bortezomib II/III
biology and identification of antigen expression pat- Histone deacetylase LBH589 Bortezomib II
terns by gene expression profiling and oncogenomics, as Vorinostat BortezomibI III
well as the advancement in mAbs engineering. Romidepsin Single I/II
It is also possible to combine monoclonal antibodies Belinostat (PDX101) Single II
with novel drugs to potentiate their activity. Lena- HSP90 KOS953 Bortezomib I/II
lidomide, for example, can markedly augment anti- IPI504 Single I/II
body-dependent cellular cytotoxicity (ADCC) induced SRC, RAC1, JNK Plitidepsin (Aplidin) Single/Bortezomib I/II
by anti-CD40 in MM.19,24 PKC Enzastaurin Bortezomib I/II
Heparanase PI-88 II
Targets within multiple myeloma cell BCL2 G3139 (Genasense) Dexamethasone III
We and others have utilized high resolution genomic Telomerase GRN163L Single I
profiles to both identify additional potential novel ther- Farnesyltransferase Tipifarnib (R115777) Bortezomib II
apeutic targets in tumor cells, as well as to define dis- MEK AZD6244 Single I/II
tinct clinical pathogenetic subgroups of MM. The sig- DNA Bendamustine Single II
naling cascades and molecular mechanisms whereby Temozolomide Single II
NF-κB,25 p38MAPK,26 telomerase,27 Ku86/70,28 and cave- O-6-benzylguanine Single II
olae29 mediate homing and adhesion to BM are now CDK Flavopiridol Single I
Multi-kinases Atiprimod Single II/II
delineated, providing the framework for therapeutic
VQD-001 Single I/II
strategies targeting these cascades. The understanding ZIO-101 Single I/II
that the MM cell growth is mediated via ERK/MAPK,26 Arsenic trioxide Single II
survival via JAK/STAT,30 drug resistance via PI3-K/Akt,31
and migration via PKC dependent signaling cascades,32 Adapted from Hideshima et al. 2007, Nature Reviews Cancer
respectively, has provided novel agents (Table 2). These
include the new proteasome inhibitors: VEGF receptor
tyrosine kinase inhibitor PTK78757, pan VEGF receptor
inhibitors GW786034B; multi-targeted kinase inhibitor
PKC-412; 2-methoxyestradiol (2ME); p38MAPK Profiling of gene and protein expression has provided
inhibitor; histone deacetylase (HDAC) inhibitors the preclinical rationale for clinical protocols combining
SAHA, romidopsine and LBH589; heat shock protein 90 novel targeted therapies. For example, our studies
(hsp90) inhibitor geldanamycin; telomerase inhibitors demonstrate that bortezomib treatment of MM cells in
GRN163; inosine monophosphate dehydrogenase vitro induces death signaling, down regulates survival
inhibitor VX944117; small molecule Akt inhibitor peri- signaling, and up regulates both ubiquitin/proteasome
fosine, PKCβ inhibitor enzastaurin; and MEK inhibitor and stress response gene transcripts. Specifically, borte-
AZD6244. The new proteasome inhibitors have irre- zomib induces Hsp90, which is not only a stress
versible proteasome inhibitor activity (PR171) or affect response protein but also plays a major role in the pro-
all three active sites in proteasome (NPI-0052), and both tein unfolding required before proteins can be degraded
have demonstrated activity against bortezomib resist- by the proteasome.33 In vitro studies show that Hsp90
ant MM cells. inhibitor 17AAG can block the Hsp90 stress response
induced by bortezomib, and thereby, increase MM cell
Development of combinations apoptosis.34 These gene microarray studies, therefore,
It is our hypothesis that drugs from multiple of these provided the framework for a clinical trial coupling
classes will need to be combined to achieve complete these agents in MM, which shows that Hsp90 inhibitor
eradication of MM cells. Our ongoing studies of their KOS953 can sensitize to, and even overcome, resistance
molecular mechanisms of action aim at providing a to bortezomib.35
framework for clinical trials of rationally designed com- In order to provide the framework for coupling these
binations to overcome drug resistance and improve novel agents in rational clinical trials, the apoptotic sig-
patient outcome. The overall goal is to develop: (i) com- naling cascades triggered in MM cells by both conven-
bination therapies optimized to enhance cytotoxicity, tional and novel agents have been characterized.7,36 For
overcome drug resistance, and allow for use of lower example, use of IMiDs with TRAIL provides dual trig-
doses with less toxicity; (ii) and provide a framework gering of caspase 8 death signaling, whereas treatment
for the development of the next generation of potent with IMiDs (lenalidomide) and bortezomib triggers
targeted therapies with higher selectivity and less toxic- both caspase 8 and caspase 9-mediated MM cell death,
ities. providing potential for synergistic activity. An ongoing

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 163 |
14th Congress of the European Hematology Association

clinical trial has demonstrated remarkable activity in cascades mediating multiple myeloma cell growth and migra-
tion. Blood 2001;98:428-35.
patients treated with lenalidomide/bortezomib, even in 6. Urashima M, Ogata A, Chauhan D, Hatziyanni M, Vidriales
patients resistant to either agent alone.37 MB, Dedera DA et al. Transforming growth factor b1:
mTOR inhibitor rapamycin sensitizes MM cells to Differential effects on multiple myeloma versus normal B
both conventional (dexamethasone) and novel thera- cells. Blood 1996;87:1928-38.
7. Hideshima T, Anderson KC. Molecular mechanisms of novel
pies;38 CCI-779 has anti-MM activity in xenograft mod- therapeutic approaches for multiple myeloma. Nat Rev
els,39 which has led to combination studies. Bortezomib Cancer 2002;2:927-37.
inhibits growth (MEK/ERK) and survival (Jak/STAT) sig- 8. Tassone P, Neri A, Burger R, Burger R, Goldmacher VS, Fram
R et al. A clinically relevant SCID-hu in vivo model of human
naling, but activates Akt, providing the preclinical multiple myeloma. Blood 2005;106:713-6.
rationale for combining bortezomib with the Akt 9. Raje N, Anderson KC. Thalidomide: A Revival Story. N Engl J
inhibitor perifosine.40 Med 1999;341:1606-9.
10. Hideshima T, Chauhan D, Shima Y, Raje N, Davies FE, Tai YT
Recent signaling studies have defined the role of the et al. Thalidomide and its analogues overcome drug resistance
aggresome in degrading ubiquitinated protein in MM, of human multiple myeloma cells to conventional therapy.
and specifically used the HDAC6 inhibitor to inhibit its Blood 2000;96:2943-50.
11. Hideshima T, Mitsiades C, Akiyama M, Hayashi T, Chauhan
transport to the aggresome for degradation. Blocking D, Richardson P et al. Molecular mechanisms mediating anti-
the aggresome induces a compensatory upregulation of myeloma activty of proteasome inhibitor PS-341. Blood 2003;
the proteasome; conversely, blocking the proteasome 101:1530-4.
with bortezomib triggers a compensatory upregulation 12. Davies F, Dring AM, Li C, Rawstron AC, Shammas MA,
O'Connor SM et al. Insights into the multistep transformation
of the aggresome. Importantly, blocking both the pro- of MGUS to myeloma using microarray expression analysis.
teasome and aggresome with bortezomib and HDAC6 Blood 2003;102:4504-11.
inhibitor, respectively, induces synergistic toxicity.41 13. Carrasco DR, Tonon G, Huang Y, Zhang Y, Sinha R, Feng B et
al. High-resolution genomic profiles defines distinct clinico-
This strategy is now under intense investigation in both pathogenetic subgroups of multiple myeloma patients. Cancer
phase II and III studies. Cell 2006;9:313-25.
Finally, gene expression profiling in correlative science 14. Bergsagel DE, Kuehl M, Zhan F, Sawyer J, Barlogie B,
Shaughnessy J. Cyclin D dysregulation: an early and unifying
studies of patients on clinical protocols will both identi- pathogenic event in multiple myeloma. Blood 2005;106:296-
fy targets of drug sensitivity versus resistance, and allow 303.
for predicting those patients most likely to respond to 15. Hideshima T, Mitsiades C, Akiyama M, Hayashi T, Chauhan
D, Richardson P et al. Molecular mechanisms mediating
conventional and novel targeted therapies. For example, antimyeloma activity of proteasome inhibitor PS-341. Blood
gene expression profiling of patient tumor samples 2003;101:1530-4.
showed genes upregulated in patients responding to 16. Mitsiades N, Mitsiades C, Richardson PG, Poulaki V, Tai YT,
bortezomib versus those patients who did not respond. Chauhan D et al. The proteasome inhibitor PS-341 potentiates
sensitivity of multiple myeloma cells to conventional
Hsp27 upregulation correlated with intrinsic or acquired chemotherapeutic agents: therapeutic applications. Blood
bortezomib resistance. Preclinical studies showed that 2003;101:2377-80.
p38MAPK inhibition downregulated Hsp27 expression 17. Orlowski RZ, Nagler A, Sonneveld P, Bladé J, Hajek R, Spencer
A et al. Randomized phase III study of pegylated liposomal
and restored bortezomib sensitivity in resistant MM cell doxorubicin plus bortezomib compared with bortezomib
lines and patient samples,26,42,43 providing the basis for a alone in relapsed or refractory multiple myeloma: combina-
trial combining these two agents. tion therapy improves time to progression. J Clin Oncol 2007;
25:3892-901.
Ultimately, it may be possible to carry out gene and 18. San Miguel J, Schlag R, Khuageva N, Dimopoulos MA, Shpil-
protein expression profiling on individual patient sam- berg O, Kropff M et al. Bortezomib plus melphalan-pred-
ples to allow selection of those agents most likely to be nisone versus melphalan-prednisone in untreated multiple
myeloma patients ineligible for stem cell transplantation. N
effective. For example, we recently compared the gene Engl J Med 2008. [Epub ahead of print]
profile of patient MM cells to normal twin plasma cells 19. Tai YT, Catley L, Mitsiades CS, Burger R, Podar K, Shringpaure
and showed surprisingly few significant differences.44 R et al. Mechanisms by which SGN-40, a humanized anti-
This data may allow selection of those combinations of CD40 antibody, induces cytotoxicity in human multiple
myeloma cells: clinical implications. Cancer Res 2004;64:2846-
agents targeting these gene products to optimize clinical 52.
response. 20. Tai YT, Dillon M, Song W, Leiba M, Li XF, Burger P et al. Anti-
CS1 humanized monoclonal antibody HuLuc63 inhibits
myeloma cell adhesion and induces antibody-dependent cellu-
lar cytotoxicity in the bone marrow milieu. Blood 2007.
References 21. Tai YT, Li SF, I. B, Song W, Neri P, Catley L et al. Role of B cell
activating factor in adhesion and growth of human multiple
1. Hideshima T, Mitsiades C, Tonon G, Richardson PG, myeloma cells in the bone marrow microenvironment. Cancer
Anderson KC. Understanding multiple myeloma pathogenesis Res 2006;66:6675-56.
in the bone marrow to identify new therapeutic targets. Nat 22. Tassone P, Gozzini A, Goldmacher V, et al. In vitro and in vivo
Rev Cancer 2007;7:585-98. antitumor activity of the maytansinoid immunoconjugate
2. Ogata A, Chauhan D, Teoh G, Treon SP, Urashima M, Schloss- hu901-N2-deacetyl-N2-(3-mercapto-1-oxopropyl)- maytan-
man RL, Anderson KC. Interleukin-6 triggers cell growth via sine against CD56+ multiple myeloma cells. Cancer Res 2004;
the ras-dependent mitogen-activated protein kinase cascade. J 64:4629-36.
Immunology 1997:2212-21. 23. Tassone P, Goldmacher V, Neri P, Gozzini A, Shammas MA,
3. Chauhan D, Uchiyama H, Akbarali Y, Urashima M, Whiteman KR et al. Cytotoxic activity of the maytansinoid
Yamamoto K, Libermann TA, Anderson KC. Multiple myelo- immunoconjugate BB4-DM1 against CD138+ multiple myelo-
ma cell adhesion-induced interleukin-6 expression in bone ma cells. Blood. 2004;104:3688-96.
marrow stromal cells involves activation of NF-kB. Blood 24. Tai YT, Li SF, Catley L, Coffey R, Breitkreutz I, Bae J et al.
1996;87:1104-12. Immunomodulatory drug lenalidomide augments anti-CD40-
4. Hideshima T, Chauhan D, Schlossman RL, Richardson PR, induced cytotoxicity in human multiple myeloma: clinical
Anderson KC. Role of TNF-alpha in the pathophysiology of implications. Cancer Res 2005;65:11712-20.
human multiple myeloma: therapeutic applications. 25. Hideshima T, Chauhan D, Richardson P, Mitsiades C,
Oncogene 2001;20:4519-27. Mitsiades N, Hayashi T et al. NF-kappa B as a therapeutic tar-
5. Podar K, Tai YT, Davies FE, Lentzsch S, Sattler M, Hideshima get in multiple myeloma. J Biol Chem 2002;277:16639-47.
T et al. Vascular endothelial growth factor triggers signaling 26. Hideshima T, Podar K, Chauhan D, Ishitsuka K, Mitsiades C,

| 164 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

Tai YT et al. p38MAPK inhibition enhances PS-341 (borte- 36. Chauhan D, Anderson KC. Apoptosis in multiple myeloma:
zomib)-induced cytotoxicity against multiple myeloma cells. therapeutic implications. Apoptosis 2001;6:47-55.
Oncogene 2004;23:8766-76. 37. Richardson PG. Phase I study of the safety and efficacy of
27. Shammas MA, Shmookler Reis Dphil RJ, Li C, Koley H, Bortezomib in combination with Revlimid in relapsed and
Hurley LH, Anderson KC, Munshi NC. Telomerase inhibition refractory myeloma: The revvel study. Haematologica 2005;90
and cell growth arrest following telomestatin treatment in (s1):PL5.04.
multiple myeloma. Clin Cancer Res 2004;10:770-6. 38. Raje N, Kumar S, Hideshima T, Ishitsuka K, Chauhan D,
28. Tai YT, Teoh G, Chauhan D, Davies FE, Chauhan D, Treon SP Mitsiades C et al. Combination of the mTOR inhibitor
et al. Ku86 variant expression and function in multiple myelo- Rapamycine and Revlimid has synergistic activity in multiple
ma cells is associated with increased sensitivity to DNA dam- myeloma. Blood 2004;104:4188-93.
age. J Immunol 2009 [Epub ahead of print] 39. Frost P, Moatamed F, Hoang B, Shi Y, Gera J, Yan H et al. In
29. Podar K, Shringapure R, Tai YT, Simoncini M, Sattler M, vivo antitumor effects of the mTOR inhibitor CCI-779 against
Ishitsuka K et al. Caveolin-1 is required for VEGF-triggered human multiple myeloma cells in a xenograft model. Blood
multiple myeloma cell migration and is targeted by borte- 2004;104:4181-7.
zomib. Cancer Res 2004;64:7500-6. 40. Hideshima T, Catley L, Yasui H, Ishitsuka K, Raje N, Mitsiades
30. Frank DA. STAT signaling in the pathogenesis and treatment C et al. Perifosine, an oral bioactive novel alkyl-lysophospho-
of cancer. Mol Med 1999;5:432-56.
31. Hideshima T, Nakamura N, Chauhan D, Anderson KC. lipid, inhibits Akt and induces in vitro and in vivo cytotoxici-
Biologic sequelae of interleukin-6 induced PI3-K/Akt signaling ty in human multiple myeloma cells. Blood 2006;107:4053-62.
in multiple myeloma. Oncogene 2001;20:5991-6000. 41. Hideshima T, Bradner J, Wong J, Chauhan D, Richardson P,
32. Podar K, Raab MS, Zhang J, McMillin D, Breitkreutz I, Tai YT Schreiber SL, Anderson KC. Small molecule inhibition of pro-
et al. Targeting PKC in multiple myeloma: in vitro and in vivo teasome and aggresome function induces synergistic anti-
effects of the novel, orally available small-molecule inhibitor tumor activity in multiple myeloma: therapeutic implications.
Enzastaurin. Blood 2007 [Epub ahead of print] Proc Natl Acad Sci USA 2005;102:8567-72.
33. Davenport EL, Moore HE, Dunlop AS, Sharp SY, Workman P, 42. Chauhan D, Li G, Hideshima T, et al. Hsp27 overcomes borte-
Morgan GJ, Davies FE. Heat shock protein inhibition is associ- zomib/proteasome inhibitor PS-341 resistance in lymphoma
ated with activation of the unfolded protein response path- cells. Cancer Res 2003;63:6174-7.
way in myeloma plasma cells. Blood 2007;110:2641-9. 43. Chauhan D, Li G, Hideshima T, et al. Heat shock protein-27
34. Mitsiades CS, Mitsiades NS, McMullan CJ, Poulaki V, Kung confers drug resistance in multiple myeloma cells. Blood 2003;
AL, Davies FE et al. Antimyeloma activity of heat shock pro- 102:3379-86.
tein-90 inhibition. Blood 2006;107:1092-100. 44. Munshi N, Hideshima T, Carrasco R, Shammas M, Auclair D,
35. Chanan-Khan A, Richardson PG, Alsina M, et al. Phase I clin- Davies F et al. Identification of genes modulated in multiple
ical trial of KOS 953 + Bortezomib in relapsed refractory mul- myeloma using genetically identical twin samples. Blood
tiple myeloma. Blood 2005;106-9a. 2004;103:1799-806.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 165 |
Multiple myeloma

Treatment options for myeloma in 2009

J. Bladé A B S T R A C T
L. Rosiñol
The incorporation of novel drugs with new mechanisms of action is resulting in an improved out-
come in patients with multiple myeloma. In the context of ASCT, the new induction regimens result
Hematology and Oncology Institute,
Hematology Department. IDIBAPS, in a higher pre-transplant cytoreduction with a higher proportion of patients achieving CR after trans-
Hospital Clínic, University of plant, this leading to a longer progression-free survival when compared with previous studies. The
Barcelona. Barcelona, Spain Allo-RIC is a promising approach that deserves further investigation in prospective trials. Major
improvements are being accomplished in the treatment of elderly patients, particularly when combin-
ing melphalan/prednisone with thalidomide (MPT), bortezomib (MPV) or lenalidomide (MPR), or with
lenalidomide/dexamethasone. In the relapse setting, the availability of thalidomide, bortezomib and
Hematology Education:
the education program for the lenalidomide has resulted in more effective salvage regimens. A number of phase III trials by different
annual congress of the European national cooperative groups, both in eligible and non-eligible patients for ASCT, are ongoing.
Hematology Association Hopefully, the results of these trials will allow the selection of a more individualized anti-myeloma
therapy, likely based on the molecular genetic status.
2009;3:166-171

ntil recently, the median survival of benefit of ASCT in terms of EFS and OS.

U patients with multiple myeloma


(MM) has been about 3 years and the
number of long-term survivors disappoint-
The question of whether ASCT is beneficial
for the majority of patients or whether the
overall benefit results from certain sub-
ingly low. The introduction of high-dose groups of patients remains to be deter-
therapy followed by stem cell support mined. It seems that the achievement of CR
(HDT/SCT), and the incorporation of novel is the most important step for a long-lasting
agents (i.e., thalidomide, lenalidomide and response and prolonged survival in patients
bortezomib) are resulting in a higher anti- with MM.2,3 On the other hand, the sensitiv-
myeloma effect and prolonged survival. This ity to the initial chemotherapy measured by
improvement is due to a better initial thera- the M-protein type at the time of transplant
py for eligible and non-eligible patients for is the most reliable predictor of CR after
HDT/SCT and/or more effective rescue regi- ASCT.2,3 Supporting this concept, in a recent
mens for patients with relapsed/refractory meta-analysis by Van de Velde et al.,4 a
disease plus better supportive measures. This strong association between maximal
review focuses on: (i) treatment of patients response to the induction therapy and long-
eligible for HDT/SCT; (ii) treatment of non- term outcome was found. The pre-trans-
eligible patients for HDT/SCT; (iii) treatment plant induction therapy with different con-
of relapsed and/or refractory disease. ventional chemotherapy regimens has
resulted in a pre- and post-transplant
Treatment of patients eligible for high-dose ther- immunofixation negative CR of 5-10% and
apy/hematopoietic stem cell transplantation about 35%, respectively, as well as in a
Autologous stem cell transplantation median survival of approximately 6 years in
Autologous stem cell transplantation the best circumstances.
(ASCT) is considered the gold standard as
part of the front-line therapy in patients with Results with novel pre-transplant induction regi-
MM younger than 65 years. The results of mens
ASCT with induction with “conventional The ideal induction chemotherapy prior to
chemotherapy” and with induction incorpo- ASCT should consist of a regimen with high
rating new agents are summarized in the fol- anti-myeloma effect, no interference with
lowing sections. stem cell collection and acceptable toxicity.
The association of thalidomide/dexam-
Results with conventional chemotherapy induc- ethasone (TD) replaced VAD and was
tion regimens approved by the US Food and Drug
Five trials comparing ASCT versus con- Administration for its use as pre-transplant
ventional chemotherapy have been fully induction regimen. In addition, two large tri-
reported.1 Two of them showed that ASCT als showed that TD is superior to dexam-
was superior in complete remission (CR), ethasone alone.5,6 The overall response rate
event-free survival (EFS) and overall survival (ORR) to TD ranges from 58-76%. However,
(OS), while the remaining three showed no the CR rate is only about 5-7%. Furthermore,

| 166 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

Table 1. Post-induction and postransplant response in rate,16,18 three showed a prolonged EFS15,16,18 and only
patients with multiple myeloma eligible for ASCT. one, a significantly longer OS in favour of tandem trans-
plant.15 Surprisingly, a recently published trial19 compar-
Regimen CR before CR after ing single transplant followed by 6 months thalidomide
ASCT (%) ASCT (%) maintenance versus double ASCT showed that the sin-
gle transplant arm was superior to tandem transplant in
Old regimens both RR and survival.19 From two of these studies, it
Dexa/VAD 5 35 seems that patients who benefit from tandem trans-
Cyclophosphamide/Dexa 7 32
plant are only those failing to achieve at least very good
VBMCP/VBAD 10 35
New regimens
partial response (VGPR) with the first transplant.15,16 The
Thalidomide/Dexa5,6 5-10 44* most consistent finding of these studies is the prolonga-
Bortezomib/Dexa8,9 12 33 tion of EFS or PFS with double transplant.15,16,18,20 The role
Bortezomib/Adria/Dexa (PAD-1)11 24 43 of maintenance,21 as well as the concept of early intensi-
Bortezomib/Adria/Dexa (PAD-2)12 11 37 fication22 after ASCT with novel drugs are interesting
Bortezomib/Thal/Dexa (VTD)13,14 21-30 43-49 and deserve prospective further investigation.

Allogeneic stem cell transplantation


The best curative approach in multiple myeloma is
thalidomide and dexamethasone would not be an opti- allogeneic transplantation. However, allogeneic trans-
mal regimen for patients with extramedullary disease, plantation with myeloablative conditioning (MAC)
due to the lack of effect of thalidomide on soft-tissue results in a transplant-related mortality (TRM) up to
plasmacytomas.7 The results of two phase II trials com- 50%, and the proportion of patients long-term disease-
bining bortezomib and dexamethasone showed a post- free and possibly cured does not exceed 10-15%.23 For
induction response rate of about 65%, with 12% this reasons, allogeneic transplantation with MAC has
immunofixation negative CR.8,9 The ORR post-trans- almost universally been replaced by the so-called dose-
plant was about 90% with 33% CR. In a large phase III reduced intensity conditioning allogeneic transplanta-
trial by the Intergroup Francophone du Myelome (IFM) tion (Allo-RIC).23-26 The current conditioning consists of
comparing bortezomib/dexamethasone versus VAD, the fludarabine/MEL-140 or fludarabine/low-dose total
immunofixation negative CR rate post-transplant was body irradiation (TBI). The TRM has decreased by 10 to
significantly higher with bortezomib/dexamethasone 20%, whereas the CR rate is about 40-50%. The inci-
than with VAD (10% vs. 19%, p<0.01).10 Thalidomide dence of acute and chronic graft-versus-host disease
and bortezomib are increasingly used in combination (cGVHD) is 30-50%, respectively. The most important
with dexamethasone or antracyclines, resulting in the predictors of outcome are the development of cGVHD
so-called triple regimens. The PAD-1 regimen (borte- and chemosensitive disease with a low tumor burden at
zomib –PS 341-, adriamycin, dexamethasone) resulted in the time of transplant.23,24 Considering that an important
a pre-transplant ORR of 95% with 24% and in a post- requisite for the success of Allo-RIC is to have achieved
transplant CR rate of 43%.11 However, about 50% of the a low tumor mass at the time of transplantation, the use
patients developed peripheral neuropathy, leading to a of ASCT to reduce tumor burden followed by Allo-RIC
reduction in the dose of bortezomib to 1 mg/m2 (PAD-2) in order to benefit from graft-versus myeloma (GVM)
in a subsequent study from the same group.12 With PAD- effect has been recently investigated with promising
2, the incidence of neuropathy significantly decreased, results.24,26 In this sense, the efficacy of a tandem ASCT
while the ORR was still 89%. However, the CR rate versus single autograft, followed by Allo-RIC in patients
after induction and transplant were lower (11% and with newly diagnosed multiple myeloma with an avail-
37%, respectively) than with PAD-1.12 Cavo et al recent- able donor have been compared in three studies.27-29 The
ly reported that bortezomib-Velcade-/thalidomide/dex- IFM group reported no difference between the two
amethasone (VTD) was superior to TD in terms of CR approaches.27 In contrast, the Italian group found an
rate both before (21% vs. 6%) and after (43% vs. 23%) increased CR rate and a significant survival advantage in
transplant.13 The Spanish PETHEMA group is comparing favour of Allo-RIC with a survival plateau beyond 4
TD versus VTD versus combination chemotherapy with years of allografting, while the tandem ASCT group
vincristine, BCNU, melphalan, cyclophosphamide, pred- observed no plateau.28 In a Spanish PETHEMA study,
nisone (VBMCP)/vincristine, BCNU, adriamycin and there were no significant differences in EFS and OS
dexamethasone (VBAD) (four cycles) plus two cycles of between the two groups. However, the curve of
bortezomib as pretransplant induction therapy.14 The patients who received an Allo-RIC showed an encour-
preliminary results show that the best regimen is VTD aging survival plateau beyond 3 years of follow-up.29
with a pre-transplant and post-transplant CR rate of With the current data, it is evident that there is a need
30% and 49%, respectively.14 Table 1 shows a summary for continuing the investigation of conditioning intensi-
of the results before and after transplant achieved with ty and pos-transplant strategies aimed at enhancing the
the so-called “old” and “new” induction regimens. GVM effect while minimizing the GVHD.30

Results of single vs. tandem double autologous stem cell Treatment of patients not eligible for high-dose
transplant therapy/hematopoietic stem cell transplantation
Five prospective randomized studies comparing single In patients not eligible for HDT/ASCT (older than 65
versus double ASCT have been conducted.15-19 Two tri- years or younger with comorbidities), the conventional
als showed a significant increase in CR or near CR therapy has consisted of alkylating-based regimens,

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 167 |
14th Congress of the European Hematology Association

Table 2. Front-line therapy studies for patients with myeloma not eligible for HDT/SCT.

Author, yr Regimen ORR (%) CR (%) EFS OS

Rajkumar et al.6 THAL/DEX vs. DEX 63 vs. 46 7.7 vs. 2.6 22.6 vs. 6.5 29 mos vs. not reached
Facon et al.32 MPT vs. MP 76 vs. 35 13 vs. 2 HR 0.56 (in favour of MPT) 51.6 vs. 33.2 mos.
Hulin et al.33 MPT vs. MP 62 vs. 31 7 vs. 1 24.1 vs 19 mos. 45.3 vs. 27.7 mos.
Palumbo et al.34 MPT vs MP 76 vs. 47 15.5 vs. 2.4 54 vs. 27% at 2 yrs 80 vs. 64% at 3 yrs
San Miguel et al.38 MPV vs. MP 71 vs. 36 30 vs. 4 24 vs. 16.6 mos. 82.6 vs 69.5% at 2 yrs
Palumbo et al.39 MPR 81 24 92% at 1 yr 100% at 1 yr
Ludwig et al.40 THAL/DEX vs. MP 68 vs. 51 14 vs. 7 43 vs. 25 mos. 58 vs. 45 mos.
Rajkumar et al.41 Len/Dex vs Len/dex 82 vs. 70- - 87 vs. 75% at 2 yrs
MP: melphalan/prednisone; MPT: melphalan/prednisone/thalidomide; MPV: melphalan/prednisone/bortezomib; THAL/DEX: thalidomide/dexamethasone;
LEN/DEX: lenalidomide/dexamethasone; LEN/dex: lenalidomide/low dose dexamethasone; MPR: melphalan/prednisone/lenalidomide; HR: hazard ratio;
Main toxicities: MP: myelosuppresion; MPT: myelosupresion, peripheral neuropathy, DVT; MPV: myelosupresion, thrombocytopenia, peripheral neuropathy;
THAL/DEX: DVT; LEN/DEX or LEN/dex: myelosupresion, DVT; MPR: myelosupresion, DVT.

In a phase II trial, the association of MP with borte-


Table 3. General considerations in the treatment of
relapsed myeloma. zomib (MPV) showed encouraging results.37 In a subse-
quent large phase 3 trial, MPV was compared with MP
Components of initial therapy – VISTA trial. 38 The ORR (71 vs. 36%), the CR rate (30
Alkylating-based vs. 4%), the PFS (median 24 vs. 16.6 months) and the OS
Dexamethasone-based (82.6% and 69.5% at 2 years follow-up) were all in
Hdt/scs favour of MPV.38 MPV was superior to MP in all prog-
Efficacy of initial therapy nostic subgroups, including patients with poor cytoge-
Degree of response netics.
Timing of relapse In a phase II study, the association of MP with
Patient status and circumstances of relapse lenalidomide – Revlimid – (MPR) was very promising,
Age, ps with 81% PR rate, including 24% immnofixation nega-
Aggressive vs “non-aggressive” relapse tive CR.39 The EFS and OS at 1 year was 92%.39 The EFS
and OS at 1 year were 92% and 100%, respectively. A
large phase III trial comparing MPR versus MP has been
mainly melphalan and prednisone (MP), dexametha- recently completed.
sone-based therapies – VAD or VAD-like regimens – or Ludwig et al.40 compared TD versus MP and found a
dexamethasone alone. With these regimens, the ORR significantly higher CR rate (68% vs. 51%). However,
was about 50% with less than 5% CR with median sur- the number of deaths during the first year was signifi-
vivals of about 3 years. With the incorporation of novel cantly higher with TD, and there was a trend towards a
agents, with different mechanisms of action than con- superior EFS and a significantly longer OS in favour of
ventional therapy, an improvement in response rate and the MP arm.40 A large ECOG trial compared lenalido-
generally in survival are consistently reported.31 mide/dexamethasone at full doses of dexamethasone
The French IFM group compared MP versus two versus lenalidomide/dexamethasone with a weekly
cycles of VAD, followed by two courses of intermediate dose of 40 mg of dexamethasone.41 Although the RR
high-dose therapy with MEL-100 versus MP-thalido- was lower with low-dose dexamethasone,41 the OS at 1
mide (MPT) in patients aged 65-75 years.32 The MPT and 2 years was significantly longer with lenalido-
regimen resulted in a significantly higher ORR and CR mide/low-dose dexamethasone because a significantly
rate, as well as in significantly longer EFS and OS. The lower toxicity and mortality.
same group compared MP versus MPT (with a dose of From the above, it seems that the association of an
thalidomide of 100 mg/day) in patients older than 75 old regimen, such as MP or dexamethasone with a new
years.33 The RR was significantly higher with MPT agent (i.e., thalidomide, bortezomib or lenalidomide) is
(62% vs. 31%). Importantly, the PFS (median 24.1 vs. 19 becoming the treatment of choice in the elderly. It also
months) and the OS (median 45.3 vs. 27.7 months) were appears that MP combinations are resulting in higher
significantly longer with MPT.33 The Italian group also RR than dexamethasone-based combinations. Concern-
reported a significantly higher ORR and CR rate, as well ing MPT versus MPV, the results in RR and survival are
as a longer PFS in favour of MPT versus MP.34 However, very similar. Once they are approved for their use as up-
the OS was not significantly different. The Nordic and front therapy, the first choice will depend on age,
the HOVON groups found a significantly longer PFS in aggressiveness of the disease, cytogenetics and patient
favour of MPT versus MP, with no differences in OS.35,36 preference. For patients with very advanced age or less
Peripheral neuropathy is the main concern in thalido- aggressive disease, MPT would be preferable. In con-
mide-containing regimens and the risk of deep vein trast, for patients aged 65-75 years with clinically
thrombosis (DVT)/pulmonary embolism makes the use aggressive disease (extensive symptomatic bone
of prophylactic anticoagulation mandatory when involvement, extramedullary spread) or with poor cyto-
thalidomide is combined with cytotoxic agents or with genetics, a bortezomib containing regimen could be
high-dose glucocorticoids. more appropriate. For patients with peripheral neuropa-

| 168 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

Table 4. Randomized trials on treatment of relapsed/refractory myeloma.

Author, yr Regimen ORR (%) CR (%) EFS OS

Richardson et al., 2005 APEX trial48 Bort. vs. Dex 38 vs. 18 6 vs. 1 6.2 vs. 3.5 80 vs. 66% at 1 yr
Orlowski et al., 2007 49
Bort.+Doxil vs. Bort. 44 vs. 41 4 vs. 2 9.3 vs. 6.5 76 vs. 65% at 15 months
Weber et al., 2007 50
Len/Dex vs. Dex 61 vs. 19.9 14.1 vs. 0.6 11.1 vs. 4.7 29.6 vs. 20.2 months
Dimopoulos et al., 200751 Len/Dex vs. Dex 60.2 vs. 24 15.9 vs. 3.4 11.3 vs. 4.7 Not reached vs. 20.6 months
ORR: overall response rate; CR: complete response; TTP: time to progression; OS: overall survival; Bort: bortezomib; Dex: dexamethasone; Len: lenalidomide

thy, a lenalidomide-based regimen (lenalidomide/low being reported. However, these studies have a number
dose dexamethasone or MPR) would be preferable. For of shortcomings, such as low number of patients, possi-
patients with renal failure, bortezomib/dexamethasone ble favorable patient selection, short follow-up, consid-
would be the initial treatment of choice.42-44 Table 2 erable toxicity, cost and limited rescue options at
shows a summary of the recent trials in the treatment of relapse. The authors of this review are inclined to use a
MM in patients not eligible for HDT/SCT. sequential therapy approach in subsequent relapses.
The selection of therapy will depend on the previous
Treatment of relapsed/refractory myeloma drug exposure, previous response, toxicity and type of
In patients with relapsed or refractory myeloma, the relapse (aggressive vs. “indolent”). In patients with
choice of the salvage therapy must depend on: (i) the chemosensitive relapse, ASCT as rescue therapy should
components of the initial therapy; (ii) the degree and be considered. Also, in patients relapsing after ASCT, a
duration of response to primary therapy (i.e., deep second HDT/SCT could be of benefit, provided that the
responses with duration longer than 2 years, relapsing duration of response with the first ASCT was longer
off therapy might benefit from pre-treatment with the than 2 years. If there is an available HLA-matched
initial option); (iii) performance status and age at the donor and the patient has chemosensitive relapse and is
time of relapse (older patients with poor performance younger than 65 years, an Alli-RC should be consid-
status should be treated with more gentle approaches); ered.23 There is an extensive review by the International
(iv) type of relapse (aggressive relapses should be treat- Myeloma Working Group on the prophylaxis of throm-
ed with bortezomib-containing regimens, while more botic events in thalidomide and lenalidomide contain-
“indolent” relapses might be treated with thalidomide ing regimens.52
or lenalidomide-based regimens, saving bortezomib for Two recent studies53,54 have shown a significant
subsequent relapses); (v) previous toxicity (i.e., patients improvement in the survival of patients with multiple
with significant peripheral neuropathy avoid thalido- myeloma since the introduction of novel agents.
mide and bortezomib) (Table 3). Although there is still a long way to go, the current data
A recent review on thalidomide therapy in MM already represent a significant advance and a real hope
emphasizes that in patients with refractory disease, the for patients with multiple myeloma.
RR to single agent thalidomide is between 30 and 40%,
that thalidomide/dexamethasone combinations induce This work has been supported in part by grants
RR ranging from 40-65% and that soft-tissue plasmacy- 06/0020/0005 and 08/0147 from Instituto Carlos III
tomas rarely respond to thalidomide.45 (Spanish Institute of Health).
In the SUMMIT study, bortezomib produced a
response rate of 35% in patients with advanced refrac-
tory myeloma.46 The APEX trial showed that borte- References
zomib was superior to dexamethasone in RR, PFS and
OS.47 An extended follow-up of this study showed that 1. Bladé J, Rosiñol L. Advances in therapy of multiple myeloma.
Current Opin Oncol 2008;20:697-704.
the OR and CR rate of bortezomib was 43 and 9%, 2. Alexanian R, Weber D, Giralt S, Dimopoulos M, Delasalle K,
respectively and that higher response quality was asso- Smith T, Champlin R et al. Impact of complete remission with
ciated with longer response duration.48 A large phase 3 intensive therapy in patients with responsive multiple myelo-
ma. Bone Marrow Transplant 2001;27:1037-43.
trial comparing bortezomib associated with pegylated 3. Nadal E, Giné E, Bladé J, steve J, Rosiñol L, Fernández-Avilés F
liposomal doxorubicin versus bortezomib alone et al. High-dose therapy/autologous stem cell transplantation
showed that the combination was superior in CR and in patients with chemosensitive multiple myeloma: predictors
VGPR, PFS and OS.49 of complete remission. Bone Marrow Transplant 2004;33:61-
4.
The combination of lenalidomide and dexamethasne 4. Van de Velde HJK, Liu X, Chen G, Cakana A, Deraedt W,
results in an ORR of about 60%, including 15% CR ver- Bayssas M. Complete response correlates with long-term sur-
sus 20-24%, ORR with 1-4% CR with dexamethasone vival and progression-free survival in high-dose therapy in
multiple myeloma. Haematologica 2007;92:1399-406.
alone.50,51 Compared with dexamethasone, the combina- 5. Rajkumar SV, Blood E, Vesole D, Fonseca R, Greipp PR. Phase
tion of lenalidomide and dexamethasone was superior III clinical trial of thalidomide plus dexamethasone compared
not only in RR but also in TTP and OS50,51 (Table 4). with dexamethasone alone in newly diagnosed multiple
myeloma: a clinical trial coordinated by the Eastern Co-
An increasing number of three and four drug combi- operative Group. J Clin Oncol 2006;24:431-6.
nations, including “old” and novel agents, showing a 6. Rajkumar SV, Rosiñol L, Hussein M, Catalano J, Jedrzejczak
high RR in patients with relapsed/refractory disease are W, Lucy L et al. A multicenter, randomized, double-blind,
placebo-controlled study of thalidomide plus dexamethasone

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 169 |
14th Congress of the European Hematology Association

versus dexamethasone as inicial therapy for newly diagnosed allogeneic transplamtation is associated with a high incidence
multiple myeloma. J Clin Oncol 2008;26:2171-7. of extramedullary relapses in multiple myeloma patients.
7. Rosiñol L, Cibeira MT, Bladé J, Esteve J, Aymerich M, Rozman Leukemia 2006;20:542-5.
M et al. Escape of extramedullary disease to the thalidomide 26. Crawley C, Iacobelli S, Björkstrand B, Apperley JF, Nieder-
effect in multiple myeloma. Haematologica 2004;89:832-6. wieser D, Gahrton G et al. Reduced-intensity conditioning for
8. Harousseau JL, Attal M, Leleu X, Troncy J, Pegourie B, Stoppa myeloma: lower nonrelapse mortality but higher relapse rates
AM et al. Bortezomib plus dexamethasone as induction treat- compared with myeloablative conditioning. Blood 2007;109:
ment prior to autologous stem cell transplantation in patients 3588-94.
with newly diagnosed multiple myeloma. Haematologica 27. Garban F, Attal M, Michallet M, Hulin C, Bourhis JH, Yakoub-
2006;91:1498-505. Agha I et al. Prospective comparison of autologous stem cell
9. Rosiñol L, Oriol A, Mateos MV, Sureda A, García-Sánchez P, transplantation followed by dose-reduced allograft (IFM99-03
Gutiérrez N et al. A phase II trial of alternating bortezomib trial) in high-risk de novo multiple myeloma. Blood 2006;107:
and dexamethasone as induction regimen prior autologous 3474-80.
stem cell transplantation in younger patients with multiple 28. Bruno B, Rotta M, Patriarca F, Mordini N, Allione B, Carne-
myeloma: efficacy and clinical implications of tumour vale-Schianca F et al. A comparison of allografting with auto-
response kinetics. J Clin Oncol 2007;25:4452-8. grafting in newly diagnosed myeloma. N Engl J Med 2007;
10. Harousseau JL, Mathiot C, Attal M, Troncy J, Pegourie B, 356:1110-20.
Stoppa AM et al. Velcade/dexamethasone versus VAD as 29. Rosiñol L, Pérez-Simón JA, Sureda A, de la Rubia J, de Arriba
induction treatment prior to autologous stem cell transplanta- F, Lahuerta JJ et al. A prospective PETHEMA study of tandem
tion in newly diagnosed multiple myeloma: updated results of autologous transplantation versus autograft followed by
the IFM 2005/01 trial. J Clin Oncol 2008;26:8505. reduced-intensity conditioning allogeneic transplantation in
11. Oakervee HE, Popat R, Curry N, Smith P, Morris C, Drake M newly diagnosed multiple myeloma. Blood 2008;112:3591-3.
et al. PAD combination therapy (PS-341/bortezomib, doxoru- 30. Cavo M. Nonmyeloablative allotransplantation for myeloma:
bicin and dexamethasone) for previously untreated patients light and shade. Blood 2007;109:3134-5.
with multiple myeloma. Br J Haematol 2005;129:755-62. 31. San Miguel JF, Harousseau JL, Joshua D, Anderson K. Individ-
12. Popat R, Oakervee HE, Curry N, et al. Long-term follow-up of ualizing treatment of patients with myeloma in the era of
PAD for untreated multiple myeloma. Haematologica 2007; novel agents. J Clin Oncol 2008;26:2761-6.
92:725. 32. Facon T, Mary JY, Hulin C, Benboubker L, Attal M, Pegourie B
13. Cavo M, Tachetti P, Patriarca F, et al. Superior complete et al. Melphalan and prednisone plus thalidomide versus mel-
response rate and progression-free survival after autologous phalan and prednisone or reduced-intensity autologous stem
transplantation with up-front Velcade-thalidomide-dexam- cell transplantation in elderly patients with multiple myeloma.
ethasone (VTD) compared with thalidomide-dexamethasone Lancet 2007;370:1209-18.
(TD) in newly diagnosed multiple myeloma. Blood 2008; 112: 33. Hulin C, Facon T, Rodon P, et al. Comparison of melphalan-
65. prednisone-thalidomide (MP-T) demonstrates a significant
14. Rosiñol L, Cibeira MT, Martínez J et al. Thalidomide/dexam- survival advantage in elderly patients > 75 years with multiple
ethasone (TD) vs. bortezomib (Velcade)/thalidomide/dexam- myeloma compared with melphalan-prednisone (MP) in a ran-
ethasone (VTD) vs. VBMCP /VBAD /Velcade as induction reg- domized double-blind, placebo controlled trial, IFM 01-01.
imen prior autologous ítem cell transplantation (ASCT) in Blood 2007;110;31a.
younger patients with multiple myeloma (MM): first results of 34. Palumbo A, Bringhen S, Liberati AM, Caravita T, Falcone A,
a prospective phase III PETHEMA/GEM trial: Blood 2008; Callea V et al. Oral melphalan, prednisone and thalidomide in
112:244. elderly patients with multiple myeloma: updated results of a
15. Attal M, Harousseau JL, Facon T, Guilhot F, Doyen C, Fuzibet randomized controlled trial. Blood 2008;112:3107-14.
JG et al. Single versus double autologous stem cell transplanta- 35. Waage A, Gimsing P, Juliusson G, et al. Melphlan-prednisone-
tion for multiple myeloma. N Engl J Med 2003; 49:2495-502. thalidomide to newly diagnosed patients with multiple
16. Cavo M, Tosi P, Zamagni E, Cellini C, Tacchetti P, Patriarca F myeloma: a placebo-controlled phase III trial. Blood 2007;110:
et al. Prospective, randomized study of single compared with 32a.
double autologous transplantation for multiple myeloma. J 36. Wiejermans P, Schaafsme M, van Norden et al. Melphalan +
Clin Oncol 2007;25:2434-41. prednisone+thalidomide in induction therapy for multiple
17. Fermand JP. High-dose therapy supported with autologous myeloma in elderly patients: final analysis of the Dutch
blood stem cell transplantation in multiple myeloma: long- Cooperative Group HOVON 49 Study. Blood. 2008;112:241.
term follow-up of prospective studies of the MAG group. 37. Mateos MV, Hernández JM, Hernández MT, Gutiérrez NC,
Haematologica 2005;90:40. Palomera L, Fuertes M et al. Bortezomib plus melphalan and
18. Sonneveld P, van der Holt B, Segeren CM, Vellenga E, prednisone in elderly untreated patients with multiple myelo-
Croockewit AJ, Verhoe GE et al. Intermediate-dose melphalan ma: up-dated time-to-events results and prognostic factors for
compared with myeloablative treatment in multiple myelo- time to progression. Haematologica 2008;93:560-5.
ma: long-term follow-up of the Dutch Cooperative Group 38. San Miguel JF, Schlag R, Khuageva N, et al. A phase 3 study
HOVON 24 trial. Haematologica 2007;92:928-35. comparing bortezomib-melphalan-prednisone (VMP) with
19. Abdelkefi A, Ladeb S, Torjman L, Othman TB, Lakhal A, melphalan-prednisone (MP) in newly diagnosed multiple
Romdhane NB et al. Single autologous stem-cell transplanta- myeloma. Blood 2007;110:31a.
tion followed by maintenance therapy with thalidomide is 39. Palumbo A, Falco P, Corradini P, Falcone A, Di Raimondo F,
superior to double autologous transplantation in multiple Giuliani N et al. Melphalan, prednisone, and lenalidomide
myeloma: results of a multicenter randomized clinical trial. treatment for newly diagnosed myeloma: a report from the
Blood 2008;111:1805-10. GIMEMA-Italian Multiple Myeloma Network. J Clin Oncol
20. Kumar A, Kharfan-Dabaja MA, Glasmacher A, Djulbegovic B. 2007;25:4459-65.
Tandem versus single autologous hematopoietic stem cell 40. Ludwig H, Tothova E, Hajek R, Drach J, Adam Z, Labar B et
transplantation for treatment of multiple myeloma: a meta- al. Thalidomide-dexamethasone compared to melphalan-
analysis of randomized controlled trials. Blood 2007;110:264a. prednisolone in elderly patients with multiple myeloma.
21. Attal M, Harousseau JL, Leyvraz S, Doyen C, Hulin C, Blood 2008; Oct 27 [Epub ahead of print]
Benboubker L et al. Maintenance therapy with thalidomide 41. Rajkumar SV, Jacobus S, Callander N, et al. A randomized trial
improves survival in multiple myeloma patients. Blood 2006; of lenalidomide plus high-dose dexamethasone versus
108:3289-94. lenalidomide plus low-dose dexamethasone in newly diag-
22. Ladetto M, Pagliano G, Avonto I, et al. Consolidation with nosed multiple myeloma (E4A03): a trial coordinated by the
bortezomib, thalidomide and dexamethasone induces molec- Eastern Cooperative Oncology Group. J Clin Oncol 2008;26:
ular remissions in autografted multiple myeloma patients. 42. Chanan-Khan AA, Kaufmann JL, Metha J, Richardson PG,
Blood 2007;110:163a. Miller KC, Lonial S et al. Activity and safety of bortezomib in
23. Bensinger W. Stem-cell transplantation for multiple myeloma multiple myeloma patients with advanced renal failure: a mul-
in the era of novel drugs. J Clin Oncol 2008;26;480-92. ticenter retrospective study. Blood 2007;109;2604-6.
24. Crawley C, Lalancette M, Szydio R, Gilleece M, Peggs K, 43. Kastritis E, Anagnostopoulos A, Roussou M, Gika D,
Mackinnon S et al. Outcomes for reduced-intensity allogeneic Matsouka C, Barmparousi D et al. Reversibility of renal failure
transplantation for multiple myeloma: an analysis of prognos- in newly diagnosed multiple myeloma patients treated with
tic factors from the Chronic Leukemia Working Party of the high dose dexamethasone and the impact of novel agents.
EBMT. Blood 2005;105:4532-9. Haematologica 2007;92:546-9.
25. Pérez-Simón JA, Sureda A, Fernández-Avilés F, Cabrera JR, 44. Ludwig H, Drach J, Graf H, Land A, Meran JG. Reversal of
Caballero D, Martino R et al. Reduced-intensity conditioning acute renal failure by bortezomib-based chemotherapy in

| 170 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

patients with multiple myeloma. Haematologica 2007;92: tion therapy improves time to progression. J Clin Oncol 2007;
1411-4. 25:3892-901.
45. Palumbo A, Facon T, Sonneveld, Bladè J, Offidani M, Gay F et 50. Weber D, Chen C, Niesvizky R, Wang M, Belch A, Stadt-
al. Thalidomide for treatment of multiple myeloma: 10 years mauer EA et al. Lenalidomide plus dexamethasone for
later. Blood 2008;111:3968-77. relapsed multiple myeloma in North America. N Engl J Med
46. Richardson PG, Barlogie B, Berenson J, Singhal S, Jagannath S, 2007;357:2133-42.
Irwin D et al. A phase 2 study of bortezomib in relapsed, 51. Dimopoulos M, Spencer A, Attal M, et al. Lenalidomide plus
refractory myeloma. N Engl J Med 2003;348:2609-17. dexamethasone for relapsed or refractory multiple myeloma.
47. Richardson P, Sonneveld P, Schuster MW, et al. Bortezomib or N Engl J Med 2007;357:2123-32.
high-dose dexamethasone for relapsed multiple myeloma. N 52. Palumbo A, Rajkumar SV, Dimopoulos MA, Richardson PG,
Engl J Med 2005;352:2487-98. San Miguel J, Barlogie B et al. Prevention of thalidomide- and
48. Richardson PG, Sonneveld P, Schuster M, Irwin D, Stadtmauer lenalidomide-associated thrombosis in myeloma. Leukemia
E, Facon T et al. Extended follow-up of a phase 3 trial in 2008;22:414-23.
relapsed multiple myeloma: final time-to-event results of the 53. Brenner H, Gondos A, Pulte D. Recent major improvement in
APEX trial. Blood 2007;110:3557-60. long-term survival of younger patients with multiple myelo-
49. Orlowski RZ, Nagler A, Sonneveld P, Bladé J, Hajek R, Spencer ma. Blood. 2008;111:2521-6.
A et al. Randomized phase III study of pegylated liposomal 54. Kumar SK, Rajkumar SV, Dispenzieri A, Lacy MQ, Hayman
doxorubicin plus bortezomib compared with bortezomib SR, Buadi FK et al. Improved survival in multiple myeloma and
alone in relapsed or refractory multiple myeloma: combina- the impact of novel therapies. Blood 2008;111:2516-20.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 171 |
Myelodysplastic syndromes

Pathogenesis and basic aspects of myelodysplastic


syndromes

N. Galili A B S T R A C T
A. Raza
The inherent heterogeneity of myelodysplastic syndromes (MDS) makes it difficult to obtain a sim-
St. Vincent’s Comprehensive plified description of the pathobiology leading to disease initiation and evolution of this universally
Cancer Center, New York, USA fatal disorder. However, it has become evident, that MDS is a result of an abnormal hematopoietic cell
interacting with an increasingly abnormal microenvironment, thus disrupting the delicate balance
Hematology Education: between proliferative and apoptotic signals that control normal stem cell function. The hallmark of
the education program the MDS marrow, high proliferation in the presence of increased apoptosis, is reflective of the devel-
for the annual congress of the opment of monoclonality with the appearance of dysplasia and cytopenia(s). Excessive inflammatory
European Hematology Association cytokines from the MDS microenvironment stimulate angiogenesis, which helps to disseminate the
dysplastic cells. Epigenetic mechanisms coupled with genomic instability, lead to aberrant immune
2009;3:172-176 response and high risk of transformation. It is the complex interplay of these and other, as of yet,
unknown factors that underlie the fatal course of MDS as described here.

yelodysplastic syndromes, a hetero- more lineages. A breakthrough in the under-

M geneous group of clonal stem cell


diseases resulting in peripheral
cytopenias with dysplastic bone marrows,
standing of the biology of the MDS marrow
came with the demonstration that, while the
marrow cells were rapidly proliferating,
which primarily affects the elderly, has there was simultaneous excessive intra-
become one of the most diagnosed hemato- medullary cytokine mediated apoptosis.3-7
logical malignancies due to the increasingly Inflammatory cytokines, notably TNF-α and
aging population. Initially, this group of dis- TGF-β, secreted by the stromal cells of the
eases was thought to be a form of pre- marrow microenvironment, as well as by
leukemia, as many patients went on to devel- other immune reactive cells, have been espe-
op acute leukemia.1 It has become clear, cially implicated in a paracrine process.7
MDS is not solely a disease of the hemato- Additionally, levels of TNF-α ligand, as well
poietic stem cell, but is a result of complex as apoptosis signaling receptors FAS and
interplay between factors derived from the TRAIL are also increased on the MDS
stromal cells of the bone marrow microenvi- hematopoietic cells,8,9 inducing an autocrine
ronment and abnormal stem cells. Thus, component to apoptosis. Activation of the
MDS has become a model for the seed and mitochondrial caspase cascade is triggered
soil hypothesis of tumor development. by these signaling pathways, resulting in
Therefore, it is important to look at each apoptosis. The marrows of patients that
component of this intricate network in order present or evolve into higher risk MDS tend
to understand why, despite many common- to have lower levels of apoptosis than those
alities found in the various subtypes, the with lower risk MDS, while retaining similar
molecular basis of MDS pathology and ther- rates of proliferation,10 and thus, are more
apy is still evolving. likely to transform. Decreased apoptosis is
attributed to accumulating mutations in this
Clonality studies molecular pathway. Additional genetic
Clonality studies using chromosomal mutations with continuing proliferation may
abnormalities together with X-chromosome also explain the gradual increase in blast cells
inactivation demonstrated that MDS is a and the resulting resistance to chemotherapy
clonal disease which most typically origi- that characterizes AML derived from MDS.
nates in a pluripotent stem cell committed to
the myeloid lineage.2 Studies have shown, Immune response
however, that in some patients, the lym- The presence of an abnormal immune
phoid lineage is also part of the diseased response in patients with MDS was first sug-
clone, suggesting that in these patients the gested by the observation that patients with
originating MDS stem cell has not yet under- aplastic anemia (AA) may evolve to MDS
gone lineage differentiation.2 that has a trisomy 8 or monsomy 7 karyo-
type.11 Patients with AA respond to treat-
Proliferation, apoptosis and cytokines ment with immunosuppressive therapy,
The MDS marrow is filled with rapidly including anti-thymocyte globulin (ATG)
proliferating cells, yet paradoxically, the and cyclosporine. MDS patients, specifically
patients present with cytopenia in one or those with trisomy 8, showed hematologic

| 172 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

improvement when treated with these drugs, suggest- likely to be mutated during disease progression. The
ing that a T-cell mediated response was active in these BCL2 family of proteins tightly controls the apoptotic
patients. Studies later showed that MDS patients with pathway as it includes both pro and anti-apoptosis pro-
trisomy 8 have expanded CD8+ T-cells of one or more teins. Levels of the pro-apoptotic proteins are increased
T-cell receptor subfamilies, and that these cells actually in low risk patients when compared to high risk, while
inhibit trisomy 8 cells in culture.12 Surprisingly, patients levels of the anti-apoptotic proteins are increased in the
with this subtype of MDS rarely transform to AML high-risk patients.23 JAK2 mutations have been detected
with ATG therapy and their cytopenias improve. This in a high percentage of patients with myeloproliferative
suggests that the trisomy 8 MDS cells retain their abili- disease, but rarely in most patients with MDS.24 This
ty to mature and when cytotoxic T-cells are reduced or gene, a tyrosine kinase, activates the JAK-STAT signal
eliminated by ATG, cytopenia can recover. Eventually, transduction pathway controlling proteins essential for
however, continued genetic instability results in disease multiple biologic functions. Many cytokines and
progression. Deregulated cytokine balance (especially growth factors are regulated by this pathway. It has
TNFα), abnormal immune response in MDS and recently been shown that JAK2 mutation is characteris-
increase in angiogenesis (see below) led to the use of tic of a subset of MDS patients with refractory anemia
thalidomide in a clinical trial.13 Thalidomide is a strong with ringed sideroblasts and thrombocytosis.25,26 In con-
inhibitor of TNFα and, has numerous immumodulatory trast, FLT3, a stimulatory protein for hematopoietic cell
effects and decreases angiogenesis. Hematologic proliferation and differentiation often found (20-25%)
response was seen in 19% of all patients and 31% of as tandem repeats in AML, is mutated in only 1-3% of
evaluable patients. Subsequently, Lenalidomide, a deriv- MDS patients.27,28 However, when present, the MDS
ative of thalidomide, was shown to improve erythro- patient is likely to transform to AML. Thus, this muta-
cytopenia in transfusion dependant low-risk MDS tion may be an early indicator of poor prognosis and
patients,14-16 and was especially effective in patients hav- transformation. Mutations of RAS and AML1 genes
ing a 5q- karyotype. Lenalidomide has been shown to have been associated with -7 MDS, which has a partic-
modulate both NK cell and T-cell activity.17 ularly poor prognosis. AML1 is a transcription factor
associated with normal hematopoiesis that has been
Angiogenesis found to be mutated several types of leukemias, includ-
Angiogenesis is mediated by a number of growth fac- ing AML.29 These patients also show constitutive activa-
tors, including vascular endothelial growth factor tion of the STAT/STAT5 signal transduction pathway
(VEGF), TNF-α and TGF-β, and a notable increase in following treatment with granulocyte colony-stimulat-
vascularity is found in the marrow of patients with ing factor, although the connection to chromosome 7
MDS.18 The cytokines that control normal marrow remains unknown. A high incidence of AML1 muta-
angiogenesis are the very cytokines that balance prolif- tions has been found in high-risk MDS patients and is
erative and apoptotic signaling in normal hemato- thought to be implicated in transformation to AML in
poiesis; the pathways deregulated in MDS. It is possi- these patients.30,31 A number of other gene mutations,
ble, therefore, that in MDS, these cytokines act to stim- including EVI1, PTPN11 and PDGFR-A are often mutat-
ulate proliferation with excess apoptosis directly in the ed in AML and rarely in MDS.32-34 The commonality is
MDS hematopoietic cell, while simultaneously stimu- that they all are essential parts of proliferative, apoptot-
lating neo-angiogenesis to support the aberrant ic or differentiation pathways.
microenvironment and to disseminate the dysplastic
cells. In addition, it is important to note that these Epigenetics
cytokines are also part of the inflammatory response While genomic mutations have been recognized as a
pathway, and thus, the pathological effects in MDS may cause of malignant transformations since the discovery
be associated with an initiating inflammatory stimula- of the Philadelphia chromosome, other biochemical
tion that cascades into eventual monoclonality and dis- events can lead to changes in the DNA without disrupt-
ease progression. ing the primary nuclei acid sequence. This modification,
referred to as epigenetics, has a profound role in genet-
Specific gene mutations in myelodysplastic syndromes ic regulation and control of gene expression. Abnormal
TP53 mutations have been detected in 5-10% of MDS methylation of a gene has the same functional effect as
patients.19-21 Wild type TP53 plays a pivotal role in the a genetic mutation, and thus provides an additional
induction of apoptotic pathway, and mutations have mechanism for malignant transformation. Two
been found predominantly in high-risk MDS patients; hypomethylating agents, 5-azacitidine and decitabine,
an observation consistent with the decreased apoptosis have been approved by the FDA for treatment of
and high risk of transformation. In addition, TP53 muta- MDS.35-37 The mechanism of action is not clear; it has
tions are often associated with complex karyotypes in been assumed that silenced tumor suppressor genes are
MDS as part of chromosome 17 deletions or transloca- reactivated upon treatment. While a number of genes
tions. Since mutations of TP53 have been shown to have been shown to be hypermethylated in patients
cause genetic instability, it is possible that the complex with MDS, including the tumor suppressor gene, p15,
karyotype is a secondary event in the evolution of response to therapy could not be correlated with
advanced disease. NRAS has been reported to be abnor- increase in p15 gene expression.37 Using methylation-
mally activated in patients with MDS (5-20%).22 Point specific polymerase chain reaction to look at promoter
mutations result in constitutive activation of the gene. methylation of regulatory genes essential to the evolu-
The family of RAS genes control signaling pathways for tion of MDS, it was possible to identify gene-silencing,
proliferation, differentiation and apoptosis, and is thus specific for both cell lineage and differentiation states.38

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 173 |
14th Congress of the European Hematology Association

Other studies using high-resolution single nucleotide Table 1.


polymorphism (SNP) arrays have shown that genomic
methylation profiles may be useful for identifying direct Reference Patient samples Cell Type Results
or indirect targets of these drugs.39,40 Additionally, bio-
chemical modifications of the histone proteins compos-
ing the chromatin structure have been found to greatly Miyazato et al.45 RAEB AC133 MDS
alter the ability of transcriptional initiating complexes to and AML separated BM samples heterogeneous
assemble on gene promoters, and thus, are another hindering analysis;
form of epigentic control of gene expression. The access Dlk gene identified
to promoter DNA is influenced by the activities of his- as MDS specific
tone acetylating (HAC) enzymes, which relax the chro-
matin structure and histone deacetylating (HDAC) Ueda et al.46 all stages AC133 Identified poor vs.
MDS separated BM good prognosis genes
enzymes, which condense the chromatin. In theory, the
use of HDAC inhibitors should restore gene expression Hofmann et al.47 low risk CD34 Large number of
of normal genes (including tumor suppressor genes) that versus separated BM deregulate genes in MDS;
have been silenced during disease evolution. Clinical tri- high risk identified 11 genes
als with HDAC inhibitors are currently underway in as class predictors
patients with MDS.41
The development of high-resolution SNP arrays has Pellagatti et al.48 RA(5q-, Neutrophils High variability between
also enabled investigators to look at the DNA structure normal), MDS samples; few genes
of the largest sub-group of MDS patients, the 50% that AML commonly deregulated
have a normal karyotype.42-44 Surprisingly, a number of in MDS; 71 genes could
patients (10-15%) with no observable clonal abnormal- discriminate 5q-; only 1
ity by karyotype had DNA that contained regions of of 71 genes maps to
uniparental disomy (UPD; regions inherited from only CDR of 5q-
one parent resulting in a neutral loss of heterozygosity),
as well as small regions of amplification and/or dele- Chen et al.49 monsomy 7, CD34 separated BM; Small number
tions. While the UPDs were found to be constitutional, trisomy 8 pooled/individual commonly de-regulated
the amplifications and deletions were acquired and samples from genes; different pathways
associated with poorer disease outcome.43 These prelim- different patients implicated for each
inary studies indicate that perhaps, the patients with karyotype; very little
UPD have inherently increased genetic instability, and overlap with results
of other studies
thus, have an increased risk for developing MDS.
Further studies are needed to confirm these findings,
but if confirmed, they point to a method to screen for
individuals at high risk of developing MDS.

Gene expression studies mapped to the commonly deleted region or CDR of


DNA microarray technology offers an unprecedented 5q–. Chen et al.49 found a small number of deregulated
opportunity to define signature profiles and several genes common to monosomy 7 and trisomy 8, but dif-
early studies have attempted to do this (Table 1). The ferent cellular pathways were implicated in the profiles
first study45 compared MDS with excess blasts (RAEB) of each karyotype.
to cells of patients with de novo AML. Dlk gene encod- The 5q deletion is the most frequent cytogenetic
ing the protein δ-like gene was identified as MDS spe- abnormality in MDS and is found in 10-15% of patients.
cific. The individual variability of MDS samples, how- The haplo-deletion is highly variable encompassing
ever, made further analysis very difficult. In a second 5q31, but two smaller regions have been defined by
study,46 this same group used a transcriptosome approach molecular studies to be associated with clinical fea-
to examine differences between normal BM and those tures.50 In a more recent expression study,51 it was noted
of MDS patients at various stages of the disease, identi- that two ribosomal genes, RBM22 and RPS14, mapping
fying poor versus good prognosis genes, which were to the CDR, were significantly down-regulated. Since
thought to contribute to the progression of MDS. another ribosomal gene, RPS19, had already been impli-
Hoffman et al.47 compared CD34+ isolated BM cells of cated in the congenital anemia of Blackfan-Diamond
low risk versus high risk MDS to normal control samples. syndrome,52 this group suggested that deregulation of
While 11 genes were identified as class (risk) predictors, these genes, and the pathway that they were part of,
a large number of genes were found to be deregulated in could be the causative abnormality in 5q- syndrome
the MDS patients. Pellagatti et al.48 looked at the expres- patients. Shortly thereafter, using RNA inhibition,
sion profile of neutrophils isolated from the peripheral RPS14 was indeed shown to be a causal gene for the 5q-
blood of 21 MDS patients [7 with del(5q) karyotype], 2 syndrome.53 To support their hypothesis that deregulat-
AMLs and normal controls. Similar to the other studies, ed ribosomal and protein synthesis pathways are a fea-
expression was highly variable but several genes were ture of 5q- syndrome, Pellagatti et al.54 looked at the
commonly deregulated. Del(5q) patients could be dis- expression of 579 ribosomal and translation related
criminated from refractory anemia (RA) patients with genes in the CD34+ cells of 5q- syndrome patients com-
normal karyotypes on the basis of 71 genes. Interesting- pared to RA patients with normal karyotype and
ly, only 5 of the 71 genes mapped to 5q and only one healthy controls. Fifty-five genes associated with ribo-

| 174 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

some and protein synthesis were differentially 5. Raza A, Gezer S, Mundle S, Gao XZ, Alvi S, Borok R, et al.
Apoptosis in bone marrow biopsy samples involving stromal
expressed with the lowest expression levels found in and hematopoietic cells in 50 patients with myelodysplastic
the 5q- syndrome patients. The rapid proliferation of syndromes. Blood 1996;87:3064-70.
red blood cells requiring continuous protein synthesis 6. Yoshida Y, Anzai N, Kawabata H. Apoptosis in myelodyspla-
would make erythropoiesis highly sensitive to disrup- sia: a paradox or paradigm. Leuk Res 1995;19:887-91.
7. Raza A, Mundle S, Shetty V, Alvi S, Chopra H, Span L, et al.
tions in ribosome biosynthesis, and explains the severe Novel insights into the biology of myelodysplastic syndromes:
anemia seen in these patients. RPS14 haploinsufficiency excessive apoptosis and the role of cytokines. Int J Hematol
explains the paradox of anemia and thrombocytosis but 1996;63:265-78.
8. Claessens YE, Bouscary D, Dupont JM, Picard F, Melle J,
not the initial clonal expansion. Recent meta-analysis Gisselbrecht S, et al. In vitro proliferation and differentiation
comparing CD34+ expression profiles of 60 cases of of erythroid progenitors from patients with myelodysplastic
non-del(5q) MDS patients with 52 normal controls syndromes: evidence for Fas-dependent apoptosis. Blood
2002;99:1594-601.
using datasets from seven independent studies in the 9. Campioni D, Secchiero P, Corallini F, Melloni E, Capitani S,
NCBI GEO database showed that the most significant- Lanza F, et al. Evidence for a role of TNF-related apoptosis-
ly deregulated class of genes in the MDS group were the inducing ligand (TRAIL) in the anemia of myelodysplastic syn-
dromes Am J Pathol 2005;166:557-63.
ribosomal protein genes.55 Thus, deregulated expression 10. Albitar M, Manshouri T, Shen Y, Liu D, Beran M, Kantarjian
of genes in a specific functional pathway provides a uni- HM, al. Myelodysplastic syndrome is not merely “preleu-
fied mechanism for the pathology of a seemingly kemia”. Blood 2002;100:791-8.
11. Maciejewski JP, Selleri C. Evolution of clonal cytogenetic
diverse disease. abnormalities in aplastic anemia. Leuk Lymphoma 2004; 45:
Our group, together with the Broad Institute took a 433-40.
different bedside to bench approach to examine gene 12. Sloand EM, Mainwaring L, Fuhrer M, Ramkissoon S, Risitano
AM, Keyvanafar K, et al. Preferential suppression of trisomy 8
expression in patients with MDS.56 Studies showed that compared with normal hematopoietic cell growth by autolo-
lenalidomide treatment elicited transfusion independ- gous lymphocytes in patients with trisomy 8 myelodysplastic
ence in 67% of patients that had a del(5q) karyotype15 syndrome. Blood 2005;106:841-51.
and 26% of patients without del(5q).16 Using pre-thera- 13. Raza A, Meyer P, Dutt D, Zorat F, Lisak L, Nascimbene F, et al.
Thalidomide produces transfusion independence in long-
py mononuclear bone marrow cells, a signature of 32 standing refractory anemias of patients with myelodysplastic
genes, which are normally expressed during later stages syndromes. Blood 2001;98:958-65.
of erythroid differentiation, was found to be down reg- 14. List A, Kurtin S, Roe DJ, Buresh A, Mahadevan D, Fuchs D, et
al. Efficacy of lenalidomide in myelodysplastic syndromes. N
ulated in responders.56 Engl J Med 2005;352:549-57.
15. List A, Dewald G, Bennett J, Giagounidis A, Raza A, Feldman
MicroRNA studies E, et al. Myelodysplastic Syndrome-003 Study Investigators.
Lenalidomide in the myelodysplastic syndrome with chromo-
The expression of microRNAs has been shown to dif- some 5q deletion. N Engl J Med 2006;355:1456-65.
ferentiate between MDS subgroups.57 In patients with 16. Raza A, Reeves JA, Feldman EJ, Dewald GW, Bennett JM,
5q- syndrome, two microRNAs (miR-145 and miR-146a Deeg HJ, et al. Phase 2 study of lenalidomide in transfusion-
dependent, low-risk, and intermediate-1 risk myelodysplastic
that target TIRAP and TRAF6) mapping to the CDR syndromes with karyotypes other than deletion 5q. Blood
appear to have biologic relevance to the disease pathol- 2008;111:86-93.
ogy, specifically affecting the 5q- syndrome pheno- 17. Chang DH, Liu N, Klimek V, Hassoun H, Mazumder A, Nimer
SD, et al. Enhancement of ligand-dependent activation of
type.58 Although preliminary, these studies are paving human natural killer T cells by lenalidomide: therapeutic
the way for an entirely novel area of investigation. implications. Blood 2006;108:618-21.
18. Zorat F, Shetty V, Dutt D, Lisak L, Nascimben F, Allampallam
Conclusion K, et al. The clinical and biological effects of thalidomide in
patients with myelodysplastic syndromes. Br J Haematol
Multiple biologic pathways have been found to be 2001;115:881-94
deregulated in MDS. The heterogeneity of the disease 19. Padua RA, Guinn BA, Al-Sabah AI, Smith M, Taylor C,
precludes development of a single agent targeting a sin- Pettersson T, et al. RAS, FMS and p53 mutations and poor clin-
ical outcome in myelodysplasias: a 10-year follow-up. Leu-
gle pathway as therapy for all patients and accounts for kemia 1998;12:887-92.
the variation of response to a given therapy. New tech- 20. Kita-Sasai Y, Horiike S, Misawa S, Kaneko H, Kobayashi M,
nologies that predict response or individualize therapeu- Nakao M, et al. International prognostic scoring system and
TP53 mutations are independent prognostic indicators for
tic strategies are promising weapons in the battle with patients with myelodysplastic syndrome. Br J Haematol 2001;
MDS. Continued basic studies into the biology of dis- 115:309-12.
ease evolution may elucidate additional unifying char- 21. Wattel E, Preudhomme C, Hecquet B, Vanrumbeke M, Que-
snel B, Dervite I, et al. p53 mutations are associated with
acteristics that will eventually lead to new therapies for resistance to chemotherapy and short survival in hematologic
this devastating disease. malignancies. Blood 1994;84:3148-57.
22. Lu D, Nounou R, Beran M, Estey E, Manshouri T, Kantarjian
H, et al. The prognostic significance of bone marrow levels of
neurofibromatosis-1 protein and ras oncogene mutations in
References patients with acute myeloid leukemia and myelodysplastic
syndrome. Cancer 2003;97:441-9.
1. Block M, Jacobson LO, Bethard WF. Preleukemic acute human 23. Parker JE, Mufti GJ, Rasool F, Mijovic A, Devereux S, Pagliuca
leukemia. J Am Med Assoc 1953;152:1018-28. A. The role of apoptosis, proliferation, and the Bcl-2-related
2. Boultwood J, Wainscoat JS. Clonality in the myelodysplastic proteins in the myelodysplastic syndromes and acute myeloid
syndromes. Int J Hematol 2001;73:411-5. leukemia secondary to MDS. Blood 2000;96:3932-8.
3. Hatfill SJ, Fester ED, Steytler JG. Apoptotic megakaryocyte 24. Kralovics R, Passamonti F, Buser AS, Teo SS, Tiedt R, Passweg
dysplasia in the myelodysplastic syndromes. Hematol Pathol JR, et al. A gain-of-function mutation of JAK2 in myeloprolif-
1992;6:87-93. erative disorders. N Engl J Med 2005;352:1779-90.
4. Raza A, Mundle S, Iftikhar A, Gregory S, Marcus B, Khan Z, et 25. Steensma DP, Dewald GW, Lasho TL, Powell HL, McClure RF,
al. Simultaneous assessment of cell kinetics and programmed Levine RL, et al. The JAK2 V617F activating tyrosine kinase
cell death in bone marrow biopsies of myelodysplastics mutation is an infrequent event in both “"atypical”" myelopro-
reveals extensive apoptosis as the probable basis for ineffec- liferative disorders and myelodysplastic syndromes. Blood
tive hematopoiesis. Am J Hematol 1995;48:143-54. 2005;106:1207-9.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 175 |
14th Congress of the European Hematology Association

26. Wang SA, Hasserjian RP, Loew JM, Sechman EV, Jones D, Hao imbalance by single-nucleotide polymorphism analysis in
S, et al. Refractory anemia with ringed sideroblasts associated low-risk myelodysplastic syndromes Blood 2007;110:3365-73.
with marked thrombocytosis harbors JAK2 mutation and 43. Gondek LP, Tiu R, O'Keefe O’Keefe CL, Sekeres MA, Theil
shows overlapping myeloproliferative and myelodysplastic KS, et al. Chromosomal lesions and uniparental disomy
features. Leukemia 2006;20:1641-4. detected by SNP arrays in MDS, MDS/MPD, and MDS-
27. Horiike S, Yokota S, Nakao M, Iwai T, Sasai Y, Kaneko H, et derived AML Blood 2008;111:1534-42.
al. Tandem duplications of the FLT3 receptor gene are associ- 44. Nowak D, Nolte F, Mossner M, Nowak V, Baldus CD, Hopfer
ated with leukemic transformation of myelodysplasia. O, et al. Genome-wide DNA-mapping of CD34+ cells from
Leukemia 1997;11:1442-6. patients with myelodysplastic syndrome using 500K SNP
28. Shih LY, Huang CF, Wang PN, Wu JH, Lin TL, Dunn P, et al. arrays identifies significant regions of deletion and uniparental
Acquisition of FLT3 or N-ras mutations is frequently associat- disomy. Exp Hematol 2009;37:215-24.
ed with progression of myelodysplastic syndrome to acute 45. Miyazato A, Ueno S, Ohmine K, Ueda M, Yoshida K, Yama-
myeloid leukemia. Leukemia 200418:466-75. shita Y, et al. Identification of myelodysplastic syndrome-spe-
29. Niimi H, Harada H, Harada Y, Ding Y, Imagawa J, Inaba T, et cific genes by DNA microarray analysis with purified
al. Hyperactivation of the RAS signaling pathway in hematopoietic stem cell fraction. Blood 2001;98:422-7.
myelodysplastic syndrome with AML1/RUNX1 point muta- 46. Ueda M, Ota J, Yamashita Y, Choi YL, Ohki R, Wada T, et al.
tions. Leukemia 2006;20:635-44. DNA microarray analysis of stage progression mechanism in
30. Harada H, Harada Y, Niimi H, Kyo T, Kimura A, Inaba T. High
incidence of somatic mutations in the AML1/RUNX1 gene in myelodysplastic syndrome. Br J Haematol 2003;123:288-96.
myelodysplastic syndrome and low blast percentage myeloid 47. Hofmann WK, de Vos S, Komor M, Hoelzer D, Wachsman W,
leukemia with myelodysplasia. Blood 2004;103:2316-24. Koeffler HP. Characterization of gene expression of CD34+
31. Nakao M, Horiike S, Fukushima-Nakase Y, Nishimura M, cells from normal and myelodysplastic bone marrow. Blood
Fujita Y, Taniwaki M, et al. Novel loss-of-function mutations 2002;100:3553-60.
of the haematopoiesis-related transcription factor, acute 48. Pellagatti A, Esoof N, Watkins F, Langford CF, Vetrie D,
myeloid leukaemia 1/runt-related transcription factor 1, Campbell LJ, et al. Gene expression profiling in the myelodys-
detected in acute myeloblastic leukaemia and myelodysplastic plastic syndromes using cDNA microarray technology Br J
syndrome Br J Haematol 2004;125:709-19. Haematol 2004;125:576-83.
32. Raza A, Buonamici S, Lisak L, Tahir S, Li D, Imran M, et al. 49. Chen G, Zeng W, Miyazato A, Billings E, Maciejewski JP,
Arsenic trioxide and thalidomide combination produces multi- Kajigaya S, et al. Distinctive gene expression profiles of CD34
lineage hematological responses in myelodysplastic syn- cells from patients with myelodysplastic syndrome character-
dromes patients, particularly in those with high pre-therapy ized by specific chromosomal abnormalities. Blood 2004; 104:
EVI1 expression. Leuk Res 2004;28:791-803. 4210-8.
33. Tartaglia M, Niemeyer CM, Fragale A, Song X, Buechner J, 50. Boultwood J, Fidler C, Strickson AJ, Watkins F, Gama S,
Jung A, et al. Somatic mutations in PTPN11 in juvenile myelo- Kearney L, et al. Narrowing and genomic annotation of the
monocytic leukemia, myelodysplastic syndromes and acute commonly deleted region of the 5q- syndrome. Blood 2002;
myeloid leukemia. Nat Genet 2003;34:148-50. 99:4638-41.
34. Christiansen DH, Desta F, Andersen MK, Pedersen-Bjergaard J. 51. Boultwood J, Pellagatti A, Cattan H, Lawrie CH, Giagounidis
Mutations of the PTPN11 gene in therapy-related MDS and A, Malcovati L, et al. Gene expression profiling of CD34+ cells
AML with rare balanced chromosome translocations. Genes in patients with the 5q- syndrome. Br J Haematol 2007; 139:
Chromosomes Cancer 2007;46:517-21. 578-89.
35. Silverman LR, Demakos EP, Peterson BL, Kornblith AB, 52. Gazda HT, Grabowska A, Merida-Long LB, Latawiec E,
Holland JC, Odchimar-Reissig R, et al. Randomized controlled Schneider HE, Lipton JM, et al. Ribosomal protein S24 gene is
trial of azacitidine in patients with the myelodysplastic syn- mutated in Diamond-Blackfan anemia. Am J Hum Genet
drome: a study of the Cancer and Leukemia Group B. J Clin 2006;79:1110-8.
Oncol 2002;20:2429-40. 53. Ebert BL, Pretz J, Bosco J, Chang CY, Tamayo P, Galili N, Raza
36. Kantarjian H, Oki Y, Garcia-Manero G, Huang X, O'Brien A, Root DE, Attar E, Ellis SR, Golub TRet al. Identification of
Kantarjian H, Oki Y, et al. Results of a randomized study of 3 RPS14 as a 5q- syndrome gene by RNA interference screen.
schedules of low-dose decitabine in higher-risk myelodysplas- Nature 2008;450:335-40.
tic syndrome and chronic myelomonocytic leukemia. Blood 54. Pellagatti A, Hellström-Lindberg E, Giagounidis A, Perry J,
2007;109:52-7. Malcovati L, Della Porta MG, et al. Haploinsufficiency of
37. Kantarjian H, Oki Y, Garcia-Manero G, Huang X, O'Brien RPS14 in 5q- syndrome is associated with deregulation of
SO’Brien S, Cortes J, et al. Results of a randomized study of 3 ribosomal- and translation-related genes. Br J Haematol 2008;
schedules of low-dose decitabine in higher-risk myelodysplas- 142:57-64.
tic syndrome and chronic myelomonocytic leukemia. Blood
2007;109:52-7. 55. Sohal D, Yeatts A, Ye K, Pellagatti A, Zhou L, Pahanish P, et al.
38. Hopfer O, Komor M, Koehler IS, Schulze M, Hoelzer D, Thiel Meta-analysis of microarray studies reveals a novel hemato-
E, et al. DNA methylation profiling of myelodysplastic syn- poietic progenitor cell signature and demonstrates feasibility
drome hematopoietic progenitor cells during in vitro lineage- of inter-platform data integration. PLoS ONE 2008;3:e2965.
specific differentiation. Exp Hematol 2007;35:712-23. 56. Ebert BL, Galili N, Tamayo P, Bosco J, Mak R, Pretz J, et al. An
39. Figueroa ME, Reimers M, Thompson RF, Ye K, Li Y, Selzer RR, erythroid differentiation signature predicts response to
et al. An integrative genomic and epigenomic approach for the lenalidomide in myelodysplastic syndrome. PLoS Med 2008;
study of transcriptional regulation. PLOS 2008;3:e1882 5:e35.
40. Kerkel K, Spadola A, Yuan E, Kosek J, Jiang L, Hod E, et al. 57. Gaken J, Mohamedali AM, Twine N, Westwood N,
Genomic surveys by methylation-sensitive SNP analysis iden- Czepulkowski B, Chehade S et al. MicroRNA Expression
tify sequence-dependent allele-specific DNA methylation. Nat Profiling of High and Low Risk MDS. Blood. (ASH Annual
Genet 2008;40:904-8. Meeting Abstracts) 2008;112;[Abstract 3645].
41. Griffiths EA, Gore SD. DNA methyltransferase and histone 58. Daniel T, Starczynowski DT, Kuchenbauer F, Argiropoulos B,
deacetylase inhibitors in the treatment of myelodysplastic Sung S, Morin R, et al. Identification of Mir-145 and Mir-146a
syndromes. Semin Hematol 2008;45:23-30. as Micrornas Involved in the Pathogenesis of 5q- Syndrome.
42. Mohamedali A, Gäken J, Twine NA, Ingram W, Westwood N, Blood. (ASH Annual Meeting Abstracts) 2008;112;[Abstract
Lea NC, et al. Prevalence and prognostic significance of allelic 853].

| 176 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Myelodysplastic syndromes

Molecular pathogenesis of myelodysplastic


syndromes: identifying new therapeutic targets
based on biology

S.D. Nimer A B S T R A C T

We are making slow but steady progress in advancing our knowledge of the molecular defects that
Division of Hematologic Oncology underlie acute leukemia and the myelodysplastic syndromes (MDS). MDS has been more difficult to
and Sloan-Kettering Institute,
Memorial Sloan-Kettering Cancer understand because of its heterogeneity. A common feature is morphologic evidence of dysplasia
Center, New York, NY, USA within the blood and marrow compartments. The basis for this and other features of the disease is
unclear. Furthermore, the significant immunologic abnormalities identified in this disease, coupled
Hematology Education: with the mixture of MDS stem cells within the dysplastic progenitor cells and normal stem and pro-
the education program genitor cells in the marrow, has made it difficult to molecularly characterize this disease. Additionally,
for the annual congress of the the lack of “MDS” cell lines and the difficulty growing MDS stem or progenitor cells in vitro or in
European Hematology Association mouse xenotransplantation studies have hampered our understanding of this disease.

2009;3:177-180

lthough the cause of MDS is usually t(3;21)(q26;q22) (also seen in patients with

A unknown, it can occur following


exposure to environmental toxins, to
alkylating agent or topoisomerase II
blast crisis CML), which rearranges the
AML1 and EVI-1 genes, fusing the N termi-
nus of AML1 with nearly all of the
inhibitor therapy that is used to treat a pri- MDS1/EVI-1 protein. Both components of
mary malignancy. Indeed, further investiga- the AML1-MDS1/EVI-1 fusion protein
tion of MDS arising in these settings may be appear to play critical roles in normal
more fruitful in the years to come. hematopoiesis, and both are required for the
Secondary MDS caused by these agents has unique effects of the fusion protein. The
two distinct time courses and often-distinct AML1 gene (now known officially as
cytogenetic abnormalities. These include the RUNX1) is involved in a variety of chromo-
loss of all or part of chromosomes 5 and 7 somal translocations, mutations and dele-
seen following alkylating agent therapy, and tions. Translocations involving RUNX1 (and
abnormalities (generally translocations) chromosome 21q22) are found in specific
involving the chromosome 11q23 region subtypes of acute leukemias, while point
seen in patients exposed to topoisomerase mutations or deletions are found in the M0
inhibitors leading to fusion of the MLL gene subtype of AML and in MDS, in particular in
with one of its more than 70 partner genes.1 therapy-related MDS.2,3 These mutations
The genes affected by chromosomal inter- generally serve as loss of function mutants,
stitial deletions or numerical changes in but some have the potential to serve as dom-
MDS have escaped characterization, even inant negatives, affecting the function of the
though the recurring cytogenetic abnormali- remaining wild type AML1 protein. RUNX1
ties have been known for decades (e.g., 5q-, expression is required for the development
7q-, 20q-, trisomy 8). Thus, for most MDS of definitive hematopoiesis in the mouse,
patients, the molecular basis of their disease and RUNX1 regulates lymphocyte develop-
is not known. However, in addition to obvi- ment, progenitor cell numbers, and
ous cytogenetic abnormalities in MDS, vari- megakaryocytic differentiation in the adult
eties of modern molecular studies of genom- mouse.4
ic integrity (e.g., SNP arrays) have shown While the role of AML1/RUNX1 in AML
other recurrent genomic abnormalities (e.g., and MDS is widely appreciated, the biology
small regions of chromosome gain or loss of the EVI-1 gene, located on 3q26, is not.
and uniparental disomy). Recently, we have The EVI-1 gene encodes both the MDS1-
gleaned much information about certain EVI-1 and EVI-1 proteins via differential pro-
genes that may be critically involved in moter usage. EVI-1, but not MDS1-EVI-1, is
MDS, and hopefully, we can use this knowl- transforming and can block hematopoietic
edge to rapidly develop better therapeutic differentiation, especially of the erythroid
approaches for patients with this disease. lineage.5 EVI-1 contains zinc finger domains
that interact with DNA and others involved
Cytogenetic abnormalities in protein-protein interactions. MDS1-EVI-1
One of the first chromosomal transloca- but not EVI-1 contains a PR domain, a region
tions described in MDS patients is the shown to contain methyltransferase activity

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 177 |
14th Congress of the European Hematology Association

in other proteins. EVI-1 has been shown to impair may silence key components of signal transduction,
GATA-1 function via protein-protein interaction and apoptosis, cell cycle progression, or DNA repair gene
this has been postulated to play a critical role in the inhi- pathways. Hypomethylating agents like decitabine (5-
bition of erythroid differentiation. However, EVI-1 has aza-2’-deoxycytidine) and 5-Azacytidine covalently
also been reported to interact with RUNX1, blocking its bind to the DNA methyltransferase enzymes, irre-
binding to DNA.6 Furthermore, EVI-1 overexpression versibly inhibiting their function. This results in the pro-
can cooperate with certain mutant forms of RUNX1 to gressive loss of methylation, which can trigger changes
generate AML from MDS.7 MDS1-EVI-1 has been in gene expression and induce cellular differentiation.
shown to function as a transcriptional repressor and to The ability of these agents to induce the DNA damage
interact with several methyltransferases, such as G9a,8 response has also been shown. Thus, the basis for the
Suv39H1,9 CtBP,10 and MBD3,11 a member of the Mi2- effectiveness of action of 5-azacitidine and decitabine in
NuRD repressor complex. MDS is not really known, nor is the reason why some
PRDM16 (aka MEL1) is an EVI-1 like gene12 and it is patients respond and others do not respond.
also involved in a translocation with RUNX1.13 Hypomethylating agents are being combined with
PRDM16 also interacts with the CtBP repressor com- histone deacetylase inhibitors in an attempt to affect
plex, which itself interacts with the LSD1 histone gene expression synergistically. For combination thera-
demethylase (LSD1= lysine specific demethylase, a H3- pies to synergize optimally, the dynamics of the
K4 demethylase).14 Thus, both the RUNX1-MDS1-EVI1 processes being inhibited needs to be considered.
and the RUNX1-MEL1 (PRDM16) fusion proteins may Histone acetylation is a rapidly reversible process,
function, at least in part, by changing the normal H3-K4 whereas DNA methylation is felt to be irreversible.
methyl activation mark to a demethylated H3-K4 mark. Thus far, histone methyltransferase inhibitors have not
This would promote the H3-K9 methylation mark and been clinically tested. Histone methylation was thought
contribute to gene repression. If so, restoration of H3- to be irreversible until multiple site-specific demethylases
K4 trimethylation, and inhibition of LSD1 and H3-K9 were reported (e.g. JMJD6 and LSD1, which act on
methylation, could be a therapeutic approach for EVI-1 methyl arginine and methyl lysine, respectively). Thus,
related, and AML1-MDS-EVI-1 related MDS or AML. both the methyltransferases and the demethylases rep-
resent potential targets.
Epigenetic deregulation in myelodysplastic syndromes MLL, NSD1, and NSD3 all have methyltransferase
In general, alterations in histone marks and DNA activity and all are involved in translocations in myeloid
methylation may also be the hallmarks of MDS and cer- disorders. These proteins contain a SET domain [named
tain other types of malignances. Given the success of because it is found in the Su(var)3-9; E(z); and Trithorax
DNA hypomethylating agents in treating MDS patients, proteins]. SET domains are found in several other can-
a clearer understanding of how gene expression is dys- cer-related proteins, including the EZH2 gene which is
regulated in this disease is essential. Therefore, under- overexpressed in prostate cancer,15 the MMSET gene
standing how transcription factors (such as RUNX1, which is overexpressed in multiple myeloma,16 and the
EVI-1, PU.1, HoxA9) regulate normal (and malignant) MLL gene. Generally, the SET domains contain the his-
hematopoiesis will be a focus of this report. Potential tone methyltransferase (HMT) activity within a protein.
candidate MDS genes affecting chromatin structure are A second class of HMTs has also been identified; they
being actively studied, as loss of these genes could have contain a PR domain rather than a SET domain.
pleiotropic effects and result in the epigenetic propaga- MLL methylates lysine 4 in histone H3, a mark gener-
tion of gene expression abnormalities from the MDS ally associated with gene activation. The region of MLL
initiating cell to its progeny. Yet, in addition to under- protein lost in the 11q23 translocation includes the SET
standing gene transcriptional regulation, a strong work- domain, which contains its methyltransferase enzymat-
ing knowledge of transcriptional elongation, post-tran- ic activity. Yet, in most cases, other activating histone
scriptional modification of protein function and riboso- marks are still found on MLL regulated genes (e.g.,
mal biogenesis are necessary to understand fully how H3K79me on the HoxA9 gene promoter) suggesting that
candidate genes contribute to MDS biology. the MLL-fusion proteins still activate gene expression,
Silencing of gene expression has been largely ascribed although they may have lost their ability to fine tune
to the processes of histone deacetylation, DNA promot- gene expression.
er hypermethylation and histone methylation. Thus,
the enzymes that catalyze these processes represent Other processes implicated in myelodysplastic syndromes
potential therapeutic targets. They include: (i) the his- The molecular basis of 5q-MDS is being explored,
tone deacetylases (HDACs), which remove acetyl especially because two-thirds of transfusion dependent
groups from lysine residues in histones; (ii) the DNA patients with 5q- MDS respond to lenalidomide, losing
methyltransferases, which physically interact with their transfusion dependency and achieving major or
HDAC complexes and catalyze methylation at CpG complete cytogenetic responses.17 Many genes have
islands within the promoters of critical growth regulato- been suggested to play a role in 5q-MDS via haploinsuf-
ry genes; and (iii) the histone methyltransferases ficiency, a mechanism that has been invoked because
(HKMTs and PRMTs) that methylate arginine or lysine deletion of candidate genes has not been accompanied
residues within the tails or globular domains of the his- by mutations in the remaining allele. RPS14 is a candi-
tones and promote a closed chromatin conformation date 5q- gene,18 as is SPARC, a protein secreted by cells
and heterochromatin formation. Aberrant methylation into the extracellular matrix.19 So are α-catenin, an intra-
of cytosines within CpG promoter regions by DNA cellular protein that may regulate cell polarity and adhe-
methyltransferases (primarily DNMT1 and DNMT3b) sion,20 and the cell cycle regulator cdc25c, which con-

| 178 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

trols the movement of cells from G2 to M. Furthermore, expression of RUNX1 (AML1) point mutant proteins in
the EGR1 transcriptional regulator, which can function retrovirally transduced bone marrow cells.7 This model
as a tumor suppressor gene, and the NPM gene, which has shown the potential synergism between EVI-1 over-
encodes a nuclear-cytoplasmic shutting protein that reg- expression and loss of RUNX1 function.
ulates the p53-p14ARF pathway, also represent intrigu-
ing candidates.21,22 However, not all of these genes are Conclusions
located within the commonly deleted region (the CDR) Much remains to be defined in the molecular patho-
on chromosome 5q. If multiple genes must be lost to genesis of MDS. The syndromes are characterized by
generate the 5q-phenotype, then the current approach defects in differentiation and defects in proliferation and
of studying genes one at a time will need adaptation in apoptosis, yet the path to their more successful treat-
order to describe the myelodysplastic syndromes fully. ment is not clear. Further study of the MDS stem or ini-
Point mutations in activating transcription factors tiating cell is essential, as is a better understanding of the
(like AML1) have been found in MDS patients, but acti- role of inflammatory mediators, and immune effector
vated kinases, such as JAK2 or RAS, have also been cells within the bone marrow microenvironment in
found. In fact activating RAS mutations may be more inhibiting hematopoiesis. Only then will the defects
frequent in patients with AML1 point mutations. The that lead to the cytopenias and the predisposition to
RAS point mutations found in MDS patients generally develop AML be understood.
involve codon12 of H-RAS or N-RAS. In addition to
RAS mutations, loss of NF1 or PTPN11, proteins,
downregulate RAS signaling are found in CMML (or References
juvenile CMML). Translocations that activate tyrosine
kinases have also been reported in MDS, but these tend 1. Krivtsov AV, Armstrong SA. MLL translocations, histone mod-
ifications and leukaemia stem-cell development. Nat Rev
to be present in patients with more proliferative fea- Cancer 2007;7:823-33.
tures to their disease. Thus, the t(5;12) that generates a 2. Harada H, Harada Y, Tanaka H, Kimura A, Inaba T. Implica-
TEL-PDGFRβ fusion protein with constitutive kinase tions of somatic mutations in the AML1 gene in radiation-
associated and therapy-related myelodysplastic syndro-
activity, frequently accompanies CMML with eosino- me/acute myeloid leukemia. Blood 2003;101:673-80.
philia. Like the TEL-PDGFRβ fusion, the JAK2V617F 3. Osato M. Point mutations in the RUNX1/AML1 gene: anoth-
mutation, when found in MDS, is associated with spe- er actor in RUNX leukemia. Oncogene 2004;23:4284-96.
4. Ichikawa M, Asai T, Saito T, Seo S, Yamazaki I, Yamagata T,
cific clinical syndromes, such as RARS with thrombocy- et al. AML-1 is required for megakaryocytic maturation and
tosis23 or in some patients with 5q- MDS.24 More than lymphocytic differentiation, but not for maintenance of
50% of patients with the RARS-T syndrome may have hematopoietic stem cells in adult hematopoiesis. Nat Med
an activating JAK2 mutation; the frequency, in part, 2004;10:299-304.
5. Kreider BL, Orkin SH, Ihle JN. Loss of erythropoietin respon-
depends on the threshold platelet count used to define siveness in erythroid progenitors due to expression of the Evi-
the syndrome. 1 myeloid-transforming gene. Proc Natl Acad Sci USA 1993;
90:6454-8.
Mouse models of myelodysplastic syndromes 6. Senyuk V, Sinha KK, Li D, Rinaldi CR, Yanamandra S,
Nucifora G. Repression of RUNX1 activity by EVI1: a new role
Several mouse models of MDS now exist. One such of EVI1 in leukemogenesis. Cancer Res 2007;67:5658-66.
model is the heterozygous NPM +/- mouse, which 7. Watanabe-Okochi N, Kitaura J, Ono R, Harada H, Harada Y,
Komeno Y, et al. AML1 mutations induced MDS and
develops a myelodysplastic syndrome similar to refrac- MDS/AML in a mouse BMT model. Blood 2008; 111:4297-
tory anemia (RA), with prominent dyserythropoiesis 308.
and erythroid hyperplasia in the bone marrow. These 8. Spensberger D, Delwel R. A novel interaction between the
mice survive to 1 year of age, comparable to their wild proto-oncogene Evi1 and histone methyltransferases,
SUV39H1 and G9a. FEBS Lett 2008;582:2761-7.
type littermates; transformation to acute leukemia has 9. Cattaneo F, Nucifora G. EVI1 recruits the histone methyltrans-
now been reported. While NPM is mutated and aber- ferase SUV39H1 for transcription repression. J Cell Biochem.
rantly located the cytoplasm in 40% of AML cases with 2008;105:344-52.
10. Palmer S, Brouillet JP, Kilbey A, Fulton R, Walker M, Crossley
normal cytogenetics,25,26 NPM mutations are generally M, et al. Evi-1 transforming and repressor activities are medi-
not found in patients with MDS.25 ated by CtBP co-repressor proteins. J Biol Chem 2001;276:
Another MDS model is based on transplanting 25834-40.
11. Spensberger D, Vermeulen M, Le Guezennec X, Beekman R,
murine bone marrow cells that have been transduced van Hoven A, Bindels E, et al. Myeloid transforming protein
with an EVI-1 expressing retrovirus. After a significant Evi1 interacts with methyl-CpG binding domain protein 3 and
period of latency, the recipient mice develop a syn- inhibits in vitro histone deacetylation by Mbd3/Mi-2/NuRD.
Biochemistry 2008;47:6418-26.
drome that mimics MDS,27 with significant erythroid 12. Mochizuki N, Shimizu S, Nagasawa T, Tanaka H, Taniwaki
defects, including greatly impaired maturation. These M, Yokota J, et al. A novel gene, MEL1, mapped to 1p36.3 is
mice do not have increased blasts, nor do they develop highly homologous to the MDS1/EVI1 gene and is transcrip-
acute leukemia. tionally activated in t(1;3)(p36;q21)-positive leukemia cells.
Blood 2000;96:3209-14.
The NUP98-HOXD13 transgenic mouse model may 13. Sakai I, Tamura T, Narumi H, Uchida N, Yakushijin Y, Hato T,
be particularly useful for studying the development and et al. Novel RUNX1-PRDM16 fusion transcripts in a patient
progression of MDS. These mice develop refractory with acute myeloid leukemia showing t(1;21)(p36;q22). Genes
Chromosomes Cancer 2005;44:265-70.
anemia with excess blasts (RAEB) that can be fatal. Mice 14. Wang J, Scully K, Zhu X, Cai L, Zhang J, Prefontaine GG, et al.
that do not die of cytopenias go on to develop AML.28 Opposing LSD1 complexes function in developmental gene
Thus, this model seems to recapitulate the human dis- activation and repression programmes. Nature 2007;446:882-
7.
ease best. 15. Varambally S, Dhanasekaran SM, Zhou M, Barrette TR,
Another MDS model that demonstrates both an MDS Kumar-Sinha C, Sanda MG, et al. The polycomb group protein
phase and progression to AML is one based on the EZH2 is involved in progression of prostate cancer. Nature
2002;419:624-9.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 179 |
14th Congress of the European Hematology Association

16. Chesi M, Nardini E, Lim RS, Smith KD, Kuehl WM, Bergsagel 22. Grisendi S, Bernardi R, Rossi M, Cheng K, Khandker L,
PL. The t(4;14) translocation in myeloma dysregulates both Manova K, et al. Role of nucleophosmin in embryonic devel-
FGFR3 and a novel gene, MMSET, resulting in IgH/MMSET opment and tumorigenesis. Nature 2005;437:147-53.
hybrid transcripts. Blood 1998;92:3025-34. 23. Ceesay MM, Lea NC, Ingram W, Westwood NB, Gäken J,
17. List A, Dewald G, Bennett J, Giagounidis A, Raza A, Feldman Mohamedali A, et al. The JAK2 V617F mutation is rare in
E, et al. Lenalidomide in the myelodysplastic syndrome with RARS but common in RARS-T. Leukemia 2006;20:2060-1.
chromosome 5q deletion. Myelodysplastic Syndrome-003 24. Ingram W, Lea NC, Cervera J, Germing U, Fenaux P, Cassinat
Study Investigators. N Engl J Med 2006;355:1456-65. B, et al. The JAK2 V617F mutation identifies a subgroup of
18. Ebert BL, Pretz J, Bosco J, Chang CY, Tamayo P, Galili N, et al. MDS patients with isolated deletion 5q and a proliferative
Identification of RPS14 as a 5q- syndrome gene by RNA inter- bone marrow. Leukemia 2006;20:1319-21.
ference screen. Nature 2008;451:335-9. 25. Falini B, Mecucci C, Tiacci E, Alcalay M, Rosati R, Pasqualucci
19. Pellagatti A, Jädersten M, Forsblom AM, Cattan H, Christens-
L, et al. Cytoplasmic nucleophosmin in acute myelogenous
son B, Emanuelsson EK, et al. Lenalidomide inhibits the malig-
nant clone and up-regulates the SPARC gene mapping to the leukemia with a normal karyotype. GIMEMA Acute
commonly deleted region in 5q- syndrome patients. Proc Natl Leukemia Working Party. N Engl J Med 2005; 352:254-66.
Acad Sci USA 2007;104:11406-11. 26. Grisendi S, Pandolfi PP. NPM mutations in acute myelogenous
20 Liu TX, Becker MW, Jelinek J, Wu WS, Deng M, Mikhalkevich leukemia. N Engl J Med 2005;352:291-2.
N, et al. Chromosome 5q deletion and epigenetic suppression 27. Buonamici S, Li D, Chi Y, Zhao R, Wang X, Brace L, et al. EVI1
of the gene encoding alpha-catenin (CTNNA1) in myeloid cell induces myelodysplastic syndrome in mice. J Clin Invest 2004;
transformation. Nat Med 2007;13:78-83. 114:713-9.
21. Joslin JM, Fernald AA, Tennant TR, Davis EM, Kogan SC, 28. Lin YW, Slape C, Zhang Z, Aplan PD. NUP98-HOXD13 trans-
Anastasi J, et al. Haploinsufficiency of EGR1, a candidate gene genic mice develop a highly penetrant, severe myelodysplastic
in the del(5q), leads to the development of myeloid disorders. syndrome that progresses to acute leukemia. Blood 2005;106:
Blood 2007;110:719-26. 287-95.

| 180 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Myelodysplastic syndromes

Risk-adapted treatment of myelodysplastic syndromes

M. Cazzola A B S T R A C T
L. Malcovati
Myelodysplastic syndromes (MDS) are usually characterized by clonal proliferation of hematopoiet-
ic cells, which partly retain their capacity to differentiate and maturate, but do so in an inefficient
Department of Hematology
Oncology, University of Pavia manner. Their clinical heterogeneity is best illustrated by the observation that these disorders range
Medical School, Pavia, Italy from indolent conditions with a near-normal life expectancy to forms approaching acute myeloid
leukemia (AML). A risk-adapted treatment strategy is mandatory for conditions showing a so highly
Hematology Education: variable clinical course, and definition of the individual risk has been based so far on the use of prog-
the education program nostic scoring systems. We have developed a prognostic model that accounts for the WHO categories,
for the annual congress of the cytogenetics and transfusion dependency. This WHO classification-based prognostic scoring system
European Hematology Association (WPSS) is able to classify patients into five risk groups showing different survivals and probabilities of
leukemic evolution. WPSS predicts survival and leukemia progression at any time during follow-up,
2009;3:181-187 and may therefore be used for implementing risk-adapted treatment strategies. The approach to a
patient with myelodysplastic syndrome should always begin with a period of observation, with
sequential peripheral blood counts – and sometimes bone marrow examinations – to assess the rate
of progression, if any. Several therapeutic tools have been proposed in the last decades but only few
survived the evidence-based criteria of efficacy. Patients with very low WPSS risk do not need any
treatment and can be just followed regularly. Transfusion dependent patients with low WPSS risk can
be treated with recombinant human erythropoietin. Responsive patients are mainly those with early
disease, inadequate endogenous erythropoietin production and low need for blood transfusion.
Transfusion-dependent patients with myelodysplastic syndrome associated with del(5q) may respond
to lenalidomide with cytogenetic remission and abolishment of transfusion requirement. However, in
2008 the EMEA Committee for Medicinal Products for Human Use was of the opinion that the bene-
fits of lenalidomide in the treatment of anemia of MDS with del(5q) did not outweigh its potential
risks, and therefore the drug has not been approved for this use in Europe. A recent survival study com-
paring azacitidine versus conventional care showed that treatment with azacitidine increases overall
survival in patients with higher-risk MDS. Thus, EMEA has recently approved azacitidine for the treat-
ment of adult patients with high-risk myelodysplastic syndrome who are not eligible for allogeneic
hematopoietic stem cell transplantation. The only treatment that can cure a patient with myelodys-
plastic syndrome is still allogeneic stem cell transplantation. It can be estimated that approximately
one third of patients receiving an allogeneic transplantation are cured with this treatment, but only a
minority of all MDS patients are eligible and have a donor.

yelodysplastic syndromes (MDSs) may be difficult to assess in a clinical set-

M are usually characterized by clonal


proliferation of hematopoietic
cells, which partly retain their capacity to
ting, and – as stated in the World Health
Organization (WHO) classification4 –
MDSs remain among the most challenging
differentiate and maturate, but do so in an of the myeloid neoplasms for proper diag-
inefficient manner.1 The bone marrow is nosis and classification.
generally hypercellular while peripheral
blood cells are variably reduced (cytope- Diagnosis and classification
nia), and several morphological abnormali- MDSs were defined and classified in 1982
ties are observed in these two compart- by the French American British (FAB)
ments.2 In addition, clones of hematopoiet- group.5 In 2001, the WHO classification of
ic cells carrying non-random cytogenetic myeloid neoplasms was developed.6 This
abnormalities are typically found in these classification has been updated recently,7
disorders.3 With time, there is a progressive and the 2008 WHO classification of myelo-
impairment in the capacity of hematopoiet- dysplastic syndromes/neoplasms is report-
ic cells to differentiate and maturate; blast ed in Table 1.
cells may appear in the bone marrow and The current diagnostic approach includes
peripheral blood, and there is an increasing- peripheral blood and bone marrow mor-
ly high risk of evolution to overt acute phology (to evaluate abnormalities of peri-
myeloid leukemia (AML). The basic com- pheral blood cells and hematopoietic pre-
ponents of the definition of MDS are clon- cursors), bone marrow biopsy (to assess
ality, ineffective hematopoiesis and pre- marrow cellularity, fibrosis and topogra-
leukemic nature. These features, however, phy), and cytogenetics (to identify non-ran-

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 181 |
14th Congress of the European Hematology Association

Table 1. WHO classification of myelodysplastic syndromes Table 2. WHO classification-based Prognostic Scoring
and peculiar entities. System (WPSS) and bone marrow fibrosis as criteria for
predicting likelihood of survival and risk of leukemic evo-
Disease lution in the individual patient with myelodysplastic syn-
drome.
2008 WHO classification
Calculation of the WPSS score and evaluation of bone marrow fibrosis
Refractory anemia (RA), refractory neutropenia (RN), refractory thrombocytopenia
Variable Variable scores
(RT), refractory cytopenia with unilineage dysplasia (RCUD)

Refractory anemia with ringed sideroblasts (RARS) 0 1 2 3

Refractory cytopenia with multilineage dysplasia (RCMD) WHO category RA, RARS, MDS RCMD RAEB-1 RAEB-2
with isolated
Refractory anemiawith excess blasts-1 (RAEB-1) deletion (5q)

Refractory anemia with excess blasts-2 (RAEB-2) Karyotype* Good Intermediate Poor −

Myelodysplastic syndrome, unclassified (MDS-U) Transfusion Absent Present − −


requirement**
Myelodysplastic syndrome associated with isolated del(5q)
Bone marrow The presence of grade 2 to 3 bone marrow fibrosis involves
fibrosis*** a shift to a one-step more advanced risk group after accounting
Peculiar entities for WHO category, karyotype, and transfusion requirement

Hypocellular myelodysplastic syndrome Definition of the individual patient’s risk group

Myelodysplastic syndrome with marrow fibrosis Risk group Score Median Risk of leukemic
survival evolution

dom chromosomal abnormalities). The combination


Very low 0 >10 years <10% at 15 years
of overt marrow dysplasia and clonal cytogenetic
abnormality allows a conclusive diagnosis of myelo- Low 1 >5 years 10–20% at 5 years
dysplastic syndrome, but this is found in only a por-
tion of patients.1 Although bone marrow biopsy may Intermediate 2 (or score ~4 years 30-40% at 5 years
be considered too invasive for elderly patients, it pro- 1 plus
vides extremely useful diagnostic and prognostic infor- marrow fibrosis)
mation regarding cellularity,8 fibrosis,9 and CD34-posi-
tive cell topography.9 High 3-4 (or score ~2 years 30% at 3 years
2 plus
Risk assessment marrow fibrosis)
The clinical heterogeneity of myelodysplastic syn-
dromes is best illustrated by the observation that these Very high 5-6 (or score ~1 year >50% at 1 year
disorders range from indolent conditions with a near- 3 to 4 plus
normal life expectancy to forms approaching AML.1 A marrow fibrosis)
risk-adapted treatment strategy is mandatory for condi-
tions showing a highly variable clinical course, and def- *Good: normal, -Y, del(5q), del(20q); Poor: complex, chromosome 7 anomalies;
Intermediate: other chromosomal abnormalities. ** Transfusion dependency: at
inition of the individual risk has been based so far on least one transfusion every 8 weeks over a period of 4 months. ***Bone marrow
the use of prognostic scoring systems. In 1997, fibrosis should be evaluated according to the European consensus criteria.
Greenberg and co-workers10 developed the Intern-
ational Prognostic Scoring System (IPSS), based on mar-
row blasts, cytogenetic pattern, and number of cytope-
nias. The IPSS was derived from a multivariable analy- fusion dependency could be considered as an inde-
sis of hematological characteristics of 816 patients at pendent indicator of the severity of the disease, and
clinical onset, and included subjects with 20-30% mar- consequently an independent prognostic factor. These
row blasts, now considered as having AML. data are in agreement with the results of a recent study
In a study on the prognostic significance of the by Greenberg and coworkers,13 which showed that the
WHO classification,11 we found that the IPSS retained severity of anemia at diagnosis is of additive prognos-
significance within the WHO subgroups. However, tic value to IPSS in terms of survival.
when accounting for blast percentage using WHO cat- In collaboration with colleagues in Düsseldorf, we
egories, the only other IPSS variable adding prognostic developed a prognostic model that accounted for the
information was found to be cytogenetics. We also WHO categories, cytogenetics and transfusion
found that transfusion dependency had a significant dependency.14 This WHO classification-based prog-
effect on survival in multivariable analysis.11 Based on nostic scoring system (WPSS) was able to classify
this and other observations1,12 we concluded that trans- patients into five risk groups showing different sur-

| 182 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

Table 3. Comparison of two different risk assessment systems (IPSS and WPSS) in 258 patients with myelodysplastic
syndrome. Within each IPSS risk group, patients have been stratified according to WPSS criteria, taking into account mul-
tilineage dysplasia, transfusion dependency and bone marrow fibrosis as indicated in Table 2.

IPSS risk group No. of patients Median overall WPSS risk group No. of patients (%) Median overall
survival* survival**

Low 71 68 Very low 26 (37) 141


Low 23 (32) 66
Intermediate 16 (23) 48
High 6 (8) 26

Intermediate 1 122 42 Very Low 6 (5) 141


Low 23 (19) 66
Intermediate 42 (34) 48
High 33 (27) 26
Very high 18 (15) 9

Intermediate 2 47 14 Intermediate 1 (2) 48


High 25 (53) 26
Very high 21 (45) 9

High 18 5 High 1 (6) 26


Very high 17 (94) 9
*Data are from Greenberg et al.10 **Data are from Malcovati et al.14

useful in clinical decision making.9 Interestingly, with-


in patients stratified according to IPSS and WPSS cate-
gories, bone marrow fibrosis involved a shift to a one-
step more advanced risk group.9 As indicated in Table
2, grade 2 to 3 marrow fibrosis can be used to better
define the individual patient’s risk group within the
WPSS. Accounting for multilineage dysplasia, transfu-
sion dependency and bone marrow fibrosis within the
WPSS categorization allows us to define the prognosis
of the individual patient with MDS more accurately as
compared with the IPSS categorization. This is mainly
noticeable for patients belonging to the IPSS low or
intermediate-1 risk groups (Table 3), that is, those
patients who pose the most difficulties in therapeutic
decision making.

Therapeutic tools currently available for myelodysplastic


syndromes
Figure 1. Cumulative risk of progressing from a WPSS risk
group to the next one (from very low to low, from low to Several therapeutic tools have been proposed in the
intermediate, from intermediate to high, and from high to last decades but only a few survived the evidence-
very high), or from the very high risk group to acute based criteria of efficacy.1 In the past years, Italian15
myeloid leukemia. Data are from the patient population
that was analyzed for defining the WPSS.14 and British16 guidelines for the therapy of myelodys-
plastic syndromes have been published. The therapeu-
tic tools currently employed in the treatment of these
disorders are reported in Table 4.
vivals and probabilities of leukemic evolution. WPSS
was shown to predict survival and leukemia progres- WPSS risk-adapted strategy
sion at any time during follow-up so, therefore, may Our WPSS risk-adapted strategy is summarized in
be used for implementing risk-adapted treatment Table 5.
strategies. Figure 1 illustrates the risk of progressing
from a WPSS risk group to the next one or to acute A) Very low WPSS risk
myeloid leukemia over time. More recently we found The approach to a patient with myelodysplastic syn-
that bone marrow fibrosis identifies a distinct sub- drome should always begin with a period of observa-
group of MDS with multilineage dysplasia, high trans- tion, with sequential peripheral blood counts – and
fusion requirement, and poor prognosis, and repre- sometimes bone marrow examinations – to assess the
sents an independent prognostic factor that may be rate of progression, if any. Patients with very low

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 183 |
14th Congress of the European Hematology Association

Table 4. Drugs and therapeutic tools currently employed in the treatment of myelodysplastic syndromes.

Therapeutic tool Current clinical use Approval by EMEA and FDA (or off-label use)

Allogeneic stem Curative treatment of relatively young Not required


cell transplantation patients with MDS who have a donor

Red cell transfusion Most widely employed treatment of anemia Not required

Iron chelation Treatment of transfusion iron overload Deferoxamine is approved for treatment of transfusion iron overload.
(deferoxamine, Deferasirox has been approved by FDA for first-line treatment
deferasirox) of transfusion iron overload and by EMEA for the treatment of this condition
when deferoxamine therapy is contraindicated or inadequate

Recombinant human Employed for treatment of anemia Despite the fact that epoetins are widely employed for treatment of
erythropoietin anemia in MDS patients, this represents an off-label use of these drugs
(different epoetins)

Granulocyte-colony Employed in combination with recombinant The use of G-CSF in the treatment of anemia in MDS patients
stimulating factor human erythropoietin for treatment of anemia. represents an off-label use of this drug
(G-CSF) Occasionally employed for treatment of granulocytopenia

Anti-thymocyte Immunosuppressive therapy in patients This represents an off-label use of both ATG and cyclosporine A
globulin (ATG) with hypocellular myelodysplastic syndrome
and cyclosporine

Lenalidomide Treatment of transfusion dependent patients In 2005, the FDA approved lenalidomide for the treatment of patients with
with myelodysplastic syndrome associated with deletion (5q) transfusion-dependent anemia due to low- or intermediate-1-risk MDS
associated with a deletion 5q cytogenetic abnormality with or without
additional cytogenetic abnormalities. In 2008, the EMEA Committee for
Medicinal Products for Human Use (CHMP) adopted a negative opinion,
recommending the refusal of marketing authorization for lenalidomide,
intended for treatment of transfusion-dependent patients with MDS
associated with del(5q).The EMEA CHMP concluded that the safety of
lenalidomide was difficult to assess, and that, in particular, it was difficult
to determine whether treatment with this drug increased the risk of
progression to acute myeloid leukemia

Azacitidine Employed mainly for treatment of patients with high-risk MDS FDA has approved azacitidine for the treatment of patients with the following
FAB MDS subtypes: refractory anemia (RA) or refractory anemia with ringed
sideroblasts (RARS) (if accompanied by neutropenia or thrombocytopenia or
requiring transfusions), refractory anemia with excess blasts (RAEB),
refractory anemia with excess blasts in transformation (RAEB-T), and
chronic myelomonocytic leukemia (CMMoL).
EMEA has approved azacitidine for the treatment of adult patients who are
not eligible for hematopoietic stem cell transplantation with:
- intermediate 2 and high risk MDS according to the IPSS;
- chronic myelomonocytic leukemia with 10-29% marrow blasts without
myeloproliferative disorder;
- acute myeloid leukemia with 20-30% blasts and multilineage dysplasia,
according to the WHO classification

Decitabine Employed mainly for treatment of patients with high-risk MDS FDA has approved decitabine for the treatment of patients with MDS
including previously treated and untreated, de novo and secondary MDS of
all FAB subtypes (RA, RARS, RAEB, RAEB-t, and CMML) and IPSS
intermediate-1, intermediate-2, and high-risk

| 184 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

Table 5. WPSS risk-adapted treatment strategy for WPSS risk include those with refractory anemia, or
myelodysplastic syndromes. refractory anemia with ringed sideroblasts, or
myelodysplastic syndrome with isolated del(5q) and
WPSS risk-group defined Therapeutic options no transfusion requirement. Their median life
as indicated in Table 2
expectancy is greater than 10 years, and as long as the
disease remains stable, is not significantly different
than that of the general population;14 these patients do
not need any treatment and can be followed regularly.
Very low risk Watchful-waiting strategy
B) Low WPSS risk, including 5q- syndrome and hypoplas-
Low risk Patients with multilineage dysplasia or intermediate
tic myelodysplastic syndrome
These patients may belong to this risk group (score
risk cytogenetics without symptomatic cytopenia may
1) because of one of the following features: multilin-
be just followed without any specific treatment eage dysplasia, intermediate risk cytogenetics, or
(watchful-waiting strategy) transfusion dependency.
Patients with multilineage dysplasia or intermediate
Transfusion-dependent patients can be treated with risk cytogenetics without symptomatic cytopenia may
be just followed without any specific treatment.
recombinant human erythropoietin: responsive
However, frequent controls (every 2 to 3 months) are
subjects are those with inadequate endogenous needed in these patients, as the 1-year risk of disease
erythropoietin productions (serum erythropoietin levels progression is about 20-25%.
lower than 100 to 200 mU/mL) and low need for Transfusion dependent patients with low WPSS risk
blood transfusion (less than 2 units per month). can be treated with recombinant human erythropoi-
Patients who fail to respond to recombinant human
etin. Responsive patients are mainly those with early
disease, inadequate endogenous erythropoietin pro-
erythropoietin may be considered for more aggressive duction (serum erythropoietin levels lower than 100 to
treatments (allogeneic stem cell transplantation and 200 mU/mL) and a low need for blood transfusion (less
azacitidine). Those who are given regular blood than 2 units per month).
transfusion should also receive iron chelation therapy The issue of transfusion-dependent patients with
myelodysplastic syndrome associated with deletion
(5q) has been examined in detail elsewhere.17 In
Transfusion-dependent patients with myelodysplastic
December 2005, the US Food and Drug Administration
syndrome associated with deletion (5q) may respond (FDA) approved lenalidomide for the treatment of patients
to lenalidomide treatment with abolishment of trans- with transfusion-dependent anemia due to low- or intermedi-
fusion requirement. In Europe, however, this not only ate-1-risk myelodysplastic syndromes associated with a dele-
represents an off-label use of this drug but more tion 5q cytogenetic abnormality with or without additional
cytogenetic abnormalities. In a substantial portion of
importantly a use that contrasts with the negative
patients with myelodysplastic syndrome and del(5q),
opinion of EMEA (related to the currently uncertain studies by List et al.18,19 indeed showed that lenalido-
safety profile of lenalidomide in this condition) mide is able to induce a cytogenetic remission and to
abolish transfusion requirement. On January 24, 2008,
Patients with hypoplastic myelodysplastic syndrome the EMEA Committee for Medicinal Products for
under the age of 60 years may be treated with
Human Use (CHMP) adopted a negative opinion, rec-
ommending the refusal of the marketing authorization
anti-thymocyte globulin of horse origin (h-ATG) and for lenalidomide intended for the treatment of anemia
cyclosporine A due to myelodysplastic syndromes, more specifically
for the treatment of transfusion-dependent patients
Intermediate, high Fit patients aged up to 65 years with intermediate to with MDS associated with del(5q), and with a low to
and very high risk very high WPSS risk are candidates for allogeneic stem intermediate risk of progressing to leukemia or death.20
Following the applicant’s request for a re-examination
cell transplantation
of the opinion, the CHMP confirmed the refusal of the
marketing authorization on May 30, 2008. The CHMP
Patients who are not eligible for allogeneic stem cell concluded that the safety of lenalidomide was difficult
transplantation may be treated with azacitidine to assess; in particular because the study did not com-
pare the medicine with any other treatment, it was dif-
ficult to determine if treatment with this drug
Patients who are not eligible for allogeneic stem cell
increased the risk of progression to acute myeloid
transplantation and/or do not respond to azacitidine leukemia. In conclusion, the CHMP was of the opinion
may be considered for experimental treatments that the benefits of lenalidomide in the treatment of
(clinical trials) anemia of MDS with del(5q) did not outweigh its
potential risks. Thus, in Europe, the administration of
Patients who do not fit into previous groups are given
lenalidomide to patients with myelodysplastic syn-
drome associated with del(5q) not only represents an
supportive therapy (regular red cell transfusion) off-label use of this drug, but also clashes with the

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 185 |
14th Congress of the European Hematology Association

European authority’s opinion that the benefits of this dine for the treatment of adult patients with high-risk
treatment do not outweigh its potential risks. In our myelodysplastic syndrome who are not eligible for
opinion, this places an enormous responsibility on the allogeneic hematopoietic stem cell transplantation
prescribing physician’s shoulders and prevents any rec- (Table 4).
ommendations being made. Patients who are not eligible for allogeneic stem cell
In patients with hypoplastic myelodysplastic syn- transplantation and those who do not respond to azac-
drome, combined treatment with anti-thymocyte itidine should be considered for participation in a clin-
globulin of horse origin (h-ATG) and cyclosporine A ical trial or given supportive therapy.
may be considered, especially in those under the age of For most patients, red cell transfusions eventually
60 years. About one third of these patients may obtain remain the mainstay of therapy. As regards this sup-
a durable response.21 portive care, pre-transfusion hemoglobin levels may
range between 7 and 9 g/dL according to the patient’s
C) Intermediate, high or very high WPSS risk performance status and co-morbidities.
At present, the only treatment that can cure a patient
with myelodysplastic syndrome is allogeneic stem cell Conclusions
transplantation.22,23 It is estimated that approximately In the last few years, our ability to define the prog-
one third of patients receiving an allogeneic transplan- nosis of the individual patient with myelodysplastic
tation are cured with this treatment, but only about 8 syndrome has improved, and a few drugs have been
to 10% of all patients are eligible and have a donor.1 approved by FDA and/or EMEA for treatment of MDS.
The decision to perform allogeneic stem cell transplan- However, allogeneic stem cell transplantation remains
tation can be based on WPSS risk, comorbidities, age, the only treatment capable of modifying the natural
and should be shared as much as possible with the history of these disorders.
patient, whose risk aversion should be taken into
account. Fit patients aged up to 65 years with interme-
diate to very high WPSS risk are candidates for allo- References
geneic stem cell transplantation. In a recent report,23 5-
1. Cazzola M, Malcovati L. Myelodysplastic syndromes--cop-
year overall survival after transplantation was 63%, ing with ineffective hematopoiesis. N Engl J Med 2005;352:
40%, and 16% for the intermediate, high and very 536-8.
high-risk groups, respectively. This may suggest that 2. Mufti GJ, Bennett JM, Goasguen J, Bain BJ, Baumann I,
patients who are eligible for allogeneic stem cell trans- Brunning R, et al. Diagnosis and classification of myelodys-
plastic syndrome: International Working Group on Morph-
plantation should be transplanted before they progress ology of myelodysplastic syndrome (IWGM-MDS) consen-
to the very high-risk group. In the estimation of the sus proposals for the definition and enumeration of
individual risk/benefit balance for allogeneic stem cell myeloblasts and ring sideroblasts. Haematologica 2008;93:
1712-7.
transplantation, besides defining disease-related and 3. Haase D, Germing U, Schanz J, Pfeilstocker M, Nosslinger T,
demographic variables, a critical task is identifying and Hildebrandt B, et al. New insights into the prognostic impact
assessing comorbidities that have a substantial impact of the karyotype in MDS and correlation with subtypes: evi-
dence from a core dataset of 2124 patients. Blood 2007;110:
on non-relapse mortality. A Hematopoietic Cell Trans- 4385-95.
plantation-specific Comorbidity Index (HCT-CI) for 4. Vardiman JW, Brunning RD, Arber DA, Le Beau MM, Porwit
predicting risk of non-relapse mortality has been A, Tefferi A, et al. Introduction and overview of the classifi-
cation of the myeloid neoplasms. In: Swerdlow SH, Campo
developed and validated.24 E, Harris NL, Jaffe ES, Pileri SA, Stein H, et al., eds. WHO
DNA hypermethylation may play a role in the pa- classification of tumours of hamatopoietic and lymphoid tis-
thogenesis of myeloid neoplasms.25 The DNA hypo- sues. Lyon: IARC; 2008. p. 18-30.
5. Bennett JM, Catovsky D, Daniel MT, Flandrin G, Galton DA,
methylating pyrimidine analogues 5-azacytidine might Gralnick HR, et al. Proposals for the classification of the
reduce hypermethylation and induce re-expression of myelodysplastic syndromes. Br J Haematol 1982;51:189-99.
key tumor suppressor genes. However, the action 6. Vardiman JW, Harris NL, Brunning RD. The World Health
mechanism of azacitidine in myelodysplastic syn- Organization (WHO) classification of the myeloid neo-
plasms. Blood 2002;100:2292-302.
dromes remains uncertain, and might be in part cyto- 7. Brunning RD, Germing U, Le Beau MM, Porwit A, Bauman I,
toxic. et al. Myelodysplastic syndromes/Neoplasms, overview. In:
A prospective randomized trial has shown that Swerdlow SH, Campo E, Harris NL, Jaffe ES, Pileri SA, Stein
H, et al, Editors. WHO classification of tumours of haema-
treatment of patients with myelodysplastic syndrome topoietic and lymphoid tissues. Lyon: IARC; 2008. p. 88-93.
with 5-azacytidine resulted in significantly higher 8. Bennett JM, Orazi A. Diagnostic criteria to distinguish hypo-
response rates, improved quality of life, reduced risk cellular acute myeloid leukemia from hypocellular myelo-
dysplastic syndromes and aplastic anemia: recommendations
of leukemic transformation, and improved survival for a standardized approach. Haematologica 2009;
compared with supportive care.26,27 On May 19, 2004 9. Della Porta MG, Malcovati L, Boveri E, Travaglino E, Pietra
the U.S. Food and Drug Administration approved D, Pascutto C, et al. Clinical relevance of bone marrow fibro-
sis and CD34-positive cell clusters in primary myelodysplas-
azacitidine as injectable suspension for the treatment tic syndromes. J Clin Oncol 2008;27:754-62.
of patients with myelodysplastic syndrome (Table 4). 10. Greenberg P, Cox C, LeBeau MM, Fenaux P, Morel P, Sanz G,
A survival study in patients with high-risk myelodys- et al. International scoring system for evaluating prognosis in
myelodysplastic syndromes. Blood 1997;89:2079-88.
plastic syndrome comparing azacitidine versus con- 11. Malcovati L, Porta MG, Pascutto C, Invernizzi R, Boni M,
ventional care has been recently completed.28 This Travaglino E, et al. Prognostic factors and life expectancy in
study showed that treatment with azacitidine increas- myelodysplastic syndromes classified according to WHO cri-
es overall survival in patients with higher-risk teria: a basis for clinical decision making. J Clin Oncol 2005;
23:7594-603.
myelodysplastic syndromes relative to conventional 12. Della Porta MG, Malcovati L, Invernizzi R, Travaglino E,
care. On December 17, 2008, EMEA approved azaciti- Pascutto C, Maffioli M, et al. Flow cytometry evaluation of

| 186 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

erythroid dysplasia in patients with myelodysplastic syn- to antithymocyte globulin. Leukemia 2007;21:1436-41.
drome. Leukemia 2006;20:549-55. 22. Martino R, Iacobelli S, Brand R, Jansen T, van Biezen A, Finke
13. Kao JM, McMillan A, Greenberg PL. International MDS risk J, et al. Retrospective comparison of reduced-intensity condi-
analysis workshop (IMRAW)/IPSS reanalyzed: impact of tioning and conventional high-dose conditioning for allo-
cytopenias on clinical outcomes in myelodysplastic syn- geneic hematopoietic stem cell transplantation using HLA-
dromes. Am J Hematol 2008;83:765-70. identical sibling donors in myelodysplastic syndromes. Blood
14. Malcovati L, Germing U, Kuendgen A, Della Porta MG, 2006; 108:836-46.
Pascutto C, Invernizzi R, et al. Time-dependent prognostic 23. Alessandrino EP, Della Porta MG, Bacigalupo A, Van Lint
scoring system for predicting survival and leukemic evolu- MT, Falda M, Onida F, et al. WHO classification and WPSS
tion in myelodysplastic syndromes. J Clin Oncol 2007;25: predict posttransplantation outcome in patients with
3503-10. myelodysplastic syndrome: a study from the Gruppo Italiano
15. Alessandrino EP, Amadori S, Barosi G, Cazzola M, Grossi A, Trapianto di Midollo Osseo (GITMO). Blood 2008;112:895-
Liberato LN, et al. Evidence- and consensus-based practice 902.
guidelines for the therapy of primary myelodysplastic syn- 24. Sorror ML, Maris MB, Storb R, Baron F, Sandmaier BM, Ma-
dromes. A statement from the Italian Society of Hematology. loney DG, et al. Hematopoietic cell transplantation (HCT)-
Haematologica 2002;87:1286-306. specific comorbidity index: a new tool for risk assessment
16. Bowen D, Culligan D, Jowitt S, Kelsey S, Mufti G, Oscier D, before allogeneic HCT. Blood 2005;106:2912-9.
et al. Guidelines for the diagnosis and therapy of adult mye- 25. Hellstrom-Lindberg E. Update on supportive care and new
lodysplastic syndromes. Br J Haematol 2003;120:187-200.
17. Cazzola M. Myelodysplastic syndrome with isolated 5q therapies: immunomodulatory drugs, growth factors and
deletion (5q- syndrome). A clonal stem cell disorder charac- epigenetic-acting agents. Hematology (Am Soc Hematol
terized by defective ribosome biogenesis. Haematologica Educ Program) 2005;161-6.
2008;93:967-72. 26. Kornblith AB, Herndon JE 2nd, Silverman LR, Demakos EP,
18. List A, Dewald G, Bennett J, Giagounidis A, Raza A, Feldman Odchimar-Reissig R, Holland JF, et al. Impact of azacytidine
E, et al. Lenalidomide in the myelodysplastic syndrome with on the quality of life of patients with myelodysplastic syn-
chromosome 5q deletion. N Engl J Med 2006;355:1456-65. drome treated in a randomized phase III trial: a Cancer and
19. List A, Kurtin S, Roe DJ, Buresh A, Mahadevan D, Fuchs D, Leukemia Group B study. J Clin Oncol 2002;20:2441-52.
et al. Efficacy of lenalidomide in myelodysplastic syndromes. 27. Silverman LR, Demakos EP, Peterson BL, Kornblith AB,
N Engl J Med 2005;352:549-57. Holland JC, Odchimar-Reissig R, et al. Randomized con-
20. European Medicines Agency. Questions and answers on rec- trolled trial of azacitidine in patients with the myelodysplas-
ommendation for the refusal of the marketing authorisation tic syndrome: a study of the cancer and leukemia group B. J
for Lenalidomide Celgene Europe. 2008 [cited 2008 June 4]; Clin Oncol 2002;20:2429-40.
Available from: www.emea.europa.eu/pdfs/human/opinion/ 28. Fenaux P, Mufti GJ, Hellstrom-Lindberg E, Santini V, Finelli
lenalidomide_27128808en.pdf - 2008-05-30 C, Giagounidis A, et al. Efficacy of azacitidine compared
21. Lim ZY, Killick S, Germing U, Cavenagh J, Culligan D, Baci- with that of conventional care regimens in the treatment of
galupo A, et al. Low IPSS score and bone marrow hypo- higher-risk myelodysplastic syndromes: a randomised, open-
cellularity in MDS patients predict hematological responses label, phase III study. Lancet Oncol 2009;10:223-32.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 187 |
Myeloproliferative disorders

The role of somatic and hereditary genetic factors in


the pathogenesis of myeloproliferative neoplasms

R. Kralovics A B S T R A C T

The variable presence of the JAK2 and MPL oncogenic mutations and diverse cytogenetic lesions in
Center for Molecular Medicine myeloproliferative disorders (MPN) accounts for the genetic heterogeneity among patients and gen-
(CeMM), Austrian Academy of
Sciences, Vienna, Austria, and erates progenitors with different genotypes within each progenitor pool. Mutation acquisition in MPN
Department of Internal Medicine I, appears to be stochastic and mutations seem to be randomly combined during the genetic evolution
Division of Hematology and Blood of the MPN stem cell clone. Patients with multiple acquisitions of JAK2 or MPL mutations are com-
Coagulation, Medical University of mon. The currently available data suggests that MPN displays an increased mutational frequency con-
Vienna, Vienna, Austria sistent with a mild form of a mutator phenotype. The genetic factors underlying the MPN mutator
phenotype are discussed.
Hematology Education:
the education program for the
annual congress of the European
Hematology Association

2009;3:188-191

ncreased activity of the myeloid lineages occur in patients with PMF and ET with a

I is the hallmark of myeloproliferative dis-


orders (MPN) phenotypes. Three major
phenotypic subtypes of MPN are distin-
frequency between 1-9% depending on the
cohort examined.13-17 Up to now, five MPL
variants have been described (MPL-W515L,
guished according to the predominant pres- MPL-W515K, MPL-S505N, MPL-A506T, and
entation of the phenotype. In polycythemia MPL-A519T), all confined to exon 10 of the
vera (PV), increased red cell mass, whereas in MPL gene. Studies in animal models (bone
essential thrombocythemia (ET), increased marrow transplant or transgenic) have clear-
platelet count dominate the phenotype. In ly shown that JAK2-V617F, JAK2-ex12, and
primary myelofibrosis (PMF), fibrosis of the MPL-W515L induce a myelopriliferative
bone marrow is the phenotype defining dis- phenotype in vivo.7,11,13,18-23 In addition, animal
ease feature. Since the clinical presentations studies revealed several interesting features
of all three MPN entities often overlap and a of these mutations. Tiedt et al. observed that
number of reactive condition masquerade as high expression level of the JAK2-V617F
MPN, precise diagnosis of the individual transgene induces a PV like phenotype in
MPN entities is often challenging and mice, whereas low expression induces an ET
requires molecular and histopathological like phenotype.21 In a bone marrow trans-
evaluation of each case. The complications plant model, Wernig et al. showed that the
associated with MPN include thrombosis myeloproliferative phenotype of mice is
and bleeding,1,2 secondary bone marrow modified depending on the genetic back-
fibrosis, and leukemic transformation.3-6 ground of mice.18

Somatic mutations in myeloproliferative Cytogenetic aberrations


disorders Monoclonal origin of the myeloid lineages
The discovery of a gain-of-function in MPN has been a fundamental part of the
somatic mutation in the JAK2 gene greatly disease pathogenesis and found to be shared
enhanced the understanding of the disease among all three types of MPN.24-28 Only a
pathogenesis and facilitated MPN diagno- proportion of ET patients are exceptions
sis.7-11 The most frequent mutation of JAK2 from this rule.29,30 Some polyclonal ET
(JAK-V617F) occurs in exon 14 in over 50% patients display positivity for the JAK2-
of MPN cases but it is not restricted to a sin- V617F mutation,31-33 ruling out the possibility
gle MPN entity. The less frequent exon 12 that this patient population represents a phe-
mutations (JAK2-ex12) were observed in PV nocopy or an unrecognized reactive condi-
patients only with a frequency of up to tion.
5%.11,12 Further examination of the To explain clonal hematopoiesis in MPN,
JAK/STAT signaling pathway and cytokine extensive cytogenetic studies were under-
receptors led to the discovery of various taken but no invariant aberration or MPN-
gain-of-function mutations in the throm- specific defect was found. Instead, cytoge-
bopoietin receptor (MPL). MPL mutations netic lesions commonly seen in other

| 188 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

myeloid malignancies were detected, especially dele- ex12.57,58 Thus, familial MPN exhibits a relatively
tions of chromosomes 5q, 13q and 20q, numerical aber- strong hereditary predisposition to acquire a disease-
rations of chromosomes 1, 8, 9, and frequent gains of causing somatic mutation, such as JAK2-V617F.
9p.34-43 Acquired uniparental disomy (or isodisomy) of Although the JAK2-V617F mutation was never
the short arm of chromosome (9pUPD) was found to be observed in the germ line, the MPL-S505N mutation
a frequent lesion in about one third of PV patients.44 was found to be both germ line inherited in a Japanese
Detailed mapping studies of 9pUPD represented one of family with autosomal dominant thrombocytosis59
the approaches that led to identification of the JAK2- and acquired somatically in patients with PMF or ET.15
V617F mutation in MPN.10 Absence of germ line JAK2-V617F mutations might be
due to reduced embryonic viability observed in JAK2-
Clonal diversity of the progenitor pool V617F transgenic mice.21
Variability of oncogenic point mutations and the spo- Recently, a population based study showed that first
radic presence of cytogenetic lesions in MPN patients degree relatives of patients with MPN have an
create a number of genotypic classes of progenitors. increased risk to develop MPN.60 The same study pro-
When in vitro cultured progenitor colonies are geno- posed a recessive mode of inheritance in familial
typed for the presence of JAK2-V617F or JAK2-ex12, MPN. The incomplete penetrance observed in MPN
three genotypic classes (wild type, heterozygous and families often makes it difficult to conclude the mode
homozygous for the mutation) are often observed in of inheritance. The majority of familial MPN cases
variable ratios.45-47 The only exception are progenitor represent either parent/child pairs or pedigrees with
clones from ET patients, in which homozygosity for affected members in each generation.55-58 Although an
JAK2-V617F is rarely observed.45 Similar complexity is autosomal dominant inheritance mode seems to be
observed in patients carrying cytogenetic aberrations, more likely than a recessive one, this issue will be
such as deletions on chromosome 20q (del20q) and resolved only after identification of the genetic factors
JAK2 or MPL mutations.47 Schaub et al. observed a vari- involved.
able acquisition order of del20q and JAK2-V617F in An interesting feature of MPN pathogenesis
patients carrying del20q and JAK2-V617F simultaneous- emerged from screening patients for JAK2 and MPL
ly.47 An even higher progenitor pool complexity consist- mutations. In certain cases, patients positive for two
ing of six different genotypic classes was reported different mutations were reported, for example, JAK2-
recently in a patient with del20q, del13q, 9pUPD, and V617F and MPL-W515L, JAK2-ex12 and JAK2-V617F,
JAK2-V617F.48 These studies demonstrated that the or two independently occurring JAK2-V617F muta-
acquisition of mutations in MPN does not follow a pre- tions.14,46,48 Considering the rarity of JAK2 and MPL
determined order but appears to be stochastic.49 mutations in the population, the mathematical likeli-
hood of two such events in the same individual are
Variability of mutational burden extremely low (one to three events in the entire world
As quantitative assays became available for analysis population) if the mutagenesis of these genes is ran-
of JAK2-V617F mutational burden (mutant allele bur- dom. Thus, we cannot rule out the possibility that
den), large differences were observed among patients. MPN exhibits a certain form of “mutator” phenotype
Mutant allele burdens for JAK2-V617F range from 0 to resulting in increased mutation frequency, which
100% (homozygosity) in PV and PMF whereas patients would provide an explanation for the acquisition of
with ET carry burdens between 0 and 50% and only multiple mutations. In an attempt to determine the
rarely exceeds 50% (heterozygosity). Low JAK2-V617F frequency of multiple acquisition of JAK2-V617F
mutational burden in ET is largely due to the absence of among MPN patients, an unequal distribution of
9pUPD in these patients.10,45 Another source of muta- JAK2-V617F mutations was observed between the
tional burden variability is the differences in population two most common JAK2 gene haplotypes.48 One of
size of mutant cells and variable ratio of heterozygous the haplotypes harbored 80% of all JAK2-V617F muta-
and homozygous cells for JAK2-V617F. Allelelic burden tions and was found to be a susceptibility locus for the
was shown to influence MPN-specific gene expression development of JAK2-V617F positive MPN. 48,61
along with a number of clinical variables, such as sec- However, the JAK2 locus was not shown to exhibit
ondary fibrosis, thrombosis, hematocrit, leukocyte and linkage with MPN phenotype in families.55 It, there-
platelet counts.50-54 Presence of 9pUPD resulting in gain fore, appears unlikely to be responsible for the famil-
of homozygosity along the short arm of chromosome 9 ial MPN predisposition, which seems to be stronger
and gain of homozygosity for JAK2-V617F is the main than the effect exerted by the 46/1 or the GGCC MPN
cause of high mutational burden in MPN. As a result, susceptibility haplotype of JAK2.48,61
secondary genetic effects might contribute to the phe- In conclusion, two types of hereditary predisposi-
notype of MPN patients with high mutational burden, tions for MPN were identified so far: one weaker pre-
including dysregulation of imprinted genes and expres- disposition exerted by a specific JAK2 haplotype and a
sion of rare recessive alleles of genes.49 stronger predisposition exerted by a yet unknown
mutation responsible for the familial MPN clustering.
Hereditary predispositions to myeloproliferative Both of these changes represent examples of a germ
disorders line “mutator” effect since they increase the likelihood
Patients with a familial history of MPN are pheno- of JAK2 mutation acquisition. It remains to be seen
typically and molecularly indistinguishable from spo- whether a somatic mutation can render a clone more
radic MPN patients.55-58 All familial MPN cases tested susceptible to acquire a JAK2 mutation and result in
to date were carrying a somatic JAK2-V617F or JAK2- the development of MPN.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 189 |
14th Congress of the European Hematology Association

genic expression of JAK2V617F causes myeloproliferative dis-


References orders in mice. Blood 2008;111:5109-17.
23. Shide K, Shimoda HK, Kumano T, Karube K, Kameda T,
1. Randi ML, Stocco F, Rossi C, Tison T, Girolami A. Thrombosis Takenaka K, et al. Development of ET, primary myelofibrosis
and hemorrhage in thrombocytosis: evaluation of a large and PV in mice expressing JAK2 V617F. Leukemia 2008;22:87-
cohort of patients (357 cases). J Med 1991;22:213-23. 95.
2. Murphy S. Therapeutic dilemmas: balancing the risks of 24. Adamson JW, Fialkow PJ, Murphy S, Prchal JF, Steinmann L.
bleeding, thrombosis, and leukemic transformation in myelo- Polycythemia vera: stem-cell and probable clonal origin of the
proliferative disorders (MPD). Thromb Haemost 1997;78:622- disease. New Eng J Med 1976 1976;295:913-6.
6. 25. Fialkow PJ, Faguet GB, Jacobson RJ, Vaidya K, Murphy S.
3. Sterkers Y, Preudhomme C, Lai JL, Demory JL, Caulier MT, Evidence that essential thrombocythemia is a clonal disorder
Wattel E, et al. Acute myeloid leukemia and myelodysplastic with origin in a multipotent stem cell. Blood 1981;58:916-8.
syndromes following essential thrombocythemia treated with 26. El Kassar N, Hetet G, Briere J, Grandchamp B. Clonality analy-
hydroxyurea: high proportion of cases with 17p deletion. sis of hematopoiesis in essential thrombocythemia: advan-
Blood 1998;91:616-22. tages of studying T lymphocytes and platelets. Blood 1997;
4. Shibata K, Shimamoto Y, Suga K, Sano M, Matsuzaki M, Ya- 89:128-34.
maguchi M. Essential thrombocythemia terminating in acute 27. Anger B, Janssen JW, Schrezenmeier H, Hehlmann R, Heimpel
leukemia with minimal myeloid differentiation--a brief review H, Bartram CR. Clonal analysis of chronic myeloproliferative
of recent literature. Acta Haematol 1994;91:84-8. disorders using X-linked DNA polymorphisms. Leukemia
5. Murphy S, Peterson P, Iland H, Laszlo J. Experience of the 1990;4:258-61.
Polycythemia Vera Study Group with essential thrombo- 28. Kreipe H, Jaquet K, Felgner J, Radzun HJ, Parwaresch MR.
cythemia: a final report on diagnostic criteria, survival, and Clonal granulocytes and bone marrow cells in the cellular
leukemic transition by treatment. Semin Hematol 1997;34:29- phase of agnogenic myeloid metaplasia. Blood 1991;78:1814-
39. 7.
6. Vardiman JW, Harris NL, Brunning RD. The World Health 29. Harrison CN, Gale RE, Machin SJ, Linch DC. A large propor-
Organization (WHO) classification of the myeloid neoplasms. tion of patients with a diagnosis of essential thrombocythemia
Blood 2002;100:2292-302. do not have a clonal disorder and may be at lower risk of
7. James C, Ugo V, Le Couedic JP, Staerk J, Delhommeau F, thrombotic complications. Blood 1999;93:417-24.
Lacout C, et al. A unique clonal JAK2 mutation leading to con- 30. Liu E, Jelinek J, Pastore YD, Guan Y, Prchal JF, Prchal JT.
stitutive signalling causes polycythaemia vera. Nature 2005; Discrimination of polycythemias and thrombocytoses by
434:1144-8. novel, simple, accurate clonality assays and comparison with
8. Baxter EJ, Scott LM, Campbell PJ, East C, Fourouclas N, PRV-1 expression and BFU-E response to erythropoietin.
Swanton S, et al. Acquired mutation of the tyrosine kinase Blood 2003;101:3294-301.
JAK2 in human myeloproliferative disorders. Lancet 2005; 31. Antonioli E, Guglielmelli P, Pancrazzi A, Bogani C, Verrucci M,
365:1054-61. Ponziani V, et al. Clinical implications of the JAK2 V617F
9. Levine RL, Wadleigh M, Cools J, Ebert BL, Wernig G, Huntly mutation in essential thrombocythemia. Leukemia 2005; 19:
BJ, et al. Activating mutation in the tyrosine kinase JAK2 in 1847-9.
polycythemia vera, essential thrombocythemia, and myeloid 32. Levine RL, Belisle C, Wadleigh M, Zahrieh D, Lee S, Chagnon
metaplasia with myelofibrosis. Cancer Cell 2005;7:387-97. P, et al. X-inactivation-based clonality analysis and quantita-
10. Kralovics R, Passamonti F, Buser AS, Teo SS, Tiedt R, Passweg tive JAK2V617F assessment reveal a strong association
JR, et al. A gain-of-function mutation of JAK2 in myeloprolif- between clonality and JAK2V617F in PV but not ET/MMM,
erative disorders. N Engl J Med 2005;352:1779-90. and identifies a subset of JAK2V617F-negative ET and MMM
11. Scott LM, Tong W, Levine RL, Scott MA, Beer PA, Stratton patients with clonal hematopoiesis. Blood 2006;107:4139-41.
MR, et al. JAK2 exon 12 mutations in polycythemia vera and 33. Kralovics R, Teo SS, Li S, Theocharides A, Buser AS, Tichelli
idiopathic erythrocytosis. N Engl J Med 2007;356:459-68. A, et al. Acquisition of the V617F mutation of JAK2 is a late
12. Pietra D, Li S, Brisci A, Passamonti F, Rumi E, Theocharides A, genetic event in a subset of patients with myeloproliferative
et al. Somatic mutations of JAK2 exon 12 in patients with disorders. Blood 2006;108:1377-80.
JAK2 (V617F)-negative myeloproliferative disorders. Blood 34. Rege-Cambrin G, Mecucci C, Tricot G, Michaux JL, Louwagie
2008;111:1686-9. A, Van Hove W, et al. A chromosomal profile of polycythemia
13. Pikman Y, Lee BH, Mercher T, McDowell E, Ebert BL, Gozo vera. Cancer Genet Cytogenet 1987;25:233-45.
M, et al. MPLW515L is a novel somatic activating mutation in 35. Diez-Martin JL, Graham DL, Petitt RM, Dewald GW.
myelofibrosis with myeloid metaplasia. PLoS Med 2006;3: Chromosome studies in 104 patients with polycythemia vera.
e270. Mayo Clinic Proc 1991;66:287-99.
14. Pardanani AD, Levine RL, Lasho T, Pikman Y, Mesa RA, Wad- 36. Mertens F, Johansson B, Heim S, Kristoffersson U, Mitelman F.
leigh M, et al. MPL515 mutations in myeloproliferative and Karyotypic patterns in chronic myeloproliferative disorders:
other myeloid disorders: a study of 1182 patients. Blood 2006; report on 74 cases and review of the literature. Leukemia
108:3472-6. 1991;5:214-20.
15. Beer PA, Campbell PJ, Scott LM, Bench AJ, Erber WN, Bareford 37. Reilly JT, Snowden JA, Spearing RL, Fitzgerald PM, Jones N,
D, et al. MPL mutations in myeloproliferative disorders: analy- Watmore A, et al. Cytogenetic abnormalities and their prog-
sis of the PT-1 cohort. Blood 2008;112:141-9. nostic significance in idiopathic myelofibrosis: a study of 106
16. Vannucchi AM, Antonioli E, Guglielmelli P, Pancrazzi A, cases. Br J Haematol 1997;98:96-102.
Guerini V, Barosi G, et al. Characteristics and clinical correlates 38. Kanfer E, Price CM, Colman SM, Barrett AJ. Erythropoietin-
of MPL 515W>L/K mutation in essential thrombocythemia. independent colony growth in polycythaemia vera is not
Blood 2008; 112:844-7. restricted to progenitor cells with trisomy of chromosome 8.
17. Chaligné R, Tonetti C, Besancenot R, Roy L, Marty C, Mossuz Br J Haematol 1992;82:773-4.
P, et al. New mutations of MPL in primitive myelofibrosis: 39. Westwood NB, Gruszka-Westwood AM, Pearson CE, Delord
only the MPL W515 mutations promote a G(1)/S-phase transi- CF, Green AR, Huntly BJ, et al. The incidences of trisomy 8,
tion. Leukemia 2008; 22:1557-66. trisomy 9 and D20S108 deletion in polycythaemia vera: an
18. Wernig G, Mercher T, Okabe R, Levine RL, Lee BH, Gilliland analysis of blood granulocytes using interphase fluorescence
DG. Expression of Jak2V617F causes a polycythemia vera-like in situ hybridization. Br J Haematol 2000;110:839-46.
disease with associated myelofibrosis in a murine bone mar- 40. Najfeld V, Montella L, Scalise A, Fruchtman S. Exploring poly-
row transplant model. Blood 2006;107:4274-81. cythaemia vera with fluorescence in situ hybridization: addi-
19. Lacout C, Pisani DF, Tulliez M, Gachelin FM, Vainchenker W, tional cryptic 9p is the most frequent abnormality detected. Br
Villeval JL. JAK2V617F expression in murine hematopoietic J Haematol 2002;119:558-66.
cells leads to MPD mimicking human PV with secondary 41. Sinclair EJ, Forrest EC, Reilly JT, Watmore AE, Potter AM.
myelofibrosis. Blood 2006;108:1652-60. Fluorescence in situ hybridization analysis of 25 cases of idio-
20. Bumm TG, Elsea C, Corbin AS, Loriaux M, Sherbenou D, pathic myelofibrosis and two cases of secondary myelofibro-
Wood L, et al. Characterization of murine JAK2V617F-positive sis: monoallelic loss of RB1, D13S319 and D13S25 loci associ-
myeloproliferative disease. Cancer Res 2006;66:11156-65. ated with cytogenetic deletion and translocation involving
21. Tiedt R, Hao-Shen H, Sobas MA, Looser R, Dirnhofer S, 13q14. Br J Haematol 2001;113:365-8.
Schwaller J, et al. Ratio of mutant JAK2-V617F to wild-type 42. Bench AJ, Nacheva EP, Champion KM, Green AR. Molecular
Jak2 determines the MPD phenotypes in transgenic mice. genetics and cytogenetics of myeloproliferative disorders.
Blood 2008;111:3931-40. Baillieres Clin Haematol 1998;11:819-48.
22. Xing S, Wanting TH, Zhao W, Ma J, Wang S, Xu X, et al. Trans- 43. Gangat N, Strand J, Lasho TL, Finke CM, Knudson RA, Parda-

| 190 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

nani A, et al. Cytogenetic studies at diagnosis in polycythemia Pancrazzi A, Ponziani V, et al. Prospective identification of
vera: clinical and JAK2V617F allele burden correlates. Eur J high-risk polycythemia vera patients based on JAK2(V617F)
Haematol 2008;80:197-200. allele burden. Leukemia 2007;21:1952-9.
44. Kralovics R, Guan Y, Prchal JT. Acquired uniparental disomy 54. Vannucchi AM, Antonioli E, Guglielmelli P, Rambaldi A,
of chromosome 9p is a frequent stem cell defect in poly- Barosi G, Marchioli R, et al. Clinical profile of homozygous
cythemia vera. Exp Hematol 2002;30:229-36. JAK2 617V>F mutation in patients with polycythemia vera or
45. Scott LM, Scott MA, Campbell PJ, Green AR. Progenitors essential thrombocythemia. Blood 2007;110:840-6.
homozygous for the V617F mutation occur in most patients 55. Kralovics R, Stockton DW, Prchal JT. Clonal hematopoiesis in
with polycythemia vera, but not essential thrombocythemia. familial polycythemia vera suggests the involvement of multi-
Blood 2006;108:2435-7. ple mutational events in the early pathogenesis of the disease.
46. Li S, Kralovics R, De Libero G, Theocharides A, Gisslinger H, Blood 2003;102:3793-7.
Skoda RC. Clonal heterogeneity in polycythemia vera patients 56. Kralovics R, Skoda RC. Molecular pathogenesis of Phila-
with JAK2 exon12 and JAK2-V617F mutations. Blood 2008; delphia chromosome negative myeloproliferative disorders.
111:3863-6. Blood Rev 2005;19:1-13.
47. Schaub FX, Jager R, Looser R, Hao-Shen H, Hermouet S, 57. Bellanne-Chantelot C, Chaumarel I, Labopin M, Bellanger F,
Girodon F, et al. Clonal analysis of deletions on chromosome Barbu V, De Toma C, et al. Genetic and clinical implications of
20q and JAK2-V617F in MPD suggests that del20q acts inde- the Val617Phe JAK2 mutation in 72 families with myeloprolif-
pendently and is not one of the pre-disposing mutations for
erative disorders. Blood 2006;108:346-52.
JAK2-V617F. Blood 2008;113:2022-7.
48. Olcaydu D, Harutyunyan A, Jäger R, Berg T, Gisslinger B, 58. Rumi E, Passamonti F, Pietra D, Della Porta MG, Arcaini L,
Pabinger I, et al. A common JAK2 haplotype confers suscepti- Boggi S, et al. JAK2 (V617F) as an acquired somatic mutation
bility to myeloproliferative neoplasms. Nat Gen 2009;AOP. and a secondary genetic event associated with disease pro-
49. Kralovics R. Genetic complexity of myeloproliferative neo- gression in familial myeloproliferative disorders. Cancer 2006;
plasms. Leukemia 2008;22:1841-8. 107:2206-11.
50. Kralovics R, Teo SS, Buser AS, Brutsche M, Tiedt R, Tichelli A, 59. Ding J, Komatsu H, Wakita A, Kato-Uranishi M, Ito M, Satoh
et al. Altered gene expression in myeloproliferative disorders A, et al. Familial essential thrombocythemia associated with a
correlates with activation of signaling by the V617F mutation dominant-positive activating mutation of the c-MPL gene,
of Jak2. Blood 2005;106:3374-6. which encodes for the receptor for thrombopoietin. Blood
51. Antonioli E, Guglielmelli P, Poli G, Bogani C, Pancrazzi A, 2004;103:4198-200.
Longo G, et al. Influence of JAK2V617F allele burden on phe- 60. Landgren O, Goldin LR, Kristinsson SY, Helgadottir EA,
notype in essential thrombocythemia. Haematologica 2008; Samuelsson J, Bjorkholm M. Increased risks of polycythemia
93:41-8. vera, essential thrombocythemia, and myelofibrosis among
52. Vannucchi AM, Antonioli E, Guglielmelli P, Pardanani A, 24577 first-degree relatives of 11039 patients with myelopro-
Tefferi A. Clinical correlates of JAK2V617F presence or allele liferative neoplasms in Sweden. Blood 2008;112:2199-204.
burden in myeloproliferative neoplasms: a critical reappraisal. 61. Jones AV, Chase A, Silver RT, Oscier D, Zoi K, Wang YL, et al.
Leukemia 2008;22:1299-307. JAK2 haplotype is a major risk factor for the development of
53. Vannucchi AM, Antonioli E, Guglielmelli P, Longo G, myeloproliferative neoplasms. Nat Genet 2009;41:446-9.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 191 |
Myeloproliferative disorders

Risk adapted approach to the treatment of primary


myelofibrosis

J. Mascarenhas A B S T R A C T
R. Hoffman Primary myelofibrosis is a clonal hematopoietic stem cell disorder associated with characteristic
clinical histopathologic and hematologic features. However, there is heterogeneity with regard to clin-
Tisch Cancer Institute, Mount Sinai ical manifestations, clinical course and overall survival. It is paramount that patients be correctly
School of Medicine, New York, USA stratified according to their risk of developing life threatening outcomes based on accepted prognos-
tic features in order that treatment plans be developed to favor the benefit versus risk of a selected
Hematology Education: therapy for an individual patient. With advances in the understanding of the molecular underpinnings
the education program for the of the myeloproliferative neoplasms, the therapeutic options for such patients have greatly increased.
annual congress of the European Treatment approaches range from watchful waiting, palliative/supportive care, implementation of rec-
Hematology Association ognized therapeutic treatments, testing experimental small molecule therapies and pursuing poten-
tially curative allogeneic stem cell transplantation. These alternative approaches available to the cli-
2009;3:192-199 nician treating PMF require decision-making based on an understanding of the biology of the disease,
risk stratification, and the potential benefits as well as risks of each of these choices.

rimary myelofibrosis, PMF, is a ence of either a WBC greater than 30×106/µL

P Philadelphia chromosome negative


myeloproliferative neoplasm (MPN). In
2008, the World Health Organization
or less than 4 ×106/µL, and a hemoglobin less
than 10 g/dL.3 A point is given for each of
these risk factors to create a Lille score of
(WHO) revised the system for classifying either 0, 1 or 2. Patients with Lille class 0, 1
myeloid malignancies, changing the term or 2 disease have median survivals of 93, 26
myeloproliferative disease or disorder to neo- and 13 months, respectively. Recently, the
plasm. Primary myelofibrosis is one of the IWG-MRT has reported a study assessing
nine MPN. The diagnosis of PMF requires prognostic variables in over 1000 PMF
meeting all three major and at least two of patients from seven institutions and found
the minor diagnostic criteria (Table 1).1 an overall median survival of 69 months.4 An
Within the umbrella of MPN, the clinical fea- age greater than 65, presence of constitution-
tures of PMF are virtually identical to those al symptoms, hemoglobin less than 10 g/dL,
of patients with post polycythemia vera leukocytosis more than 25×106/µL and cir-
myelofibrosis (post-PV MF) and post essen- culating peripheral blood blast count greater
tial thrombocythemia myelofibrosis (post- than or equal to 1% were all identified as
ET MF). Diagnostic criteria for these entities negative risk factors by multivariate analy-
have also been created by an International sis. Four risk groups with distinct patterns of
Working Group on Myelofibrosis Research survival were identified. Patients were classi-
and Treatment (IWG-MRT).2 It has been the fied as low risk, intermediate-1, intermedi-
practice of most investigators to apply simi- ate-2, and high risk with median survivals of
lar therapeutic strategies to patients suffer- 135, 95, 48 and 27 months, respectively.
ing from each of these entities. Importantly, JAK2V617F mutational status
was not associated with a particular prog-
Risk stratification nostic score or survival time. It is important
A number of reports have identified risk to emphasize that both the Lille and the
factors predictive of survival in PMF IWG-MRT prognostic scoring systems are
patient.3-10 Progressive marrow failure, trans- predictive of survival based on clinical fea-
formation to acute myeloid leukemia and tures at the time of diagnosis. Whether these
the consequences of portal hypertension are criteria are of use as a patient’s clinical course
the major causes of death in PMF.3 After test- changes over time and places the patient in a
ing the predictive value of a number of these different prognostic group remains
risk factors, prognostic scoring systems have unknown. Although this is common prac-
been created with which to predict survival. tice, this approach has never been validated.
Such systems are extremely valuable in Whether these scoring systems are equally
determining appropriate therapeutic strate- effective in predicting the outcome of
gies to be used for the treatment of individ- patients with post-PV/ET MF has also not
ual patients. The most widely used system is been well documented. The median overall
the Lille scoring system, which classifies survival of patients with post-PV/ET MF has
patients into three groups based on the pres- been reported to be 90 months.11 Older age,

| 192 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

anemia and prior history of PV have been reported to be


Table 1. Proposed criteria for primary myelofibrosis.
adverse prognostic characteristics. The presence of
unfavorable cytogenetic abnormalities (clones other
than 13q- or 20q-) at time of transformation from PV/ET
to MF has been identified as the most significant
adverse prognostic factor for survival of this patient
population.11
The relative utility of the Lille and IWG-MRT scoring
systems in clinical practice has yet to be determined.
During the remainder of this text, we will rely on the
Lille system for prognostication and risk stratification
for defining appropriate treatment plans.

Treatment
We hypothesize that the treatment of PMF patients
should be tailored to the individual patient based on the
risk stratification tools described. For the purposes of
this discussion and acknowledging that the various
prognostic scoring systems have never been compared
in a prospective fashion, we have chosen to use to the
Lille classification system to outline a risk based treat-
ment approach in PMF. There are no randomized trials
available at present to provide data, which could be
used to construct an evidence based algorithm for the
treatment of PMF. The recommendations made in this
manuscript represent the opinions of the authors and
the cumulative experience reported in the medical liter-
ature.

Standard of care
The present strategies employed as the standard of
care for patients with PMF are directed toward treating Androgen therapy has been shown to be an effective
the clinical symptoms and laboratory abnormalities as class of agents in the treatment of anemia in MF.
they affect the quality of life of an individual patient. Danazol, a synthetic attenuated androgen, given at an
None of these interventions is known to alter the natu- initial dose of 600 mg/day and tapered to a minimum
ral history of this disease. effective dose at 6 months was associated with a
response rate of 37 to 55% with a median time to
Anemia response between 3 to 6 months.17,18 The absence of a
Symptomatic patients with MF frequently present transfusion requirement and higher hemoglobin prior to
with anemia and require intervention. It is important initiation of danazol were associated with a favorable
that B12, folate, and iron deficiency as possible contrib- response to therapy. The most frequent toxicity of
utory factors be identified and corrected. Beyond trans- transaminitis improved with dose reduction.17
fusional support for the acutely symptomatic patient, An unexpected finding in a retrospective review of
various erythropoiesis stimulating agents (ESA) have 311 PMF patients treated at the Mayo Clinic found that
been used to minimize or eliminate the need for trans- prior therapy with ESA or danazol was associated with
fusion therapy. The role of recombinant erythropoietin a higher risk for leukemic transformation (LT).19 The
(rEPO) in the amelioration of anemia and the associated contention that the use of an ESA may, in fact, act as a
symptoms is somewhat controversial. Studies favoring surrogate marker for a more aggressive form of PMF
the use of rEPO suggest that MF patients with low may not be valid considering that other agents
serum erythropoietin levels, limited transfusional employed to treat anemia were not found to be associ-
requirements, female sex and older age are most likely ated with LT. It remains uncertain whether ESA therapy
to respond to therapy.12 In a study of 61 patients with has an effect of the evolution of such patients to acute
MF, 87% had serum erythropoietin levels appropriate to leukemia.
the degree of anemia, supporting the concept that rEPO Occasionally chemotherapeutic agents can alleviate
would unlikely be beneficial to the majority of anemic the degree of anemia in PMF patients. Oral melphalan,
MF patients.13 However, a 40 to 55% overall response for instance, has been reported to result in 60% of
rate using darbepoetin or rEPO in MF has been reported severely anemic patients without transfusion require-
in the literature. ESA should remain the first line thera- ments having a significant elevation of hemoglobin
py for anemic patients with serum erythropoietin levels while nearly 38% of transfusion dependent patients
less than 125 U/L at a rEPO dose of at least 350 becoming transfusion independent with such therapy.20
U/kg/week.13-15 Concomitant interferon alpha therapy Based on the anti-angiogenic and immunomodulato-
with rEPO has been associated with unexpected favor- ry activity of thalidomide, this agent has been used
able response rate which deserves further prospective alone and in combination with prednisone to treat PMF
evaluation.16 patients with anemia. The use of this agent is tempered

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 193 |
14th Congress of the European Hematology Association

by a toxicity profile of sedation, constipation, neuropa- quality of life (QOL). Hydroxyurea is a ribonucleotide
thy, myelosuppression and increased thrombotic poten- reductase inhibitor with cytoreductive potential with
tial. A pooled-analysis of 5 phase II studies in MF using the advantage of relieving constitutional symptoms and
standard dose thalidomide showed a reduction in 6 reversing advancing splenomegaly in MF. Hydroxyurea
point severity score in 45% of patients, and worsening of has not been shown to have any advantage in selective-
this score in 20%, with 66% discontinuing treatment ly decreasing the malignant clone as assessed by
before 6 months due to intolerance of therapy.21 In a ran- JAK2V617F allele burden31 and, therefore, does not hold
domized multicenter phase II B setting in MF, thalido- potential for modifying the natural history of this dis-
mide at 400 mg daily failed to reach endpoints of ease. The success of hydroxyurea in effectively control-
improving anemia and reducing transfusion require- ling splenomegaly has not been well documented in the
ments.22 Low dose thalidomide at 50 mg/day, combined literature. Its use is largely empiric and frequently com-
with a prednisone taper, demonstrated an objective clin- plicated by excessive hematopoietic toxicity, often
ical response in 62% of patients with anemia with 95% requiring the use of alternative strategies, including
of patients able to complete 3 months of therapy.23,24 splenectomy.
More recently, lenalidomide, a more potent thalidomide Splenectomy is associated with long-term improve-
analogue with greater myelosuppressive effects and less ment in symptomatic splenomegaly, anemia, portal
sedative and neurotoxic effects has been explored in hypertension, and severe thrombocytopenia in approx-
clinical trials for MF. Phase II studies with lenalidomide imately 80%, 50%, 40% and 30% of cases, respective-
as a single agent in MF were initially seen to produce ly.32 Splenectomy is not always a viable option in
overall response rates of 22%, 33%, and 50% for ane- patients that have a poor performance status and carries
mia, splenomegaly and thrombocytopenia, respective- a risk of postoperative bleeding of 14%, infection of
ly.24,25 As expected, grade 3/4 neutropenia and thrombo- 10%, and thrombosis of 10% that contribute to an over-
cytopenia were observed, but increased thromboembol- all fatal complication rate of approximately 7%.32-34 An
ic events were not seen. In a cooperative group study, alternative approach is low dose radiation to the spleen,
the combination of prednisone and lenalidomide did which can potentially result in a transient reduction in
not improve the response rate for anemia as compared spleen size and improvement in symptoms in patients
to lenalidomide alone. Lenalidomide therapy was com- that are considered poor candidates for splenectomy.
plicated by grade 3/4 myelosuppression in 45% of Rebound thrombocytosis and compensatory hepato-
cases.25 Given the degree of myelosuppression observed megaly also complicate splenectomy, and postoperative
in this study and the comparable response rates seen thrombocytosis can increase the risk of perioperative
with thalidomide therapy, these investigators conclud- thrombosis and compromise survival.34 A post splenec-
ed that lenalidomide should be reserved as second line tomy rate of LT has been reported of 16.3% and the risk
therapy for anemic patients that fail thalidomide thera- of LT appears to be highest in MF patients with marked-
py or posses the 5q- abnormality. Pomalidomide, CC- ly increased spleen size and thrombocytopenia prior to
4047, a newer IMiD, which has far less neurotoxicity is surgery.34 Most patients require hydroxyurea therapy to
being investigated in early clinical trials in patients with prevent postoperative compensatory hepatomegaly. In
MF. Pomalidomide alone or in combination with short patients resistant to hydroxyurea, 2-chlorodeoxyadeno-
course prednisone showed a 25% response rate for ane- sine (2-CdA), a purine nucleoside analogue has been
mia and increased the platelet count by more than 50% shown to decrease hepatomegaly and thrombocytosis
in 43% of patients with platelet counts less than in post-splenectomy MF patients in a pilot study of nine
100×109/L.26 WBC less than 4×109/L were the most pow- patients.35
erful predictor of response but no patients experienced
a significant reduction in splenomegaly. It is sobering Experimental therapies
that in this trial, a cohort of patients received pred- Most clinical trials for patients with advanced forms
nisone therapy alone, which was associated with a 19% of MF do not discriminate between patients with PMF
response rate in anemic patients. We conclude that or post-PV/ET MF. These clinical trials are designed for
pomalidomide requires further careful evaluation. the enrollment of intermediate/high risk or sympto-
There are no randomized trials specifically investigat- matic patients. Response criteria created by the IWG-
ing the benefit of iron chelation therapy in PMF. Case MRT and the European Myelofibrosis Network (EUM-
reports indicate that effective reduction in ferritin levels NET) have enabled standardized evaluation of these
can be achieved and additionally, this may enhance ery- agents by objective endpoints.36,37
thropoiesis.27-29 A very recent analysis of transfusion
dependent PMF patients receiving iron chelation thera- JAK2 inhibitors
py, demonstrated a survival benefit favoring chelation JAK2 inhibitors are a class of small molecules that
therapy over those not receiving chelation therapy.30 block the JAK-STAT pathway resulting from the
Based on the available data in PMF, iron chelation ther- JAK2V617F gene mutation, and to some degree, wild
apy should be considered in transfusion dependent type JAK2. Clinical trials with several candidate mole-
patients with a ferritin more than 1000 and an extended cules, including Cep-701, XL019, INCB018424,
expected survival. TG101348 have been pursued. Each trial has reported
some success in reaching clinical endpoints, including
Organomegaly reduction in spleen size and improvement in constitu-
Splenomegaly is a frequent complication of PMF, tional symptoms.38-40 The excitement about JAK2V617F
which is associated with a hypercatabolic state and con- inhibition has been tempered by the realization that a
stitutional symptoms that compromise the patient’s significant diminution in mutant allele burden or

| 194 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

improvement in the degree of anemia has been rarely inhibitor, LBH589 is also being evaluated in phase I/II
observed.38 The Myelofibrosis Symptom Assessment studies in MF based on encouraging data using this oral
Form (MFSAF) is a proposed new instrument designed drug in a trial of patients with advanced hematological
to better capture the disease related symptoms and malignancies. Two PMF patients in these trials experi-
assess the QOL and can be utilized as a metric in future enced a reduction in the degree of splenomegaly, while
therapeutic trials.41 These JAK2 inhibitors may also play one achieved transfusion independence.50
an important role in reversing anorexia and cancer
cachexia by suppressing the pro-inflammatory cytokine Anti-angiogenic agents
state, reducing splenomegaly and reversing the hypo- The tyrosine kinase (TK) associated with the VEGFR
cholesterolemia characteristic of PMF.42 is another potential PMF therapeutic target, since this
MF is a chronic inflammatory state with marked ele- entity is associated with increased marrow micro-vessel
vation in pro-inflammatory cytokines irrespective of the density and increased serum VEGF levels. SU5416 is an
JAK2 status or degree of splenomegaly. INCB18424 has intravenous small molecule TKI that can inhibit VEGFR,
been shown to dramatically and quickly reduce these among other TKs, and has been tested in refractory
levels, which has been shown to correlate with MPNs, including a phase II study, which included three
improvement in constitutional symptoms and cachex- PMF patients with one patient obtaining a partial
ia.43 Myelosuppression, gastrointestinal toxicity and response.51 PTK787/ZK 222584 is a novel oral small mol-
neurotoxicity have been reported with the use of specif- ecule inhibitor of the VEGFR tyrosine kinase that was
ic JAK2 inhibitors. Although the primary effect of these shown in a pilot study of 29 MF patients to result in a
agents appears to be on hematopoiesis, JAK-STAT sig- CR in a single patient and clinical improvement (CI) in
naling plays a role in the biological regulatory mecha- five patients.52 Bevacizumab, a recombinant humanized
nisms involving a variety of other organ systems, which monoclonal antibody against VEGF, is also currently
could account for the toxicities observed. Since the being investigated in a phase II trial. These agents will
presently evaluated inhibitors are not specific to the likely have more clinical relevance in combination ther-
mutated from of JAK2, these toxicities are not surpris- apy with other agents.
ing. At this time, the relative clinical efficacy of each of Aplidin, a marine-derived depsipeptide with both
the JAK2 inhibitors has not been compared, thereby anti-apoptotic and anti-angiogenic properties, has been
making it impossible to identify the most effective shown to reduce BM MVD in a murine MF model
member of this class of drugs. through downregulation of VEGF and TGFβ and may
even impact megakaryocyte proliferation and the for-
Chromatin modifying agents mation of reticulin fibrosis.53 This drug will soon enter
The epigenetic silencing of genes through DNA clinical trials in MF.
hypermethylation and histone deacetylation likely
plays a role in the pathogenesis of PMF and provides a Interferon
different set of therapeutic targets.44 The nucleoside ana- Recombinant interferon (IFN) has both anti-tumor
logue 5-azacitidine (5-AZA), a DNA methyltransferase and immunomodulatory activity and has been investi-
inhibitor with proven clinical efficacy in MDS, had a gated extensively in various formulations in the treat-
response rate of 24% in MF patients.45 5-AZA given ment of MPNs.54 Pegylated interferon alpha is produced
with an alternative dosing schedule for 5 days was also by covalent addition of ethylene glycol polymers to
attempted and failed to achieve even a single clinical rIFNα, resulting in a decreased immunogenicity and an
response.46 Decitabine in case reports47 and in our hands increased half-life due to decreased clearance. Both
(unpublished data) has demonstrated clinical activity therapy with rIFNα and PEG-IFN have not enjoyed the
resulting in significant reduction in splenomegaly, same favorable clinical response rates in PMF as has
improvement in the degree of anemia and constitution- been achieved in PV and ET patients.54 Some investiga-
al symptoms, and even a durable CR in a single patient tors have suggested that treatment with IFN during
in LT. Myelosuppression is the major adverse event that early phases of the disease, so called pre-fibrotic form,
has been encountered. The sequential exposure of PMF might delay disease progression.55 Such an approach
CD34+ cells in vitro to Decitabine and trichostatin A has will require testing in a randomized trial before it can be
been reported to result in a reduction in total number of implemented.
assayable cells, CD34+ cells, hematopoietic progenitor
cells (HPC), and selective reduction in JAK2V617F+ Combination therapy
HPCs and HPCs containing cytogenetic abnormalities.48 Combination therapy in cancer treatment is a stan-
The sequential exposure of these agents also upregulat- dard approach based upon the use of drugs that affect
ed CD34+ CXCR4 expression, restoring the ability of different targets and at the same time have non-overlap-
CD34+ PMF cells to migrate in response to the SDF-1. ping organ toxicities. Such an approach has not been
These data provide a very compelling rationale to eval- previously explored in PMF. Since none of the standard
uate sequential chromatin modifying agents in future therapeutic options or experimental therapies outlined
clinical trials of PMF patients. above has similar therapeutic effectiveness as displayed
ITF2357, a class I/II HDAC inhibitor, was investigat- by imatinib, the paradigm of CML might not be appli-
ed in PV/ET and MF patients in a phase IIa study, result- cable to PMF. These obstacles have lead several investi-
ing in a median reduction of JAK2V617F mutant allele gators to explore the use of combination therapy for
burden of 8% in 10 of 17 patients. Two major and two PMF. For instance, a combination of the pan HDAC
moderate clinical responses were achieved by EUMNET inhibitor LBH589 or heat shock protein (HSP90)
criteria in the 13 MF patients treated.49 The pan HDAC inhibitor AU922 with the JAK2 inhibitor TG101209 in

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 195 |
14th Congress of the European Hematology Association

primary MPD cells or JAK2V617F+ HEL cell line showed measures, antimicrobials and immunosuppressive ther-
enhanced and even synergistic levels of apoptosis and apies. However, in a smaller review of SCT in 27 MF
attenuation of JAK2V617F, p-STAT-3, p-STAT-5, p-AKT, patients in Sweden over 22 years, a TRM of 10% and
Bcl-xl levels.56 In addition, upregulation of anti-apoptot- 30% was seen in RIC and fully MA regimens, respec-
ic proteins in erythroblasts and megakaryocytes have tively.67 One can conclude from these studies that both
been observed in MPN, suggesting to some that selec- MA and RIC allogeneic SCT can be curative for patients
tive inhibitors of these pathways might be effective with PMF. Future prospective trials are, therefore, clear-
components of a combination drug therapy for PMF. ly required to define the choice of conditioning regimen,
These encouraging laboratory findings will surely be the source of donor cells, and the selection of appropri-
followed by in vivo pilot studies attempting to improve ate PMF candidates for allogeneic SCT.
upon the current success seen with JAK2 inhibitors Currently, it is our practice to follow younger patients
alone in MF. with low risk disease for signs and symptoms of disease
progression and only resort to transplantation with dis-
Stem cell transplantation ease progression as reflected with advancing Lille score.
The only known curative approach for PMF is allo- The appropriateness of this strategy is uncertain. A long
geneic transplantation (SCT). The first step is determin- interval between diagnosis and transplant has been
ing who is an appropriate candidate for SCT and the found to be a negative predictor of TRM in multivariate
second is determining the optimal time to proceed. The analysis in a retrospective review and may actually sup-
effective completion of these decision-making process- port the use of SCT earlier in low risk patients before
es requires an appreciation of the risks involved in the progression of disease occurs.68 An argument could be
procedure and the risk of disease evolution to a terminal made that some young patients with low risk disease
event. For the purposes of this discussion, we will use may benefit from SCT upfront. At this time, we would
the Lille prognostic score system to risk stratify in order advocate watchful waiting (WW) for low risk disease in
to optimize the application of SCT in MF patients. It any age group outside of a clinical trial. It remains
must be emphasized that, although very encouraging, uncertain whether younger PMF patients with interme-
this modality of treatment remains experimental at this diate/high risk disease by Lille classification should pur-
time and that patients who are interested in being eval- sue a MA-SCT or RIC SCT; this question warrants a
uated for SCT would likely be best served by participat- prospective study.
ing in a clinical trial. Due to the high transplant related Although the presence of the JAK2V617F mutation
mortality (TRM) (27-48%) and poor overall survival has not been shown to influence either the relapse rate
(OS) (41-64%) associated with myeloablative SCT or survival after SCT, JAK2V617F has been used to
(MA-SCT) in a disease that preferentially affects those assess minimal residual disease after SCT and the allele
older than 45 years of age, reduced intensity condition- burden is inversely correlated with donor-cell
ing SCT (RIC-SCT) offers a reduced treatment related chimerism.66,69 Kroger et al. demonstrated that in 22 MF
mortality (TRM) (10%). It has a favorable complete patients receiving SCT, 78% achieved real time PCR
remission rate (CR) and overall survival (OS) of 76% negativity, while four of the five residual JAK2V17F pos-
and 86%, respectively.57-60 Initial reports of lower TRM itive patients still achieved a CR by IWG criteria.66 One
and successful engraftment with RIC-SCT61,62 prompted JAK2V617F positive patient post transplant was able to
the first prospective pilot study of 21 MF patients with eliminate the evidence of the mutation after receiving a
a median age of 53, and some as old as 70 years of age.63 donor lymphocyte infusion. This data underscores the
Conditioning with fludarabine, busulphan and ATG and potential role of monitoring the JAK2V617F gene muta-
utilizing related donors, if available, resulted in a medi- tion as a clonal marker of the depth of response post-
an time to leukocyte engraftment of 16 days with 95% transplant, and it may even be used to guide additional
complete donor chimerism at day 100. Treatment relat- immunotherapy.
ed mortality was 16% at 1 year and 19% of patients Concern regarding donor cell sequestration and
experienced grade III/IV acute GVHD while maintain- increased transfusional requirements in patients with
ing a 3 year estimated OS of 84%. A busulfan/fludara- MF and splenomegaly has prompted investigation into
bine based RIC regimen was investigated in a prospec- the role of splenectomy prior to SCT. This has been
tive study by the European Group for Blood and evaluated in a limited fashion and prior splenectomy
Marrow Transplantation (EBMT) in 104 patients with a has not been shown to be associated with an obvious
median age of 55 years of age demonstrating a 3 year survival advantage.70 Moreover, both a progressive
OS and EFS of 70% and 55%, respectively.64 reduction in splenomegaly and the degree of marrow
Additionally, a graft versus myelofibrosis effect has fibrosis can be observed over a 12 month period follow-
been reported to occur following donor lymphocyte ing RIC-SCT.71 A statistically significant prolongation in
infusions in patients with graft rejection.65,66 ANC recovery was seen in patients with massive
Most investigators have concluded that the improved splenomegaly (>30 cm) of 19 days compared to 13 days
results with allogeneic stem cell transplantation for PMF in patients with smaller spleens. Full donor chimerism
are a consequence of RIC and or the use of PBSC grafts. was achieved in all the patients indicating successful
However, a retrospective review of 100 MF patients engraftment is possible, even in the face of extensive
treated with SCT over 20 years in 26 Italian centers did splenomegaly. These results suggest that the risk associ-
not observe an advantage in TRM or OS favoring RIC ated with splenectomy may not be warranted despite a
over MA regimens. Interestingly, it is not clear that a longer period of neutropenia after RIC-SCT and we
reduction in the TRM seen after 1996 was a result of the would not recommend splenectomy prior to SCT unless
introduction of RIC regimens or improved supportive done to specifically address splenomegaly associated

| 196 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

5. Tam CS, Abruzzo LV, Lin KI, Cortes J, Lynn A, Keating MJ,
Table 2. Proposed approach to treatment of primary et al. The role of cytogenetic abnormalities as a prognostic
myelofibrosis in 2009. marker in primary myelofibrosis: Applicability at the time of
diagnosis and later during disease course. Blood 2009;[Epub
Age Standard of Care Alternative options ahead of print].
6. Cervantes F, Barosi G, Demory JL, Reilly J, Guarnone R, Du-
Lille Class O <45 WW SCT on protocol priez B, et al. Myelofibrosis with myeloid metaplasia in
45-65 WW SCT on protocol young individuals: disease characteristics, prognostic factors
and identification of risk groups. Br J Haematol 1998;102:
>65 WW SMT on protocol/rIFN 684-90.
7. Ahmed A, Chang CC. Chronic idiopathic myelofibrosis:
Lille Class I/II < 45 SCT on protocol SMT on protocol clinicopathologic features, pathogenesis, and prognosis.
45-65 SCT on protocol SMT on protocol Arch Pathol Lab Med 2006;130:1133-43.
>65 Conventional therapy SMT on protocol/ SCT 8. Dingli D, Schwager SM, Mesa RA, Li CY, Tefferi A. Pro-
gnosis in transplant-eligible patients with agnogenic myeloid
on protocol up to age 70 metaplasia: a simple CBC-based scoring system. Cancer
2006; 106:623-30.
WW: watchful waiting; SCT: stem cell transplant; SMT: small molecule therapy; 9. Elliott MA, Verstovsek S, Dingli D, Schwager SM, Mesa RA,
rIFN: recombinant interferon α. Li CY, et al. Monocytosis is an adverse prognostic factor for
survival in younger patients with primary myelofibrosis.
Leuk Res 2007;31:1503-9.
signs and symptoms. The actuarial probability of devel- 10. Mesa RA, Huang J, Schwager S, Pardanani A, Hussein K,
oping LT in PMF over 100 months from diagnosis has Dingli D, et al. Hypocholesterolemia is independently asso-
been estimated at approximately 20%, which is much ciated with decreased survival in patients with primary
higher than that observed in PPV-MF or PET-MF at 9% myelofibrosis: an analysis of lipid profiles in 558 myelopro-
liferative patients. Blood 2007;110:2548.
and 4%, respectively.72 The leukemic phenotype is usu- 11. Dingli D, Schwager SM, Mesa RA, Li CY, Dewald GW,
ally myeloid and is uniformly resistant to standard Tefferi A. Presence of unfavorable cytogenetic abnormalities
induction therapies with a median survival of 2.6 is the strongest predictor of poor survival in secondary
months.73 The role of SCT in transformed disease has myelofibrosis. Cancer 2006;106:1985-9.
12. Rodriguez JN, Martino ML, Dieguez JC, Prados D. rHuEpo
not been clarified and emphasis should be made on for the treatment of anemia in myelofibrosis with myeloid
timely institution of SCT prior to transformation. metaplasia. Experience in 6 patients and meta-analytical
In the era of rationally designed targeted molecular approach. Haematologica 1998;83:616-21.
13. Barosi G, Liberato LN, Guarnone R. Serum erythropoietin in
therapeutics, one could envision the use of JAK2 patients with myelofibrosis with myeloid metaplasia. Br J
inhibitors and/or chromatin modifying agents either as Haematol 1993;83:365-9.
a means to reduce splenomegaly and transfusion 14. Cervantes F, Alvarez-Larran A, Hernandez-Boluda JC, Sureda
requirements prior to SCT or as consolidative therapy A, Torrebadell M, Montserrat E. Erythropoietin treatment of
the anaemia of myelofibrosis with myeloid metaplasia:
post transplant to suppress the re-emergence of minimal results in 20 patients and review of the literature. Br J
residual disease. More than ever, it is imperative to refer Haematol 2004;127:399-403.
MF patients to centers offering early phase clinical trials 15. Cervantes F, Alvarez-Larran A, Hernandez-Boluda JC, Sureda
A, Granell M, Vallansot R, et al. Darbepoetin-alpha for the
and transplant protocols. A collective attempt to share anaemia of myelofibrosis with myeloid metaplasia. Br J
experience and data in order to provide the best thera- Haematol 2006;134:184-6.
peutic option to each individual patient in a disease 16. Hasselbalch HC, Clausen NT, Jensen BA. Successful treat-
characterized by biological and clinical heterogeneity ment of anemia in idiopathic myelofibrosis with recombi-
nant human erythropoietin. Am J Hematol 2002;70:92-9.
should be the goal of the international hematology com- 17. Cervantes F, Alvarez-Larran A, Domingo A, Arellano-
munity. Outlined in Table 2 is the approach of the Rodrigo E, Montserrat E. Efficacy and tolerability of danazol
authors in providing care to patients with PMF. It is our as a treatment for the anaemia of myelofibrosis with
myeloid metaplasia: long-term results in 30 patients. Br J
hope that new data generated in well-designed clinical Haematol 2005; 129:771-5.
trials will allow us to provide evidence-based recom- 18. Cervantes F, Hernandez-Boluda JC, Alvarez A, Nadal E,
mendations in the future. Montserrat E. Danazol treatment of idiopathic myelofibrosis
with severe anemia. Haematologica 2000;85:595-9.
19. Huang J, Li CY, Mesa RA, Wu W, Hanson CA, Pardanani A,
et al. Risk factors for leukemic transformation in patients
References with primary myelofibrosis. Cancer 2008;112:2726-32.
20. Petti MC, Latagliata R, Spadea T, Spadea A, Montefusco E,
1. Tefferi A, Thiele J, Orazi A, Kvasnicka HM, Barbui T, Aloe Spiriti MA, et al. Melphalan treatment in patients with
Hanson CA, et al. Proposals and rationale for revision of the myelofibrosis with myeloid metaplasia. Br J Haematol 2002;
World Health Organization diagnostic criteria for poly- 116:576-81.
cythemia vera, essential thrombocythemia, and primary 21. Giovanni B, Michelle E, Letizia C, Filippo B, Pier Paolo P,
myelofibrosis: recommendations from an ad hoc interna- Giuseppe V, et al. Thalidomide in myelofibrosis with
tional expert panel. Blood 2007;110:1092-7. myeloid metaplasia: a pooled-analysis of individual patient
2. Barosi G, Mesa RA, Thiele J, Cervantes F, Campbell PJ, data from five studies. Leuk Lymphoma 2002;43:2301-7.
Verstovsek S, et al. Proposed criteria for the diagnosis of 22. Abgrall JF, Guibaud I, Bastie JN, Flesch M, Rossi JF, Lacotte-
post-polycythemia vera and post-essential thrombocy- Thierry L, et al. Thalidomide versus placebo in myeloid
themia myelofibrosis: a consensus statement from the metaplasia with myelofibrosis: a prospective, randomized,
International Working Group for Myelofibrosis Research and double-blind, multicenter study. Haematologica 2006;91:
Treatment. Leukemia 2008;22:437-8. 1027-32.
3. Dupriez B, Morel P, Demory JL, Lai JL, Simon M, Plantier I, 23. Mesa RA, Steensma DP, Pardanani A, Li CY, Elliott M,
et al. Prognostic factors in agnogenic myeloid metaplasia: a Kaufmann SH, et al. A phase 2 trial of combination low-dose
report on 195 cases with a new scoring system. Blood 1996; thalidomide and prednisone for the treatment of myelofibro-
88:1013-8. sis with myeloid metaplasia. Blood 2003;101:2534-41.
4. Cervantes F, Dupriez B, Pereira A, Passamonti F, Reilly JT, 24. Tefferi A, Cortes J, Verstovsek S, Mesa RA, Thomas D, Lasho
Morra E, et al. New prognostic scoring system for primary TL, et al. Lenalidomide therapy in myelofibrosis with
myelofibrosis based on a study of the International Working myeloid metaplasia. Blood 2006;108:1158-64.
Group for Myelofibrosis Research and Treatment. Blood 25. Mesa RA, Yao X, Cripe LD, Li CY, Tefferi A, Tallman MS.
2009;113:2895-901. Lenalidomide and prednisone for primary and post poly-

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 197 |
14th Congress of the European Hematology Association

cythemia vera/essential thrombocythemia myelofibrosis RC, Scherle PA, et al. The clinical phenotype of myelofibro-
(MF): an Eastern Cooperative Oncology Group (ECOG) sis encompasses a chronic inflammatory state that is favor-
Phase II Trial. Blood 2008;112:1753. ably altered by INCB018424, a selective inhibitor of JAK1/2.
26. Tefferi A, Verstovsek S, Barosi G, Passamonti F, Roboz GJ, Blood 2008;112:2804.
Gisslinger H, et al. Pomalidomide therapy in anemic patients 44. Wang JC, Chen C, Dumlao T, Naik S, Chang T, Xiao YY, et
with myelofibrosis: results from a Phase-2 randomized al. Enhanced histone deacetylase enzyme activity in primary
Multicenter Study. Blood 2008;112:663. myelofibrosis. Leuk Lymphoma 2008;49:2321-7.
27. Vreugdenhil G, Smeets M, Feelders RA, van Eijk HG. Iron 45. Quintas-Cardama A, Tong W, Kantarjian H, Thomas D,
chelators may enhance erythropoiesis by increasing iron Ravandi F, Kornblau S, et al. A phase II study of 5-azacitidine
delivery to haematopoietic tissue and erythropoietin for patients with primary and post-essential thrombo-
response in iron-loading anaemia. Acta Haematol 1993; cythemia/polycythemia vera myelofibrosis. Leukemia 2008;
89:57-60. 22:965-70.
28. Smeets ME, Vreugdenhil G, Holdrinet RS. Improvement of 46. Mesa RA, Verstovsek S, Rivera C, Pardanani A, Hussein K,
erythropoiesis during treatment with deferiprone in a patient Lasho T, et al. 5-Azacitidine has limited therapeutic activity
with myelofibrosis and transfusional hemosiderosis. Am J in myelofibrosis. Leukemia 2009;23:180-2.
Hematol 1996;51:243-4. 47. Danilov AV, Relias V, Feeney DM, Miller KB. Decitabine is an
29. Jensen PD, Heickendorff L, Pedersen B, Bendix-Hansen K, effective treatment of idiopathic myelofibrosis. Br J
Jensen FT, Christensen T, et al. The effect of iron chelation Haematol 2008; 145:131-2.
on haemopoiesis in MDS patients with transfusional iron 48. Shi J, Zhao Y, Ishii T, Hu W, Sozer S, Zhang W, et al. Effects
overload. Br J Haematol 1996;94:288-99. of chromatin-modifying agents on CD34+ cells from
30. Leitch H, Ezzat H, Rollins MD, Goodman TA, Leger CS, patients with idiopathic myelofibrosis. Cancer Res 2007; 67:
Wong DH, et al. Improved survival in red blood cell transfu- 6417-24.
sion dependent patients with primary myelofibrosis (PMF) 49. Rambaldi A, Dellacasa CM, Salmoiraghi S, Spinelli O, Ferrari
receiving iron chelation therapy. Blood 2008;112:1748. ML, Gattoni E, et al. A Phase 2A study of the histone-
31. Larsen TS, Pallisgaard N, de Stricker K, Moller MB, deacetylase inhibitor ITF2357 in patients with Jak2V617F
Hasselbalch HC. Limited efficacy of hydroxyurea in lower- positive chronic myeloproliferative neoplasms. Blood 2008;
ing of the JAK2 V617F allele burden. Blood 2008; 112:1750. 112:100.
32. Mesa RA, Nagorney DS, Schwager S, Allred J, Tefferi A. 50. Ottmann OG, Spencer A, Prince HM, Bhalla KN, Fischer T,
Palliative goals, patient selection, and perioperative platelet Liu A, et al. Phase IA/II study of oral panobinostat (LBH589),
management: outcomes and lessons from 3 decades of a novel pan-deacetylase inhibitor (DACi) demonstrating effi-
splenectomy for myelofibrosis with myeloid metaplasia at cacy in patients with advanced hematologic malignancies.
the Mayo Clinic. Cancer 2006;107:361-70. Blood 2008;112:958.
33. Malmaeus J, Akre T, Adami HO, Hagberg H. Early postoper- 51. Giles FJ, Cooper MA, Silverman L, Karp JE, Lancet JE,
ative course following elective splenectomy in haematologi- Zangari M, et al. Phase II study of SU5416--a small-molecule,
cal diseases: a high complication rate in patients with myelo- vascular endothelial growth factor tyrosine-kinase receptor
proliferative disorders. Br J Surg 1986;73:720-3. inhibitor in patients with refractory myeloproliferative dis-
34. Tefferi A, Mesa RA, Nagorney DM, Schroeder G, Silverstein eases. Cancer 2003;97:1920-8.
MN. Splenectomy in myelofibrosis with myeloid metapla- 52. Giles FJ, List AF, Carroll M, Cortes JE, Valickas J, Chen BL, et
sia: a single-institution experience with 223 patients. Blood. al. PTK787/ZK 222584, a small molecule tyrosine kinase
2000; 95:2226-33. receptor inhibitor of vascular endothelial growth factor
35. Pardanani A, Phyliky RL, Li CY, Tefferi A. 2-Chlorodeoxy- (VEGF), has modest activity in myelofibrosis with myeloid
adenosine therapy for disseminated Langerhans cell histiocy- metaplasia. Leuk Res 2007;31:891-7.
tosis. Mayo Clin Proc 2003;78:301-6. 53. Verrucci M, Pancrazzi A, Aracil M, Martelli F, Guglielmelli P,
36. Barosi G, Bordessoule D, Briere J, Cervantes F, Demory JL, Paz MF, et al. Aplidin improves megakaryocytopoiesis and
Dupriez B, et al. Response criteria for myelofibrosis with halts neo-angiogenesis in the Gata1low murine model of
myeloid metaplasia: results of an initiative of the European myelofibrosis. Blood 2008;112:2787.
Myelofibrosis Network (EUMNET). Blood 2005;106:2849- 54. Quintas-Cardama A, Kantarjian HM, Giles F, Verstovsek S.
53. Pegylated interferon therapy for patients with Philadelphia
37. Tefferi A, Barosi G, Mesa RA, Cervantes F, Deeg HJ, Reilly JT, chromosome-negative myeloproliferative disorders. Semin
et al. International Working Group (IWG) consensus criteria Thromb Hemost 2006;32:409-16.
for treatment response in myelofibrosis with myeloid meta- 55. Levy B, Vandris K, Adriano F, Goldman J, Silver RT. Recom-
plasia, for the IWG for Myelofibrosis Research and binant interferon α (rIFNα) may retard progression of early
Treatment (IWG-MRT). Blood 2006;108:1497-503. primary myelofibrosis (PM) by reducing splenomegaly and
38. Verstovsek S, Kantarjian HM, Pardanani AD, Burn T, Vaddi by changing marrow morphology. Blood 2008;112:1758.
K, Redman J, et al. Characterization of JAK2 V617F Allele 56. Wang Y, Fiskus W, Natarajan K, Jillella A, Quadt C, Ataja P,
Burden in Advanced Myelofibrosis (MF) Patients: No et al. Co-Treatment with JAK2 inhibitor TG101209 and
Change in V617F:WT JAK2 Ratio in Patients with High panobinostat or hsp90 inhibitor AUY922 attenuates mutant
Allele Burdens despite Profound Clinical Improvement JAK2-V617F levels and activity in human myeloproliferative
Following Treatment with the JAK Inhibitor, INCB018424. disorder cells. Blood 2008;112:3739.
Blood 2008; 112:2802. 57. Guardiola P, Anderson JE, Bandini G, Cervantes F, Runde V,
39. Shah NP, Olszynski P, Sokol L, Verstovsek S, Hoffman R, List Arcese W, et al. Allogeneic stem cell transplantation for
AF, et al. A Phase I Study of XL019, a Selective JAK2 agnogenic myeloid metaplasia: a European Group for Blood
Inhibitor, in patients with primary myelofibrosis, post-poly- and Marrow Transplantation, Societe Francaise de Greffe de
cythemia vera, or post-essential thrombocythemia myelofi- Moelle, Gruppo Italiano per il Trapianto del Midollo Osseo,
brosis. Blood 2008;112:98. and Fred Hutchinson Cancer Research Center Collaborative
40. Verstovsek S, Kantarjian HM, Pardanani AD, Thomas D, Study. Blood 1999;93:2831-8.
Cortes J, Mesa RA, et al. The JAK inhibitor, INCB018424, 58. Rondelli D, Barosi G, Bacigalupo A, Prchal JT, Popat U,
demonstrates durable and marked clinical responses in pri- Alessandrino EP, et al. Allogeneic hematopoietic stem-cell
mary myelofibrosis (PMF) and post-polycythemia/essential transplantation with reduced-intensity conditioning in inter-
thrombocythemia myelofibrosis (Post PV/ETMF). Blood mediate- or high-risk patients with myelofibrosis with
2008; 112:1762. myeloid metaplasia. Blood 2005;105:4115-9.
41. Mesa RA, Schwager S, Radia D, Cheville A, Hussein K, 59. Deeg HJ, Gooley TA, Flowers ME, Sale GE, Slattery JT,
Niblack J, et al. Assessment and monitoring of constitutional Anasetti C, et al. Allogeneic hematopoietic stem cell trans-
symptoms in patients with myelofibrosis: a proposed new plantation for myelofibrosis. Blood 2003;102:3912-8.
instrument the Myelofibrosis Symptom Assessment Form 60. Daly A, Song K, Nevill T, Nantel S, Toze C, Hogge D, et al.
(MFSAF). Blood 2008;112:1754. Stem cell transplantation for myelofibrosis: a report from
42. Mesa RA, Verstovsek S, Kantarjian HM, Pardanani AD, two Canadian centers. Bone Marrow Transplant 2003;32:35-
Friedman S, Newton R, et al. INCB018424, a Selective 40.
JAK1/2 Inhibitor, Significantly Improves the compromised 61. Devine SM, Hoffman R, Verma A, Shah R, Bradlow BA,
nutritional status and frank cachexia in patients with Stock W, et al. Allogeneic blood cell transplantation follow-
myelofibrosis (MF). Blood 2008;112:1760. ing reduced-intensity conditioning is effective therapy for
43. Tefferi A, Kantarjian HM, Pardanani AD, Mesa RA, Newton older patients with myelofibrosis with myeloid metaplasia.

| 198 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

Blood 2002;99:2255-8. tion in myelofibrosis using conventional or reduced-intensi-


62. Hessling J, Kroger N, Werner M, Zabelina T, Hansen A, ty conditioning regimens. Br J Haematol 2006;135:367-73.
Kordes U, et al. Dose-reduced conditioning regimen fol- 68. Patriarca F, Bacigalupo A, Sperotto A, Isola M, Soldano F,
lowed by allogeneic stem cell transplantation in patients Bruno B, et al. Allogeneic hematopoietic stem cell transplan-
with myelofibrosis with myeloid metaplasia. Br J Haematol tation in myelofibrosis: the 20-year experience of the
2002;119:769-72. Gruppo Italiano Trapianto di Midollo Osseo (GITMO).
63. Kroger N, Zabelina T, Schieder H, Panse J, Ayuk F, Stute N, Haematologica 2008;93:1514-22.
et al. Pilot study of reduced-intensity conditioning followed 69. Ditschkowski M, Elmaagacli AH, Trenschel R, Steckel NK,
by allogeneic stem cell transplantation from related and Koldehoff M, Beelen DW. No influence of V617F mutation in
unrelated donors in patients with myelofibrosis. Br J JAK2 on outcome after allogeneic hematopoietic stem cell
Haematol 2005; 128:690-7. transplantation (HSCT) for myelofibrosis. Biol Blood
64. Kroeger N, Holler E, Kobbe G, Bornhaeuser M, Schwerdt- Marrow Transplant 2006;12:1350-1.
feger R, Nagler A, et al. Dose-reduced conditioning followed 70. Li Z, Gooley T, Applebaum FR, Deeg HJ. Splenectomy and
by allogeneic stem cell transplantation in patients with hemopoietic stem cell transplantation for myelofibrosis.
myelofibrosis. Results from a Multicenter Prospective Trial
of the Chronic Leukemia Working Party of the European Blood 2001;97:2180-1.
Group for Blood and Marrow Transplantation (EBMT). 71. Ciurea SO, Sadegi B, Wilbur A, Alagiozian-Angelova V,
Blood 2007; 110:683. Gaitonde S, Dobogai LC, et al. Effects of extensive spleno-
65. Byrne JL, Beshti H, Clark D, Ellis I, Haynes AP, Das-Gupta E, megaly in patients with myelofibrosis undergoing a reduced
et al. Induction of remission after donor leucocyte infusion intensity allogeneic stem cell transplantation. Br J Haematol
for the treatment of relapsed chronic idiopathic myelofibro- 2008;141:80-3.
sis following allogeneic transplantation: evidence for a 'graft 72. Cervantes F, Tassies D, Salgado C, Rovira M, Pereira A,
vs. myelofibrosis' effect. Br J Haematol 2000;108:430-3. Rozman C. Acute transformation in nonleukemic chronic
66. Kroger N, Badbaran A, Holler E, Hahn J, Kobbe G, Born- myeloproliferative disorders: actuarial probability and main
hauser M, et al. Monitoring of the JAK2-V617F mutation by characteristics in a series of 218 patients. Acta Haematol
highly sensitive quantitative real-time PCR after allogeneic 1991; 85:124-7.
stem cell transplantation in patients with myelofibrosis. 73. Mesa RA, Li CY, Ketterling RP, Schroeder GS, Knudson RA,
Blood 2007; 109:1316-21. Tefferi A. Leukemic transformation in myelofibrosis with
67. Merup M, Lazarevic V, Nahi H, Andreasson B, Malm C, myeloid metaplasia: a single-institution experience with 91
Nilsson L, et al. Different outcome of allogeneic transplanta- cases. Blood 2005;105:973-7.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 199 |
Myeloproliferative disorders

Interferon in the treatment of myeloproliferative


disorders

J.-J. Kiladjian A B S T R A C T
Interferon (IFN), the first cytokine discovered, has a wide range of biological properties, including
Assistance Publique, Hôpitaux de immunomodulatory, pro-apoptotic and anti-angiogenic activities that rapidly raised interest in its
Paris, Hôpital Avicenne, Service therapeutic use in malignancies. In addition, IFN-receptor characterization was pivotal in the discov-
d’Hématologie Clinique, Bobigny, ery of the JAK/STAT signaling pathway. Among the large IFN family, IFNα2 is mainly used in therapy.
France; Assistance Publique, Many clinical trials have shown remarkable efficacy of IFNα in bcr-abl-negative myeloproliferative
Hôpitaux de Paris, Hôpital disorders (MPD), especially polycythemia vera (PV), and essential thrombocythemia (ET). In those dis-
Saint-Louis, Centre d’Investigation eases, IFNα induces hematological responses in about 80% of patients, and is able to reduce
Clinique, Paris, France; Université splenomegaly, relieve pruritus and other constitutional symptoms. Yet, its use is limited by toxicity,
Paris 7, Denis Diderot, Paris, France leading to early treatment discontinuation in about 20% of the patients. However, its lack of leuke-
mogenic potential, and its possible use during pregnancy have already made IFNα the drug of choice
Hematology Education: for younger MPD patients. In addition, several studies have shown a probable selective effect of IFNα
the education program for the on the malignant PV and ET clones. In an increasing number of patients, cytogenetic remissions, rever-
annual congress of the European sions to polyclonal hematopoiesis, and more recently, induction of JAK2V617F complete molecular
Hematology Association remissions have been reported. These observations may widen the indications of IFNα in myeloprolif-
erative disorders.
2009;3:200-207

nterferon (IFN) was the first cytokine dis- Janus kinase (JAK)/signal transducers and

I covered, identified 50 years ago by Isaacs


and Lindenmann.1 They found a secreted
factor produced by influenza-virus infected
activators of transcription (STAT) signaling
pathway.

chick cells able to transfer a virus-resistant Biology of interferons


state to previously uninfected cells. This fac- The human family of IFNs includes seven
tor was named IFN because of its ability to species.8 Interferon proteins are historically
interfere with viral growth. Soon, IFNs were characterized as type I (viral) IFNs, or type II
found to be produced in many animals, tis- (immune) IFNs. The latter type consists of a
sues, and cells. About ten mammalian inter- unique member, IFN-γ. Both types I and II
feron species have been discovered, includ- IFNs exert their actions through cognate
ing seven in humans (Table 1).2 IFNs belong receptor complexes, IFNAR and IFNGR
to the class 2 α-helical cytokines that have respectively, present on cell surface mem-
existed in early chordates for about 500 mil- branes.9,10 The most important and best stud-
lion years.3 They play important roles in ied among type I IFNs is IFN-α.
innate and adaptative immunity, which The human IFN-α family comprises 14
raised interest in their therapeutic use. genes, including 2 pseudo-genes or allelic
Production and purification of human leuko- forms (IFNAP22 and IFNA10). Thus in all,
cyte IFNs obtained after infection of human there are 13 proteins expressed from these
white blood cells (WBC) with viruses genes, but only 12 distinct IFN-α and allelic
already allowed their use in clinical trials in forms (the protein produced from IFNA13
the late seventies.4 Such preparation howev- gene being identical to that produced from
er, did not allow complete purity, and IFNA1). Of these, IFN-α2 has been very pre-
although leukocyte interferon consisted pre- dominantly used as a therapeutic agent to
dominantly of IFN-α, small amounts of date.
other IFN species were present. In 1980, IFNs elicit many biological effects that can
cloning of recombinant human IFN-α and even be opposite in different cell types. For
IFN-β allowed production of large amounts example, type I IFN inhibits proliferation
of IFN for characterization, biological activi- and is proapoptotic for many cell types,11 yet
ty studies, and clinical trials.5-7 It is only 20 it prolongs the survival of memory T cells.12
years after their discovery that 2 IFN species, Such a functional diversity can, at least in
IFN-α and -β, could be satisfactorily purified part, be explained by diversity of the IFN
using high performance liquid chromatogra- receptor itself, and that of the signaling path-
phy allowing their analysis and characteriza- ways it may use.
tion.8 In addition, early availability of recom- The type I IFN receptor (IFNAR) complex
binant IFNs offered an opportunity to inves- is unique among cytokine receptors in the
tigate how cytokines induce gene expres- number of cognate ligands, including the 13
sion, culminating in the discovery of the IFN-α subtypes, as well as β, w, ε, and κ

| 200 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

Table 1. Published trials of interferon in polycythemia Vera.

Author, year Number Reduction Freedom Discontinuations Discontinuations Type of IFN


of patients of PHL (%) from PHL (%) 1st year (%) total

Cacciola, 199169 11 9 (82) 5 (45) 0 NA NA


Cimino, 199370 13 10 (77) 4 (31) 0 NA α2b
Finelli, 199371 13 11 (85) 11 (85) 3 (23%) NA NA
Turri, 199172 11 7 (64) 4 (36) 0 0 α2a
Papineschi, 199473 11 9 (82) 8 (73) NA NA α2a
Sacchi, 199474 22 21 (95) 21 (95) 0 0 hl-IFN
Muller, 199575 15 7 (47%) NA 4 (27%) 6 (40%) α2b
Taylor, 199676 17 14 (82) 9 (53) 2 (12%) 6 (35%) NA
Foa, 199877 38 19 (50) 11 (29) 11 (29%) 16 (42%) α2a
Gilbert, 199878 31 NA NA NA 7 (23%) α2b
Stasi, 199879 18 17 (94) 11 (61) 0 NA hl-IFN
Heis, 199980 32 28 (87) 2 (6%) 4 (12%) 10 (31%) NA
Radin, 200381 12 5 (42) 1 (8) NA NA NA
Silver, 200634 55 55 (100) 53 (96) NA 8 (14%) α2a, α2b
Samuelsson, 200682 21 7/9 (78) 4/9 (44) NA 7/23 (30%) peg-α2b
Kiladjian, 200823 37 37 (100) 36 (97) 3 (8%) 13 (35%) peg-α2a
Total 349 260/318 (82%) 182/303 (60%) 27/227 (12%) 82/281 (29%)
NA: data not available, PHL: phlebotomy, hl-IFN: human leukocyte interferon.

species. Conventional type I IFN receptor is comprised Activities and therapeutic use of interferons
of two chains, IFNAR1 and IFNAR2c. IFN binding to IFNs have a wide range of biological activities. They:
this complex induces activation by phosphorylation of (i) stimulate cytotoxic activity of several immune cells
pre-associated JAKs in their cytoplasmic portions: Tyk2, (T-cells, natural killer cells, monocytes, macrophages,
associated to IFNAR1, and Jak1, associated to IFNAR2c. dendritic cells);13 (ii) increase the expression of tumor-
This provides docking sites on the receptor complex for associated surface antigens and other surface molecules,
STAT-1 and -2 proteins that are in turn phosphorylated. such as major histocompatibility complex (MHC) class
Activated STAT-1/STAT-2 heterodimers translocate to I antigens, amplifying the recognition of infected or
the nucleus, associate with IRF-9 (IFN regulatory factor- transformed cells by immune-effectors;14 (iii) induce
9) to form the trimeric ISGF-3 (IFN-stimulated gene fac- and/or activate proapoptotic genes and proteins includ-
tor-3) complex that binds to the ISRE (IFN-stimulated ing TRAIL, caspases, Bak, and Bax; (iv) repress antiapop-
response element) enhancer family element within the totic genes like Bcl-2, IAP (inhibitor of apoptosis pro-
promoters of interferon-regulated genes, leading to their tein); (v) modulate cell differentiation; (vi) display
transcription. However, diversity of IFNAR signaling is antiangiogenic activity.2 Accordingly, the evaluation of
probably in part achieved by the activation of other IFNs as therapeutic agents focused mainly on cancers
pathways, including other STATs (in particular STAT-3) and viral diseases.
and non-STAT proteins.11 These alternative signaling The U.S. Food and Drug Administration approved
human IFN-α2a and IFN-α2b (allelic versions of IFN-α2)
pathways include CrkL, Rap1, MAP-kinases, Vav,
for the treatment of hairy cell leukemia in 1986. About
RAC1, PI 3-kinase, IRS1 and -2, PMRT1, and Sin1.
10 years later, IFN-β1a and IFN-β1b were approved for
Some mechanisms also exist to downregulate IFNs
the treatment of multiple sclerosis. Clinical trials for
signaling. Indeed, if the diversity of signals generated by IFN-α, before and after approval, focused on cancers
IFNs aim at protecting the host against infection and and viral diseases. To date, IFN-α is approved for the
malignant transformation, disproportionate signaling treatment of hairy cell leukemia, malignant melanoma,
may be harmful by inducing leukopenia, and autoim- follicular lymphoma, condylomata acuminata (genital
mune manifestations. Several negative regulatory mole- warts), AIDS-related Kaposi sarcoma, and chronic hep-
cules have been identified, including SOCS-1 atitis B and C. In addition, use of IFN-α is frequent in
(Suppressor Of Cytokine Signaling-1), UBP43 (a type I many cancers, especially in bladder and renal cancers.
IFN-inducible cysteine protease), and SHP-2. SOCS-1
inhibits IFN signaling by binding the c-terminal region Rationale for IFN-α in the treatment of myeloproliferative
of IFNAR1 and blocking the interaction with Tyk2, disorders
UBP43 blocks the JAK1/IFNAR2 interaction, whereas Many activities of IFN-α provide a rationale for its
SHP-2 is implicated in nuclear STAT dephosphorylation, therapeutic use in MPDs. First, IFN-α inhibits in vitro
which appears to be critical for STAT nuclear export, proliferation of hematopoietic progenitors. It has been
another mechanism of regulation of STAT activity. shown that IFN-α can markedly reduce the colony-
SOCS proteins are also thought to regulate JAKs activi- forming ability of erythroid, granulocytic, and mega-
ty by binding to their JH1 catalytic loop and targeting karyocytic progenitors in polycythemia vera (PV) and
those kinases for degradation. primary myelofibrosis (PMF).15-17 Some of these in vitro

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 201 |
14th Congress of the European Hematology Association

studies even suggested that clonal MPD progenitors Table 2. Published trials of interferon in essential thrombo-
might be more sensitive to IFN-α than their normal cythemia.
counterparts might. This possible selective effect on the
MPD clone is further supported by several case reports Author, Year Number Response Discontinuation Type
showing reversion from monoclonal to polyclonal pat- of patients rate (%) n (%) of IFN
terns of hematopoiesis (based on X-chromosome inacti-
vation pattern studies) or disappearance of a chromoso-
Bellucci, 198827 12 NA 4 (33) α2a
mal abnormality present before treatment in IFN-α-
Giles, 198828 18 100 0 α2a &α2b
treated patients.18-21 Using quantification of JAK2V617F Gugliotta, 198983 10 100 NA α2a
mutated alleles in circulating granulocytes as a marker Lazzarino, 198984 26 86 9 (35) α2b
of minimal residual disease, our group has also demon- Giralt, 199185 13 69 NA α2b
strated that IFN-a markedly decreased the proportion of Gisslinger, 199186 20 85 10 (50) α2c
circulating mutated cells in a large cohort of patients.22,23 Sacchi, 199187 35 85 4 (11) α2b
The megakaryocytic lineage seems particularly sensitive Turri, 199172 10 70 1 (10) α2a
to IFN-α, with clear morphological and biochemical Seewann, 199188 19 80 6 (30) α2b
changes of megakaryocytes (MK) in IFN-treated Kasparu, 199289 14 86 0 α2b
patients.24 Furthermore, IFN-α is able to directly repress Rametta, 199490 25 92 NA α2b
megakaryopoiesis by inhibiting thrombopoietin- Berte, 199691 12 83 NA α2a & α2b
induced Mpl receptor signaling.25 Finally, IFN-α antago- Sacchi, 199892 11 100 1 (9) α
nizes platelet-derived growth factor (PDGF) and inhibits Radin, 200381 17 88% NA α2
the growth of marrow-derived fibroblasts, two activi- Alvarado, 200393 11 100 2 (18) peg-α2b
ties that may be useful in treating myelofibrosis. Saba, 200594 20 75 3 (15) α2a
Although the exact mechanisms of action of this Langer, 200595 36 75 13 (36) peg-α2b
cytokine on MPD clones are unknown, theoretically Samuelsson, 200682 21 70 11 (55) peg-α2b
IFN-a could, by acting at the level of the hematopoietic Jabbour, 200796 13 70 NA peg-α2b
stem cell and by enhancing the autologous immune Total 343 84 23
response against transformed cells, eradicate the MPD NA: data not available.
clone in selected cases.

Results of clinical trials using IFN-α in myeloproliferative for therapy. There was no biological evidence to antici-
disorders pate better efficacy of one of those isoforms over the
Clinical trials have shown clear efficacy of IFN-α in other in MPD, and, indeed, efficacy and toxicity profiles
the treatment of all types of MPDs. The pioneer study of these two drugs when used in Ph-negative MPD
of Silver in 1988 was followed by many studies demon- were similar (Tables 1 to 3). More recently, pegylated
strating the clinical efficacy of IFN-α in controlling forms of IFN have been developed. Addition of a poly-
myeloid proliferation and relieving pruritus and other ethyleneglycol (peg) tail produces several benefits,
constitutional symptoms in PV.26 Similar efficacy was at including enhanced plasma half-life, lower toxicity, and
the same time shown in ET.27,28 This review will focus increased drug stability and solubility, without affecting
on the three main Ph-negative MPD: PV, Essential therapeutic activity.29 Use of pegylated IFN translated to
Thrombocythemia (ET), and PMF, and will not analyze improvement for patients, mainly due to longer inter-
IFN-α effects in other MPDs, including chronic myelo- vals between administrations (weekly instead of every
genous leukemia (CML), hypereosinophilic syndromes, 24 to 48 hours). Surprisingly, although pegylation of
and systemic mastocytosis. drugs usually results in a certain decrease in side effects
Many clinical studies have been performed since due to reduced relative peak concentration after each
those first works using various commercial forms of injection, such benefit was not found for IFN as studies
IFN-α. Of note, to date none of these forms is registered comparing standard versus pegylated IFN in hepatitis,
by FDA, EMEA or other countries’ health agencies for and CML patients showed similar rates of toxicity and
Ph-negative MPD. Most of the published studies used treatment withdrawal using one or the other form.30,31
standard IFN-α2a or 2b, which were the first available More than 700 PV, ET or PMF, treated with IFN have

Table 3. Published trials of interferon in primary myelofibrosis.

Author, Year Number of patients Response rate (%) Spleen size reduction Discontinuation (%) Type of IFN
(% of patients)

Gilbert, 199878 22 NA 58 46 α2b


Tefferi, 2001 (35) 11 0 18 64 α2
Heis-Vahidi-Fard, 2001 (97) 9 0 20 67 γ
Radin, 2003 (81) 31 3 33 NA α2
Jabbour, 2007 (96) 11 9 NA 26 peg-α2b
Total 84 3 32 51
NA: data not available.

| 202 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

been published, as shown in Tables 1, 2, and 3, which Hematological toxicity


summarize IFN studies that included more than 10 Hematological toxicity on erythroid and megakary-
patients. Meta-analysis of these studies is difficult. First, ocytic lineages is commonly observed during IFN treat-
various forms of IFN-α were used, and one cannot ment of hepatitis patients (even requiring sometimes
exclude (even though it is unlikely) a differential effect treatment with recombinant erythropoietin), but is very
of those forms on response rate and/or disease evolu- rare in PV and ET. In contrast, PMF patients often expe-
tion. Furthermore, heterogeneous response criteria were rience worsening of anemia or thrombocytopenia dur-
used. Harmonization of response criteria to therapy in ing IFN therapy, requiring treatment discontinuation in
PV and ET, as published for PMF,32,33 would be very use- a substantial number of cases. Neutropenia is rarely
ful, and the MPD working group of the European reported as a limiting toxicity in MPD treatment, again
LeukemiaNet has developed consensus response criteria with the exception of PMF. The most common hemato-
that may be used in future studies. logical side effect of IFN is lymphopenia, which is
Results of published studies show that IFN-α is effec- reversed after IFN treatment discontinuation.8 However,
tive at controlling myeloproliferation in PV and ET. In clinical complications potentially related to lymphope-
almost all studies, IFN-α was able to normalize platelet nia (like viral infections) were not reported in MPD
counts and leukocytosis rapidly, and to reduce erythro- studies.
cytosis, allowing reduction in the need for phle-
botomies within a few months. In both diseases, an Other toxicities
objective response was observed in about 80% of Some chronic IFN toxicities are difficult to prevent or
patients, including complete freedom from phle- control. They include fatigue and musculoskeletal pain,
botomies in PV in 60% of patients (Tables 1 and 2). In two of the most frequent reasons for treatment with-
addition, IFN-α was also able to reduce PV-associated drawal. Depression is seen less often, but a history of
pruritus in a significant number of cases, and appears to depression or psychiatric disorder constitutes a con-
be the most efficient drug for this symptom. However, traindication to IFN therapy. Less frequently reported,
toxicity of IFN was relatively significant, leading to but often noticed by patients’ community, is subtler
treatment discontinuation in almost one quarter of mood changes, like anxiety, irritability, and sullenness,
patients (see below). which may impact social life. Other rare side effects
include skin toxicity (mild injection site reactions, or
Although in vitro data suggested that IFN-α was a
more generalized exanthema or urticaria), hair loss, gas-
good candidate drug for treating bone marrow fibrosis,
trointestinal toxicity, including nausea, diarrhea, and
clinical trials in PMF have been disappointing (Table 3).
weight loss, liver, cardiac and neurologic toxicities.
Almost no objective response was obtained in the 84
An intriguing set of complications of chronic IFN ther-
reported PMF patients. In 30% spleen size decreased on
apy is the development of autoimmune abnormalities,
treatment, while in almost the same proportion of
ranging from single asymptomatic autoantibodies to
patients, it increased. Importantly, IFN toxicity in these overt autoimmune disease. Hypothyroidism, for exam-
studies was much higher than in PV or ET studies, lead- ple, is a rare but well-known complication of IFN treat-
ing to rapid treatment discontinuation (usually after 3 to ment. In addition, other types of autoimmune abnormal-
6 months only) in more than 50% of the patients. One ities have been described in IFN-treated CML patients,
of the main limiting toxicities of IFN in PMF, contrary to including antinuclear autoantibodies, immune-mediated
PV or ET studies, appeared to be worsening of cytope- hemolysis or thrombocytopenia, polyarthritis, glomeru-
nias. lonephritis, and connective tissue diseases.36-39 Some
authors have raised the hypothesis that development of
Side effects of IFN-α autoimmune phenomena in IFN-treated CML patients
One of the main concerns regarding wide use of IFN- could parallel and reflect the anti-leukemic efficacy of
α is toxicity. As shown in Tables 1 and 2, the average the drug, as patients presenting with autoimmune
rate of discontinuation because of toxicity in PV and ET abnormalities had earlier and more frequent cytogenet-
trials with IFN-α was around 25%, almost half of those ic responses.36,37 Outcome of those immunological
withdrawals occurring during the first year of therapy. abnormalities is usually favorable, autoantibodies disap-
A wide range of side effects has been reported with pearing after discontinuation of IFN-α, but they some-
standard IFN-α. times require specific treatment, such as hormone
replacement in patients developing thyroiditis.
Flu like symptoms
Common flu-like symptoms are experienced by most IFN-α and leukemic evolution
patients, including headache, malaise, fever, chills, One major advantage of IFN-α is that it clearly
fatigue, myalgia, back and joint pain. These symptoms appears to be non-leukemogenic, especially compared
usually appear 1 to 3 hours after administration, disap- to most available cytoreductive agents. Indeed, concern
pear within hours, and are generally manageable by pre- has appeared in recent years in MPD regarding the long-
ventive use of paracetamol. In addition, they gradually term risk of hematological evolution to acute leukemia
decrease in intensity within a few weeks of treatment and myelodysplastic syndromes (AL/MDS). Increased
initiation. These symptoms are clearly dose-dependent, risk of AL/MDS has been found to be associated with
and can be prevented or at least limited by starting IFN the use of alkylating agents and radioactive phosphorus
therapy at low doses.34 The frequency and intensity of in randomized clinical trials.40-43 By contrast, a leuke-
those flu-like symptoms are usually less marked using mogenic potential of HU has not been demonstrated in
pegylated forms of IFN. clinical trials, although there is evidence showing that

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 203 |
14th Congress of the European Hematology Association

sequential use of HU and other cytoreductive drugs inhibitors in CML taught us that molecular complete
(alkylating agents, radioactive phosphorus) increases remission does not necessarily equate to cure.62
the risk of AL/MDS.43-46 In addition, several retrospec- Reduction of the JAK2 mutated clone may, however,
tive studies did not find an increased risk of AL in PV impact disease evolution. For example, the percentage
and ET treated with HU alone,47-49 and similar conclu- of circulating mutated JAK2 allele was shown to be sig-
sions were drawn from a recent meta-analysis of HU nificantly higher in PV and ET patients who developed
therapy in sickle cell anemia.50 Still, very long term myelofibrosis, and may be significantly higher in
results of prospective studies in HU treated MPD patients who develop vascular complications.63,64 Some
patients with more than 10 years of follow-up showed authors suggested that IFNα could delay myelofibrosis
a cumulative incidence of AL/MDS of more than 10% development in PV patients34 based on in vitro data, a
beyond 12 years of follow-up, suggesting that the risk benefit that could be mediated by reduction of the pro-
of leukemic evolution could be higher than previously portion of JAK2V617F mutated cells. In addition, the
reported in studies with shorter follow-up.51 The rela- selective effect of IFNa on mutated granulocytes may
tive contribution of HU and of the natural very long- impact the incidence of thrombosis, as it has been
term evolution of the disease in the pathogenesis of shown that leukocyte activation and hemostatic
those late AL/MDS remains unclear. These findings changes were correlated with the presence of the JAK2
have, however, lead published guidelines in ET and PV mutation.63,65,66 Accordingly, the incidence of thrombosis
to recommend the use of clearly non-leukemogenic in IFNα-treated patients was consistently found to be
drugs, like IFNα in younger patients requiring cytore- lower than expected during follow-up.34,55,56 However,
ductive therapy.52,53 such possible reduction in thrombotic risk with IFN has
yet to be demonstrated in a properly designed con-
Has IFN-α a selective effect on myeloproliferative clones? trolled trial.
As for all hematological malignancies, a future pri-
mary objective of Ph-negative MPD treatment should Conclusion and perspectives
be cure; a goal that cannot be achieved with currently Many studies over the last 20 years have shown
available drugs, which have never been shown to influ- remarkable efficacy of IFNa in PV and ET, but its use
ence the natural history of these diseases positively. The remained limited in clinical practice, due to side effects,
situation may change with the development of JAK2 and maybe in some cases to the lack of experience of
inhibitors54 but, to date, only IFNα has been able to physicians with management of IFNα treatment.
induce cytogenetic remissions or reversion from mono- Indeed, in expert hands, low toxicity with long-term use
clonal to polyclonal hematopoiesis in some patients,18-21 of this drug has been reported.34
suggesting that it could eradicate the malignant clone Although many biological properties of IFNα may
and possibly cure the disease in selected cases. By mon- account for its efficacy in MPD, the exact mechanisms
itoring JAK2V617F mutation in 37 PV patients treated of action remain unknown.
with peg-IFNα-2a, we showed that V617F mutation To date, in published guidelines indications of IFNα
could be reduced to undetectable levels (using a PCR for the treatment of PV and ET have generally remained
assay with 1% sensitivity) in circulating granulocytes in limited to high risk patients younger than 40 years, and
24% of patients (molecular remission).23 Such reduction of to pregnancy.52,53 Indeed, when cytoreduction is neces-
the JAK2 mutated clone with IFNα may explain report- sary during pregnancy, many publications have docu-
ed cases of hematological remissions in ET and PV mented the safety of IFNα in MPD mothers, as well as
patients lasting for months after treatment discontinua- fetuses.67,68 However, there are very few data available
tion.55,56 Molecular complete remission with peg-IFNα- on the long-term safety of IFNα in children exposed to
2a has also been described in a case report,57 showing this drug during pregnancy. Recent results suggest,
that JAK2V617F was no longer detectable after only 2 however, that indications of treatment with IFNα
months of treatment, (which was continued for an addi- should be broader. First, the possibility of a higher inci-
tional month) but rapidly re-appeared after peg-IFNα-2a dence of progression to MDS and acute leukemia in the
discontinuation. Comparison with our results suggests very long-term follow-up of PV and ET51 suggests that
that peg-IFNα-2a should not be discontinued as soon as non-leukemogenic drugs like IFNα should be more
molecular CR is reached if one aims at eliminating the widely used in those disorders. An additional advantage
mutated clone. of IFNα may be its ability to reduce specifically the
Reduction of the JAK mutated clone, however, does MPD clone in a large proportion of PV patients, and
not imply disease cure. First, the sensitivity limit of the even to induce complete molecular remissions.23
technique we used (1%) remained low. Furthermore, Confirmation of those findings may widen the indica-
studies comparing clonality (based on X-chromosome tions of IFNα in JAK2-mutated PV and ET, without
inactivation patterns or coexisting cytogenetic abnor- restricting treatment to patients at high risk of vascular
malities) to JAK2 mutant allele frequency suggest that complications. At least, the large experience with IFN
JAK2V617F could be a secondary genetic event in some accumulated for 20 years in MPD treatment we have
PV patients.58-61 If so, elimination of JAK2V617F would described in this review should prompt investigators to
only reflect the evolution of subclones. On the other initiate a direct comparison of this drug versus hydrox-
hand, a theoretical advantage of IFNa compared to more yurea in a randomized trial. Such a trial could answer
specific inhibitors is that being non-specific of any genet- the question of the best current cytoreductive treatment
ic alteration, it may have some activity against MPD in MPDs, in terms of hematological and molecular
clones regardless of their underlying molecular defects. response, as well as reduction of short-term vascular
Finally, experience with IFN or tyrosine-kinase risk, and of long-term risk of evolution to AL/MDS.

| 204 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

The current development of JAK2 inhibitors raises the 18. Hino M, Futami E, Okuno S, Miki T, Nishizawa Y, Morii H.
hope that molecularly targeted therapy may benefit Possible selective effects of interferon α-2b on a malignant
clone in a case of polycythemia vera. Ann Hematol 1993;
MPD patients by reducing the kinase hyper-activity that 66:161-2.
may lead to disease that is more aggressive with higher 19. Liu E, Jelinek J, Pastore YD, Guan Y, Prchal JF, Prchal JT.
incidence of vascular events and of hematological evo- Discrimination of polycythemias and thrombocytoses by
novel, simple, accurate clonality assays and comparison with
lution. Early results of phase 1 to 2 trials with these new PRV-1 expression and BFU-E response to erythropoietin.
agents already seem to indicate that they will play a role Blood 2003;101:3294-301.
in the therapy of symptomatic PMF patients. Their role 20. Massaro P, Foa P, Pomati M, LaTargia ML, Iurlo A, Clerici C,
in the treatment of PV and ET will certainly be studied, et al. Polycythemia vera treated with recombinant interfer-
on-α2a: evidence of a selective effect on the malignant clone.
but one should keep in mind that the vast majority of Am J Hematol 1997;56:126-8.
ET and PV have excellent response rates and significant 21. Messora C, Bensi L, Vecchi A, Longo R, Giacobbi F,
reduction of the vascular risk using conventional pallia- Temperani P, et al. Cytogenetic conversion in a case of poly-
tive therapies, such as HU. The use of much more cythaemia vera treated with interferon-α. Br J Haematol
1994;86:402-4.
expensive drugs with unknown long-term risk will be 22. Kiladjian JJ, Cassinat B, Turlure P, Cambier N, Roussel M,
justified only if they can cure or significantly alter natu- Bellucci S, et al. High molecular response rate of poly-
ral disease history, and in particular, the very long-term cythemia vera patients treated with pegylated interferon α-
2a. Blood. 2006;108:2037-40.
risk of evolution to AL/MDS. 23. Kiladjian JJ, Cassinat B, Chevret S, Turlure P, Cambier N,
Roussel M, et al. Pegylated interferon-α-2a induces complete
hematologic and molecular responses with low toxicity in
References polycythemia vera. Blood 2008;112:3065-72.
24. Chott A, Gisslinger H, Thiele J, Fritz E, Linkesch W,
1. Isaacs A, Lindenmann J. Virus interference. I. The interferon. Radaszkiewicz T, et al. Interferon-α-induced morphological
Proc R Soc Lond B Biol Sci 1957;147:258-67. changes of megakaryocytes: a histomorphometrical study on
2. Pestka S, Krause CD, Walter MR. Interferons, interferon-like bone marrow biopsies in chronic myeloproliferative disor-
cytokines, and their receptors. Immunol Rev 2004;202:8-32. ders with excessive thrombocytosis. Br J Haematol 1990;74:
3. Krause CD, Pestka S. Evolution of the Class 2 cytokines and 10-6.
receptors, and discovery of new friends and relatives. 25. Wang Q, Miyakawa Y, Fox N, Kaushansky K. Interferon-α
Pharmacol Ther 2005;106:299-346. directly represses megakaryopoiesis by inhibiting throm-
4. Cantell K, Hirvonen S, Kauppinen HL, Myllyla G. Production bopoietin-induced signaling through induction of SOCS-1.
of interferon in human leukocytes from normal donors with Blood 2000;96:2093-9.
the use of Sendai virus. Methods Enzymol 1981;78:29-38. 26. Silver RT. Recombinant interferon-α for treatment of poly-
5. Derynck R, Content J, DeClercq E, Volckaert G, Tavernier J, cythaemia vera. Lancet 1988;2:403.
Devos R, et al. Isolation and structure of a human fibroblast 27. Bellucci S, Harousseau JL, Brice P, Tobelem G. Treatment of
interferon gene. Nature 1980;285:542-7. essential thrombocythaemia by α2a interferon. Lancet 1988;
6. Maeda S, McCandliss R, Gross M, Sloma A, Familletti PC, 2:960-1.
Tabor JM, et al. Construction and identification of bacterial 28. Giles FJ, Singer CR, Gray AG, Yong KL, Brozovic M, Davies
plasmids containing nucleotide sequence for human leuko- SC, et al. α-interferon therapy for essential thrombocy-
cyte interferon. Proc Natl Acad Sci USA 1980;77:7010-3. thaemia. Lancet 1988;2:70-2.
7. Nagata S, Taira H, Hall A, Johnsrud L, Streuli M, Ecsodi J, et 29. Kozlowski A, Charles SA, Harris JM. Development of pegy-
al. Synthesis in E. coli of a polypeptide with human leuko- lated interferons for the treatment of chronic hepatitis C.
cyte interferon activity. Nature 1980;284:316-20. BioDrugs 2001;15:419-29.
8. Pestka S. The interferons: 50 years after their discovery, there 30. Glue P, Fang JW, Rouzier-Panis R, Raffanel C, Sabo R, Gupta
is much more to learn. J Biol Chem 2007;282:20047-51. SK, et al. Pegylated interferon-α2b: pharmacokinetics, phar-
9. de Weerd NA, Samarajiwa SA, Hertzog PJ. Type I interferon macodynamics, safety, and preliminary efficacy data.
receptors: biochemistry and biological functions. J Biol Hepatitis C Intervention Therapy Group. Clin Pharmacol
Chem 2007;282:20053-7. Ther 2000;68:556-67.
10. Krause CD, Mei E, Xie J, Jia Y, Bopp MA, Hochstrasser RM, 31. Michallet M, Maloisel F, Delain M, Hellmann A, Rosas A,
et al. Seeing the light: preassembly and ligand-induced Silver RT, et al. Pegylated recombinant interferon α-2b vs
changes of the interferon gamma receptor complex in cells. recombinant interferon α-2b for the initial treatment of
Mol Cell Proteomics 2002;1:805-15. chronic-phase chronic myelogenous leukemia: a phase III
11. Platanias LC. Mechanisms of type-I- and type-II-interferon- study. Leukemia 2004;18:309-15.
mediated signalling. Nat Rev Immunol 2005;5:375-86. 32. Barosi G, Bordessoule D, Briere J, Cervantes F, Demory JL,
12. Tough DF, Sun S, Zhang X, Sprent J. Stimulation of naive and Dupriez B, et al. Response criteria for myelofibrosis with
memory T cells by cytokines. Immunol Rev 1999;170:39-47. myeloid metaplasia: results of an initiative of the European
13. Ortaldo JR, Mason A, Rehberg E, Moschera J, Kelder B, Myelofibrosis Network (EUMNET). Blood 2005;106:2849-
Pestka S, et al. Effects of recombinant and hybrid recombi- 53.
nant human leukocyte interferons on cytotoxic activity of 33. Tefferi A, Barosi G, Mesa RA, Cervantes F, Deeg HJ, Reilly JT,
natural killer cells. J Biol Chem 1983;258:15011-5. et al. International Working Group (IWG) consensus criteria
14. Greiner JW, Guadagni F, Noguchi P, Pestka S, Colcher D, for treatment response in myelofibrosis with myeloid meta-
Fisher PB, et al. Recombinant interferon enhances monoclon- plasia, for the IWG for Myelofibrosis Research and
al antibody-targeting of carcinoma lesions in vivo. Science Treatment (IWG-MRT). Blood 2006;108:1497-503.
1987; 235:895-8. 34. Silver RT. Long-term effects of the treatment of poly-
15. Carlo-Stella C, Cazzola M, Gasner A, Barosi G, Dezza L, cythemia vera with recombinant interferon-α. Cancer
Meloni F, et al. Effects of recombinant α and γ interferons on 2006;107:451-8.
the in vitro growth of circulating hematopoietic progenitor 35. Tefferi A, Elliot MA, Yoon SY, Li CY, Mesa RA, Call TG, et
cells (CFU-GEMM, CFU-Mk, BFU-E, and CFU-GM) from al. Clinical and bone marrow effects of interferon α therapy
patients with myelofibrosis with myeloid metaplasia. Blood in myelofibrosis with myeloid metaplasia. Blood 2001;
1987;70:1014-9. 97:1896.
16. Castello G, Lerza R, Cerruti A, Cavallini D, Bogliolo G, 36. Sacchi S, Kantarjian H, O'Brien S, Cohen PR, Pierce S, Talpaz
Pannacciulli I. The in vitro and in vivo effect of recombinant M. Immune-mediated and unusual complications during
interferon α-2a on circulating haemopoietic progenitors in interferon α therapy in chronic myelogenous leukemia. J
polycythaemia vera. Br J Haematol 1994;87:621-3. Clin Oncol 1995;13:2401-7.
17. Dudley JM, Westwood N, Leonard S, Eridani S, Pearson TC. 37. Steegmann JL, Requena MJ, Martin-Regueira P, De La
Primary polycythaemia: positive diagnosis using the differ- Camara R, Casado F, Salvanes FR, et al. High incidence of
ential response of primitive and mature erythroid progeni- autoimmune alterations in chronic myeloid leukemia
tors to erythropoietin, interleukin 3 and α-interferon. Br J patients treated with interferon-α. Am J Hematol 2003; 72:
Haematol 1990;75:188-94. 170-6.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 205 |
14th Congress of the European Hematology Association

38. Wandl UB, Nagel-Hiemke M, May D, Kreuzfelder E, Kloke 57. Ishii T, Xu M, Zhao Y, Hu WY, Ciurea S, Bruno E, et al.
O, Kranzhoff M, et al. Lupus-like autoimmune disease Recurrence of clonal hematopoiesis after discontinuing pegy-
induced by interferon therapy for myeloproliferative disor- lated recombinant interferon-α2a in a patient with poly-
ders. Clin Immunol Immunopathol 1992;65:70-4. cythemia vera. Leukemia 2007;21:373-4.
39. Zuffa E, Vianelli N, Martinelli G, Tazzari P, Cavo M, Tura S. 58. Kiladjian JJ, Elkassar N, Cassinat B, Hetet G, Giraudier S,
Autoimmune mediated thrombocytopenia associated with Balitrand N, et al. Essential thrombocythemias without
the use of interferon-α in chronic myeloid leukemia. V617F JAK2 mutation are clonal hematopoietic stem cell dis-
Haematologica 1996;81:533-5. orders. Leukemia 2006;20:1181-3.
40. Treatment of polycythaemia vera by radiophosphorus or 59. Kralovics R, Teo SS, Li S, Theocharides A, Buser AS, Tichelli
busulphan: a randomized trial. "Leukemia and Hemato- A, et al. Acquisition of the V617F mutation of JAK2 is a late
sarcoma" Cooperative Group, European Organization for genetic event in a subset of patients with myeloproliferative
Research on Treatment of Cancer (EORTC). Br J Cancer disorders. Blood 2006;108:1377-80.
1981;44:75-80. 60. Nussenzveig RH, Swierczek SI, Jelinek J, Gaikwad A, Liu E,
41. Berk PD, Wasserman L, Fruchtman S, Goldberg JT. Verstovsek S, et al. Polycythemia vera is not initiated by
Treatment of polycythaemia vera, a summary of clinical JAK2V617F mutation. Exp Hematol 2007;35:32-8.
trends conducted by the polycythaemia vera sub-group. In: 61. Rumi E, Passamonti F, Pietra D, Della Porta MG, Arcaini L,
Wasserman L, Berk PD, editors. Treatment of Polycythaemia Boggi S, et al. JAK2 (V617F) as an acquired somatic mutation
Vera, a Summary of Clinical Trends Conducted by the and a secondary genetic event associated with disease pro-
Polycythaemia Vera Study Group. Philadelphia, PA: W.B. gression in familial myeloproliferative disorders. Cancer
Saunders, 1995. p. 166-94. 2006; 107:2206-11.
42. Najean Y, Rain JD. Treatment of polycythemia vera: the use 62. Rousselot P, Huguet F, Rea D, Legros L, Cayuela JM, Maarek
of hydroxyurea and pipobroman in 292 patients under the O, et al. Imatinib mesylate discontinuation in patients with
age of 65 years. Blood 1997;90:3370-7. chronic myelogenous leukemia in complete molecular remis-
43. Najean Y, Rain JD. Treatment of polycythemia vera: use of sion for more than 2 years. Blood 2007;109:58-60.
32P alone or in combination with maintenance therapy using 63. Passamonti F, Rumi E, Pietra D, Della Porta MG, Boveri E,
hydroxyurea in 461 patients greater than 65 years of age. Pascutto C, et al. Relation between JAK2 (V617F) mutation
The French Polycythemia Study Group. Blood 1997;89:2319- status, granulocyte activation, and constitutive mobilization
27. of CD34+ cells into peripheral blood in myeloproliferative
44. Finazzi G, Caruso V, Marchioli R, Capnist G, Chisesi T, disorders. Blood 2006;107:3676-82.
Finelli C, et al. Acute leukemia in polycythemia vera: an 64. Vannucchi AM, Antonioli E, Guglielmelli P, Longo G,
analysis of 1638 patients enrolled in a prospective observa- Pancrazzi A, Ponziani V, et al. Prospective identification of
tional study. Blood 2005;105:2664-70. high-risk polycythemia vera patients based on JAK2(V617F)
45. Murphy S, Peterson P, Iland H, Laszlo J. Experience of the allele burden. Leukemia 2007;21:1952-9.
Polycythemia Vera Study Group with essential thrombo- 65. Falanga A, Marchetti M, Vignoli A, Balducci D, Barbui T.
cythemia: a final report on diagnostic criteria, survival, and Leukocyte-platelet interaction in patients with essential
leukemic transition by treatment. Semin Hematol 1997;34: thrombocythemia and polycythemia vera. Exp Hematol
29-39. 2005;33:523-30.
46. Sterkers Y, Preudhomme C, Lai JL, Demory JL, Caulier MT, 66. Falanga A, Marchetti M, Vignoli A, Balducci D, Russo L,
Wattel E, et al. Acute myeloid leukemia and myelodysplastic Guerini V, et al. V617F JAK-2 mutation in patients with
syndromes following essential thrombocythemia treated essential thrombocythemia: relation to platelet, granulocyte,
with hydroxyurea: high proportion of cases with 17p dele- and plasma hemostatic and inflammatory molecules. Exp
tion. Blood 1998;91:616-22. Hematol 2007;35:702-11.
47. Gangat N, Strand J, Li CY, Wu W, Pardanani A, Tefferi A. 67. Griesshammer M, Struve S, Harrison CM. Essential throm-
Leucocytosis in polycythaemia vera predicts both inferior bocythemia/polycythemia vera and pregnancy: the need for
survival and leukaemic transformation. Br J Haematol 2007; an observational study in Europe. Semin Thromb Hemost
138:354-8. 2006; 32:422-9.
48. Gangat N, Wolanskyj AP, McClure RF, Li CY, Schwager S, 68. Harrison C. Pregnancy and its management in the
Wu W, et al. Risk stratification for survival and leukemic Philadelphia negative myeloproliferative diseases. Br J
transformation in essential thrombocythemia: a single insti- Haematol 2005; 129:293-306.
tutional study of 605 patients. Leukemia 2007;21:270-6. 69. Cacciola E, Giustolisi R, Guglielmo P, Di Raimondo F,
49. Passamonti F, Rumi E, Pungolino E, Malabarba L, Bertazzoni Calogero RD. Recombinant interferon α in the treatment of
P, Valentini M, et al. Life expectancy and prognostic factors polycythemia vera. Blood 1991;77:2790-1.
for survival in patients with polycythemia vera and essential 70. Cimino R, Rametta V, Matera C, Mele G, Mettivier V, Ferrara
thrombocythemia. Am J Med 2004;117:755-61. F. Recombinant interferon α-2b in the treatment of poly-
50. Lanzkron S, Strouse JJ, Wilson R, Beach MC, Haywood C, cythemia vera. Am J Hematol 1993;44:155-7.
Park H, et al. Systematic review: Hydroxyurea for the treat- 71. Finelli C, Gugliotta L, Gamberi B, Vianelli N, Visani G, Tura
ment of adults with sickle cell disease. Ann Intern Med 2008; S. Relief of intractable pruritus in polycythemia vera with
148:939-55. recombinant interferon alfa. Am J Hematol 1993;43:316-8.
51. Kiladjian JJ, Rain JD, Bernard JF, Briere J, Chomienne C, 72. Turri D, Mitra ME, Di Trapani R, Lipari MG, Perricone R,
Fenaux P. Long-term incidence of hematological evolution in Cajozzo A. α-interferon in polycythemia vera and essential
three French prospective studies of hydroxyurea and pipo- thrombocythemia. Haematologica 1991;76:75-7.
broman in polycythemia vera and essential thrombo- 73. Papineschi F, Bucalossi A, Capochiani E, Benedetti E,
cythemia. Semin Thromb Hemost 2006;32:417-21. Bramanti E, Dastoli G, et al. Recombinant α2a interferon and
52. Barbui T, Barosi G, Grossi A, Gugliotta L, Liberato LN, polycythemia vera: clinical results and biological evaluation
Marchetti M, et al. Practice guidelines for the therapy of by means of Fourier-transform infrared microspectroscopy.
essential thrombocythemia. A statement from the Italian Eur J Haematol 1994;53:213-7.
Society of Hematology, the Italian Society of Experimental 74. Sacchi S, Leoni P, Liberati M, Riccardi A, Tabilio A, Tartoni P,
Hematology and the Italian Group for Bone Marrow et al. A prospective comparison between treatment with
Transplantation. Haematologica 2004;89:215-32. phlebotomy alone and with interferon-α in patients with
53. McMullin MF, Bareford D, Campbell P, Green AR, Harrison polycythemia vera. Ann Hematol 1994;68:247-50.
C, Hunt B, et al. Guidelines for the diagnosis, investigation 75. Muller EW, de Wolf JT, Egger R, Wijermans PW, Huijgens PC,
and management of polycythaemia/erythrocytosis. Br J Halie MR, et al. Long-term treatment with interferon-α2b for
Haematol 2005;130:174-95. severe pruritus in patients with polycythaemia vera. Br J
54. Pardanani A. JAK2 inhibitor therapy in myeloproliferative Haematol 1995;89:313-8.
disorders: rationale, preclinical studies and ongoing clinical 76. Taylor PC, Dolan G, Ng JP, Paul B, Collin R, Reilly JT.
trials. Leukemia 2008;22:23-30. Efficacy of recombinant interferon-α (rIFN-α) in polycy-
55. Lengfelder E, Berger U, Hehlmann R. Interferon alpha in the thaemia vera: a study of 17 patients and an analysis of pub-
treatment of polycythemia vera. Ann Hematol 2000;79:103- lished data. Br J Haematol 1996;92:55-9.
9. 77. Foa P, Massaro P, Caldiera S, LaTargia ML, Iurlo A, Clerici C,
56. Lengfelder E, Griesshammer M, Hehlmann R. Interferon-α in et al. Long-term therapeutic efficacy and toxicity of recombi-
the treatment of essential thrombocythemia. Leuk nant interferon-α 2a in polycythaemia vera. Eur J Haematol
Lymphoma 1996;22 Suppl 1:135-42. 1998;60:273-7.

| 206 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

78. Gilbert HS. Long term treatment of myeloproliferative dis- 88. Seewann HL, Zikulnig R, Gallhofer G, Schmid C. Treatment
ease with interferon-α-2b: feasibility and efficacy. Cancer of thrombocytosis in chronic myeloproliferative disorders
1998;83:1205-13. with interferon α-2b. Eur J Cancer 1991;27 Suppl 4:S58-62;.
79. Stasi R, Venditti A, Del Poeta G, Conforti M, Brunetti M, 89. Kasparu H, Bernhart M, Krieger O, Lutz D. Remission may
Bussa S, et al. Role of human leukocyte interferon-α in the continue after termination of rIFN α-2b treatment for essen-
treatment of patients with polycythemia vera. Am J Med Sci tial thrombocythemia. Eur J Haematol 1992;48:33-6.
1998;315:237-41. 90. Rametta V, Ferrara F, Marottoli V, Matera C, Mettivier V,
80. Heis N, Rintelen C, Gisslinger B, Knobl P, Lechner K, Giss- Cimino R. Recombinant interferon α-2b as treatment of
linger H. The effect of interferon α on myeloproliferation essential thrombocythaemia. Acta Haematol 1994;91:126-9.
and vascular complications in polycythemia vera. Eur J
Haematol 1999;62:27-31. 91. Berte R, Vallisa D, Ferrari B, Civardi G, Sbolli G, Cavanna L.
81. Radin AI, Kim HT, Grant BW, Bennett JM, Kirkwood JM, Low-dose interferon α treatment in essential thrombo-
Stewart JA, et al. Phase II study of α2 interferon in the treat- cythemia. Eur J Haematol 1996;56:104-5.
ment of the chronic myeloproliferative disorders (E5487): a 92. Sacchi S, Gugliotta L, Papineschi F, Liberati AM, Rupoli S,
trial of the Eastern Cooperative Oncology Group. Cancer Delfini C, et al. α-interferon in the treatment of essential
2003;98:100-9. thrombocythemia: clinical results and evaluation of its bio-
82. Samuelsson J, Hasselbalch H, Bruserud O, Temerinac S, logical effects on the hematopoietic neoplastic clone. Italian
Brandberg Y, Merup M, et al. A phase II trial of pegylated Cooperative Group on ET. Leukemia 1998;12:289-94.
interferon α-2b therapy for polycythemia vera and essential 93. Alvarado Y, Cortes J, Verstovsek S, Thomas D, Faderl S,
thrombocythemia: feasibility, clinical and biologic effects, Estrov Z, et al. Pilot study of pegylated interferon-α2b in
and impact on quality of life. Cancer 2006;106:2397-405. patients with essential thrombocythemia. Cancer Chemo-
83. Gugliotta L, Bagnara GP, Catani L, Gaggioli L, Guarini A, ther Pharmacol 2003;51:81-6.
Zauli G, et al. In vivo and in vitro inhibitory effect of α-inter- 94. Saba R, Jabbour E, Giles F, Cortes J, Talpaz M, O'Brien S, et
feron on megakaryocyte colony growth in essential throm- al. Interferon α therapy for patients with essential thrombo-
bocythaemia. Br J Haematol 1989;71:177-81. cythemia: final results of a phase II study initiated in 1986.
84. Lazzarino M, Vitale A, Morra E, Gagliardi A, Bernasconi P, Cancer. 2005;103(12):2551-7.
Torromeo C, et al. Interferon α-2b as treatment for 95. Langer C, Lengfelder E, Thiele J, Kvasnicka HM, Pahl HL,
Philadelphia-negative chronic myeloproliferative disorders Beneke H, et al. Pegylated interferon for the treatment of
with excessive thrombocytosis. Br J Haematol 1989;72:173-
7. high risk essential thrombocythemia: results of a phase II
85. Giralt M, Rubio D, Cortes MT, San Miguel J, Steegmann JL, study. Haematologica 2005;90:1333-8.
Serena J, et al. α interferon in the management of essential 96. Jabbour E, Kantarjian H, Cortes J, Thomas D, Garcia-Manero
thrombocythaemia. Eur J Cancer 1991;Suppl 4:S72-4. G, Ferrajoli A, et al. PEG-IFN-α-2b therapy in BCR-ABL-neg-
86. Gisslinger H, Chott A, Scheithauer W, Gilly B, Linkesch W, ative myeloproliferative disorders: final result of a phase 2
Ludwig H. Interferon in essential thrombocythaemia. Br J study. Cancer 2007;110:2012-8.
Haematol 1991;79 Suppl 1:42-7. 97. Heis-Vahidi-Fard N, Forberg E, Eichinger S, Chott A, Lechner
87. Sacchi S, Tabilio A, Leoni P, Riccardi A, Vecchi A, Messora C, K, Gisslinger H. Ineffectiveness of interferon-γ in the treat-
et al. Interferon α-2b in the long-term treatment of essential ment of idiopathic myelofibrosis: a pilot study. Ann Hematol
thrombocythemia. Ann Hematol 1991;63:206-9. 2001;80:79-82.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 207 |
Pediatric hematological malignancies

Myelodysplastic syndromes in children


and adolescents

C. M. Niemeyer A B S T R A C T

Accounting for 4% of hematopoietic neoplasia in childhood, myelodysplastic syndromes (MDS) in


Division of Paediatric Haematology children are far less common than in adults. There is a great heterogeneity in presentation and clini-
and Oncology, Department of
Paediatrics and Adolescent cal course. Apart from inherited bone marrow failure disorders, the genetic changes predisposing chil-
Medicine, Albert-Ludwigs-University, dren to MDS are largely obscure. Monosomy 7 is the most common chromosomal aberration, often
Freiburg, Germany occurring as sole abnormality. Refractory cytopenia is the most common subtype accounting for about
50% of the cases. The recent edition of the World Health Organization (WHO) classification of
hematopoietic neoplasia has incorporated refractory cytopenia (RC) of childhood as provisional enti-
ty. While children with RC and a normal karoytype or trisomy 8 can experience a long stable course
Hematology Education:
the education program for the and may be candidates for a watch-and-wait strategy or immunosuppressive therapy, patients with
annual congress of the European monosomy 7, 7q- or complex karoytypes generally have rapidly progressive disease and are candidates
Hematology Association for hematopoietic stem cell transplantation (HSCT) early in the course of their disease. Similarly, the
therapeutic approach in children for MDS with elevated blast count is primarily curative based on
2009;3:208-213 HSCT. Research on genetic conditions predisposing to MDS in young age, such as inherited syndromes
with bone marrow failure may give important insights into MDS pathogenesis.

yelodysplastic syndromes (MDS) the subdivision in RAEB I (blasts 5-9%) and

M are a heterogeneous group of clonal


hematopoietic stem cells disorders
characterized by ineffective hematopoiesis,
RAEB II (blasts 10-19%), as suggested for
MDS in adults is useful in children. There is
consensus that the relationship between
morphological dysplasia and a variable risk advanced MDS and de novo AML is better
of transformation to MDS-related leukemia. defined by biological and clinical behaviour
MDS belong to the most frequent bone mar- than by blast count.8 Consequently, myeloid
row (BM) disorders in adults, with a crude disease with low blast count and cytogenet-
incidence of about 4/100.000/year.1 In chil- ic abnormalities typically associated with de
dren and adolescents, MDS is rare, account- novo AML is classified as AML. In addition,
ing 4% of hematopoietic malignancies disease with rapid increase in marrow blasts
(Table 1) corresponding to an annual inci- or organ infiltration should be considered de
dence estimated at 1.8 children per million novo AML, which is by far the more com-
children aged 0 to 14 years.2,3 The dypslastic mon disorder. Children with RAEB generally
prodrome of acute myeloid leukemia (AML) have relatively stable counts for weeks or
in infants with Down syndrome is excluded months. A similar biological behaviour is
from these population-based studies of MDS noted in some children with myelodyspla-
and are classified with myeloid leukemia of sia, and blasts 20-29% are considered refrac-
Down syndrome.4,5 tory anemia with excess blasts in transfor-
As expected, there are a number of differ- mation (RAEB-T) in the French-American-
ences between adult and pediatric MDS. British Cooperative Group classification.9
Refractory anemia (RA) with ring siderob- Because these children may not benefit from
lasts and MDS associated with del(5q) chro- intensive chemotherapy, upfront HCST has
mosome are exceedingly rare in children. been suggested by some investigators.8
While in adults with RA, anemia is frequent- MDS after prior chemo- or radiation ther-
ly the main presenting symptom, in child- apy, prior acquired aplastic anemia, in inher-
hood cases of MDS without increased blast ited BM failure disorders and in familial dis-
count, neutropenia and thrombocytopenia ease is generally classified as secondary
are prominent.6 Therefore, refractory cytopenia MDS. All other cases are referred to as pri-
(RC) is felt more suitable for childhood MDS mary MDS, although it is reasonable to
without excess blast count. RC is the most assume that most of these disorders are sec-
common subtype of childhood MDS, ondary to some yet unknown genetic predis-
accounting for more than half of the cases.7 position. These presumed underlying genet-
The importance of multilineage dysplasia in ic changes may also give rise to subtle phe-
RC is unknown. notypic abnormalities observed in many
Refractory anemia with excess blasts children with primary MDS.
(RAEB) in children has similar morphological
and immunophenotypical features than that Genetics
observed in adults. It is unknown whether Monosomy 7 is the most common cyto-

| 208 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

genetic abnormality in childhood MDS, seen in Table 1. Incidence of hematological malignancies in chil-
approximately 30% of the cases.7,10-12 While mono- dren 0 to 14 years. Combined data from Denmark 1980-
somy 7 is associated with a poor prognosis in child- 1991 and British Columbia 1982–1996.2,3
hood AML13 and MDS in adults,14 it is not an unfavor-
able feature in childhood MDS.15 After monosomy 7, Annual incidence
trisomy 8 and trisomy 21 are the most common N % per million
numerical abnormalities. Constitutional trisomy 21 is
usually clinically obvious when present, whereas con-
ALL 815 79 38.5
stitutional trisomy 8 mosaicism may remain unrecog- AML1 115 11 5.4
nized16 and should be tested when trisomy 8 is found MDS1 38 4 1.8
in the bone marrow (BM). Chromosomal abnormali- Myeloid leukemia of DS 19 2 0.9
ties detected in children enrolled in Study 98 of the JMML 25 2 1.2
European Working Group of MDS in Childhood CML 13 1 0.6
(EWOG-MDS) are depicted in Table 3. PV/ET2 3 0 0.1
RAS mutations are detected in childhood MDS at Unclassified 3 0 0.1
low frequency.17 Mutations of the transcription factor, Total 1031 100 48.7
RUNX1 (AML1), with a consequential decrease in its
1
trans-activation potential have been described in AML Excluding Down syndrome; (DS) 2PV: polycythemia vera; ET: essential
thrombocythemia.
M0, therapy related AML, RAEB, RAEB-T, and AML
following MDS and suggests a loss of function as a
mechanism for transformation.18,19 MDS patients with
RUNX1 mutations frequently harbor somatic muta-
tions in genes of the RAS signaling pathway. topenia, and pancytopenia with radioulnar synostosis,
Mutations of TP53 are present in a significant propor- cannot be distinguished from RC by morphology or
tion of cases of MDS and are especially associated histology alone; they have to be excluded by careful
with therapy related MDS.20 physical examination for skeletal and other organ
abnormalities, past and family history and appropriate
Primary myelodysplastic symdrome without increase in laboratory tests. The distinction between RC and SAA
blast count (refractory cytopenia) may be challenging. In contrast to RC, SAA presents
Diagnosis with adipocytosis of the bone marrow spaces with
MDS with less than 5% blasts in the BM is particu- few scattered myeloid cells, absence of erythroid
larly difficult to diagnose, because dysplasia of islands with increased numbers of immature erythrob-
hematopoietic cells is frequently observed in associa- lasts and without any micromegakaryocytes.21
tion with infections, metabolic disorders, nutritional Following immunosuppressive therapy (IST), the his-
deficiencies, and a variety of other diseases. In the tological pattern of SAA can no longer be separated
absence of a cytogenetic marker, the clinical course has from that observed in RC.
to be carefully evaluated before a diagnosis of RC can
be established. Most children present with symptoms Therapy
related to pancytopenia, but in up to 20% of patients, The median time to progression for children with
no clinical signs or symptoms are reported.6 The mean RC and monosomy 7 is less than 2 years.6 Sponta-
corpuscular volume (MCV) of red cells and hemoglo- neous disappearance of monosomy 7 and cytopenia
bin F are usually elevated, but they are also high in the has been noted in some infants,22,23 but remains a rare
majority of patients with inherited BM failure disor- event. In contrast to monosomy 7, patients with tri-
ders, in some patients with acquired severe aplastic somy 8 and other karyotypes may experience a long
anemia (SAA) at diagnosis, and in most patients with stable course of their disease.6
SAA during their clinical course. On PB smears, red
cells usually are macrocytic with anisopoikilocytosis. Watch-and-wait strategy
There may be giant platelets. Neutrophils with pseudo For children with a normal karyotype or chromoso-
Pelger Huet nuclei and/or hypogranulation can often mal abnormalities other than monosomy 7, 7q- or a
be noted.21 complex karyotype, and absence of transfusion
Like in adults, MDS without increase in blast count dependency or neutropenia, a watch-and-wait strategy
in children can present with cytopenia and a hyper- can be appropriate.
plastic BM; however, three quarters of cases in chil-
dren are hypoplastic (Figure 1). The morphological pic- Immunosuppressive therapy
ture of RC with hypocellular BM is similar to that If in these above patients, cytopenia necessitates
observed in cases with normo- or hypercellualr BM. treatment, current therapy options include HSCT or
There is a patchy pattern of erythropoiesis with imma- IST. It has been suggested that autoimmunity con-
ture precursors accompanies by a sparse granulo- tributes to cytopenia of MDS,24 and approximately 35
poiesis and significantly decreased or absent mega- to 50% of adults with MDS have clinically relevant
karyocytes (Table 3). At least two trephine biopsies are responses to IST.25 In children with hypoplastic RC
recommended to detect the characteristic histological with normal karyotype treated with IST, overall and
pattern. Inherited BM failure disorders, such as failure-free survival at 3 years was 89% and 55%,
Fanconi anemia, dyskeratosis congenita, Shwachman respectively.26 However, the long-term outcome
Diamond syndrome, amegakaryocytic thrombocy- remains unknown.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 209 |
14th Congress of the European Hematology Association

Table 2. Results of cytogenetics in study EWOG-MDS 98 for


patients with primary myelodysplastic syndrome (MDS)
and secondary MDS following chemo- or radiation therapy,
inherited BM failure disorder, acquired aplastic anemia or
in familial disease (update of 55).

Karyotype Primary MDS (%) Secondary MDS (%)

RC RAEB/RAEB-T RC RAEB /RAEB-T


(N= 126) (N=86) (N=23) (N=44)

Normal 77 45 39 27
Monosomy 7 12 29 22 32
(+/- 1 additional)
Other aberrations (<3) 10 17 22 11
≥3 aberrations 1 8 17 30
Figure 1. Bone marrow cellularity and karyotype in chil- RC: refractory cytopenia; RAEB: refractory anemia with excess blasts;
dren with primary refractory cytopenia (unpublished data RAEB-T: RAEB in transformation; EWOG-MDS: European Working Group
from EWOG-MDS. of Myelodysplastic Syndrome in Childhood.

Hematopoietic stem cell transplantation New agents


For patients with RC and monosomy 7, 7q- or com- Treatment of adults with MDS has changed dra-
plex karyotypes allogeneic HSCT from an HLA-identi- matically after new agents, such as azacitidine,
cal sibling donor (MSD) or HLA-compatible unrelated decitabine and lenalidomie became available. Lenali-
donor (UD) is recommended early in the course of the domide, an immunomodulatory drug derived from
disease. Following different high intensity regimens the the parent compound thalidomide, has a dramatic
3 to 5-year Kaplan-Meier estimate of event-free survival effect demonstrated in adult patients with del(5q), a
is 75 to 80%.27,28 Recurrence of the original disease is subtype unknown in pediatric MDS. In addition, it is
rarely observed, while transplant-related mortality is becoming increasingly clear that aberrant DNA
the major cause of failure. methylation contributes to the malignant phenotype
HSCT with reduced intensity conditioning may be in MDS. Few investigators have started to tackle the
able to reduce the risk of early and late toxicity. However, epigenetic profile of MDS in childhood.30,31 Because of
this may be counterbalanced by an increased incidence of small cohorts, and the fact that, in contrast to adults,
delayed engraftment, graft failure, mixed chimerism and most children with MDS will receive a hematopoiet-
relapse. Strahm et al.29 report on a pilot study on 19 chil- ic transplant, results obtained in adult MDS will be
dren with RC transplanted following a fludarabine-based difficult to verify in the pediatric age group.
reduced intensity regimen. Overall and event-free sur- Nevertheless, it is plausible to believe that exciting
vival at 3 years was 0.84 and 0.74, respectively. Infections findings with new agents will be in store for child-
were the most frequently observed complication. These hood MDS as well.
results are comparable to those of patients transplanted
following high intensity preparative regimens.

Table 3. Minimal diagnostic criteria for refractory cytopenia of childhood.

Erythropoiesis Granulopoiesis Megakaryopoiesis

Bone marrow aspirate Dysplastic changes* Dysplastic changes# in at Unequivocal micromegakaryocytes,


and/or megaloblastoid least 10% of granulocytic other dysplastic changes§
changes in at least 10% precursors and neutrophils in variable numbers
of erythroid precursors

Bone marrow biopsy A few clusters of at least 20 No minimal diagnostic criteria Unequivocal micromegakaryocytes;
erythroid precursors. Stop in immunohistochemistry is
maturation with increased numbers of obligatory (CD61, CD41),
proerythroblasts. Increasednumbers other dysplastic changes§ in variable numbers
of mitoses.

Peripheral blood Dysplastic changes# in at least


10% of neutrophils
*Abnormal nuclear lobulation, multinuclear cells, nuclear bridges; #Pseudo-Pelger-Huet cells, hypo-or agranularity, giant bands (in cases of severe neutropenia this cri-
teria may not be fulfilled). §Megakaryocytes of variable size with separated nuclei or round nuclei. The absence of megakaryocytes will not exclude refractory cytopenia
of childhood.

| 210 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

Primary myelodysplastic syndrome with increased blast Myelodysplastic syndrome after acquired aplastic anemia
count MDS develops in 10 to 15% of those children with
Allogeneic HSCT is the treatment of choice in chil- aplastic anemia not treated with HSCT.49,50 The initial
dren with MDS and increased blast count. Since most diagnosis of SAA has to be questioned when secondary
patients receive HSCT, the International Prognostic MDS/AML is diagnosed within a few months after
Scoring System (IPSS) for MDS in adults based upon presentation. Risk factors for clonal disease in SAA are
weighted data on BM blasts percentage, cytopenia and not well clarified, but some reports showed that older
cytogenetics is of limited value in children.32 Whether age, splenectomy, multiple courses of IST, duration of
intensive chemotherapy prior to stem cell transplanta- G-CSF therapy, and unresponsiveness to IST are related
tion should routinely be employed is highly controver- with high risk of clonal evolution.49,50
sial.28,33-35 In the United States and United Kingdom, chil-
dren with RAEB and RAEB-T are generally included in Therapy-related myelodysplastic syndrome
pediatric AML trials.33,34 For MDS patients enrolled, Hematological, therapy-related secondary MDS
most AML studies found significant morbidity and mor- (tMDS) after chemo- or radiotherapy can manifest as
tality associated with induction chemotherapy, a com- RC, high grade MDS, or CMML.15 Between 7% to 18%
plete remission rate of less than 60%, many relapses, of MDS cases in children are tMDS.7,37 The pathophysi-
and an overall survival of less than 30%.34,36,37 Data from ology of the development of tMDS is supposed to be
the European Working Group of MDS in Childhood similar in children, adolescents and adults. However, in
(EWOG-MDS) on 101 children with primary RAEB, children and adolescents, the classical distinction
RAEB-T or MDR-AML given allogeneic HSCT after a between alkylator-type and topoisomerase II inhibitor-
preparative regimen consisting of BU 16 mg/kg, CY 120 type tMDS does neither exists clinically51 nor cytogenet-
mg/kg and L-PAM 140 mg/m2 indicate that AML-type ically.52 HSCT is the therapy of choice, but outcome
therapy prior to the grafting procedure does not prolong remains poor.51,53
survival.35 Allogeneic HSCT can cure about half the chil-
dren with advanced primary MDS.27,38 Toxicity of the Familial myelodysplastic syndrome
procedure and relapse rate contribute equally to the Familial occurrence of MDS, especially with -7/7q-,
number of events. Children receiving a graft from an has been reported in a number of cases. Some families
unrelated donor have previously suffered a higher TRM show discordance for -7;15 therefore, it is uncertain
and a lower EFS rate, but more recent studies have whether -7 per se increase the risk for familiar cases.
shown survival following UD-HSCT comparable to The inherited predisposing locus in familial MDS or
MSD-HSCT.39 AML with -7/7q- may not be located on chromosome
7.54 Familial MDS does also occur without -7/7q-.7
Secondary myelodysplastic syndrome
MDS in inherited bone marrow failure disorders Outlook
The risk of myeloid neoplasias in individuals with Clinical progress in rare hematological disorder like
inherited BM failure disorders depends on the specific childhood MDS will only be obtained by enrolling
disorder and the underlying genetic lesion. Among the patients in clinical studies and preserving sufficient
inherited BM failure disorders Fanconi anemia (FA) is material for bio banks. MDS in inherited BM failure dis-
most frequent, followed by hematopoietic neoplasia. orders and familial disease may serve as a model system
MDS or AML develop in as many as 50% of the patients for studying the succeeding genetic changes in leukemic
with FA before the age of 40 years.40 For patients with stem cells.
severe congenital neutropenia, an incidence of
MDS/AML development of about 13% at 8 years of G- The author wishes to acknowledge the relevant discussions
CSF treatment has been reported.41 In most SCN patients and contributions in literature of many colleagues who are
with MDS/AML, acquired mutations in the G-CSF active in several committees and task forces, including that of
receptor are noted, and partial or complete loss of chro- the Dutch Childhood Oncology Group, the International
mosome 7 is found in more than half.42,43 Pediatric AML Group, the International BFM Study Group,
MDS may occur in as many as one third of patients and the International Consortium on Childhood APL.
with Shwachman-Diamond syndrome,44 but less fre-
quently in patients with Diamond-Blackfan anemia.45,46 It
is noteworthy that not all BM failure syndromes predis- References
pose to the development of MDS; for example, patients
with dyskeratosis congenita develop BM failure in 95% 1. Germing U, Strupp C, Kundgen A, Bowen D, Aul C, Haas R,
of the cases, but MDS is rare.47 Heterozygous mutations Gattermann N. No increase in age-specific incidence of
of the transcription factor RUNX1, which is a common myelodysplastic syndromes. Haematologica 2004;89:905-10.
2. Hasle H, Kerndrup G, Jacobsen BB. Childhood myelodysplas-
target for chromosomal translocation in actue leukemia, tic syndrome in Denmark: incidence and predisposing condi-
cause familial thrombocytopenia, with a propensity to tions. Leukemia 1995;9:1569-72.
develop acute myelogenous leukemia (FTP/AML syn- 3. Hasle H, Wadsworth L, Massing BG, McBride M, Schultz KR.
A population-based study of childhood myelodysplastic syn-
drome).48 There are no solid data indicating that avanced drome in British Columbia, Canada. Br J Haematol 1999; 106:
MDS occurring in patients with inherited BM failure dis- 1027-32.
order other than Fanconi anemia differ from MDS in 4. Baumann I, Niemeyer CM, Bennett J, Shannon K, Thiele J.
Myeloid proliferations related to Down syndrome. In:
other children. The patients should be included in series Swerdlow S, Campo, E, Harris, NL, Jaffe ES, Pileri SA, Stein H,
of MDS and the type and the frequency of constitution- et al. editors. WHO Classification of Tumours of Haemato-
al abnormalities should be reported. poietic and Lymphoid Tissues. 4th edition. ARC.
5. Stark J, Baumann I, Creutzig U, Harbott J, Michalova K,

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 211 |
14th Congress of the European Hematology Association

Niemeyer C. Getting the numbers straight in pediatric MDS: ulin as immune-modulating therapy in patients with low-risk
distribution of subtypes after exclusion of Down syndrome. myelodysplastic syndromes. Leukemia 2004;18:460-5.
Pediatr Blood Cancer 2008;50:435-6. 26. Yoshimi A, Baumann I, Fuhrer M, Bergstrasser E, Gobel U,
6. Kardos G, Baumann I, Passmore SJ, Locatelli F, Hasle H, Sykora KW, et al. Immunosuppressive therapy with anti-thy-
Schultz KR, et al. Refractory anemia in childhood: a retrospec- mocyte globulin and cyclosporine A in selected children with
tive analysis of 67 patients with particular reference to mono- hypoplastic refractory cytopenia. Haematologica 2007; 92:
somy 7. Blood 2003;102:1997-2003. 397-400.
7. Passmore SJ, Chessells JM, Kempski H, Hann IM, Brownbill 27. Yusuf U, Frangoul HA, Gooley TA, Woolfrey AE, Carpenter
PA, Stiller CA. Paediatric myelodysplastic syndromes and PA, Andrews RG, et al.Allogeneic bone marrow transplanta-
juvenile myelomonocytic leukaemia in the UK: a population- tion in children with myelodysplastic syndrome or juvenile
based study of incidence and survival. Br J Haematol 2003; myelomonocytic leukemia: the Seattle experience. Bone
121:758-67. Marrow Transplant 2004;33:805-14.
8. Hasle H, Niemeyer CM, Chessells JM, Baumann I, Bennett JM, 28. Locatelli F, Nöllke P, Fischer A. Hematopoietic stem cell trans-
Kerndrup G, et al. A pediatric approach to the WHO classifi- plantation after a myeloablative conditioning regimen in chil-
cation of myelodysplastic and myeloproliferative diseases. dren with refractory cytopenia: results of a retrospective
Leukemia 2003;17:277-82. analysis from the EWOG-MDS group. Leuk Res 2007;31:S39-
9. Bennett JM, Catovsky D, Daniel MT, Flandrin G, Galton 39[Abstract].
DAG, Gralnick HR, et al. Proposed revised criteria for the clas- 29. Strahm B, Locatelli F, Bader P, Ehlert K, Kremens B, Zintl F, et
sification of acute myeloid leukemia. A report of the French- al. Reduced intensity conditioning in unrelated donor trans-
American-British Cooperative Group. Ann Intern Med 1985; plantation for refractory cytopenia in childhood. Bone
103:620-5. Marrow Transplant. 2007;40:329-33
10. Harbott J, Haas OA, Kerndrup G, Michalová K, Sainati L, Slater 30. Flotho C. Is childhood MDS an epigenetic disease? Leuk Res
R, et al. Cytogenetic evaluation of children with MDS and 2007;31:743.
JMML. Results of the European working group of childhood 31. Vidal DO, Paixão VA, Brait M, Souto EX, Caballero OL, Lopes
MDS (EWOG-MDS). Leukemia 2000;14:961. LF, Vettore AL. Aberrant methylation in pediatric myelodys-
11. Luna-Fineman KM, Shannon SK, Atwater J, Davis M, plastic syndrome. Leuk Res 2007;31:75-81.
Masterson J, Ortega, et al . Myelodysplastic and myeloprolif- 32. Hasle H, Baumann I, Bergstrasser E, Fenu S, Fischer A, Kardos
erative disorders of childhood: a study of 167 patients. Blood G, et al. The International Prognostic Scoring System (IPSS) for
1999;93:459-66. childhood myelodysplastic syndrome (MDS) and juvenile
12. Groupe Francais de Cytogénétique Hématologique. Forty-four myelomonocytic leukemia (JMML). Leukemia 2004;18:2008-
cases of childhood myelodysplasia with cytogenetics, docu- 14.
mented by the Groupe Francais de Cytogénétique Héma- 33. Woods WG, Barnard DR, Alonzo TA, Buckley JD, Kobrinsky
tologique. Leukemia 1997;11:1478-85. N, Arthur DC, et al. Prospective study of 90 children requiring
13. Woods WG, Kobrinsky N, Buckley JD, Lee JW, Sanders J, treatment for juvenile myelomonocytic leukemia or
Neudorf S, et al. Timed-sequential induction therapy improves myelodysplastic syndrome: a report from the Children’s
postremission outcome in acute myeloid leukemia: a report Cancer Group. J Clin Oncol 2002;20:434-40.
from the Children's Cancer Group. Blood 1996;87:4979-89. 34. Webb DK, Passmore SJ, Hann IM, Harrison G, Wheatley K,
14. Greenberg P, Cox C, Le Beau MM, Fenaux P, Morel P, Sanz G, Chessells JM. Results of treatment of children with refractory
Sanz M, et al. International scoring system for evaluating prog- anaemia with excess blasts (RAEB) and RAEB in transforma-
nosis in myelodysplastic syndromes. Blood. 1997;89:2079-88. tion (RAEBt) in Great Britain 1990-99. Br J Haematol 2002;
15. Hasle H, Aricò M, Basso G, Biondi A, Cantù-Rajnoldi A, 117:33-9.
Creutzig U, et al. Myelodysplastic syndrome, juvenile myelo- 35. Zecca M, Nollke P, Fischer A. Hematopoietic stem cell trans-
monocytic leukemia, and acute myeloid leukemia associated plantation for advanced primary MDS in children: results of a
with complete or partial monosomy 7. Leukemia 1999; 13: retrospective analysis from the EWOG-MDS group. Leuk Res
376-85. 2007;31:S38-9.
16. Hasle H, Clausen N, Pedersen B, Bendix-Hansen K. 36. Chan GC, Wang WC, Raimondi SC, Behm FG, Krance RA,
Myelodysplastic syndrome in a child with constitutional tri- Chen G, et al. Myelodysplastic syndrome in children: differen-
somy 8 mosaicism and normal phenotype. Cancer Genet tiation from acute myeloid leukemia with a low blast count.
Cytogenet 1995;79:79-81 Leukemia 1997;11:206-11.
17. Jekic B, Novakovic I, Lukovic L, Kuzmanovic M, Popovic B, 37. Sasaki H, Manabe A, Kojima S, Tsuchida M, Hayashi Y, Ikuta
Pastar I, et al. Low frequency of NRAS and KRAS2 gene muta- K, et al. Myelodysplastic syndrome in childhood: a retrospec-
tions in childhood myelodysplastic syndromes. Cancer Genet tive study of 189 patients in Japan. Leukemia. 2001;15:1713-
Cytogenet 2004;154:180-2. 20.
18. Harada H, Harada Y, Niimi H, Kyo T, Kimura A, Inaba T. High 38. Stary J, Locatelli F, Niemeyer CM. Stem cell transplantation for
incidence of somatic mutations in the AML1/RUNX1 gene in aplastic anemia and myelodysplastic syndrome. Bone Marrow
myelodysplastic syndrome and low blast percentage myeloid Transplant 2005;35 Suppl 1:S13-6.
leukemia with myelodysplasia. Blood 2004;103:2316-24. 39. Deeg HJ, Storer B, Slattery JT, Anasetti C, Doney KC, Hansen
19. Chen CY, Lin LI, Tang JL, Bo BS, Tsay W, Chou WC, et al. JA, et al. Conditioning with targeted busulfan and cyclophos-
RUNX1 gene mutation in primary myelodysplastic syndrome- phamide for hemopoietic stem cell transplantation from relat-
the mutation can be detected early at diagnosis or acquired ed and unrelated donors in patients with myelodysplastic syn-
during disease progression and is associated with poor out- drome. Blood 2002;100:1201-7.
come. Br J Haematol 2007;139:405-14. 40. Kutler DI, Singh B, Satagopan J, Batish SD, Berwick M,
20. Pedersen-Bjergaard J, Christiansen DH, Desta F, Andersen MK. Giampietro PF, et al. A 20-year perspective on the Intern-
Alternative genetic pathways and cooperating genetic abnor- ational Fanconi Anemia Registry (IFAR). Blood 2003; 101:
malities in the pathogenesis of therapy-related myelodysplasia 1249-56.
and acute myeloid leukemia. Leukemia 2006;20:1943-49. 41. Dale DC, Cottle TE, Fier CJ, et al. Severe chronic neutropenia:
21. Baumann I, Niemeyer CM, Bennett J, Shannon K, Thiele J: treatment and follow-up of patients in the Severe Chronic
Myelodysplastic syndromes in childhood. In: Swerdlow S, Neutropenia International Registry. Am J Hematol 2003;72:82-
Campo, E, Harris, NL, Jaffe ES, Pileri SA, Stein H, Thiele J, 93.
Vardiman JV, editors. WHO Classification of Tumours of 42. Tidow N, Pilz C, Teichmann B, Müller-Brechlin A, Germes-
Haematopoietic and Lymphoid Tissues. 4th edition. ARC. hausen M, Kasper B, et al.Clinical relevance of point mutations
22. Mantadakis E, Shannon KM, Singer DA, Finklestein J, Chan in the cytoplasmatic domain of the granulocyte-colony stimu-
KW, Hilden JM, et al. Transient monosomy 7: a case series in lating factor gene in patients with severe congenital neutrope-
children and review of the literature. Cancer 1999;85:2655-61. nia. Blood 1997;88:2369-75.
23. Parker TM, Klaassen RJ, Johnston DL. Spontaneous remission 43. Link DC, Kunter G, Kasai Y, Zhao Y, Miner T, McLellan MD,
of myelodysplastic syndrome with monosomy 7 in a young et al. Distinct patterns of mutations occurring in de novo AML
boy. Cancer Genet Cytogenet 2008;182:122-5. versus AML arising in the setting of severe congenital neu-
24. Barrett J, Saunthararajah Y, Molldrem J. Myelodysplastic syn- tropenia. Blood 2007;110:1648-55.
drome and aplastic anemia: distinct entities or diseases linked 44. Dror Y. Schwachman-Diamond syndrome. Pediatr Blood
by a common pathophysiology? Semin Hematol 2000;37:15- Cancer 2005;45:892-901.
29. 45. Janov AJ, Leong T, Nathan DG, Guinan EC. Diamond-
25. Stadler M, Germing U, Kliche KO, Josten KM, Kuse R, Blackfan anemia. Natural history and sequelae of treatment.
Hofmann WK, et al. A prospective, randomised, phase II study Medicine 1996;75:77-8.
of horse antithymocyte globulin vs rabbit antithymocyte glob- 46. Lipton JM, Atsidaftos E, Zyskind I, Vlachos A. Improving clin-

| 212 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

ical care and elucidating the pathophysiology of Diamond 51. Ammann R, Zecca M, Betts D, et al. Characteristics and out-
Blackfan anemia: an update from the Diamond Blackfan come of therapy-related MDS developing after primary cancer
Anemia Registry. Pediatr Blood Cancer 2006;46:558-64. in children. Blood 2007;106:706[Abstract].
47. Vulliamy TJ, Marrone A, Knight SW, Walne A, Mason PJ, 52. Pedersen-Bjergaard J, Andersen MK, Christiansen DH, Nerlov
Dokal ISW, et al. Mutations in dyskeratosis congenita: their C. Genetic pathways in therapy-related myelodysplasia and
impact on telomere length and the diversity of clinical presen- acute myeloid leukemia. Blood 2002;99:1909-12.
tation Blood 2006;107:2680-5. 53. Tsurusawa M, Manabe A, Hayashi Y, Akiyama Y, Kigasawa H,
48. Owen CJ, Toze CL, Koochin A, Forrest DL, Smith CA, Stevens Inada H, et al. Therapy-related myelodysplastic syndrome in
JM, et al. Five new pedigrees with inherited RUNX1 muta- childhood: a retrospective study of 36 patients in Japan. Leuk
tions causing familial platelet disorder with propensity to
myeloid malignancy. Blood 2008:112:4639-45. Res 2005;29:625-32.
49. Locasciulli A, Arcese W, Locatelli F, Di Bona E, Bacigalupo A. 54. Gao Q, Horwitz M, Roulston D, Hagos F, Zhao N, Freireich EJ,
Treatment of aplastic anaemia with granulocyte-colony stim- et al. Susceptibility gene for familial acute myeloid leukemia
ulating factor and risk of malignancy. Lancet 2001;357:43-4. associated with loss of 5q and/or 7q is not localized on the
50. Kojima S, Ohara A, Tsuchida M, Kudoh T, Hanada R, commonly deleted portion of 5q. Genes Chromosomes
Okimoto Y, et al. Risk factors for evolution of acquired aplas- Cancer 2000;28:164-72.
tic anemia into myelodysplastic syndrome and acute myeloid 55. Pui CH, Schrappe M, Ribeiro RC, Niemeyer CM. Childhood
leukemia after immunosuppressive therapy in children. Blood and adolescent lymphoid and myeloid leukemia. Hematology
2002; 100:786-90. Am Soc Hematol Educ Program 2004:118-45.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 213 |
Pediatric hematological malignancies

Pediatric acute myeloid leukemia

G.J.L. Kaspers A B S T R A C T
Pediatric acute myeloid leukemia (AML) has become a curable disease, with the possibility of cur-
Pediatric Oncology/Hematology, ing about two thirds of the patients. However, many events still occur; side-effects are very signifi-
VU University Medical Center, cant, and late-effects can be severe and even life-threatening. Thus, treatment of pediatric AML still
Amsterdam, the Netherlands needs to improve further. While most study groups agree on several principles of treatment, many
unanswered questions and even controversies remain. This also justifies further clinical studies on the
Hematology Education: optimal treatment of pediatric AML. Fortunately, biotechnical developments provide novel treatment
the education program for the targets and targeted drugs, and intensified international collaboration enables such clinical research.
annual congress of the European This makes it realistic to expect major improvements in the treatment of pediatric AML in the next
Hematology Association 10-20 years.
2009;3:214-220

Pediatric acute myeloid leukemia (AML) increased drug resistance by the bulk of
concerns up to 20% of all childhood AML cells. Third, late mortality in 5-10% of
leukemias and has become a curable disease patients (in complete remission) is mainly
over the past three to four decades. While caused by treatment-related complications,
most children with AML still died in the such as infections, bleeding, graft-versus-
early 1970s, recent reports indicate probabil- host-disease, second malignancies and car-
ities of long-term survival of more than diac failure. Fourth, relapsed disease is the
60%.1 This improvement, which unfortu- most frequent event, occurring in 30-40% of
nately is limited to the more privileged coun- patients. These four main types of events
tries, and has certainly not been achieved also indicate how we should improve out-
worldwide, cannot be attributed to the come in pediatric AML.
introduction of many new antileukemic Meanwhile, several main issues remain in
drugs or to innovative treatment modalities. pediatric AML, which will be discussed here
In fact, the drugs to treat AML being used to some extent. First, there is no consensus
nowadays, by most if not all groups have on several elements in the treatment of pedi-
been available for decades. Thus, other fac- atric AML. These include the need for high-
tors should explain the much higher cure dose cytarabine at induction, the need for a
rates. These include the identification of typ- third or even fourth drug in addition to
ical AML-drugs, such as cytarabine and cytarabine and an anthracycline at induc-
anthracyclines, the treatment of patients tion, the optimal number of treatment cours-
according to detailed protocols, improved es, the maximum cumulative dose of anthra-
dosing and scheduling of the drugs, better cyclines to be given in relation to the risk of
risk-group stratification and risk-adapted cardiotoxicity (and for that matter, the safest
therapy, and major improvements in sup- anthracycline), prophylaxis for central nerv-
portive care. The latter includes blood prod- ous system (CNS) relapse, the best risk-
uct support, prevention and treatment of group stratification, and the need for allo-
infections, and temporary admissions to the geneic hematopietic stem cell transplanta-
pediatric intensive care unit to support vital tion (allo-SCT), and if needed, in which
body functions. All of this means that patients. Second, now that it seems that cure
improvements in the treatment of AML can rates are reaching a plateau, we need the
theoretically be achieved in less-privileged introduction of novel agents and innovative
countries as well, if the drugs and supportive treatment modalities. This is especially true
care are available for the majority of in view of the significant late-effects often
patients. An international effort by different found in survivors of pediatric AML. Finally,
study groups to report on long-term clinical with improved outcome and the identifica-
outcome in pediatric AML resulted in a large tion of more and more subgroups, the need
number of excellent manuscripts.1 These for international collaboration increases,
papers clearly illustrate the main events that which is also a challenge.
occur in pediatric AML. First, early death in Since acute promyelocytic leukemia (APL)
2-10% of patients is mainly caused by infec- and myeloid leukemia of Down syndrome
tious and bleeding complications. Second, have become well-recognised separate enti-
refractory or resistant disease in 3-19% (usu- ties, these will not be extensively discussed
ally 5-10%) of patients is caused by in this contribution.

| 214 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

Current controversies in the treatment of newly diagnosed Table 1. Current controversies in the treatment of
pediatric acute myeloid leukemia pediatric acute myeloid leukemia.
Do we need an additional drug at induction? Many groups
use etoposide, or etoposide plus thioguanine, in addi- Need for a third or even fourth drug at induction
tion to the classical combination of cytarabine and Dose of cytarabine at induction
daunorubicin (or another anthracycline/anthracene- Total number of courses of combination chemotherapy
dione). However, it has not convincingly been shown Optimal cumulative dose of anthracyclines
Best anthracycline (including anthracenediones) in terms of cardiotoxicity
that such additional drugs are required at induction.
Need for and type of prophylaxis for central nervous system relapses
Indeed, several pediatric groups still apply a two drug Indications for allogeneic stem cell transplantation
induction. A direct comparison of etoposide and
thioguanine, added to cytarabine and daunorubicin, did
not reveal statistically significant differences in remis-
sion rates, event-free or overall survival.2 The issue of
additional drugs has been addressed in the past by sev- How many courses of chemotherapy are required? Most
eral groups but large, more recent randomized studies in groups now use multiple courses of intensive
children are lacking. Study MRC AML15 recently ran- chemotherapy, first as induction, followed by consoli-
domized conventional induction with cytarabine, dation therapy. The minimum total number is four, as
daunorubicin and etoposide against FLAG plus idaru- applied by the MRC group, and results of that group are
bicin, enrolling both children and adults. In addition, the among the best in the world. Study MRC AML12 did
value of adding gemtuzumab ozogamicin to conven- not show a statistically significant benefit for five cours-
tional induction chemotherapy was studied. Mature es as compared to four, but that question was continued
results are eagerly awaited. Today, complete remissions to be studied in AML15, and pediatric results have not
rates nor event-free or overall survival rates differ to yet been reported. Higher-risk patients theoretically
such an extent between study groups that the best strat- may benefit from an additional (fifth) course of
egy becomes apparent. However, a comparison of dif- chemotherapy, but such a risk-group adapted therapy
ferent schedules in relation to the rate of resistant dis- has not been proven better in pediatric AML. The
ease and prognosis suggested that a cumulative anthra- NOPHO is studying the role of two doses of gem-
cycline dose of less than 100 mg/m2 after one induction tuzumab ozogamicine as post-consolidation therapy,
course is associated with a worse outcome.3 which one might consider an extra course of
Theoretically, we should aim for high quality complete chemotherapy. The Japanese group reported that they
remissions with as low as possible amounts of minimal could safely reduce the number of consolidation cours-
residual disease. Then, it is reasonable to combine drugs es from eight to six to five for standard-risk patients and
with different mechanisms of action, as long as toxicity to six for higher-risk patients.10,11 One may conclude that
remains manageable. between four and six courses of chemotherapy in total
What is the best moment for high-dose cytarabine? Most are usually needed in pediatric AML. Depending on the
groups now assume that high-dose cytarabine should be content of each course, the number of courses will also
incorporated in the treatment, although there are no influence the cumulative dose of anthracyclines or relat-
recent randomized studies in pediatric AML reported to ed drugs. It seems fair to state that the optimal cumula-
support that policy. An older study by the POG with tive dose is unknown. On the other hand, it is quite
overall inferior outcome compared to currently achiev- clear that cumulative doses of anthracyclines above 300
able survival rates did not reveal an improvement with mg/m2 are associated with increasing risk of cardiotoxi-
higher doses of cytarabine.4 However, in adult AML, it city.12
has been shown that high-dose cytarabine at induction Do we need maintenance chemotherapy? Two ran-
results in better postremission outcome.5,6 High-dose domised studies have shown that maintenance
cytarabine as post-remission strategy benefited adults chemotherapy did not improve overall survival.13,14
especially, with core binding factor or normal karyotype In fact, both studies even suggested that maintenance
AML.7 Knowing that pediatric AML on average concerns might be worse because of a lower salvage rate of
better risk, and thus, more chemosensitive patients than relapsed AML cases. Therefore, maintenance therapy
adults with AML, it seems reasonable to extrapolate should not be given, although one cannot exclude the
these data and to indeed incorporate higher-dose of possibility that some patients might actually benefit
cytarabine in our protocols. Moreover, preclinical studies from it.
have shown that AML cells from some patients are more Do we need prophylaxis of CNS relapse? The incidence
resistant to cytarabine, and that in some patients the of AML cells in the cerebrospinal fluid at initial diagno-
active transport of cytarabine into the AML cells is lim- sis is different among different studies, but in the range
ited; a mechanism which is necessary in case of lower of 5-30%. Moreover, CNS involvement at relapse is
concentrations of cytarabine.8,9 Both mechanisms of observed in up to 10% of patients, while it was up to
resistance theoretically can be overcome by higher dose 20% in studies in which CNS prophylaxis had not been
cytarabine. However, these studies do not identify the used.15 Therefore, there are good arguments to apply
optimal moment to use high-dose cytarabine. Again, the prophylaxis of CNS relapse. Most groups successfully
recently closed study MRC AML15 may provide infor- use intrathecal chemotherapy, in that their CNS relapse
mation on this issue, since it does include a comparison rates or cumulative incidence of any relapse do not
of standard low dose and high-dose cytarabine at induc- seem to be higher than that reported by the BFM-AML
tion, although unfortunately other differences in that group, which still uses cranial irradiation. The latter pol-
induction therapy may confound the results. icy is based on a single randomized study, and cranial

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 215 |
14th Congress of the European Hematology Association

Table 2. Incidence and prognostic significance of FLT3-internal tandem duplication in pediatric acute myeloid leukemia.

Authors/ Patient FLT3-ITD FLT-ITD p value Remarks


Year number positivity negativity
and outcome and outcome

Iwai 94 5 (5%) => 89 (95%) => 0.004


1999 5-yr EFS 20% 5-yr EFS 60%

Kondo 64 7 (11%) => 57 (89%) => 0.003 FLT3-ITD positive patients especially more induction failures
1999 5-yr EFS 14% 5-yr EFS 69%

Xu 87 12 (14%) => 75 (86%) => ? FLT3-ITD positive patients: more than 50% induction failure rate
1999 4-yr OS 0% ?

Meshinchi 91 15 (16%) => 76 (84%) => FLT3-ITD positive subgroup more induction failures and more relapses
2001 8-yr EFS 7% 8-yr EFS 44% 0.002
8-yr OS 13% 8-yr OS 50% 0.002

Liang 80 9 (11%) 71 (89%) n.s. Only 6 non-APL patients; all 3 cases with FLT3-ITD and AR>2.0 died
2002

Arrigoni 45 10 (22%) => 35 (78%) => 0.033


2003 5-yr EFS 20% 5-yr EFS 49%

Zwaan 234 27 (12%) => 207 (88%) => FLT3-ITD positive patients with AR>median had a significantly worse outcome
2003 CR rate 70% CR rate 88% 0.01 than FLT3 wild type patients, which was not found for FLT3-ITD positive
5-yr EFS 29% 5-yr EFS 46% 0.005 patients with AR<median (2-yr EFS 44 vs 61%, p=0.26)
5-yr OS 32% 5-yr OS 58% 0.037

Kang 61 4 (7%) => DFS 57 (93%) => ?


2005 0% DFS 52%

Meshinchi 630 77 (12%) => 553 (88%) => <0.001 FLT3-ITD positive patients with AR >0.4 had PFS of 16% vs.72% in patients
2006 4-yr PFS 31% 4-yr PFS 55% with AR<0.4 (p=0.001)

Meshinchi 630 77 (12%) 553 (88%) FLT3-ITD positive patients with a longer ITD length had a worse RFS (19 vs 51%,
2008 p=0.035), OS from CR (21% vs. 67% (p=0.006), and OS at 4 yr (17% vs 51%,
p=0.006) than those with a shorter ITD length

Shimada 158 17 (13%) => 141 (87%) =>


2008 3-yr RR 52% 3-yr RR 30% <0.005
3-yr DFS 40% 3-yr DFS 67% <0.003
3-yr OS 35% 3-yr OS 84% <0.001

Hollink 276 53 (19%) => 223 (81%) => 0.05 Within patients with NPM1 mutation, FLT3-ITD presence or absence: no
2009 5-yr EFS 25% 5-yr EFS 44% 0.04 prognostic impact; similar outcome for ITD positive patients with AR >0.4 and
<0.4
5-yr OS 42% 5-yr OS 63%
APL: acute promyelocytic leukemia; AR: allelic ratio (amount of FLT3 with ITD divided by wild type FLT3); CR: complete remission; DFS: disease-free survival;
EFS: event-free survival; n.s.: not significant; OS: overall survival; PFS: progression-free survival; RFS: relapse-free survival; RR: relapse rate.

irradiation provided a statistically significant benefit Do we need allo-SCT in CR1? There have been no ran-
only when randomized and non-randomized patients domized studies on this important question. There
were pooled, and mainly concerned less bone marrow seems to be an increasing difference in the actual use of
relapses.16 Indeed, a St. Jude study showed that even allo-SCT in CR1 between the USA and European
CNS leukemia at initial diagnosis can be successfully groups. While COG still advocates its use in newly
treated with chemotherapy only.17 In conclusion, pro- diagnosed AML according to its most recent protocol,
phylaxis of CNS relapse is indicated using intrathecal except for core-binding factor leukemia, European
chemotherapy, either single-agent cytarabine or cytara- groups tend to reserve allo-SCT for patients who
bine in combination with prednisolone and methotrex- relapse. Several study groups have done intention-to-
ate, while there seems no place for prophylactic cranial treat analyses, comparing the outcome between
irradiation in the setting of current intensive chemother- patients with and those without a suitable donor. Most
apy. studies seem to demonstrate a decreased incidence of

| 216 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

Table 3. Incidence and type of recurrent cytogenetic and molecular abnormalities in pediatric acute myeloid leukemia.
Actual percentages differ slightly between groups and original studies, except for the t(15;17) translocation reflecting
acute promyelocytic leukemia, the incidence of which is highly dependent on the ethnicity of the population being stud-
ied.

Type of abnormality Percentage of cases Clinical relevance

Cytogenetic subtype
Normal karyotype 20 prognosis average
t(8;21) 12 good prognosis, especially in terms of overall survival
t(9;11) 7 prognosis differs per study group and additional risk factors; MRD marker
trisomy 8 8 prognosis average
trisomy 21 6 prognosis excellent in case of Myeloid Leukemia of Down syndrome
other 11q23 abnormalities 6 heterogeneous group; MRD marker
Inv(16) 6 prognosis good; MRD marker
t(15;17) 5 (may be much higher) prognosis excellent; enables tailored therapy; MRD marker
monosomy 7 3 prognosis poor
7q deletion 2 prognosis average
t(6;9) 2 prognosis supposed to be poor
monosomy 5/5q- 1-2 prognosis supposed to be poor
t(9;22) 0-1 prognosis supposed to be poor
other 21 heterogeneous

Molecular subtype
RAS mutation 17 prognosis average; marker for MRD?
FLT3-ITD 12 prognosis poor; treatment target; MRD monitoring, but be aware of instability
WT1 mutation 12 prognosis poor; suitable for MRD monitoring
KIT mutation 11 prognosis poor in some studies; treatment target; marker for MRD?
NPM1 mutation 8 prognosis good, might depend on FLT3 status; marker for MRD?
CEBPα mutation 6 prognosis good in some recent (unpublished) studies; marker for MRD?
PTPN11 mutation 5 potential treatment target; marker for MRD?
FLT3-TKD 5 treatment target; marker for MRD monitoring?
MRD; minimal residual disease. For all potential treatment targets and MRD markers, the possibility of (in-) stability of the target/marker during the course of the
disease must be taken into account.

relapses in the donor group, but this favourable effect is it is not superior to chemotherapy only.25,26
diminished by increased treatment-related mortality What is a good risk and what a poor risk AML patient?
and perhaps decreased salvageability of patients that This discussion becomes especially important if risk-
relapse after allo-SCT in CR1. Thus, overall survival group directed therapy is being used. Most groups only
rates are similar (at least, at a statistical level) in “donor” use prognostic risk factors for the decision whether a
versus “no-donor” groups. This is especially true in patient is eligible for an allo-SCT or not. Probably the
studies in which chemotherapy alone results in event- only exceptions are the clinical studies done by the St.
free survival rates above 50% and in overall survival Jude’s consortium. In view of the potential use of risk
rates above 60%. In other words, if chemotherapy factors for risk-group directed therapy, it is remarkable
results are suboptimal, the chance that allo-SCT shows that various groups interpret the prognostic significance
a benefit increases and vice versa. Together with the of several such clinical and cell-biological factors differ-
costs associated with allo-SCT and its more severe side- ently. For instance, t(9;22) is considered to be a poor risk
effects and late effects than associated with chemother- factor by the BFM-AML Group, NOPHO and Japanese
apy only,18,19 it seems preferable to apply allo-SCT in JPSLG, but as average by the MRC. Also, the t(9;11) is
CR2 only. This is further supported by recent analyses considered to be a poor risk factor by the BFM-AML
reported by the BFM-AML Group, MRC and even group, as good by NOPHO and as average by MRC and
COG, that poor risk AML patients do not benefit from JPLSG. As final example, NOPHO does not consider
allo-SCT.20-22 The COG group recently reported that FLT3-internal tandem duplication (ITD) to be a poor risk
intermediate risk AML patients benefit from allo-SCT, factor, while the BFM-AML Group and JPLSG do. Table
while good and poor risk patients did not.21 However, 2 summarizes the currently available studies on the
the overall survival with allo-SCT in that intermediate prognostic significance of FLT3-ITD in pediatric AML,
risk group of 62% seems to be in the same range that is as an example of an intensively studied novel prognos-
being achieved by other groups with chemotherapy tic factor.27-38 The studies clearly indicate a worse out-
only.20,23,24 Obviously, innovative therapies should be come associated with FLT3-ITD. However, length of
tested in patients identified to have poor risk AML at ITD, allelic ratio and the concurrent NPM1 status
initial diagnosis. This is very briefly discussed below. should probably be taken into account for risk-group
Autologous “SCT” (reinfusion would be more appropri- directed therapy based on FLT3 status.
ate) is not discussed here. Randomized studies showed Partly the differences between groups in how they

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 217 |
14th Congress of the European Hematology Association

interpret the prognostic significance of a biological fea- Relapsed acute myeloid leukemia
ture reflect differences in implementation of novel data The most frequent event for newly diagnosed
in ongoing and new protocols. It is also likely that dif- patients with AML is a relapse, occurring in about one
ferent conclusions are drawn based on own treatment in three children. Bone marrow usually is involved, and
protocols, which could be explained by chance or by an the CNS in up to 10% of cases (including combined
actual influence of the treatment itself on the prognostic relapses). Arbitrarily defined as within or after 1 year
significance of a given factor. However, it also reflects from initial diagnosis, about 50% of patients have an
the lack of solid data on the prognostic significance of early or late relapse. Outcome from relapse is signifi-
many of these factors in pediatric AML, such as seems cantly better in case of a longer duration of CR1.45,46
true; for example, t(9;22), t(6;9) and multilineage dyspla- However, the international study Relapsed AML
sia. Therefore, international retrospective and prospec- 2001/01 showed early treatment response to be an even
tive studies are necessary to clarify these issues. In fact, more important prognostic factor.47 In that study, early
such collaborative studies have been done and are ongo- response is determined by morphological examination
ing. A good example is the effort coordinated by Hasle of the bone marrow obtained at day 28 from start of
reinduction, and defined as good in case of less than
et al., on the prognostic significance of monosomy and
20% AML blasts, and poor in case of 20% or more of
7q-.39 Ongoing analyses focus on MLL gene rearranged
such blasts. Day 28 in practice is between days 28 and
AML and on the reasons for the clinical heterogeneity
42 from the start of reinduction therapy. In addition to
among AML patients with t(8;21). In general, important treatment response, favorable cytogenetics t(8;21) and
prognostic factors are initial treatment response and inv(16), as initially identified in studies with newly diag-
cytogenetic and molecular abnormalities. The latter bio- nosed children, also seems to be a favorable prognostic
logical factors are summarized in Table 3. factor at relapsed AML (48 and unpublished data).
Molecular abnormalities may also provide leukemia- Study Relapsed AML 2001/01 and several other recent-
specific treatment targets, and have been studied more ly published studies report a probability of long-term
recently.28,40-43 The definition of a good and a poor risk survival from relapsed AML of more than 30%, as sum-
patient will become more important if we can actually marized by Goemans et al.18 Therefore, reinduction
design more tailored therapies, as briefly discussed chemotherapy should be offered to all children and ado-
below in the section “Innovative treatment of pediatric lescents with relapsed AML who can tolerate intensive
AML.” However, instability of treatment targets should treatment. However, patients that respond poorly to the
be taken into account, as has been described for FLT3- first course of reinduction chemotherapy, patients that
ITD.44 do not achieve a second complete remission, and
Are there subgroups of AML that should be treated differ- patients that relapse again should be offered more
ently? There is compelling evidence that APL should be experimental therapy in that setting of a very dismal
treated with drugs, including ATRA, at induction, con- prognosis. Fortunately, innovative treatment seems
solidation and maintenance therapy. Traditionally, APL achievable within 10-20 years from now.
has been treated with high doses of anthracyclines.
However, cumulative doses of more than 600 mg/m2 of Innovative treatment of pediatric acute myeloid leukemia
anthracyclines do not seem justified in such a curable Significant improvements have already been achieved
disease, in view of the severe cardiac late effects that with conventional drugs that have been available for the
may be the consequence. Therefore, the international treatment of AML for decades. Therefore, it seems real-
consortium on childhood (ICC) APL recently developed istic to expect major advances with the development of
ICC APL Study 01, in which ATRA is used intensively, new drugs and especially novel treatment modalities.
together with intermediate-dose cyarabine and anthra- Discussion of all of these challenges is beyond the scope
cyclines at maximum cumulative doses of 355 mg/m2 of this manuscript, but has been reviewed elsewhere.49
for standard risk patients and of 405 mg/m2 for high-risk In brief, innovative treatment will consist of at least four
patients. In addition, minimal residual disease monitor- different modalities. First, the development of novel but
conventional drugs. A good example might be clofara-
ing will be used to enable treatment of disease persist-
bine, a relatively novel nucleoside analogue that is being
ing or relapsing at the molecular level. The protocol
studied in both children and adults with AML. Second,
includes guidelines for such situations, as well as for
the design of drugs with novel mechanisms of action,
frank relapse and CNS relapses. The study will open in often targeted at leukemia-specific abnormalities.
2009. Another group now treated separately by most Typical examples are monoclonal-antibody mediated
groups according to an international protocol concerns treatment, such as with gemtuzumab ozogamicin (tar-
Myeloid Leukemia of Down syndrome. This entity typ- geting CD33-positive cells) and with tyrosine kinase
ically occurs in children below the age of 5 years, who inhibitors (targeting e.g., Flt3- or kit-mutated AML
have the GATA1 mutation in their AML cells. With that cells). Third, improving the graft-versus-leukemia effect
approach, probability of survival for this special sub- of donor cells without an increase in graft-versus-host-
group is above 85%. In the near future, it is likely that disease. Finally, the development of vaccination and
AML patients with activating type I mutations, such as other immunotherapy approaches. Innovative treat-
FLT3-ITD and KIT mutations, will be treated with ment will become more and more tailored and person-
chemotherapy plus tyrosine kinase inhibitors. This will alized, which necessitates the need for large-scale, and
be discussed below in the section “Innovative treatment thus, international collaboration. Such collaborative
of pediatric AML”. efforts will be the only way to enrol a sufficient number
of patients with a specific subtype of AML in a given

| 218 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

study. Despite such an international setting, random- diagnosed children and adolescents with acute myeloid
leukemia: a Childrens Cancer Group study. French LAME
ized studies in large cohorts of patients will become (Leucémie Aiguë Myéloblastique Enfant) Cooperative Group.
more often impossible because of limited patient num- J Clin Oncol 1994;12:2367-77.
bers eligible for subgrou p-directed therapy. Thus, we 15. Pui CH, Schrappe M, Ribeiro RC, Niemeyer CM. Childhood
also need innovative trial designs. Fortunately, interna- and adolescent lymphoid and myeloid leukemia. Hematology
Am Soc Hematol Educ Program 2004:118-145.
tional large-scale collaboration has been realised in the 16. Creutzig U, Ritter J, Zimmermann M, Schellong G. Does cra-
past 10-20 years, especially in the setting of the nial irradiation reduce the risk for bone marrow relapse in
International BFM Study Group, the so-called acute myelogenous leukemia? Unexpected results of the
Childhood Acute Myelogenous Leukemia study BFM-87. J
International Pediatric AML Group, and the Clin Oncol 1993;11:279-86.
International Consortium for Childhood APL. These 17. Abbott BL, Rubnitz JE, Tong X, Srivastava DK, Pui CH,
collaborations, together with the efforts of many pro- Ribeiro RC, et al. Clinical significance of central nervous sys-
tem involvement at diagnosis of pediatric acute myeloid
fessionals in pediatric AML will undoubtedly further leukemia: a single institution’s experience. Leukemia 2003;
improve the outcome of this disease in terms of cure, 17:2090-6.
side-effects and late effects. 18. Goemans BF, Tamminga RY, Corbij CM, Hählen K, Kaspers
GJ. Outcome for children with relapsed acute myeloid
leukemia in the Netherlands following initial treatment
between 1980 and 1998: survival after chemotherapy only?
References Haematologica 2008;93:1418-20.
19. Leung W, Hudson MM, Strickland DK, Phipps S, Srivastava
1. Kaspers GJL, Creutzig U. Pediatric acute myeloid leukemia: DK, Ribeiro RC, et al. Late effects of treatment in survivors of
International progress and future directions. Leukemia childhood acute myeloid leukemia. J Clin Oncol 2000;18:
2005;19:2025-9. 3273-9.
2. Stevens RF, Hann IM, Wheatley K. Marked improvements in 20. Gibson BE, Wheatley K, Hann IN, Stevens RF, Webb D, Hills
outcome with chemotherapy alone in paediatric acute RK, et al. Treatment strategy and long-term results in pediatric
myeloid leukaemia: results of the United Kingdom Medical patients treated in consecutive UK AML trials. Leukemia 2005;
Research Council’s 10th AML trial. MRC Childhood 19:2130-8.
Leukaemia Working Party. Br J Haematol 1998;101:130-40. 21. Horan JT, Alonzo TA, Lyman GH, Gerbing RB, Lange BJ,
3. Kaspers GJL, Ravindranath Y. Acute Myeloid Leukemia in Ravindranath Y, et al. Impact of disease risk on efficacy of
children and adolescents. In: Degos L, Griffin JD, Linch DC, matched related bone marrow transplantation for pediatric
Löwenberg B, editors. Textbook of Malignant Haematology. acute myeloid leukemia: the Children’s Oncology Group. J
London: Martin Dunitz; 2005. p. 617-32. Clin Oncol 2008;26:5797-801.
4. Ravindranath Y, Steuber CP, Krischer J, Civin CI, Ducore J, 22. Reinhardt D, Kremens B, Zimmermann M. No improvement
Vega R, et al. High-dose cytarabine for intensification of early of overall-survival in children with high-risk acute myeloid
therapy of childhood acute myeloid leukemia. J Clin Oncol leukemia by stem cell transplantation in 1st complete remis-
1991;9:572-80. sion. Blood 2006;108:99a[Abstract].
5. Bishop JF, Matthews JP, Young GA, Bradstock K, Lowenthal 23. Creutzig U, Zimmermann M, Lehrnbecher T, et al. Less toxic-
RM. Intensified induction chemotherapy with high dose ity by optimizing chemotherapy, but not by addition of gran-
cytarabine and etoposide for acute myeloid leukemia: a ulocyte colony-stimulating factor in children and adolescents
review and updated results of the Australian Leukemia Study with acute myeloid leukemia: results of AML-BFM 98. J Clin
Group. Leuk Lymphoma 1998;28:15-27. Oncol 2006;24:4499-506.
6. Weick JK, Kopecky KJ, Appelbaum FR, Head DR, Kingsbury 24. Lie SO, Abrahamsson J, Clausen N, Graf N, Hermann J,
LL, Balcerzak SP, et al. A randomized investigation of high- Niemeyer CM, et al. Long-term results in children with AML:
dose versus standard-dose cytosine arabinoside with daunoru- NOPHO-AML Study Group--report of three consecutive tri-
bicin in patients with previously untreated acute myeloid als. Leukemia 2005;19:2090-100.
leukemia: a Southwest Oncology Group study. Blood 1996; 25. Ravindranath Y, Yeager AM, Chang MN, Steuber CP, Krischer
88:2841-51. J, Graham-Pole J, et al. Autologous bone marrow transplanta-
7. Bloomfield CD, Lawrence D, Byrd JC, Carroll A, Pettenati MJ, tion versus intensive consolidation chemotherapy for acute
Tantravahi R, et al. Frequency of prolonged remission duration myeloid leukemia in childhood, Pediatric Oncology Group. N
after high-dose cytarabine intensification in acyte myeloid Engl J Med 1996;334:1428-34.
leukemia varies by cytogenetic subtype. Cancer Res 1998; 58: 26. Woods WG, Neudorf S, Sanders J, Sanders J, Buckley JD,
4173-9. Barnard DR, et al. A comparison of allogeneic bon marrow
8. Hubeek I, Stam RW, Peters GJ, Broekhuizen R, Meijerink JP, transplantation, autologous bone marrow transplantation, and
van Wering ER, et al. The human equilibrative nucleoside aggressive chemotherapy in children with acute myeloid
transporter 1 mediates in vitro cytarabine sensitivity in child- leukemia in remission: a report from the Children’s Cancer
hood acute myeloid leukaemia. Br J Cancer 2005;93:1388-94. Group. Children's Cancer Group. Blood 2001;97:56-62.
9. Zwaan CM, Kaspers GJL. Possibilities for tailored and target- 27. Arrigoni P, Beretta C, Silvestri D, Rossi V, Rizzari C, Valsecchi
ed therapy in paediatric acute myeloid leukaemia. Br J MG, et al. FLT3 internal tandem duplication in childhood
Haematol 2004;127:264-79. acute myeloid leukaemia: association with hyperleucocytosis
10. Tomizawa D, Tabuchi K, Kinoshita A, Hanada R, Kigasawa H, in acute promyelocytic leukaemia. Br J Haematol 2003;120:89-
Tsukimoto I, , et al. Repetitive cycles of high-dose cytarabine 92.
are effective for childhood acute myeloid leukemia: long-term 28. Hollink IH, Zwaan CM, Zimmermann M, Arentsen-Peters TC,
outcome of the children with AML treated on two consecutive Pieters R, Cloos J, et al. Favorable prognostic impact of NPM1
trials of Tokyo Children’s Cancer Study Group. Tokyo gene mutations in childhood acute myeloid leukemia, with
Children's Cancer Study Group. Pediatr Blood Cancer 2007; emphasis on cytogenetically normal AML. Leukemia 2009;23:
49:127-32. 262-70.
11. Tsukimoto I, Tawa A, Horibe K, et al. Intensive regimens with 29. Iwai T, Yokota S, Nakao M, Okamoto T, Taniwaki M,
an increasing dose density of cytarabine improves the out- Onodera N, et al. Internal tandem duplication of the FLT3 gene
come in childhood acute myeloid leukemia. J Clin Oncol 2009 and clinical evaluation in childhood acute myeloid leukemia.
[Epub ahead of print] The Children’s Cancer and Leukemia Study Group, Japan.
12. Kremer LC, van Dalen EC, Offringa M, Ottenkamp J, Voûte Leukemia 1999;13:38-43.
PA. Anthracycline-induced clinical heart failure in a cohort of 30. Kang HJ, Hong SH, Kim IH, Park BK, Han KS, Cho HI, et al.
607 children: long-term follow-up study. J Clin Oncol 2001; Prognostic significance of FLT3 mutations in pediatric non-
19:191-6. promyelocytic acute myeloid leukemia. Leuk Res 2005; 29:
13. Perel Y, Auvrignon A, Leblanc T, Michel G, Reguerre Y, 617-23.
Vannier JP, et al. Treatment of childhood acute myeloblastic 31. Kondo M, Horibe K, Takahashi Y, Matsumoto K, Fukuda M,
leukemia: dose intensification improves outcome and mainte- Inaba J, et al. Prognostic value of internal tandem duplication
nance is of no benefit: multicenter studies of the French LAME of the FLT3 gene in childhood acute myelogenous leukemia.
(Leucémie Aiguë Myéloblastic Enfant) Cooperative Group. Med Pediatr Oncol 1999; 33:525-9.
Leukemia 2005;19:2082-9. 32. Liang DC, Shih LY, Hung IJ, Yang CP, Chen SH, Jaing TH, et al.
14. Wells RJ, Woods WG, Buckley JD, et al. Treatment of newly Clinical relevance of internal tandem duplication of the FLT3

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 219 |
14th Congress of the European Hematology Association

gene in childhood acute myeloid leukemia. Cancer 2002; 94: 41. Goemans BF, Zwaan CM, Miller M, Zimmermann M, Harlow
3292-8. A, Meshinchi S, et al. Mutations in KIT and RAS are frequent
33. Meshinchi S, Woods WG, Stirewalt DL, Sweetser DA, events in pediatric core-binding factor acute myeloid
Buckley JD, Tjoa TK, et al. Prevalence and prognostic signifi- leukemia. Leukemia 2005;19:1536-42.
cance of Flt3 internal tandem duplication in pediatric acute 42. Hollink IH, van den Heuvel-Eibrink MM, Zimmermann M,
myeloid leukemia. Blood 2001;97:89-94. Balgobind BV, Arentsen-Peters ST, Alders M, et al. Clinical rel-
34. Meshinchi S, Alonzo T, Stirewalt DL, Zwaan M, Zimmerman evance of Wilms’ tumor 1 gene mutations in childhood acute
M, Reinhardt D, et al. Clinical implications of FLT3 mutations myeloid leukemia. Blood 2009;[Epub ahead of print]
in pediatric AML. Blood 2006;108:3654-61. 43. Liang DC, Shih LY, Huang CF, Hung IJ, Yang CP, Liu HC, et al.
35. Meshinchi S, Stirewalt DL, Alonzo TA, Boggon TJ, Gerbing
RB, Rocnik JL, et al. Structural and numerical variation of CEBPα mutations in childhood acute myeloid leukemia.
FLT3/ITD in pediatric AML. Blood 2008;111:4930-3. Leukemia 2005;19:410-4.
36. Shimada A, Taki T, Tabuchi K, Taketani T, Hanada R, Tawa 44. Cloos J, Goemans BF, Hess CJ, van Oostveen JW, Waisfisz Q,
A, et al. Tandem duplications of MLL and FLT3 are correlated Corthals S, et al. Stability and prognostic influence of FLT3
with poor prognoses in pediatric acute myeloid leukemia: a mutations in paired initial and relapsed AML samples.
study of the Japanese childhood AML Cooperative Study Leukemia 2006;20:1217-20.
Group. Pediatr Blood Cancer 2008;50:264-9. 45. Stahnke K, Boos J, Bender-Götze C, Ritter J, Zimmermann M,
37. Xu F, Taki T, Yang HW, Hanada R, Hongo T, Ohnishi H, et al. Creutzig U. Duration of first remission predicts remission
Tandem duplication of the FLT3 gene is found in acute lym- rates and long-term survival in children with relapsed acute
phoblastic leukaemia as well as acute myeloid leukaemia but myelogenous leukemia. Leukemia 1998;12:1534-8.
not in myelodysplastic syndrome or juvenile chronic myeloge- 46. Webb DKH. Management of relapsed acute myeloid
nous leukaemia in children. Br J Haematol 1999;105:155-62. leukaemia. Br J Haematol 1999;106:851-9.
38. Zwaan CM, Meshinchi S, Radich JP, Veerman AJ, Huismans 47. Kaspers GJL, Zimmermann M, Reinhardt D. Prognostic signif-
DR, Munske L, et al. FLT3 internal tandem duplication in 234 icance of time to relapse in pediatric AML: results from the
children with acute myeloid leukemia (AML): prognostic sig- International randomised Phase III Study Relapsed AML
nificance and relation to cellular drug resistance. Blood
2003;102:2387-94. 2001/01. Blood 2008;112:976.
39. Hasle H, Alonzo TA, Auvrignon A, Behar C, Chang M, 48. Webb D, Wheatley K, Harrison G, Stevens RF, Hann IM.
Creutzig U, et al. Monosomy 7 and deletion 7q in children and Outcome for children with relapsed acute myeloid leukemia
adolescents with acute myeloid leukemia: an international ret- following initial therapy in the Medical Research Council
rospective study. Blood 2007;109:4641-7. (MRC) AML 10 trial. MRC Childhood Leukaemia Working
40. Goemans BF, Zwaan CM, Martinelli S, Harrell P, de Lange D, Party. Leukemia 1999;13:25-31.
Carta C, et al. Differences in the prevalence of PTPN11 muta- 49. Kaspers GJL, Zwaan CM. Pediatric acute myeloid leukemia:
tions in FAB M5 paediatric acute myeloid leukaemia. Br J towards high-quality cure of all patients. Haematologica 2007;
Haematol 2005;130:801-3. 92:1519-32.

| 220 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Pediatric hematological malignancies

Pediatric myeloproliferative neoplasms

M.L. Randi A B S T R A C T
M.C. Putti1
The increasing knowledge accumulated over recent years in adults with Philadelphia-negative
myeloproliferative disorders (Ph-MPD) has prompted better evaluation of the rare pediatric essential
Dept of Medical and Surgical thrombocythemia (ET), polycythemia vera (PV) and primary idiopathic myelofibrosis (PMF). A few cases
Sciences, Internal Medicine; of PV, ET and PMF in children were published in the English literature and an up-date of these works
1
Dept of Pediatrics, Clinic is here given. The experience recently collected by the Italian Pediatric Hemato-Oncology Association
Hemato-Oncology, University of
Padua Medical School, Italy is also reported. Overall, the findings suggest that pediatric Ph-negative MPD are heterogeneous dis-
eases and before the diagnosis is made, hereditary disorders have to be excluded. Because JAK2 and
other genes mutations are only detectable in a minority of children with sporadic forms, complemen-
Hematology Education: tary markers, such as clonality of hematopoieis, spontaneous erythroid colony growth, and so on
the education program for the should be performed for the diagnosis and new tests have to be identified. Diagnostic criteria that fit
annual congress of the European
pediatric ET, PV and PMF have to be considered. Prognosis in children is still unknown and treatment
Hematology Association
guidelines need to be established; therefore, prospective observations and clinical trials are needed.
2009;3:221-226

hronic Philadelphia-negative myelo- detailed that these diseases are classified as

C proliferative diseases (Ph-MPD), now


better defined as myeloproliferative
neoplasms, are clonal1,2 pathologies charac-
chronic myeloproliferative neoplasm, as
well as chronic myelogenous leukemia and
less common forms (chronic neutrophilic
terized by an increased proliferation of mul- leukemia, chronic eosinophilic leukemia/
tipotent hematopoietic progenitors and a hypereosinophilic syndrome and unclassifi-
prolonged clinical course. They are frequent- able chronic myeloproliferative diseases).
ly complicated by thrombotic and hemor- Regarding pediatric Ph-MPD, a greater
rhagic events and a possible transformation interest has emerged only recently, mainly
to myelofibrosis and/or acute leukemia.3 In on ET. This is not surprising if we consider
the past, they were considered as orphan that this is the Ph-MPD, which manifests
diseases, but the recent identification of typ- not only in median-advanced age but also in
ical mutations of Ph-MPD in adults gave young people.5,16,17 In about 20% of cases, it
new interest to these diseases. In the spring is diagnosed in people below 40 years of
of 2005, five different research groups4-8 age.18
identified the presence of the V617F muta-
tion in the gene of Janus Kinase 2 (JAK2) in Essential thrombocythemia
most Ph-MPD patients. This is a protein ET is an acquired Ph-MPD characterized
involved in the signaling transduction. This by a sustained increase of platelet count over
mutation determines a constitutive activa- the normal level. It is more common (dou-
tion of JAK2, which continuously stimulates ble) in females than in males; its incidence is
STAT54-7 leading to proliferation of normally 1.5/100,000 persons/year and the median
maturing cells. Later, other less common diagnostic age is 65-70 years.19 After the dis-
biological markers in Ph-MPD were found: covery of JAK2V617F mutation in about
exon 12 JAK29 and thrombopoietin receptor 50% of ET14 and of MPLW515K mutation in
(MPL)10 mutations. about 1%,10 the WHO3 developed new diag-
From a clinical point of view, the discov- nostic criteria, which in the presence of a
ery of the biological markers greatly sustained platelet count over 450x109/L,
improved the specificity of the diagnostic large and mature megakaryocytes prolifera-
criteria for Ph-MPD. For a long time, the tion in bone marrow, without PV, PMF,
available criteria were those proposed by the chronic myelogenous leukemia, myelodys-
Polycythemia Vera Study Group (PVSG).11,12 plastic syndromes and causes of re-
In 2001, the first World Health Organization active/secondary thrombocytosis, gave a
(WHO) criteria were published13 and they first line importance to the presence of a
have been improved recently.3 The relevance clonal marker, such as JAK2V617F. An
given to JAK2V617F is justified by the increased platelet number over 500x109/L is
demonstration that this mutation is now extremely common in children to such an
known to occur in virtually all patients extent that a secondary/reactive thrombocy-
affected by Polycythemia Vera (PV) and in tosis is found in 5-6% of hospitalized chil-
half or little more of those affected by essen- dren.20,21 This is mainly due to asplenic con-
tial thrombocythemia (ET) and primary ditions (splenectomy) infections, Kawasaki
myelofibrosis (PMF).14 The WHO13 also syndrome, autoimmunity, tumors, drugs,

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 221 |
14th Congress of the European Hematology Association

Figure 1. Age distribution at diag-


nosis of ET in 350 patients diag-
nosed in University of Padua over
the last 10 years. Pediatric
patients represent 3% of total;
while diagnoses in females are
distributed all over pediatric age,
all males were diagnosed in schol-
ar age. It has to be underlined that
our departments are third level
centres for myeloproliferative neo-
plasms

surgical procedures and iron deficiency. Some other bral district) and two of them died (one myocardial infarc-
pediatric thrombocytosis are found in children from tion and one multiple vein thrombosis).39 Platelet func-
kindreds with familial thrombocytosis.22 Finally, a small tional studies27 did not give evidence of significant alter-
number of children are affected by primary thrombocy- ations as in adults,40 serum EPO was found to be within
tosis; it has been evaluated that 0.09/1,000,000 chil- normal limits,27,41 and no patients with spontaneous ery-
dren/year23 are affected by ET. This means that ET in throid colony (EEC) growth in vitro were found.27
childhood is 60 times lower than in adults.18 The distri- Moreover, in 2004 our group (42) evaluated seven spo-
bution of ET diagnosis in different ages in our experi- radic cases of children with ET and a platelet count over
ence is shown in Figure 1. 900x109/L with normal plasma thrombopoietin (TPO)
The first report of hemorrhagic thrombocythemia in a level, without finding any mutation of TPO or of MPL
child was published in 1960.24 The patient was treated genes.
with radiophosphorus (32P) and died of acute leukemia. A The use of drugs was extremely variable: within the
total of 94 cases, 23 case reports and 10 small series with patients in whom the therapy adopted was reported, 12
less than 15 patients each25-34 and 2 reviews21,35 have been did not receive any drug, 1 received warfarin, 5 aspirin, 2
published on the topic. The children are characterized by alpha-interferon (α-IFN) (in 1 case with aspirin), 5 hydrox-
young age, high platelets counts frequent positive familial yurea, 3 busulphan, 1 radiophosphorus, 2 anagrelide (in 1
history and a quite severe clinical picture, with about case after α-IFN) and 1 polychemotherapy.
20% of hemorrhagic or thrombotic complications. Rare The Italian Pediatric Hemato-Oncology Association
cases of transformation are also reported, possibly related (AIEOP) recently43 collected a retrospective cohort of 90
to drugs use.24,36-38 Evolution to myelofibrosis was found in children (under 16 years of age), all diagnosed to be affect-
three patients (one treated with hydroxyurea and two ed by ET in agreement with the criteria in use at the time
untreated) and to acute leukemia in two (one treated with of the first diagnosis, followed in 14 different pediatric
busulphan and one with phosphorus).32 centers, plus 16 cases pertaining to 6 different families.22
The mean patients’ age at diagnosis was 7.44 years This represents the largest series ever published on this
(range 2 months to 16 years) with only four infants. There topic. The main clinical features of Italian pediatric ET, as
were some more females (1.5:1), the follow-up ranged well as the treatment adopted are summarized in Table 1.
between 10 months and 16 years, the platelet was 600- Not all the diagnostic procedures were used in this
4000x109/L, with half cases with a platelet count over cohort, in particular, the bone marrow biopsy, which is
1500x109/L. In 35 patients, splenomegaly was described. not so easy in children as in adults, because it needs a pro-
It is important to note that some reported cases belong to found sedation. The biological studies were all performed
families with thrombocythemia and these children seem in three centers (Padua, Rome and Turin) utilizing the cen-
to have less thrombotic events then those with sporadic tralization system in use for leukemia studies in Italy.
forms. The risk of hemorrhagic and thrombotic events The work concluded that ET might be heterogeneous,
seem to be higher in children with a platelet count over with different features from adult ET. The children seem
1.500-2.000x109/L. Symptoms were described in 40% of to have a mild clinical pictures (even with high platelets
children, the most common of which was headache. One counts and long follow-up) and headache was the com-
fifth of patients developed major vascular complications monest symptom. Vascular complications are infrequent
(five arterial and nine venous, mainly in splancnic or cere- and do not correlate with a high leukocyte count at diag-

| 222 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

nosis in agreement with Passamonti44 but not with Table 1. Main clinical and laboratory data of 90 Italian
Carobbio45 in adult ET. The myeloproliferative origin of patients with ET.
ET in pediatric age is confirmed only in a minority of chil-
dren46,47 and its correlation with thrombosis, mainly in N° of patients 90
unusual veins, is suggestive as in adults ET.48 While about
one quarter of children with ET carry the JAK2V617F
mutation, no mutation of MPL515 was found, while in Males/Females 28/62
adults with sporadic ET, it is demonstrated in about 1 to Age at diagnosis (range) 6.75 years (1 month-17 years)
Follow-up (years)(range) 5 (2-11)
3% of cases.10 It is interesting to note that the presence of
Plts×109/L (range) 1260 (611-4020)
MPL mutations in ET seems to represent a significant risk WBC109/L 11.75
factor for microvessels disturbances.49 JAK2V617F 16/65 (24%)
Regarding treatment (Table 2), through interview to the EEC positive 17/28
different pediatricians in the different centers, we con- Monoclonality 15/32
cluded that, in absence of any guidelines for ET in chil- MPL/TPO mutation none
dren, the physician’s aims were: (i) to resolve symptoms; No signs, symtoms or complications 52 (55%)
(ii) to reduce platelets at least to 500 to 600 x109/L. Budd Chiari syndrome 2
Low dose aspirin (50-100 mg/daily) have been used Deep Venous Thrombosis 2
alone or associated with cytoreductive agents, mainly Headache 20
hydroxyurea; in one patient aspirin was suspended due to Paresthesia 3
epistaxis. Two patients refused the use of a-IFN and 1 Nosebleed 2
anagrelide for headache. Over a median follow up of 5 Abdominal pain 4
years, no case of transformation under hydroxyurea or Splenomegaly 19
other treatments was observed. Peripheral neuropathy 1
Not surprisingly, in this retrospective study, treatment
has been heterogeneous, not following any established
criteria50-52 and each pediatrician chose based on their own
experience. Aspirin was used mainly in patients with
headache, between cytoreductive drugs hydroxyurea38,53 Table 2. Treatment adopted in 90 Italian children with ET.
and anagrelide were preferred54,55 to α-IFN.56 It is remark- The extreme variability of the options of different physi-
able that a recent paper57 demonstrates a significant cians is evident.
reduction of JAK2V617F allele burden in ET patients after Treatment adopted N° of patients
first year of hydroxyurea therapy. Therefore, considering
that the leukemogenic potential of hydroxyurea in ET is None never 28
still now debated,58 we think that it may be useful to re- Low dose aspirin (ASA) only 16
consider the use of this drug in young patients with ET Warfarin only 1
and a previous major thrombotic event. α-IFN only 3
The observation of the results of these treatments dial Anagrelide only 2
in some relevant questions: Is anti-aggregation necessary? Hydroxyurea only 9
Is it dangerous with the common high level of platelets α-IFN + aspirin 2
counts of children? Is cytoreduction necessary? Which Anagrelide +ASA 3
long-term drug toxicity is predictable in ET children?59 Hydroxyurea + ASA 3
Hydroxyurea >anagrelide 5
Polycythemia vera Hydroxyurea > α-IFN > anagrelide 1
Polycthemias or erythrocytosis are characterized by the Hydroxyurea > anagrelide > α-IFN 2
expansion of erythrocyte compartment in the peripheral α-IFN > anagrelide 3
blood reflected by an increase of erythrocyte count, α-IFN > anagrelide >α-IFN 1
hemoglobin content and hematocrit. Absolute erythrocy- α-IFN > hydroxyurea 4
α-IFN > anagrelide >hydroxyurea 1
tosis,11,60 as defined by the increase of total red cell mass,
Hydroxyurea >α-IFN 3
must be distinguished from relative erythrocytosis caused Pipobroman > hydroxyurea > anagrelide 2
by a severe reduction of the plasma volume. Plateltpheresis > hydroxyurea > anagrelide >α-IFN 1
Absolute secondary erythrocytosis is driven by increase
of erythropoietin (EPO),61 which may be due to physio- α-IFN: α-interferon.
logic response to tissue hypoxia or abnormal EPO produc-
tion or a deregulation of the oxygen-dependent EPO syn-
thesis. The more common causes of secondary erythro- thropoietic compartment is expanded independently of
cytsis in children are renal diseases (cancers, transplanta- EPO level, is extremely rare in pediatric patients. In this
tions, and malformations), EPO-producing tumors group of diseases, primary familial and congenital poly-
(hemangioblastoma, hepatic adenoma) and Down syn- cythemia (PFCP) caused by a number of mutations in
drome.62 The congenital forms of secondary erythrocyto- cytoplasmic domain of the erythropoietin receptor (EPO-
sis may be due to abnormal hemoglobin with high oxy- R) gene70 and polycythemia vera (PV), the only clonal
gen affinity,63 to 2 to 3 bisphosphoglycerate deficiency,64 acquired primary erythrocytosis, are comprehended.62 PV
to altered oxygen-sensing pathway genes (HIF-1-α, is one of the Ph-MPD, its incidence is of 2/100,000 per-
PHD2, VHL).65-69 sons/year19 and almost all the patients carry the acquired
Primary erythrocytosis, a condition in which the ery- JAK2V617F point mutation;14 a somatic gain-of function

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 223 |
14th Congress of the European Hematology Association

mutations affecting JAK2 exon 12 is identified in 25% of mastocytosis, histiocytosis), rheumatic diseases (juvenile
PV patients.9,71 The 2008 WHO3 criteria established that rheumatoid arthritis, systemic lupus erythematosus),
patients with increased hemoglobin or hematocrit, or ele- sickle cell anemia, infectious diseases, vitamin D deficien-
vated RBC mass, in the presence of JAK2V617F or similar cy rickets and severe combined immunodeficiency.
mutation are affected by PV. In the few cases without any At present, PMF has been described, as our best knowl-
mutation, a minor criterion is needed (1-bone marrow tri- edge, in 11 children (7 males and 4 females),32,87-89 mainly
lineage myeloproliferation or 2-subnormal serum EPO in children of Indian or neighboring countries origins.72,90,91
level, or 3-spontaneous EEC growth). The age at diagnosis ranged between 3 months and 13
Few cases of PV in childhood have been published in the years, and almost all were in pre-school age. The numer-
English literature. Before 2005, about 30 cases in children ous neonatal cases suggested a congenital pathogenesis
and adolescents were described.62 In recent years, we found beginning in utero.87 In all the published data, the diagno-
only 18 children with PV;72-75 all presented with plethora sis was performed with a dry tap and the observation a
and hematocrit levels over 65%. The research of JAK2 hypercellular bone marrow with excess of reticulin and
mutations were performed in 17 of these children: 10 had abnormal megakaryocytes. Almost all children had ane-
classic V617F mutation, 2 had exon 12 mutations (in one mia (Hb 50-90 gr/L) with teardrops poikilocytosis. They
case PV developed after treatment for large cell anaplastic had normal to mildly elevated leucocyte counts (5 to
lymphoma)75 and the remaining 5 were JAK2 wild type. 35x109/L), variable platelet counts (10 to 1970x109/L) and
A new case of PV in a 13-year-old girl was found in extramedullary hematopoiesis with or without
Padua University. The patient suffers for headache and splenomegaly and/or hepatomegaly, but no karyotype
undergoes periodical phlebotomies. She has EEC sponta- alteration. In two patients, spontaneous EEC were found.
neous formation and carries JAK2V617F mutation with From a clinical point of view, two patients had spleno-
an allele burden of 35%. Similarly, three other cases in portal thrombosis and one girl died soon after diagnosis
Rome have an allele burden lower than 50% significantly for septicemia while other patients had a long (3 to 21
lower than 13 ET patients.76 years) and relatively benign course. Therefore, the clinical
No indications are available regarding the best manage- and laboratory findings suggested that PMF presenting in
ment of PV in children. Most authors used phlebotomies infancy may represent a distinct entity compared with
to maintain the Ht below 50%,60 but, for example, the use the disease in adults and a conservative approach to clin-
of low-dose aspirin77 is scarce. ical management has been recommended.87
Currently, the AIEOP group has found only one 13-
Myelofibrosis year-old girl affected by PMF in cellular phase (WBC
PMF is a rare Ph-MPD (estimated incidence 0.4- 19.42x109/L, platelets 2444x109/L, Hb 103 g/L). The diag-
0.7/100,000 persons/year)79 usually affecting elderly peo- nosis was performed in agreement with WHO criteria; in
ple reducing the life expectancy.79 PMF manifests clinical- particular, the patient had splenomegaly (16 cm longitudi-
ly as anemia, splenomegaly due to extramedullary nal diameter), teardrops poikilocytosis and increased cir-
hematopoiesis, leukoerythroblastosis and constitutional culating CD34+ cells (73x106/L); her bone marrow was
symptoms.80,81 It implies an increase in the bone marrow hypercellular with a great increase of reticulin and the
fiber with reticulin and/or collagen deposition,82 which presence of anomalous megakaryocytes in clusters. Both
occurs in response to a clonal proliferation of hematopoi- JAK2 and MPL genes were wild type, spontaneous EEC
etic stem cells. It also leads to a profound hyperplasia of were positive while clonality evaluation resulted skew-
morphologically abnormal megakaryocytes (MK) and ing. At present, the girl is treated with anagrelide. A pre-
populations of monocytes that release fibrogenic growth vious therapy with low dose aspirin was discontinued
factors. PMF is characterized by the constitutive mobi- because of gastric ulcer.92
lization of CD34+ cells, as well as endothelial progenitor The treatment of patients with PMF is largely palliative
cells into the peripheral blood.83 Approximately 50% of and therapeutic interventions are mostly used in the
patients with PMF are JAK2V617F positive4 and 5% har- patients with symptoms. A number of drugs have been
bour MPLW515L mutations.84,85 As well as for other Ph- used in PMF, but, at present, they have not been shown to
MPD, and also for PMF, the WHO3 has recently devel- alter the natural history of PMF;93 however, targeted ther-
oped diagnostic criteria. All major criteria are requested: apies for PMF, that is, small molecule inhibitors of JAK2,
(i) proliferation of atypical megakaryocytes with reticulin are now under evaluation to assess their efficacy and their
and/or collagen fibrosis or increased granulocytic prolifer- potential associated toxicities.94,95
ation with decreased erytrhopoiesis; (ii) not meeting
WHO criteria for chronic myelogenous leukemia, PV, Overall, the findings collected show that polycythemia,
myelodysplasia or other myeloid neoplasms; (iii) demon- thrombocythemia and myelofibrosis in childhood are
stration of clonal marker or no evidence of reactive mar- heterogeneous diseases. In the diagnostic screening, the
row fibrosis, together with at least 2 minor criteria ((i) leu- first step should be to investigate the possible presence of
coerythroblastosis; (ii) increased serum LDH; (iii) anemia; a familial occurrence.96 Because genes mutations are
(iv) palpable spleen). PMF and secondary myelofibrosis is detectable in a minority of children, other tests have to be
extremely rare in childhood.86 The last was described as performed (e.g., clonality, EEC) for the diagnosis and new
evolution of neoplastic diseases (myeloproliferative neo- markers have to be searched; diagnostic criteria that fit for
plasms, myelodysplastic syndromes, reticulum cell sarco- pediatric ET, PV and PMF have to be considered.
ma, neuroblastoma, Fanconi anemia, acute leukemia, Prognosis in children is still unknown and treatment
mainly megakaryocytic leukemia carrying t(1;22) translo- guidelines need to be established: therefore, prospective
cation, Hodgkin disease, rabdomyosarcoma, systemic observations and clinical trials are needed.

| 224 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

27. Randi ML, Putti MC, Fabris F, Sainati L, Zanesco L, Girolami A.


References Features of essential thrombocythemia in childhood: a study of
five children. Br J Haematol 2000;108:86-9.
1. Adamson JW, Fialkow PJ, Murphy S, Prchal JF, Steinmann L. 28. Yang RC, Qian LS. Essential thrombocythemia in children: a
Polycythemia vera: stem-cell and probable clonal origin of the report of 9 cases. Br J Haematol 2000;110:1002-14.
disease. N Engl J Med 1976;295:913-6. 29. Florensa F, Zamora L, Besses C, Ortega JJ, Bastida P, Toll, T et
2. Fialkow PJ, Faguet GB, Jacobson RJ, Vaidya K, Murphy S. al. Cultures of myeloid progenitor cells in pediatric essential
Evidence that essential thrombocythemia is a clonal disorder thrombocythemia. Leukemia 2002;16:18767.
with origin in a multipotent stem cell. Blood 1981;58:916-9. 30. Kratz CP, El-Monheim A, Manke EM, Rister M, Rogge T,
3. Tefferi A, Vardiman JW. Classification and diagnosis of myelo- Niemeyer CM. Retrospective analysis of the clinical course of
proliferative neoplasm: the 2008 world health organization cri- 12 children given the diagnosis essential thrombocythemia.
teria and point-of-care diagnostic algorithms. Leukemia 2008; Klin Padiatr 2004;216:349-55.
22:14-22. 31. Lackner H, Urban C, Benesch M, Moser A, Sovinz P, Schwinger
4. Kralovics R, Passamonti F, Buser AS, Teo SS, Tiedt R, Passweg W, et al. Long-term use of anagrelide in the treatment of chil-
JR, et al. A gain-of-function mutation of JAK2 in myeloprolifer- dren with essential thrombocythemia. Eur J Haematol 2006;
ative disorders. N Engl J Med 2005;352:1779-90. 77:358-9.
5. Baxter EJ, Scott LM, Campbell PJ, East C, Fourouclas N, 32. El-Moneim AA, Kratz CP, Boll S, Rister M, Pahl H, Niemeyer
Swanton S, et al. Acquired mutation of the tyrosine kinase CM. Essential versus reactive thrombocythemia in children:
JAK2 in human myeloproliferative disorders. Lancet 2005; 365: retrospective analysis of 12 cases. Pediatr Blood Cancer 2007;
1054-61. 49:52-5.
6. Levine RL, Wadleigh M, Cools J, Ebert BL, Wernig G, Huntly 33. Veselovska J, Pospisilova D, Pekova S, Horvathova M, Solna R,
BJ, et al. Activating mutation in the tyrosine kinase JAK2 in Cmejlova J, et al. Most pediatric patients with essential throm-
polycythemia vera, essential thrombocythemia, and myeloid bocythemia show hypersensitivity to erythropoietin in vitro,
metaplasia with myelofibrosis. Cancer Cell 2005;7:387-97. with rare JAK2V617F-positive erythroid colonies. Leuk Res
7. James C, Ugo V, Le Couedic JP, Staerk J, Delhommeau F, Lacout 2008;32:369-77.
C, et al. A unique clonal JAK2 mutation leading to constitutive 34. Nakatani T, Imamura T, Ishida H, Wakaizumi K, Yamamoto T,
signaling causes polycythemia vera. Nature 2005; 434:1144-8. Otabe O, et al. Frequency and clinical features of the
8. Zhao R, Xing S, Li Z, Fu X, Li Q, Krantz SB, et al. Identification Jak2V617F mutation in pediatric patients with sporadic essen-
of an acquired JAK2 mutation in polycythemia vera. J Biol tial thrombocythemia. Pediatr Blood Cancer 2008;51:802-5.
Chem 2005;280:22788-92. 35. Dror Y, Blanchette VS. Essential thrombocythemia in children.
9. Scott LM, Tong W, Levine RL, Scott MA, Beer PA, Stratton MR, Br J Haematol 1999;107:691-8.
et al. JAK2 exon 12 mutations in polycythemia vera and idio- 36. Fickers M, Speck B. Thrombocythemia. Acta Haematologica
pathic erythrocytosis. N Engl J Med 2007;356:459-68. 1974;51:257-65.
10. Pardanani AD, Levine RL, Lasho T, Pikman Y, Mesa RA, 37. Amato D, Freedman MH. Eleven-year follow-up of primary
Wadleight M et al. MPL515 mutations in myeloproliferative thrombocythemia in a child. J Pediatr 1984;104:639.
and other myeloid disorders: a study of 1182 patients. Blood 38. Kikawa Y, Fukumoto Y, Obata K, Tamura S, Takeuchi M,
2006;108:2472. Tanizawa A, et al. Successful treatment of essential thrombo-
11. Berlin NI. Diagnosis and classification of polycythemia. Semin cythemia evolving into agnogenic myeloid metaplasia with
Hematol 1975;12:339-51. interferon-α. J Pediatr Hematol Oncol 1998;20:463-6.
12. Murphy S, Peterson P, Iland H, Laszlo J. Experience of the poly- 39. Mitus AJ, Barbui T, Shulman LN, Rosenthal DS, Viero P,
cythemia vera study group with essential thrombocythemia: a Cortellazzo S, et al. Haemostatic complications in young
final report on diagnostic criteria, survival and leukemic transi- patients with essential thrombocythemia. Am J Med 1990; 88:
tion by treatment. Semin Hematol 1997;34:29-39. 371-5.
13. Vardiman JW, Harris NL, Brunning RD. The World Health 40. Fabris, F, Randi ML, Casonato A, Dal Bo Zanon R Bonvicini P,
Organization (WHO) classification of the myeloid neoplasms. Girolami A. Clinical significance of beta-thromboglobulin in
Blood 2002;100:2292-302. patients with high platelet count. Acta Haematologica 1984;7:
14. Verstovsek S, Silver RT, Cross NC, Tefferi A. Jak2V617F muta- 343-6.
tional frequency in polycythemia vera: 100%, >90%, less? 41. Chan GC, Ma SK, Ha SY, Chan LC, Lau YL. Childhood essen-
Leukemia 2006;20:2067. tial thrombocythemia without evidence of myeloproliferation:
15. Randi ML, Rossi C, Fabris F, Girolami A. Essential thrombo- how many investigations should be done? Br J Haematol 2000;
cythemia in young adults: major thrombotic complications and 110:1002.
complications during pregnancy. A follow-up study in 68 42. Randi ML, Putti MC, Pacquola E, Luzzatto G, Zanesco L, Fabris
patients. Clin Appl Thromb Hemost 2000;6:31-5. F. Normal thrombopoietin and its receptor (c-mpl) genes in chil-
16. Alvarez-Larran A, Cervantes F, Bellosillo B, Giralt M, Julia A, dren with essential thrombocythemia. Pediatr Blood Cancer
Hernandez-Boluda JC, et al. Essential thrombocythemia in 2004;43:1-4.
young individuals: frequency and risk factors for vascular 43. Putti MC, Giona F, Consarino C, Fiori GM, Giordano P, Lado-
events and evolution to myelofibrosis in 126 patients. gana S, et al. Retrospective evaluation of 90 children with
Leukemia 2007;21:1218-23. essential thrombocythemia: the AIEOP experience. Blood
17. Tefferi A, Fonseca R, Pereira DL, Hoagland HC. A long term 2008;112:664.
retrospective study of young women with essential thrombo- 44. Passamonti F, Rumi E, Arcaini L, Boveri E, Elena C, Pietra D, et
cythemia. Mayo Clin Proc 2001;76:22-8. al. Prognostic factors for thrombosis, myelofibrosis and leu-
18. Jensen MK, De Nully Brown P, Nielsen OJ, Hasselbach HC. kemia in essential thrombocythemia:a study of 605 Patients.
Incidence, clinical features and outcome of essential thrombo- Haematologia 2008;93:1645-51.
cythemia in a well defined geographical area. Eur J Hematol 45. Carobbio A, Finazzi G, Guerrini V, Spinelli O, Delaini E,
2000;65:132-9. Marchioli R, et al. Leukocytosis and risk stratification for
19. Johansson P. Epidemiology of myeloproliferative disorders thrombosis in essential thrombocythemia: interaction with
polycythemia vera and essential thrombocythemia. Semin treatment, standard risk stratification and JAK2 mutation
Thromb Hemost 2006;32:171-3. assessment in essential thrombocythemia. J Clin Oncol 2008;
20. Sutor AH. Thrombocytosis in childhood. Semin Thromb 26:2732-6.
Hemost 1995;2:330-9. 46. Randi ML, Putti MC, Scapin M, Pacquola E, Tucci F, Micalizzi
21. Dame C, Sutor AH. Primary and secondary thrombocytosis in C, et al. Pediatric patients with essential thrombocythemia are
childhood. Br J Haematol 2005;129:165-77. mostly polyclonal and V617FJAK2 negative. Blood 2006; 108:
22. Teofili L, Giona F, Marini M, Cenci T, Guidi F, Torti L, et al. 3600-2.
Markers of myeloproliferative diseases in childhood with poly- 47. Teofili L, Giona F, Martini M, Cenci T, Guidi F, Torti L, et al.
cythemia vera and essential thrombocythemia. J Clin Oncol The revised WHO criteria for Ph-negative myeloproliferative
2007;26:1048-53. diseases are not appropriate for the diagnostic screening of
23. Hasle H. Incidence of essential thrombocythemia in children. childhood polycythemia vera and essential thrombocythemia.
Br J Haematol 2000;110:751 Blood 2007;110:3384-86.
24. Ozer FL, Traux WE, Miesch DC, Levin WC. Primary hemor- 48. Campbell PJ, Scott LM, Buck G, Wheatley K, East CL, Marsden
rhagic thrombocythemia. Am J Med. 1960;28:807-23. JT, et al. Definition of subtypes of essential thromboythemia
25. Michiels JJ, Van Genderen PJJ. Essential thrombocythemia in and relation to polycythemia vera based on JAK2 V617F muta-
childhood. Semin Thromb Hemost 1997;23:295-301. tion status: a prospective study. Lancet 2005;366:1945-53.
26. Dror Y, Zipursky A, Blanchette VS. Essential thrombocythemia 49. Vannucchi AM, Antonoli E, Guglielmelli P, Pancrazi A, Guerini
in children. J Pediatr Hematol Oncol 1999;21:356-63. V, Barosi G, et al. Characteristics and clinical correlates of MPL

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 225 |
14th Congress of the European Hematology Association

515W>L/K mutation in essential thrombocythemia. Blood Sharathkumar A, et al: A basic classification and a comprehen-
2008;112:844-7. sive examination of pediatric myeloproliferative syndromes. J
50. Barbui T, Barosi G, Grossi A, Gugliotta L, Liberato LN, Pediatr Hematol Oncol 2005;27:192-6.
Marchetti M, et al. Practice guidelines for the therapy of essen- 73. Park MJ, Shimada A, Asada H, Koike K, Tsuchida M, Hayashi
tial thrombocythemia. A statement from the Italian Society of Y. Jak2 mutation in a boy with polycythemia vera, but not in
Hematology, the Italian Society of Experimental Hematology other pediatric hematologic disorders. Leukemia 2006; 20:
and the Italian Group for Bone Marrow Transplantation. 1453-4.
Haematologica 2004;89:215-32. 74. Teofili L, Foà R, Giona F, Larocca LM. Childhood polycy-
51. Barbui T, Finazzi G. Therapy for polycythemia vera and essen- themia vera and essential thrombocythemia does their patho-
tial thrombocythemia is driven by the cardiovascular risk. genesis overlap with that of adults. Haematologica 2008;
Semin Thromb Hemost 2007;33:321-9. 93:169-72.
52. Finazzi G, Barbui T. Evidence and expertise in the management 75. Cario H, Schwartz K, Herter JM, Komrska V, Mc Mullin MF,
of polycythemia vera and essential thrombocythemia. Minkov M, et al. Clinical and molecular characterization of a
Leukemia 2008;22:1494-502. prospectively collected cohort of children and adolescents
53. Finazzi G, Ruggeri M, Rodeghiero F, Barbui T. Efficacy and with polycythemia vera. Br J Haematol 2008;142:622-6.
safety of long-term use of hydroxyurea in young patients with 76. Teofili L, Cenci T, Martini M, Capodimonti S, Torti L, Giona F,
essential thrombocythemia and a high risk of thrombosis. et al. The mutant JAK2V617F allele burden in children with
Blood 2003;101:3749. essential thrombocythemia. Br J Haematol [Epub ahead of
54. Storen E, Tefferi A. Long term use of anagrelide in young print as doi: 10.1111/j.1365-2141.2009.07591]
patients with essential thrombocythemia. Blood 2001;97:863- 77. Landolfi R, Marchioli R, Kutti J, Gisslinger H, Tognoni G,
6. Patrono C, et al: Efficacy and safety of low dose aspirin in
55. Mazzucconi MG, Redi R, Bernasconi S, Bizzoni L, Dragoni F, polycythemia vera. N Engl J Med. 2004;350:114-24.
Latagliata L, et al. A long term study of young patients with 78. Mesa RA, Silverstein MN, Jacobsen SJ, Wollan PC, Tefferi A.
essential thrombocythemia treated with anagrelide. Haema- Population based incidence and survival figures in essential
tologica 2004;89:1306-13. thrombocythemia and agnogenic myeloid metaplasia: an
56. Szegedi I, Benko I, Mero G, Prinzinger A, Kappelmayer J, Kiss Olmsted County study, 1975–1995. Am J Hematol 1999; 61:
C. Long-lasting partial remission by Interferon-α treatment in 10-5.
a child with essential thrombocythemia. Pediatr Blood Cancer 79. Rozman C, Giralt M, Feliu E, Rubio D, Cortes MT. Life
2007;49:744-7. expectancy of patients with chronic non-leukemic myelopro-
57. Girodon F, Schaeffer C, Cleyrat C, Mounier M, Lafont I, Dos liferative disorders. Cancer 1991;67:2658-63.
Santos F, et al. Frequent reduction or absence of detection of 80. Barosi G, Hoffman R. Idiopathic myelofibrosis. Semin
the JAK2-mutated clone in JAK2V617F-positive patients with- Hematol 2003;42:248-58.
in the first years of hydroxyurea therapy. Haematologica 81. Abdel-Wahab OI, Levine RL. Primary myelofibrosis: update
2008;93:1723-27. on definition, pathogenesis, and treatment. Ann Rev Med
58. Randi ML, Fabris F, Girolami A. Leukemia and myelodysplasia 2009;60:233-45.
in patients with essential thrombocythemia treated with cyto- 82. Thiele J, Kvasnicka HM. Myelofibrosis in chronic myeloprolif-
toxic agent. Haematologica 1999;84:1049-50. erative disorders: dynamics and clinical impact. Histol
59. Randi ML, Putti MC. Essential thrombocythemia in children: Histopathol 2006;21:1367-78.
is a treatment needed? Expert Opin Pharmacother 2004;5: 83. Massa M, Rosti V, Ramajoli I, Campanelli R, Pecci A, Viarengo
1009-14. G, et al. Circulating CD34+,CD133+, and vascular endothelial
60. Pearson TC. Messinezy M, Westwood N, Green AR, Bench AJ, growth factor receptor 2-ositive endothelial progenitor cells in
Green AR, et al. A Polycythemia Vera Updated: Diagnosis, myelofibrosis with myeloid metaplasia. J Clin Oncol 2005;
Pathobiology, and Treatment. Hematology Am Soc Hematol 23:5688-95.
Educ Program. 2000:51-68.
84. Pikman Y, Lee BH, Mercher T, McDowell E, Ebert BL, Gozo
61. Messinezy M, Westwood NB, El-Hemaidi I, Marsden JT,
Sherwood RS, Pearson TC. Serum erythropoietin values in M, et al. MPLW515L is a novel somatic mutation in myelofi-
erythrocytoses and in primary thrombocythaemia. Br J brosis with myeloid metaplasia. PLoS Med 2006;3:e270.
Haematol 2002;117:47-53. 85. Schnittger S, Bacher U, Haferlach C, Beelen D, Bojko P,
62. Cario H. Childhood polycythemias/erythrocytoses: classifica- Dengler R, et al. Characterization of 35 new cases with four
tion, diagnosis, clinical presentation and treatment. Ann different MPLW515 mutations and essential thrombo-
Hematol 2005;84:137-45. cythemia or primary myelofibrosis. Haematologica 2009; 94:
63. Wajcman H, Galacteros F. Abnormal hemoglobins with high 141-4.
oxigen affinity and erythrocytosis. Hematol Cell Ther 1996; 86. Naithani R, Tyagi S, Choudhry VP: Secondary myelofibrosis
38:305-12. in children. J Pediatr Hematol Oncol 2008;30:196-8.
64. Hoyer JD, Allen SL, Beutler E, Kubik W, West C, Fairbanks VF. 87. Altura RA, Head DR, Wang WC. Long-term survival of infants
Erythrocytosis due to biphosphoglycerate-mutase deficiency with idiopathic myelofibrosis. Br J Haematol 2000;109:459-62.
with concurrent glucose-6- phosphate dehydrogenase (G- 88. Okada T, Sasaki F, Minagawa N, Naito S, Kobayashi R, Kubota
6PD) deficiency. Am J Hematol 2004;75:205-8. M, et al. Postoperative portal and splenic vein thrombosis in
65. Prchal JT. Pathogenetic mechanisms of polycythemia vera and children: identification of risk factors. Pediatr Surg Int 2005;
congenital polycythemic disorders. Semin Hematol 2001; 36: 21:918-21.
10-20. 89. Pulini S, D’Amico E, Basilico R, Mereu M, Bacci F, Spadano A,
66. Pastore YD, Jedlickova K, Guan Y, Liu E, Fahner J, Hasle H, et et al. Portal venous thrombosis in a young patient with idio-
al. Mutations of von-Hipple-Lindau tumor-suppressor gene pathic, myelofibrosis and intrahepatic extramedullary
and congenital polycythemia. Am J Human Genet 2003; 73: hematopoiesis: a difficult diagnosis, prognosis and manage-
412-9. ment. Leukemia 2007;21:2373-5.
67. Randi ML, Murgia A, Putti MC, Martella M, Casarin A, 90. Sekhar M, Prentice HG, Popat U, Anderson D, Janmohammed
Opocher G, et al. Low frequency of VHL gene mutations in R, Roberts I, et al. Idiopathic myelofibrosis in children. Br J
young individuals with polycythemia and high serum erythro- Haematol 1996;93:394-7.
poietin. Haematologica 2005;90:689-91. 91. Walia M, Mehta R, Paul P, Saluja S, Kapoor S, Sharma M.
68. Percy MJ, Furlow PW, Beer PA, Lappin TR, McMullin MF, Lee Idiopathic myelofibrosis with generalized periostitis in a 4-
FS. A novel erythrocytosis-associated PHD2 mutation sug- year-old girl. Clin Lab Osberv 2005;27:278-82.
gests the location of a HIF binding groove. Blood 2007; 110: 92. Zanazzo GA, Bua J. Mielofibrosi con metaplasia mieloide
2193-6. (MMM) ad esordio precoce. AIEOP meeting. Ancona. 2008,
69. Percy MJ, Furlow PW, Lucas GS, Li X, Lappin TR, McMullin 93. Hoffman R, Rondelli D. Biology and treatment of primary
MF, et al. A gain-of-function mutation in the HIF2A gene in myelofibrosis. Hematology Am Soc Hematol Educ Program
familial erythrocytosis. N Engl J Med 2008;358:162-8. 2007;346-54.
70. Arcasoy MO, Karayal AF, Segal HM, Sinning JG, Forget BG. A 94. Cervantes F, Mesa R, Barosi G. New and old treatment modal-
novel mutation in the erythropoietin receptor gene is associat- ities in primary myelofibrosis. Cancer J 2007;13:377-83.
ed with familial erytrhocytosis. Blood 2002;99:3066-9. 95. Levine RL, Gilliland DG. Myeloproliferative disorders. Blood
71. Percy MJ, Scott LM, Erber WN, Harrison CN, Reilly JT, Jones 2008;112:2190-8.
FG, et al. The frequency of JAK2 exon 12 mutations in idio- 96. Teofili L, Foà R, Giona F, Larocca LM. Childhood polycy-
pathic erythrocytosis patients with low serum erythropoietin themia vera and essential thrombocythemia: does their patho-
levels. Haematologica 2007;92:1585-9. genesis overlap with that of adult patients? Haematologica
72. Gassas A, Dole JJ, Weitzman S, Freedman MH, Hitzler JK, 2008; 93:169-72.

| 226 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Red cell disease

Red cell membrane disorders

N. Mohandas A B S T R A C T

During the last three decades, molecular, biochemical and biophysical studies have enabled the
New York Blood Center, development of detailed insights into altered membrane function in inherited red cell membrane dis-
New York, USA
orders. This paper highlights our current understanding of disorders involving either altered membrane
assembly (hereditary spherocytosis, hereditary elliptocytosis and hereditary ovalocytosis) or altered
membrane transport function (hereditary stomatocytosis). Mutations in multiple genes account for
Hematology Education: hereditary spherocytosis and elliptocytosis, while a single genetic defect accounts for all cases of
the education program for the hereditary ovalocytosis. The structural basis for altered membrane function of spherocytosis and ellip-
annual congress of the European
tocytosis but not of ovalocytosis has been delineated. Loss of vertical linkage between lipid bilayer and
Hematology Association
membrane skeleton leads to membrane loss in spherocytosis, while weakening of lateral linkages
2009;3:227-232 between skeletal proteins leads to membrane fragmentation and surface area loss in elliptocytosis. The
severity of anemia in both these disorders is related to extent of membrane loss and splenectomy ame-
liorates the anemia. Ovalocytosis is associated with mild anemia. In terms of membrane transport
defects, two distinct phenotypes have been identified: dehydrated stomatocytosis with mild to mod-
erate anemia and overhydrated stomatocytosis with moderate to severe anemia. Molecular basis for
stomatocytoses has not been fully defined. Splenectomy is contraindicated in both forms of stomato-
cytosis.

n order to perform its function of oxygen

I
Structural organization of normal red cell membrane
delivery, the discoid human red cell The structural organization of the mem-
undergoes extensive passive deformation brane is responsible for endowing the cell
during repeated passage through the narrow with its ability to undergo repeated exten-
capillaries of the microvasculature during its sive reversible deformations while main-
120-day life span in the circulation. taining its structural integrity during its life
Decreased cellular deformability, not only span in circulation. The red cell membrane
compromises the ability of the red cell to is highly elastic, responds rapidly to applied
perform its function of oxygen delivery opti- stresses and is capable of undergoing large
mally, but also leads to its premature membrane extensions without fragmenta-
removal from circulation by the spleen. The tion. These unusual membrane material
importance of red cell deformability was properties are the result of a composite
first recognized by Anton van Leeuwenhoek structure, in which a plasma membrane
in 1675 when he stated that when he was envelope composed of amphiphilic lipid
greatly disordered, the globules of his blood molecules is anchored to a two dimensional
appeared hard and rigid, but grew softer and elastic network of skeletal proteins through
more pliable as his health returned: whence he tethering sites (transmembrane proteins)
infers that in a healthy body they should be soft embedded in the lipid bilayer (Figure 1).3,6,7
and flexible (Van Leeuwenhoek, 1675). This There is also evidence suggesting a direct
truly prescient observation made more than interaction of skeletal proteins with the
three centuries ago has since been extensive- anionic phospholipids.8,9
ly validated. The lipid bilayer is composed of equiva-
During the last four decades, comprehen- lent amounts of cholesterol and phospho-
sive biochemical, biophysical and molecular lipids. A significant feature of bilayer lipid
biological studies on red cells from normal organization is that various phospholipids
individuals and individuals with various red are asymmetrically distributed with phos-
membrane disorders have enabled the devel- phatidylcholine and sphingomyelin local-
opment of detailed molecular insights into ized predominantly in the outer monolayer,
structural basis for membrane function in while most of phosphatidylethanolamine
health and disease.1-7 This presentation high- and all of phosphatidylserine (PS) and phos-
lights our current understanding of the phoinositides are localized in the inner
molecular and structural basis for various red monolayer.10 This asymmetric distribution
cell membrane disorders, and discusses how of phospholipids is functionally relevant
these new insights have contributed to the since both PS and phosphoinositides inter-
understanding of their pathophysiology and act with spectrin and protein 4.1R, thereby
clinical manifestations. anchoring the skeletal network to the bilay-

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 227 |
14th Congress of the European Hematology Association

er. Recent studies documented that spectrin binding to Hereditary spherocytosis


PS enhances membrane mechanical stability11 while Hereditary spherocytosis (HS) is a common inherited
phosphoinositide binding to 4.1R regulates its interac- hemolytic anemia that occurs in all racial ethnic groups
tion with transmembrane proteins band 3 and gly- and is particularly common in individuals of northern
cophorin C, and thus, linkage of the lipid bilayer to the European ancestry, affecting approximately 1 person in
skeletal network.12 3000.32,33 HS is commonly associated with dominant
More than 50 transmembrane proteins of varying inheritance (75%), although nondominant and recessive
abundance, ranging from a few hundred to approxi- inheritance (25%) has been reported. According to com-
mately million copies per red cell, have been identi- mon hematological parameters, including hemoglobin
fied.13 Of direct relevance to structural integrity of the and reticulocyte counts, disease severity is classified as
membrane are membrane proteins, band 3, glycophrin mild, moderate, moderately severe and severe. About
C and RhAG that link the bilayer to the spectrin based 20% of HS patients have mild HS with compensated
membrane skeleton. Band 3 and RhAG link the bilayer hemolysis with minimal spherocytosis, near normal
to the membrane skeleton through the interaction of hemoglobin levels, slight reticulocytosis (<6%), and
their cytoplasmic domains with ankyrin,14,15 while gly- mild splemenomegaly. Many of these individuals
cophorin C links through its interaction with protein escape detection until adulthood when complications
4.1R.16-18 The linkages play a key role in regulating cohe- related to chronic hemolysis, such as gallstones occur.
sion between the bilayer and the membrane skeleton.19 Moderate HS is the largest group of HS patients, com-
Loss of linkages results in lipid loss and decreased prising about 60% of cases. In this group, the hemoglo-
membrane surface area leading to decreased cellular bin level is between 8-11 g/dL and reticulocytes in most
deformability, a key contributor to decreased cell sur- cases are greater than 8%. The incidence of palpable
vival.2,7 In contrast, an increased number of linkages splenomegaly varies from about 50% in young children
leads to increased membrane cohesion and increased to 75 to 95% in older children and adults. A small group
membrane rigidity.19-21 of patients (approximately 10%) have moderately
The major protein constituents of the membrane severe HS with low hemoglobin values (6 to 8 g/dL),
skeletal network are α- and β-spectrin, actin, protein reticulocytosis (>15%) and intermittent need for trans-
4.1R, adducin, dematin, tropomyosin and tropomod- fusions. Approximately 3 to 5% have severe HS with
ulin.7,22-23 α- and β-spectrin form an anti-parallel het- life-threatening anemia requiring regular transfusions.
They usually have recessive HS.
erodimer through strong lateral interaction of the C-ter-
A common feature of all forms of HS is loss of mem-
minus of α-spectrin with the N-terminus of β-spec-
brane surface area and resultant change in cell shape
trin.24 Spectrin tetramer, the major structural compo-
from discocytes to stomatocytes to spherocytes. As red
nent of the two-dimension skeletal network is formed
cells with decreased membrane surface area and
by the lateral interaction of the single helical repeat at
reduced cellular deformability are unable to traverse
N-terminus of α-spectrin of one spectrin dimer with a
effectively through the splenic cords into the sinuses,
two helical repeat at C-terminus of β-spectrin of the they are sequestered in the red pulp of the spleen and
second dimer.25,26 The spectrin dimer-dimer interaction removed from circulation. Importantly, the severity of
is dynamically regulated in intact red cell membranes the disease is related to extent of decreased membrane
and the loss of avidity of the interaction leads to surface area. Splenectomy significantly reduces the
decreased membrane mechanical stability.27,28 The other severity of anemia by increasing the circulatory life span
end of the 100nm long spectrin dimer forms a junction- of spherocytes.
al complex with actin and protein 4.1R.7,29-31 The mechanistic basis for membrane loss in HS is the
Based on detailed analysis of normal and variously result of the defective anchoring of the skeletal network
modified red cells, the following concepts regarding the to the membrane because of defects in several proteins
structural basis for membrane material properties have (Table 1). Reduced anchoring because of deficiencies of
evolved. The unfolding and refolding of distinct spec- transmembrane proteins that link the bilayer to mem-
trin repeats, as well as transient dissociation and re- brane skeleton (band 3 or RhAG), of anchoring proteins
association of spectrin dimers accounts for remarkable (ankyrin or protein 4.2), or of spectrin leads to decreased
elasticity of the normal red cell membrane. The vertical membrane cohesion with resultant loss of membrane
linkages between bilayer and membrane skeleton play surface area. Thus, HS is the result of defects in any of
a critical role in maintaining membrane cohesion, while the protein components involved in vertical linkages
the lateral linkages between spectrin dimers and between skeletal network and the membrane (Figure 1).
between spectrin-actin-protein 4.1R are the dominant Ankyrin deficiency is the most common cause of HS
regulators of membrane mechanical stability. in Northern European populations, accounting for
Maintenance of both membrane cohesion and mem- approximately 50-60% of cases but it is found in only 5-
brane mechanical stability is critical for the red cell to 10% of HS cases in Japan.32-34 Ankyrin mutations cause
maintain its redundant surface area that is critical for it both dominant and recessive HS, and patients with
to undergo extensive deformations. Mutations in vari- ankyrin defects have prominent spherocytosis with
ous membrane and skeletal proteins that result in either clinical severity ranging from mild to severe depending
decreased membrane cohesion or membrane mechani- on the extent of membrane loss. Since ankyrin links β-
cal stability lead to membrane surface area loss, spectrin to band 3, it is not surprising that ankyrin defi-
decreased red cell life span and resultant anemia in a ciency leads to a proportional and secondary decrease in
variety of inherited red cell membrane disorders. membrane associated spectrin. Isolated spectrin defi-
ciency due to mutations in either α- or β-spectrin

| 228 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

Table 1. Gene mutation in inherited human red cell mem- on peripheral blood smears.41,42 Rh deficiency accounts
brane disorders. for less than 1% of HS cases. The molecular basis for
approximately 10% of HS cases has yet to be estab-
Spherocytosis Elliptocytsis Ovalocytosis lished.
Ankyrin 50/60% α- spectrin 65% Band 3 100% Hereditary elliptocytosis
α- and β- spectrin 20% β- spectrin 30% Hereditary elliptocytosis (HE) is a relatively common,
Band 3 15-20% Protein 4.1 5% clinically heterogeneous disorder characterized by pres-
Protein 4.2 <5%
ence of elliptically-shaped red cells on peripheral blood
Rh complex <1%
No defect identified 10%
smears,43 with prevalence approaching 2% in West
Africa, a malaria endemic region. Inheritance of HE is
autosomal dominant. The clinical presentation of HE
ranges from asymptomatic carrier to severe, life-threat-
ening anemia, with a few reported case of hydrops
account for 20% of HS cases.32,33 Patients with β-spectrin fetalis. The overwhelming majority of HE is asympto-
defects typically have mild to moderately severe HS, matic but approximately 10% of patients have moder-
while patients with α-spectrin suffer from severe HS. ate to severe anemia. Typically, individuals heterozy-
Band 3 deficiency is found in approximately 15-20% of gous for an elliptocytic variant have asymptomatic ellip-
HS patients, presenting with a phenotype of a mild to tocytosis while individuals with homozygous or com-
moderate anemia, although a few cases of severe HS pound heterozygous for HE variants experience mild to
have also been reported.33,35-38 Band 3 mutations are severe anemia. Poikylocytes and fragmented red cells, in
dominantly inherited. Mushroom-shaped or “pincered” addition to elliptocytes is a feature of red cell morphol-
red cells, in addition to spherocytes may be seen on ogy in HE cases with severe anemia.
peripheral blood smear. Recessive HS due to homozy- A distinguishing feature of all forms of HE is a
gous mutations in protein 4.2 gene is common in Japan mechanically unstable membrane, which results in pro-
but is rare in other populations, accounting for less than gressive transformation of cell shape from discocytes to
5% of HS cases.33,39-40 Rh deficiency associated with elliptocytes during circulation, and in severe cases to
absent or markedly reduced RhAG expression is associ- membrane fragmentation and generation of cells with
ated with mild to moderate hemolytic anemia associat- reduced membrane surface area and abnormal morphol-
ed with the presence of stomatocytes and spherocytes ogy. Red cells with decreased membrane surface area, as

Figure 1. A schematic representation of red cell membrane. The membrane is a composite structure in which a plasma
membrane envelope composed of amphiphilic lipid molecules is anchored to a two dimensional elastic network of skele-
tal proteins through tethering sites (transmembrane proteins) embedded in the lipid bilayer. Of particular relevance to
the present review is the vertical interaction involving cytoplasmic domains of band 3 and RhAG, ankyrin, protein 4.2 and
β-spectrin and the lateral linkages in the membrane skeletal network involving spectrin self-association and spectrin-
actin junctional complex.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 229 |
14th Congress of the European Hematology Association

result of membrane fragmentation are sequestered and some protection against malaria.
removed from circulation by the spleen. As with HS, Marked increase in membrane rigidity is a distin-
the severity of the disease is related to the extent of the guishing feature of ovalocytic red cell membranes.20 In
decrease in membrane surface area. Since splenic terms of clinical manifestations, it is important to note
sequestration is the dominant mechanism responsible that, in spite of a marked increase in rigidity, most
for reduced life span of fragmented red cells with affected individuals experience minimal hemolysis. In
reduced membrane surface area, splenectomy reduces all cases of ovalocytosis studied to date, only one muta-
the severity of anemia by increasing the circulatory life tion has been identified – a genomic deletion of 27bp
span of fragmented red cells. encoding amino acids 400 to 408 located at the bound-
The mechanistic basis for decreased membrane ary of the cytoplasmic and first transmembrane domain
mechanical stability in HE is weakened lateral linkages of band 3.20,49,50 Thus, hereditary ovalocytosis is unique
in membrane skeleton due to either defective spectrin among red cell membrane disorders in that the identical
dimer-dimer interaction or a defective spectrin-actin- mutation in a single gene is responsible for the pheno-
protein 4.1R junctional complex (Figure 1). Reduced type (Table 1). Although a number of hypotheses have
avidity of lateral interactions due to defects in α-spec- been proposed regarding how mutation in band 3 could
trin, β-spectrin or protein 4.1R lead to decreased mem- lead to increased membrane rigidity, the precise mecha-
brane mechanical stability. Thus, HE is the result of nistic basis for increased membrane rigidity of ovalo-
defects in any of the protein components involved in cytes has yet to be established.
lateral linkages in the skeletal network (Table 1).
Mutations in α-spectrin are the most common cause of Red cell membrane transport defects:
HE, accounting for approximately 65% of the cases. hereditary stomatocytoses
Missense mutations in the amino-terminal region of α- In addition to playing a critical role in regulating the
spectrin that is involved in spectrin dimer-dimer interac- key membrane material properties of deformability and
tion are the most frequent, while mutations in other membrane integrity, membrane proteins play a critical
parts of α-spectrin, that are not directly involved in role in regulating cell volume homeostasis. As with the
spectrin self association have also been identified.43,44 requirement for maintenance of normal membrane sur-
Mutations is β-spectrin account for 30% of HE cases.43 face area for optimal cell function, there is a similar
Point mutations, as well as truncations in the carboxyl- requirement for maintenance of normal cell volume.
terminus of β-spectrin that impair spectrin self-associa- Loss of the ability of the red cell to regulate its volume
tion have been identified. Heterozygous mutations in compromises its ability to perform its function optimal-
this region of β-spectrin are associated with variable ly. An increase in net cation content, with an accompa-
clinical severity, while in the homozygous state they nying increase in cell water content leads to increased
are fatal or near-fatal. Importantly, in almost all cases of cell volume, thereby reducing redundant surface area
spectrin mutations associated with HE, the degree of and generation of undeformable stomatocytic and sphe-
spectrin self-association disruption correlates with clin- rocytic red cells that are sequestered by the spleen. A
ical severity – the larger the degree of disruption of decrease in net cation content, with an accompanying
spectrin self-association, more severe the clinical phe- loss of cell water leads to decreased cell volume, and
notype. Quantitative deficiency of protein 4.1R, as well hence, increased cytoplasmic viscosity that compromis-
as qualitative defects in protein 4.1R account for 5% of es the ability of the cell to undergo rapid deformation
HE cases43,45,46 In all these instances, the defects in pro- needed for optimal oxygen delivery. Dehydrated red
tein 4.1R lead to a weakened spectrin-actin junctional cells with increased cytoplasmic viscosity appear to tra-
complex, leading to decreased membrane mechanical verse the spleen albeit less effectively.
stability. Two distinct phenotypes of inherited red cell mem-
When originally described, hereditary pyropoikilo- brane disorders with membrane transport defects have
cytes (HPP) was thought to be a distinct entity due to been identified: dehydrated hereditary stomatocytosis
increased thermal sensitivity of the red cells and the (xerocytosis) (DHS) and overhydrated hereditary stom-
unusual morphological features that were similar to atocytosis (OHS).51 DHS may appear alone or in con-
those seen in blood smears in severe thermal burns.47 junction with other clinical manifestations, including
However, subsequent molecular studies have estab- pseudohyperkalemia and/or perinatal fluid effusions.
lished that HPP is a subset of HE due to either homozy- The inheritance pattern of DHS is autosomal dominant.
gous or compound heterozygous mutations in spectrin DHS alone is associated with well-compensated anemia
leading to severe disruption of spectrin self-associa- with borderline macrocytosis and a mild to moderately
tion.43 enlarged spleen. Blood smears show stomatocytosis but
usually less than 10%. The distinctive feature of DHS is
Hereditary ovalocytosis cell dehydration with a resultant increase in MCHC and
Hereditary ovalocytosis is very common in malaria decreased osmotic resistance. OHS is associated with
endemic areas in Melanesia, Malaysia, Philippines, uncompensated hemolytic anemia with frank macrocy-
Indonesia and Southern Thailand. In endemic areas, its tosis and reticulocytosis. In contrast to DHS, stomato-
prevalence ranges from 5-25%.48 Ovalocytosis charac- cytes are a major feature of red cell morphology on
terized by the presence of oval-shaped red cells on blood smears. The distinctive feature of OHS is
blood smears is dominantly inherited. To date, only het- increased cell hydration with resultant increase in MCV,
erozygotes have been identified in high prevalent decreased MCHC and increased osmotic fragility. In
regions implying that homozygosity leads to embryon- contrast to HS, the increased osmotic fragility is not the
ic or fetal lethality. Hereditary ovalocytosis provides result of cell surface area but increased cell volume with

| 230 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

normal surface area. The inheritance pattern of OHS is action between band 3 and glycophorin A in human erythro-
cytes: immobilization of band 3 induced by antibodies to gly-
autosomal dominant. In contrast to our detailed under- cophorin A. Biophys J 1994;66:1726-32.
standing of the molecular basis for HS, HE and heredi- 22. Bennett V. The spectrin-actin junction of erythrocyte mem-
tary ovalocytosis, the molecular basis for HHS and OHS brane skeletons. Biochim Biophys Acta. 1989;988:107-21.
has yet to be fully defined.52-54 23. Bennett V, Baines AJ. Spectrin and ankyrin-based pathways:
metazoan inventions for integrating cells into tissues. Physiol
While splenectomy is highly beneficial in the manage- Rev 2001;81:1353-92.
ment of HS and HE patients with moderately severe to 24. Ursitti JA, Kotula L, DeSilva TM, Curtis PJ, Speicher DW.
severe anemia, it is contraindicated in both forms of Mapping the human erythrocyte beta-spectrin dimer initiation
site using recombinant peptides and correlation of its phasing
hereditary stomatocytosis since venous thromboembol- with the α-actinin dimer site. J Biol Chem 1996;271:6636-44.
ic and other complications occur following splenecto- 25. Speicher DW, DeSilva TM, Speicher KD, Ursitti JA, Hembach
my.55,56 The mechanistic basis for this complication has P, Weglarz L. Location of the human red cell spectrin tetramer
binding site and detection of a related "closed" hairpin loop
yet to be defined. dimer using proteolytic footprinting. J Biol Chem 1993; 268:
4227-35.
26. DeSilva TM, Peng KC, Speicher KD, Speicher DW. Analysis of
human red cell spectrin tetramer (head-to-head) assembly
References using complementary univalent peptides. Biochemistry 1992;
1. Mohandas N, Chasis JA, Shohet SB. The influence of mem- 31:10872-8.
brane skeleton on red cell deformability, membrane material 27. Liu SC, Palek J. Spectrin tetramer-dimer equilibrium and the
properties, and shape. Semin Hematol 1983;20:225-42. stability of erythrocyte membrane skeletons. Nature 1980;
2. Mohandas N, Chasis JA. Red blood cell deformability, mem- 285:586-8.
brane material properties and shape: regulation by transmem- 28. An X, Lecomte MC, Chasis JA, Mohandas N, Gratzer W.
brane, skeletal and cytosolic proteins and lipids. Semin Shear-response of the spectrin dimer-tetramer equilibrium in
Hematol 1993;30:171-92. the red blood cell membrane. J Biol Chem 2002;277:31796-
3. Mohandas N, Evans E. Mechanical properties of the red cell 800.
membrane in relation to molecular structure and genetic 29. Ungewickell E, Bennett PM, Calvert R, Ohanian V, Gratzer
defects. Annu Rev Biophys Biomol Struct 1994;23:787-818. WB. In vitro formation of a complex between cytoskeletal
4. Discher DE. New insights into erythrocyte membrane organi- proteins of the human erythrocyte. Nature 1979;280:811-4.
zation and microelasticity. Curr Opin Hematol 2000;7:117-22. 30. Karinch AM, Zimmer WE, Goodman SR. The identification
5. Delaunay J. The molecular basis of hereditary red cell mem- and sequence of the actin-binding domain of human red blood
brane disorders. Blood Rev 2007;21:1-20. cell β-spectrin. J Biol Chem 1990;265:11833-40.
6. An X, Mohandas N. Disorders of red cell membrane. Br J 31. An X, Debnath G, Guo X, Liu S, Lux SE, Baines A, et al.
Haematol 2008;141:367-75. Identification and functional characterization of protein 4.1R
7. Mohandas N, Gallagher PG. Red cell membrane: past, present, and actin-binding sites in erythrocyte β spectrin: regulation of
and future. Blood 2008;112:3939-48. the interactions by phosphatidylinositol-4,5-bisphosphate.
8. Rybicki AC, Heath R, Lubin B, Schwartz RS. Human erythro- Biochemistry 2005; 44:10681-88.
cyte protein 4.1 is a phosphatidylserine binding protein. J Clin 32. Eber S, Lux SE. Hereditary spherocytosis--defects in proteins
Invest 1988;81:255-60. that connect the membrane skeleton to the lipid bilayer.
9. An X, Guo X, Sum H, Morrow J, Gratzer W, Mohandas N. Semin Hematol 2004;41:118-41.
Phosphatidylserine binding sites in erythroid spectrin: location 33. Perrotta S, Gallagher PG, Mohandas N. Hereditary spherocy-
and implications for membrane stability. Biochemistry 2004; tosis. Lancet. 2008;372:1411-26.
43:310-5. 34. Eber SW, Gonzalez JM, Lux ML, Scarpa AL, Tse WT, Dornwell
10. Zwaal RF, Schroit AJ. Pathophysiologic implications of mem- M, et al. Ankyrin-1 mutations are a major cause of dominant
brane phospholipid asymmetry in blood cells. Blood 1997; 89: and recessive hereditary spherocytosis. Nat Genet 1996;13:
1121-32. 214-8.
11. Manno S, Takakuwa Y, Mohandas N. Identification of a func- 35. Jarolim P, Murray JL, Rubin HL, Taylor WM, Prchal JT, Ballas
tional role for lipid asymmetry in biological membranes: SK, et al. Characterization of 13 novel band 3 gene defects in
Phosphatidylserine-skeletal protein interactions modulate hereditary spherocytosis with band 3 deficiency. Blood 1996;
membrane stability. Proc Natl Acad Sci USA 2002;99:1943-8. 88:4366-74.
12. An X, Zhang X, Debnath G, Baines AJ, Mohandas N. 36. Dhermy D, Galand C, Bournier O, Boulanger L, Cynober T,
Phosphatidylinositol-4,5-biphosphate (PIP2) differentially reg- Schismanoff PO, et al. Heterogenous band 3 deficiency in
ulates the interaction of human erythrocyte protein 4.1 (4.1R) hereditary spherocytosis related to different band 3 gene
with membrane proteins. Biochemistry 2006;45:5725-32. defects. Br J Haematol 1997;98:32-40.
13. Reid ME, Mohandas N. Red blood cell blood group antigens: 37. Tanner MJ. Band 3 anion exchanger and its involvement in
structure and function. Semin Hematol 2004;41:93-117. erythrocyte and kidney disorders. Curr Opin Hematol 2002;
14. Bennett V. Proteins involved in membrane--cytoskeleton asso- 9:133-9.
ciation in human erythrocytes: spectrin, ankyrin, and band 3. 38. Perrotta S, Borriello A, Scaloni A, De Franceschi L, Brunati
Methods Enzymol 1983;96:313-24. AM, Turrini F, et al. The N-terminal 11 amino acids of human
15. Nicolas V, Le Van Kim C, Gane P, et al. Rh-RhAG/ankyrin-R, a erythrocyte band 3 are critical for aldolase binding and protein
new interaction site between the membrane bilayer and the phosphorylation: implications for band 3 function. Blood
red cell skeleton, is impaired by Rh(null)-associated mutation. 2005;106:4359-66.
J Biol Chem 2003;278:25526-33. 39. Bouhassira EE, Schwartz RS, Yawata Y, Ata K, Kanzaki A, Qiu
16. Reid ME, Takakuwa Y, Conboy J, Tchernia G, Mohandas N. JJ, et al. An alanine-to-threonine substitution in protein 4.2
Glycophorin C content of human erythrocyte membrane is cDNA is associated with a Japanese form of hereditary
regulated by protein 4.1. Blood 1990;75:2229-34. hemolytic anemia (protein 4.2NIPPON). Blood 1992;79:1846-
17. Marfatia SM, Lue RA, Branton D, Chishti AH. In vitro binding 54.
studies suggest a membrane-associated complex between ery- 40. Yawata Y, Kanzaki A, Yawata A, Doerfler W, Ozcan R, Eber
throid p55, protein 4.1, and glycophorin C. J Biol Chem 1994; SW. Characteristic features of the genotype and phenotype of
269:8631-4. hereditary spherocytosis in the Japanese population. Int J
18. Marfatia SM, Leu RA, Branton D, Chishti AH. Identification of Hematol 2000;71:118-35.
the protein 4.1 binding interface on glycophorin C and p55, a 41. Ballas SK, Clark MR, Mohandas N, Colfer HF, Caswell MS,
homologue of the Drosophila discs-large tumor suppressor Bergren MO, et al. Red cell membrane and cation deficiency in
protein. J Biol Chem 1995;270:715-9. Rh null syndrome. Blood 1984; 63:1046-55.
19. Butler J, Mohandas N, Waugh RE. Integral protein linkage and 42. Cartron JP. Rh nlood group system and molecular basis of Rh-
the bilayer-skeletal separation energy in red blood cells. deficiency. Baillieres Best Pract Res Clin Haematol 1999;
Biophys J 2008;95:1826-36. 12:655-89.
20. Mohandas N, Winardi R, Knowles D, Leung A, Parra M, 43. Gallagher PG. Hereditary elliptocytosis: spectrin and protein
George E, et al. Molecular basis for membrane rigidity of 4.1R. Semin Hematol 2004;41:142-64.
hereditary ovalocytosis. A novel mechanism involving the 44. Delaunay J, Dhermy D. Mutations involving the spectrin het-
cytoplasmic domain of band 3. J Clin Invest 1992;89:686-92. erodimer contact site: clinical expression and alterations in
21. Knowles DW, Chasis JA, Evans EA, Mohandas N. Cooperative specific function. Semin Hematol 1993;30:21-33.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 231 |
14th Congress of the European Hematology Association

45. Tchernia G, Mohandas N, Shohet S. Deficiency of skeletal in hereditary ovalocytosis. J Mol Biol 1992;223:949-58.
membrane protein band 4.1 in homozygous hereditary ellip- 51. Delaunay J. The hereditary stomatocytoses: genetic disorders
tocytosis. Implications for erythrocyte membrane stability. J of the red cell membrane permeability to monovalent cations.
Clin Invest 1981;68:454-60. Semin Hematol 2004;41:165-72.
46. Marchesi SL, Conboy J, Agre P, Letsinger JT, Marchesi VT, 52. Bruce LJ, Robinson HC, Guizouarn H, Borgese F, Harrison P,
Speicher DW, et al. Molecular analysis of insertion/deletion King MJ, et al. Monovalent cation leaks in human red cells
mutations in protein 4.1 in elliptocytosis. I. Biochemical iden- caused by single amino-acid substitutions in the transport
tification of rearrangements in the spectrin/actin binding domain of the band 3 chloride-bicarbonate exchanger, AE1.
domain and functional characterizations. J Clin Invest 1990; Nat Genet 2005;37:1258-63.
86:516-23. 53. Iolascon A, Perrotta S, Stewart GW. Red blood cell membrane
47. Zarkowsky HS, Mohandas N, Speaker CB, Shohet SB. A con- defects. Rev Clin Exp Hematol. 2003;7:22-56.
genital haemolytic anaemia with thermal sensitivity of the 54. Bruce LJ. Red cell membrane transport abnormalities. Curr
erythrocyte membrane. Br J Haematol 1975;29:537-43. Opin Hematol 2008;15:184-90.
48. Amato D, Booth PB. Hereditary ovalocytosis in Melanesians. 55. Stewart GW, Amess JA, Eber SW, Kingswood C, Lane PA,
P N G Med J 1977;20:26-32. Smith BD, et al. Thrombo-embolic disease after splenectomy
49. Jarolim P, Palek J, Amato D, Hassan K, Sapak P, Nurse GT, et for hereditary stomatocytosis. Br J Haematol 1996;93:303-10.
al. Deletion in erythrocyte band 3 gene in malaria-resistant 56. Jais X, Till SJ, Cynober T, Garcia G, Tchernia G, Dartevelle P,
Southeast Asian ovalocytosis. Proc Natl Acad Sci USA 1991; et al. An extreme consequence of splenectomy in dehydrated
88:11022-6. hereditary stomatocytosis: gradual thrombo-embolic pul-
50. Schofield AE, Tanner MJ, Pinder JC, Clough B, Bayley PM, monary hypertension and lung-heart transplantation.
Nash GB, et al. Basis of unique red cell membrane properties Hemoglobin 2003;27:139-47.

| 232 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Red cell disease

Predicting clinical severity in sickle cell anemia

M.H. Steinberg A B S T R A C T

Sickle cell anemia is noted for its clinical heterogeneity. Some patients almost continually seem to
Department of Medicine, Boston suffer from disease-related complications. Others have far fewer complications. Some individuals die
University School of Medicine and
the Center of Excellence in Sickle young; others survive until the 8th or 9th decade. The ability to predict the phenotype of an individ-
Cell Disease, Boston Medical Center, ual with sickle cell anemia would allow a reliable prognosis and could guide therapeutic decision-
Boston, MA., USA making. Some risk factors for individual disease complications are known but are insufficiently pre-
cise to use for individual prognostic purposes. Progress has been made toward estimating the overall
disease severity, at least in adults. Genetic association studies, which attempt to link gene polymor-
Hematology Education:
phisms with selected disease subphenotypes, may eventually provide useful means of predicting the
the education program for the
annual congress of the European likelihood of certain complications and allow more individualized treatment.
Hematology Association

2009;3:233-237

ickle cell anemia, a monogenic disorder interest was HbF, SNPs in this gene were

S caused by homozygosity for a single β-


globin gene (HBB) mutation (glu6val), is
phenotypically heterogeneous. Persuasive
associated with HbF concentration in nor-
mal individuals, patients with β thalassemia
trait, β thalassemia intermedia, and sickle
evidence suggests that to a first approxima- cell anemia.1-2,5 Patients with sickle cell ane-
tion, the pathophysiology of disease has mia, who were homozygous for the C allele
components related to intravascular hemoly- of SNP rs 766432 had an average of 7% HbF
sis and proliferative vasculopathy, and to compared with 3% HbF in patients
sickle cell-related vasoocclusion. This pro- homozygous for the A allele.1 The associa-
vides many loci where the disease pheno- tion of BCL11A with HbF in at least four dif-
type can be influenced by modifying genes ferent populations, are consistent with an
(Figure 1). Accurately forecasting the severi- ancient variant that is highly conserved
ty of sickle cell disease might require know- among ancestral populations. SNPs at the
ing which genes are associated with its BCL11A and HBS1L-MYB loci, along with a
hemolytic and vascular complications, and SNP 5’ to HBG2 creating an Xmn I restric-
how variants of these genes interact among tion site (rs7482144), accounted for more
themselves and with the environment to than 20% of the variation in HbF and were
determine the course of disease. associated with painful events in two sickle
cell anemia populations.3
Fetal hemoglobin (HbF) Hydroxyurea is the sole agent available for
HbF (α2γ2) is the most thoroughly studied specifically treating the complications of
and powerful genetic modulator of sickle sickle cell anemia. Predicting an individual’s
cell anemia. Its exclusion from the HbS poly- HbF response to hydroxyurea treatment
mer inhibits HbS polymerization, the initia- would aid in the selection of patients for
tor of most pathophysiology. Among treatment and reduce toxicity from unfruit-
patients with sickle cell anemia, HbF con- ful dose escalation.
centrations vary from 0.1-30%. At least two Twenty-nine candidate genes were stud-
well-characterized quantitative trait loci ied in sickle cell anemia patients treated with
(QTL) loci on chromosomes 6q and 2p are hydroxyurea. SNPs in genes within the
associated with HbF concentration, and it is 6q22.3-23.2 and 8q11-q12 linkage peaks,
likely that others exist.1-3 In K562 cells, over- and the ARG2, FLT1, HAO2 and NOS1
expression of MYB inhibites γ-globin gene genes were associated with the HbF
expression. In northern European families, response to this agent.6
polymorphisms within and 5’ to HBS1L are In GWAS of hydroxyurea-treated patients,
strongly associated with F cell levels, chromosome 20 had more significant SNPs
accounting for 17.6% of the F cell variance.4 than expected at random, especially in
BCL11A (2p16.1) codes for a zinc finger tran- CST9, one of a family of protease
scription factor. In genome-wide association inhibitors.7 CST9 is tagged by three SNPs
studies (GWAS), where the phenotype of (rs2983639, rs2983640, rs10485646); two of

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 233 |
14th Congress of the European Hematology Association

Figure 1. Pathophysiology of sickle


cell disease. The mutation, HBB
glu6val causes the production of HbS
that polymerizes when deoxygenated.
HbS polymer damages the membrane
cytoskeleton, causing reduced cation
and water content, and altered distri-
bution of membrane lipids. Sickle cells
interact with endothelium and other
blood cells, causing vasoocclusion and
a variable number lyse intravascularly
releasing hemoglobin that scavenges
plasma nitric oxide (NO). NO, by bind-
ing soluble guanylate cyclase, converts
cGTP to GMP, relaxes vascular smooth
muscle and causes vasodilatation.
Reduced endothelial NO bioavailabilty
also impairs inhibition of platelet acti-
vation and aggregation and transcrip-
tional repression of cell adhesion mol-
ecule genes. Oxygen radicals and pro-
tein nitration potentially further limit
NO bioavailability and activates
endothelium. Lysed red cells also liber-
ate arginase that destroys L-arginine,
the substrate for NO production.

which were associated with significant positive changes gene studies, polymorphisms in genes of the TGF-
in HbF after treatment and one with significant negative β/BMP pathway appear involved in several subpheno-
changes of HbF. Although individually these SNPs did types of disease, and could reflect the hitherto unappre-
not reach genome-wide significance, cumulatively they ciated role of this very large pathway in the pathobiol-
provide strong evidence of association, as the probabil- ogy of disease.10
ity that they are all simultaneously associated by chance
was 10-4. Furthermore, significant variants in other Stroke
genes that belong to the same family of type 2 cysteine About 10-15% of patients with sickle cell anemia
protease inhibitors, CTS3 and CTS5, also had SNPs have overt strokes and larger numbers have silent cere-
associated with HbF. Small sample size and the large brovascular disease. A family predisposition to stroke in
number of SNPs tested in GWAS suggests caution until sickle cell disease suggested that inherited modulation
these results are replicated. of this phenotype was possible.11 Among the genes
A small guanosine triphosphate (GTP)-binding pro- associated with stroke in sickle cell anemia are vascular
tein, secretion-associated and RAS-related (SAR1A) pro- adhesion molecule-1 (VCAM1),12-14 interleukin 4 recep-
tein is inducible by hydroxyurea, and might play a piv- tor gene (IL4R), tumor necrosis factor α gene (TNFA) and
otal role in induction of γ-globin gene expression via its α adrenergic receptor 2 (ADRB2), and low density
role in erythroid maturation. Polymorphisms in the lipoprotein receptor (LDLR).13,15-16
SAR1A promoter were associated with differences in To examine the interactions among genes and their
HbF levels or the HbF response to hydroxyurea in SNPs, and to develop a prognostic model for stroke in
patients with sickle cell anemia.8 sickle cell anemia, a Bayesian network was developed
to analyze SNPs in candidate genes in 1398 unrelated
α thalassemia subjects with sickle cell anemia.17 SNPs in 11 genes and
In patients with sickle cell anemia and α thalassemia, four clinical variables, including α thalassemia and HbF,
the concentration of HbS is reduced, lessening the poly- interacted in a complex network of dependency to
merization potential of HbS and decreasing hemolysis. modulate the risk of a stroke. This network of interac-
Vasoocclusive events that are highly dependent on the tions included three genes, BMP6, TGFBR2, TGFBR3,
intensity of hemolysis, such as stroke, leg ulcer and pri- with a functional role in the TGF-β/BMP pathway and
apism benefit from the coexistence of α thalassemia. P-selectin (SELP). The model predicted the occurrence
Painful episodes, acute chest syndrome and osteonecro- of a stroke in unrelated individuals with 98.2% accura-
sis are either minimally affected, or their prevalence is cy. The predictive accuracy of this stroke model is a step
increased by coincident α thalassemia.9 toward the development of prognostic tests better able
to identify patients at risk for stroke.
Predictors of disease severity Gene expression studies have also contributed to
The diversity of sickle cell anemia cannot be understanding the predisposition to stroke.18 When sub-
explained by HbF and α-globin gene-linked modulation jects at risk for stroke, estimated by the presence of
alone. Other modifying genes must affect the pathobi- Circle of Willis disease or having had a stroke, were
ology of sickle cell anemia and while candidate genes compared with controls, transcripts in genes of inflam-
association studies have been informative, GWAS stud- mation-related pathways expressed in blood-outgrowth
ies, with their promise of capturing more genetic vari- endothelial cells were most strongly associated with
ability, are starting to be reported. Based on candidate stroke or predisposition to stroke.

| 234 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

Priapism sporadic pulmonary hypertension. In 111 sickle cell dis-


Priapism occurs in about 40% of males with sickle cell ease patients screened for pulmonary hypertension,
anemia. A strong association was found between the genes in the TGF-β/BMP superfamily, including
prevalence of priapism, the severity of hemolysis and ACVRL1, BMPR2, and BMP6 were associated with
the presence of α thalassemia.19 Polymorphisms in TRJV.29 One of these SNP in BMP6, (rs 449853), was also
Klotho (KL) showed an association with priapism.20 KL associated with stroke and bacteremia in other stud-
has a role in NO biochemistry and directly or indirectly ies.17,24
promotes endothelial NO production. In another study, Acute chest syndrome is the second most frequent
priapism was associated with SNPs in TGFBR3, AQP1 disease complication, and occurs in about 60% of
and the adhesion molecule, ITGAV..21 cases.30 Both an increased and decrease risk of having
acute chest syndrome was associated with a T786C
Osteonecrosis SNP in the endothelial NO synthase gene (NOS3); how-
Osteonecrosis is found in nearly half of all adults with ever, this observation has not been replicated.31-33
sickle cell anemia. Its prevalence might be increased by Exhaled NO levels were reduced in patients with acute
concurrent α thalassemia. When subjects with chest syndrome compared with controls, and this was
osteonecrosis were compared with controls, significant associated with the number of AAT repeats in intron 20
associations were observed with SNPS in BMP6, of NOS1.34 In follow-up, the ATT repeat polymorphism
ANXA2, TGFBR2, TGFBR3, EDN1, ERG, KL and ECE1.22 in NOS1 was associated with acute chest syndrome
Some of these genes might play a role in bone metabo- only in patients without asthma (itself, a risk factor for
lism. KL is also a glycosyl hydrolase in a negative regu- acute chest syndrome).33 A T8002C SNP in the endothe-
latory network of the vitamin D endocrine system. lin-1 gene (EDN1) was associated with an increased risk
Bone morphogenetic proteins, including BMP6, are of acute chest syndrome in 173 children with sickle cell
pleiotropic secreted proteins structurally related to TGF- anemia detected at birth and followed longitudinally.32
β and activins and are participate in bone formation and These studies examined small numbers of cases and
development. ANXA2, a member of the calcium- controls, were limited to children, and few SNPs were
dependent phospholipid-binding protein family, is examined.
involved in osteoblast mineralization. In 1422 subjects with sickle cell anemia, the popula-
tion was dichotomized into children aged less than or
Leg ulcers equal to 5 years and older children and adults.35 There
Sickle cell leg ulcers develop in about 10% of patients were 170 acute chest syndrome cases and 884 controls
and are related to hemolysis. In candidate gene associ- in patients aged less than 5 years and 388 cases and 819
ation studies, associations were found with SNPs in KL, controls in older individuals. Using time-to-first event in
TEK and several genes in the TGF-β/BMP signaling an age, gender, leukocyte, reticulocyte and platelet
pathway.23 count-adjusted analysis, and controlling for the false dis-
covery rate, one SNP in TGFBR3 and one in LD with
Bacteremia SMAD7 were significantly associated with acute chest
Infection, bacteremia and sepsis are common events syndrome in both patient groups.
in sickle cell anemia. SNPs in candidate genes have been
associated with an increased risk of sepsis in other dis- Hyperbilirubinemia and gallstones
eases. In a case control study, bacteremia in sickle cell Promoter polymorphisms in the uridine diphosphate-
anemia was associated with SNPs and haplotypes of glucuronosyltransferase 1A (UGT1A) gene were associ-
genes of the TGF-β/BMP pathway, such as BMP6, ated with unconjugated hyperbilirubinemia and Gilbert
TGFBR3, BMPR1A, SMAD6 and SMAD3.24 syndrome. Children with sickle cell disease had a signif-
icantly higher mean bilirubin level if they carried the 7/7
Renal disease UGT1A genotype compared with the wild type 6/6 or
With aging, sickle nephropathy becomes common. the 6/7 genotypes; patients with the 7/7 genotype were
Patients with sickle cell anemia associated with a Bantu more likely to have had a cholecystectomy.36-37
haplotype were more likely to develop renal failure,
perhaps because they had the lowest levels of HbF.25,26 Erythrocyte glucose-6-phosphate dehydrogenase (G-6-PD)
SNPs in selected candidate genes were also associated deficiency
with glomerular filtration rate (GFR). Tagging SNPs in G-6-PD deficiency was not associated with differen-
about 70 genes of the TGF-β/BMP pathway were stud- tial survival, increased hemolysis, or a higher incidence
ied and four SNPs in BMPR1B, a BMP receptor gene, of acute anemic episodes.38 Using DNA-based methods
showed significant associations with GFR. The TGF- to detect the GdA- allele of G6PD unequivocally, blood
β/BMP pathway has been associated with the develop- counts were similar in patients with and without G-6-
ment of diabetic nephropathy, which has some features PD deficiency, although the hemoglobin concentration
in common with sickle cell nephropathy.27 was lower in sickle cell anemia with the GdA- gene.39
While little, if any, modulation of the phenotype of
Pulmonary disease sickle cell anemia by coincident G-6-PD deficiency is
Pulmonary hypertension, estimated by tricuspid apparent, perhaps the right phenotype has not been
regurgitant jet velocity (TRJV) is a major risk factor for studied. Patients with sickle cell anemia have impaired
premature death in sickle cell anemia.28 Mutations in flow-dependant and independent vasodilation.40 This
bone morphogenetic protein receptor 2 (BMPR2) and a might be a consequence of intravascular hemolysis and
few other genes were associated with both familial and oxidant stress. Adequate availability of G-6-PD is need-

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 235 |
14th Congress of the European Hematology Association

ed to maintain both NO levels and preserve the proper some SNPs discovered with this strategy tag genes asso-
redox milieu. A G-6-PD deficient phenotype could be ciated with susceptibility to diabetes (CDKN2A), and
present in critical vascular tissues in G-6-PD deficient regulation of insulin-like growth factor (IGF2BP1).
individuals and perhaps even in sickle cell disease
patients with a normal G-6-PD genotype.41 Perhaps, the Conclusions
hyperaldosteronism of sickle cell anemia might impair Personalized medicine is a goal of the future. By cap-
vascular reactivity by decreasing endothelial G-6-PD turing the genetic heterogeneity associated with the
activity.42 complications of sickle cell anemia, it might be possible
very early in life to build predictive models that esti-
Painful episodes mate the likelihood of the clinical events most responsi-
Besides HbF concentration and a possible role of α ble for disease morbidity and mortality. These models
thalassemia, a genetic basis for the heterogeneous distri- could be revised for different populations and environ-
bution of painful episodes among patients has not yet ments, and take into account clinical events and labora-
been described. Patient response to opioid analgesics tory measures. GWAS and deep sequencing of candi-
varies considerably, and the efficacy of these drugs is date genes to capture uncommon genetic variants
known to depend on genetic variability of their catabol- require large populations and careful validation studies,
ic enzymes and receptors.43 not an easy task in a rare disease like sickle cell anemia.
Compound phenotypes and integrated measure of disease
severity References
Few genetic association studies have used compound
phenotypes. To produce an integrated disease pheno- 1. Sedgewick A, Timofeev N, Sebastiani P, et al. BCL11A (2p16)
is a major HbF quantitative trait locus in three different pop-
type, a Bayesian network model was developed that ulations. Blood Cells Mol Dis 2008;41:255-8.
describe the complex associations of 25 clinical and lab- 2. Uda M, Galanello R, Sanna S, Lettre G, Sankaran VG, Chen W,
oratory variables, deriving a score to define disease et al. Genome-wide association study shows BCL11A associ-
ated with persistent fetal hemoglobin and amelioration of the
severity as the risk of death within five years.44 The phenotype of β-thalassemia. Proc Natl Acad Sci USA 2008;
genetic basis of severity was examined by using this 105:1620-5.
score as a phenotype and studying the association of 3. Lettre G, Sankaran VG, Bezerra MA, Araújo AS, Uda M, Sanna
S, et al. DNA polymorphisms at the BCL11A, HBS1L-MYB,
795 SNPs in 320 candidate genes in mild and severe dis- and β-globin loci associate with fetal hemoglobin levels and
ease.45 Positive associations were validated in a smaller pain crises in sickle cell disease. Proc Natl Acad Sci USA 2008;
independent patient sample. Among the SNPs in genes 105:11869-74.
associated with disease severity, some associations, like 4. Thein SL, Menzel S, Peng X, Best S, Jiang J, Close J, et al.
Intergenic variants of HBS1L-MYB are responsible for a major
with TGFBR3, confirmed previous findings and sup- quantitative trait locus on chromosome 6q23 influencing fetal
ported the conjecture that dysregulation of the TGF- hemoglobin levels in adults. Proc Natl Acad Sci USA 2007;
β/BMP signaling pathway might modulate disease 104:11346-51.
5. Menzel S, Garner C, Gut I, Matsuda F, Yamaguchi M, Heath S,
severity. Some associated genes had a less obvious role et al. A QTL influencing F cell production maps to a gene
in the pathobiology of disease, but were strongly asso- encoding a zinc-finger protein on chromosome 2p15. Nat
ciated with the phenotype of severity. SNPs in these Genet 2007;39:1197-9.
6. Ma Q, Wyszynski DF, Farrell JJ, Kutlar A, Farrer LA, Baldwin
genes were also associated with normal aging. Perhaps CT, et al. Fetal hemoglobin in sickle cell anemia: genetic deter-
increased oxidative stress and sickle vasculopathy cause minants of response to hydroxyurea. Pharmacogenomics J
accelerated tissue damage that is modulated by genes 2007;7:386-94.
similar to those involved in the aging process. 7. Timofeev N, Sebastiani P, Hartley SH, Baldwin CT, Steinberg
MH. Fetal hemoglobin in sickle cell anemia: a Genome-wide
In GWAS of patients aged over 18 years, 49 SNPs were association study of the response to hydroxyurea. Blood 2008:
strongly associated with the severity score and 15 asso- 2471.
ciations were reproduced in an independent sample. To 8. Kumkhaek C, Taylor JG 6th, Zhu J, Hoppe C, Kato GJ, Rodgers
GP. Fetal haemoglobin response to hydroxycarbamide treat-
boost the power of the sample size, a complementary ment and SAR1A promoter polymorphisms in sickle cell
discovery approach was used to leverage the pleiotropic anaemia. Br J Haematol 2008;141:254-9.
effects of genes. As some complications of sickle cell 9. Taylor JG 6th, Nolan VG, Mendelsohn L, Kato GJ, Gladwin
MT, Steinberg MH. Chronic hyper-hemolysis in sickle cell
anemia resemble aging diseases, it was hypothesized anemia: association of vascular complications and mortality
that protective gene variants associated with mild dis- with infrequent vasoocclusive pain. PLoS One 2008;3:e2095.
ease might also be associated with delayed aging in the 10. Bertolino P, Deckers M, Lebrin F, Ten DP. Transforming
growth factor-β signal transduction in angiogenesis and vascu-
general population, and that deleterious gene variants lar disorders. Chest 2005;128:585S-590S.
associated with severe disease might be associated with 11. Driscoll MC, Hurlet A, Styles L, McKie V, Files B, Olivieri N,
early mortality. Significant associations identified in a et al. Stroke risk in siblings with sickle cell anemia. Blood
GWAS of exceptional longevity of more than 700 cente- 2003;101:2401-4.
12. Taylor JG 6th, Tang DC, Savage SA, Leitman SF, Heller SI,
narians and 1800 referent subjects were compared with Serjeant GR, et al. Variants in the VCAM1 gene and risk for
the results in sickle cell anemia. Several SNPs that were symptomatic stroke in sickle cell disease. Blood 2002; 100:
more frequent in long-lived individuals were also more 4303-9.
13. Hoppe C, Klitz W, Cheng S, Apple R, Steiner L, Robles L, et al.
frequent in patients with mild disease. Among these Gene interactions and stroke risk in children with sickle cell
associations were SNPs in the gene KCNQ1 that is asso- anemia. Blood 2004;103:2391-6.
ciated with long and short QT syndrome.46 Prolonged 14. Kutlar A, Brambilla D, Clair B, et al. Candidate gene polymor-
phisms and their association with TCD velocities in children
QT interval is more common in sickle cell anemia than with sickle cell disease. Blood 2007;110:133a[Abstract].
in controls and could be a risk factor for the unexpected 15. Styles LA, Hoppe C, Klitz W, Vichinsky E, Lubin B, Trachten-
sudden death that is common in adults. Interestingly, berg E. Evidence for HLA-related susceptibility for stroke in
children with sickle cell disease. Blood 2000;95:3562-7.

| 236 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

16. Hoppe C, Klitz W, Noble J, Vigil L, Vichinsky E, Styles L. acute chest syndrome and painful vaso-occlusive crises in chil-
Distinct HLA associations by stroke subtype in children with dren with sickle cell anemia. Haematologica 2006;91:1277-8.
sickle cell anemia. Blood 2002;101:2865-9. 33. Duckworth L, Hsu L, Feng H, Wang J, Sylvester JE, Kissoon N,
17. Sebastiani P, Ramoni MF, Nolan V, Baldwin CT, Steinberg MH. et al. Physician-diagnosed asthma and acute chest syndrome:
Genetic dissection and prognostic modeling of overt stroke in associations with NOS polymorphisms. Pediatr Pulmonol
sickle cell anemia. Nat Genet 2005;37:435-40. 2007;42:332-8.
18. Milbauer LC, Wei P, Enenstein J, et al. Genetic endothelial sys- 34. Sullivan KJ, Kissoon N, Duckworth LJ, Sandler E, Freeman B,
tems biology of sickle stroke risk. Blood 2008;111:3872-9. Bayne E, et al. Low exhaled nitric oxide and a polymorphism
19. Nolan VG, Wyszynski DF, Farrer LA, Steinberg MH. in the NOS I gene is associated with acute chest syndrome.
Hemolysis-associated priapism in sickle cell disease. Blood Am J Respir Crit Care Med 2001;164:2186-90.
2005;106:3264-7.
20. Nolan VG, Baldwin C, Ma Q, Wyszynski DF, Amirault Y, 35. Martinez-Castaldi C, Nolan VG, Baldwin CT, et al. Asso-
Farrell JJ, et al. Association of single nucleotide polymor- ciation of genetic polymorphisms in the TGFα pathway with
phisms in KLOTHO with priapism in sickle cell anemia. Br J the acute chest syndrome of sickle cell disease. Blood 2007;
Haematol 2005;128:266-72. 110;666a.
21. Elliott L, Ashley-Koch AE, De Castro L, Jonassaint J, Price J, 36. Fertrin KY, Melo MB, Assis AM, Saad ST, Costa FF. UDP-glu-
Ataga KI, et al. Genetic polymorphisms associated with pri- curonosyltransferase 1 gene promoter polymorphism is asso-
apism in sickle cell disease. Br J Haematol 2007;137:262-7. ciated with increased serum bilirubin levels and cholecystecto-
22. Baldwin C, Nolan VG, Wyszynski DF, Ma QL, Sebastiani P, my in patients with sickle cell anemia. Clin Genet 2003; 64:
Embury SH, et al. Association of klotho, bone morphogenic 160-2.
protein 6, and annexin A2 polymorphisms with sickle cell 37. Passon RG, Howard TA, Zimmerman SA, Schultz WH, Ware
osteonecrosis. Blood 2005;106:372-5. RE. Influence of bilirubin uridine diphosphate-glucuronosyl-
23. Nolan VG, Adewoye A, Baldwin C, Wang L, Ma Q, Wy- transferase 1A promoter polymorphisms on serum bilirubin
szynski DF, et al. Sickle cell leg ulcers: associations with levels and cholelithiasis in children with sickle cell anemia. J
haemolysis and SNPs in Klotho, TEK and genes of the TGF- Pediatr Hematol Oncol 2001;23:448-51.
β/BMP pathway. Br J Haematol 2006;133:570-8. 38. Steinberg MH, West MS, Gallagher D, Mentzer W. Effects of
24. Adewoye AH, Nolan VG, Ma Q, Baldwin C, Wyszynski DF, glucose-6-phosphate dehydrogenase deficiency upon sickle
Farrell JJ, et al. Association of polymorphisms of IGF1R and cell anemia. Blood 1988;71:748-52.
genes in the transforming growth factor-β/bone morphogenet- 39. Bouanga JC, Mouélé R, Préhu C, Wajcman H, Feingold J,
ic protein pathway with bacteremia in sickle cell anemia. Clin
Galactéros F. Glucose-6-phosphate dehydrogenase deficiency
Infect Dis 2006;43:593-8.
25. Powars DR, Elliott Mills DD, Chan L. Chronic renal failure in and homozygous sickle cell disease in Congo. Hum Hered
sickle cell disease: Risk factors, clinical course, and mortality. 1998;48:192-7.
Ann Intern Med 1991;115:614-20. 40. Belhassen L, Pelle G, Sediame S, Bachir D, Carville C, Bucherer
26. Powars DR, Chan L, Schroeder WA. βS-gene-cluster haplo- Cet al. Endothelial dysfunction in patients with sickle cell dis-
types in sickle cell anemia: clinical implications. Am J Pediatr ease is related to selective impairment of shear stress-mediat-
Hematol Oncol 1990;12:367-74. ed vasodilation. Blood 2001;97:1584-9.
27. Nolan VG, Ma Q, Cohen HT, Adewoye A, Rybicki AC, 41. Forgione MA, Loscalzo J, Holbrook M, et al. The A326G (A+)
Baldwin C, et al. Estimated glomerular filtration rate in sickle variant of the glocose-6-phosphate dehydrogenase gene is
cell anemia is associated with polymorphisms of bone mor- associated with endothelial dysfunction in African Americans.
phogenetic protein receptor 1B (BMPR1B). Am J Hematol J Am Coll Cardiol 2003;41:249a[Abstract].
2007;82:179-84. 42. Leopold J, Dam A, Maron BA, Scribner AW, Liao R, Handy
28. Gladwin MT, Sachdev V, Jison ML, Shizukuda Y, Plehn JF, DE, et al. Aldosterone impairs vascular reactivity by decreas-
Minter K, et al. Pulmonary hypertension as a risk factor for ing glucose-6-phosphate dehydrogenase activity. Nat Med
death in patients with sickle cell disease. N Engl J Med 2004; 2007;13:189-97.
350:886-95. 43. Lotsch J, Skarke C, Liefhold J, Geisslinger G. Genetic predic-
29. Ashley-Koch AE, Elliott L, Kail ME, De Castro LM, Jonassaint tors of the clinical response to opioid analgesics: clinical utili-
J, Jackson TL, et al. Identification of genetic polymorphisms ty and future perspectives. Clin Pharmacokinet 2004;43:983-
associated with risk for pulmonary hypertension in sickle cell 1013.
disease. Blood 2008;111:5721-6.
30. Vichinsky EP, Neumayr LD, Earles AN, Williams R, Lennette 44. Sebastiani P, Nolan VG, Baldwin CT, Abad-Grau MM, Wang
ET, Dean D, et al. Causes and outcomes of the acute chest L, Adewoye AH, et al. A netwok model to predict the risk of
syndrome in sickle cell disease. N Engl J Med 2000;342:1855- death in sickle cell disease. Blood 2007;110:2727-35.
65. 45. Sebastiani P, Wang L, Perls T, et al. A repertoire of genes mod-
31. Sharan K, Surrey S, Ballas S, Borowski M, Devoto M, Wang ifying the risk of death in sickle cell anemia. Blood 2007; 110:
KF, et al. Association of T-786C eNOS gene polymorphism 52a[Abstract].
with increased susceptibility to acute chest syndrome in 46. Sebastiani P, Timofeev N, Hartley SH, et al. Genome-wide
females with sickle cell disease. Br J Haematol 2004;124:240-3. association studies suggest shared polymorphisms are associ-
32. Chaar V, Tarer V, Etienne-Julan M, Diara JP, Elion J, Romana ated with severity of sickle cell anemia and exceptional
M. ET-1 and ecNOS gene polymorphisms andsusceptibility to longevity. Blood 2008;1446a[Abstract].

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 237 |
Red cell disease

Diagnosis and management of erythrocytosis

M. F. McMullin A B S T R A C T

An erythrocytosis is present when there is an increase in the number of circulating red cells.
Department of Haematology, Erythrocytoses are classified as primary where there is an intrinsic problem in the erythroid compart-
Queen’s University Belfast, Belfast,
N. Ireland ment, and secondary where an external factor is driving the red cell production. They are also classi-
fied as congenital or acquired. Investigation starts with a clear history, examination and establishment
of a true erythrocytosis. The diagnosis of polycythemia vera should be made if current criteria are ful-
Hematology Education: filled. Measurement of the erythropoietin level can then direct further investigation. A low erythro-
the education program for the poietin level should lead to investigation of the erythropoietin-signalling pathway, whereas an elevat-
annual congress of the European
ed erythropoietin level would lead to investigations for all secondary causes. Management considera-
Hematology Association
tions include administration of aspirin, venesection to reduce the hematocrit and a specific target
2009;3:238-241 hematocrit for venesection but there is little objective evidence to support decisions.

he term erythrocytosis refers to an

T
Primary erythrocytosis
increase in the number of circulating A primary congenital erythrocytosis
differentiated red cells. This means results from mutations of the EPO receptor.
that the total amount of red cells (red cell An abnormal receptor in the erythroid cell
mass) is increased. As red cells make up the behaves abnormally in response to EPO
vast majority of the circulating cells, the (after it attaches to the receptor) and ulti-
hematocrit (Hct) is a reflection of the red cell mately results in increased red cells. A num-
mass, and, as red cells contain hemoglobin ber of mutations, all of which lead to a trun-
(Hb), an elevated Hct or Hb raises the possi- cation of the receptor and failure to abort
bility of an erythrocytosis. A Hb above 18.5 EPO signalling, have been described in a
dl in a male or 16.5 g/dL in a female, or a Hct number of families.3
above 0.52 in a male or 0.48 in a female is Polycythemia vera is an acquired primary
judged to be elevated, thus, raising the pos- erythrocytosis, as there is an intrinsic defect
sibility of an erythrocytosis. However, to in the erythroid progenitor cell. In the vast
establish the unequivocal presence of an majority of cases, this has been shown to be
absolute erythrocytosis, it is necessary to due to a mutation in the JAK2 gene (either
show that the measured red cell mass is the V671F mutation or rarely an exon 12
greater that 125% of predicted for an indi- mutation)4-8 leading to constitutively active
vidual’s height and weight.1 Those who intracellular signalling and cell proliferation.
have a raised Hct but do not demonstrate an
increase in red cell mass have an apparent Secondary erythrocytosis
erythrocytosis. It is clear than a Hct of 0.60 The cell has a mechanism for oxygen sens-
or more in a male or 0.56 or more in a female ing. In normoxia, oxygen activates the prolyl
is always associated with an increased red hydroxlases (PHDs), which then hydroxlate
cell mass,2 and, with these results, a red cell the a subunit of the transcription factor,
mass investigation is not required. Hypoxia Inducible Factor (HIF). This then
leads to binding of the Von Hippel Lindau
Causes of an absolute erythrocytosis (VHL) ubiquitination of HIF and destruction
An erythrocytosis can be classified as pri- of the proteins. However, in the presence of
mary or secondary. In a primary erythrocy- hypoxia, instead of going down this path-
tosis, an intrinsic defect in the erythroid pro- way, HIF-α associates with its b subunit and
genitor cells is associated with an enhanced the complex then binds to the hypoxia
response to cytokines. Whereas in a second- response elements within the genome, acti-
ary erythrocytosis, the increased red cell vating downstream genes, including EPO.
mass results from factors external to the ery- Increased EPO leads to increased red cell
throid progenitor cells, such as increased production, an appropriate response to the
erythropoietin (EPO) production from any hypoxia. Alteration in any of the genes in
cause. Both primary and secondary causes the oxygen-sensing pathway may lead to
can be congenital or acquired (for a full list the protein behaving abnormally, and even
see Table 1). in normoxia, driving the HIF down the

| 238 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

Table 1. Causes of an absolute erythrocytosis.

Primary erythrocytosis
Congenital
Erythropoietin receptor mutation
Acquired
Polycythemia vera

Secondary erythrocytosis
Congenital
VHL gene mutation (Chuvash erythrocytosis)
PHD2 mutations
HIF-2α mutations
High oxygen-affinity hemoglobin
Bisphosphoglycerate mutase deficiency
Acquired
EPO mediated
Hypoxia driven Figure 1. An initial approach to the investigation of erythro-
Central hypoxic process cytosis.
Chronic lung disease
Right-to-left cardiopulmonary vascular shunts
Carbon monoxide poisoning
Smoker’s erythrocytosis erythrocytosis.14,15
Hypoventilation syndromes including sleep apnea Anything, which would shift the oxygen dissociation
(High-altitude habitat) curve to the left, resulting in hemoglobin with high oxy-
Local renal hypoxia gen affinity leads to tissue hypoxia and secondary ery-
Renal artery stenosis throcytosis. High oxygen affinity hemoglobins, of which
End-stage renal disease over 90 variants have been described, can have this effect.
Hydronephrosis There are also rare mutations of biphosphoglycerate
Renal cysts (polycystic kidney disease) mutase, leading to deficiency of 2, 3 biphosphoglycerate
Post-renal transplant erythrocytosis and shifting the oxygen dissociation curve to the left.16
Pathologic EPO production An acquired secondary erythrocytosis can arise from
Tumors any process, which results in hypoxia, thus increasing
Cerebellar hemangioblastoma EPO production. This can be a central hypoxia, for exam-
Meningioma ple, altitude, lung disease, right to left cardiopulmonary
Parathyroid carcinoma/adenomas shunts or smoking. The hypoxia can also arise at a local
Hepatocellular carcinoma
renal level from a variety of renal disorders, such as renal
Renal cell cancer
artery stenosis. Pathological production of EPO has been
Pheochromocytoma
Uterine leiomyomas described for a variety of different tumors, for example, in
Exogenous EPO cerebellar hemangioblastoma. Erythrocytosis will also
Drug associated result from exogenous EPO or androgen administration.17
Erythropoietin administration There then remains a group of individuals with an
Androgen administration absolute erythrocytosis in whom no cause can be identi-
Idiopathic erythrocytosis fied and this is termed idiopathic erythrocytosis.

EPO: erythropoietin. Diagnostic pathway


The first step in any diagnostic pathway is to undertake
a complete history and examination. During this process,
hypoxic pathway, ultimately leading to increased EPO the causes of erythrocytosis should be considered and
production. A number of mutations in the genes for the likely factors explored. For example, the patients with
proteins in the oxygen-sensing pathway have been severe respiratory disease should be identified, and this
described in families with erythrocytosis. Thus, these can dictate further investigation (Figure 1).
congenital abnormalities are the cause of the increased Secondly, the presence of an erythrocytosis must also
red cell production. A large group of individuals be clearly established. A consistently raised Hb and Hct
described in the Chuvashia region of Russia have a sin- should be observed. In those with an isolated erythrocy-
gle mutation in the VHL gene, C598T, which alters tosis without an immediately obvious cause, further
amino acid 200 from arginine to tryptophan.9 A further investigation, including red cell mass is required to be sure
cluster with the same mutation was discovered in the that an absolute erythrocytosis is present.2
island of Ischia, Italy,10 and a disparate group, all of The main primary cause is of course polycythemia vera
Pakistani or Bangladeshi origin, have been identified.11 and the criteria for this diagnosis have been recently sim-
Several kindreds with a mutation in the PHD2 gene, plified. The United Kingdom (UK) group examined the
producing functionally abnormal protein, leading to evidence, and agreed that the presence of an elevated Hct
erythrocytosis have been described.12-13 Similarly a num- or red cell mass and a JAK2 mutation was sufficient to
ber of families and individuals with erythrocytosis have make this diagnosis.18 The World Health Organization
been shown to a have mutated HIF-2a gene leading to (WHO) group also redefined their criteria, and require a

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 239 |
14th Congress of the European Hematology Association

raised Hb or red cell mass, a JAK2 mutation and one Table 2. Investigations of an erythrocytosis with a normal
minor criteria from bone marrow biopsy changes, serum or raised EPO level.
EPO below the normal range, or in vitro endogenous ery-
throid colony formation.19 In those without a JAK2 muta- Oxygen sensing pathway gene mutations
tion, more extensive criteria are required, including VHL
changes in white cell and platelet counts.18,19 PHD2
Therefore, screening for the JAK2 mutations should be HIF-2α
an initial investigation in those who do not have an obvi- Hemoglobin electrophoresis/globin gene sequencing
ous secondary cause and in all those in whom poly- P50
Biphosphoglycerate mutase level
cythemia vera is a possibility. Bone marrow biopsy and
Pulse oximetry
cytogenetics may not be required in those who have an
Carboxyhemoglobin levels
obvious polycythemia vera but should be considered if CXR
there is any doubt about the diagnosis, and when further Radiology of head, chest and abdomen
investigating an erythrocytosis. Sleep studies
Having established the presence of an erythrocytosis
the next procedure, the EPO level should be considered.
A subnormal EPO level leads to the suspicion of an abnor-
mality in the EPO signalling pathway or a primary cause, reduce the JAK2 mutant level with induction of molecu-
whereas an elevated EPO level or one, which is inappro- lar remission.25 No vascular events occurred. A number of
priately normal for a raised Hb suggests an abnormality in JAK2 inhibitors are in development and are likely to be
the oxygen sensing pathway or a secondary cause (Figure available in the future for therapy of polycythemia vera.
1). TG101348 is a small, orally administered molecule,
Further investigation in those with a subnormal EPO which inhibits JAK2. In a phase myelofibrosis I study, it
level should include an EPO receptor mutation screen and showed reduction in splenomegaly.26 XL019 in another
testing for JAK2 exon 12 mutations. oral highly selective JAK2 inhibitor, which in phase I stud-
In those with a raised or inappropriately normal EPO ies in myelofibrosis, showed reduction in splenomegaly.27
level, an extensive list of investigations can be considered In similar phase I/II studies, INCB018424, a potent selec-
(Table 2). The order and extent to which these investiga- tive JAK2 inhibitor, showed reduction in spenomegaly
tions should be carried out is driven by the history and and marked improvement in constitutional symptoms.28
examination. In those with an obvious secondary cause, CEP-701 has JAK2 inhibitory activity and is showing ini-
further investigation may not be necessary, whereas in tially encouraging responses in a study of patient with
the young patient with a marked erythrocytosis, exten- polycythemia vera and essential thrombocythemia.29A
sive investigation may be required in order to attempt to histone deacetylase inhibitor ITF2357 has JAK2 inhibito-
identify the cause. ry activity has also shown encouraging responses in
myelofibrosis or polycythemia vera and essential throm-
Management bocythemia refractory to other therapy.30 More trials and
Elevated red cell masses cause an increase in blood vis- compounds are in development. In those with a pure ery-
cosity, which may cause thromboembolic events. throcytosis from other causes, there is a paucity of evi-
Therefore, management must consider reduction of the dence to guide management. However, some factors can
red cell mass, and therefore, reduction of the viscosity. be considered. A study with controls of those homozy-
The Hct reflects the whole blood viscosity most acurate- gous for the Chuvash VHL mutation found that those
ly;20 therefore, the Hct level should be considered and the with the mutation had an increased morbidity and mor-
target for reduction. tality from thromboembolic events. History of venesec-
In polycythemia vera, a retrospective study found a tion did not significantly influence the event rate but the
relationship between the Hct and thromboembolic events numbers who were not venesected were very small.31
and showed that there was an increased event rate unless If venesection is to be undertaken, there is no guidance
the Hct was reduced to less that 0.45.21 This study led to about the target Hct to aim for and, therefore, the original
the recommendation that, in polycythemia vera, the Hct evidence to reduce to 0.45 is the only information avail-
should be maintained below 0.45. Venesection can be able to guide management. This may be very difficult to
used to achieve this target. However, in the large achieve and maintain in those starting with very elevated
European Collaboration on Low-dose Aspirin in Hct. Aspirin at low dose is also often considered in those
Polycythemia Vera (ECLAP) prospective study, the Hct with a presumed thromboembolic risk. In the Chuvash
did not correlate with the rate of occurrence of thrombot- group, aspirin use was associated with an increased but
ic events.22 In this study, the incidence of thrombotic non-significant risk of thrombosis,31 whereas in the ran-
events, mainly myocardial infarction, was related to domised study ECLAP, it was shown to be beneficial.32
white cell counts greater that 15×109/L.23 Low dose aspirin is relatively safe and it is likely that most
Therapy in polycythemia vera is directed to reducing all patients will be offered this therapy in the absence of a
the blood counts with the aim of reducing the likelihood specific contraindication. In many of the specific second-
of thromboembolic events, and transformation to ary erythrocytoses, there is limited evidence suggesting
myelofibrosis or acute leukemia. Guidelines have been some control of the Hct with venesection, taking into
developed providing advice on management, taking into account other events in the patient’s history. In the case of
account the patient’s age and risk factors.24 However, the familial defects, performance of other affected family
JAK2 mutation provides a specific and measurable target members may be beneficial. Judicious limits for the
for therapy. Pegylated interferon has been shown to reduction of Hct to much higher levels than 0.45 have

| 240 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

been suggested.24 13. Percy MJ, Furlow PW, Beer PA, Lappin TR, McMullin MF, Lee
FS. A novel erythrocytosis-associated PHD2 mutation sug-
In the congenital defects of the oxygen-sensing path- gests the location of a HIF binding groove. Blood 2007; 110:
way studies, including the largest cohort from Chuvashia, 2193-6.
no tumors were reported.31 However, a new mutation in 14. Percy MJ, Furlow PW, Lucas GS, Li X, Lappin TR, McMullin
PHD2, H374R, was recently described in association with MF, et al. A Gain-of-Function mutation in the HIF2A gene in
familial erythrocytosis. N Eng J Med 2008;335:52-8.
a paraganglioma. There was loss of heterozygosity of 15. Percy MJ, Beer PA, Campbell G, Dekker AW, Green AR,
PHD2 in the tumor, suggesting the PHD2 could be a Oscier D, et al. Novel exon 12 mutations in the HIF2a gene
tumor-suppressor gene.33 This patient was first seen with associated with erythrocytosis. Blood 2008;111:5400-2.
erythrocytosis at the age of 30 years. The tumor was first 16. Rosa R, Prehu MO, Beuzard Y, Rosa J. The first case of a com-
plete deficiency of diphosphoglycerate mutase in human ery-
seen 13 years later. Many of the other recently described throcytes. J Clin Invest 1978;162:907-15.
patients with oxygen sensing pathway mutations are 17. McMullin MF. The classification and diagnosis of erythrocyto-
much younger so observation and screening for tumors sis. Int J Labor Hematology 2008;30:447-59.
may become an issue in management. 18. McMullin MF, Reilly JT, Campbell P, Bareford D, Green AR,
Harrison CN, et al. Amendment to the guideline for the diag-
nosis and investigation of polycythaemia/erythrocytosis.
Conclusion British Committee for Standards in Haematology. Br J
In polycythemia vera management, venesection and Haematol 2007;138:821-2.
19. Thiele J, Kvasnicka HM, Orazi A. Polycythaemia vera. In
low dose aspirin have benefits. Cytoreduction depend- WHO classification of tumours of haematopoietic and lym-
ing on the risk, group and age should be considered. phoid tissues. IARC, Lyon. 2008. p 40- 3.
JAK2 inhibitors are likely to be used in the future. In 20. Pearson TC, Grimes AJ, Slater NGP, Wetherley-Mein G.
other erythrocytosis, venesection can be used to reduce Viscosity and iron deficiency in treated polycythaemia. Br J
Haematol 1981;49:123-7.
the Hct. There is conflicting evidence as to the benefits 21. Pearson TC, Wetherley-Main G. Vascular occlusive episodes
of this strategy and how much the level of Hct should and venous haematocrit in primary proliferative polycy-
be reduced. In the absence of other guidance, a thera- thaemia. Lancet 1978;2:1219-22.
peutic target of 0.45 should be considered. Low dose 22. Di Nisio M, Barbui T, Di Gennaro L, Borrelli G, Finazzi G,
Landolfi R, et al. The haematocrit and platelet target in poly-
aspirin may also be considered. cythaemia vera. European Collaboration on Low-dose Aspirin
in Polycythemia Vera (ECLAP) Investigators. Br J Haematol
2007;136:249-59.
References 23. Landolfi R, Di Gennaro L, Barbui T, De Stefano V, Finazzi G,
Marfisi R, et al. Leukocytosis as a major risk factor in patients
1. Pearson TC, Guthrie DL, Simpson J, Chinn S, Barosi G, Ferrant with polycythaemia vera. European Collaboration on Low-
A, et al. Interpretation of measured red cell mass and plasma Dose Aspirin in Polycythemia Vera (ECLAP). Blood 2007; 109:
volume in adults: Expert Panel on Radionuclides of the 2446-52.
International Council for Standardization in Haematology. Br 24. McMullin MF, Reilly JT, Campbell P, Bareford D, Green AR,
J Haematol 1995;89:748-56. Harrison CN, et al. Amendment to the guideline for the diag-
2. Johansson PL, Soodabeh S-K, Kutti J. An elevated venous nosis and investigation of polycythaemia/erythrocytosis.
haemoglobin concentration cannot be used as a surrogate British Committee for Standards in Haematology. Br J
marker for absolute erythrocytosis: a study of patients with Haematol 2007;138:821-2.
polycythaemia vera and apparent polycythaemia. Br J 25. Kiladjian J-J, Cassinat B, Chevret S, Turlure P, Cambier N,
Haematol 2005;129:701-5. Roussel M, et al. Pegylated interferon-α-2a induces complete
3. Percy MJ. Genetically heterogeneous origins of idiopathic ery- hematologic and molecular responses with low toxicity in
throcytosis. Hematology 2007;12;131-9. polycythemia vera. Blood 2008;122:3065-72.
4. Baxter EJ, Scott LM, Campbell PJ, East C, Fourouclas N, 26. Parandani AD, Gitib J, Jamieson C. A phase I study of
Swanton S, et al. Acquired mutation of the tryrosine kinase TG101348, an orally biovailable JAK2-selective inhibitor, in
JAK2 in human myeloproliferative disorders. Lancet 2005; patient myelofibrosis. Blood 2008;99[Abstract].
365:1054-61. 27. Shah NP, Olszynski P, Sokol L, et al. A phase I study of XL019,
5. James C, Ugo V, Le Couédic JP, Staerk J, Delhommeau F, a selective JAK2 inhibitor, in patients with primary myelofi-
Lacout C, et al. A unique clonal JAK2 mutation leading to con- brosis, polycythaemia vera, or post-essential thrombo-
stitutive signalling causes polycythemia vera. Nature 2005; cythemia myelofibrosis. Blood 2008;98[Abstract].
434:1144-8. 28. Verstovsek S, Kantarjian HM, Pardanani AD. The JAK2
6. Kralovics R, Passamonti F, Buser AS, Teo SS, Tiedt R, Passweg inhibitor, INCB18424, demonstrated durable and marked clin-
JR, et al. A gain-of-function mutation of JAK2 in myeloprolif- ical responses in primary myelofibrosis (PMF) and post-poly-
erative disorders. N Engl J Med 2005;352:1779-80. cythemia/essential thrombocythemia myelofibrosis (Post
7. Levine RL, Wadleigh M, Ebert BL, Wernig G, Huntly BJ, et al. PV/ET-IMF) Blood. 2008:1762[Abstract].
Activating mutation in the tyrosine kinase JAK2 in poly- 29. Moliterno AR, Roboz GL, Carroll M. An open-label study of
cythemia vera, essential thrombocythemia, and myeloid CEP-701 in patients with JAK2 V617F-positive polycythemia
metaplasia with myelofibrosis. Cancer Cell 2005;7:387-97. vera and essential thrombocytosis. Blood 2008;99[Abstract].
8. Scott LM, Tong W, Levine RL, Scott MA, Beer PA, Stratton 30. Rambaldi A, Dellacasa CM, Salmoiraghi S. A phase 2A study
MR, et al. JAK2 exon 12 mutations in polycythemia vera and
idiopathic erythrocytosis. N Eng J Med 2007;356:459-68. of the histone-deacetylase inhibitor ITF2357 in patients with
9. Ang SO, Chen H, Gordeuk VR, Sergueeva AI, Polyakova LA, JAK2 V617F positive chronic myeloproliferative neoplasms.
Miasnikova GY, et al. Endemic polycythaemia in Russia: Blood 2008;100[Abstract].
mutation in the VHL gene. Blood Cells Mol Dis 2002;28:57-62. 31. Gordeuk VR, Sergueeva AI, Miasnikova GY, Okhotin D,
10. Perrotta S, Novili B, Ferraro M, Migliaccio C, Borriello A, Voloshin Y, Choyke PL, et al. Congenital disorder of oxygen
Cucciolla V, et al. Von Hippel-Lindau-dependent polycythe- sensing: association of the homozygous Chuvash poly-
mia is endemic on the island of Ischia: identification of a novel cythaemia VHL mutation with thromobosis and vascular
cluster. Blood 2006;107:514-9. abnormalities but not tumors. Blood 2004;103:3924-32.
11. Percy MJ, McMullin MF, Jowitt SN, Potter M, Treacy M, 32. Landolfi R, Marchioli R, Kutti J, Gisslinger H, Tognoni G,
Watson WH, et al. Chuvash-type congenital polycythemia in Patrono, et al. Efficacy and safety of low-dose aspirin in poly-
4 families of Asian and Western European ancestry. Blood cythemia vera. Collaboration on Low-Dose Aspirin in
2003; 102:1097-9. Polycythemia Vera Investigators. N Engl J Med 2004;350:114-
12. Percy MJ, Zhao Q, Flores A, Harrison C, Lappin TR, Maxwell 24.
PH, et al. A family with erythrocytosis establishes a role for 33. Ladroue C, Carcenac R, Leporrier M, Gad S, Le Hello C,
prolyl hydroxylase domain protein 2 in oxygen homeostasis. Galateau-Salle F, et al. PHD2 mutation and congenital erythro-
Proc Nat Acad Sci USA 2006;103:654-9. cytosis and paaganglioma. N Engl J Med 2008; 359:2685-92.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 241 |
Stem cell transplantation

Immunobiology of reduced intensity conditioning


in hematopoietic stem cell transplantation

F. Baron1 A B S T R A C T
B. Sandmaier2,3 Reduced-intensity and truly nonmyeloablative conditioning regimens followed by allogeneic
hematopoietic stem cell transplantation (SCT) have been evaluated in patients with hematological
1
Giga-research, division of malignancies who were not considered candidates for conventional SCT because of age or medical
Hematology, University of Liège, comorbidities. Reduced intensity conditioning regimens have been aimed at both eliminating host-
Belgium; 2Fred Hutchinson Cancer versus-graft reactions (graft rejection) and producing major anti-tumor effects. Conversely, nonmye-
Research Center; 3University of loablative conditioning regimens have relied on optimization of pre- and post-transplant immunosup-
Washington, Seattle, WA, USA pression to overcome host-versus-graft reactions, thereby allowing engraftment and eradication of
tumors nearly exclusively via immune-mediated graft-versus-tumor effects. Remarkably, a minimally
Hematology Education: toxic regimen of 2 Gy total body irradiation with or without fludarabine followed by postgrafting
the education program for the immunosuppression with cyclosporine and mycophenolate mofetil has assured engraftment rates
annual congress of the European almost similar to those after myeloablative conditioning. While nonmyeloablative and reduced-inten-
Hematology Association sity SCT have been associated with reduced regimen-related toxicities and have been curative for
many patients with otherwise fatal hematological malignancies, challenges have remained in regard
2009;3:242-249 to acute graft-versus-host disease, infections, and disease progression.

Allogeneic hematopoietic stem cell of graft-versus-tumor effects came from the


transplantation and graft-versus-tumor effects observations by Kolb et al. and Slavin et al.
Most hematological malignancies display that donor lymphocyte infusions (DLI) were
a dose-response susceptibility to radiation able to eradicate the malignancy in a number
therapy and alkylating agents. Because mye- of patients who relapsed with chronic
loablation is the dose-limiting toxicity of myeloid leukemia5 or acute lymphoblastic
many of these agents, allogeneic hematopoi- leukemia (ALL)6 after allogeneic SCT. Since
etic stem cell transplantation (SCT) was first then, DLI has also proven to be effective in a
developed as a way to rescue patients from number of patients with acute myeloid
severe myelosuppression occurring after leukemia (AML),7 multiple myeloma (MM),8
administration of high-dose chemoradio- chronic lymphocytic leukemia (CLL),9 non-
therapy.1 Despite major improvements in hodgkin lymphoma (NHL),10 and Hodgkin
supportive care in the last 40 years, the non- lymphoma (HL).11
hematopoietic toxicities of high-dose condi-
tioning has restricted its use to patients Nonmyeloablative and reduced-intensity
younger than 55-60 years. Given that medi- conditioning
an ages at diagnosis for patients with most Two key observations in 1997 led to the
hematologic malignancies range from 65-70 emergence of nonmyeloablative and
years,1 the majority of such patients could reduced-intensity conditioning, respective-
not benefit from potentially curative allo- ly. First, Storb et al. demonstrated that opti-
geneic SCT. Further, some patients have mizing post-transplant immunosuppression
medical cormorbidities, which exclude them could not only control GVHD but also graft
from high-dose conditioning. rejection.12 Secondly, Giralt et al. and Slavin
Since the late 1970s, it has become appar- et al. showed that administration of purine
ent that an important part of the efficacy of analogs, such as fludarabine provided suffi-
allogeneic SCT was mediated by immune cient immunosuppression to allow sus-
reactions of the graft itself against recipient tained engraftment of HLA-matched allo-
tumor cells, termed graft-versus-tumor geneic stem cells.13,14 Based on these obser-
effects.2,3 Indeed, patients who developed vations, several groups of investigators
acute and/or chronic graft-versus-host dis- developed a number of reduced intensity13-16
ease (GVHD) had lower risks of relapse than or truly nonmyeloablative17-20 conditioning
those who did not.2-4 In addition, patients regimens for allogeneic SCT. Reduced inten-
given syngeneic SCT and those given T-cell sity conditioning regimens have been aimed
depleted grafts had substantially higher risk at eliminating host-versus-graft reactions
of relapse than those receiving unmanipulat- (graft rejections) and producing major anti-
ed grafts from allogeneic donors.4 tumor effects. Most reduced-intensity con-
Furthermore, a direct evidence of the power ditioning regimens have combined fludara-

| 242 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

bine with relatively high doses of busulfan (8 mg/kg),21


melphalan (140 mg/m2),22 or thiotepa.16 Conversely,
nonmyeloablative conditioning regimens have relied
on optimization of pre- and post-transplant immuno-
suppression to overcome host-versus-graft reactions,
thereby allowing engraftment,12,23 while eradication of
tumors has depended nearly exclusively on the graft-
versus-tumor effects.24 Examples of nonmyeloablative
conditioning regimens include low-dose (2 Gy) TBI
with or without added fludarabine,18 8 Gy total lym-
phoid irradiation with anti-thymocyte globulin
(ATG),25 or fludarabine with cyclophosphamide.17
Figure 1 shows commonly used conditioning regimens
in relation to their immunosuppressive and myelosup-
pressive properties.
Typical complications of high-dose therapy, such as
nausea, vomiting, pancytopenias, mucositis, and new- Figure 1. Commonly used conditioning regimens in rela-
onset alopecia have been observed with most of tion to their immunosuppressive and myelosuppressive
reduced-intensity conditioning regimens, and sinu- properties.83 Please note that this classification is not
based on direct experimentation, and is thus hypothetical.
soidal obstructive syndrome has also been seen, Ale, alemtuzumab; ATG, antithymocyte globulin; Bu8,
although less frequently than after myeloablative con- busulfan 8 mg/kg; Bu16, busulfan 16 mg/kg; Cy,
ditioning.21,22,26-29 Conversely, nonmyeloablative regi- cyclophosphamide; Cy120, cyclophosphamide 120
mg/kg; Cy200, cyclophosphamide 200 mg/kg; F, fludara-
mens produced only mild myelosuppression and few bine; Flag-Ida, fludarabine/cytosine arabinoside/idaru-
regimen-related toxicities,30 and have been associated bicin; M, melphalan; M 140, melphalan 140 mg/m2; M
with a low 100-day incidence of nonrelapse mortality, 180, melphalan 180 mg/m2; TBI, total body irradiation;
TLI, total lymphoid irradiation; TT, thiotepa. (From
even in elderly patients and those with comorbid con- Sandmaier BM, Storb R. "Reduced-intensity conditioning
ditions.18 Typically, many transplants following non- followed by hematopoietic cell transplantation for hemato-
myeloablative conditioning could be carried out entire- logic malignancies." In: Appelbaum FR, Forman SJ, Negrin
ly in the outpatient setting.31 RS, Blume KG (eds): Thomas' Hematopoietic Cell
Transplantation. Oxford, UK: Wiley-Blackwell, p. 1043-
No randomized study thus far has compared non- 1058, 2009. Used with permission from the authors.
myeloablative versus reduced-intensity conditioning
regimens. A recent analysis by the Société Française de
Greffe de Moelle et de Thérapie Cellulaire (SFGM-TC)
retrospectively compared outcomes in patients given Non-myeloablative versus myeloablative conditioning
grafts after 2 Gy TBI with or without added fludarabine In order to determine the relative contributions of
(n=255; nonmyeloablative conditioning) or after flu- conditioning intensity and graft-versus-host reactions to
darabine, busulfan plus ATG (n=465; reduced-intensity transplant related complications, several retrospective
conditioning) as treatment for various hematological studies compared transplant-related toxicities after non-
malignancies.32 Three-year overall and disease-free sur- myeloablative versus myeloablative SCT. Not surpris-
vivals were both similar in the two groups. ingly, the hematological changes after nonmyeloabla-
tive conditioning were milder than those seen after
Engraftment kinetics myeloablative conditioning.42 Specifically, neutrophil
By definition, nonmyeloablative and reduced-intensi- counts remained above 500 throughout the transplant
ty conditioning regimens usually lead to an initial state period in a high proportion of patients given nonmye-
of mixed chimerism defined as co-existence of loablative conditioning, while temporary grade IV neu-
hematopoiesis of host and donor origin.33 Several factors tropenia was the rule in patients given reduced-intensi-
have been associated with faster donor T-cell engraft- ty or myeloablative conditioning. Further, patients given
ment, including higher intensity of the conditioning reg- nonmyeloablative or reduced-intensity conditioning
imen,33,34 having received prior myelosuppressive required less platelet and red blood cell transfusions
chemotherapy,35,36 the use of peripheral blood stem cells than those given myeloablative conditioning.43
(PBSC) instead of marrow as a stem cell source,20,37,38 a Similarly, liver, kidney, gastro-intestinal, and lung toxic-
high number of CD34+ and T-cells in the graft,35,37,38 and ities were significantly reduced with nonmyeloablative
intense post-grafting immunosuppression.39 conditioning.44-47
Further, several authors have demonstrated correla- Three large retrospective studies from the European
tions between engraftment kinetics and clinical events. Group for Blood and Marrow Transplantation (EBMT)
Specifically, high levels (>50%) of donor T- and NK-cell compared SCT outcomes of patients given various mye-
chimerism 1 month after SCT have each been associat- loablative versus various reduced-intensity/nonmye-
ed with lower risk of graft rejection.18,36 While high day loablative conditioning regimens as treatment for
14 to 42 donor T-cell chimerism levels were associated AML,48 myelodysplastic syndrome (MDS),49 or CLL50
with high incidence of acute GVHD,36,40 high donor NK (Figure 2). Obviously, these studies were limited by the
cell levels had no such association.41 Conversely, high fact that fitter patients were probably more often pro-
donor NK cell41 and T cell40,41 chimerism levels early after posed myeloablative regimens, while older and sicker
SCT have each been associated with low relapse risk patients were probably more often given nonmyeloab-
and good progression-free survival. lative or reduced-intensity conditioning. Nevertheless,

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 243 |
14th Congress of the European Hematology Association

Figure 2. Retrospective studies comparing SCT outcomes Figure 3. (A) Impact of acute and chronic GVHD and of
after reduced-intensity (RIC) or myeloablative conditioning achievement of full donor T-cell chimerism (Ach. FDC) on
as treatment for myelodysplastic syndrome (MDS),49 acute relapse risk in 322 patients reported in ref. 24 given grafts
myeloid leukemia (AML),48 or chronic lymphocytic after 2 Gy TBI with or without fludarabine. (B) Impact of
leukemia (CLL).50 TRM, transplant related mortality. The chronic GVHD on relapse risk according to disease group
black bars show the hazard ratio and the grey bars the in the same study. The black bars show the hazard ratio
95% confidence intervals. and the grey bars the 95% confidence intervals.

these studies found similar disease-free and overall sur- p=0.7, respectively). In contrast, among patients with
vival in the two groups of patients, since nonrelapse comorbidities (HCT-CI score ≥1) at SCT, the use of non-
mortality was lower in nonmyeloablative patients, but myeloablative conditioning was associated with lower
relapse rate was lower in myeloablative recipients.48-50 nonrelapse mortality (HR: 0.19; p<0.001) and better
Further, Scott et al. compared efficacy of SCT after overall survival (HR: 0.33; p=0.007) in multivariate
nonmyeloablative (with 2 Gy TBI with or without analyses. These observations suggest that the HCT-CI
added fludarabine; n=38) or myeloablative conditioning score might help in determining which patients might
(with busulfan [16 mg/kg, targeted to 800 to 900 ng/mL] benefit from nonmyeloablative or reduced-intensity
and cyclophosphamide [120 mg/kg; n=112]) in patients conditioning, and which others could safely receive
with MDS over 40 years of age.51 In multivariate analy- myeloablative regimens.
ses adjusted for IPSS group and comorbidity at SCT, 3-
year overall survival [(HR=1.2, p=0.56), progression-free Graft-versus-host disease and graft-versus-tumor effects
survival (HR=1.1, p=0.60), progression rate (HR=1.3, after nonmyeloablative conditioning
p=0.43), and nonrelapse mortality (HR=1.0, p=0.94)] did Graft-versus-host disease
not differ significantly between the two groups of Several studies have shown slightly lower day-100
patients. incidence of acute GVHD after nonmyeloablative than
More recently, Sorror et al. compared outcomes after myeloablative conditioning.48,51,55,56 In contrast, the
among patients with lymphoma or CLL given either incidence of chronic GVHD has been comparable in
nonmyeloablative (n = 152, consisting of 2 Gy TBI with patients given nonmyeloablative or myeloablative con-
or without added fludarabine) or myeloablative (n = 68) ditioning.48,51,55,56
conditioning.52 Outcomes were stratified by the Some reduced-intensity conditioning regimens have
hematopoietic cell transplantation (HCT)-specific co- used in vivo T-cell depletion of the grafts (with either
morbidity index (HCT-CI).53,54 Among patients without anti-thymocyte globulin (ATG) or alemtuzumab) in
comorbidity at SCT (HCT-CI = 0), survival and nonre- order to decrease the incidence of acute and chronic
lapse mortality were comparable for patients given non- GVHD. While these strategies achieved their goal,15,57
myeloablative or myeloablative conditioning (p=0.7 and increased incidences of both infections (due to slower

| 244 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

immune recovery)58 and disease relapse59 were


observed, especially when high doses of these agents
were used.32 Based on murine experiments, the Stanford
University group investigated an innovative nonmye-
loablative regimen that favored the presence of a high
proportion of regulatory NK-T cells.60 This regimen
combined total lymphoid irradiation (TLI, 8 Gy) with
ATG (Thymoglobulin, 7.5 mg/kg total dose), and post-
grafting immunosuppression with mycophenolate
mofetil and cyclosporine. Initial results in 37 patients
with various hematological malignancies indicated that
this regimen was indeed associated with a low inci-
dence of acute and chronic GVHD, while graft-versus-
tumor effects were apparently preserved,25 some late
relapses did occur.61

Association between graft-versus-host disease and


graft-versus-tumor effects
There has been a close relationship between GVHD
and graft-versus-tumor responses after myeloablative
conditioning.2,4,62,63 We investigated whether such a rela-
tionship existed for patients given nonmyeloablative
conditioning in 322 patients given grafts from HLA-
matched related (n=192) or unrelated (n=130) donors
after 2 Gy TBI with or without added fludarabine.24
Grade II, III and IV acute GVHD occurred in 44%, 11%
and 3% of the patients, respectively, while extensive
chronic was seen in 56% of the patients. While grades
II and III-IV acute GVHD had no significant impact on
relapse/progression, they were associated with
increased non-relapse mortality and decreased progres-
sion-free survival. Conversely, extensive chronic GVHD
was associated with decreased relapse/progression
(p=0.006; Figure 3), and better progression-free survival Figure 4. Susceptibility of disease groups to graft-versus-
(p=0.003). The greatest reduction of relapse risk in tumor effects. Relapse rates per patient year during the
patients with chronic GVHD was observed in the sub- first 2 years after SCT corrected for follow-up and compet-
group of patients with AML or MDS (p<0.001; Figure 3), ing nonrelapse mortality in a cohort of 834 consecutive
patients reported in ref 69 given grafts after 2 Gy TBI with
while a similar trend was observed in patients with or without added fludarabine: (A) in patients in complete
lymphoma or CLL (p=0.12). In contrast, the study failed remission (CR) at SCT. (B) In patients not in CR at SCT. CLL,
to show a significant association between GVHD and chronic lymphocytic leukemia; NHL-LG, low-grade non-
Hodgkin lymphoma; MM, multiple myeloma; MCL, mantle
relapse risk in patients with MM (p=0.41). cell lymphoma; NHL-HG, high-grade NHL; ALL, acute lym-
Confirming these observations, several groups of phoblastic leukemia; AML, acute myeloid leukemia; HL,
investigators observed a significant association between Hodgkin lymphoma; MDS-RA, myelodysplastic syndrome
occurrence of chronic GVHD and low relapse risk and with refractory anemia; MPD, myeloproliferative disease;
CMML, chronic myelomonocytic leukemia.
high progression-free survival in patients with AML or
MDS. Specifically, Blaise et al. analyzed outcomes of 33
patients with AML in first complete remission (CR)
receiving allogeneic SCT from HLA-identical siblings survival. In contrast, acute GVHD was not associated
following reduced-intensity conditioning with fludara- with a lower risk of relapse.
bine, busulfan and ATG.64 In a landmark analysis start- The impact of chronic GVHD on relapse risk in
ing on day 100, occurrence of chronic GVHD was asso- patients with MM has been more controversial. Indeed,
ciated with a lower risk of relapse (0% versus 44%, two studies found lower risk of relapse and better pro-
p=0.007) and better disease-free survival (95% versus gression-free survival in MM patients who developed
53%, p=0.007). More recently, Valcarcel et al. studied chronic GVHD in time-dependent Cox analyses,59,66 while
data from 93 patients with AML (n=59) or MDS (n=34) two others failed to find such an association.67,68 Finally, a
given allogeneic SCT after a reduced intensity condi- recent analysis from the EBMT demonstrated in a land-
tioning consisting of fludarabine and busulfan, and post- mark analysis that Hodgkin lymphoma patients who had
grafting immunosuppression with cyclosporine plus chronic GVHD before 9 months after SCT had lower risk
methotrexate or mycophenolate mofetil.65 The 4-year of relapse than patients who did not (p=0.008).11
overall and disease-free survival rates were 45% and
43%, respectively. The 4-year cumulative incidence of Disease susceptibility to graft-versus-tumor effects
chronic GVHD was 53% (45% extensive), and its devel- In order to assess the susceptibility of the different
opment was the major factor associated with lower hematological malignancies to graft-versus-tumor
relapse incidence and improved overall and disease-free effects, Kahl et al. analyzed the relapse risk according to

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 245 |
14th Congress of the European Hematology Association

disease characteristics in a cohort of 834 consecutive (150 mg/m2), cyclophosphamide (29 mg/kg), and 2 Gy
patients (median age, 55 years; range, 5 to 74 years). TBI in 68 patients with advanced hematologic malig-
They were given related (n=498) or unrelated (n=336) nancies (n=67), or paroxysmal nocturnal hemoglobin-
grafts after 2 Gy TBI alone (n=171) or combined with uria (n=1).71 Postgrafting immunosuppression consisted
fludarabine (90 mg/m2; n=663).69 Besides the variation in of MMF, tacrolimus, and cyclophosphamide, the latter
tumor burden, which had a major impact on the relapse given at a dose of 50 mg/kg on day 3 or on days 3 and
risk, graft-versus-tumor effects were most impressive in 4 after SCT. Nine patients (13%) had graft rejection,
patients with indolent NHL, CLL and mantle cell lym- while 59 achieved sustained donor engraftment. The
phoma. Specifically, relapse rates per patient year (PY) 200-day cumulative incidences of grades II-IV and III-IV
during the first 2 years after SCT (corrected for follow- acute GVHD were 34% and 6%, respectively, while
up and competing nonrelapse mortality) were between chronic GVHD occurred in less than 30% of the
0.0 and 0.24 in patients with CLL and MM in CR (most patients. With a median follow-up of 2 years, 2-year
of whom received tandem autologous/allogeneic SCT), overall and disease-free survivals were 36% and 26%,
low-grade or mantle cell NHL in CR or in partial remis- respectively.
sion (PR), and high-grade NHL in CR (Figure 4). In con-
trast, patients with advanced myeloid and advanced Conclusions and perspectives
aggressive lymphoid malignancies had rates of more Nonmyeloablative and reduced-intensity condition-
than 0.50. Patients with lymphoproliferative diseases ing regimens have allowed older patients, those who
not in CR (except Hodgkin lymphoma and high-grade had failed a prior myeloablative SCT,72 and those with
NHL) and myeloid malignancies in CR had rates of 0.26 comorbidity to benefit from the potentially curative
to 0.37. The exact reasons for the variable graft-versus- graft-versus-tumor effects. While graft-versus-tumor
tumor effects are unclear, but might include variability effects were most impressive in patients with CLL,
in kinetics of tumor cell growth, presentation and den- indolent NHL or mantle cell lymphoma, patients with
sity of minor-histocompatibility (and perhaps tumor- chemosensitive aggressive lymphoma or Hodgkin lym-
specific) target antigens on tumor cells, susceptibility of phoma, and those with MDS or myeloprolifarative dis-
tumor cells to cytotoxic cell kill, or access of tumor to orders with less than 5% marrow blasts, or acute
donor cytotoxic cells. leukemias in complete remission also had encouraging
Nonmyeloablative or reduced-intensity conditioning for results following nonmyeloablative or reduced-intensi-
ty conditioning.25 Anti-tumor responses in some disease
cord blood or HLA-haploidentical stem cell transplantation
types required extended periods, with some patients
Given that HLA-matched donors can only be found
achieving a CR more than 1 year after SCT.18,24
for 50-80% of patients, depending on their ethnic
Occurrence of chronic GVHD has been associated with
group, there has been a considerable interest to extend
lower risk of relapse and better progression-free survival
the use of nonmyeloablative or reduced-intensity condi-
in several studies, and particularly so in patients with
tioning to cord blood or HLA-haploidentical SCT. Due
AML or MDS.
to greater degrees of histoincompatibility, the use of
such alternative donors has been associated with For patients with non-malignant diseases, ongoing
increased risks of both graft rejection and GVHD. efforts are directed at further decreasing the intensity of
Brunstein et al. investigated the feasibility of unrelated the conditioning regimen needed to achieve sustained
cord blood transplantation after nonmyeloablative con- donor engraftment. Promising strategies aimed at reduc-
ditioning consisting of fludarabine (200 mg/m2), ing the intensity of host immune responsiveness include
cyclophosphamide (50 mg/kg), and 2 Gy TBI.70 Data pre-transplant administration of donor cells together
from 110 patients (median age 51 [range, 17-69] years) with T-cell costimulation blocking agents,73,74 or pre-
with hematologic malignancies (n=106) or aplastic ane- transplant administration of radiolabeled monoclonal
mia (n=4) given one (n=17) or two (n=93) unrelated cord antibodies directed against T-cell receptors.75
blood units have been recently analyzed.70 Thirty-nine For patients with hematological malignancies, ongo-
patients not given myelosuppressive chemotherapy in ing efforts are directed at better preventing acute
the 6 months preceding SCT were also given ATG. GVHD, better treating extensive chronic GVHD, and
Postgrafting immunosuppression consisted of MMF and preventing relapse in patients with aggressive malignan-
CSP. Cord blood units were predominantly 1- or 2- cies by reducing tumor burden at the time of SCT,
HLA-antigen mismatched with the recipient. Primary through preceding cytoreductive autologous SCT in the
neutrophil recovery occurred in 92% of patients at a cases of multiple myeloma,76 or the addition of radiola-
median of 12 days (range, 0-32) after SCT, while the beled monoclonal antibodies against CD20 or CD45 in
cumulative incidence of sustained donor engraftment case of advanced non-Hodgkin lymphoma,77 or myeloid
was 85%. The cumulative incidence of grade II-IV and malignancies.78 The addition of disease-targeted thera-
grade III-IV acute GVHD were 59% and 22%, respec- py, such as imatinib, thalidomide, bortezomib, lenalido-
tively, while chronic GVHD was seen in 23% of mide or monoclonal antibodies after transplant seems
patients. Three-year overall and progression-free sur- also promising.9,78-81 Finally, recent progress in large scale
vivals were 45% and 38%, respectively. The most fre- identification of minor-histocompatibility antigens82
quent causes of death were disease relapse/progression might allow the adoptive transfer of T-cell populations
(n=27), infection (n=12) and GVHD (n=6). specifically directed against recipient minor-histocom-
O’Donnell et al. investigated feasibility of unmanipu- patibility antigens expressed only by hematopoietic
lated HLA-haploidentical marrow transplantation after (including tumor) recipient cells, thus increasing graft-
nonmyeloablative conditioning combining fludarabine versus-tumor effects without inducing GVHD.

| 246 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

donor T-cell chimerism precedes alloimmune responses. Blood


References 1999;94:3234-41.
20. Maris MB, Niederwieser D, Sandmaier BM, Storer B, Stuart
1. Storb R. Allogeneic hematopoietic stem cell transplantation - M, Maloney D, et al. HLA-matched unrelated donor hemato-
yesterday, today, and tomorrow. Exp Hematol 2003;31:1-10. poietic cell transplantation after nonmyeloablative condition-
2. Weiden PL, Flournoy N, Thomas ED, Prentice R, Fefer A, ing for patients with hematologic malignancies. Blood 2003;
Buckner CD, et al. Antileukemic effect of graft-versus-host 102:2021-30.
disease in human recipients of allogeneic-marrow grafts. N 21. Or R, Shapira MY, Resnick I, Amar A, Ackerstein A, Samuel S,
Engl J Med 1979;300:1068-73. et al. Nonmyeloablative allogeneic stem cell transplantation
3. Weiden PL, Sullivan KM, Flournoy N, Storb R, Thomas ED, for the treatment of chronic myeloid leukemia in first chronic
and the Seattle Marrow Transplant Team. Antileukemic effect phase. Blood 2003;101:441-5.
of chronic graft-versus-host disease. Contribution to improved 22. Giralt S, Thall PF, Khouri I, Wang X, Braunschweig I, Ippolitti
survival after allogeneic marrow transplantation. N Engl J Med C, et al. Melphalan and purine analog-containing preparative
1981;304:1529-33. regimens: reduced-intensity conditioning for patients with
4. Horowitz MM, Gale RP, Sondel PM, Goldman JM, Kersey J, hematologic malignancies undergoing allogeneic progenitor
Kolb H-J, et al. Graft-versus-leukemia reactions after bone cell transplantation. Blood 2001;97:631-7.
marrow transplantation. Blood 1990;75:555-62. 23. Storb R, Yu C, Barnett T, Wagner JL, Deeg HJ, Nash RA, et al.
5. Kolb HJ, Mittermüller J, Clemm Ch, Holler G, Ledderose G, Stable mixed hematopoietic chimerism in dog leukocyte anti-
Brehm G, et al. Donor leukocyte transfusions for treatment of gen-identical littermate dogs given lymph node irradiation
recurrent chronic myelogenous leukemia in marrow trans- before and pharmacologic immunosuppression after marrow
plant patients. Blood 1990;76:2462-5. transplantation. Blood 1999;94:1131-6.
6. Slavin S, Naparstek E, Nagler A, Ackerstein A, Kapelushnik J, 24. Baron F, Maris MB, Sandmaier BM, Storer BE, Sorror M,
Or R. Allogeneic cell therapy for relapsed leukemia after bone Diaconescu R, et al. Graft-versus-tumor effects after allogene-
marrow transplantation with donor peripheral blood lympho- ic hematopoietic cell transplantation with nonmyeloablative
cytes. Exp Hematol 1995;23:1553-62. conditioning. J Clin Oncol 2005;23:1993-2003.
7. Kolb HJ. Graft-versus-leukemia effects of transplantation and 25. Lowsky R, Takahashi T, Liu YP, Dejbakhsh-Jones S, Grumet
donor lymphocytes. Blood 2008;112:4371-83. FC, Shizuru JA, et al. Protective conditioning for acute graft-
8. Lokhorst HM, Schattenberg A, Cornelissen JJ, Thomas LLM, versus-host disease. N Engl J Med 2005;353:1321-31.
Verdonck LF. Donor leukocyte infusions are effective in 26. Morris E, Thomson K, Craddock C, Mahendra P, Milligan D,
relapsed multiple myeloma after allogeneic bone marrow Cook G, et al. Outcomes after alemtuzumab-containing
transplantation. Blood 1997;90:4206-11. reduced-intensity allogeneic transplantation regimen for
9. Khouri IF, Lee MS, Saliba RM, Andersson B, Anderlini P, relapsed and refractory non-Hodgkin lymphoma. Blood 2004;
Couriel D, et al. Nonablative allogeneic stem cell transplanta- 104:3865-71.
tion for chronic lymphocytic leukemia: impact of rituximab on 27. Girgis M, Hallemeier C, Blum W, Brown R, Lin H-S, Khoury
immunomodulation and survival. Exp Hematol 2004;32:28-35. H, et al. Chimerism and clinical outcomes of 110 unrelated
10. Bloor AJ, Thomson K, Chowdhry N, Verfuerth S, Ings SJ, Cha- donor bone marrow transplants who underwent conditioning
kraverty R, et al. High response rate to donor lymphocyte with low-dose, single-exposure total body irradiation and
infusion after allogeneic stem cell transplantation for indolent cyclophosphamide. Blood 2005;105:3035-41.
non-Hodgkin lymphoma. Biol Blood Marrow Transplant 28. Schetelig J, Thiede C, Bornhauser M, Schwerdtfeger R, Kiehl
2008; 14:50-8. M, Beyer J, et al. Evidence of a graft-versus-leukemia effect in
11. Robinson SP, Sureda A, Canals C, Russell N, Caballero D, Baci- chronic lymphocytic leukemia after reduced-intensity condi-
galupo A, et al. Reduced intensity conditioning allogeneic tioning and allogeneic stem-cell transplantation: the
stem cell transplantation for Hodgkin's lymphoma: identifica- Cooperative German Transplant Study Group. J Clin Oncol
tion of prognostic factors predicting outcome. Haematologica 2003;21:2747-53.
2009;94:230-8. 29. Corradini P, Dodero A, Zallio F, Caracciolo D, Casini M,
12. Storb R, Yu C, Wagner JL, Deeg HJ, Nash RA, Kiem H-P, et al. Bregni M, et al. Graft-versus-lymphoma effect in relapsed
Stable mixed hematopoietic chimerism in DLA-identical litter- peripheral T-cell non-Hodgkin's lymphomas after reduced-
mate dogs given sublethal total body irradiation before and intensity conditioning followed by allogeneic transplantation
pharmacological immunosuppression after marrow transplan- of hematopoietic cells. J Clin Oncol 2004;22:2172-6.
tation. Blood 1997;89:3048-54. 30. Sorror ML, Maris MB, Storer B, Sandmaier BM, Diaconescu R,
13. Giralt S, Estey E, Albitar M, van Besien K, Rondón G, Flowers C, et al. Comparing morbidity and mortality of HLA-
Anderlini P, et al. Engraftment of allogeneic hematopoietic matched unrelated donor hematopoietic cell transplantation
progenitor cells with purine analog-containing chemotherapy: after nonmyeloablative and myeloablative conditioning: influ-
harnessing graft-versus-leukemia without myeloablative ther- ence of pretransplant comorbidities. Blood 2004;104:961-8.
apy. Blood 1997;89:4531-6. 31. Sandmaier BM, Storb R. Nonmyeloablative therapy and
14. Slavin S, Nagler A, Naparstek E, Kapelushnik Y, Aker M, hematopoietic cell transplantation for hematologic disorders.
Cividalli G, et al. Nonmyeloablative stem cell transplantation In: Blume KG, Forman SJ, Appelbaum FR, editors. Thomas’
and cell therapy as an alternative to conventional bone mar- Hematopoietic Cell Transplantation. 3rd edition. Oxford (UK):
row transplantation with lethal cytoreduction for the treat- Blackwell Publishing Ltd; 2004. p. 1164-76.
ment of malignant and nonmalignant hematologic diseases. 32. Michallet M, Le QH, Mohty M, Prebet T, Nicolini F, Boiron
Blood 1998;91:756-63. JM, et al. Predictive factors for outcomes after reduced inten-
15. Kottaridis PD, Milligan DW, Chopra R, Chakraverty RK, sity conditioning hematopoietic stem cell transplantation for
Chakrabarti S, et al. In vivo CAMPATH-1H prevents graft-ver- hematological malignancies: a 10-year retrospective analysis
sus-host disease following nonmyeloablative stem cell trans- from the Societe Francaise de Greffe de Moelle et de Therapie
plantation. Blood 2000;96:2419-25. Cellulaire. Exp Hematol 2008;36:535-44.
16. Corradini P, Tarella C, Olivieri A, Gianni AM, Voena C, Zallio 33. Baron F, Sandmaier BM. Chimerism and outcomes after allo-
F, et al. Reduced-intensity conditioning followed by allograft- geneic hematopoietic cell transplantation following nonmye-
ing of hematopoietic cells can produce clinical and molecular loablative conditioning. Leukemia 2006;20:1690-700.
remissions in patients with poor-risk hematologic malignan- 34. de Lima M, Anagnostopoulos A, Munsell M, Shahjahan M,
cies. Blood 2002;99:75-82. Ueno N, Ippoliti C, et al. Nonablative versus reduced-intensi-
17. Khouri IF, Keating M, Körbling M, Przepiorka D, Anderlini P, ty conditioning regimens in the treatment of acute myeloid
O’Brien S, et al. Transplant-lite: induction of graft-versus- leukemia and high-risk myelodysplastic syndrome: dose is rel-
malignancy using fludarabine-based nonablative chemothera- evant for long-term disease control after allogeneic
py and allogeneic blood progenitor-cell transplantation as hematopoietic stem cell transplantation. Blood 2004; 104:865-
treatment for lymphoid malignancies. J Clin Oncol 1998; 16: 72.
2817-24. 35. Carvallo C, Geller N, Kurlander R, Srinivasan R, Mena O,
18. McSweeney PA, Niederwieser D, Shizuru JA, Sandmaier BM, Igarashi T, et al. Prior chemotherapy and allograft CD34+ dose
Molina AJ, Maloney DG, et al. Hematopoietic cell transplanta- impact donor engraftment following nonmyeloablative allo-
tion in older patients with hematologic malignancies: replac- geneic stem cell transplantation in patients with solid tumors.
ing high-dose cytotoxic therapy with graft-versus-tumor Blood 2004;103:1560-3.
effects. Blood 2001;97:3390-400. 36. Baron F, Baker JE, Storb R, Gooley TA, Sandmaier BM, Maris
19. Childs R, Clave E, Contentin N, Jayasekera D, Hensel N, MB, et al. Kinetics of engraftment in patients with hematolog-
Leitman S, et al. Engraftment kinetics after nonmyeloablative ic malignancies given allogeneic hematopoietic cell transplan-
allogeneic peripheral blood stem cell transplantation: full tation after nonmyeloablative conditioning. Blood 2004; 104:

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 247 |
14th Congress of the European Hematology Association

2254-62. Maloney DG, et al. Hematopoietic cell transplantation-specif-


37. Baron F, Storb R. Allogeneic hematopoietic cell transplantation ic comorbidity index as an outcome predictor for patients
as treatment for hematological malignancies: a review. Semin with acute myeloid leukemia in first remission: Combined
Immunopathol 2004:26:71-94. FHCRC and MDACC experiences. Blood 2007;110:4608-13.
38. Baron F, Maris MB, Storer BE, Sandmaier BM, Panse JP, 55. Mielcarek M, Martin PJ, Leisenring W, Flowers MED, Maloney
Chauncey TR, et al. High doses of transplanted CD34+ cells DG, Sandmaier BM, et al. Graft-versus-host disease after non-
are associated with rapid T-cell engraftment and lessened risk myeloablative versus conventional hematopoietic stem cell
of graft rejection, but not more graft-versus-host disease after transplantation. Blood 2003;102:756-62.
nonmyeloablative conditioning and unrelated hematopoietic 56. Couriel DR, Saliba RM, Giralt S, Khouri I, Andersson B, de
cell transplantation. Leukemia 2005;19:822-8. Lima M, et al. Acute and chronic graft-versus-host disease
39. Maris MB, Sandmaier BM, Storer BE, Maloney DG, Shizuru after ablative and nonmyeloablative conditioning for allogene-
JA, Agura E, et al. Unrelated donor granulocyte colony-stimu- ic hematopoietic transplantation. Biol Blood Marrow
lating factor-mobilized peripheral blood mononuclear cell Transplant 2004;10:178-85.
transplantation after nonmyeloablative conditioning: the 57. Mohty M, Bay JO, Faucher C, Choufi B, Bilger K, Tournilhac
effect of postgrafting mycophenolate mofetil dosing. Biol O, et al. Graft-versus-host disease following allogeneic trans-
Blood Marrow Transplant 2006;12:454-65. plantation from HLA-identical sibling with antithymocyte
40. Mohty M, Avinens O, Faucher C, Viens P, Blaise D, Eliaou JF. globulin-based reduced-intensity preparative regimen. Blood
Predictive factors and impact of full donor T-cell chimerism 2003;102:470-6.
after reduced intensity conditioning allogeneic stem cell trans- 58. Chakrabarti S, Mackinnon S, Chopra R, Kottaridis PD, Peggs
plantation. Haematologica 2007;92:1004-6. K, O’Gorman P, et al. High incidence of cytomegalovirus infec-
41. Baron F, Petersdorf E, Gooley T, Sandmaier B, Malkki M, tion after nonmyeloablative stem cell transplantation: poten-
Chaunce T, et al. What is the role for donor NK cells after non- tial role of Campath-1H in delaying immune reconstitution.
myeloablative conditioning? Biol Blood Marrow Transplant Blood 2002;99:4357-63.
2009;15:580-8. 59. Crawley C, Lalancette M, Szydlo R, Gilleece M, Peggs K,
42. Weissinger F, Sandmaier BM, Maloney DG, Bensinger WI, Mackinnon S, et al. Outcomes for reduced-intensity allogene-
Gooley T, Storb R. Decreased transfusion requirements for ic transplantation for multiple myeloma: an analysis of prog-
patients receiving nonmyeloablative compared with conven- nostic factors from the Chronic Leukemia Working Party of
tional peripheral blood stem cell transplants from HLA-identi- the EBMT. Blood 2005;105:4532-9.
cal siblings. Blood 2001;98:3584-8. 60. Lan F, Zeng D, Higuchi M, Huie P, Higgins JP, Strober S.
43. Baron F, Vanstraelen G, Beguin Y. Transfusions after nonmye- Predominance of NK1.1+TCR αβ+ or DX5+TCR αβ+ T cells
loablative or reduced-intensity conditioning regimens. in mice conditioned with fractionated lymphoid irradiation
Leukemia 2006;20:2081-6. protects against graft-versus-host disease: “natural suppressor”
44. Hogan WJ, Maris M, Storer B, Sandmaier BM, Maloney DG, cells. J Immunol 2001;167:2087-96.
Schoch HG, et al. Hepatic injury after nonmyeloablative con- 61. Lowsky R, Stockerl-Goldstein K, Laport G. Clinical outcomes
ditioning followed by allogeneic hematopoietic cell transplan- following allogeneic hematopoietic cell transplantation (HCT)
tation: a study of 193 patients. Blood 2004;103:78-84. using nonmyeloablative host conditioning with total lym-
45. Parikh CR, Schrier RW, Storer B, Diaconescu R, Sorror ML, phoid irradiation and anti-thymocyte globulin confirm a low
Maris MB, et al. Comparison of ARF after myeloablative and incidence of graft versus host disease (GVHD) and retained
nonmyeloablative hematopoietic cell transplantation. Am J graft anti-tumor activity. Blood 2006;108:182a[Abstract].
Kidney Dis 2005;45:502-9. 62. Collins RH, Shpilberg O, Drobyski WR, Porter DL, Giralt S,
46. Fukuda T, Hackman RC, Guthrie KA, Sandmaier BM, Boeckh Champlin R, et al. Donor leukocyte infusions in 140 patients
M, Maris MB, et al. Risks and outcomes of idiopathic pneumo- with relapsed malignancy after allogeneic bone marrow trans-
nia syndrome after nonmyeloablative and conventional condi- plantation. J Clin Oncol 1997;15:433-44.
tioning regimens for allogeneic hematopoietic stem cell trans- 63. Kolb HJ, Schmidt C, Barrett AJ, Schendel DJ. Graft-versus-
plantation. Blood 2003;102:2777-85. leukemia reactions in allogeneic chimeras. Blood 2004; 103:
47. Chien JW, Maris MB, Sandmaier BM, Maloney DG, Storb RF, 767-76.
Clark JG. Comparison of lung function after myeloablative 64. Blaise DP, Boiron JM, Faucher C, Mohty M, Bay JO, Bardoux
and 2 Gy of total body irradiation-based regimens for VJ, et al. Reduced intensity conditioning prior to allogeneic
hematopoietic stem cell transplantation. Biol Blood Marrow stem cell transplantation for patients with acute myeloblastic
Transplant 2005;11:288-96. leukemia as a first-line treatment. Cancer 104:1931-8.
48. Aoudjhane M, Labopin M, Gorin NC, Shimoni A, Ruutu T, 65. Valcarcel D, Martino R, Caballero D, Martin J, Ferra C, Nieto
Kolb H-J, et al. Comparative outcome of reduced intensity and JB, et al. Sustained remissions of high-risk acute myeloid
myeloablative conditioning regimen in HLA identical sibling leukemia and myelodysplastic syndrome after reduced-inten-
allogeneic haematopoietic stem cell transplantation for sity conditioning allogeneic hematopoietic transplantation:
patients older than 50 years of age with acute myeloblastic chronic graft-versus-host disease is the strongest factor
leukaemia: a retrospective survey from the Acute Leukemia improving survival. J Clin Oncol 2008;26:577-84.
Working Party (ALWP) of the European group for Blood and 66. Kennedy GA, Durrant S, Butler J, Morton J, Western R, Bartlett
Marrow Transplantation (EBMT). Leukemia 2005;19:2304-12. ML, et al. Outcome of myeloablative allogeneic stem cell
49. Martino R, Iacobelli S, Brand R, Jansen T, van Biezen A, Finke transplantation in multiple myeloma with a 153Sm-EDTMP-
J, et al. Retrospective comparison of reduced-intensity condi- based preparative regimen. Leukemia 2005;19:879-80.
tioning and conventional high-dose conditioning for allogene- 67. Bruno B, Rotta M, Patriarca F, Mattei D, Allione B, Carnevale-
ic hematopoietic stem cell transplantation using HLA-identical Schianca F, et al. Non-myeloablative allografting for newly
sibling donors in myelodysplastic syndromes. Blood 2006; diagnosed multiple myeloma: the experience of the Gruppo
108:836-46. Italiano Trapianti di Midollo. Blood 2009;113:3375-82.
50. Dreger P, Brand R, Milligan D, Corradini P, Finke J, Lam- 68. Rotta M, Storer BE, Sahebi F, Shizuru JA, Bruno B, Lange T, et
bertenghi DG, et al. Reduced-intensity conditioning lowers al. Long-term outcome of patients with multiple myeloma
treatment-related mortality of allogeneic stem cell transplanta- after autologous hematopoietic cell transplantation and non-
tion for chronic lymphocytic leukemia: a population-matched myeloablative allografting. Blood 2009;113:3383-91.
analysis. Leukemia 2005;9:1029-33. 69. Kahl C, Storer BE, Sandmaier BM, Mielcarek M, Maris MB,
51. Scott BL, Sandmaier BM, Storer B, Maris MB, Sorror ML, Blume KG, et al. Relapse risk among patients with malignant
Maloney DG, et al. Myeloablative vs nonmyeloablative allo- diseases given allogeneic hematopoietic cell transplantation
geneic transplantation for patients with myelodysplastic syn- after nonmyeloablative conditioning. Blood 2007;110:2744-8.
drome or acute myelogenous leukemia with multilineage dys- 70. Brunstein CG, Barker JN, Weisdorf DJ, Defor TE, Miller JS,
plasia: a retrospective analysis. Leukemia 2006;20:128-35. Blazar BR, et al. Umbilical cord blood transplantation after
52. Sorror ML, Storer BE, Maloney DG, Sandmaier BM, Martin PJ, nonmyeloablative conditioning: impact on transplantation
Storb R. Outcomes after allogeneic hematopoietic cell trans- outcomes in 110 adults with hematologic disease. Blood 2007;
plantation with nonmyeloablative or myeloablative regimens 110:3064-70.
for treatment of lymphoma and chronic lymphocytic 71. O’Donnell PV, Luznik L, Jones RJ, Vogelsang GB, Leffell MS,
leukemia. Blood 2008;111:446-52. Phelps M, et al. Nonmyeloablative bone marrow transplanta-
53. Sorror ML, Maris MB, Storb R, Baron F, Sandmaier BM, tion from partially HLA-mismatched related donors using
Maloney DG, Storer B. Hematopoietic cell transplantation posttransplantation cyclophosphamide. Biol Blood Marrow
(HCT)-specific comorbidity index: a new tool for risk assess- Transplant 2002;8:377-86.
ment before allogeneic HCT. Blood 2005;106:2912-9. 72. Baron F, Storb R, Storer BE, Maris MB, Niederwieser D,
54. Sorror ML, Giralt S, Sandmaier BM, de Lima M, Shahjahan M, Shizuru JA, et al. Factors associated with outcomes in allo-

| 248 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

geneic hematopoietic cell transplantation with nonmyeloabla- tion and peripheral blood stem cell infusion for elderly
tive conditioning after failed myeloablative hematopietic cell patients with advanced acute myeloid leukemia (AML) or
transplantation. J Clin Oncol 2006;24:4150-7. high-risk myelodysplastic syndrome (MDS). Blood 2005; 106:
73. Storb R, Yu C, Zaucha JM, Deeg HJ, Georges G, Kiem H-P, et 119a[Abstract].
al. Stable mixed hematopoietic chimerism in dogs given donor 79. Kroger N, Shimoni A, Zagrivnaja M, Ayuk F, Lioznov M,
antigen, CTLA4Ig, and 100 cGy total body irradiation before Schieder H, et al. Low-dose thalidomide and donor lympho-
and pharmacologic immunosuppression after marrow trans- cyte infusion as adoptive immunotherapy after allogeneic
plant. Blood 1999;94:2523-9. stem cell transplantation in patients with multiple myeloma.
74. Jochum C, Beste M, Zellmer E, Graves SS, Storb R. CD154 Blood 2004;104:3361-3.
blockade and donor-specific transfusions in DLA-identical 80. Gopal AK, Pagel JM, Rajendran JG, Maloney DG, Appelbaum
marrow transplantation in dogs conditioned with 1-Gy total FR, Sorror ML, et al. Improving the efficacy of reduced inten-
body irradiation. Biol Blood Marrow Transplant 2007;13:164- sity allogeneic transplantation for lymphoma using radiother-
71. apy. Biol Blood Marrow Transplant 2006;12:697-702.
75. Bethge WA, Wilbur DS, Storb R, Hamlin DK, Santos EB, 81. van de Donk NW, Kröger N, Hegenbart U, Corradini P, Miguel
Brechbiel MW, et al. Selective T-cell ablation with bismuth-
213-labeled anti-TCRαβ as nonmyeloablative conditioning for JF, Goldschmidt H, et al. Remarkable activity of novel agents
allogeneic canine marrow transplantation. Blood 2003; 101: bortezomib and thalidomide in patients not responding to
5068-75. donor lymphocyte infusions following nonmyeloablative allo-
76. Maloney DG, Molina AJ, Sahebi F, Stockerl-Goldstein KE, geneic stem cell transplantation in multiple myeloma. Blood
Sandmaier BM, Bensinger W, et al. Allografting with nonmye- 2006;107:3415-6.
loablative conditioning following cytoreductive autografts for 82. Kamei M, Nannya Y, Torikai H, Kawase T, Taura K, Inamoto
the treatment of patients with multiple myeloma. Blood. Y, et al. HapMap scanning of novel human minor histocom-
2003;102:3447-54. patibility antigens. Blood 2008 Sept 22. [Epub ahead of print as
77. Pagel JM, Hedin N, Subbiah K, Meyer D, Mallet R, Axworthy doi: 10.1182/blood-2008-07-171678]
D, et al. Comparison of anti-CD20 and anti-CD45 antibodies 83. Sandmaier BM, Storb R. Reduced-intensity conditioning fol-
for conventional and pretargeted radioimmunotherapy of B- lowed by hematopoietic cell transplantation for hematologic
cell lymphomas. Blood 2003;101:2340-8. malignancies. In: Appelbaum FR, Forman SJ, Negrin RS, Blume
78. Pagel JM, Appelbaum FR, Sandmaier BM, et al. 131I-anti- KG, editors. Thomas’ Hematopoietic Cell Transplantation. 4th
CD45 antibody plus fludarabine, low-dose total body irradia- ed. Oxford (UK): Wiley-Blackwell; 2009. p. 1043-58.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 249 |
Stem cell transplantation

Reduced intensity conditioning as a platform


for immunotherapy

J. Barrett A B S T R A C T

Reduced intensity conditioning (RIC) regimens offer the patient a stem cell transplant (SCT) with
Allogeneic Stem Cell Transplantation low regimen-related toxicity but preserved graft-versus-leukemia (GVL) effects. In the last decade, a
Section, Hematology Branch, NHLBI,
NIH, Bethesda, MD, USA wide variety of regimens has been developed and it is now possible to refine protocol design to achieve
desired properties of suppression and intensity. RIC regimens have some unique immunological fea-
tures, which can favor GVL while controlling graft-versus-host disease (GVHD). Current regimens and
Hematology Education: strategies provide GVL and graft-versus-tumor (GVT) effects whose efficacy varies according to dis-
the education program for the ease, with chronic leukemia and lymphomas being the most susceptible to immune control. While
annual congress of the European much has been learned through RIC SCT about ways to manipulate immune recovery and GVL/GVT
Hematology Association effects, both acute and chronic GVHD remain an issue requiring better ways of separating GVHD from
antimalignant effects. Here, concepts of RIC transplantation in relation to their immunotherapeutic
2009;3:250-255 potential are discussed, and disease-specific RIC transplant approaches to maximize malignant dis-
ease control and cure are described.

Introduction malignant disease states and strategies to


Over 10 years ago, encouraged by grow- further refine and augment the GVL effect of
ing evidence that the graft-versus-leukemia RIC regimens.
(GVL) effect from donor lymphocyte infu-
sions (DLI) could be harnessed as a potent Immune reconstitution after reduced intensity
immunological weapon, investigators began conditioning regimens
to develop conditioning regimens for Immediately after allogeneic SCT, host
leukemia stem cell transplantation (SCT), antigen presenting cells (APC) induce allore-
which achieved lymphocyte engraftment sponses in the incoming donor lymphocytes,
and the GVL effect without using myeloab- initiating in graft-versus-host disease
lation to minimize residual malignancy.1 (GVHD) and GVL reactions. The cytokine
This conceptual change in perception of milieu (the so-called cytokine storm induced
how SCT cures malignancy is illustrated in by the conditioning regimen and a surge of
Figure 1. Reduced intensity conditioning lymphocyte growth factors, notably IL-15
(RIC) regimens (whether or not myeloabla- induced by lymphopenia)3 tends to boost the
tion is achieved) have extended the use of alloresponse. Subsequently, alloresponding
SCT to older and debilitated patients with and engrafting donor lymphocytes displace
hematological malignancies, who cannot any residual host immune cells and marrow
withstand classical myeloablative regimens, cells.4 Lineage-specific chimerism studies can
and to patients with solid tumors, where be used to track separate recovery of donor
intensive myeloablative treatment is ineffec- lymphopoiesis and myelopoiesis and loss of
tive and only adds unwanted toxicity.2 host cells.5 In children who have a function-
There are now over 40 published RIC reg- ing thymus before SCT, recovery of the thy-
imens varying in the relative degree of mus after transplant speeds the acquisiton of
immunosuppression or myelosuppression full T cell competence and favors donor-host
they achieve.1 They fall into two broad cate- tolerance. Acute and chronic GVHD and the
gories: non-myeloablative (e.g., fludarabine immunosuppressive agents used to prevent
and cyclophosphamide) and partially or or treat the disease are the major limits to
completely myeloablative (e.g., fludarabine complete and rapid immune reconstitution.4
or other purine analogs with melphalan, RIC transplants have particular impact on
busulfan or total body irradiation and non- immune recovery, which can be manipulat-
purine-analog regimens). In each of these ed to optimize GVL effects and transplant
categories, additional treatments are some- outcome. It was thought that the lower
times included, such as antilymphocyte anti- intensity conditioning might protect patients
bodies. This diversity reflects the ability to from GVHD because it was argued that the
select separately the degree of immunosup- cytokine storm (from regimen-induced tis-
pression and myelosuppression desired for sue damage) would be less. In fact, acute
different transplant situations. Here, we dis- GVHD is not mitigated by the regimen, and
cuss the nature of the immune recovery that risk factors for acute GVHD appear to be
occurs after RIC regimens, the approaches comparable after standard and RIC trans-
available to optimize GVL effects in specific plants.6 One factor, which may increase the

| 250 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

risk and severity of acute and chronic GVHD after RIC Myelofibrosis
transplantation, is the less complete eradication of host This myeloproliferative disorder, common in older
APC by non-myeloablative regimens.7 While RIC regi- individuals, appears to be especially susceptible to erad-
mens may favor thymic recovery, T cell repertoire nor- ication by RIC regimens. Not only is the recipient mar-
malization and tolerance,7,8 it could be argued that the row rapidly replaced by engrafting donor cells but the
establishment of tolerance may, in fact, be unfavorable fibrosis resolves, making it unnecessary to perform
for GVL effects, which require intolerant T cells to attack splenectomy before SCT.21,22
leukemia cells.4 Chimerism has been extensively stud-
ied in RIC transplants. The patterns of T cell and
myeloid cell recovery are distinct and regimen-specific.
Pure immunoablative regimens result in an early switch
to 100% donor T cells, while hematopoietic recovery is
100% host, switching within a few months to 100%
donor myelopoiesis as the incoming donor immune
function eliminates host marrow through a graft-vs-
marrow mechanism.2,5 The switch to donor marrow
often heralds GVL and graft-versus-tumor (GVT)
effects.2 RIC regimens with more myelosuppressive
intensity achieve earlier and more complete donor
myeloid chimerism. Also, fully myeloablative RIC regi-
mens show 100% donor myeloid reconstitution imme-
diately on engraftment.9 Similarly, by varying the
immunosuppressive intensity of the conditioning the
degree of donor-host T cell, chimerism can be modulat-
ed. Figure 2 segregates conditioning regimens with dif-
ferent immunosuppressive and myeloablative capacity.

Reduced intensity conditioning regimens in specific malig- Figure 1. Classical and “reduced intensity” concepts of the
nancies relationship between conditioning regimens, engraftment
and GVL effects. (A) Classical conditioning (e.g.,
Chronic myelocytic leukemia Cyclophosphamide and TBI 12Gy) results in rapid and
The exquisite sensitivity of chronic myelocytic complete engraftment of marrow and T cells (slower if the
leukemia (CML) relapsing after SCT to achieving a sec- graft is T cell depleted). Conditioning eliminates most of
the disease burden while the GVL effect is perceived only
ond cure following DLI has led several investigators to to eliminate minimal residual disease. (B) In the reduced
use RIC SCT for this leukemia.10-13 Disappointingly, intensity conditioning, a larger burden of leukemia
truly myeloablative regimens have not been found to remains and is only eradicated after DLI which converts
mixed T cell and myeloid chimerism to full donor engraft-
reliably eradicate chronic phase CML, although regi- ment. The neutrophil and platelet nadir after conditioning
mens, including reduced doses of busulfan confer GVL may not be profound (depending on myeloablative intensi-
effects without increased toxicity.10 The arrival of tyro- ty of the regimen) and the GVL effect is perceived to be a
significant component of disease eradication.
sine kinase inhibitors to treat CML has restricted SCT to
advanced CML where again RIC regimens appear
unsuitable.14 The attractive idea of combining targeted
treatment with imatinib with RIC SCT to eradicate
residual disease has not been formally explored.
However, the observation that combined treatment
with DLI and imatinib to treat relapsed CML is better
than either agent used alone would support the idea of
a useful synergy using both modalities.14,15

Acute myeloblastic leukemia and myelodysplastic syndrome


While there is universal agreement that RIC regimens
have made it possible to safely transplant Acute
myeloblastic leukemia (AML) and myelodysplastic syn-
drome (MDS) patients in their 60s, it is now clear that
lower transplant-related mortality (TRM) from reduced
intensity is offset by increased relapse,16 rendering RIC
approaches unsuccessful in advanced myeloid malig-
nancies.17 Strategies to reduce relapse in high risk AML
and MDS patients include pretreatment with chemo- Figure 2. Relationship between regimen intensity,
therapy (e.g., the fludarabine/amsacrine based FLAMSA immunosuppression and myelosuppression. Single agents
and low dose TBI are inadequate to ensure engraftment,
regimen) to reduce tumor burden,18 targeted therapy while immunoablative regimens ensure engraftment with-
with the anti CD33 immunotoxin gemtuzemab out myeloablation. Increasing intensity achieves increas-
ozogamicin prior to SCT,19 or radiolabled anti CD45 ing myelosuppression, such that some RIC regimens are
fully myeloablative without the extra toxicity of classical
given during conditioning.20 “full dose” regimens.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 251 |
14th Congress of the European Hematology Association

Chronic lymphocytic leukemia Not all reduced intensity conditioning regimens are safer
This chronic leukemia is very susceptible to the GVL There is a limit to the amount of dose reduction in
effect. Even advanced and high-risk forms of CLL with RIC regimens. For example, early regimens using only
bulky disease at transplant can show durable responses 200 cGy total body irradiation were associated with
to SCT. RIC SCT has been widely used to treat this graft rejection. When fludarabine was combined with
older population of patients.23,24 It is concluded that out- low dose TBI, engraftment was improved.42 The safety
come is favored because of a lower treatment-related of RIC regimens hinges as much on the prophylactic
mortality (TRM) while the GVL effect can control dis- regimen for GVHD as the regimen-related toxicity, and
ease. However, mixed chimerism delays disease some RIC protocols have a higher risk of severe acute,
response and may predispose to relapse, which reached as well as chronic GVHD. In addressing safety of RIC
31% in a large European series.25 SCT, attention must be focused on the entire procedure
to include adequate GVHD prevention. In this regard,
Non-Hodgkin’s lymphoma correctly dosed alemtuzemab appears to offer a better
RIC SCT has been extensively explored in both low alternative to standard methotrexate/cyclosporine
grade (follicular) and high grade NHL.26,27 Low grade GVHD prophylaxis.43
NHL appears to be uniquely susceptible to GVL effects
after RIC transplants. Even patients with high-grade Myeloablation does not correlate with increased toxicity
NHL who have relapsed after autologous SCT can Although non-myeloablative SCT may be sufficient
achieve prolonged remissions and possible cure.28,29 to achieve cures in some chronic leukemias and low
grade lymphomas, abundant data shows that in AML
Hodgkin’s disease and MDS, the RIC regimens reduce TRM only at the
RIC regimens are often the only practical approach to price of increased relapse. While DFS may still be favor-
SCT in patients with HD where allogeneic SCT is used able because of the low TRM compared with higher
in high risk patients with multiply relapsed and multi- intensity regimens, it has to be concluded that regimen
ply treated disease. There is clear evidence for a GVL intensity does contribute to leukemia control. The com-
effect in HD but lack of prospective comparative stud- bination of fludarabine and busulfan is being increasing-
ies makes it difficult to determine the regimen intensity ly used in RIC regimens. This regimen is myeloablative
that confers the optimum balance between disease con- but well tolerated at busulfan doses about 50% of stan-
trol and toxicity.30 dard regimens.44,45 Studies where the dose of busulfan is
cautiously advanced further are in order in high risk
Multiple myeloma patients receiving RIC SCT to find the optimum balance
There is a modest GV-myeloma effect in MM. between GVL effect and regimen related toxicity.
Although progression free survivals are often compara-
ble, relapse and progression is greater after RIC regi- GVHD remains a major problem
mens than after full intensity regimens.31,32 The most Despite earlier assumptions to the contrary, it is now
promising strategy to improve outcome has been to agreed that RIC SCT carry at least as high as risk of
uncouple the tumor reduction from the GV-myeloma acute and chronic GVHD as standard SCT.6 Therefore,
effect by prior intensive therapy and autologous SCT the GVL effect is compromised by the need to give
followed by a RIC SCT.33 GVHD prophylaxis. The negative impact of GVHD pro-
phylaxis on relapse is clear from two trials in myeloab-
Solid tumors lative SCT, which demonstrated a significant reduction
Complete remissions in metastatic renal cell cancer in relapse in patients receiving low dose CSA compared
following fludarabine/cyclophosphamide conditioning with those who received standard dose CSA.46,47 The
and allo SCT34,35 paved the way to numerous studies of humanized monoclonal antibody anti CD52 (alem-
RIC SCT in breast,36 colon,37 ovarian,38 pancreatic,39 tuzemab) has been used to deplete T cells in the donor
hepatic40 cancers and sarcomas.41 It is generally agreed and the recipient. Studies have shown that alemtuzum-
that there is a GVT effect in some of these advanced ab decreases acute and chronic GVHD, while conserv-
malignancies. Notably relationships between GVHD ing GVL/GVT. However, the beneficial effect is sched-
and GVT effects and regression of metastases after stop- ule and dose sensitive with best results seen in adminis-
ping immunosuppression in the context of conditioning tering the antibody early in conditioning at lower
regimens that are unlikely to cause tumor shrinkage is doses.48 Other promising GVHD prophylactic regimens
strong evidence for GVT effects in many cancers.2 are the combination of tacrolimus, sirolimus and low
Unfortunately, only a minority of patients are respon- dose methotrexate,49 and cyclosporine combined with
ders and durable complete remissions appear to be con- mycophenolate mofetil.50
fined to patients with renal cancer.
Mixed chimerism has mixed benefits
What we have learned about immune function in reduced Some investigators have used regimens, which initial-
intensity conditioning stem cell transplantation? ly caused a mixed chimeric T cell state (carrying a low
These results from a wide diversity of studies over 12 risk of GVHD development), followed by DLI to con-
years (unfortunately mostly non-randomized and some vert to full donor T cell chimerism a few months later.51
conflicting)1 allow us to draw some general conclusions While the DLI can deliver powerful antitumor effects,
about the factors regulating immune function, GVHD the major problem with the DLI given in the first few
and GVL after RIC SCT, and point the way towards months after SCT is the rapid development of acute and
optimizing disease-specific RIC transplant regimens. chronic GVHD. Thus, at worst both GVHD and GVL

| 252 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

are simply delayed. The persistence of mixed phomas, they may also replace full intensity SCT in
chimerism carries a worse prognosis in some studies.52 younger patients. They are being further explored to
Individual donor-host variations make it impossible to reduce toxicity in haploidentical SCT.63 Their persisting
design a regimen that reliably achieves mixed limitation is one common to all allogeneic SCT
chimerism and DLI does not always result in full donor approaches, namely the separation of GVHD and GVL
T cell reconstitution because a full lymphocyte compart- effects.4 New approaches, such as selective T cell deple-
ment (with reflexly low levels of lymphocyte growth tion have already been explored in the context of RIC
factors) is not favorable to further engraftment and SCT in older patients to reduce the risk from GVHD in
expansion of infused T cells.53 Reversal of mixed this high-risk group.64 Further boosting of GVL/GVT
chimerism can be achieved more reliably by treating effects could be achieved using vaccines or adoptive
with fludarabine to create lymphopenia prior to DLI.54 transfer of NK cells or tumor specific T cells.4
However, mixed chimerism is inherently unstable, car-
rying the risk of graft rejection when donor T cells are Conclusions
in the minority and acute GVHD when full chimerism Since their introduction more than a decade ago, RIC
is achieved. regimens have come to be applied to a wide group of
malignant and non-malignant disorders. In the process,
Preemptive DLI is more effective at controlling disease post- we have developed a greater understanding of the rela-
SCT tive roles of conditioning agents in achieving immuno-
Although DLI is highly effective at treating relapse in ablation and myelosuppression and it is now possible to
chronic leukemias the control of relapsed acute refine protocol design to achieve desired properties of
leukemias and other diseases disappointing. Giving DLI suppression and intensity. Studies of immune reconsti-
preemptively is a logical means of improving the man- tution after RIC SCT has taught us the impact of donor
agement of persisting disease that has not yet pro- chimerism on outcome and improved the way to con-
gressed to hematological relapse55-57 but randomized trol immune recovery and deliver DLI effectively. Future
studies to validate this approach are lacking. regimens incorporating targeted agents and immuno-
therapy strategies, such as vaccines, adoptive cell trans-
Diseases vary in their susceptibility to GVT effects fer, selective T cell depletion and administration of
As described above GVT/GVL effects vary widely in cytokines should further improve outcome of this type
different disease states. Thus, RIC treatment strategies of transplant.
have to be tailored to specific disease states.

Control of disease benefits from pretransplant tumor control References


Carella was the first to propose separating a prelimi-
nary intensive therapy with autologous SCT to reduce 1. Barrett AJ, Savani BN. Stem cell transplantation with reduced-
intensity conditioning regimens: a review of ten years experi-
bulky disease followed after a recovery period by a RIC ence with new transplant concepts and new therapeutic
transplant to confer GVL.58 This approach appears agents. Leukemia 2006;20:1661-72.
promising for treatment of multiple myeloma, a disease, 2. Lundqvist A, Childs R. Allogeneic hematopoietic cell trans-
which historically had a high TRM after full intensity plantation as immunotherapy for solid tumors: current status
and future directions. J Immunother 2005;28:281-8.
SCT. But it is not yet clear whether this approach is 3. Boyiadzis M, Memon S, Carson J, Allen K, Szczepanski MJ,
superior to the tandem autologous SCT.59 A similar Vance BA, et al. Up-regulation of NK cell activating receptors
approach in older MDS patients appears promising. following allogeneic hematopoietic stem cell transplantation
under a lymphodepleting reduced intensity regimen is associ-
Kolb et al. have used a fludarabine/amsacrine regimen ated with elevated IL-15 levels. Biol Blood Marrow Transplant
(Flamsa) followed after a 5 day rest period by a condi- 2008;14:290-300.
tioning regimen of fludarabine and melphalan.18 4. Barrett AJ. Understanding and harnessing the graft-versus-
Although, this study was not randomized, the leukaemia effect. Br J Haematol 2008;142:877-88
5. Childs R, Clave E, Contentin N, Jayasekera D, Hensel N,
leukemia-free survival of 73% for this high risk patient Leitman S, et al. Engraftment kinetics after nonmyeloablative
group was extremely encouraging. allogeneic peripheral blood stem cell transplantation: full
Targeted treatments are promising partners for RIC donor T-cell chimerism precedes alloimmune responses. Blood
1999;94:3234-41.
regimens: in the last few years, non-chemotherapeutic 6. Remberger M, Mattsson J, Hassan Z, Karlsson N, LeBlanc K,
agents with low toxicity, which primarily target the Omazic B, et al. Risk factors for acute graft-versus-host disease
malignancy, have begun to be incorporated into RIC grades II-IV after reduced intensity conditioning allogeneic
stem cell transplantation with unrelated donors: a single cen-
SCT. In addition to incorporating them into the condi- tre study. Bone Marrow Transplant 2008;41:399-405.
tioning regimen agents, such as imatinib for CML60 and 7. Jiménez M, Martínez C, Ercilla G, Carreras E, Urbano-Ispízua
sirolumus which, as well as being a prophylactic agent A, Aymerich M, et al. Reduced-intensity conditioning regimen
for GVHD, has a suppressive effect on B cell lym- preserves thymic function in the early period after hematopoi-
etic stem cell transplantation. Exp Hematol 2005;33:1240-8.
phomas61 and bortezomib for MM,62 can be given post 8. Hentschke P, Omazic B, Mattsson J, Näsman-Björk I,
SCT to augment the effect of DLI. Lundkvist I, Gigliotti D, et al. T-cell receptor Vbeta repertoire
after myeloablative and reduced intensity conditioning allo-
geneic haematopoietic stem cell transplantation. Scand J
Future developments Immunol 2005;61:285-94.
RIC regimens have made SCT available to a potential- 9. Baron F, Little MT, Storb R. Kinetics of engraftment following
ly large cohort of older patients with hematological allogeneic hematopoietic cell transplantation with reduced-
intensity or nonmyeloablative conditioning. Blood Rev 2005;
malignancies and patients with solid tumors who do 19:153-64.
not respond to high dose myeloablative treatments. In 10. Or R, Shapira MY, Resnick I, Amar A, Ackerstein A, Samuel S,
susceptible diseases, such as selected low grade lym- et al. Nonmyeloablative allogeneic stem cell transplantation
for the treatment of chronic myeloid leukemia in first chronic

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 253 |
14th Congress of the European Hematology Association

phase. Blood 2003;101:441-5. loablative conditioning for aggressive lymphoma. Bone


11. Sloand E, Childs RW, Solomon S, Greene A, Young NS, Barrett Marrow Transplant 2008;42 Suppl 1:S35-S36
AJ. The graft-versus-leukemia effect of nonmyeloablative 27. Smith SM. Reduced-intensity transplantation for lymphoma.
stem cell allografts may not be sufficient to cure chronic myel- Curr Treat Options Oncol 2006;7:295-305
ogenous leukemia. Bone Marrow Transplant 2003;32:897-901 28. Sorror ML, Storer BE, Maloney DG, Sandmaier BM, Martin PJ,
12. Crawley C, Szydlo R, Lalancette M, Bacigalupo A, Lange A, Storb R. Outcomes after allogeneic hematopoietic cell trans-
Brune M, et al. Chronic Leukemia Working Party of the EBMT. plantation with nonmyeloablative or myeloablative condi-
Outcomes of reduced-intensity transplantation for chronic tioning regimens for treatment of lymphoma and chronic lym-
myeloid leukemia: an analysis of prognostic factors from the phocytic leukemia. Blood 2008;111:446-52.
Chronic Leukemia Working Party of the EBMT. Blood 2005; 29. Armand P, Kim HT, Ho VT, Cutler CS, Koreth J, Antin JH, et
106:2969-76. al. Allogeneic transplantation with reduced-intensity condi-
13. Faber E, Koza V, Vitek A, Mayer J, Sedlacek P, Zak P, et al. tioning for Hodgkin and non-Hodgkin lymphoma: importance
Czech National Hematopoietic Stem Cell Transplantation of histology for outcome. Biol Blood Marrow Transplant
Registry. Reduced-intensity conditioning for allogeneic stem 2008;14:418-25.
cell transplantation in patients with chronic myeloid leukemia 30. Peggs KS, Anderlini P, Sureda A. Allogeneic transplantation for
is associated with better overall survival but inferior disease- Hodgkin lymphoma. Br J Haematol 2008;143:468-80.
free survival when compared with myeloablative conditioning 31. Bensinger WI. Is there still a role for allogeneic stem-cell trans-
- a retrospective study of the Czech National Hematopoietic plantation in multiple myeloma? Best Pract Res Clin Haematol
Stem Cell Transplantation Registry. Neoplasma 2007;54:443- 2007;20:783-95
6. 32. de Lavallade H, El-Cheikh J, Faucher C, Fürst S, Stoppa AM,
14. Poiré X, Artz A, Larson RA, Kline J, Odenike O, Rich E, et al. Coso D, et al. Reduced-intensity conditioning allogeneic SCT
Allogeneic stem cell transplantation with alemtuzumab-based as salvage treatment for relapsed multiple myeloma. Bone
conditioning for patients with advanced chronic myelogenous Marrow Transplant 2008;41:953-60.
leukemia. Leuk Lymphoma 2009;50:85-91. 33. Rosiñol L, Pérez-Simón JA, Sureda A, de la Rubia J, de Arriba
15. Savani BN, Montero A, Kurlander R, Childs R, Hensel N, F, Lahuerta JJ, et al. Programa para el Estudio y la Terapéutica
Barrett AJ. Imatinib synergizes with donor lymphocyte infu- de las Hemopatías Malignas y Grupo Español de Mieloma
sions to achieve rapid molecular remission of CML relapsing (PETHEMA/GEM). A prospective PETHEMA study of tandem
after allogeneic stem cell transplantation. Bone Marrow autologous transplantation versus autograft followed by
Transplant 2005;36:1009-15. reduced-intensity conditioning allogeneic transplantation in
16. Craddock CF. Full-intensity and reduced-intensity allogeneic newly diagnosed multiple myeloma. Blood 2008;112:3591-3.
stem cell transplantation in AML. Bone Marrow Transplant 34. Childs R, Chernoff A, Contentin N, Bahceci E, Schrump D,
2008;41:415-23. Leitman S, et al. Regression of metastatic renal-cell carcinoma
17. Gutierrez-Aguirre CH, Cantú-Rodríguez OG, Gonzalez-Llano after nonmyeloablative allogeneic peripheral-blood stem-cell
O, Salazar-Riojas R, Martinez-González O, Jaime-Pérez JC, et transplantation. N Engl J Med 2000;343:750-8
al. Non-myeloablative hematopoietic stem cell transplantation 35. Peres E, Abidi MH, Mellon-Reppen S, Klein J, Braun T, Abella
is of limited value in advanced or refractory acute myeloblas- E, et al. Reduced intensity transplantation for metastatic renal
tic leukemia. The Mexican experience. Hematology 2007; 12: cell cancer with 2-year follow-up. J Immunother 2007;30:562-
193-7. 6.
18. Schmid C, Schleuning M, Hentrich M, Markl GE, Gerbitz A, 36. Ueno NT, Rizzo JD, Demirer T, Cheng YC, Hegenbart U,
Tischer J , et al. High antileukemic efficacy of an intermediate Zhang MJ, et al. Allogeneic hematopoietic cell transplantation
intensity conditioning regimen for allogeneic stem cell trans- for metastatic breast cancer. Bone Marrow Transplant 2008;
plantation in patients with high-risk acute myeloid leukemia 41:537-45.
in first complete remission. Bone Marrow Transplant 2008; 41: 37. Kojima R, Kami M, Hori A, Murashige N, Ohnishi M, Kim
721-7. SWet al. Reduced-intensity allogeneic hematopoietic stem-cell
19. Bornhäuser M, Illmer T, Oelschlaegel U, Schetelig J, Orde- transplantation as an immunotherapy for metastatic colorectal
mann R, Schaich M, et al. Gemtuzumab ozogamicin as part of cancer. Transplantatio 2004;78:1740-6.
reduced-intensity conditioning for allogeneic hematopoietic 38. Blaise D, Bay JO, Faucher C, Michallet M, Boiron JM, Choufi
cell transplantation in patients with relapsed acute myeloid B, et al. Reduced-intensity preparative regimen and allogeneic
leukemia. Clin Cancer Res. 2008;14:5585-93 stem cell transplantation for advanced solid tumors. Blood
20. Pagel JM, Appelbaum FR, Eary JF, Rajendran J, Fisher DR, 2004;103:435-41.
Gooley T, et al. 131I-anti-CD45 antibody plus busulfan and 39. Sakamaki H, Harada M. Allo-SCT using reduced-intensity
cyclophosphamide before allogeneic hematopoietic cell trans- conditioning against advanced pancreatic cancer: a Japanese
plantation for treatment of acute myeloid leukemia in first survey. Bone Marrow Transplant2008;42:99-103.
remission. Blood 2006;107:2184-91. 40. Conrad R, Remberger M, Cederlund K, Ringdén O, Barkholt L.
21. Ciurea SO, Sadegi B, Wilbur A, Alagiozian-Angelova V, A comparison between low intensity and reduced intensity
Gaitonde S, Dobogai LC, et al. Effects of extensive spleno- conditioning in allogeneic hematopoietic stem cell transplan-
megaly in patients with myelofibrosis undergoing a reduced tation for solid tumors. Haematologica 2008;93:265-72.
intensity allogeneic stem cell transplantation. Br J Haematol 41. Secondino S, Carrabba MG, Pedrazzoli P, Castagna L, Spina F,
2008;141:80-3. Grosso F, et al. European Group for Blood and Marrow
22. Kröger N, Thiele J, Zander A, Schwerdtfeger R, Kobbe G, Transplantation Solid Tumors Working Party. Reduced inten-
Bornhäuser M, et al. MDS-Subcommittee of the Chronic sity stem cell transplantation for advanced soft tissue sarco-
Leukaemia Working Party of the European Group for Blood mas in adults: a retrospective analysis of the European Group
and Marrow Transplantation. Rapid regression of bone mar- for Blood and Marrow Transplantation. Haematologica 2007;
row fibrosis after dose-reduced allogeneic stem cell transplan- 92:418-20.
tation in patients with primary myelofibrosis. Exp Hematol 42. Laport GG, Sandmaier BM, Storer BE, Scott BL, Stuart MJ,
2007;35:1719-22. Lange T, et al. Reduced-intensity conditioning followed by
23. Hoogendoorn M, Jedema I, Barge RM, van Luxemburg-Heijs allogeneic hematopoietic cell transplantation for adult patients
SA, Beaumont F, Marijt EW, et al. Characterization of graft- with myelodysplastic syndrome and myeloproliferative disor-
versus-leukemia responses in patients treated for advanced ders. Biol Blood Marrow Transplant 2008;14:246-55
chronic lymphocytic leukemia with donor lymphocyte infu- 43. Delgado J, Pillai S, Benjamin R, Caballero D, Martino R,
sions after in vitro T-cell depleted allogeneic stem cell trans- Nathwani A, et al. The effect of in vivo T cell depletion with
plantation following reduced-intensity conditioning. alemtuzumab on reduced-intensity allogeneic hematopoietic
Leukemia 2007;21:2569-74. cell transplantation for chronic lymphocytic leukemia. Biol
24. Boyiadzis M, Foon KA, Pavletic S Hematopoietic stem cell Blood Marrow Transplant 2008;14:1288-97
transplantation for chronic lymphocytic leukemia: potential 44. Chunduri S, Dobogai LC, Peace D, Saunthararajah Y, Quigley
cure for an incurable disease. Expert Opin Biol Ther 2007; 7: J, Chen YH, et al. Fludarabine/i.v. BU conditioning regimen:
1789-97. myeloablative, reduced intensity or both? Bone Marrow
25. Dreger P, Brand R, Hansz J, Milligan D, Corradini P, Finke J, et Transplant 2008;41:935-40.
al. Chronic Leukemia Working Party of the EBMT. Treatment- 45. Andersson BS, de Lima M, Thall PF, Wang X, Couriel D,
related mortality and graft-versus-leukemia activity after allo- Korbling M, et al. Once daily i.v. busulfan and fludarabine (i.v.
geneic stem cell transplantation for chronic lymphocytic Bu-Flu) compares favorably with i.v. busulfan and cyclophos-
leukemia using intensity-reduced conditioning. Leukemia phamide (i.v. BuCy2) as pretransplant conditioning therapy in
2003;17:841-8. AML/MDS. Biol Blood Marrow Transplant 2008;14:672-84.
26. Maloney D. Allogeneic transplantation following nonmye- 46. Fagioli F, Bacigalupo A, Frassoni F, Van Lint MT, Occhini D,

| 254 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

Gualandi F, et al. Allogeneic bone marrow transplantation for Blood Marrow Transplant 2003;9:257-65
acute myeloid leukemia in first complete remission: the effect 56. Massenkeil G, Nagy M, Lawang M, Rosen O, Genvresse I,
of FAB classification and GVHD prophylaxis. Bone Marrow Geserick G, et al. Reduced intensity conditioning and prophy-
Transplant 1994;13:247-52. lactic DLI can cure patients with high-risk acute leukaemias if
47. Pession A, Locatelli F, Zecca M, Rondelli R, Prete A, Bonetti F, complete donor chimerism can be achieved. Bone Marrow
et al. Cyclosporine-A as GVHD prophylaxis in allogeneic BMT Transplant 2003;31:339-45.
for childhood acute leukemia. AIEOP-BMT group. Bone 57. Glass B, Nickelsen M, Dreger P, Claviez A, Hasenkamp J, Wulf
Marrow Transplant 1998;21 Suppl 2 :S50-2. G, Trümper L, et al. Reduced-intensity conditioning prior to
48. Giralt S. The role of alemtuzumab in nonmyeloablative allogeneic transplantation of hematopoietic stem cells: the
hematopoietic transplantation. Semin Oncol 2006;33 (2 Suppl need for T cells early after transplantation to induce a graft-
5):S36-43. versus-lymphoma effect. Bone Marrow Transplant 2004; 34:
49. Alyea EP, Li S, Kim HT, Cutler C, Ho V, Soiffer RJ, et al. Siro-
limus, tacrolimus, and low-dose methotrexate as graft-versus- 391-7.
host disease prophylaxis in related and unrelated donor 58. Carella AM, Beltrami G, Corsetti MT, Nati S, Musto P, Scal-
reduced-intensity conditioning allogeneic peripheral blood zulli P, et al. Reduced intensity conditioning for allograft after
stem cell transplantation. Biol Blood Marrow Transplant 2008; cytoreductive autograft in metastatic breast cancer. Lancet
14:920-6. 2005;366:318-20.
50. Pérez-Simón JA, Martino R, Caballero D, Valcarcel D, Rebollo 59. Sinha R, Lonial S. Novel treatment approaches for patients
N, de la Cámara R, et al. Grupo Español de Trasplante with relapsed and refractory multiple myeloma. Curr Treat
Hematopoyético (GETH). Reduced-intensity conditioning Options Oncol 2006;7:246-57.
allogeneic transplantation from unrelated donors: evaluation 60. Olavarria E, Siddique S, Griffiths MJ, Avery S, Byrne JL, Piper
of mycophenolate mofetil plus cyclosporin A as graft-versus- KP, et al. Posttransplantation imatinib as a strategy to post-
host disease prophylaxis. Biol Blood Marrow Transplant 2008; pone the requirement for immunotherapy in patients under-
14:664-71. going reduced-intensity allografts for chronic myeloid
51. Slavin S.Reduced-intensity conditioning or nonmyeloablative leukemia. Blood 2007;110:4614-7.
stem cell transplantation: introduction, rationale, and historic 61. Armand P, Gannamaneni S, Kim HT, Cutler CS, Ho VT,
background. Semin Oncol 2004;31:1-3. Koreth J, et al. Improved survival in lymphoma patients receiv-
52. Pérez-Simón JA, Caballero D, Diez-Campelo M, Lopez-Pérez ing sirolimus for graft-versus-host disease prophylaxis after
R, Mateos G, Cañizo C, et al. Chimerism and minimal resid- allogeneic hematopoietic stem-cell transplantation with
ual disease monitoring after reduced intensity conditioning reduced-intensity conditioning. J Clin Oncol 2008;26:5767-74.
(RIC) allogeneic transplantation. Leukemia 2002;16:1423-3. 62. El-Cheikh J, Michallet M, Nagler A, de Lavallade H, Nicolini FE,
53. Mohty M, Avinens O, Faucher C, Viens P, Blaise D, Eliaou JF. Shimoni A, et al. High response rate and improved graft-ver-
Predictive factors and impact of full donor T-cell chimerism
after reduced intensity conditioning allogeneic stem cell trans- sus-host disease following bortezomib as salvage therapy after
plantation. Haematologica 2007;92:1004-6. reduced intensity conditioning allogeneic stem cell transplanta-
54. Miller JS, Weisdorf DJ, Burns LJ, Slungaard A, Wagner JE, tion for multiple myeloma. Haematologica 2008;93:455-8.
Verneris MR, et al. Lymphodepletion followed by donor lym- 63. Lang P, Handgretinger R. Haploidentical SCT in children: an
phocyte infusion (DLI) causes significantly more acute graft- update and future perspectives. Bone Marrow Transplant
versus-host disease than DLI alone. Blood 2007;110:2761-3. 2008;42 Suppl 2:S54-9
55. Peggs KS, Mackinnon S, Williams CD, D'Sa S, Thuraisun- 64. Solomon SR, Mielke S, Savani BN, Montero A, Wisch L,
daram D, Kyriakou C, et al. Reduced-intensity transplantation Childs R, et al. Selective depletion of alloreactive donor lym-
with in vivo T-cell depletion and adjuvant dose-escalating phocytes: a novel method to reduce the severity of graft-ver-
donor lymphocyte infusions for chemotherapy-sensitive sus-host disease in older patients undergoing matched sibling
myeloma: limited efficacy of graft-versus-tumor activity. Biol donor stem cell transplantation. Blood 2005;106:1123-9.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 255 |
Stem cell transplantation

The role of reduced intensity conditioning in


allogeneic hematopoietic stem cell transplantation

A. Gratwohl A B S T R A C T

Reduced intensity conditioning (RIC) has obtained wide acceptance over the last decade in allo-
Hematology, University Hospital, geneic hematopoietic stem cell transplantation (HSCT). It has extended this therapy to older patients
University of Basel, Basel,
Switzer-land and opened new possibilities for patients with comorbidities. Nearly a quarter of all allogeneic HSCT
in Europe are currently prepared with RIC. RIC HSCT has given proof of principle for the power of
immunotherapy: on its own, a healthy immune system can control a malignant disease. RIC reduces
Hematology Education: mortality due to the conditioning; it remains a full allogeneic transplant. It does not reduce risk of
the education program for the death from GvHD or infection and is associated with an increased risk of rejection and relapse. Overall
annual congress of the European
survival is not necessarily improved compared to standard conditioning. RIC HSCT has its primary place
Hematology Association
in situations where this benefit of reduced TRM is not offset by increased rejection or relapse, for
2009;3:256-260 example, primarily in older patients with disease in remission. Its value is currently being tested in a
prospective randomised EBMT study for elderly patients with AML in their first complete remission.

Introduction occlusive syndrome, diffuse alveolar hemor-


Hematopoietic stem cell transplantation rhage. It can also induce severe mucositis or
(HSCT) has become an established therapy failure of near every organ (Figure 2). There
for many congenital or acquired severe dis- has been substantial improvement in sur-
orders of the hematopoietic system, as well vival over the last decades.6,7 These
as for chemo- radio-, and immuno-sensitive improved outcome results were primarily
malignancies.1,2 It is estimated that more due to a significant reduction of infectious
than 50,000 such procedures are performed death. Disappointingly, there has been little
annually all over the world. The risk factors improvement in reduction of relapse and in
have been defined and results have signifi- reduction of death from GvHD since the
cantly improved over the last decades. early introduction of calcineurine inhibitors
Allogeneic and autologous stem cells from in the early 1980s. It is understandable that
bone marrow, peripheral blood or cord there is a substantial interest on reducing the
blood are used depending on the disease, detrimental effects from conditioning.
clinical condition and availability of a donor.
Still, HSCT remains associated with signifi- History of reduced intensity conditioning
cant early and late morbidity and mortality, hematopoietic stem cell transplantation
and this is illustrated in Figure 1. Early mor- The basic concepts of allogeneic HSCT
tality increases with increasing degree of were outlined very early in the history and
mismatching, and many years might be they still hold true.8 The purpose of the con-
required to compensate for the early years of ditioning is threefold: (i) to immunosuppress
life lost due to the immunological complica- the recipient in order to accept the trans-
tions of the allogeneic stem stell product. In plant; (ii) to eradicate the disease; (iii) to cre-
contrast, relapse remains the key problemat- ate space for the incoming hematopoietic
ic of autologous HSCT. The important role stem cells. Engraftment itself is a complex
of the immunological complications is fur- process and influenced by the degree of
ther illustrated by the best results of syn- immunosuppression before and after the
geneic HSCT and the difference between transplant, and by the stem cell source, the
matched and mismatched allogeneic donors. cell dose and the composition of the stem
We still lack the tools to separate the detri- cell product. Control of the disease is influ-
mental consequences of GvHD from the enced by the degree of conditioning, the
beneficial effects of graft-versus-disease graft product, the degree of engraftment,
effects on a routine basis.1 presence or absence of graft-versus-host
Causes of death after an HSCT include reactions and by the post-transplant treat-
disease related (relapse) or transplant related ment.1,8 The importance of the role of both
complications.1,5 The latter can be subdivid- conditioning and graft-versus-leukemia
ed in immunological complications (rejec- effects to control the disease was shown
tion or graft-versus-host disease), death from very early in history. It is nicely illustrated by
infection or death due to the conditioning the first clinical description of the disappear-
itself. High dose chemotherapy or total body ance of a refractory acute leukemia follow-
irradiation (TBI) can directly cause endothe- ing the transfusion of granulocytes for
lial damage syndromes, for example, veno patients with a severe infection in neutrope-

| 256 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

Figure 2. Principle causes of death after allogeneic HSCT.


Curves represent survival and causes of death of patients
transplanted for early disease.
Figure 1. Outcome of HSCT by donor type. Results are
based on near 200,000 autologous and allogeneic HSCT
reported to thee EBMT.

nia, which was accompanied by a skin rash, liver abnor- transplants up to metacrine transplants. There was also
malities and diarrhea:9 graft-versus-host disease by its an intensive debate between myeloablative versus
own can induce graft-versus-leukemia and eliminate the immunoablative conditioning strategies.27-32
disease.10-13 Still, early results in the 1970s were disap-
pointing. Transplants for leukemia were primarily based Current status of reduced intensity conditioning
on single dose total body irradiation alone. The majori- hematopoietic stem cell transplantation
ty of the patients who did engraft and who did survive Today, RIC HSCT is an established form of treatment
the early complications eventually relapsed. It was the and nearly a quarter of all allogeneic HSCT performed
concept of combining total body irradiation with last year in Europe were RIC HSCT (Figure 3).33 The
chemotherapy (cyclophosphamide and TBI) or giving concept is used all over Europe, even though some
intensive radiomimetic chemotherapy (CyBu, BACT), countries apply it more and others to a lesser extent
which led to the success.14 Subsequent concepts in the (Figure 4). The general term RIC has become established
1980s concentrated on increasing conditioning and find- and the distinction is no longer between im-muno-sup-
ing the maximum tolerated conditioning regimen. All of pression or myelosuppression. All drugs have both
the many attempts failed. They all successfully reduced immuno- and myelosuppressive properties in common.
relapse. This occurred at the expense of increased trans- Focus is primarily on the total intensity. This explains
plant related mortality and the net survival was never why the separation between standard and reduced
improved. intensity conditioning is not a clear cutoff. It has also
The reobservation of the powerful effects of donor become clear that, for different diseases, different arbi-
lymphocyte infusion in the 1990s brought a complete trary distinc-tions might be made.
shift.15-19 Focus was on smarter rather than “stronger” Within its definition’s committee, the EBMT has
transplants.20 Carefully conducted animal studies defined which regimens with doses equal or be-low
defined a minimal dose of conditioning required for sta- these limits should be classified as RIC HSCT
ble engraftment in dogs. A dose as low as 200 cGy TBI (www.ebmt.org). In brief, conditioning regimens, which
was found sufficient when accompanied by combined include total body irradiation below the dose of 600
immunosuppression either with cyclosporine and cGy (fractionated) +/- a purine analog and +/- ATG or
methotrexate or mycophenolate mofetil.21 These exper- with a dose of less than 60 mg/kg of cyclophosphamide
iments formed the basis for the now traditional Seattle- or alternatively, less than 8 mg/kg busulphan should be
Leipzig mini-transplant conditioning regimen.22-25 The con- considered as RIC for leukemias. The same applies for
cept was rapidly adopted when initial studies showed patients with lymphoma, where in addition condition-
surprisingly good early results, such as those published ing regimens, using melphalan of a dose of less than 140
very early on patients with chronic myeloid leukemia.26 mg/m2 +/- purine analogue +/- campath or ATG should
It was followed by an over-abundance of re-duced be considered as RIC HSCT. High dose fludarabine (90
intensity conditioning regimens, to such an extent that mg/m2 i.v. with TBI 2 Gy) should also be considered as
the diversity of RIC probably exceeds the number of RIC. For myelomas, melphalan less than 100 mg/m2, +/-
transplant teams. Initially, there was also a wealth of purine analogue +/- ATG is considered RIC. Any dose of
names for this new form of conditioning, from mini- cyclophosphamide less than 1,200 mg/m2 +/- ATG is
transplants, micro-transplants, non-myelo-ablative con-sidered RIC for aplastic anemia and nonconstitu-

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 257 |
14th Congress of the European Hematology Association

complete remission in patients with active disease prior


to the transplant. RIC HSCT does reduce early toxicity
and has expanded the age limit. Still, there are no com-
parative studies yet between RIC and standard condi-
tioning regimens for similar patient categories. There
are indications that incidence of acute and chronic
GvHD is probably similar if patients without engraft-
ment are excluded.33 The use of RIC HSCT is associated
with a higher incidence of graft rejection, specifically if
bone marrow is used as stem cell source.22 Peripheral
blood is, therefore, the recommended stem cell source
for RIC HSCT in the lower intensity spectrum. There
Figure 3. Number of allogeneic HSCT reported to the EBMT are also indications that RIC HSCT has made little
activity survey office from 1990 to 2007 and numbers of impact on the rate of infectious complications. There is
RIC allogeneic HSCT.
one exception: the risk of lethal septicemias within the
first 30 days is lower in patients with RIC HSCT. There
is clearly a reduced rate of days with neutropenia in
tional bone marrow failures syndromes. For solid patients with RIC compared to standard HSCT. There is
tumors busulphan, less than 8 mg/kg +/- TBI up to 600 one early complication, potentially lethal, which is
cGy +/- purine analogue and +/- ATG or cyclophos- more frequent in RIC HSCT. The risk of early acute
phamide less than 60 mg/kg should be considered as hemolysis should not be neglected in situations of a
RIC HSCT. There are no general recommendations for minor blood group barrier. Specifically after peripheral
other conditioning approaches or other disease cate- blood transplants, rapidly expanding donor lympho-
gories. There are no prospective comparative studies. cytes can induce hyperacute red blood cell lysis.38 With
Feasibility of RIC HSCT has been documented for a follow-up of more than 5 years in many disease cate-
patients with acute leukemias, chronic leukemias, gories, RIC HSCT appears to be associated with a high-
myelodysplastic and myeloproliferative syndromes, er incidence of relapse. Few longterm remissions are
lymphomas and multiple myelomas.27-32,34 RIC can main- observed for patients transplanted with RIC in very
tain complete remission for many years and induce advanced disease.

% RIC transplants 2007

no allogeneic hematopoietic stem cell


transplantation/no report

no RIC

1-25

25.1-40

>40

Algeria
Iran
Israel
Lebanon
Saudi Arabia
South Africa
Tunisia

Figure 4. Proportion of RIC HSCT amongst allogeneic HSCT reported to the EBMT activity survey office in 2007.

| 258 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

RIC HSCT can be applied to patients with high risks plications and changes over calendar time. Bone Marrow
Transplant 2005;36:757-69.
as measured by EBMT risk score or to patients with 6. Viollier R, Socié G, Tichelli A, Bacigalupo A, Korthof ET,
multiple comorbidities or a high Sorror score. However, Marsh J, et al. Recent improvement in outcome of unrelated
RIC HSCT does not abrogate the risk score or the donor transplantation for aplastic anemia. Bone Marrow
comorbidity or Karnofsky score. Patients with a high Transplant 2008;41:45-50.
7. Gratwohl A, Brand R, Apperley J, Crawley C, Ruutu T,
risk score or a high comorbidity score have a worse out- Corradini P, et al. Chronic Leukemia Working Party of the
come than patients with a low risk score or with no European Group for Blood and Marrow Transplantation.
comorbidities, whether they receive a transplant with Allogeneic hematopoietic stem cell transplantation for chron-
ic myeloid leukemia in Europe 2006: transplant activity, long-
standard or RIC approach.38 term data and current results. An analysis by the Chronic
RIC HSCT has also been discussed as a new tool to Leukemia Working Party of the European Group for Blood and
avoid late complications of an allogeneic HSCT in chil- Marrow Transplantation (EBMT). Haematologica 2006; 91:
513-21.
dren.39 Specifically, some late effects, such as cataract 8. Thomas ED, Storb R, Clift RA, Fefer A, Johnson L, Neiman PE,
formation, growth retardation or fertility are directly et al. Bone-marrow transplantation. N Engl J Med 1975;292:
affected by conditioning intensity. Feasibility of RIC 895-902.
9. Mathé G. Leukocyte transfusions. In: Bone Marrow
HSCT in children with malignant disorders has been Transplantation and Leukocyte Transfusions. Mathé G, Amiel
documented. RIC HSCT appears less promising in con- JL, Schwarzenberg L, editors. American Lecture Series No.
genital disorders such as hemoglobinopathies, where 793, Springield (ILL): CT Thomas. p 104-130
10. Weiden PL, Flournoy N, Thomas ED, Prentice R, Fefer A,
the risk of rejection remains high. It remains open Buckner CD, et al. Antileuke-mic effect of graft-versus-host
whether the improvement in late effects remains in bal- disease in human recipients of allogeneic-mar-row grafts. N
ance with the risk of late relapse. Engl J Med 1979;300:1068-73.
11. Weiden PL, Storb R, Tsoi MS, Graham TC, Lerner KG,
Thomas ED. Infusion of donor lymphocytes into stable canine
Outlook radiation chimeras: implications for mechanism of transplan-
The place of RIC appears most successful in patients tation tolerance. J Immunol 1976;116:1212-9.
with advanced age where early transplant related mor- 12. Mathé G, Amiel JL, Schwarzenberg L, Cattan A, Schneider M.
Adoptive immuno-therapy of acute leukemia: experimental
tality caused by the conditioning reduces the probabili- and clinical results. Cancer Res 1965;25:1525-31.
ty of a better survival compared to a conventional non- 13. Barnes DW, Corp MJ, Loutit JF, NeaL FE. Treatment of murine
transplant strategy the most. This applies primarily to leukaemia with X rays and homologous bone marrow; prelim-
inary communication. Br Med J 1956 2:626-7.
patients with acute leukemia above the age of 60 years. 14. Thomas ED, Buckner CD, Clift RA, Fefer A, Johnson FL,
Survival prospects without a transplant are limited but Neiman PE, et al. Marrow transplantation for acute nonlym-
not nil. If RIC can maintain complete remissions at the phoblastic leukemia in first remission. N Engl J Med 1979;
301:597-9.
expense of a limited amount of early toxicity and mor- 15. Kolb HJ, Schattenberg A, Goldman JM, Hertenstein B,
tality it might become standard of care. Preliminary Jacobsen N, Arcese W, et al. Graft-versus-leukemia effect of
pilot data from non-controlled studies indicate that the donor lymphocyte transfusions in marrow grafted patients.
European Group for Blood and Marrow Transplantation
traditional 20% survival rate at 5 years without a trans- Working Party Chronic Leukemia. Blood 1995;86:2041-50.
plant could be increased to 40 to 50% with RIC HSCT. 16. Kolb HJ. Graft-versus-leukemia effects of transplantation and
No comparative data are available yet.34,40 A prospective donor lymphocytes. Blood. 2008;112:371-83.
controlled European wide study has been organised by 17. Giralt SA, Kolb HJ. Donor lymphocyte infusions. Curr Opin
Oncol 1996;8:96-102.
the EBMT. Patients with AML in first complete remis- 18. Slavin S, Naparstek E, Nagler A, Ackerstein A, Kapelushnik J,
sion after induction chemotherapy and with an HLA- Or R. Allogeneic cell therapy for relapsed leukemia after bone
identical sibling or unrelated donor will be randomized marrow transplantation with donor peripheral blood lympho-
cytes. Exp Hematol 1995;23:1553-62.
to early RIC HSCT in first CR or to conventional con- 19. Slavin S, Naparstek E, Nagler A, Ackerstein A, Samuel S,
solidation chemotherapy. If the study can successfully Kapelushnik J, et al. Allogeneic cell therapy with donor periph-
be conducted, we might know in 5 years from now eral blood cells and recombinant human interleukin-2 to treat
leukemia relapse after allogeneic bone marrow transplan-
whether RIC HSCT will hold the promises it gives tation Blood 1996;87:2195-204.
today. 20. Slavin S. Immunotherapy of cancer with alloreactive lympho-
cytes. N Engl J Med 2000;14;802-3.
21. Storb R, Yu C, Wagner JL, Deeg HJ, Nash RA, Kiem HP, et al.
Stable mixed hematopoietic chimerism in DLA-identical litter-
References mate dogs given sublethal total body irradiation before and
pharmacological immunosuppression after marrow transplan-
1. Copelan EA. Hematopoietic stem-cell transplantation. N Engl tation. Blood 1997;89:3048-54.
J Med 2006;354:1813-26. 22. Niederwieser D, Maris M, Shizuru JA, Petersdorf E, Hegenbart
2. Appelbaum FR. Hematopoietic-cell transplantation at 50. N U, Sandmaier BM, et al. Low-dose total body irra-diation (TBI)
Engl J Med 2007;357:1472-5. and fludarabine followed by hematopoietic cell transplanta-
3. Gratwohl A, Baldomero H, Schwendener A, Rocha V, tion (HCT) from HLA-matched or mismatched unrelated
Apperley J, Frauendorfer K, et al. The EBMT activity survey donors and postgrafting immunosuppression with cyclo-
2007 with focus on allogeneic HSCT for AML and novel cellu- sporine and mycophenolate mofetil (MMF) can induce durable
lar therapies. Bone Marrow Transplant. 2009;26 [In press] complete chimerism and sustained remissions in patients with
4. Gratwohl A, Baldomero H, Schwendener A, Gratwohl M, hematological diseases. Blood 2003;10:1620-9.
Apperley J, Niederwieser D, et al. Joint Accreditation 23. McSweeney PA, Niederwieser D, Shizuru JA, Sandmaier BM,
Committee of the International Society for Cellular Therapy; Molina AJ,Maloney DG, et al. Hematopoietic cell transplanta-
European Group for Blood and Marrow Transplantation; tion in older patients with hematologic malignancies: replac-
European Leukemia Net. Predictability of hematopoietic stem ing high-dose cytotoxic therapy with graft-versus-tumor
cell transplantation rates. Haematologica 2007:92:1679-86. effects. Blood 2001;97:3390-400.
5. Gratwohl A, Brand R, Frassoni F, Rocha V, Niederwieser D, 24. Baron F, Storb R, Storer BE, Maris MB, Niederwieser D,
Reusser P, et al. Acute and Chronic Leukemia Working Parties; Shizuru JA, et al. Factors associated with outcomes in allo-
Infectious Diseases Working Party of the European Group for geneic hematopoietic cell transplantation with nonmyeloabla-
Blood and Marrow Transplantation.Cause of death after allo- tive conditioning after failed myelo-ablative hematopoietic
geneic haematopoietic stem cell transplantation (HSCT) in cell transplantation. J Clin Oncol 2006;24:4150-7.
early leukaemias: an EBMT analysis of lethal infectious com- 25. Hegenbart U, Niederwieser D, Sandmaier BM, Maris MB,

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 259 |
14th Congress of the European Hematology Association

Shizuru JA, Greinix H, et al. Treatment for acute myelogenous 33. Gratwohl A, Baldomero H, Passweg J, Urbano-Ispizua A.
leukemia by low-dose, total-body, irradiation-based condi- European Group for Blood and Marrow Transplantation
tioning and hematopoietic cell transplantation from related (EBMT). Accreditation Committee Increasing use of reduced
and unrelated donors. J Clin Oncol 2006;24:444-53. intensity conditioning transplants: report of the 2001 EBMT
26. Or R, Shapira MY, Resnick I, Amar A, Ackerstein A, Samuel S, activity survey. Bone Marrow Transplant 2002;30:813-31.
et al. Nonmyeloablative allogeneic stem cell transplantation 34. Appelbaum FR. Allogeneic hematopoietic cell transplantation
for the treatment of chronic myeloid leukemia in first chronic for acute myeloid leuke-mia when a matched related donor is
phase. Blood 2003;101:441-5. not available. Hematology Am Soc Hematol Educ Program
27. Rotta M, Storer BE, Sahebi F, Shizuru JA, Bruno B, Lange T, et 2008;2008:412-7.
al. Long-term outcome of patients with multiple myeloma
after autologous hematopoietic cell transplantation and non- 35. Mielcarek M, Martin PJ, Leisenring W, Flowers ME, Maloney
myeloablative allografting. Blood 2009;113:3383-91. DG, Sandmaier BM, et al. Graft-versus-host disease after non-
28. Sobecks RM, Dean R, Rybicki LA, Chan J, Theil KS, Macklis myeloablative versus conven-tional hematopoietic stem cell
R, et al. 400 cGy TBI with fludarabine for reduced-intensity transplantation. Blood 2003;102:756-62.
conditioning allogeneic hematopoietic stem cell transplanta- 36. Junghanss C, Marr KA, Carter RA, Sandmaier BM, Maris MB,
tion. Bone Marrow Transplant 2008;42:715-22. Maloney DG, et al. Incidence and outcome of bacterial and
29. Maris MB, Niederwieser D, Sandmaier BM, Storer B, Stuart fungal infections- following nonmyeloablative compared with
M, Maloney D, et al. HLA-matched unrelated donor hemato- myeloablative allogeneic hematopoietic stem cell transplanta-
poietic cell transplantation after nonmyeloablative condition- tion: a matched control study. Biol Blood Marrow Transplant
ing for patients with hema-tologic malignancies. Blood 2003; 2002;8:512-20.
102:2021-30. 37. Sorror ML, Giralt S, Sandmaier BM, De Lima M, Shahjahan M,
30. Giralt S, Estey E, Albitar M, van Besien K, Rondón G, Ander- Maloney DG, et al. Hematopoietic cell transplantation specif-
lini P, et al. Engraftment of allogeneic hematopoietic progeni- ic comor-bidity index as an outcome predictor for patients
tor cells with purine analog-containing chemotherapy: har- with acute myeloid leukemia in first remission: combined
nessing graft-versus-leukemia without myeloablative therapy. FHCRC and MDACC experiences. Blood 2007;110:4606-13.
Blood 1997;89:4531-6. 38. Kimura F, Sato K, Kobayashi S, Ikeda T, Sao H, Okamoto S, et
31. Crawley C, Szydlo R, Lalancette M, Bacigalupo A, Lange A, al. Japan Marrow Donor Program. Impact of ABO-blood
Brune M, et al. Chronic Leukemia Working Party of the EBMT. group incompatibility on the outcome of recipients of bone
Outcomes of reduced-intensity transplantation for chronic
myeloid leukemia: an analysis of prognostic factors from the marrow transplants from unrelated donors in the Japan
Chronic Leukemia Working Party of the EBMT. Blood 2005; Marrow Donor Program. Haematologica 2008;93:1686-93.
106:2969-76. 39. Yaniv I, Stein J. EBMT Paediatric Working Party. Reduced-
32. Crawley C, Lalancette M, Szydlo R, Gilleece M, Peggs K, intensity conditioning in children: a reappraisal in 2008. Bone
Mackinnon S, et al. Chromic Leukaemia Working Party of the Marrow Transplant 2008;41:18-22.
EBMT. Outcomes for re-duced-intensity allogeneic transplan- 40. Niederwieser D, Lange T, Cross M, Basara N, Al-Ali H.
tation for multiple myeloma: an analysis of prognostic factors Reduced intensity conditioning (RIC) haematopoietic cell
from the Chronic Leukaemia Working Party of the EBMT. transplants in elderly patients with AML. Best Pract Res Clin
Blood 2005;105:4532-9. Haematol 2006;19:825-38.

| 260 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Thrombosis

Genetics of thrombosis

P.H. Reitsma A B S T R A C T

Venous thrombosis is a common episodic disease with a complex etiology. The mean annual inci-
Einthoven Laboratory for dence of venous thrombosis is about 1:1000, but there is a steep age gradient. In the etiology, both
Experimental Vascular Medicine,
Departments of Thrombosis and environmental and genetic risks are important. The environmental risk factors include medical condi-
Hemostasis and Nephrology, Leiden tions, such as surgery, prolonged bed rest and cancer, but also life style conditions, varying from obe-
University Medical Center, Leiden, sity and oral contraceptive use to long-haul air travel. A variety of genetic risk factors is known for
The Netherlands venous thrombosis. Arbitrarily these are often divided between classical genetic risk factors and ‘other’
genetic risk factors. There are six classical risk factors and these include deficiencies of the natural
anticoagulants protein C, protein S and antithrombin, factor V Leiden, prothrombin 20210, and blood
group non-O. With the exception of blood group non-O, these risk factors are quite rare in the gener-
Hematology Education: al population, but their associated relative risks are high, varying from 2 for blood group non-O to per-
the education program for the haps 20 antithrombin deficiency. The remaining risk factors consist predominantly of single for
annual congress of the European nucleotide polymorphisms (SNPs) in a variety of genes encoding coagulation proteins. The risk alleles
Hematology Association of these SNPs are often quite common, but the associated relative risks are low.
2009;3:261-265

Introduction also in veins. Deep venous thrombosis


The blood coagulation system is responsi- (DVT) and pulmonary embolism (PE), com-
ble for stemming the loss of blood in the monly referred to as venous thromboem-
case of vascular damage. In almost all indi- bolism (VTE), are common complications
viduals, the system is quite effective and during and after hospitalization for acute
excessive blood loss in therefore uncom- medical illness or surgery, but also often
mon. On the other hand, the capacity to occur spontaneously. The overall incidence
form blood clots brings the risk that blood of VTE, which increases with age, is approx-
clotting inadvertently leads to blood clots in imately 1:1000 in Western Europe and the
the circulation per se: thrombosis. Such United States (USA).1,2 The results of the
blood clots often occur in the arterial circu- VITAE study reported that the annual bur-
lation of individuals affected by atheroscle- den in the European Union (EU) of fatal and
rosis. non-fatal symptomatic VTE comprises
Atherosclerosis is an inflammatory degen- 435,000 cases of PE and 684,000 cases of
erative disease of the vessel wall. The degen- documented symptomatic DVT.3 The yearly
eration of the vessel wall leads to so-called number of VTE-related deaths was estimat-
atherosclerotic plaques with procoagulant ed at 543,500.
properties. The procoagulant potential of Pulmonary embolism accounts for 5 to 10%
the plaque becomes operational when the of deaths in hospitalized patients, making
endothelium on the plaque surface is dam- VTE the most common preventable cause of
aged, for example by plaque rupture, leading in-hospital death.
to the formation of a blood clot. This clot In the United Kingdom, an independent
may occlude the artery leading to myocar- Government Health Select Committee
dial infarction or ischemic stroke. In essence, enquiry in 2007 estimated that VTE in hos-
the blood clotting reactions on the damaged pitalized patients causes 25,000 deaths each
plaque surface are not different from the year(http://www.dh.gov.uk/en/Publicationsand
reactions that occur when a blood vessel is statistics/Lettersandcirculars/Dearcolleagueletter
damaged by a trauma, in which case the pri- s/DH_073957). The Committee noted that
mary driving force is also the exposure of deaths from VTE in hospital exceed the
strongly procoagulant proteins and surfaces combined total of deaths from breast cancer,
to blood. The reaction that follows is only to AIDS and traffic accidents. This mortality is
a limited extend sensitive to subtle varia- also over 25 times greater than the annual
tions in the concentrations of the coagula- deaths from methicillin resistant
tion proteins, which explains why levels of Staphylococcus aureus (MRSA) and more than
coagulations proteins, whether determined five times the total of all hospital-acquired
genetically or by environmental factors, only infections.
play a minor role in determining risk for These data give a strong indication of the
arterial thrombosis. burden of VTE in the EU, and underline that
Blood clots not only occur in arteries but actions to improve the prevention of VTE in

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 261 |
14th Congress of the European Hematology Association

primary, secondary and tertiary care are badly needed. coagulation cascade except for factor VIIa, for which
The need for action is also supported by the U.S. Surgeon there is no inhibitor when it is not in complex with tis-
General’s Call to Action released 15 September 2008 sue factor and factor Xa.
(http://www.surgeongeneral.gov/topics/deepvein/calltoaction/cal
l-to-action-on-dvt-2008.pdf). Deficiencies of natural anticoagulants
The risk of venous thrombosis is determined partly The coagulation balance will shift towards increased
by the delicate balance between procoagulant and anti- clot formation when coagulation inhibitors are present
coagulant pathways, but it also involves a poorly in low amounts. Based on this, one would predict that
defined anatomic substrate in the proximal leg veins. inhibitor deficiencies would increase the risk of throm-
The age-gradient of thrombosis incidence is steep, botic disease. Indeed, this appears to be the case. In
increasing from less than 1:10,000 in the young to 1:100 1965, it was already established by Egeberg et al. that a
per year in the elderly. Precise mechanisms for this age partial deficiency of antithrombin leads to an increased
gradient remain largely unknown.4 Prominent among risk of thrombosis.12 A complete deficiency of
hypothesized factors are age related metabolic changes, antithrombin has not been observed in humans and is
steroid hormone use, obesity and cancer. generally considered to be incompatible with life.
The basal risk for thrombosis is strongly influenced A similar pattern is observed for deficiencies of pro-
by inherited risk factors. The notion that genetics are tein C and protein S. Shortly after the discovery of these
important for venous thrombosis dates back to observa- anticoagulant proteins, it was established that complete
tions on familial occurrence in 1956.5 In the ensuing 50 deficiency of either protein C or protein S leads to lethal
years, stepwise progress was made in the identification purpura fulminans immediately after birth, whereas a
of inherited gene variations that, at least partly, partial deficiency increases the risk for venous thrombo-
explained the familial occurrence of thrombosis. sis.13-15
Moreover, we have learned now that in individuals The magnitude of the increased risk of protein C, S,
with an isolated thrombosis, genetic risk factors may and antithrombin deficiency is not known with certain-
also play a role in determining the risk profile. ty but has been estimated to be 10 to 20-fold. This esti-
mate is mostly based on observations in anecdotal fam-
Natural anticoagulants ilies rather than on large case-control studies. The rea-
The procoagulant forces of the clotting system are son why these latter studies have failed to yield reliable
kept in balance by several anticoagulant mechanisms. risk estimates is that the deficiency states are quite rare
The first operates at the level of the initiation of blood in the normal population (1 in 500 to 1 in 1000 at most),
coagulation and involves the soluble plasma protein tis- indicating that only very large studies will suffice for a
sue factor pathway inhibitor (TFPI).6 TFPI can bind to reliable estimate.16,17
the factor Xa when it is still in complex with tissue fac- After cloning of the genes encoding protein C, S and
tor-factor VIIa on the cell membrane. antithrombin it became possible to search for the genet-
Second, there is a set of serine protease inhibitors (ser- ic alterations that lead to a deficiency state. As expect-
pins) that are capable of irreversible inhibition of acti- ed, hundreds of different mutations have been docu-
vated coagulation enzymes. Best known among these is mented that all lead to loss of function of the gene in
antithrombin, that inhibits the activity of, not only the question.18-20 It almost appears as if every family has its
procoagulant thrombin, but also of factors IXa and Xa.7 own private mutation, and in that respect, the genetic
The inhibitory capacity of antithrombin is greatly architecture of deficiencies of natural anticoagulants is
enhanced by heparin. Heparin cofactor II inhibits very much like the genetic architecture of for example
thrombin rapidly in the presence of dermatan sulfate, hemophilia B. The spectrum of mutations is kept in
heparan sulfate, or heparin.8 Protein Z-dependent pro- databases like the Human Gene Mutation Database
tease inhibitor inhibits factors Xa and XIa of the coagu- (http://www.hgmd.cf.ac.uk/ac/). The large variety of muta-
lation cascade.9 Its name implies that it requires protein tions makes it difficult to do routine genetic testing for
Z, another circulating protein, to function properly, but protein C, S or antithrombin deficiency. Therefore, the
this only applies to its inhibition of factor Xa. Finally, diagnosis of these deficiency states in a thrombophilia
CI-inhibitor functions as a potent inhibitor for the coag- screening wholly relies on plasma assays. This is quite
ulation factors XIa and XIIa.10 straightforward for antithrombin and protein C, but can
Next, there is the so-called protein C anticoagulant be unreliable for protein S. These complications are
system.11 When the blood coagulation system is silent, beyond the scope of this review.
this anticoagulant system does not operate. It becomes The other components of the natural anticoagulant
active when thrombin, the key enzymatic end-product system are not clearly related to an increased risk of
of the clotting system, binds to the endothelial cell venous thrombosis but they may be related to other dis-
membrane protein thrombomodulin (TM). Once bound eases. Levels of heparin cofactor II are probably associ-
to TM, thrombin loses its capacity to cleave fibrinogen. ated with atherosclerosis.21 In some strains of mice,
Instead, thrombin will cleave protein C to form activat- heparin cofactor II deficiency is lethal, and neointima
ed protein C. This reaction is enhanced by the endothe- formation is increased in heterozygotes.22 One early
lial protein C receptor (EPCR). APC is able to proteolyt- report described deleterious nonsense mutations in ZPI,
ically inactivate the procoagulant co-factors factor Va which were associated with venous thrombosis.23 This
and factor VIIIa in the presence of its co-factor protein result could not be confirmed in a recent meta-analy-
S. sis.24 Complete deficiency of C1 inhibitor leads to
Taken together, these anticoagulant mechanisms angioedema, probably because of effects on the comple-
cover each serine protease and activated co-factor of the ment system.25 Heterozygosity for C1 inhibitor defects

| 262 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

seems to be associated with age-related macular degen- The prevalence of factor V Leiden is high in the
eration.26 Caucasian population. In LETS, the prevalence in the
Several studies have tried to relate levels of TFPI to Dutch population was estimated at 3%. In other ethnic
venous thrombosis. Low levels of TFPI seem to be relat- groups, like the Chinese and Japanese, factor V Leiden
ed to venous thrombosis, but homozygosity or het- is extremely rare.33
erozygosity for true deficiency states have not been Not long after the discovery of APC resistance and
described.27 factor V Leiden, a second unique mutation in a gene
The other components of the protein C anticoagulant encoding a pro-coagulant protein, prothrombin, was
system, thrombomodulin and EPCR have also been found.34 This mutation, often called FII G20210A, was
studied, and again, no true deficiency states have been later shown to increase the level of prothrombin mRNA
documented in humans. Both are membrane receptors by improving 3’ end formation of mature mRNA.35 This
of the endothelium. Shedding of these receptors into effect on mRNA metabolism leads to elevated levels of
the blood does occur and several assays are available to plasma prothrombin. The increase is about 20% in het-
estimate levels of soluble TM and EPCR. The relation- erozygotes, and in such individuals, the relative risk for
ship between these levels and venous thrombosis is not venous thrombosis is increased by about a factor of
very strong. Several missense mutations and polymor- three.34 By analogy with factor V Leiden, the thrombot-
phisms in the TM gene have been found, but the rela- ic tendency in homozygotes is relatively modest, the
tionship with venous thrombosis has not been proven prevalence in the general population is quite high
unequivocally.28 There is one frequent polymorphism in (~1%), and the mutation is present primarily in Cau-
the EPCR gene that is quite strongly related to soluble casians and not in other races.36
levels of EPCR that may be a weak risk factor for As noted above, the notion that procoagulant factors
venous thrombosis.29 could carry a risk for venous thrombosis was only gen-
erally accepted after the discovery of APC resistance
Gain of function mutations in procoagulant proteins and factor V Leiden. This is surprising in light of the fact
Not only a (partial) deficiency of coagulation that already in 1969, Jick et al. noted that ABO blood
inhibitors tips the hemostatic balance in the direction of group played an important role in venous thrombotic
venous thrombosis. An excess of pro-coagulant factors risk, in particular within the context of oral contracep-
may do the same. It was not until the discovery of tive use.37 It appeared that non-O individuals had a high-
Factor V Leiden that this notion became generally er risk than individuals with blood group O. This was
accepted. later confirmed in more formal studies and the risk asso-
The discovery of this mutation was based on seminal ciated with non-O is estimated to be around two.38
findings of Dahlbäck et al. in a family with what is now A probable explanation for this increased risk can be
called APC resistance.30 The unusual plasma phenotype based on seminal observations that date back to 1964.
in this family became evident in assays where the acti- Preston and Barr reported in that year that coagulation
vated partial thromboplastin time (APTT) was meas- factor VIII levels were higher in individuals with non-O
ured in both the absence and presence of activated pro- blood group than in those with blood group O.39 Later
tein C (APC). In most individuals, the addition of APC research showed that this was due to an effect of blood
markedly prolonged the APTT, but in members of the group on levels of Von Willebrand Factor, the carrier
family that Dahlbäck et al had identified the prolonga- protein of factor VIII.
tion was much less. Later studies showed that APC To summarize, genetically determined increases in
resistance is due to a mutation in coagulation factor V procoagulant function increase the risk for venous
within one of the cleavage sites for APC, and this muta- thrombosis. Factor V Leiden, PT 20210A and non-O
tion was named factor V Leiden.31 Although activated blood group are the major risk factors with relative risks
factor V Leiden can still be degraded by APC through varying between seven and two. The risk factors are
alternative cleavage sites, inactivation is less efficient. much more prevalent in the population than those
From the perspective of protein function, this mutation involved in anticoagulant function.
is a gain-of-function mutation because the pro-coagu-
lant potential of factor V Leiden is stronger than that for Single nucleotide polymorphisms
normal factor V. Since there are fewer possibilities to The genetic findings in procoagulant and anticoagu-
improve protein function, there is little or no hetero- lant proteins described above were obtained in relative-
geneity in the genetic basis of APC resistance; in other ly straightforward family and case-control studies using
words, the plasma phenotype almost always goes with simple genetic analysis tools. In the meantime, The
the same mutation. Human Genome Project has taught us that the human
The relative risk for venous thrombosis associated genome contains millions of variations that set one indi-
with factor V Leiden was first estimated in the Leiden vidual apart from another. Most of these differences
Thrombophilia Study (LETS) and found to be around take the form of common single nucleotide polymor-
seven.31 Although the estimates vary somewhat in sub- phisms (SNPs). In theory, some of the common SNPs
sequent studies, these original results were essentially may alter the risk for venous thrombosis, and the chal-
confirmed. Homozygotes for factor V Leiden have a rel- lenge that is before us is to sift out these relatively few
atively mild phenotype, at least compared to the severe functional SNPs from the millions of SNPs that are irrel-
phenotype of homozygotes for deficiencies of natural evant. During the past years, SNPs that influence throm-
anticoagulants.32 Compared to heterozygous deficiency botic risk have indeed been found. The discovery plat-
of natural anticoagulants, the risk of homozygous factor forms used a combination of family and case-control
V Leiden is quite potent. studies with either candidate gene or whole genome

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 263 |
14th Congress of the European Hematology Association

screening. 7. Olson ST, Bjork I. Regulation of thrombin activity by anti-


The results include SNPs in procoagulant genes, such thrombin and heparin. Semin Thromb Hemost 1994; 20:
373-409.
as fibrinogen (alpha, beta, and gamma), factor VIII, 8. Rau JC, Beaulieu LM, Huntington JA, Church FC. Serpins in
Tissue Factor, factor IX, factor X, factor XI, factor XIII thrombosis, hemostasis and fibrinolysis. J Thromb Haemost
(both the A and B subunit), glycoprotein 6, and 2007; 5 Suppl 1:102-15.
9. Broze GJ, Jr. Protein Z-dependent regulation of coagulation.
Thrombin Activated Fibrinolysis Inhibitor (TAFI). Also Thromb Haemost 2001;86:8-13.
common SNPs in the natural anticoagulants were found 10. Pratt CW, Macik BG, Church FC. Protein C inhibitor: purifi-
for TFPI, TM, protein C, and antithrombin.40,41 Al- cation and proteinase reactivity. Thromb Res 1989;53:595-
together, there is now reasonably convincing data for 602.
11. Esmon CT. The protein C pathway. Chest 2003;124(3
about 20 SNPs – mostly, if not exclusively, located in Suppl):26S-32S.
coagulation genes – that influence thrombotic risk. As 12. Egeberg O. Inherited antithrombin III deficiency causing
the large genome wide association studies that are still thrombophilia. Thromb Diath Haemorrh 1965;13:516-30.
under way become finished this number is likely to 13. Griffin JH, Evatt B, Zimmerman TS, Kleiss AJ, Wideman C.
Deficiency of protein C in congenital thrombotic disease. J
increase considerably and perhaps also SNPs in genes Clin Invest 1981;68:1370-3.
not commonly associated with hemostasis and throm- 14. Comp PC, Nixon RR, Cooper MR, Esmon CT. Familial pro-
bosis will be found. tein S deficiency is associated with recurrent thrombosis. J
Clin Invest 1984;74:2082-8.
The risk SNPs that were discovered share two com- 15. Bertina RM, Broekmans AW, van der Linden IK, Mertens K.
mon features. Almost by definition, the risk alleles have Protein C deficiency in a Dutch family with thrombotic dis-
a relatively high prevalence in the population, with ease. Thromb Haemost 1982;48:1-5.
extremes up to 80%. Secondly, the risk for venous 16. Lane DA, Mannucci PM, Bauer KA, Bertina RM, Bochkov NP,
Boulyjenkov V, et al. Inherited thrombophilia 2. Thromb
thrombosis that goes with these risk factors is relative- Haemost 1996;76:824-34.
ly low, that is, in the order of 1.5-fold. 17. Lane DA, Mannucci PM, Bauer KA, Bertina RM, Bochkov
NP, Boulyjenkov V, et al. Inherited thrombophilia 1. Thromb
Conclusions Haemost 1996;76:651-62.
18. Lane DA, Ireland H, Olds RJ, Thein SL, Perry DJ, Aiach M.
This review discussed how research into the genetic Antithrombin III: A database of mutations. Thromb
background of venous thrombosis slowly shifted from Haemost 1991;66:657-61.
discovering proteins and genes based on plasma assays 19. Reitsma PH, Bernardi F, Doig RG, Gandrille S, Greengard JS,
Ireland H, et al. Protein C deficiency: a database of muta-
to whole-genome studies aimed at determining the risk tions, 1995 update. On behalf of the Subcommittee on
associated with common genetic variation in the form Plasma Coagulation Inhibitors of the Scientific and
of SNPs. At the same time, we have seen attention shift Standardization Committee of the ISTH. Thromb Haemost
from genetic abnormalities that go with high risk, for 1995;73:876-89.
20. Gandrille S, Borgel D, Ireland H, Lane DA, Simmonds R,
example, 20-fold for antithrombin deficiency, to those Reitsma PH, et al. Protein S deficiency: a database of muta-
with a much smaller risk. The overall result is that we tions. For the Plasma Coagulation Inhibitors Subcommittee
now know about 20 genetic risk factors for venous of the Scientific and Standardization Committee of the
International Society on Thrombosis and Haemostasis.
thrombosis. This is providing us with unprecedented Thromb Haemost 1997;77:1201-14.
insight into the genetic architecture of this common but 21. Aihara K, Azuma H, Takamori N, Kanagawa Y, Akaike M,
complex disease. It is tempting to assume that the pres- Fujimura M, et al. Heparin cofactor II is a novel protective
ent knowledge of so many genetic risk factors easily factor against carotid atherosclerosis in elderly individuals.
Circulation 2004;109:2761-5.
leads to robust predictive models for venous thrombo- 22. Aihara K, Azuma H, Akaike M, Ikeda Y, Sata M, Takamori N,
sis. However, work in progress indicates that this is et al. Strain-dependent embryonic lethality and exaggerated
almost certainly not the case. The challenge which vascular remodeling in heparin cofactor II-deficient mice. J
remains before us is to translate these insights into Clin Invest 2007;117:1514-26.
23. Van de Water N, Tan T, Ashton F, O'Grady A, Day T,
approaches that can be used in the doctor’s office; for Browett P, et al. Mutations within the protein Z-dependent
example, for better tailoring of dose or duration of anti- protease inhibitor gene are associated with venous throm-
coagulant treatment. boembolic disease: a new form of thrombophilia. Br J
Haematol 2004; 127:190-4.
24. Dentali F, Gianni M, Lussana F, Squizzato A, Cattaneo M,
Ageno W. Polymorphisms of the Z protein protease inhibitor
References and risk of venous thromboembolism: a meta-analysis. Br J
Haematol 2008;143:284-7.
1. Oger E. Incidence of venous thromboembolism: a communi- 25. Cugno M, Zanichelli A, Foieni F, Caccia S, Cicardi M. C1-
ty-based study in Western France. EPI-GETBP Study Group. inhibitor deficiency and angioedema: molecular mechanisms
Groupe d'Etude de la Thrombose de Bretagne Occidentale. and clinical progress. Trends Mol Med 2009;[Epub ahed of
Thromb Haemost 2000;83:657-60. print].
2. Naess IA, Christiansen SC, Romundstad P, Cannegieter SC, 26. Ennis S, Jomary C, Mullins R, Cree A, Chen X, Macleod A,
Rosendaal FR, Hammerstrom J. Incidence and mortality of et al. Association between the SERPING1 gene and age-relat-
venous thrombosis: a population-based study. J Thromb ed macular degeneration: a two-stage case-control study.
Haemost 2007;5:692-9. Lancet 2008;372:1828-34.
3. Cohen AT, Agnelli G, Anderson FA, Arcelus JI, Bergqvist D, 27. Dahm A, Van Hylckama Vlieg A, Bendz B, Rosendaal F,
Brecht JG, et al. Venous thromboembolism (VTE) in Europe. Bertina RM, Sandset PM. Low levels of tissue factor pathway
The number of VTE events and associated morbidity and inhibitor (TFPI) increase the risk of venous thrombosis.
mortality. Thromb Haemost 2007;98:756-64. Blood 2003;101:4387-92.
4. Silverstein RL, Bauer KA, Cushman M, Esmon CT, Ershler 28. Espana F, Medina P, Navarro S, Estelles A, Aznar J. Inherited
WB, Tracy RP. Venous thrombosis in the elderly: more ques- abnormalities in the protein C activation pathway.
tions than answers. Blood 2007;110:3097-101. Pathophysiol Haemost Thromb 2002;32:241-4.
5. Jordan FL, Nandorff A. The familial tendency in thrombo- 29. Saposnik B, Lesteven E, Lokajczyk A, Esmon CT, Aiach M,
embolic disease. Acta Med Scand 1956;156:267-75. Gandrille S. Alternative mRNA is favored by the A3 haplo-
6. Crawley JT, Lane DA. The haemostatic role of tissue factor type of the EPCR gene PROCR and generates a novel soluble
pathway inhibitor. Arterioscler Thromb Vasc Biol 2008; 28: form of EPCR in plasma. Blood 2008;111:3442-51.
233-42. 30. Dahlbäck B, Carlsson M, Svensson PJ. Familial thrombophil-

| 264 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

ia due to a previously unrecognized mechanism character- mation: a new genetic mechanism contributing to hereditary
ized by poor anticoagulant response to activated protein C: thrombophilia. Nat Genet 2001;28:389-92.
prediction of a cofactor to activated protein C. Proc Natl 36. Rosendaal FR, Doggen CJ, Zivelin A, Arruda VR, Aiach M,
Acad Sci USA 1993;90:1004-8. Siscovick DS, et al. Geographic distribution of the 20210 G
31. Bertina RM, Koeleman BPC, Koster T, Rosendaal FR, Dirven to A prothrombin variant. Thromb Haemost 1998;79:706-8.
RJ, De Ronde H, et al. Mutation in blood coagulation factor 37. Jick H, Slone D, Westerholm B, Inman WH, Vessey MP,
V associated with resistance to activated protein C. Nature Shapiro S, et al. Venous thromboembolic disease and ABO
1994;369:64-7. blood type. A cooperative study. Lancet 1969;1:539-42.
32. Rosendaal FR, Koster T, Vandenbroucke JP, Reitsma PH. 38. Koster T, Blann AD, Briët E, Vandenbroucke JP, Rosendaal
High risk of thrombosis in patients homozygous for factor V FR. Role of clotting factor VIII in effect of von Willebrand
Leiden (activated protein C resistance). Blood 1995;85:1504- factor on occurrence of deep-vein thrombosis. Lancet 1995;
8. 345:152-5.
33. Zivelin A, Griffin JH, Xu X, Pabinger I, Samama M, Conard 39. Preston AE, Barr A. The Plasma Concentration of Factor Viii
J, et al. A single genetic origin for a common caucasian risk in the Normal Population. Ii. The Effects of Age, Sex and
factor for venous thrombosis. Blood 1997;89:397-402. Blood Group. Br J Haematol 1964;10:238-45.
34. Poort SR, Rosendaal FR, Reitsma PH, Bertina RM. A com- 40. Smith NL, Hindorff LA, Heckbert SR, Lemaitre RN,
mon genetic variation in the 3'-untranslated region of the Marciante KD, Rice K, et al. Association of genetic variations
prothrombin gene is associated with elevated plasma pro- with nonfatal venous thrombosis in postmenopausal
thrombin levels and an increase in venous thrombosis. Blood women. JAMA 2007;297:489-98.
1996; 88:3698-703. 41. Bezemer ID, Bare LA, Doggen CJ, Arellano AR, Tong C,
35. Gehring NH, Frede U, Neu-Yilik G, Hundsdoerfer P, Vetter B, Rowland CM, et al. Gene variants associated with deep vein
Hentze MW, et al. Increased efficiency of mRNA 3' end for- thrombosis. JAMA 2008;299:1306-14.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 265 |
Thrombosis

Venous thrombosis: from bench to bedside and back

T. Baglin A B S T R A C T

The laboratory has a critical role in the diagnosis and management of venous thromboembolism.
Department of Haematology, Increasingly, diagnosis is dependent on diagnostic algorithms that incorporate D-dimer measurement.
Addenbrooke’s NHS Trust,
Cambridge, UK Monitoring and adjusting the intensity of anticoagulation determines efficacy and safety, and recent
studies indicate that the quality of control of oral anticoagulant therapy determines the long-term risk
of post thrombotic syndrome. The identification of heritable and acquired thrombophilic defects has
helped us to understand the mechanism of venous thromboembolism and appreciate the importance
Hematology Education: of gene-environment interaction, although testing for a limited number of heritable thrombophilic
the education program for the
defects has not been shown to have clinical utility so far. Recent studies indicate that measurement
annual congress of the European
Hematology Association of the global activity of the coagulation system, either using biomarkers or measuring the thrombin
generating potential, may have useful clinical predictive value for recurrent thrombosis.
2009;3:266-270 Pharmacogenetic studies incorporating rapid genotyping may improve anticoagulant therapy with
vitamin K antagonists. Finally, whilst a simple dichotomous testing strategy for heritable throm-
bophilic defects has not been shown to have useful clinical predictive value, future assessment of the
intermediate phenotype by global coagulation tests combined with genome wide mutation and SNP
detection may provide a complimentary approach to the quantification of both thrombotic and bleed-
ing risks.

Introduction tive approach to reliance on imaging tech-


Venous thrombosis (VT), or venous niques alone for diagnosis and decision mak-
thromboembolism (VTE), comprises deep ing in suspected cases of VTE has been
vein thrombosis (DVT) with or without adopted. This relies on integrated strategies
symptomatic pulmonary embolus (PE). The that incorporate clinical assessment and
incidence of a first episode of VT is 1.5 per measurement of D-dimer.5 The emphasis of
1000 person-years,1 with a per-person life- methods is on high sensitivity and the safe
time incidence of 5%.2 The clinical utility of exclusion of VTE in a significant number of
the atology laboratory has advanced signifi- patients, thus reducing the need for imaging.
cantly with laboratory tests having an For measurement of D-dimer, fast single
increasing clinical utility in diagnosis, moni- samples assays are now available, producing
toring and prognosis (clinical utility is results in less than 30 minutes with sensitiv-
defined as the likelihood that a test will lead ity in excess of 95%. For high sensitivity
to an improved health outcome). Venous assays, the negative predictive value (NPV) is
thrombosis is a multicausal disease. close to 100% and for intermediate sensitiv-
Understanding the genetic risk factors and ity assays, the NPV is close to 100% when
gene-environment interactions is key to the assay is combined with a pre-test proba-
understanding why an individual develops bility score.6
thrombosis at a specific point in time.3 The The cut-off level used for exclusion of VTE
risk factors for venous thrombosis and arte- is not the upper limit of normal in a healthy
rial atherothrombosis overlap but the mate- population. Rather, each assay has a desig-
rial contribution of each differs between the nated cut-off for VTE exclusion. For some
two diseases. Hypercoagulability resulting assays, the cut-off has been validated in
from variation in the genetic control of management studies. The specificity of
blood coagulation produces a greater materi- D-dimer is inevitably low because D-dimer
al contribution to venous thrombosis. levels are elevated by infection, inflamma-
However, it is becoming increasingly recog- tion, cancer, surgery, trauma, atherosclerosis,
nised that both genetic and acquired risk fac- bleeding and pregnancy. Consequently, the
tors are common denominators for venous specificity is lower in certain populations,
and arterial thrombosis.4 including the elderly and hospitalised
patients. The Vidas® D-dimer is one of the
Diagnosis most extensively evaluated assays and con-
As VTE is common, diagnostic clinical sus- sistently produces a 100% NPV for either
picion is high and consequently the preva- DVT or PE regardless of the pre-test proba-
lence of the disease in suspected cases is low bility score.6 An extensive meta-analysis
at about 10% and 20%. Because of cost and completed in 2007 calculated assay perform-
a high number of negative tests, an alterna- ance from 184 articles, including 328

| 266 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

D-dimer test evaluations, and showed that automated Pharmacogenetics


quantitative ELISAs and turbidometric assays generally The aim of pharmacogenetic-based dosing of oral
have a higher sensitivity than whole blood agglutina- vitamin K antagonists (VKA) is to improve safety and
tion assays.7 effectiveness. Warfarin induction, rather than mainte-
In patients with previous DVT or suspected PE and a nance therapy, is the time when the INR is most likely
suspected recurrence a negative high sensitivity to be out of range and when the risk of thrombosis and
D-dimer assay result safely excludes recurrence.8,9 bleeding is greatest.20,21 Previously discussed, early sub-
therapeutic anticoagulation also increases the risk of
Monitoring recurrent VTE and the development of PTS.
Heparin Common single nucleotide polymorphisms (SNPs)
The usual treatment of VTE requires therapeutic anti- in CYP2C9 genes are associated with altered kinetic
coagulation, with additional thrombolysis in up to 5% activity of CYP2C9 with lower warfarin maintenance
of patients presenting with PE. Immediate heparin ther- dose requirements. Two common SNPs in the vitamin
apy results in a low risk of fatal PE and progression of K epoxide reductase gene (VKORC1 6853 and -1639)
DVT.10 A therapeutic APTT can be achieved quicker by correlate with the dose of warfarin required to achieve
commencing UFH according to the patient’s weight.11 therapeutic anticoagulation, as measured by the INR.22
However, with doses of UFH exceeding 30,000 units In patients with mutant alleles, there is an increased
per 24 hours, it is now uncertain if continued adjust- risk of overanticoagulation and bleeding during induc-
ment of heparin dose in response to the APPT improves tion of therapy.23-26 The influence of mutant alleles on
safety and efficacy.12 Nevertheless, current guidelines overanticoagulation and bleeding during long term
recommend monitoring and dose adjustment of thera- therapy is less.27,28 Studies of the combined effect of
py with UFH.13,14 CYP2C9 and VKORC1 polymophisms indicate that
In many countries, UFH has been superseded by low approximately 50-70% of the warfarin dose require-
molecular weight heparin (LMWH). Treatment with ment is determined by these genetic variants but a
LMWH is at least as effective as UFH, and low risk prospective randomized trial of genotype-guided ver-
patients can be treated as outpatients.14 The APTT is rel- sus standard warfarin dosing in patients initiating war-
atively insensitive to LMWH. Anti-Xa activity can be farin did not demonstrate a difference in percentage of
used to monitor LMWH but there are significant limita- INRs out of range.29 In a recent study comparing clini-
tions.13 It is a major advantage of LMWH that therapeu- cal and pharmacogenetic induction algorithms in
tic anticoagulation does not require routine monitoring patients undergoing orthopedic surgery the time in
or dose adjustment. However, in patients with renal fail- therapeutic range, measured by linear interpolation,
ure, pregnancy and young children monitoring with an was higher in the pharmacogenetic cohort than the
anti-Xa assay is advised. A target peak anti-Xa level of clinical cohort and this was associated with fewer
1.0-2.0 IU/mL is suggested for once daily subcutaneous adverse clinical events.30 Commercial platforms for
administration and 0.6-1.0 IU/mL for twice daily.14 rapid genotyping are now available.31 Further studies
will be needed to clarify the clinical utility of pharma-
Oral vitamin K antagonists cogenetic testing for induction and maintenance thera-
Continued treatment with an oral vitamin K antago- py with oral VKA.32
nist (VKA), such as warfarin, will prevent more than
95% of recurrent episodes of VTE.15,16 VKA therapy is Prognosis
monitored by the International Normalised Ratio (INR), After a first episode of VTE, patients are 40 times
with a target INR of 2.5 for the majority of patients. The more likely to suffer a further event compared with pre-
risk of VTE recurrence during treatment is higher when viously unaffected individuals.33 Recurrent DVT or PE is
the INR is subtherapeutic.16 It has recently been demon- associated with a high case-fatality rate and an
strated that achievement of a sustained intensity of increased likelihood of PTS and pulmonary hyperten-
early anticoagulation is required to reduce the risk of sion, respectively. Therefore, secondary prevention of
post thrombotic syndrome (PTS),17 and possibly also VTE significantly reduces the burden of disease.
prevent recurrent VTE after treatment is stopped.18 Point However, recurrent VTE is only prevented for as long as
of care testing and patient self-management offer anticoagulation is continued and so anticoagulation
opportunities for improved anticoagulant care in some must be continued indefinitely in patients at high risk.
locations. In view of the risk of anticoagulant-related bleeding,
Van Dongen et al. reported that patients who have an there is a need to improve risk stratification for recur-
INR less than 2.0 for more than 50% of the time are rent VTE, and hence target long-term therapy at
almost three-times as likely to develop PTS after DVT.17 patients considered to be at greatest risk. Similarly,
After adjustment, the single most important risk factor accurate individualised estimates of anticoagulant-relat-
for PTS was recurrent ipsilateral DVT but there was ed bleeding are required in order to estimate the benefit
almost a 3-fold increased risk when more than 50% of and risk trade-off with continued anticoagulation. The
the time was spent with an INR less than 2.0. Whilst risk of recurrent VTE in an individual can be determined
ipsilateral recurrence was the stronger risk, only 7% of from a variety of interacting clinical, laboratory and
patients suffered ipsilateral recurrence whilst 30% of imaging results. It is becoming apparent that measure-
the time was spent below an INR of 2.0. A relationship ment of the global activity of the coagulation system,
between subtherapeutic anticoagulation and the devel- using either biomarkers34 or measuring the thrombin
opment of PTS was reported in a retrospective analysis generating potential,35-38 may have useful clinical predic-
but VKA therapy was not standardised by the INR.19 tive value for VTE recurrence.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 267 |
14th Congress of the European Hematology Association

Hypercoagulability (D-dimer and thrombin generation) common in unselected patients and their relatives,
One month after completion of treatment of a first despite the fact that clinical utility was unproven.54 It is
episode of VTE, 40% of patients have increased levels now apparent that:
of D-dimer and these patients are at significantly greater (i) recurrence on treatment is not more likely in unse-
risk of recurrence.34,39 Measurement of D-dimer levels lected patients with heritable thrombophilia;55
following cessation of anticoagulant therapy has a high (ii) testing for heritable thrombophilia does not use-
negative predictive value (NPV) for recurrent venous fully predict likelihood of recurrence after stopping anti-
thrombosis with 96% of patients with a low D-dimer coagulant treatment in unselected patients;56,57
being recurrence free at 2 years.34 The positive predic- (iii) testing for inherited thrombophilia has not been
tive value (PPV) has less clinical utility. In patients with shown to prevent VTE recurrence;58
a first episode of unprovoked venous thrombosis and an (iv) systematic review of the risk of recurrent venous
elevated D-dimer after stopping anticoagulant therapy, thromboembolism in patients heterozygous for the fac-
there is still a likelihood of being free from recurrence of tor V Leiden has shown a modest risk not justifying life
approximately 90% at 2 years.34 Therefore, it is debat- long anticoagulation;59
able whether a high D-dimer result alone is an indica- (v) in patients with deficiency of a natural anticoagu-
tion for prolonged anticoagulation.40,41 Patients at high lant (antithrombin, protein C, protein S deficiency), the
risk of recurrence might be better identified by a combi- risk of recurrence is uncertain but relative risks of recur-
nation of clinical and laboratory factors.42,43 Furthermore, rence in relation to deficiency of a natural anticoagulant
the predictive value of D-dimer may be improved by appear to be less than 2.0 in unselected patients;56,57,60
using different cut-offs according to patient characteris- (vi) in young selected patients, the presence of defi-
tics, including age.44 Additional cohort and management ciency of antithrombin, protein C or protein S predicts
studies are still required to determine the clinical utility a high rate of recurrence61 but selection criteria need to
of D-dimer testing, including the value of measurement be defined;
during anticoagulant treatment. (vii) cohort studies have demonstrated a low risk of
D-dimer measurement reflects in vivo thrombin gener- primary events in prospectively followed affected
ation that has occurred. Potentially complimentary to asymptomatic relatives of patients with the factor V
this measurement is analysis of the tissue factor-throm- Leiden or prothrombin mutations.62
bin response coupling mechanism by measurement of Consequently indiscriminate testing for heritable
the in vitro thrombin generating capacity.45-47 This indi- thrombophilia is not mandatory and decisions regarding
cates how much relative thrombin an individual may duration of anticoagulation (lifelong or not) should be
generate in response to a pre-determined stimulus. made with reference to whether or not an episode of
Several recent studies have demonstrated a higher rate VTE was provoked and the presence of other risk fac-
of recurrent VTE in patients with increased thrombin tors, regardless of whether a heritable thrombophilic
generation after unprovoked VTE.35-38 Interestingly, D- defect is present.
dimer and the thrombin generating potential are inde-
pendent predictors of risk of recurrence35,36 suggesting Laboratory testing for acquired thrombophilia
they may be measuring different aspects of hypercoag- Antiphospholipid syndrome (APS) is diagnosed when
ulability. Future studies will evaluate the clinical predic- thrombosis or pregnancy loss occurs in association with
tive value of these measurements in conjunction with positive laboratory tests for antiphospholipid antibody.
clinical risk factors. In contrast to heritable thrombophilia, thrombosis in
A strong association between a short APTT and the patients with APS commonly occurs in the arterial, as
risk of VTE is well recognised.48 The Activated throm- well as venous circulation. Whilst there is a higher rate
boplastin Time (APTT) is a simple global assay of of recurrent VTE in patients with APS, the risk-benefit
thrombin generation, albeit of limited physiological rel- of long-term anticoagulation has not yet been clarified.
evance. Nevertheless two studies have demonstrated an Therefore, it is recommended that the intensity and
association between short APTT values and recurrent duration of treatment should be determined on an indi-
VTE.49,50 However, after correction for levels of factors vidual basis, taking into account the presence of addi-
VIII, IX and XI there was no independent predictive tional risk is factors, the severity of the presenting event
value. High factor VIII and IX levels have previously and the risk of bleeding on warfarin, rather than just the
been reported as independent predictors of VTE recur- diagnosis of APS.63
rence.51,52 Paroxysmal nocturnal oglobinuria (PNH) is associated
with venous and arterial thrombosis with the greatest
Genetic testing risk is in patients with large PNH clones (PNH granulo-
Heritable thrombophilia describes an inherited ten- cytes >50%, 44% versus 5%).64 Oral anticoagulation
dency for venous thrombosis, with or without associat- effectively prevents thrombosis and long-term anticoag-
ed pulmonary embolus. In addition to case-control stud- ulation with a target INR of 2.5 is recommended.65
ies, family pedigree studies have reported a risk of An acquired point mutation (1849G>T, JAK2V617F) is
venous thrombosis in association with heritable throm- present in the majority of Ph-negative myeloprolifera-
bophilia in thrombosis-prone families. However, the tive disorders (MPD).66 Thrombosis is the main cause of
reported rates of thrombosis in these studies are consid- mortality and morbidity in MPD and age over 60 years
ered to be overestimates of the true risk due to selection is an additional risk factor. The contribution of co-exis-
bias. Nevertheless, a causal link between heritable tent heritable thrombophilia to thrombosis risk has not
thrombophilia and venous thrombosis is unequivocal.3,53 yet been determined. In patients with MPD, the degree
In the 1980s and 1990s, thrombophilia testing became of thrombocytosis does not predict thrombosis risk.67

| 268 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

This is also the case, even in essential thrombocytia.68 Thrombotic events during oral anticoagulant treatment:
results of the inception-cohort, prospective, collaborative
This does not exclude an effect of platelets, independent ISCOAT study: ISCOAT study group (Italian Study on
of the actual count, on thrombosis risk. An elevated Complications of Oral Anticoagulant Therapy). Thromb
white count is associated with thrombosis risk69 which Haemost 1997;78:1438-43.
may relate to the observed reduction in thrombosis risk 17. van Dongen CJ, Prandoni P, Frulla M, Marchiori A, Prins MH,
Hutten BA. Relation between quality of anticoagulant treat-
with cytoreductive therapy. There is conflicting evi- ment and the development of the postthrombotic syndrome. J
dence regarding the association between JAK2V617F Thromb Haemost 2005;3:939-42.
and thrombois risk in MPD patients and the association 18. Palareti G, Legnani C, Cosmi B, Guazzaloca G, Cini M,
Mattarozzi S. Poor anticoagulation quality in the first 3
requires clarification. months after unprovoked venous thromboembolism is a risk
factor for long-term recurrence. J Thromb Haemost 2005;
Conclusions 3:955-61.
19. Ziegler S, Schillinger M, Maca TH, Minar E. Post-thrombotic
The association between the bench and bedside is a syndrome after primary event of deep venous thrombosis 10
two-way association. High quality clinical care is to 20 years ago. Thromb Res 2001;101:23-33.
dependent on a high standard of laboratory practice and 20. Beyth RJ, Quinn L, Landefeld CS. A multicomponent interven-
tion to prevent major bleeding complications in older patients
clinical practice informs research and development in receiving warfarin. A randomized, controlled trial. Ann Intern
the laboratory. Given the importance of laboratory Med2000;133:687-95.
results for diagnosis and management, continued satis- 21. Douketis JD, Foster GA, Crowther MA, Prins MH, Ginsberg
factory performance in internal quality control and JS. Clinical risk factors and timing of recurrent venous throm-
boembolism during the initial 3 months of anticoagulant ther-
external quality assurance is necessary to prevent apy. Arch Intern Med 2000;160:3431-6.
adverse clinical outcomes. 22. Gage BF. Pharmacogenetics-based coumarin therapy.
Hematology Ame Soc Hematol Educ Prog 2006:467-73.
23. Higashi MK, Veenstra DL, Kondo LM, Wittkowsky AK,
Srinouanprachanh SL, Farin FM, et al. Association between
References CYP2C9 genetic variants and anticoagulation-related out-
comes during warfarin therapy. J Am Med Assoc 2002; 287:
1. Naess IA, Christiansen SC, Romundstad P, Cannegieter SC, 1690-8.
Rosendaal FR, Hammerstrom J. Incidence and mortality of 24. Margaglione M, Colaizzo D, D'Andrea G, Brancaccio V,
venous thrombosis: a population-based study. J Thromb Ciampa A, Grandone E, et al. Genetic modulation of oral anti-
Haemost 2007;5:692-9. coagulation with warfarin. Thromb Haemost 2000;84:775-8.
2. Silverstein MD, Heit JA, Mohr DN, Petterson TM, O'Fallon 25. Visser LE, van Schaik RH, van Vliet M, Trienekens PH, De
WM, Melton LJ. Trends in the incidence of deep vein throm- Smet PA, Vulto AG, et al. The risk of bleeding complications
bosis and pulmonary embolism: a 25-year population-based in patients with cytochrome P450 CYP2C9*2 or CYP2C9*3
study. Arch Intern Med 1998;158:585-93. alleles on acenocoumarol or phenprocoumon. Thromb
3. Rosendaal FR. Venous thrombosis: a multicausal disease. Haemost 2004;92:61-6.
Lancet 1999;353:1167-73. 26. Voora D, Eby C, Linder MW, Milligan PE, Bukaveckas BL,
4. Agnelli G, Becattini C. Venous thromboembolism and athero- McLeod HL, et al. Prospective dosing of warfarin based on
sclerosis: common denominators or different diseases? J cytochrome P-450 2C9 genotype. Thromb Haemost 2005; 93:
Thromb Haemost 2006;4:1886-90. 700-5.
5. Wells PS. Integrated strategies for the diagnosis of venous 27. Aithal GP, Day CP, Kesteven PJ, Daly AK. Association of poly-
thromboembolism. J Thromb Haemost 2007;5 Suppl 1:41-50. morphisms in the cytochrome P450 CYP2C9 with warfarin
6. Righini M, Perrier A, De Moerloose P, Bounameaux H. D- dose requirement and risk of bleeding complications. Lancet
Dimer for venous thromboembolism diagnosis: 20 years later. 1999;353:717-9.
J Thromb Haemost 2008;6:1059-71. 28. Taube J, Halsall D, Baglin T. Influence of cytochrome P-450
7. Di Nisio M, Squizzato A, Rutjes AW, Buller HR, Zwinderman CYP2C9 polymorphisms on warfarin sensitivity and risk of
AH, Bossuyt PM. Diagnostic accuracy of D-dimer test for over-anticoagulation in patients on long-term treatment.
exclusion of venous thromboembolism: a systematic review. J Blood 2000;96:1816-9.
Thromb Haemost 2007;5:296-304. 29. Anderson JL, Horne BD, Stevens SM, Grove AS, Barton S,
8. Le Gal G, Righini M, Roy PM, Sanchez O, Aujesky D, Perrier Nicholas ZP, et al. Randomized trial of genotype-guided ver-
A, et al. Value of D-dimer testing for the exclusion of pul- sus standard warfarin dosing in patients initiating oral antico-
monary embolism in patients with previous venous thrombo- agulation. Circulation 2007;116:2563-70.
embolism. Arch Intern Med 2006;166:176-80. 30. Lenzini PA, Grice GR, Milligan PE, Dowd MB, Subherwal S,
9. Rathbun SW, Whitsett TL, Raskob GE. Negative D-dimer Deych E, et al. Laboratory and clinical outcomes of pharmaco-
result to exclude recurrent deep venous thrombosis: a manage- genetic vs. clinical protocols for warfarin initiation in orthope-
ment trial. Ann Intern Med 2004;141:839-45. dic patients. J Thromb Haemost 2008;6:1655-62.
10. Douketis JD, Kearon C, Bates S, Duku EK, Ginsberg JS. Risk of 31. King CR, Porche-Sorbet RM, Gage BF, Ridker PM, Renaud Y,
fatal pulmonary embolism in patients with treated venous Phillips MS, et al. Performance of commercial platforms for
thromboembolism. Am Med Assoc 1998;279:458-62. rapid genotyping of polymorphisms affecting warfarin dose.
11. Raschke RA, Reilly BM, Guidry JR, Fontana JR, Srinivas S. The Am J Clin Path 2008;129:876-83.
weight-based heparin dosing nomogram compared with a 32. Flockhart DA, O'Kane D, Williams MS, Watson MS, Flockhart
"standard care" nomogram. A randomized controlled trial. DA, Gage B, et al. Pharmacogenetic testing of CYP2C9 and
Ann Intern Med 1993;119:874-81. VKORC1 alleles for warfarin. Genet Med 2008;10:139-50.
12. Anand SS, Bates S, Ginsberg JS, Levine M, Buller H, Prins M, 33. Kearon C. Natural history of venous thromboembolism.
et al. Recurrent venous thrombosis and heparin therapy: an Circulation 2003;107 23 Suppl 1:I22-30.
evaluation of the importance of early activated partial throm- 34. Verhovsek M, Douketis JD, Yi Q, Shrivastava S, Tait RC,
boplastin times. Arch Intern Med 1999;159:2029-32. Baglin T, et al. Systematic review: D-dimer to predict recurrent
13. Baglin T, Barrowcliffe TW, Cohen A, Greaves M. Guidelines disease after stopping anticoagulant therapy for unprovoked
on the use and monitoring of heparin. Br J Haematol 2006; venous thromboembolism. Ann Intern Med 2008;149:481-90.
133:19-34. 35. Besser M, Baglin C, Luddington R, van Hylckama Vlieg A,
14. Kearon C, Kahn SR, Agnelli G, Goldhaber S, Raskob GE, Baglin T. High rate of unprovoked recurrent venous thrombo-
Comerota AJ. Antithrombotic therapy for venous throm- sis is associated with high thrombin-generating potential in a
boembolic disease: American College of Chest Physicians prospective cohort study. J Thromb Haemost 2008;6:1720-5.
Evidence-Based Clinical Practice Guidelines (8th Edition). 36. Eichinger S, Hron G, Kollars M, Kyrle PA. Prediction of recur-
Chest 2008;133:454S-545S. rent venous thromboembolism by endogenous thrombin
15. Hutton B, Prins MH. Duration of treatment with vitamin K potential and D-dimer. Clin Chem 2008;54:2042-8.
antagonists in symptomatic venous thromboembolism. In: 37. Hron G, Eichinger S, Weltermann A, Minar E, Bialonczyk C,
The Cochrane database of Systematic Reviews 2000: The Hirschl M, et al. Family history for venous thromboembolism
Cochrane Collaboration; 2000. and the risk for recurrence. Am J Med 2006;119:50-3.
16. Palareti G, Manotti C, A DA, Pengo V, Erba N, Moia M, et al. 38. Tripodi A, Legnani C, Chantarangkul V, Cosmi B, Palareti G,

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 269 |
14th Congress of the European Hematology Association

Mannucci PM. High thrombin generation measured in the Mackinnon B, et al. Influence of thrombophilia on risk of
presence of thrombomodulin is associated with an increased recurrent venous thromboembolism while on warfarin:
risk of recurrent venous thromboembolism. J Thromb Results from a randomized trial. Blood 2008;112:4432-6.
Haemost 2008;6:1327-33. 56. Baglin T, Luddington R, Brown K, Baglin C. Incidence of recur-
39. Palareti G, Cosmi B, Legnani C, Tosetto A, Brusi C, Iorio A, et rent venous thromboembolism in relation to clinical and
al. D-dimer testing to determine the duration of anticoagula- thrombophilic risk factors: prospective cohort study. Lancet.
tion therapy. New Engl J Med 2006;355:1780-9. 2003;362:523-6.
40. Baglin T. Value of D-dimer testing to decide duration of anti-
coagulation after deep vein thrombosis: not yet. J Thromb 57. Christiansen SC, Cannegieter SC, Koster T, Vandenbroucke JP,
Haemost 2006;4:2530-2. Rosendaal FR. Thrombophilia, clinical factors, and recurrent
41. Cosmi B. Value of D-dimer testing to decide duration of anti- venous thrombotic events. J Am Med Assoc 2005;293:2352-
coagulation after deep vein thrombosis: yes. J Thromb 61.
Haemost 2006;4:2527-9. 58. Coppens M, Reijnders JH, Middeldorp S, Doggen CJ,
42. Baglin T, Palmer C, Luddington R, Baglin C. Unprovoked Rosendaal FR. Testing for inherited thrombophilia does not
recurrent venous thrombosis: prediction by D-dimer and clin- reduce recurrence of venous thrombosis. J Thromb Haemost
ical risk factors. J Thromb Haemost 2008;6:577-82. 2008;6:1474-7.
43. Rodger MA, Kahn SR, Wells PS, Anderson DA, Chagnon I, Le 59. Ho WK, Hankey GJ, Quinlan DJ, Eikelboom JW. Risk of recur-
Gal G, et al. Identifying unprovoked thromboembolism rent venous thromboembolism in patients with common
patients at low risk for recurrence who can discontinue antico- thrombophilia: a systematic review. Arch Intern Med 2006;
agulant therapy. Can Med Assoc J 2008;179:417-26. 166:729-36.
44. Legnani C, Palareti G, Cosmi B, Cini M, Tosetto A, Tripodi A. 60. De Stefano V, Simioni P, Rossi E, Tormene D, Za T, Pagnan A,
Different cut-off values of quantitative D-dimer methods to
predict the risk of venous thromboembolism recurrence: a et al. The risk of recurrent venous thromboembolism in
post-hoc analysis of the PROLONG study. Haematologica patients with inherited deficiency of natural anticoagulants
2008;93:900-7. antithrombin, protein C and protein S. Haematologica 2006;
45. Baglin T. The measurement and application of thrombin gen- 91:695-8.
eration. Br J Haematol 2005;130:653-61. 61. Lijfering WM, Brouwer JL, Veeger NJ, Bank I, Coppens M,
46. Hemker HC. Recollections on thrombin generation. J Thromb Middeldorp S, et al. Selective testing for thrombophilia in
Haemost 2008;6:219-26. patients with first venous thrombosis. Results from a retro-
47. Mann KG, Brummel-Ziedins K, Undas A, Butenas S. Does the spective family cohort study on absolute thrombotic risk for
genotype predict the phenotype? Evaluations of the hemosta- currently known thrombophilic defects in 2479 relatives.
tic proteome. J Thromb Haemost 2004;2:1727-34. Blood 2009;[Epub ahead of print].
48. Tripodi A, Chantarangkul V, Martinelli I, Bucciarelli P, 62. Langlois NJ, Wells PS. Risk of venous thromboembolism in rel-
Mannucci PM. A shortened activated partial thromboplastin atives of symptomatic probands with thrombophilia: a sys-
time is associated with the risk of venous thromboembolism. tematic review. Thromb Haemost 2003;90:17-26.
Blood 2004;104:3631-4. 63. Greaves M, Cohen H, MacHin SJ, Mackie I. Guidelines on the
49. Hron G, Eichinger S, Weltermann A, Quehenberger P,
Halbmayer WM, Kyrle PA. Prediction of recurrent venous investigation and management of the antiphospholipid syn-
thromboembolism by the activated partial thromboplastin drome. Br J Haematol 2000;109:704-15.
time. J Thromb Haemost 2006;4:752-6. 64. Hall C, Richards S, Hillmen P. Primary prophylaxis with war-
50. Legnani C, Mattarozzi S, Cini M, Cosmi B, Favaretto E, farin prevents thrombosis in paroxysmal nocturnal hemoglo-
Palareti G. Abnormally short activated partial thromboplastin binuria (PNH). Blood 2003;102:3587-91.
time values are associated with increased risk of recurrence of 65. Baglin TP, Keeling DM, Watson HG. Guidelines on oral anti-
venous thromboembolism after oral anticoagulation with- coagulation (warfarin): third edition--2005 update. Br J
drawal. Br J Haematol 2006;134:227-32. Haematol 2006;132:277-85.
51. Kyrle PA, Minar E, Hirschl M, Bialonczyk C, Stain M, 66. Austin SK, Lambert JR. The JAK2 V617F mutation and throm-
Schneider B, et al. High plasma levels of factor VIII and the risk bosis. B J Haematol 2008;143:307-20.
of recurrent venous thromboembolism. N Engl J Med 2000; 67. Di Nisio M, Barbui T, Di Gennaro L, Borrelli G, Finazzi G,
343:457-62. Landolfi R, et al. The haematocrit and platelet target in poly-
52. Weltermann A, Eichinger S, Bialonczyk C, Minar E, Hirschl M, cythemia vera. Br J Haematol 2007;136:249-59.
Quehenberger P, et al. The risk of recurrent venous throm-
68. Lengfelder E, Hochhaus A, Kronawitter U, Hoche D, Queisser
boembolism among patients with high factor IX levels. J
Thromb Haemost 2003;1:28-32. W, Jahn-Eder M, et al. Should a platelet limit of 600x109/L be
53. Rosendaal FR, Bovill EG. Heritability of clotting factors and used as a diagnostic criterion in essential thrombocythaemia?
the revival of the prothrombotic state. Lancet 2002; 359:638-9. An analysis of the natural course including early stages. Br J
54. Greaves M, Baglin T. Laboratory testing for heritable throm- Haematol 1998;100:15-23.
bophilia: impact on clinical management of thrombotic dis- 69. Landolfi R, Di Gennaro L, Barbui T, De Stefano V, Finazzi G,
ease annotation. Br J Haematol 2000;109:699-703. Marfisi R, et al. Leukocytosis as a major thrombotic risk factor
55. Kearon C, Julian JA, Kovacs MJ, Anderson DR, Wells P, in patients with polycythemia vera. Blood 2007;109:2446-52.

| 270 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Thrombosis

New anticoagulants

A. Kleinjan A B S T R A C T
H.R. Büller
Although the efficacy of low molecular weight heparin and vitamin K antagonists is undisputed,
shortcomings of these anticoagulants prompted the search for new ones. New anticoagulants for the
Department of Vascular Medicine,
Academic Medical Center, prevention of venous thrombosis act upon different stages of the coagulation cascade. Targeting the
Amsterdam, The Netherlands initiation phase is promising, but potential applications are limited. Many factor Xa inhibitors are
under development. Fondaparinux is one of the pentasaccharides, indirect factor Xa inhibitors, which
have been approved for several indications. Rivaroxaban is the direct factor Xa inhibitor in the most
advanced stage of development. Parenteral direct thrombin inhibitors have been approved for limited
Hematology Education: indications. Dabigatran etexilate is the oral direct thrombin inhibitor in phase III stage of development.
the education program for the Inhibitors of factors in the amplification phase of the coagulation could potentially be interesting
annual congress of the European because they do not completely block thrombin generation. Drugs specifically targeted at factor XI are
Hematology Association also being developed. Recombinant activated protein C and thrombomodulin act upon natural antico-
agulant pathways. Because of the arrival of these new anticoagulants, the field of anticoagulant treat-
2009;3:271-275 ment may change drastically within a few years.

Introduction inhibits factor Xa and thrombin. LMWH


Arterial and venous thromboembolic inhibits factor Xa more than thrombin. In
events are frequently encountered and cause case of bleeding, protamine sulphate is an
significant morbidity and mortality. The antidote for unfractioned heparin and to a
introduction of anticoagulants more than 50 lesser extent for LMWH.
years ago meant a significant step forward in Both compounds are highly active and
the therapeutic management of these dis- their efficacy in the prevention and treat-
eases. Anticoagulants currently used for the ment of venous and arterial thrombosis is
prevention of venous thrombosis are low undisputed. Limitations of existing parenter-
molecular weight heparins (LMWH) and al anticoagulants, such as users’ conven-
fondaparinux. Vitamin K antagonists, ience, risk for bleeding and need for dose
LMWH and fondaparinux are the main adjustments, prompted the search for new
drugs for the treatment of symptomatic parenteral anticoagulants. Shortcomings of
venous thrombosis. For the prevention and existing oral anticoagulants, such as the need
treatment of arterial thrombosis anti-platelet for monitoring and bridging therapy and the
drugs and again vitamin K antagonists, bleeding risk stimulated the search for new
LMWH and fondaparinux are the current oral antithrombotic agents. Because of the
drugs of choice. better knowledge of how the coagulation
The FDA approved Vitamin K antagonists cascade functions, specifically targeted drugs
in 1954. These are taken orally and inhibit were developed.1
the synthesis of the vitamin K dependent Here we will review the new anticoagu-
clotting factors (factors II, VII, IX and X). lants for the prevention and treatment of
Because of the unpredictable anticoagulant venous thrombosis, with the coagulation
effect, the intensity has to be monitored, cascade as a guide (Figure 1).
with dose adjustments when necessary.
Vitamin K antagonists need four to five days The coagulation system
until a steady effect has been reached. In The process of clot formation is divided in
case immediate protection is needed, bridg- three parts, which do not occur consecutive-
ing therapy with a parenteral anticoagulant ly but rather simultaneously: primary hemo-
should be employed. The effect of vitamin K stasis involving platelet activation and aggre-
antagonists can be reversed by oral vitamin gation, coagulation cascade activation result-
K and factor concentrates. ing in fibrin formation and fibrinolysis.
Discovered in 1916, unfractionated Venous thrombi are formed under low shear
heparin was the most commonly used par- conditions and are mainly composed of fib-
enteral anticoagulant until recently. Low rin and red blood cells. Therefore, to prevent
molecular weight heparin (LMWH) is a or treat venous thrombosis, targeting the
derivative of unfractioned heparin, which is coagulation cascade is the most appropriate
given subcutaneously once or twice daily. approach.
Both unfractioned heparin and LMWH The coagulation cascade consists of a few
increase the activity of antithrombin, which overlapping stages, in which inactive

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 271 |
14th Congress of the European Hematology Association

enzymes become activated and subsequently activate increase the affinity for antithrombin. Direct factor Xa
the next factor in the cascade (Figure 1). The initiation inhibitors also target factor Xa within the prothrombi-
of coagulation is triggered by tissue factor (TF), which nase complex, while indirect inhibitors only act against
binds factor VIIa and as a complex, activates factor X. free factor Xa.
Activated factor X then converts small amounts of pro-
thrombin to thrombin. These small amounts of throm- Indirect factor Xa inhibitors
bin are sufficient to activate the amplification loops via All indirect factor Xa inhibitors are administered sub-
factors V, VIII, IX and XI. Finally, thrombin converses cutaneously; however, because of the long half-life of
fibrinogen to fibrin. To prevent a hypercoagulable state, some of the compounds, they may also be suitable for
procoagulant activity is regulated by anticoagulant long-term indications.
pathways, such as activated protein C, thrombomod- Fondaparinux is the protype of the pentasaccharides,
ulin, antithrombin and tissue factor pathway inhibitor.2 which are synthetic analogs of a part of the heparin
Theoretically, new anticoagulants can be targeted at molecule, namely the five sugars that bind to
each of these steps; however, not all steps are equally antithrombin. Fondaparinux was first tested in patients
attractive. after hip and knee operations in phase II and III trials. A
meta-analysis of four large phase III trials, including
Classical evaluation of new anticoagulants 7344 patients undergoing hip or knee replacement, con-
The clinical evaluation of new anticoagulant drugs cluded that fondaparinux showed a benefit over enoxa-
follows a typical pattern. First, the new drug is tested in parin without increasing the incidence of clinically rele-
vitro, in animal models and in healthy volunteers. vant bleeding. This beneficial effect of fondaparinux
Thereafter, clinical dose finding trials are set up in was on total venous thromboembolism (VTE), includ-
orthopedic patients to evaluate the efficacy of the novel ing asymptomatic VTE, while rates of symptomatic
compound for the prevention of thrombosis after knee VTE were comparable in most studies.6
or hip replacement. In this group of patients, asympto- The Matisse studies compared fondaparinux to
matic thrombosis occurs frequently and can be easily enoxaparin for the initial treatment of deep venous
detected by venography 7-10 days postoperatively. This thrombosis or pulmonary embolism. Both studies
allows the establishment, relative to control, of the found that fondaparinux was equally safe and effective
most effective dose. Furthermore, a non-safe dose is as enoxaparin or unfractioned heparin for this indica-
also quickly identified. The second group of patients in tion.7,8 Also, in post-operative prevention of venous
which drugs are subsequently evaluated, comprises of thrombo-embolism after general surgery, fondaparinux
patients with symptomatic venous thrombosis. Again, was shown to be effective. Fondaparinux has been
usually initially with dose finding studies, followed by approved for these indications. Fondaparinux has also
large phase III studies. Finally, when the optimum dose been approved in arterial thrombosis, most notably
is known, the drug is tested in patients with or with a acute coronary syndromes.9-11 A phase III trial in patients
risk for arterial thrombosis, such as atrial fibrillation and undergoing bypass surgery is planned.
the acute coronary syndrome. Idraparinux is a similar pentasaccharide with a long
half-life, which makes once weekly administration pos-
Initiation sible and, therefore, was evaluated for the treatment of
Conceptually, targeting TF, factor VIIa or the complex VTE. The Van Gogh DVT and PE trials were phase III
is promising; however, most studies were disappointing trials comparing idraparinux with LMWH or vitamin K
and potential applications are limited. A recombinant antagonists for 3 to 6 months in patients with sympto-
TF-pathway inhibitor named tifacogin was evaluated in matic deep venous thrombosis (DVT) or pulmonary
sepsis patients, after animal studies showed an effect on embolism (PE). In the DVT patients, the rates of recur-
mortality. A large phase III trial in patients with severe rent VTE were similar in both groups, while clinically
sepsis comparing tifacogin to placebo could not demon- relevant bleeds were initially less frequent with idra-
strate an effect of tifacogin on 28-day mortality, while parinux. In the PE patients, however, idraparinux was
there was a significant higher risk of bleeding.3 less effective than LMWH and vitamin K antagonists.12
NAPc2, a protein originally isolated from a hook- The Van Gogh extension study aimed to evaluate the
worm, binds to factor X or Xa, and as a complex, efficacy of idraparinux for an additional 6 months of
inhibits TF bound factor VIIa. Promising results were treatment after patients completed 6 months of treat-
found with recombinant NAPc2 in a phase II dose- ment with idraparinux or a vitamin K antagonist. In this
response trial in patients undergoing elective knee study, idraparinux produced a 73% relative reduction in
replacement surgery and in a phase II study in arterial the frequency of recurrent VTE, but at the cost of a rate
thrombosis. No phase III studies have been set up in of 3.7 % major bleeding. After this study, safety con-
these patient groups so far.4,5 cerns halted further development of idraparinux.13
The development of active site-blocked factor VIIa Biotynalated idraparinux (SSR-126517-E) is derived
has been halted after disappointing results in patients from idraparinux and, therefore, exhibits the same bio-
undergoing elective percutaneous coronary intervention logical charactistics as idraparinux. The added biotin
(PCI). molecule to idraparinux facilitates direct neutralisation
by the infusion of avidin, an egg protein, as was demon-
Factor Xa strated in the EQUINOX trial.14 SSR12517E is now
Many factor Xa inhibitors are under development. undergoing phase III evaluation in patients with symp-
Factor Xa inhibitors are divided into agents, which tomatic PE, after bridging therapy with heparin or
directly target factor Xa and indirect drugs, which LMWH. In addition, this drug is currently being com-

| 272 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

Figure 1. The new anticoagulants and the various levels in the coagulation cascade they act upon.

pared to warfarin for the prevention of stroke in a phase rates. Phase III studies for the treatment of VTE and for
III trial (Borealis study). stroke prevention in patients with atrial fibrillation are
SR 123781A comprises of a pentasaccharide plus a ongoing.
thrombin binding tetrasaccharide and, therefore, has Apixaban is another direct oral Xa inhibitor, currently
both thrombin and Xa inhibiting activity. This drug is in in phase III development: one phase III trial in orthope-
phase II evaluation for patients after knee arthroplasty.15 dic surgery has just been completed and showed prom-
ising results.20 Other phase III studies for the prevention
Direct factor Xa inhibitors of VTE in hospitalized patients, in stroke prevention
A lot of attention is presently going to direct oral Xa and treatment of VTE are currently recruiting patients.
inhibitors. Rivaroxaban is the oral Xa inhibitor in the Results will probably become available within two to
most advanced stage of development. It has a rapid three years. Many other oral direct Xa inhibitors are in
onset of action and a half-life of 5 to 9 hours, which phase II development.21
does not necessitate the use of bridging with parenteral
anticoagulants. Four phase II trials evaluated rivaroxa- Thrombin
ban in patients undergoing knee or hip surgery and The new anticoagulants in this class are direct throm-
showed that rivaroxaban was at least as effective as bin inhibitors. Indirect thrombin inhibitors, such as
enoxaparin, while higher doses of rivaroxaban caused LMWH and heparin who act by stimulating antithrom-
more major bleedings.16 bin, will not be described here.
The RECORD phase III trials compared rivaroxaban Parenteral thrombin inhibitors have been approved
with enoxaparin in the same patient population. All for limited indications. Hirudin and argatroban are reg-
three studies demonstrated that rivaroxaban was signif- istered for the treatment of patients with heparin-
icantly more effective than enoxaparin in the prophy- induced thrombocytopenia, and bivalirudin is licensed
laxis of VTE after knee or hip replacement surgery, with as an alternative to heparin in percutaneous cutaneous
comparable bleeding rates.17-19 Interestingly, rivaroxaban interventions. Two new parenteral direct thrombin
in a dose of 10 mg once daily was started post-opera- inhibitors are in phase II development. The oral direct
tively. thrombin inhibitor, which provided proof of concept,
The evaluation of rivaroxaban for the treatment of was ximelagatran. However, because of liver toxicity,
DVT is in an advanced stage. Phase II studies showed at the drug was removed from the market, after having
least non-inferiority of rivaroxaban compared to been licensed briefly in Europe. Currently, dabigatran
LMWH plus vitamin K antagonists, with low bleeding etexilate is the most advanced in clinical development in

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 273 |
14th Congress of the European Hematology Association

this class of anticoagulants. Phase II studies in patients Natural anticoagulant mechanisms


undergoing hip or knee replacement surgery provided Activated protein C inactivates the cofactors Va and
evidence for the efficacy of dabigatran in this patient VIIIa and therefore, is an other interesting target for
group. After that, three phase III trials were undertaken. new antithrombotics. Activated protein C also has anti-
The RE-MODEL trial included 2076 patients undergo- inflammatory actions, which makes it suitable for use in
ing knee replacement, which were randomized to dabi- sepsis patients, who suffer from both thrombotic and
gatran or enoxaparin.22 The primary outcome, a combi- inflammatory complications. Recombinant activated
nation of symptomatic and asymptomatic VTE and protein C, called drotrecogin α, is already licensed for
total mortality, was not significantly different between use in severe sepsis. This approval was based on a trial
both groups. Also, bleeding rates were comparable. The in 1690 patients with severe sepsis, which compared
RE-NOVATE trial in 3494 patients undergoing hip drotrecogin alfa to placebo and found a 19% reduction
replacement reported efficacy and bleeding rates of in mortality at 28 days. However, the rate of major
dabigatran similar to enoxaparin.23 The RE-MOBILIZE bleeding was higher in the treated group compared to
in patients undergoing knee replacement surgery con- the placebo group (3.5% vs. 2%, p=0.06). After
cluded that dabigatran was inferior to enoxaparin, but approval, two additional phase III trials in adults with
in this trial a higher (North America) dose of enoxaparin sepsis and a low risk of death and in children with sep-
was used compared to the other phase III trials.24 In sis were prematurely halted because of lack of effect
patients with atrial fibrillation, dabigatran was found to with an increased risk of bleeding.32,33
be promising in phase II trials; therefore, the RE-LY trial Recombinant thrombomodulin (ART-123) binds to
in patients with atrial fibrillation was designed. The thrombin and, thereby converts thrombin from a proco-
results of this study will become available soon. The agulant enzyme into a potent activator of protein C. A
RE-COVER and RE-MEDY trials will compare dabiga- phase II dose-finding trial has been reported in patients
tran with warfarin for the treatment and secondary pre- undergoing elective hip surgery. In this study, ART-123
vention of patients with acute symptomatic VTE. seemed to have antithrombotic potential; however, the
real value of ART-123 needs to be determined in a ran-
Amplification phase domized controlled trial.34
Inhibitors of actors in the amplification phase of the
coagulation cascade, mainly mediated through throm- The future of anticoagulant therapy
bin, could potentially be interesting, because they do Although heparin and VKAs have been used for a
not completely block thrombin generation. Therefore, long time and, especially heparin, has been improved in
they theoretically might have fewer bleeding complica- the last ten years, these drugs certainly have drawbacks.
tions.25-28 At these points, anticoagulant therapy can be improved
TB-402 is a partial inhibitor of factor VIII. Phase I by the new anticoagulants. Dabigatran, rivaroxaban and
studies have been completed and a phase II study in apixaban are the drugs closest to implementation into
prophylaxis after total knee replacement is currently clinical practise. The first two have recently been
underway. The results of this study may provide ‘proof licensed for thromboprophylaxis in orthopedic surgery
of principle’ for the efficacy of factor VIII inhibition as a in Europe. However, because of the limited patient
viable anticoagulant. numbers in which these drugs have been tested, unex-
Two different factor IXa inhibitors have been evaluat- pected adverse effects may still arise. The most recent
ed. TTP 889 is an oral active direct factor IXa inhibitor. example is ximelagatran, which first seemed to be very
The FIXIT study, a phase II randomized placebo con- promising, but was withdrawn from the market
trolled study in patients after hip fracture surgery because of unexpected liver toxicity. Another potential
demonstrated that TTP 889 was not effective in reduc- drawback for most newly developed anticoagulants is
ing the incidence of venous thromboembolism. the lack of a specific antidote and the lack of a way to
Strikingly, little bleeding complications were reported. monitor compliance. Also, costs for the health system
The development was subsequently stopped.29 RB006 is will likely rise with these new drugs.
another parenteral factor IXa inhibitor, with a specifical- It is difficult to predict whether LMWH and vitamin
ly designed antidote called RB007. In phase I studies, K antagonists will keep playing an important role in the
RB006 showed rapid anticoagulant activity which could future. In specific subgroups such as cancer patients,
be reversed by RB007.30 Because of the potential for LMWH will probably stay the preferred choice for long-
rapid reversal, this drug is being developed, for example, term treatment and secondary prevention, because of its
for use in cardiac surgery, and a phase II study is ongo- possible advantageous effects on cancer progression.
ing in patients undergoing PCI. These additional advantageous effects remain to be
Lately, factor XI is being investigated as a target for evaluated for the new anticoagulants.
anticoagulant therapy. In mice models, factor XI knock-
out resulted in significantly reduced thrombus forma-
tion, which was more profound than the effect of a high References
dose of heparin, without an increase in bleeding risk. 1. Geerts WH, Bergqvist D, Pineo GF, Heit JA, Samama CM,
Studies in humans show a correlation with high levels Lassen MR, et al. Prevention of venous thromboembolism:
of factor XI with arterial and venous thrombosis, while American College of Chest Physicians Evidence-Based Clinical
Practice Guidelines (8th ed). Chest 2008;1336 Suppl:381S-453S.
this factor is thought to have a relatively small role in 2. Furie B, Furie BC. Mechanisms of thrombus formation. N Engl
normal hemostasis. Presently, different research groups J Med 2008;359:938-49.
are developing drugs specifically targeted at factor XI.31 3. Abraham E, Reinhart K, Opal S, Demeyer I, Doig C,
Rodriguez AL, et al. Efficacy and safety of tifacogin (recombi-

| 274 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

nant tissue factor pathway inhibitor) in severe sepsis: a ran- 20. Buller H, Deitchman D, Prins M, Segers A. Efficacy and safety
domized controlled trial. JAMA 2003;290:238-47. of the oral direct factor Xa inhibitor apixaban for symptomatic
4. Lee A, Agnelli G, Buller H, Ginsberg J, Heit J, Rote W, et al. deep vein thrombosis. The Botticelli DVT dose-ranging study.
Dose-response study of recombinant factor VIIa/tissue factor J Thromb Haemost 2008;6:1313-8.
inhibitor recombinant nematode anticoagulant protein χ2 in 21. Lassen MR, Davidson BL, Gallus A, Pineo G, Ansell J,
prevention of postoperative venous thromboembolism in Deitchman D. The efficacy and safety of apixaban, an oral,
patients undergoing total knee replacement. Circulation 2001; direct factor Xa inhibitor, as thromboprophylaxis in patients
104:74-8. following total knee replacement. J Thromb Haemost 2007;
5. Lee AY, Vlasuk GP. Recombinant nematode anticoagulant pro-
tein χ2 and other inhibitors targeting blood coagulation factor 5:2368-75.
VIIa/tissue factor. J Intern Med 2003;254:313-21. 22. Eriksson BI, Dahl OE, Rosencher N, Kurth AA, van Dijk CN,
6. Turpie AG, Bauer KA, Eriksson BI, Lassen MR. Fondaparinux Frostick SP, et al. Oral dabigatran etexilate vs. subcutaneous
vs enoxaparin for the prevention of venous thromboembolism enoxaparin for the prevention of venous thromboembolism
in major orthopedic surgery: a meta-analysis of 4 randomized after total knee replacement: the RE-MODEL randomized
double-blind studies. Arch Intern Med 2002;162:1833-40. trial. J Thromb Haemost 2007;5:2178-85.
7. Buller HR, Davidson BL, Decousus H, Gallus A, Gent M, 23. Eriksson BI, Dahl OE, Rosencher N, Kurth AA, van Dijk CN,
Piovella F, et al. Subcutaneous fondaparinux versus intra- Frostick SP, et al. Dabigatran etexilate versus enoxaparin for
venous unfractionated heparin in the initial treatment of pul- prevention of venous thromboembolism after total hip
monary embolism. N Engl J Med 2003;349:1695-702. replacement: a randomised, double-blind, non-inferiority trial.
8. Buller HR, Davidson BL, Decousus H, Gallus A, Gent M, Lancet 2007;370:949-56.
Piovella F, et al. Fondaparinux or enoxaparin for the initial 24. Ginsberg JS, Davidson BL, Comp PC, Francis CW, Friedman
treatment of symptomatic deep venous thrombosis: a ran- RJ, Huo MH, et al. Oral thrombin inhibitor dabigatran etexi-
domized trial. Ann Intern Med 2004;140:867-73. late vs North American enoxaparin regimen for prevention of
9. Oldgren J, Wallentin L, Afzal R, Bassand JP, Budaj A, venous thromboembolism after knee arthroplasty surgery. J
Chrolavicius S, et al. Effects of fondaparinux in patients with Arthroplasty 2009;24:1-9.
ST-segment elevation acute myocardial infarction not receiv-
ing reperfusion treatment. Eur Heart J 2008;29:315-23. 25. Gailani D, Renne T. The intrinsic pathway of coagulation: a
10. Yusuf S, Mehta SR, Chrolavicius S, Afzal R, Pogue J, Granger target for treating thromboembolic disease? J Thromb
CB, et al. Effects of fondaparinux on mortality and reinfarction Haemost 2007;5:1106-12.
in patients with acute ST-segment elevation myocardial 26. Doggen CJ, Rosendaal FR, Meijers JC. Levels of intrinsic coag-
infarction: the OASIS-6 randomized trial. JAMA 2006; 295: ulation factors and the risk of myocardial infarction among
1519-30. men: Opposite and synergistic effects of factors XI and XII.
11. Yusuf S, Mehta SR, Chrolavicius S, Afzal R, Pogue J, Granger Blood 2006;108:4045-51.
CB, et al. Comparison of fondaparinux and enoxaparin in 27. Wang X, Smith PL, Hsu MY, Gailani D, Schumacher WA,
acute coronary syndromes. N Engl J Med 2006;354:1464-76. Ogletree ML, et al. Effects of factor XI deficiency on ferric
12. Buller HR, Cohen AT, Davidson B, Decousus H, Gallus AS, chloride-induced vena cava thrombosis in mice. J Thromb
Gent M, et al. Idraparinux versus standard therapy for venous Haemost 2006;4:1982-8.
thromboembolic disease. N Engl J Med 2007;357:1094-104. 28. Wang X, Cheng Q, Xu L, Feuerstein GZ, Hsu MY, Smith PL, et
13. Buller HR, Cohen AT, Davidson B, Decousus H, Gallus AS, al. Effects of factor IX or factor XI deficiency on ferric chloride-
Gent M, et al. Extended prophylaxis of venous thromboem- induced carotid artery occlusion in mice. J Thromb Haemost
bolism with idraparinux. N Engl J Med 2007;357:1105-12. 2005;3:695-702.
14. Buller HR, Destors JM, Gallus AS, Prins MH, Raskob G.
Idrabiotaparinux, a Biotinylated Long-Acting Anticoagulant, 29. Eriksson BI, Dahl OE, Lassen MR, Ward DP, Rothlein R, Davis
in the Treatment of Deep Venous Thrombosis (EQUINOX G, et al. Partial factor IXa inhibition with TTP889 for preven-
Study): Safety, Efficacy, and Reversibility by Avidin. Blood tion of venous thromboembolism: an exploratory study. J
2008. Thromb Haemost 2008;6:457-63.
15. Savi P, Herault JP, Duchaussoy P, Millet L, Schaeffer P, Petitou 30. Chan MY, Rusconi CP, Alexander JH, Tonkens RM,
M, et al. Reversible biotinylated oligosaccharides: a new Harrington RA, Becker RC. A randomized, repeat-dose, phar-
approach for a better management of anticoagulant therapy. J macodynamic and safety study of an antidote-controlled fac-
Thromb Haemost 2008;6:1697-706. tor IXa inhibitor. J Thromb Haemost 2008;6:789-96.
16. Eriksson BI, Borris LC, Dahl OE, Haas S, Huisman MV, Kakkar 31. Lin J, Deng H, Jin L, Pandey P, Quinn J, Cantin S, et al. Design,
AK, et al. A once-daily, oral, direct Factor Xa inhibitor, rivarox- synthesis, and biological evaluation of peptidomimetic
aban (BAY 59-7939), for thromboprophylaxis after total hip inhibitors of factor XIa as novel anticoagulants. J Med Chem
replacement. Circulation 2006;114:2374-81. 2006;49:7781-91.
17. Eriksson BI, Borris LC, Friedman RJ, Haas S, Huisman MV, 32. Abraham E, Laterre PF, Garg R, Levy H, Talwar D, Trzaskoma
Kakkar AK, et al. Rivaroxaban versus enoxaparin for thrombo- BL, et al. Drotrecogin alfa (activated) for adults with severe
prophylaxis after hip arthroplasty. N Engl J Med 2008; sepsis and a low risk of death. N Engl J Med 2005;353:1332-41.
358:2765-75.
33. Bernard GR, Vincent JL, Laterre PF, LaRosa SP, Dhainaut JF,
18. Kakkar AK, Brenner B, Dahl OE, Eriksson BI, Mouret P, Muntz
J, et al. Extended duration rivaroxaban versus short-term Lopez-Rodriguez A, et al. Efficacy and safety of recombinant
enoxaparin for the prevention of venous thromboembolism human activated protein C for severe sepsis. N Engl J Med
after total hip arthroplasty: a double-blind, randomised con- 2001;344:699-709.
trolled trial. Lancet 2008;372:31-9. 34. Kearon C, Comp P, Douketis J, Royds R, Yamada K, Gent M.
19. Lassen MR, Ageno W, Borris LC, Lieberman JR, Rosencher N, Dose-response study of recombinant human soluble thrombo-
Bandel TJ, et al. Rivaroxaban versus enoxaparin for thrombo- modulin (ART-123) in the prevention of venous thromboem-
prophylaxis after total knee arthroplasty. N Engl J Med 2008; bolism after total hip replacement. J Thromb Haemost 2005; 3:
358:2776-86. 962-8.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 275 |
Transfusion

Transfusion policies and practices in alloimmunized


sickle cell patients

J.M. Moulds A B S T R A C T

The alloimmunization rate in patients with sickle cell disease is higher than in most transfused
LifeShare Blood Centers, patients and it appears this is the result of many factors. Clearly increasing patient age and number
Shreveport, LA, USA
of transfusions, as well as being female affect this rate. Limited blood group phenotype matching has
been controversial in the past but is becoming the standard of care in large centers, resulting in
decreased rates of alloimmunization once implemented. More recently, extended genotype matching
Hematology Education: has become available due to advances in molecular technology. High throughput microarrays allow
the education program for the for more complete patient blood group typing, as well as finding matched donors. Genotyping has the
annual congress of the European
added benefit of locating compatible donors for patients with complex antibodies. Since delayed
Hematology Association
transfusion reactions and hyperhemolysis pose a severe risk for SCD patients, genotype matched
2009;3:276-281 donors may provide a safer blood product. However, in some instances of hyperhemolysis, withhold-
ing transfusion or short-term corticosteroid treatment may be best for patient management. The
introduction of molecular genetics into the medical field has not only changed our approach to the
treatment of sickle cell disease (SCD) but promises to significantly improve the transfusion needs of
SCD patients and their quality of life in the future. This manuscript will cover three relevant areas,
including the causes of alloimmunization and its prevention, unusual alloantibody specificities that
may be unrecognized in the SCD patients and delayed transfusion reactions resulting in hyperhemol-
ysis – an often unrecognized syndrome in transfused SCD patients.

Alloimmunization in sickle cell disease Alloantibody frequency and specificity


In the United States, an estimated 80,000 Chronic transfusion, however, carries risk
individuals have been diagnosed with sick- including transmission of infectious disease,
le cell disease (SCD), yet there remains a iron overload and alloimmunization to red
significant lack of funding both for research cell, platelet and HLA antigens. Published
and clinical care.1 This has adversely affect- figures regarding the incidence of red cell
ed the adoption of policies regarding stan- alloimmunization in sickle cell patients vary
dard of care for these patients. Accordingly, greatly ranging from 4-40%.5-9 It is clear that
specialized centers for Sickle Cell research many factors can influence the observed
and treatment have been established and alloimmunization rate, including increasing
are addressing some of the major health age, number of transfusions, and racial dis-
issues of SCD; for example, strokes. parity, as well as the techniques used for
It is well established that strokes during antibody detection. For example, Rh blood
early childhood result in significant morbid- group antibodies are enhanced using
ity and mortality among SCD patients. enzyme treated reagent cells but this is not a
Based on an abnormal transcranial doppler, routine technique in all blood banks. Studies
Adams et al.2 showed that blood transfusion that do not use this technique could under-
was very useful in preventing the first estimate the percent of immunized patients.
stroke among children who were at great- One constant among all of the transfusion
est risk. In fact, the results from the Stroke studies is the antibody specificities that
Prevention Trial in Sickle Cell Anemia develop in these transfused patients. In
(STOP) were so convincing regarding the almost all studies, the most frequent anti-
benefit of red cell transfusion that the study bodies are anti-E, anti-Kell and anti-C.5,10-12
was actually halted before its scheduled This is in part due to the high frequency (35-
completion date. 40%) of the Ro (cDe) blood type in Blacks
A follow-up study, in which transfusions making them at risk to develop anti-C and -
were discontinued, showed a high rate of E. It has been a common assumption that
reversion to abnormal blood-flow velocities the high rate of allloimmunization in SCD
and stroke.3-4 patients is due to racial differences between
Thus, the continued use of red cell trans- patients and donors.10 In fact, some investi-
fusion has become generally accepted for gators have suggested using intra-racial
the treatment of high-risk SCD patients. transfusions for SCD patients to reduce the
exposure rate to foreign blood group anti-

| 276 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

gens.13,14 A study by Noumsi and Moulds,15 comparing


SCD patients in West Africa to the Southern United
States (US), found that anti-E and anti-C occurred with Mali
USA
a similar incidence in Mali where all the donors were
Black (Figure 1). This suggests that donors, Black or
White, must be matched for the Rh antigens in order to
avoid an antibody response. Another study from Sao
Paulo, Brazil reported that their mixed race donor pop-
ulation had a high incidence of the C antigen, and that
the most frequent antibodies in their SCD patients were
antibodies in the Rh and Kell systems.5

Phenotype matching
The argument for and against phenotype matching of
blood donors and patients has been ongoing for the last
20 years but with no consensus policy. Arguments
against matching cite cost as the major drawback.16-17 Figure 1. Comparison of the alloimmunization rate for spe-
Reports from small or single institution studies suggest- cific antibody specificities in SCD patients from Mali and
ed that matching of blood donors to recipient pheno- the USA.
type reduced the incidence of alloimmunization and
that cost was justified by improved clinical response.14,18
However, it was the publication from a large multi-cen-
ter trial that provided the most convincing data for phe- for rapid, high-throughput genotyping of patients and
notype matching.19 In the US, the National Institutes of donors has caused us to re-evaluate cost-related argu-
Health now recommend phenotype matching for multi- ments against phenotype matching. The BloodGen
ply transfused SCD patients (www.nhlbi.nih.gov/health/ project in Europe developed microarrays, not only for
prof/blood/sickle/sc_mngt.pdf) and almost all of the ABO and Rh (including weak D and D variants), but
Comprehensive Sickle Cell Centers (CSCC) routinely also for the other important minor blood groups, includ-
perform patient phenotyping to include ABO, Rh, Kell, ing Kell, Duffy, Kidd, MNSs, Diego, Dombrock and
Duffy, Kidd, Lewis, Lutheran, P1 and the MNS blood Colton. In the US and South America, the bead-chip
group systems.20 Although providing at least C, E, and microarray is used for red cell, platelet and low-resolu-
Kell compatible red cells has become the standard of tion HLA genotyping. The red cell chip does not have
care at these institutions, it is not widely accepted. A ABO or D, but does include Cc, Ee, Kell, Duffy, Kidd,
survey of over 1,000 hospital laboratories in North MNSs, Diego, Dombrock, LW, Scianna, Colton and
America found that only 1 in 4 performed phenotype HgbS. Our 2008 cost analysis study of the bead-chip
matching for SCD patients.21 Indeed, in our survey of assay found that genotyping was actually less expensive
the 128 hospitals served by LifeShare Blood Centers than serological typing, and more genotypes could be
(which covers three states in the Southern US) only determined in a shorter period of time. Ninety-five
26% provide phenotyped matched blood.22 Many labo- donors could be typed in 6 hours, as opposed to 18
ratories are beginning to adopt a two-tiered approach to hours for serology; thus, allowing technologists to per-
phenotype matching. All chronically transfused SCD form other revenue generating tasks.
patients are matched for C, E and Kell (limited pheno- Thus, the goal of extended genotyping matched
type), which is not difficult to achieve. However, some donors for multi-transfused SCD patients is now with-
(including the CSCCs) provide more fully (extended) in reach. Molecular techniques for genotyping have the
matched units, that is, Duffy, Kidd, MNS, once the added advantage in that they are not influenced by
patient has made an alloantibody. recent transfusions as are serological methods.25 Yet
there remains opposition to genotype matching because
Extended genotype matching of the assumption that donors will not be available.
The common arguments against provision of pheno- Zhang et al.26 described a fulfillment model, which was
type matched units is that: (i) there is no way currently tested in two Southern US populations. Using actual
to predict who will be a responder; (ii) it is not cost- patient and donor data, they showed that the fulfill-
effective; (iii) the needed blood types are not readily ment of blood orders using donors matched for 14 anti-
available for most blood centers.16,23-24 In pediatric SCD gens could be achieved in 48-53% for site A and 59 to
patients, minimal matching can reduce alloimmuniza- 65% in site B. A feasibility study performed in Hong
tion by 65%.17 Castro et al.16 demonstrated that limited Kong27 stated that, by providing genotyped matched
phenotype matched blood could have prevented alloim- units, the antibody screen could be eliminated resulting
munization in 53.3% of SCD patients they studied, in a savings of approximately $14 million US dollars.
while extended matching would have reduced this in
70.8%. However, these authors conclude that matching Other factors influencing alloimunization
was impractical for long term-use because it relied heav- Even though the STOP trials used prospective red cell
ily on phenotypes not found in the majority of phenotype matching, alloimmunization was greatly
(Caucasian) blood donors. The rising cost of reagents reduced but not entirely eliminated.19 This suggests that
made these very real concerns in the US. However, the other factors may be involved in the immune response.
advent of molecular techniques, especially microarrays From the investigation of over 3,000 patients enrolled in

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 277 |
14th Congress of the European Hematology Association

the Comprehensive Study for Sickle Cell Disease, Rosse the D antigen; however, due to the lost epitopes they
et al.28 reported that the immunization rate was affected can form allo-anti-D. In addition, the DAR gene appears
by increased age, female sex and the presence of other to be linked to a RHCE variant known as ceAR.
abnormal hemoglobins. This was confirmed in a study Individuals who are homozygous for these alleles have
by Murao and Viana29 in Brazil, but not in the the unique phenotype known as hrS–. The hrS– pheno-
Netherlands.30 Female sex is a also a risk factor for type can also be the result of other variant alleles, for
alloimmunization in non-SCD patients,31 occurring in a example, ceMO.41,42 These individuals may produce an
2:1 ratio in West Africa (G. Noumsi, unpublished data). anti e-like antibody when transfused and if so, geno-
This most likely can be explained by the added anti- typed-matched blood may only be available through
genic exposure from pregnancies. In support of this idea rare donor programs.43
is the fact that there is no sex bias for alloimmunization Another variant RHD gene, known as r’s, is actually a
rates in pediatric patients.32 hybrid gene with exons 4-8 and part of exon 3 being
Investigations of HLA and immunization have been contributed by RHCE.44-46 If the r’s gene is paired with an
contradictory with some investigators reporting a role Ro or r gene, the patient will be genetically C negative
for HLA B-35.33,34 The presence of white blood cells, and may make allo anti-C, even though the RBCs may
which carry HLA antigens, appears detrimental by type as C+.47,48 We have observed this situation in at
affecting the type 2 immune response. Blumberg et al.35 least a half dozen patients, so we now routinely geno-
postulated that by reducing the leukocytes and platelets type for r’s, and when this scenario occurs, we transfuse
found in whole blood there would be a decreased fre- C negative units. When a patient is homozygous for r,s
quency of alloimmunization. These authors were their red cells will lack the hrB antigen and they are also
proved correct and leukoreduced blood products, that at risk to make anti-hrB or other complex Rh antibodies.
are also hemoglobin S negative, have become the stan- R2R2 units will be compatible since they lack hrB but one
dard of care in the US for transfusion of SCD patients. runs the risk of stimulating an anti-E. When this occurs,
Finally, recent work by Hendrickson and colleagues rare E-hrB negative blood may only be obtainable
has demonstrated that inflammation may contribute to through rare donor programs.
the heightened immune response.18,36 Using a murine
model of transfusion, they showed that induction of Dombrock blood group
inflammation with poly(I:C) induced significant con- The major antigens in the Dombrock system include:
sumption of transfused cells by splenic dendritic cells. Doa, Dob, Gya, Hy and Joa, all of which have been iden-
Inflammation has been well documented in SCD and its tified at the molecular level.49 The Hy– and Jo(a–) phe-
role in alloimmunization, as well as hyperhemolysis notypes are rare but have been found more frequently
deserves further study. in Blacks descended from Africa.50,51 The antibodies are
often weak and difficult to identify but they can cause
Unique antibody specificities and blood group hemolytic transfusion reactions.52 Compatible donors
phenotypes in Blacks are best identified by molecular methods. Using
Autoantibodies microarray, we have found the frequency of Hy- and
It is not unusual for SCD patients to have, at some Jo(a–) to be 1/1,000 and 1/100, respectively, in Southern
point in time, a demonstrable warm autoantibody, US Black blood donors (JMM, unpublished data). Of
sometimes having apparent Rh blood group specificity.37 note, we identified 3 Jo(a–) SCD patient in the first 75
This makes it difficult to identify underlying alloanti- that were genotyped.
bodies and makes transfusion even riskier. Once again,
extended genotyped units can provide a safer alterna- MNSs blood group
tive to random unit transfusions. Perhaps a more com- The MNSs and U blood group antigens are carried on
pelling argument for genotype matching for SCD comes glycophorin A and B. Deletion of the genes encoding
from recent data showing that alloimmunization is a GYPB results in the S-s-U- phenotype. Some red cells
contributing risk factor for autoimmune hemolytic ane- that type as S-s- may be weakly U+ and are known as
mia.38,39 Furthermore, correctly identifying antibody U+var. Both types are virtually exclusive to Blacks and,
specificities in these complex cases may often be diffi- thus, can pose a problem if found in a SCD patient
cult due to unique blood group types found in Blacks, needing transfusions. Storry and Reid reported that anti-
some of which are described in the following sections. U were heterogeneous depending if the individual had
no GYPB or a hybrid GYPB-GYPA protein.53 Allo anti-U
RH blood group has caused both hemolytic and delayed hemolytic
This is not intended to be a review of blood groups transfusion reactions (DHTR). These are sometimes dif-
but rather a brief description of some of the unusual ficult to recognize in the SCD patient, often being con-
genotypes found in SCD patients. These unusual geno- fused with acute chest syndrome or infection. DHTR
types can result in the production of complex antibody will be dealt with in more detail in the following sec-
specificities in transfused SCD patients and can ulti- tion.
mately delay obtaining compatible blood.
Partial D phenotypes are characterized by lost epi- Hyperhemolysis in transfused sickle cell patients
topes, and arise from the replacement of RHD exons by Incidence
their counterparts in the RHCE gene. Several of these There are many reports in the literature of SCD
are found more frequently in Black SCD patients, patients having delayed transfusion reactions and hyper-
including DIIIa and DAR.40 The DAR gene has mutations hemolysis, that is, the post-transfusion hemoglobin drops
in exon 4, 5 and 7, resulting in the weak expression of lower than pre-transfusion.54-59 Hyperhemolysis can

| 278 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

Table 1. Sickle cell hemolytic transfusion syndrome.61 course, would add to the proinflammatory mileau.
Furthermore, Test and Woolworth66 found that sickled
• Acute or delayed hemolytic transfusion reaction cells had a defect in the regulation of the complement
• Marked reticulocytopenia membrane attack complex particularly in the most
• Development of more severe anemia post-transfusion in the absence of dense or older cells. Thus, membrane perturbations aris-
bleeding ing from the release of oxygen radicals, paired with
• Immune hemolysis of autologous red cells defective complement regulatory control may con-
• Subsequent transfusions may exacerbate the anemia and be fatal tribute to increased RBC destruction in the sickle cell
• Patients may have multiple alloantibodies or autoantibodies patient.
• Serological studies may not provide an explanation for hemolysis The hyperhemolysis syndrome bears striking similar-
• Recovery, as manifested by reticulocytosis and increasing hematocrit occurs ity to the severe anemia found in malaria where the fall
following the withholding of additional transfusions in hematocrit is more than can be explained by the rup-
• Similar symptoms may occur with future transfusion
ture of parasitized cells alone. In both situations, it
appears that there is reactive lysis or innocent bystander
hemolysis, that is, immune destruction of RBCs that are
not sensitized with immunoglobulin. Thompson and
occur both in pediatric and adult patients and is often, Lachmann64 found that acute phase sera often exhibited
but not always the result of a delayed transfusion reac- this phenomenon and showed that these sera could
tion. Alloantibodies will appear in the serum 1 to 2 activate C5-C6-C7 causing lysis without the early com-
weeks post transfusion but the direct antiglobulin test plement components being bound.
(DAT) may or may not be positive. Antibody specifici-
ties associated with DHTR include C, E, Fya, Jka, Jkb and Conclusions
S, but in some cases there are no detectable antibod- The application of molecular genotyping to provide
ies.17,59 It is important that the manifestation of this syn- better-matched red cell units to multiply transfused
drome be recognized because failure to do so may result sickle cell patients, holds promise to help reduce all-
in inappropriate patient management. loimmunization. However, this will require increased
public awareness of SCD, a better knowledge of moti-
Clinical findings vating factors for Black blood donors and targeted
Multiple factors contribute to the clinical picture recruitment programs.67 Furthermore, a consensus will
found in hyperhemolysis, including recent transfusion, need to be achieved and standards developed regard-
presence of allo or autoantibodies, persistent reticulocy- ing the matching of red cell antigens.21,67 Delayed
topenia and splenic sequestration.17 Laboratory results transfusion reactions and hyperhemolysis, however,
may include increased total bilirubin, LDH, RDW and remain a risk of multiple transfusions. Education of
nucleated red cells in the peripheral circulation.60 The physicians and medical staff regarding transfusion
patient often presents with pain and/or fever. Table 1 medicine practices is paramount to providing better
shows the criteria for hyperhemolysis as defined by care for the SCD patient, and the hematologist should
Petz et al.61 for the syndrome they call sickle cell hemolyt- be an integral part of this effort.
ic transfusion reaction. Often SCD patients having a
hemolytic transfusion reaction are misdiagnosed as hav-
ing typical vaso-occlusive or aplastic sickle cell crises. References
However, a DHTR should be suspected when the
patient has recurrent or severe sickle cell crises follow- 1. Smith LA, Oyeku SO, Homer C, Zuckerman B. Sickle Cell
ing transfusion. Treatment for patients having hyperhe- Disease: A question of equity and quality. Pediatrics 2006;
117:1763-70.
molysis are; (i) withholding of further transfusions; (ii) a 2. Adams RG, McKie V, Hsu L. Prevention of a first stroke by
short course of corticosteroids, for example, 1 to 2 transfusion in children with abnormal results of transcra-
mg/kg of prednisone daily.61,62 Although there is prom- nial Doppler ultrasonography. N Engl J Med 1998;339:5-11.
ising research and a few case reports in the literature 3. Lee MT, Piomelli S, Granger S, Miller ST, Harkness S,
Brambilla DJ, Adams RJ, STOP Study Investigators. Stroke
regarding the use of intravenous immune globulin for prevention trial in sickle cell anemia (STOP): extended fol-
the treatment of hyperhemolysis, a recent guideline low-up and final results. Blood 2006;108:847-52.
from Canada did not recommend its use.63 4. Adams RJ, Brambilla DJ, STOP Study Investigators.
Discontinuing prophylactic transfusions used to prevent
stroke in sickle cell disease. N Engl J Med 2005;353:2769-
Theories regarding hyperhemolysis 78.
The syndrome of hyperhemolysis may now be better 5. Moreira JG, Bordin JO, Kuroda A, Kerbauy J. Red blood
defined but its exact cause is unknown. Several theories cell alloimmunization in sickle cell disease: the influence of
racial and antigenic pattern differences between donors
have been put forth including: (i) sequestration followed and recipients in Brazil. Am J Hematol 1996;52:197-200.
by intravascular hemolysis secondary to adhesion to 6. Blumberg N, Peck K, Ross K, Avila E. Immune response to
endothelial cells via inflammatory mediators; (ii) reor- chronic red cell transfusion. Vox Sang 1983;44:212-17.
ganization of the phospholipid bilayer of sickled cells 7. Coles SM, Klein HG, Holland PV. Alloimmunization in
two multitransfused patient populations. Transfusion
and possible exposure of crypt antigens; (iii) innocent 1981;21: 462-7.
bystander hemolysis due to reactive lysis.62,64 The true 8. Davies SC, McWilliam AC, HewittE, Devenish A,
explanation may be a combination of these. For exam- Brozovic M. Red cell alloimmunization in sickle cell dis-
ple, it has been reported that phosphatidylserine is ease. Br J Haematol 1986;63:241-5.
9. Bashwari LA. Red cell alloimmunization in sickle-cell
exposed on the outer membrane surface of sickled red anaemia patients. East Mediterr Health J 2007;3:1181-9.
cells leading to complement activation.58,65 This, of 10. Vichinsky EP, Earles A, Johnson RA, Hoag MS, Williams A,

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 279 |
14th Congress of the European Hematology Association

Lubin B. Alloimmunization in sickle cell anemia and trans- cell disease. Clin Immunol Immunopathol 1986;38:178-83.
fusion of racially unmatched blood. N Engl J Med 1990; 34. Reisner EG, Kostyu DO, Phillips G, Walker C, Dawson
322: 1617-21. DV. Alloantibody responses in multiply transfused sickle
11. Sarnaik S, Schornack J, Lusher JM. The incidence of devel- cell patients. Tissue Antigens 1987;30:161-6.
opment of irregular red cell antibodies with sickle cell ane- 35. Blumberg N, Heal JM, Gettings KF. WBC reduction of RBC
mia. Transfusion 1986;6:249-52. transfusions is associated with a decreased incidence of
12. Orlina AR, Unger PJ, Koshy M. Post-transfusion alloimmu- RBC alloimmunization. Transfusion 2003;43:945-52.
nization in patients with sickle cell disease. Am J Hematol 36. Hendrickson JE, Desmarets M, Deshpande SS, Chadwick
1978;5:101-6 TE, Hillyer CD, Roback JD, et al. Recipient inflammation
13. Sosler SD, Jilly BJ, Saporito C, Koshy M. A simple, practi- affects the frequency and magnitude of immunization to
cal model for reducing alloimmunization in patients with transfused red blood cells. Transfusion 2006;46:1526-36.
sickle cell disease. Am J Hematol 1993;43:103-6. 37. Castellino SM, Combs MR, Zimmerman SA, Issitt PD,
14. Tahhan HR, C. Holbrook T, Braddy LR, Brewer LD, Ware RE. Erythrocyte autoantibodies in paediatric patients
Christie JD. Antigen-matched donor blood in the transfu- with sickle cell disease receiving transfusion therapy: fre-
sion management of patients with sickle cell disease. quency, characteristics and significance. Br J Haematol
Transfusion 1994;34:562-9. 1999;104:189-94.
15. Noumsi GT, Tounkara A, Guindo YG, Moulds JM. 38. Ahrens N, Pruss A, Kahne A, Kiesewetter H, Salama A.
Incidence of alloimmunization in Blacks from Africa vs. Coexistence of autoantibodies and alloantibodies to red
the USA. Transfusion 2006;46 Suppl:157A. blood cells due to blood transfusion. Transfusion 2007;47:
16. Castro O, Sandler SG, Houston-Yu P, Rana S. Predicting the 813-6.
effect of transfusing only phenotype matched RBCs to 39. Young PP, Uzielblo A, Trulock W, Lublin DM, Goodnough
patients with sickle cell disease: theoretical and practical LT. Autoantibody formation after alloimmunization: are
implications. Transfusion 2002;42:684-90. blood transfusions a risk factor for autoimmune hemolytic
17. Aygun B, Padmanabhan S, Paley C, Chandrasekaran V. anemia? Transfusion 2004;44:67-72.
Clinical significance of RBC alloantibodies and autoanti- 40. Hemker MB, Ligthart PC, Berger L, van Rhenen DJ, van der
bodies in sickle cell patients who received transfusions. Schoot CE, Maaskant-van Wijk PA. DAR, a new RhD vari-
Transfusion 2002;42:43. ant involving exons, 4, 5 and 7, often in linkage disequilib-
18. Hendrickson JE, Chadwick TE, Roback JD, Hillyer CD, rium with ceAR, a new Rhce variant frequently found in
Zimring JC. Inflammation enhances comsumption and African Blacks. Blood 1999;94:4337-42.
presentation of transfused RBC antigens by dendritic cells. 41. Noizat-Pirenne F, LePennec PY, Ansart-Pirenne H, Juszczak
Blood 2007;110:2736-43. PC, Patereau C, Verdier M, Babinet J, Roussel M, Rouger
19. Vichinsky EP, Luban NLC, Wright E, Olivieri N, Driscol Nl, P, Cartron JP. Two new alleles of the RHCE gene in Black
Pegelow CH, et al. Prospective RBC phenotype matching individuals: the RHce allele ceMO and the RHcE allele
in a stroke-prevention trial in sickle cell anemia: a multi- cEMI. Br J Haematol 2001;113:672-9.
center transfusion trial. Transfusion 2001;41:1086-92. 42. Castilho L, Rios M, Rodriquez A, Pellegrino J Sr, Saad ST,
20. Afenyi-Annan A, Willis MS, Konrad TR, Lottenberg R. Costa FF. High frequency of partial DIIIa and DAR alleles
Blood bank management of sickle cell patients at compre- found in sickle cell disease patients suggests increased risk
hensive sickle cell centers. Transfusion. 2007;47:2089-97. of alloimmunization. Transfus Med 2005;15:49-55.
21. Osby M, Shulman IA. Phenotype matching of donor red 43. Vege S, Westhoff CM. Molecular characterization of GYPB
blood cell units for nonalloimmunized sickle cell disease and RH in donors in the American Rare Donor Program.
patients: a survey of 1182 North American laboratories. Immunohematology 2006;22:143-7.
Arch Pathol Lab Med 2005;129:190-3. 44. GL Daniels, Faas BHW, Green CA, Smart E, Maaskant-van
22. Moulds JM, Allen TI. A regional survey of transfusion serv- Wijk PA, AvenT ND, Zondervan HA, von dem Borne AEG
ices and their adherence to the sickle cell disease transfu- Kr, van der Schoot CE. The VS and V blood group poly-
sion protocol. Transfusion 2006;46:156A-157A. morphisms in Africans: serologic and molecular analysis.
23. Blumberg N, Ross K, Avila E, Peck K. Should chronic trans- Transfusion 1998;38:951-8.
fusions be matched for antigens other than ABO and 45. Faas BHW, Beckers EAM, Wildoer P, Ligthart PC,
Rho(D)? Vox Sang 1984;47:205-8. Overbeeke MAM. Molecular background of VS and weak
24. Flickinger C. In search of red blood cells for alloimmunized C expression in blacks. Transfusion 1997;37:38-44.
patients with sickle cell disease. Immunohematology 2009; 46. Noizat-Pirenne F, Lee K, Le Pennec PY, Simon P, Kazup P,
22:136-42. Rouzaud AM, et al. Rare RHCE phenotypes in black indi-
25. Castilho L, Rios M, Blanco C, Pellegrino J, Alberto FL, Saad viduals of Afro-Carribean origin: identification and trans-
ST, et al. DNA-based typing of blood groups for the man- fusion safety. Blood 2002;100:4223-31.
agement of multiply-transfused sickle cell disease patients. 47. Westhoff CM, Vege S, Nance S, Flickinger C. Transfusion
Transfusion 2002;42:232-8. complicated by variant C antigen in sickle cell disease.
26. Zhang Y, HashmI G, Seul M, Collins C, Sapp C, Borges M, Transfusion 2006;46 Suppl:157A.
et al. Random selection of donor units for extended pheno- 48. Lomas C, Storry JR, Spruell P, Moulds M. Apparent allo-C
type determination and efficient allocation to prospective produced by r’s people: five examples. Transfusion 1994;4
recipients. Transfusion 2008;48:198A. Suppl:25S.
27. Lau FY, Wong R, Chan NP, Chui CH, Ng E, Ng MH, Cheng 49. Rios M, Hue-Roye K, Oyen R, Miller J, Reid ME. Insights
G. Provision of phenotyped-matched blood units: no need into the Holley- and Joseph- phenotypes. Transfusion
for pre-transfusion antibody screening. Haematologica 2002; 42:52-8.
2001; 86:742-8. 50. Baleottie W, Rios M, Hashmi G, Fabron A, Pelligrino J,
28. Rosse WF, Gallagher D, Kinney TR, Castro O, Dosik H, Castilho L. Dombrock gene analysis in Brazilian people
Moohr J, et al. Transfusion and alloimmunization in sickle reveals novel alleles. Vox Sang 2006;91:81-7.
cell disease. Blood 1990;76:1431-7. 51. Moulds JM, Noumsi GT, Chen Q, Moulds JJ. New
29. Murao M, Viana MB. Risk factors for alloimmunization by Dombrock haplotypes of African origin. Transfusion
patients with sickle cell disease. Braz J Med Biol Res 2005; 2006;46 Suppl:139A.
38:675-82. 52. Reid ME. The Dombrock blood group system: a review.
30. Fluit CR, Kunst VA, Drenthe-Schonk AM. Incidence of red Transfusion 2003;43:107-14.
cell antibodies after multiple blood transfusion. 53. Storry JR, Reid ME. Characterization of antibodies pro-
Transfusion 1990; 30:532-5. duced by S-s- individuals. Transfusion 1996;36:512-6.
31. Bauer MP, Wiersum-Osselton J, Schipperus M, Vanden- 54. Cox JV, Steane E, Cunningham G, Frenkel EP. Risk of
broucke JP, Briet E. Clinical predictors of alloimmunization alloimmunization and delayed hemolytic transfusion reac-
after red blood cell transfusion. Transfusion 2007; 47:2066- tions in patients with sickle cell disease. Arch Intern Med
71. 1988; 148:2485-9.
32. Talano JA, Hillery CA , Gottschall JL, Baylerian DM, Scott 55. King KE, Shirey RS, Lankiewics MW, Young-Ramsaran J,
JP. Delayed hemolytic transfusion reaction/hyperhemoly- Ness PM. Delayed hemolytic transfusion reactions in sick-
sis syndrome in children with sickle cell disease. Pediatrics le cell disease: simultaneous destruction of recipients’ red
2003; 111:661-5. cells. Transfusion 1997;37:376-81.
33. Alarif L, Castro O, Ofosu M, Dunston G, Scott RB. HLA- 56. Diamond WJ, Brown FL, Bitterman P. Delayed hemolytic
B35 is associated with red cell alloimmunization in sickle transfusion reaction presenting as sickle-cell crisis. Ann

| 280 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

Intern Med 1980;93:231-4. ment of sickle cell disease. Blood 1993;81:1109-23.


57. Cullis JO, Win N, Dudley JM, Kayentao K. Post-transfusion 63. Anderson D, Ali K, Blanchette V, Brouwers M, Couban S,
hyperhemolysis in a patient with sickle cell disease: use of Radmoor P, et al. Guidelines on the use of intravenous
steroids and intravenous immunoglobulin to prevent fur- immune globulin for hematologic conditions. Transfus
ther red cell destruction. Vox Sang 1995;69:355-7. Med Rev 2007;21 Suppl 1:S9-S56.
58. Salama, A, Mueller-Eckhardt C. Delayed hemolytic trans- 64. Thompson RA, Lachmann PJ. Reactive lysis: the comple-
fusion reactions. Evidence for complement activation ment-mediated lysis of unsensitized cells. I. The character-
involving allogeneic and autologous red cells. Transfusion ization of the indicator factor and its identification as C7.
1984;24:188-93. J Exp Med 1970;131:629-41.
59. King KE, Shirey RS, Lankiewics MW, Young-Ramsaran J,
65. Mold C, Tamerius JD, Phillips G. Complement activation
Ness PM. Delayed hemolytic transfusion reactions in sick-
le cell disease: simultaneous destruction of recipients’ red during painful crisis in sickle cell anemia. Clin Immunol
cells. Transfusion 1997;37:376-81. Immunopathol 1995;76:314-20.
60. Ballas SM, Marcolina MJ. Hyperhemolysis during the evo- 66. Test ST, Woolworth VS. Defective regulation of comple-
lution of uncomplicated acute painful episodes in patients ment by the sickle erythrocyte: evidence for a defect in
with sickle cell disease. Transfusion 2006;46:105-10. control of membrane attack complex formation. Blood
61. Petz LD, Calhoun L, Shulman IA, Johnson C, and Herron 1994;83:842-52.
RM. The sickle cell hemolytic transfusion reaction syn- 67. Hassell K, Pace B, Wang W, Kulkarni R, Luban N, Johnson
drome. Transfusion 1997;37:382-92. CS, et al. Sickle cell disease summit: from clinical and
62. Wayne AS, Kevy SV, Nathan DG. Transfusion manage- research disparity to action. Am J Hematol 2008:84:39-45.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 281 |
Transfusion

Transfusion problems in stem cell transplant patients

D.H. Pamphilon A B S T R A C T

Stem cell transplant (SCT) patients may require intensive blood component support. Transfusions
NHS Blood and Transplant, Bristol, can be complicated by transmission of viral and bacterial infections, transfusion-associated (TA)-
UK
GVHD, febrile non-hemolytic transfusion reactions (FNHTR) and transfusion-related acute lung injury
(TRALI). Alloimmunisation (AI) to red cell antigens may cause difficulties in selecting compatible blood,
Hematology Education: whilst AI to the human leukocyte antigens (HLA) present on platelets may cause refractoriness to sub-
the education program for the sequent platelet transfusions. It is essential to define robust transfusion policies and procedures, and
annual congress of the European these should be regularly audited. This article briefly reviews the blood components available for
Hematology Association
transfusion and discusses the use of granulocyte transfusions in the setting of SCT, the impact of
2009;3:282-287 reduced-intensity conditioning (RIC) transplantation on transfusion requirements, the prevention of
cytomegalovirus (CMV) transmission and the management of ABO-mismatched transplants.

This paper is modified from the


chapter on transfusion support first
published in The EBMT Handbook
2008 with permission of the EBMT
(ref 28).

Regulatory requirements for blood and blood HTLV-1 and 2, CMV and syphilis. Blood
components in Europe services routinely test blood for:
The European Union (EU) Directive • Hepatitis B: hepatitis B surface antigen
2002/98/EC sets standards for the collection, (HbsAg)*
testing, processing, storage and distribution • Hepatitis C: hepatitis C antibodies (anti-
of human blood and blood components.1 It HCV)*
requires that Blood Establishments should • Human Immunodeficiency Virus 1 and 2:
be licensed, and this is of importance for HIV 1 + 2 antibodies (anti-HIV 1+2)*
both Blood Centers in EU countries that • Human T- Lymphotropic Virus 1 and 2:
undertake these activities, as well as hospi- HTLV 1 + 2 antibodies (anti-HTLV-1+2)
tals that collect and issue, for example, gran- • Syphilis
ulocytes for transfusion. The most impor- *These tests are mandated by the EU Blood
tant aspects of the Directive are: (i) the fate Directive (2002/98/EC).1
of each unit of all blood components should Blood donors may also be tested for anti-
be recorded and this record kept for 30 HBc, that is, anti-hepatitis B core antigen,
years. i.e. vein-to-vein traceability; (ii) robust alanine aminotransferase (ALT): a surrogate
quality systems should be in place; the pro- marker of hepatitis C, HCV-RNA by PCR for
cessing of blood and blood components hepatitis C and p24 antigen for HIV-1.
should be undertaken by licensed blood Testing for anti-CMV antibody to identify
establishments (see above); (iii) training CMV seronegative donors is done on a pro-
should be provided for hospital transfusion portion of blood donations, and must be suf-
laboratory staff, and (iv) hemovigilance sys- ficient to identify enough CMV seronegative
tems should be established to include the components for transfusion to those patients
reporting of adverse events for whom it is appropriate.
Establishments are licensed by the Bacterial contamination is a relatively
Competent Authorities in EU Member common occurrence, with an incidence esti-
States following inspection by a regulatory mated at 0.05 to 0.5% of components.2 The
body – in the United Kingdom (UK) this is sources of bacteria are donor bacteremia and
the Medicines and Healthcare Products contamination with bacteria present on the
Regulatory Authority (MHRA). Reports of skin at the time of donation or present in
compliance must be submitted. blood packs. Screening tests for bacteria in
platelet concentrates (PC) using automated
Testing of donated blood for infectious blood culture systems, for example,
disease markers BacT/ALERT have been evaluated,2 and are
A number of microbial agents may be now used routinely by some transfusion
transmitted by blood transfusion. These services. PC are not issued until at least 48
include hepatitis B and C, HIV-1 and 2, hours after collection but the storage period

| 282 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

may be extended to 7 days once sterility has been eval- Table 1. Adverse effects of transfused leukocytes.
uated. The risk of bacterial transmission is also mini-
mized by careful donor selection, meticulous attention
to sterility during venepuncture, diversion of the first 30 HLA alloimmunisation causing
mL of blood collected – which contains most of the bac- FNHTR
teria – away from the primary collection pack, and Refractoriness to random donor platelets
sterility during preparation of blood components. Graft rejection
Bacterial contamination should be suspected in any Shortened/red cell survival
patient who develops a febrile reaction characterized by
fever, chills and/or hypotension. Transmission of microorganisms
Microbiological testing does not completely remove CMV
the risk of infection, although the chance of infection in HTLV – 1 / 11
the UK after transfusion of screened blood components Toxoplasma gondii
Yersinia enterocolitica
from known/previously tested donors is estimated to be
less than 1 in 2×106 for HIV-1, HBV and HCV. This risk Immunomodulation
will vary somewhat according to the donor selection GVHD
and testing policies that are operative within a Blood Activation of viruses in host cells e.g., HIV – 1
Service. Immune suppression of T- and NK-cell functions
Prevention of cytomegalovirus transmission Affecting the quality of stored blood
A proportion of SCT patients are CMV seropositive Microaggregate formation
pre-transplant or have seropositive donors. They Metabolic deterioration during storage
require regular screening by PCR and antigenemia test-
ing, together with ganciclovir therapy where appropri-
ate to minimize the impact of virus reactivation and
prevent clinical infection post-transplant. All CMV refractoriness. Refractoriness is not always prevented,
seronegative SCT patients, with CMV seronegative since in more than 50% of cases, it results from
donors (neg/neg), and patients with hematological and increased platelet destruction due to non-immune caus-
other disorders who are likely to proceed to a trans- es, which include fever, splenomegaly, DIC and ampho-
plant, should receive blood components that have a tericin therapy.12
minimal risk of causing CMV acquisition.3 Studies show AI is also associated with a higher incidence of graft
that the use of CMV seronegative components is asso- failure in patients with severe aplastic anemia.
ciated with an incidence of CMV infection in CMV Filtration of blood or its components is best performed
neg/neg SCT of between 1 to 4%.4-7 CMV is transmitted in blood centers and hospital blood banks. Data from
via leucocytes and leucodepletion also minimizes the studies where leucocytes were filtered from blood com-
risk of CMV transmission.4-6,8 CMV seronegative and ponents at the bedside show that this might be ineffec-
leucodepleted blood components are probably of equiv- tive in preventing or reducing FNHTR, AI and refractori-
alent efficacy but this view is not generally accepted.6,9 ness.13
Further evidence from prospective randomized con- Indications for leucodepleted blood components14
trolled studies (PRCT) using pre-storage leucodepleted include: (i) pre-transplant in patients with SAA to
blood components is required. Centers must establish reduce the likelihood of graft failure; (ii) pre- and post-
their own policies. transplant to prevent recurrent FNHTR; (iii) pre-and
post-SCT to minimise HLA AI and platelet refractori-
Leucodepleted blood components ness. This is optional since there is no evidence of a sig-
Transfused leucocytes cause alloimmunisation (AI) to nificant impact on important clinical outcome meas-
HLA class 1 antigens in a proportion of patients. This ures, such as survival post-SCT, except in patients with
may be manifested clinically as FNHTRs, although SAA. Nonetheless, many blood services have imple-
these may also be caused by antibodies to neutrophils, mented leucodepletion of a large proportion or, in some
platelets, plasma proteins, and by cytokines such as cases, all of their blood components. In the UK, univer-
interleukin (IL)-1, IL-6, IL-8 and tumor necrosis factor sal leucodepletion was implemented in 1999, with the
(TNF)-alpha, which accumulate in stored blood compo- aim of minimizing the risk of transfusion-associated
nents, especially PC. HLA AI may cause accelerated transmission of the causative agent of variant
destruction of HLA incompatible transfused platelets. Creutzfeld-Jakob disease (vCJD) (iv) as an alternative to
This is clinically manifest as a failure to achieve a satis- CMV seronegative components.
factory increment after platelet transfusion (refractori-
ness). A summary of the adverse effects of transfused Gamma-irradiation of blood components and TA-GVHD
leucocytes is shown in Table 1.10,11 HLA incompatible third party leucocytes contained
Donor dendritic cells (DC), which are present in red in donated blood components can engraft and initiate
cell and platelet transfusions, appear to be responsible an alloreactive response after transfusion. This can
for sensitisation to HLA. Studies show that removal of cause TA-GVHD, manifest clinically by fever, rash,
leucocytes to less than 5×106 per blood component pre- diarrhea, jaundice and pancytopenia, and this is fatal in
vents primary HLA AI in more than 97% of patients more than 90% of cases, so prevention is essential.
with hematological malignancies. The use of leucode- Donor leucocytes are inactivated by gamma-irradiation
pleted components also reduces secondary AI and of 2500 cGy, and all components for SCT recipients

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 283 |
14th Congress of the European Hematology Association

should be irradiated from the time that conditioning Table 2. Indications for irradiated blood components.
therapy is started. In addition, HLA matched PC should
be irradiated, as should those from family members, Allogeneic HSC recipients from time of conditioning therapy for 6 months or
since HLA haplotype sharing may result in TA-GVHD, until the lymphocyte count is 1×109/L in the absence of chronic GVHD
even in immunocompetent patients. A summary of the Allogeneic HSC donors
indications for blood component irradiation is shown
in Table 2. Autologous HSC recipients (from 7 days before harvest until 3 months
Platelets show normal functional characteristics post transplant)
through 5 days storage after irradiation with doses up to All donations from HLA-matched donors or 1st or 2nd degree relatives
5000 cGy. Red cells leak potassium during storage and
this is made worse by irradiation. Therefore, storage is All patients with Hodgkins disease at any stage of therapy
limited to 14 days after 2500 cGy. TA-GVHD has been All patients treated with purine analogues, e.g., fludarabine
shown to occur after 1500-2000 cGy, and this dose
All patients with congenital immunodeficiency states
range is not recommended.15

Red cell transfusion policies


Red cells should be matched for ABO and Rhesus D A higher threshold of 20×109/L should be used in
type.1 Extended phenotyping may be necessary in patients with fever, sepsis, splenomegaly and other
patients, e.g., those with sickle cell disease, who have well-established causes of increased platelet consump-
formed red cell alloantibodies after previous transfu- tion.
sions. Red cells should be cross-matched against the If an invasive procedure is planned, e.g. central line
patient’s serum by standard techniques prior to transfu- insertion, the platelet count should be more than
sion. Thresholds should be defined for hemoglobin and 50×109/L. PC should be transfused when there is signif-
PCV below which red cell transfusions are always icant clinical bleeding, irrespective of the platelet count.
given. Suggested arbitrary cut-off points are Hb less PC is contraindicated in patients with TTP. In adults
than 8.0 g/dL and PCV less than 25%. the usual dose of platelets is 3 x 1011 (an adult therapeu-
In adults 1 unit of red cells raises the Hb by 1.0 g/dL, tic dose – ATD) in a volume of 200 to 300 mL.
whereas in children the volume of blood to be trans- Children more than 30 kg receive one ATD. Children
fused is derived from the formula: volume to transfuse less than 30 kg are given 10 mL/kg.
= increase in Hb (g/dL) required x 4 x weight (kg) These policies apply to both SCT patients, as well as
These policies apply to both SCT patients, as well as being accepted recommendations for patients with
being accepted recommendations for patients with bone marrow failure who require supportive transfu-
bone marrow failure who require supportive transfu- sions.16-18
sions. Platelet transfusions can be monitored by looking for
cessation of bleeding, measuring the platelet count the
Platelet transfusion policies following day (values <20×109/L suggest refractoriness)
Current practice, based on the results of randomized or by measuring the platelet count at between 10 to 60
studies is to transfuse platelets concentrates (PC) pro- minutes post-transfusion where the corrected count
phylactically when the platelet count is less than increment (CCI) should be more than 7.5 (absolute
10x109/L. A recent Cochrane Systematic Review con- value).
cluded that, whilst there is no reason to change current If the patient is refractory to transfusion of PC, sam-
practice, blood products may become scarce, and fur- ples should be taken to test for HLA antibodies. Testing
ther trials should be undertaken to compare prophylac- is not required pre-transplant in all patients, although
tic versus therapeutic platelet transfusion; that is, PC some centers recommend it as a screening test. If HLA
should only be given when there is clinical bleeding.16 In antibodies are detected, HLA-matched platelets collect-
autologous HPCT, this has been found to be safe.17 ed by apheresis of HLA-typed donors should be used in
Fewer PC transfusions are required in RIC allografted these patients. If the CCI is less than 7.5 following
patients compared to those who receive full myeloabla- transfusion of HLA matched PC and the patient is not
tive conditioning.18 Best current practice is: PC should be bleeding, then withhold all platelet transfusions. If the
ABO and Rh compatible wherever possible since ABO CCI using well-HLA-matched PC is less than 7.5 and/or
incompatibility may reduce the expected count incre- bleeding persists, then possible causes include non-
ment (CI) by 10-30%. immune causes of refractoriness or, less commonly,
Group O PC should be tested for high titre anti-A, B, platelet-specific antibodies. Platelet transfusions will be
and if positive, should only be transfused to group O required in bleeding patients – sometimes 2 to 3 times
recipients to avoid hemolysis caused by passive admin- daily to prevent life-threatening hemorrhage. There is
istration of the antibody. no evidence that therapies such as IVIG improve out-
If Rh D positive platelets are given to an Rh D nega- come in this situation
tive patient, then give 250 iu polyclonal anti-Rh (D)
immunoglobulin. Since the chance of Rh immunization Granulocyte transfusion
is probably less than 5%, this may be omitted and the Granulocyte transfusions (GT) are prepared by pool-
patients serum screened for immune red cell antibodies, ing buffy coats from whole blood donations or they
or prior to a red cell transfusion. Studies show that a may be collected by the apheresis of steady state
threshold of 10×109/L in stable thrombocytopenic healthy donors who may be family members or unre-
patients is optimal for prophylactic platelet transfusion. lated volunteers. Granulocyte apheresis donors must

| 284 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

Figure 1. Diagram showing


the blood groups of red cells
platelets and FFP used for
transfusion support in ABO
missmatched SCT.

have a full medical assessment and testing to select Donor/recipient ABO incompatibility and transfusion
those that are ABO compatible and CMV appropriate. support
The granulocyte content is in the range 5-10×109 unit Approximately 15 to 25% of HLA identical sibling
for both these preparations. Mobilized granulocytes donor/recipient pairs differ for ABO blood groups and
can be collected from donors who receive G-CSF (5-10 the figure is higher in alternative donor transplants. In
µg/kg) and/or dexamethasone (8 mg) – both given 12 to myeloablative transplants, ABO incompatibility is asso-
24 hours before – to increase the number that can be ciated with an increased risk of delayed red cell engraft-
collected during a standard apheresis procedure. ment, pure red cell aplasia (PRCA), hemolysis and
Current practice is to give only G-CSF, since the use of increased transfusion requirements.18 There are also
steroids has been associated with the development of reports of increased platelet transfusion requirements.
posterior subcapsular cataracts,19 and the two methods ABO mismatch does not affect neutrophil engraftment,
give similar yields.20 This strategy gives a granulocyte the incidence of graft rejection, GVHD, disease progres-
yield of 10-100×109 per unit, and data available so far sion or overall survival.23
indicates that significant granulocyte increments, for RIC transplants are associated with decreased red cell
example, 1-2×109/L can be obtained.21 By contrast, it is and PC, usage18 and it has been demonstrated with
unusual to observe such increments with buffy coat or chimerism studies that early erythroid progenitors
unmobilised granulocytes. All granulocyte products engrafted as promptly as myeloid progenitors.24
must be irradiated prior to transfusion to prevent TA- However, as with myeloablative SCT, engraftment of
GVHD. Cross-matching is also required. A recent mature red cells is delayed, and cases of PRCA have
Cochrane Systemic Review indicated that there is cur- been reported. ABO mismatch is associated with
rently inconclusive evidence from PRCTs to support or increased red cell transfusion requirements.25 Recipient
refute the use of GT in neutropenic patients. Further plasma cells produce anti-donor ABO alloagglutinins
PRCT are required before definitive recommendations and after RIC SCT, the rate of decline of anti-donor
can be made.22 There is recent anecdotal evidence that hemagglutinins takes twice as long, leading to more
prophylactic administration of granulocytes may hemolysis.26 In one report of 40 patients who had RIC
reduce the incidence of severe fungal infections after SCT, ABO mismatch was associated with one death
BMT, but currently few centers use such transfusions due to hemolysis, three cases of PRCA, six cases of
and further studies are needed.21 Furthermore, granulo- thrombotic microangiopathy (three fatal), an increase in
cyte transfusions increase the likelihood of HLA immu- rehospitalisation days, relapse or disease progression
nization and platelet refractoriness. Granulocyte trans- and higher TRM.27 By contrast, other reports do not
fusions are probably best reserved for patients with show an inferior outcome.24-26
granulocyte counts less than 0.2×109/L and document-
ed bacterial or fungal infections not responding to at Definitions
least 3 days of appropriate antimicrobial therapy, in sit- Major ABO incompatibility is defined as the presence
uations where the granulocyte count is not expected to in the recipients plasma of anti- A, -B or -A,B alloagglu-
recover within 7 days. tinins reactive with the donor’s red cells, e.g., donor

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 285 |
14th Congress of the European Hematology Association

group A and recipient group O. ated and, in addition, may be CMV seronegative and
Minor ABO incompatibility is defined as the presence leucodepleted, provide optimum transfusion support
of anti-A,-B or -A,B alloagglutinins in the donors plasma and minimize the chance of adverse effects.
reactive with the recipient’s red cells, e.g., donor group
O and recipient group A.
Major plus minor, also referred to as bidirectional, References
ABO compatibility is defined as the presence in both
the donor and recipients plasma of anti-A, -B or -A,B 1. European Union Directive 2002/98/EC http://eurlex.
europa.eu/LexUriServ/site/en/oj/2003/l_033/l_03320030208e
alloagglutinins reactive with recipient and donor cells n00300040.pdf
respectively, e.g., donor group A and recipient group B. 2. Ramírez-Arcos S, Jenkins C, Dion J, Bernier F, Delage G,
Goldman M. Canadian experience with detection of bacterial
Incompatible stem cell graft infusion contamination in apheresis platelets. Transfusion 2007; 47:
421-9.
If the titre of anti-A and/or anti-B, is less than 1:64, 3. Bowden RA, Slichter SJ, Sayers M, Weisdorf D, Cays M,
then bone marrow or PBPC grafts may be infused with- Schoch G, et al. A comparison of filtered leokocyte- reduced
and cytomegalovirus (CMV) seronegative blood products for
out any modification. At higher titres, red cells should the prevention of transfusion – associated CMV infection after
be removed from the graft. Marrow processing or HPC marrow transplant. Blood 1995;86:3598-603.
collection on certain apheresis machines, such as the 4. Pamphilon DH, Rider JR, Barbara JA, Williamson LMP.
GAMBRO Spectra, usually results in red cell contamina- Prevention of transfusion-transmitted cytomegalovirus infec-
tion. Transfus Med 1999;9:115-23.
tion of less than 5 mL and ABO incompatibility may be 5. Ljungman P, Larsson K, Kumlien G, Aschan J, Barkholt L,
ignored. Likewise, stem cells that are separated on den- Gustafsson-Jernberg A, et al. Leukocyte depleted, unscreened
sity gradients and washed can also be infused without blood products give a low risk for CMV infection and disease
in CMV seronegative allogeneic stem cell transplant recipients
regard to ABO incompatibility. with seronegative stem cell donors. Scand J Infect Dis 2002;
Plasma may be removed from the transplant in cases 34:347-50.
of minor ABO mismatch where the alloagglutinin titre 6. Nichols WG, Price TH, Gooley T, Corey L, Boeckh M. Trans-
fusion-transmitted cytomegalovirus infection after receipt of
is high to avoid acute hemolysis in the recipient. leukoreduced blood products. Blood 2003;101:4195-200.
Delayed hemolytic transfusion reactions may follow 7. Foot AB, Pamphilon D, Caul EO, Roome AP, Hunt LP, Cornish
the infusion of donor HSC where there is a minor ABO JM, et al. Cytomegalovirus infection in recipients of related
mismatch. This is because of a secondary (anamnestic) and unrelated donor bone marrow transplants no evidence of
increased incidence in patients receiving unrelated donor
immune response mediated via memory B cells in the grafts. Br J Haematol 1998;102:671-7.
graft against recipient ABO antigens. A rise in anti-A, -B 8. Ronghe MD, Foot AB, Cornish JM, et al. The impact of trans-
or -A,B titre is seen together with anemia and jaundice. fusion of leukodepleted platelet concentrates on CMV disease
after allogeneic stem cell transplantation. Br J Haematol 2002;
This phenomenon is rarely, if ever, seen when bone 118:1124-7.
marrows are depleted of alloreactive T-lymphocytes 9. Narvios AB, de Lima M, Shah H, Steward CG, Carrington D,
using strategies, such as alemtuzumab (CAMPATH-1) Goulden N, et al. Transfusion of leukoreduced cellular blood
components from cytomegalovirus-unscreened donors in allo-
antibody or CD34 positive cell selection since B-cells are geneic hematopoietic transplant recipients: analysis of 72
also depleted. recipients. Bone Marrow Transplant 2005;36:499-501
10. Blajchman MA. Immunomodulation and blood transfusion.
Blood groups used for transfusion support Am J Ther 2002;9:389-95.
11. Blajchman MA. The clinical benefits of the leukoreduction of
Before transplant, recipient-type red cells and platelets blood products. J Trauma 2006;60 6 Suppl:S83-90.
should be given but after the transplant, the situation is 12. Slichter SJ. Platelet refractoriness and alloimmunization.
more complicated (Figure 1). Leukemia1998; Suppl 1:S51-3
13. Williamson LM, Wimperis JZ, Williamson P, Copplestone JA,
For major ABO mismatch, use group O red cell prod- Gooi HC, Morgenstern GR, et al. Bedside filtration of blood
ucts, irrespective of ABO group until recipient ABO products in the prevention of HLA alloimmunizzation- a
antibodies are undetectable, and the antiglobulin test is prospective randomised study. Alloimmunisation Study
Group. Blood 1994; 83:3028-35.
negative, and platelets and plasma from donors of the 14. BCSH. Blood Transfusion Task Force Guidelines on the clini-
recipients ABO type until recipient red cells are no cal use of leucocyte-depleted blood components. Transfus
longer detected. Med 1998;8:59-71.
For minor ABO mismatch, use red cells of the donor 15. BCSH Blood Transfusion Task Force. Guidelines on γ irradia-
tion of blood components for the prevention of transfusion-
type, i.e., group O throughout. Give platelets and plas- associated graft-versus-host disease. Transfus Med 1996; 6:
ma of recipient type until recipient-type red cells are no 261-71.
longer detected. 16. Stanworth SJ, Hyde C, Heddle N, Rebulla P, Brunskill S,
Murphy MF. Prophylactic platelet transfusion for haemorrhage
For major and minor ABO mismatch, use group O red after chemotherapy and stem cell transplantation. Cochrane
cells until recipient ABO antibodies are undetectable Database Syst Rev 2004; 18:CD004269.
and the antiglobulin test is negative, then switch to 17. Wandt H, Schaefer-Eckart K, Frank M, Birkmann J, Wilhelm
donor type. For platelets and plasma use group AB until M. A therapeutic platelet transfusion strategy is safe and fea-
sible in patients after autologous peripheral blood stem cell
recipient red cells are undetectable. transplantation. Bone Marrow Transplant 2006;37:387-92.
Following graft rejection, revert to recipient-type red 18. Weissinger F, Sandmaier BM, Maloney DG, Bensinger WI,
cells and platelets. Gooley T, Storb R. Decreased transfusion requirements for
patients receiving nonmyeloablative compared with conven-
tional peripheral blood stem cell transplants from HLA-identi-
Conclusions cal siblings. Blood 2001;98:3584-8.
Transfusion support in BMT patients requires special 19. Brocklebank JT, Harcourt RB, Meadow SR. Corticosteroid –
induced cataracts in idiopathic nephrotic syndrome. Arch Dis
consideration and carefully defined policies. The use of Child 1982;57:30-4.
high quality blood components, which have a high 20. Heufth HG, Goudeva L, Sel S, Blasczyk R. Equivalent mobi-
degree of microbiological safety, are also gamma-irradi- lization and collection of granulocytes for transfusion after
administration of glycosylated G-CSF (3 µg/kg) plus dexam-

| 286 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

ethasone versus glycosylated G-CSF (12 µg/kg) alone. haematopoietic stem cell transplantation. Br J Haematol 2002;
Transfusion 2002;42:928-34. 119:740-50.
21. Kerr JP, Liakopolou E, Brown J, Cornish JM, Fleming D, 25. Canals C, Muñiz-Díaz E, Martínez C, Martino R, Moreno I,
Massey E, et al. The use of stimulated granulocyte transfu- Ramos A, et al. Impact of ABO incompatibility on allogeneic
sions to prevent recurrence of past severe infections after allo- peripheral blood progenitor cell transplantation after reduced
geneic stem cell transplantation. Br J Haematol 2003;123:114- intensity conditioning. Transfusion 2004;44:1603-11.
8 26. Griffith LM, McCoy JP Jr, Bolan CD, Stroncek DF, Pickett AC,
22. Stanworth SJ, Massey E, Hyde C, Brunskill S, Lucas G, Navar- Linton GF, et al. Persistence of recipient plasma cells and anti-
rete C, et al. Granulocyte transfusions for treating infections in donor isohaemagglutinins in patients with delayed donor ery-
patients with neutropenia or neutrophil dysfunction.
thropoiesis after major ABO incompatible non-myeloablative
Cochrane Database Syst Rev 2005;CD005339
23. Helbig G, Stella-Holowiecka B, Wojnar J, Krawczyk M, haematopoietic cell transplantation. Br J Haematol 2005; 128:
Krzemien S, Wojciechowska-Sadus M, et al. Pure red-cell apla- 668-75.
sia following major and bi-directional ABO-incompatible allo- 27. Worel N, Kalhs P, Keil F, Prinz E, Moser K, Schulenburg A, et
geneic stem cell transplantation: recovery of donor-derived al. ABO mismatch increases transplant-related morbidity and
erythropoiesis after long-term treatment using different thera- mortality in patients given nonmyeloablative allogeneic HPC
peutic strategies. Ann Hematol 2007;86:677-83. transplantation. Transfusion 2003;43:1153-61
24. Maciej Zaucha J, Mielcarek M, Takatu A, Little MT, Gooley T, 28. Pamphilon DH. Transfusion Policy in the EBMT Handbook.
Baker J, et al. Engraftment of early erythroid progenitors is not 5th Edition. Haemopoietic Stem Cell Transplantation 2008. p.
delayed after non-myeloablative major ABO-incompatible 146-62.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 287 |
Transfusion

Transfusion issues in neonatal and pediatric


hematology

N.L.C. Luban Introduction to physiologic decreases in erythropoietin


Blood component transfusion is integral to (EPO) levels, the hgb concentrations decreas-
the treatment of infants and children cared es to a nadir of 10-12 gm/dL at aproximately
George Washington University
School of Medicine and Health for by a wide range of generalists, pediatri- 2 to 3 months before a secondary rise. This
Sciences Professor, Pediatrics cians, neonatologists, surgeons, intensivists, is referred to as the physiologic anemia of
& Pathology, Children's National and hematologists/oncologists. Technolo- infancy. In preterm infants, this anemia is
Medical Center, Washington, DC, gical advances in blood collection, separa- more significant with mean nadirs of 8
USA tion, preservation and storage have resulted gm/dL in very low birth weight (VLBW)
in red blood cell, platelet, white blood cell, infants (birthweight: 1.0-1.5 kg), and 7
and plasma products, which are superior to gm/dL in extremely low birth weight
Hematology Education: whole blood used in the past. Advances in (ELBW) infants (birthweight: <1 kg). This is
the education program for the donor selection, infectious disease testing, the result of lower hgb concentrations at
annual congress of the European use of leukoreduction (LR) filters and gamma birth, frequent blood sampling, low total
Hematology Association irradiation ensure that products are less like- blood volume to blood sampling ratio,
ly to cause adverse reactions, but issues increased risk for other comorbidities, and
2009;3:288-293 remain. Areas of particular concern include diminished capacity of the premature infant
poorly defined indications, unintentional to increase EPO.2 Tissue hypoxia ensues
adverse and long-term consequences, and when peripheral vascular resistance increas-
the need to alter products to meet the es. This is, at least, partly due to the infant’s
unique needs of children with specific diag- inability to improve left ventricular stroke
noses. Transfusion practices in infants less volume without an increase in heart rate
than 4 months of age differ markedly from when greater than 10% of blood volume has
transfusion in older children, which more been lost. Infants with significant cardiac or
closely resemble adult practices. respiratory disease generally receive more
Neonates have a small total blood volume aggressive RBC transfusion therapy.3,4
when compared to older children and adults Normal ranges of hgb and hematocrit have
with high blood volume per kilogram of been established for full-term and premature
body weight. Their immature organs and infants; however, guidelines for red cell
relative immunologic immaturity predispos- transfusion therapy remain controversial
es them to specific hazards of transfusion because of a paucity of randomized con-
including metabolic complications, infec- trolled studies addressing appropriate
tions and immuno modulation.1 Their rapid neonatal transfusion triggers. Guidelines for
growth is coupled with a limited capacity to replacement transfusion therapy in neonates
expand their blood volume. The infant’s are given in Table 1.
response to stresses, such as hypothermia, Two recent studies attempted to address
hypovolemia, hypoxia, acidosis, and comor- high versus low threshold transfusion guide-
bidities, warrant special considerations lines based on level of respiratory support in
before and during the transfusion process. ELBW and VLBW infants. Although differ-
This monograph will concentrate on issues ent in design and outcome, neither study
relevant to newborns. clearly established an appropriate hemoglo-
bin target. While the multi-institutional
Transfusion of RBCs Canadian Premature Infants in Need of
RBC transfusions in newborns are admin- Transfusion (PINT) study5 demonstrated no
istered to replace acute or chronic blood loss advantage for liberal transfusion practices,
or treat anemia of prematurity. Blood loss the American study6 suggested that restric-
can result from hemorrhage or phlebotomy. tive transfusion was associated with, apneic
While iatrogenic losses from phlebotomy more episodes, intraparenchymal brain
can be considerable, they can be minimized hemmorhage, and periventricular leukoma-
by combining judicious testing strategies, lacia. Unfortunately, the question of long-
sampling from indwelling catheters, micro- term neurodevelopmental outcome has not
tizing laboratory assays and implementing yet been addressed by either study. The use
point-of-care testing. of recombinant human EPO (rhEPO) has
been proposed to decrease neonatal transfu-
Indications sion burden, donor exposure, anemia of pre-
In the first few days of life, full term maturity, as well as other comorbidities.
infants have an elevated hemoglobin (hgb) Meta-analysis of late (>8 days of life) rhEPO
concentration (14-20 gm/dL); however, due use involving 1302 preterm infants showed

| 288 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

Table 1. Transfusion guidelines for RBCs in infants less Table 2. Platelet transfusion guidelines in neonates and
than 4 months of age.1,24 older children.1,24

Hematocrit <20% with low reticulocyte count and symptoms of anemia With Thrombocytopenia
(tachycardia, tachypnea, poor feeding) • Platelet count 5,000 to 10,000/µL with failue of platelet production
• Platelet count <30,000/µL in neonate with failure of platelet production
Hematocrit <30% and any of the following: • Platelet count <50,000/µL in stable premature infant with:
• On <35% oxygen hood a. Active bleeding, or
• On oxygen by nasal cannula b. Before an invasive procedure with failure of platelet production
• On continuous positive airway pressure and/or intermittent mandatory • Platelet count <100,000/µL in sick premature infant with:
ventilation on mechanical ventilation with mean airway pressure a. Active bleeding, or
<6 cm of water b. Before an invasive procedure in patient with DIC
• With significant tachycardia or tachypnea (heart rate>80 beats/minute for 24
Without Thrombocytopenia
hours, respiratory rate>80 beats/minute for 24 hours)
• Active bleeding in association with qualitative platelet defect
• With significant apnea or bradycardia (>6 episodes in 12 hours or 2 episodes
• Unexplained excessive bleeding in a patient undergoing cardiopulmonary
in 24 hours requiring bag and mask ventilation while receiving therapeutic
bypass
doses of methylxanthines)
• Patient undergoing ECMO with:
• With low weight gain (<10 g/day observed over 4 days while
a. A platelet count of <100,000/µL
receiving >100 kcal/kg/day)
b. Higher platelet counts and bleeding
Hematocrit <35% and either of the following:
• On >35% oxygen hood DIC : disseminated intravascular coagulation; ECMO: extracorporeal membrane
oxygenation
• On continuous positive airway pressure/intermittent mandatory ventilation
with mean airway pressure ≥6-8 cm of water
Hematocrit <45% and either of the following:
• On extracorporeal membrane oxygenation Pretransfusion testing and product selection
• With congenital cyanotic heart disease Prior to RBC transfusion, a blood sample is obtained
for ABO and Rh determination (blood group and type)
and to screen for antibodies against blood group anti-
gens.12 Because antibodies identified in the neonate’s
no clear benefit in terms of significantly decreasing blood are most often of maternal origin, maternal blood
donor exposures, nor was there any effect on preva- can often serve as the source of serum/plasma for the
lence of cormorbidities. Although late administration of antibody screen. A newborn’s ABO group is assigned
rhEPO reduced the number of RBC transfusions and the solely on the testing of the patient’s RBCs for the A and
total transfused volume of RBCs per infant, the clinical B antigens (forward typing), because the isohemagglu-
impact of these results is trivial (<1 transfusion per tinins anti-A and anti-B are not present in the serum at
infant and 7 mL/kg of RBCs). Avoiding the use of RBC birth. Cord blood specimens for infant blood type deter-
transfusions was not demonstrated because many mination should not be used because contamination
infants had received one or more transfusion prior to with Wharton’s jelly often hinders correct grouping, and
receiving rhEPO at study entry.7 A subsequent meta- because proper identification of the specimen in the
analysis designed to assess the effectiveness and safety delivery room may be problematic.
of early initiation of rhEPO (<8 days of life) in 1825 pre- If the newborn’s antibody screen is negative, ABO-
mature infants also showed no evidence of any substan- and Rh-specific RBCs may be transfused. Neither the
tial benefits with regard to donor blood exposure. antibody screen nor crossmatching need to be per-
However, a statistically significant increased risk of formed during the infant’s hospitalization during the
first 4 months of life. If the screening identifies passive-
retinopathy of prematurity (>grade 3) was noted in
ly acquired maternal blood group antibodies, as in Rh
neonates who received early rhEPO therapy. Again,
hemolytic disease of the newborn, then O-negative red
RBC transfusion was not avoided when RBCs trans-
cells should be transfused until repeat testing is negative
fused prior to study entry was considered.8,9 Therefore, for antibodies reacting against the ABO- or Rh-specific
no conclusive evidence currently exists that either early units. When antibodies to other RBC antigens other
or late rhEPO use offers any clear benefit in regard to than D are detected, blood should be selected that is
donor exposure or improvement of morbidity and/or ABO and Rh specific but antigen negative for the iden-
mortality in preterm infants, and that early rhEPO use tified antibody. In cases when reconstituted WB is need-
may increase the risk of ROP in the VLBW neonatal ed for large volume transfusion procedures (i.e.,
population. The use of rhEPO as a neuroprotectant is exchange transfusions, cardiopulmonary bypass,
under investigation. ECMO), the neonate may be given plasma that is ABO
Another area of interest is long-term outcomes follow- compatible with their RBCs, but receive RBCs that are
ing transfusion. Patient outcomes, especially mortality, compatible with maternal serum. This may mean that
have been studied in adults, leading to recommendations the ABO group of the RBC and FFP units are different.
for lowering transfusion triggers. Only a couple of stud- In circumstances of multiple transfusions access ABO
ies have addressed pediatric patients10,11 but neither were group, we advise crossmatching of red blood cell units.
designed to specifically address neonates nor the long- An alternative used by some transfusion centers entails
term adverse effects. This is an area of increasing interest, the use of low isohemagglutinin titer, group-O WB, if
which warrants large scale multi-institutional studies. available.2

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 289 |
14th Congress of the European Hematology Association

In infants older than 4 months of age, repeat testing Table 3. Tranfsuion guidelines for plasma products in
for blood group, Rh-type and antibody screening is per- neonates and older children.1,24
formed within 72 hours of each red blood cell transfu-
sion if the patient has received a transfusion during the FFP
last 3 months, or if the history is uncertain or unavail- • Support during treatment of DIC
able.2 Reverse ABO blood group testing, which detects • Replacement therapy:
anti-A and anti-B antibodies, is frequently deferred for a. When specific factor concentrates are not available, including but not limited
the first 6 months of life because this testing is often to, antithrombin, protein C or S deficiency, and Factor II, Factor V, Factor X, and
weak or nonreactive in babies less than 1 year of age. Factor XI deficiencies
RBC viability and functional activity require that b. During therapeutic plasma exchange when FFP is indicated (cryopoor asthma,
RBCs be preserved in additive solutions that support plasma from which the cryoprecipitate has been removed)
their metabolic demands. All anticoagulant/preservative • Reversal of warfarin in an emergency situation, such as before an invasive
(AP) solutions contain citrate, phosphate, and dextrose procedure with active bleeding
(CPD), which function as an anticoagulant, a buffer and Note: FFP is not indicated for volume expansion or enhancement of wound
a source of RBC metabolic energy, respectively; the healing.
addition of mannitol and adenine as preservatives to
anticoagulant solutions increases the shelf life of RBCs Cryoprecipitate
from 21 days (CPD) to 35 days (CPDA-1) and to 42 days • Hypofibrinogenemia or dysfibrinogenemia with active bleeding
for newer AP solutions (AS1, AS3, AS5) by stabilizing • Hypofibrinogenemia or dysfibrinogenemia, undergoing an invasive procedure
the RBC membrane and maintaining 2.3-diphospho- • Factor XIII deficiency with active bleeding or undergoing an invasive procedure
glycerate (DPG) and adenosine triphosphate (ATP)
in the absence of Factor XIII concentrate
within the stored RBCs. Studies have shown that blood
• Limited directed-donor cryoprecipitate for bleeding episodes in small children
collected in AP solutions is safe and as efficacious as
with hemophilia A (when recombinant and plasma-derived Factor VIII products
CPDA-1 RBCs in increasing the Hct for neonates receiv-
ing small volume (10-15 mL/kg) RBC transfusions.13,14 are not available
These products can be dispensed as small aliquots from • In the preparation of fibrin sealant
one RBC unit (300-350 mL) to one or more neonates • von Willebrand’s disease with active bleeding but only when both of the
who require multiple transfusions in order to decrease following are true:
donor exposure and to conserve RBC inventory. Sterile a. Deamino-D arginine vasopressin (DDAVP) is contraindicated, not available, or
connecting devices ensure that the original RBC unit does not elicit response.
remains a closed system and transfer packs or syringe b. Virus-inactivated plasma-derived Factor VIII concentrate (which contains von
sets permit multiple aliquots to be removed. Studies Willebrand factor) is not available
have shown that CPDA-1 and AS-3-preserved split RBC
packs effectively limit donor exposures, and are safe for
use in neonatal small volume transfusions until 35 days
of storage.15 However, clinical studies have neither con- 50,000/uL. However, this study did not address bleed-
firmed or refuted the effect of an AP on metabolic ing risk or transfusion benefit for neonates with platelet
abnormalities in massive transfusion for the neonate. counts below 50,000/mL.17
Therefore, many experts recommend avoiding RBCs A number of platelet transfusion guidelines for the
stored in AP solutions for large volume transfusions newborn have been proposed. Given the lack of clinical
until such data have been published.16 Because hypo- trials addressing absolute bleeding risk in thrombocy-
thermia can trigger hypoglycemia, metabolic acidosis topenia caused by different etiologies, these recommen-
and apnea in line blood warmers should be used when dations are based on expert panel recommendations
infants receive large volume transfusions. garnered from clinical experience. Becasue of concerns
for the outcomes secondary to intraventricular hemor-
Platelet transfusion rhage (IVH) in the sick neonate, neonatologists have tra-
Indications ditionally adopted an aggressive platelet threshold for
Platelet transfusions are administered to neonates transfusion. Murray et al. retrospectively studied 53
therapeutically or prophylactically to prevent the hem- neonates (44 preterm) with severe thrombocytopenia,
orrhagic complications of thrombocytopenia. Neonates and concluded that a threshold of 30,000/mL, without
have different risks of bleeding given the same degree of other risk factors or previous IVH, is safe for the major-
thrombocytopenia. For example, thrombocytopenic ity of neonates.18 Thus, a generally accepted transfusion
neonates with neonatal alloimmune thrombocytopenia trigger for platelet count less than 30,000/mL has been
(NAIT) have a high risk of major bleeding, whereas endorsed for neonates without other risk factors.
those with sepsis or necrotizing enterocolitis, and those However, some propose a higher trigger (<50,000/mL)
with intra-uterine growth retardation have an interme- for ELBW neonates within the first week of life, clinical-
diate and low risk of major hemorrhage, respectively. ly unstable neonates, and neonates with NAIT (using
Platelet functions either intrinsic or secondary to drugs HPA-compatible platelet products).3,19-20 Platelet transfu-
or comorbidities like coagulopathy are likely causes for sions are also indicated to treat hemorrhage associated
these discrepancies. The only randomized controlled with acquired (i.e., ECMO, cardiopulmonary bypass,
trial addressing whether platelet transfusions reduce uremia) or congenital qualitative platelet abnormalities
major bleeding in neonate found no benefit of maintain- (i.e., Glanzman thrombasthenia, Bernard-Soulier syn-
ing a normal platelet count (>150,000/uL) in preterm drome), even when the platelet count is within the nor-
neonates compared to those maintained at greater than mal range.

| 290 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

Pretransfusion testing and product selection tion and release. Transfusion of granulocytes has been
Platelets express ABO but not Rh antigens. They employed in the treatment of neonatal sepsis; however,
should be ABO-group specific whenever possible, due their efficacy is controversial. Meta-analysis of the safe-
to reports of intravascular hemolysis following transfu- ty and efficacy of granulocyte infusion adjunctive to
sion of ABO-incompatible platelets in infants and chil- antimicrobial therapy in the treatment of septic neu-
dren.21 Platelets do not routinely require crossmatching. tropenic neonates failed to show a reduction in morbid-
If ABO-incompatible platelets must be used, plasma ity or mortality.25
removal via volume reduction or washing, or selecting
low isohemagglutinin (anti-A, anti-B) titer units are Granulocyte preparations
options. Routine volume reduction methods for all Granulocyte concentrates for neonatal transfusion are
neonates should be avoided because approximately prepared by automated leukopheresis of healthy stimu-
20% of platelets are lost in the final product, which is lated (Dexamethasone +/-GCSF) donors, and contain 1-
resuspended in either saline or compatible plasma.22 2×109 neutrophils per kilogram in a volume of 10 to 15
Although Rh matching does not affect post-transfusion mL/kg. Treatment should be continued daily until clini-
platelet survival, RBCs are present in small amounts of cal improvement or neutrophil count recovery (ANC
whole blood derived platelet concentrates, which can >3000/mL in first week of life; >1500/mL thereafter). All
cause Rh sensitization in an Rh-negative recipient. granulocytes should be gamma irradiated and CMV
Administration of Rh immune globulin (RhIG) should negative, since leukodepletion is contraindicated.
be strongly considered for any Rh-negative neonate, Because granulocyte concentrates have a significant
especially females, within 72 hours of exposure to Rh- amount of RBCs (Hct: 15-20%), the component must be
positive RBCs through a platelet transfusion. The rec- ABO, Rh, and crossmatch compatible with the intend-
ommended dose is 120 IU RhIG per mL of RBCs trans- ed neonatal recipient. Because granulocytes must be
fused, administered intramuscularly (90 IU RhIG per mL transfused within 24 hours of collection, FDA-mandat-
of RBCs intravenously).23 When aliquots of apheresis ed testing for blood products will not be completed
platelets, which are virtually RBC free are used, there before the product is released for administration; there-
should be limited to no concern for Rh sensitization. fore the risks and benefits of transfusing an untested
blood product must be weighed by the medical team
Component definitions and the parents of the infant.2,24
A whole blood derived platelet unit contains at least Unique risks to granulocyte transfusion include pul-
5.5×1010 platelets in 50- 70 mL of plasma. Apheresis monary reactions from mild transient respiratory dis-
platelets , also called single-donor platelets (SDP), con- tress, severe pulmonary edema, hypoxia, ARDS and a
tain a minimum of 3×1011 platelets in approximately 250 high incidence of febrile transfusion reactions.
mL (range: 200-400 mL) of plasma; the equivalent of an Pulmonary complications have been reported in 4% of
estimated six units of WB-derived platelets. SDPs are transfused infants, and severe pulmonary reactions
split for neonatal use and offer the advantage of avoid- resembling TRALI have been reported.25,26 Mild to mod-
ing multiple donor exposures. Platelets are optimally erate reactions occur in 25-50% and severe reactions in
stored at 20-24ºC under constant agitation, and have a about 1% of all granuloctye transfusions.2
shelf life of 5 days after collection. Transfusion of 10-15 Although granulocyte colony-stimulating factor (G-
mL/kg of platelets for neonates, or 0.1-0.2 Unit/kg for CSF) and granulocyte-macrophage colony-stimulating
children over 10 kg, should yield a platelet increment of factor (GM-CSF) have been used successfully to stimu-
50,000/mL to 100,000/mL if no predisposing risk factors late neutrophil numbers, and IVIG has been attempted
for refractoriness exist.24 It is important to account for to augment traditional antimicrobial therapy to support
device related dead space (10-30 mL) when issuing the septic neonates, data on efficacy are inconclusive.27,28
product, as this can be considerable in relation to the Due to the lack of clear consensus on their impact on
overall platelet dose. The expected rise in the platelet improving host defense mechanisms and improving
count after transfusion may not be met because of outcomes, these adjuncts to standard antimicrobial and
destructive thrombocytopenia, which occur with antifungal therapy need further investigation.
splenomegaly, fever, sepsis, disseminated intravascular
coagulation (DIC), bleeding, or antibiotic therapy. Transfusion of plasma
Immune-mediated causes of platelet refractoriness, such Indications
as alloantibodies to platelet specific antigens (i.e. HPA- FFP is used primarily to treat acquired coagulation fac-
1a, 5b) in NAIT, and autoantibodies to common platelet tor deficiencies because of DIC, liver failure, vitamin K
antigens in ITP, require consideration in this population, deficiency from malabsorbtion, biliary disease or war-
so that appropriate management can be initiated.2 farin therapy, or dilutional coagulopathy from massive
transfusion. It can also be used for specific factor
Granulocyte transfusion replacement in congenital factor deficiencies (i.e., factor
Indications II, V, X, XI, protein C, protein S, antithrombin, etc.)
Indication VLBW neonates are susceptible to over- when specific factor concentrates or recombinant prod-
whelming bacterial infection. Many factors contribute ucts are either not manufactured or unavailable.2,3,12,24 FFP
to this risk, including disruption of mucosal barriers, is not indicated for volume expansion, enhancement of
hypogammaglobulinemia, and qualitative neutrophil wound healing, or as first-line treatment for congenital
defects. Furthermore, preterm neonates may become factor deficiencies when either a virally-inactivated
neutropenic during sepsis due to a marginal myeloid plasma derived factor concentrate or recombinant fac-
storage pool and poor myeloid progenitor cell prolifera- tor is available.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 291 |
14th Congress of the European Hematology Association

Plasma preparations difficult to obtain depending on donor demographics.


Plasma can be prepared by either WB separation or by Because CMV is harbored within WBC s, decreasing
apheresis. When the plasma product is frozen to -18ºC leukocyte number and viability would theoretically
or colder within 8 hours of collection it is labeled as reduce transmission of CMV; LR (<5×106 WBCs per
fresh frozen plasma (FFP), (approximately 250-300 mL), unit) has been shown to be effective in preventing CMV
and can be stored at this temperature for up to 1 year.2 infection in adults with hematopoietic malignancies,
In order to conserve plasma inventory and limit donor neonates and patients post stem cell transplant. It has
exposures for infants who receive only fractions of an been widely debated whether LR is as efficacious as
FFP unit, plasma can be separated into a system of mul- CMV-seronegative blood. In a landmark study, Bowden
tiple satellite bags, and frozen as aliquots. Once thawed, et al.33 found equivalent rates of post transfusion CMV
FFP can be further subdivided into aliquots for multiple infection in an allogeneic hematopoietic stem cell popu-
neonates via sterile connecting devices, stored at 1-6ºC lation (1.4% for seronegative versus 2.4% for LR).
for up to 5 days and transfused as thawed plasma (TP).21 Although this study’s conclusions have been debated
Although TP has approximately 40% Factor V and VIII widely, no formal consensus on the debate of equiva-
(heat labile factors) activity, effective hemostasis is lency has been formulated. A subsequent study by
maintained at this level, making TP clinically similar to Nichols et al.35 demonstrated that, although LR platelet
FFP. Plasma transfusions should be ABO-compatible products were deemed similar to CMV-seronegative
with the neonate’s RBCs to avoid passive transfer of iso- products in regard to transfusion transmission of CMV,
hemagglutinins from ABO-incompatible plasma which LR depleted RBCs were not. The authors warned
can result in hemolysis.21,24 Because the freezing process against the practice of abandoning dual inventory blood
renders the frozen-thawed plasma component free of products for CMV-seronegative and seropositive units.
viable leukocytes, FFP is not screened for CMV anti- Nonetheless, variable practices currently exist.35 Many
body, nor are leukoreduction and irradiation necessary institutions use algorithms based on pre-transplant
for the prevention of CMV reactivation and TA-GVHD, serostatus of recipient, serostatus of the hematopoietic
respectively. However, passive administration of anti- stem cell donor, and the donor demographics within the
body to CMV in FFP may cause CMV IgG testing to area. It is unlikely that further randomized controlled
become positive in the transfused neonate. This does trials will be performed to assess comparability of CMV
not represent CMV infection, and the antibody disap- seronegative versus leukoreduced products.
pears in a time course consistent with the 21-day half-
life of γ-globulin.29 Transfusion-associated graft-versus-host-disease
FFP is typically dosed at 10-15 mL/kg and will replace Transfusion-associated graft versus host disease (TA-
approximately 10-30% of most factors immediately fol- GVHD) occurs when an immunosuppressed or immun-
lowing transfusion. However, the half lives of different odeficient patient receives cellular blood products,
factor(s) vary which impacts dosing intervals.24 which possess immunologically competent lympho-
Furthermore, vitamin K dependent factors are lower in cytes. The transfused donor lymphocytes are able to
neonates prolonging both the PT and aPTT. Correlation proliferate and engraft in the immunologic incompetent
of laboratory values to clinical status is important when recipient because they are unable to detect and reject
devising an FFP dosing schedule for a coagulopathic foreign cells. The degree of similarity between HLA
infant. antigens also increases the ability of donor lymphocytes
to engraft with the recipient. This fact explains why TA-
Specialized products GVHD can occur in situations of directed donation
Leukoreduction of blood components among family members. In the event where the donor
Current third-generation LR filters consistently pro- is homozygous and the recipient is heterozygous for an
vide a 3-4 log or 99.9% reduction of white blood cell HLA antigen, donor lymphocytes may escape immune
content to less than 5×106 white blood cells and, with surveillance and engraft in the immunocompetent host,
some filters, less than 1×106 per product. This LR step is resulting in TA-GVHD. This situation may also occur in
best performed prestorage with good quality control populations with limited HLA variability, thus necessi-
techniques. Febrile nonhemolytic transfusion reactions tating universal gamma-irradiation of all cellular compo-
are typically caused by reactions to donor white blood nents in specific situations.
cells or to cytokines present in the product, and are Clinical symptoms of TA-GVHD include fever and an
reduced when in pre-storage LR products are used.30,31-32 erythematous rash, which may progress to bullae and
Alloimmunization to foreign HLA class I antigens is also desquamation and diarrhea developing within 3 to 30
reduced. LR has also been used to reduce transmission days of receiving cellular blood components. Because
of cytomegalovirus (CMV) in high-risk patient popula- the hematopoietic progenitor cells are particularly
tions. Recipient groups at increased risk for post-trans- affected, severe cytopenia usually is present. Mild hep-
fusion CMV related morbidity and mortality include: atitis to fulminant liver failure may occur. Mortality for
• Premature, seronegative, neonates less than 1250 g, TA-GVHD is 90% in the pediatric population. Patients
who require blood component support at high risk of TA-GVHD include:
• Recipients of hematopoietic stem cell and solid- • Patients with congenital immunodeficiencies, par-
organ transplants ticularly of cellular immunity
• Fetuses who receive intrauterine transfusions • Those receiving intrauterine transfusion followed
• Other severely immunocompromised individuals.1,24 by neonatal exchange transfusion
Historically, the use of CMV-seronegative blood was • Bone marrow transplant recipients
considered as the gold standard; such products are often • Recipients of HLA-matched cellular components, or

| 292 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

blood components from blood-related donors 12. Price TH, editor. Standards for Blood Banks and Transfusion
services. 25th edition. Bethesda (MD, USA): American
• Patients with hematologic malignancies and cancer Association of Blood Banks; 2008. p. 41-7.
patients undergoing intense chemotherapy or immuno- 13. Strauss RG, Burmeister LF, Johnson K, Cress G, Cordle D.
modulatory therapy (i.e. Fludarabine, and other purine Feasibility and safety of AS-3 red blood cells for neonatal
analogs).35 transfusions. J Pediatr 2000;136:215-9.
14. Jain R, Jarosz C. Safety and efficacy of AS-1 red blood cell use
Neonates, especially those who are extremely prema- in neonates. Transfus Apher Sci 2001;24:111-5.
ture, are considered by many to be at high risk for TA- 15. Mangel J, Goldman M, Garcia C, Spurll G. Reduction of donor
GVHD. Whereas some neonatal centers irradiate all cel- exposures in premature infants by the use of designated ade-
nine-saline preserved split red blood cell packs. J Perinatol
lular blood products for infants less than 4 months of 2001;6:363-7.
age, others only irradiate blood products given to 16. Luban NL, Strauss RG, Hume HA. Commentary on the safety
preterm infants born at less than 1.2 kg.36 However, of red cells preserved in extended storage media for neonatal
transfusions. Transfusion 1991;31:229-35.
given the lack of clinical studies on the incidence of TA- 17. Andrew M, Vegh P, Caco C. A randomized, controlled trial of
GVHD in the neonatal population, and concern for fail- platelet transfusions in thrombocytopenic premature infants, J
ing to recognize an infant with an undiagnosed congen- Pediatr 1993;123:285-91.
18. Murray NA, Howarth LJ, McCloy MP, Letsky EA, Roberts IA.
ital immunodeficiency, there exists no standard of care Platelet transfusion in the management of severe thrombocy-
regarding irradiation of blood products for otherwise topenia in neonatal intensive care unit patients. Transfus Med
non-high risk infants born greater than 1.2 kg. 2002;12:35-41.
TA-GVHD can be prevented by γ-irradiation of cellu- 19. Roberts I, Murray NA. Neonatal thrombocytopenia. Semin
Fetal Neonatal Med 2008;13:256-64.
lar blood components at a minimum of 2,500 cGy using 20. Blanchette VS, Kühne T, Hume H, Hellmann J. Platelet trans-
cesium, cobalt or linear accelerators as radiation fusion therapy in newborn infants. Transfus Med Rev 1995;
sources.2 Because in vivo recovery of irradiated RBCs is 9:215-30.
21. Angiolillo A, Luban NL. Hemolysis following an out-of-group
decreased compared to non-irradiated RBCs at 42 days platelet transfusion in an 8-month-old wih Langerhans cell his-
of storage, the FDA recommends a 28-day expiration tiocytosis. J Pediatr Hematol Oncol 2004;26:267-9.
for irradiated RBCs.12 Potassium and free hemoglobin 22. Moroff G, Friedman A, Robkin-Kline L, Gautier G, Luban NL.
Reduction of the volume of stored platelet concentrates for
are also increased after irradiation and storage of RBCs. use in neonatal patients. Transfusion 1984;24:144-6.
Therefore, it is preferable to irradiate in a time frame 23, Kennedy MS. Perinatal issues in transfusion practice. In
close to administration, especially for neonates who are Roback JD, editor. Technical Manual of the American
Association of Blood Banks. 16th ed. Bethesda (MD):
sensitive to large potassium loads. Irradiation of American Association of Blood Banks; 2008. p. 625-37.
platelets and granulocytes does not affect the function 24. Josephson CD. Neonatal and pediatric transfusion practice. In
of component products. Roback JD, editor. Technical Manual of the American
Association of Blood Banks. 16th ed. Bethesda (MD):
American Association of Blood Banks; 2008. p. 639-63.
25. O'Connor JC, Strauss RG, Goeken NE, Knox LB. A near-fatal
References reaction during granulocyte transfusion of a neonate.
Transfusion 1988;28:173-6.
1. Luban NL, Wong EC. Hazards of transfusion. In: Arceci RJ, 26. Sanchez R, Toy P. Transfusion related acute lung injury: A
Hann IM, Smith OP, editors. Pediatric Hematology. 3rd ed. pediatric perspective. Pediatr Blood Cancer 2005;45:248-55.
2006. p. 724-44. 27. Sandberg K, Fasth A, Berger A, Eibl M, Isacson K, Lischka A,
2. Roseff SD, editor. Pediatric Transfusion: A physician’s hand- et al. Preterm infants with low immunoglobin G have
book. 2nd edition. Bethesda (MD): American Association of increased risk of neonatal sepsis but do not benefit from pro-
Blood Banks; 2006. p. 1-52. phylactic immuoglobin G. J Pediatr 2000;623-8.
3. Roseff SD, Luban NL, Manno CS. Guidelines for assessing 28. Carr R, Modi N, Doré C. G-CSF and GM-CSF for treating or
appropriateness of pediatric transfusion. Transfusion 2002; 42: preventing neonatal infections. Cochrane Database Syst Rev
1398-413. 2003;3:CD003066.
4. Widness JA. Treatment and prevention of neonatal anemia. 29. Robitaille N, Hume HA, Blood components and fractionated
NeoRev 2008;9:e526-33. plasma products: preparations, indications, and administra-
5. Kirpalani H, Whyte RK, Andersen C, Asztalos EV, Heddle N, tion. In: Arceci RJ, Hann IM, Smith OP editors. Pediatric
Blajchman MA, et al. The Premature Infants in Need of Hematology. 3rd edition. 2006. p. 693-706.
Transfusion (PINT) study: a randomized, controlled trial of a 30. Yazer MH, Podlosky L, Clarke G, Nahirniak SM. The effect of
restrictive (low) versus liberal (high) transfusion threshold for prestorage LR on the rates of febrile nonhemolytic transfusion
extremely low birth weight infants. J Pediatr 2006;149:301-7. reactions to PC and RBC. Transfusion 2004;44:10-5.
6. Bell EF, Strauss RG, Widness JA, Mahoney LT, Mock DM, 31. Paglino JC, Pomper GJ, Fisch GS, Champion MH, Snyder EL.
Seward VJ, et al. Randomized trial of liberal versus restrictive Reduction of febrile but not allergic reactions to red cells and
guidelines for red blood cell transfusion in preterm infants. J platelets following conversion to universal prestorage LR.
Pediatr 2005; 115:1685-91. Transfusion 2004;44:16-24.
7. Aher S, Ohlsson A. Late erythropoietin for preventing red 32. King TE, Tanz W, Shirey S. Universal LR decreases the inci-
blood cell transfusion in preterm and/or low birth weight dence of febrile nonhemolytic transfusion reactions to red
infants. Cochrane Database Syst Rev 2006;3:CD004868. cells. Transfusion 2004;44:25-9.
8. Mainie P. Is there a role for erythropoietin in neonatal medi- 33. Bowden RA. A comparison of filtered leukocyte-reduced and
cine? Early Hum Dev 2008;84:525-32. cytomegalovirus (CMV) seronegative blood products for the
9. Ohlsson A, Aher SM. Early erythropoietin for preventing red prevention of transfusion-associated CMV infection after mar-
blood cell transfusion in preterm and/or low birth weight row transplant. Blood 2003;86:3598-603.
infants. Cochrane Database Syst Rev 2006;3:CD004863. 34. Nichols WG, Price TH, Gooley T, Corey L, Boeckh M.
10. Slonim AD, Joseph JG, Turenne WM, Sharangpani A, Luban Transfusion-transmitted cytomegalovirus infection after
NL. Blood transfusion in children: a multi-institutional analy- receipt of leukoreduced blood products. Blood 2003;101:4195-
sis of practices and complications. Transfusion 2008;48:73-80. 220.
11. Lacroix J, Hébert PC, Hutchison JS, Hume HA, Tucci M, 35. Pisciotto PT, Luban NL. Complications of neonatal transfu-
Ducruet T, et al. Transfusion strategies for patients in pediatric sion. In: Popovsky MA, editor. Transfusion Reactions. 3rd edi-
intensive care units. TRIPICU Investigators; Canadian Critical tion. 2007;459-99.
Care Trials Group; Pediatric Acute Lung Injury and Sepsis 36. Strauss RG. Data-driven blood banking practices for neonatal
Investigators Network. N Engl J Med 2007;356:1609-19. RBC transfusions. Transfusion 2000;40:1528-40.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 293 |
Uncommon lymphoproliferative disorders

Natural killer cell disorders

G. Semenzato A B S T R A C T
R. Zambello Natural killer (NK) cell neoplasms account for less than 5% of all groups of lymphoid malignancies.
The recently updated World Health Organization classification in 2008 has made a great effort in
Padua University School of revisiting and ordering a series of disorders previously included in the pool of NK cell neoplasms.
Medicine, Department of Clinical Accordingly, blastic NK-cell lymphoma, which in the previous classification likely included blastic plas-
and Experimental Medicine, macytoid dentritic cell neoplasms, has been provisionally changed into NK cell lymphoblastic
Hematology and Clinical leukemia/lymphoma, since rare cases of bona fide immature NK lymphoid tumours have been report-
Immunology Branch, Padova, Italy ed in the literature. In addition, the new provisional entity defined Chronic Lymphoproliferative
Disorders (CLPD) of NK cells has been added to the list of mature T-cell and NK-cell neoplasms that
already included Aggressive NK cell leukemia and Extranodal NK/T lymphoma, nasal type. These last
Hematology Education: two NK cell tumours are more frequent in Asia and Central and South America, are often associated
the education program for the with Epstein-Barr virus (EBV) infection and show a very aggressive clinical course. By contrast, CLPD
annual congress of the European of NK cells are prevalent in Europe and USA, and show an indolent clinical course over a long period
Hematology Association of time, with some cases displaying a spontaneous remission. This paper focuses on recent concepts
2009;3:294-301 and progress in clinicopathologic features, pathogenesis, treatment approaches and outcomes of NK
cell disorders.

Introduction Classification of natural killer disorders


Natural killer (NK) cells represent the NK cell neoplasms are a rare and hetero-
major component of innate immune system geneous group of disorders with a broad
and mediate a MHC non-restricted cytotox- spectrum of morphologic, immunopheno-
icity against tumor cells and bacterial or viral typic, and clinical features. The World
infected cells.1-3 NK cells account for 10% of Health Organization (WHO) classification
peripheral blood lymphocytes, showing typ- encompasses four distinct entities, two of
ically large granular lymphocyte (LGL) mor- them are still provisional: (i) NK cell lym-
phology. NK cells are derived in bone mar- phoblastic leukemia/lymphoma (provision-
row from hematopoietic stem cells through al);8 (ii) chronic lymphoproliferative disor-
the intermediate developmental stages of der of NK cells (provisional);9 (iii) aggressive
lymphoid stem cells, bipotential T/NK pro- NK cell leukemia;10 (iv) extranodal NK/T-
genitor cells, and committed NK progenitor cell lymphoma, nasal type.11 In addition,
cells.4-6 For this reason, NK cells express vari- based on morphology, immunophenotype,
ably T-lineage-associated antigens (CD2 functional NK cell activity, and expression
and/or CD7). By definition, NK cells are sur- of cytotoxic molecules, NK cell neoplasms
face CD3-negative and myeloperoxidase can be divided into immature and mature
(MPO)-negative, and have germline configu- categories.12-16
ration of T-cell receptor (TCR) and In recent years, rare cases of lymphoblas-
immunoglobulin (Ig) genes.1-3 CD16, CD56, tic lymphomas/leukemia (LBL) arising from
and CD57 are NK-associated antigens. immature NK cells have been reported,13,15
Among these three markers, CD56 (a neural although the lack of suitable markers for
cell adhesion molecule) is most consistently immature NK cells mentioned above makes
expressed.3 Major advances in understanding it difficult to distinguish NK-cell lympho-
the actual role of NK cells in immune blastic lymphoma (LBL) from precursor T-
defence have been related to the discovery cell LBL. It is also worth mentioning that
and molecular characterization of numerous the plasticity of hematopoietic-cell lineage
surface receptors that play a crucial role in seems greater than previously thought, and
cell function.7 A key example is represented relations between phenotypically dissimilar
by the HLA-class I specific Ig killer-like neoplastic disorders are being reassessed. In
receptors (KIR) that play a crucial role in reg- contrast, it is believed that chronic lympho-
ulating NK cell function and are also useful proliferative disorder of NK cells, aggressive
in recognition of NK cell subsets. In this way, NK cell leukemia and extranodal NK cell
the molecular mechanisms that allow NK lymphoma and nasal type all originate from
cell to spare normal cells and kill tumor- or mature NK cells.12,17,18
virus-infected cells have now been clarified.
Table 1 summarizes the nomenclature of
most relevant NK receptors.

| 294 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

Precursor NK cell neoplasms Table 1. Nomenclature of natural killer receptor antigens.


NK cell lymphoblastic leukemia/lymphoma, provisional
A considerable confusion in the literature concerns NK Receptor specificity
this type of disorders, mostly due to the definition of CD designation* Gene Molecule mAb
NK cell leukemia according to the expression of CD56
antigen. Many cases reported as NK cell leukemia are Killer Immunoglobulin-like Receptors (KIR)
indeed cases of plasmacytoid dendritic cell leukemia19,20 CD158a KIR2DL1 p58.1 EB6
or acute myeloid leukemias.19,21 As a matter of fact, there CD158h KIR2DS1 P50.1 EB6
are rare reports of CD56+ immature lymphoid tumours CD158b1 KIR2DL2 p58.2 GL183
that are characterized by a blastic appearance of neo- CD158b2 KIR2DL3 p58.3 GL183
plastic cells with a CD4–/CD56+/CD13–/CD33– pheno- CD158j KIR2DS2 p50.2 GL183
type, lacking expression of CD3, CD19, CD20, and CD158i KIR2DS4 p50.3 FES172
MPO.22-25 These patients frequently presented with CD158d KIR2DL4 p49 ?
CD158e1 KIR3DL1 p70 Z27
leukemia and lymphadenopathy without skin involve-
CD158e2 KIR3DS1 p70 Z27
ment and were negative for EBV. TCR and/or Ig genes
CD158k KIR3DL2 p140 Q66
were in a germline configuration in all cases in which
the tests were performed. Outcomes of these patients Leukocyte Ig-like receptors/immunoglobulin like Ttranscripts (LIR/ILT)
were unfavorable. The immature morphology with NK CD85j LIR1/ILT2 LIR1/ILT2 F278
cell-associated phenotype and genotype suggests that CD85d LIR2/ILT4 LIR2/ILT4 42D1
these tumors represent true precursor NK cell neo-
plasms. Killer Lectin-like receptors (KLR)
Some well-characterized cases of NK precursor CD94 KP43 CD94 XA185
tumours with lymphomatous presentation that CD159a NKG2A/B NKG2A/B Z199
expressed CD94 1A transcripts have been reported.16 CD159c NKG2C NKG2C P25
Recent advances in the developmental biology of T cells CD314 NKG2D NKG2D BAT221
and NK cells indicate that both cell types are derived
from a common T/NK cell thymic precursor. Interleukin Natural cytotoxicity receptors (NCR)
15 (IL-15) and transcription factor ID2 are essential for CD335 NKp46 NKp46 BAB281
the NK cell lineage to diverge from the T cell lineage.15 CD336 NKp44 NKp44 Z231
CD94 1A, a distal promoter of the CD94 molecule (an CD337 NKp30 NKp30 AZZ20
NK cell receptor), is activated only by IL-15.26,27 Lin et al.16 NC NKp80 NKp80 MA152
recently reported that CD94 1A is the predominant
form found in immature NK cells and it is expressed in Co-receptors
CD244 2B4 2B4 C1.7.1
TCR-ve LBL (NK lineage LBL) but not in TCR+ve LBL (T
CD328 AIRM1 p75 Z176
lineage LBL). By studying 21 patients with LBL, and
based on the expression of CD94 1A transcripts and the NC: Not clustered. Regular characters indicate inhibiting molecules while italic
lack of TCR, these investigators identified seven denotes activating receptors. *CD are indicated according to 8th Human
Leukocyte Differentiation Workshop (HLDA8).91
patients with LBL of immature NK cell origin (CD94
1A+, TCR–). It is noteworthy that those NK-LBLs
occurred in young patients and had better outcomes as
compared with patients who had T-LBL (CD94 1A-, They are the most common type among primary
31,32

TCR+); none of the tumors were positive for CD56. lymphomas of the nasal cavity.33 The site of disease is
Thus, the use of CD94 1A associated with TCR appears primarily in the midline and includes the nasal cavity in
to be more precise than CD56 for identifying an imma- more than 80% of cases. The tumour is locally invasive
ture NK cell neoplasm. It is hoped that a wider availabil- and might infiltrate surrounding tissues and organs,
ity of more specific NK markers, including panels of such as oropharynx, palate, orbits, untill the appearance
KIR, KLR, and LIR1 will help to clarify this issue8 that of the characteristic midfacial destructive lesions, the so
still remains provisional. called lethal midline granuloma.31-33 Common symptoms
include nasal discharge, nasal obstruction, purulent rhi-
Mature natural killer cell neoplasms norrhea, epistaxis and local swelling of the nasal bridge.
Extranodal NK/T cell lymphoma, nasal type The tumours may be destructive, leading to the highly
Since they share the same histology, the WHO classi- characteristic midline perforation.
fication groups both nasal NK cell lymphoma and Extranasal NK cell lymphomas represent the counter-
extranasal NK cell lymphoma in the same category,11 part of nasal NK cell lymphomas, and involve any other
even if these lymphomas might have different clinical part of the body. Men are predominantly affected, and
manifestations, treatment approaches, and prog- the median age of presentation is the fifth decade.
noses.15,29 Although most cases are genuine NK-cell neo- Primary sites of involvement include the skin, gastroin-
plasms, the term NK/T rather than NK is used because testinal tract, salivary glands, spleen, and testis.34
this entity also includes cytotoxic T-cell neoplasms.30 Patients with extranasal NK cell lymphoma are more
Nasal and extranasal NK cell lymphoma are more preva- likely to exhibit an advanced stage of disease with sig-
lent in Asia, Mexico, and Central and South America11,29 nificantly higher general involvement, high levels of lac-
and are rare in Western countries. tate dehydrogenase and a significantly decrease in
Nasal NK/T cell lymphomas refer to tumours that hemoglobin and platelet count as compared with
occur in the nose and the upper aerodigestive tract.17,18, patients who have nasal NK cell lymphoma.34 The his-

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 295 |
14th Congress of the European Hematology Association

Figure 1. Hypothesis on the


pathogenetic events leading to
CLPD of NK cell.

tologic features are similar, regardless of the involved ly basophilic cytoplasm contains fine or coarse
sites. Mucosal sites often show ulceration. A diffuse azurophilic granules. Nuclei show slightly immature
infiltrate of lymphoid cells is found in association with chromatin pattern and inconspicuous or distinct nucle-
tissue necrosis and coagulation. An angiocentric and oli. These cells are sCD3–, cCD3+CD56+CD16+, CD57–,
angiodestructive growth pattern with associated fibri- CD94+ with a germline configuration of β and γ genes
noid changes in the blood vessels is frequently of TCR. In tissue sections, the neoplastic infiltrate is dif-
observed. In most patients, the neoplastic cells are char- fuse and destructive, with lymphoid cell population
acterized by medium-sized cells or a mixture of small usually appearing monomorphous.33 Necrosis, apopto-
and large lymphoid cells, with a moderate amount of sis, angioinvasion, and angiodestruction are common
cytoplasm, irregular or elongated nuclei, granular or findings.10,34,39,40 Although clonal EBV is found in tumour
vescicular chromatin, and inconspicous, small nucleoli. cells in most patients, and EBV is considered to be the
These cells express the CD45+ sCD3–cCD3+CD56+, lack- etiological agent,17 little is known about the mechanisms
ing myeloid and B lymphoid markers. Association with though which EBV infection triggers clonal proliferation
EBV can be demonstrated in nearly all patients.35 Using of NK cells. Several chromosomal abnormalities have
in situ hybridization technique, EBV-encoded RNA can been reported and the finding of the same chromosomal
be found in neoplastic cells and Southern Blot analysis abnormality involving del(6q) in aggressive NK-cell
can detect monoclonal proliferation of EBV. Analysis of leukemia and in extranodal NK/T cell lymphoma pro-
the terminal repeat region of the EBV genome indicates vides a biological link between these two diseases.41,42
that the virus is in a clonal episomal form. Other than However, array-based comparative genomic hybridiza-
providing an indirect proof of the clonal nature of the tion analysis demonstrated clear genetic differences
lymphoid proliferation, this finding suggests that the between aggressive NK-cell leukemia and extranodal
EBV might play an etiologic role in mature NK cell neo- NK/T cell lymphoma, suggesting that they are two sep-
plasms rather than simply being a bystander.33,36,37 arate entities.43

Aggressive natural killer cell leukemia Chronic lymphoproliferative disorder of natural killer cells
First characterized by Fernandez et al.38 aggressive NK (provisional)
cell leukemia is a systemic disease, again more common The chronic lymphoproliferative disorders of NK cells
in Asians than in Caucasians.10 It is characterized by the (CLPD-NK) are included among the novelties of revised
presence of neoplastic NK cells in the peripheral blood, WHO classification published in September 2008.9
bone marrow, liver and spleen and by a rapidly progres- These rare and heterogeneous disorders are character-
sive clinical course with poor prognosis. There is an ized by a chronic expansion of mature looking NK-cells
equal sex incidence in men and women. The disease (usually more than 2,000 µL) in peripheral blood for
typically affects young to middle-aged adults with a more than 6 months,44-48 without a clearly identified
median age in the third decade. At presentation, cause. Patients are usually adults with a mean age of 60
patients usually are very compromised with systemic years without sexual and racial predisposition.49,50
symptoms, liver dysfunction, and hepatosplenomegaly Recently, several studies have been published focusing
sometimes accompanied by systemic lymphadenopa- on the pathogenetic mechanisms of this disease and the
thy. In contrast to extranodal NK cell lymphoma, skin current concepts on this topic are reported in Figure 1.
lesions are uncommon. Disseminated intravascular NK cell activation in response to an unknown stimulus,
coagulation and hemophagocytic syndrome are often likely of viral origin, is postulated to play a role in the
seen during the course of disease.17,18 The clinical pro- initial steps of CLPD-NK by selecting NK clones.51-54 In
gression is devastating despite treatment, and most particular, although no prototypical HTLV infection was
patients survive for only days to weeks. Morphological- demonstrated in these patients, the evidence that sera
ly, in this disorder the leukemic cells are slightly larger from a series of patients from Europe and USA reacted
than normal LGLs.17 An ample amount of pale or slight- with the recombinant HTLV env protein p21E suggests

| 296 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

Table 2. Clinical and biological features of natural killer cell neoplasms.

NK cell Extranodal NK/T-cell Aggressive NK cell Chronic


lymphoblastic lymphoma, nasal type leukemia lymphoproliferative
leukemia/lymphoma disorder of NK cells

Age Pediatric patients Middle age adult Young to Middle aged adult Adults (6th decade)
Geographic distribution Worldwide Asia Asia Worldwide

Cell origin Immature NK cell Mature NK cell Mature NK cell Mature NK cell
Relevant phenotype sCD3-CD4-CD13- CD2+sCD3- sCD3- CD3-
CD33- CD16-CD7+ cCD3ε+CD56+CD16- cCD3ε±CD56+CD16± CD8±CD16+CD56+CD57+
CD2+CD56+CD3ε+ CD57- CD57-, CD94+ KIRs±CD94/NKG2A±
EBV association No Yes Yes No

Clinical course Aggressive Aggressive Very aggressive Indolent


Prognosis Poor Poor Poor Good

that exposure to a protein containing homology to density on patients’ NK cells; this antigen is usually
BA21 may be important in the pathogenesis of this lym- associated with the inhibitory subunit NKG2A,
phoproliferative disorder.55 In contrast with other although in some cases the association CD94/NKG2C
mature NK cell neoplasms, EBV is not usually detected has been reported.66 Patients’ NK cells characteristically
within pathological GL.50,52,54 It is believed that bone express functional β and γ chains of IL-2/IL-15 receptor,
marrow, which is frequently involved in CLPD-NK which are strictly related to the role of these cytokines
patients, represents the setting where the putative incit- in the pathogenesis of disease.57
ing antigen could reside, and dendritic cells (DC) have Most patients are asymptomatic, and the disease has
been suggested to represent the target of infection in a chronic indolent clinical course, similar to that report-
these patients.56 In fact, analysis of bone marrow biop- ed for patients with T LGL leukemia.44,47,48 In some cases,
sies of patients demonstrated a topographic distribution this disorder is associated with other conditions, includ-
of DCs and NK cells that indicates contact between the ing pure red cell aplasia, vasculitic syndromes, solid and
two cell types.56 DC are also likely to represent the hematologic tumors, splenectomy, neuropathy and
source of IL-15, which is crucial in the mechanisms sus- autoimmune disorders.47-51,53,67 Recently, in patients with
taining the maintenance of NK proliferation.57 IL-15 has chronic myeloid leukemia, the association has been
been demonstrated to mediate its activity through tar- reported between treatment with dasatinib and the
geted proteasome degradation of Bid expression, thus development of CLPD-NK; the hypothesis having been
preventing apoptosis of NK cells. A genetic susceptibili- suggested that the development of CLPD-NK might
ty for this disease has been suggested and related to the have a therapeutical effect on Ph+ leukemic cells.68
detection in these patients of type B KIR gene reper- Systemic symptoms, such as cytopenia (mostly neu-
toire, which is characterized by a high number of acti- tropenia and anemia), are rare. Lymphoadenopathy,
vating genes.58-60 In fact, the expression of NK receptors, hepatomegaly, splenomegaly and cutaneous lesions are
mostly represented by KIR, is altered in patients with uncommon.67 Occasionally, patients present with a
CLPD-NK. A restricted pattern of KIR expression has slowly progressive increase of peripheral blood NK cells
usually been reported in these patients, which is charac- and with organ involvement. In rare cases, the disease
terized either by a dominant expression of a relevant transforms to aggressive NK cell leukemia69 and cases
KIR, or by a lack of KIR expression.58,60-65 A typical fea- with EBV positive NK cells tend to evolve.17 These EBV
ture of these patients is the preferential expression of positive patients usually suffer from chronic active EBV
the KIR activating receptor isoforms,58,60 and this pattern infection and should be carefully monitored for the
correlates with a reduced expression of other activating emergence of clonal cells.69,70 Several cases with a spon-
receptors,60 such as natural cytotoxicity receptors taneous complete remission have been reported.51,53,69,72
(NCR). Together with a bias towards activating KIR Cytologically, the circulating cells show typical GL mor-
expression, a deep silencing of inhibitory KIR through phology, with moderate amount of pale cytoplasm that
increased gene metylation has been reported. contains more than or equal to three azurophilic gran-
Biochemical studies on the mechanisms sustaining the ules.48,49 Bone marrow biopsy is characterized by inter-
growth of NK cells in these patients have demonstrated stitial infiltration of cells with small nuclei and pale
a role of RAS farnesil transferase,65 with clinical implica- cytoplasm, which are difficult to recognize without the
tions.65 Pathological NK cells express CD16 and usually help of immunohistochemical techniques. Cytogenetic
low levels of CD56 and CD57. As expected, cells is normal in most cases,53,67,69 and the germ line configu-
espress TIA1, granzyme and perforins, which correlate ration of TCR is demonstrated, as expected for normal
with the cytotoxic potential of these cells displayed in in NK cells. Clonality of proliferating cells is difficult to
vitro cytotoxic assays. CD94 antigen is expressed at high detect in these patients. As an indirect marker of clonal-

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 297 |
14th Congress of the European Hematology Association

ity, the analysis of restriction fragment length polymor- drugs (in particular L-asparaginase) have been success-
phism (RLFP) has been used to demonstrate the clonali- fully used in relapsed or refractory patients83,84 and oth-
ty in some but not all patients.51,73-74 The evidence of a ers are currently under investigation,85-87 warranting
restricted pattern of clonally distributed KIR genes promising upfront use in extranasal and advanced nasal
expression by proliferating NK cells might also provide disease.
indirect demonstration of clonality.60,61 In rare cases, in Aggressive NK cell leukemia is a catastrophic disease
which EBV can be demonstrated in plasmid form with- with an almost uniform mortality. A few patients have
in NK cells, the clonality of cells might be easily exam- a clinical response with conventional chemotherapy,29
ined by Southern Blot analysis using probes recognizing although the response is typically transient. Survival is
the EBV terminal repeats.70,75 measured in days to weeks. Allogeneic bone marrow
Within post-transplant lymphoproliferative disorders transplantation reportedly results in short-term remis-
(PTLD), WHO classification also identifies as provision- sion in a few patients.80-82
al a group of conditions, which are indistiguishable Patients with CLPD-NK usually have an indolent clin-
from that of the immunocompetent host apart from the ical course and respond to immunosuppressive therapy,
feature that they occur as a consequence of immuno- with cyclosporin (3-5 mg/kg/day), low doses
suppression.76 Patients are usually recipients of a solid methotrexate (usually 15 mg/week) or cyclophos-
organ, bone marrow, or stem cell allograft.76 Among phamide (50 mg/day), associated or not to low doses of
these, the group of monomorphic PTLD provisionally steroid.44-49 Because of the potential long-term side
includes B- and T/NK-cell disorders. Clinical features effects of immunosuppressive therapy, limiting specific
and prognosis, morphology, immunophenotype and therapy only to patients with symptomatic disease is
genetics are consistent with those reported for the same recommended.
disorder in immunocompetent host.
New therapeutic strategies
Treatment and prognosis Considering the poor efficacy of current therapies for
NK cell lymphoblastic leukemia/lymphoma, provisional NK cell neoplasms, novel approaches must be consid-
A standard treatment protocol for immature NK cell ered to improve survival. Chemotherapeutic agents cur-
neoplasms has not been established because of the rently being tested in cutaneous T-cell lymphoma
paucity of patients. Current chemotherapy strategies for (CTCL) and peripheral T-cell lymphoma (PTCL), such
non-Hodgkin’s lymphoma or acute lymphoblastic as gemcitabine, liposomal doxorubicin, as well as the
leukemia (ALL) were the most commonly used. purine analogs, such as fludarabine and cladribine and
However, the overall outcomes were dismal. Two pedi- nelarabine,88 represent possible new agents to be consid-
atric patients who received non-Hodgkin lymphoma ered for NK cell treatment protocols. Furthermore, the
therapy and ALL therapy, respectively, followed by allo- use of histone deacetylase inhibitors (depsipeptide and
geneic bone marrow transplantation achieved complete vorinostat) is being tested in CTCL.89 Monoclonal anti-
remission for 3 years.16 Patients who did not undergo bodies like alemtuzumab have some activity in PTCL
bone marrow transplantation died of disease between 6 and are being investigated in combination with other
months and 35 months. Further studies are necessary to therapies.88 In addition, a better understanding of the
determine whether increased survival can be obtained signaling pathways activated in NK cell neoplasms
with aggressive chemotherapy followed by HSCT. It is could identify other biologically targeted agents as
possible that allogeneic HSCT could provide additional potential candidates for inclusion in NK cell treatment
graft versus leukemia/lymphoma benefit. protocols. Good responses have been obtained in
patients with symptomatic CLPD-NK with the use of
Mature natural killer cell neoplasms RAS farnesyltransferase inhibitor (FTI), tipifarnib
The clinical outcome of patients with nasal NK cell (Zarnestra®, Johnson & Johnson), in accordance to the
lymphoma is variable. Most observational studies have findings of constitutively active signaling of the
consistently demonstrated that radiotherapy is superior Ras/MAPK/ERK pathway.65 In addition, the proteasome
to chemotherapy alone in patients with stage I/II dis- inhibitor bortezomib has been reported to display anti-
ease.77,78 Some patients with early-stage disease are cancer activity against aggressive NK leukemia and
cured by radiation therapy. It has been demonstrated extranodal NK/T cell lymphoma in vitro and in vivo,85,86,90
that radiotherapy, either as initial treatment or as part of opening new therapeutical perspectives for these
the chemotherapy regimen, is the single most important patients.
key to a successful outcome.79-81 However, some
patients with early-stage disease have early local or sys- Conclusions and future perspectives
temic recurrences and die of the disease. For patients In order to get insight into the characteristics of NK-
with stage III/IV disease, chemotherapy is the treatment lineage disorders, the understanding of development
of choice.29 In several published series, the median sur- pathways of normal NK cell is mandatory. Impressive
vival of patients with advanced-stage disease was progress made in understanding the expression and
approximately 12 months.82 Extranasal NK cell lym- functional role of NKR has contributed to our knowl-
phomas are clinically aggressive. Because the disease edge of the characteristics of proliferating cells in
may be disseminated, chemotherapy usually is the ini- patients with CLPD. The distinctive features between
tial choice of treatment. The response is poor, and most cytotoxic T and NK cells are not fully understood and
patients die within 6 months after diagnosis. The long- DNA profiling using purified normal and pathological T
term remission rate with allogeneic bone marrow trans- and NK cells will help to address this issue and would
plantation is less than 10%.31 Other non-anthracycline likely help in understanding the differences of clinical

| 298 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

behavior between cytotoxic T and NK derived tumors. 16. Lin CW, Liu TY, Chen SU, Wang KT, Medeiros LJ, Hsu SM.
Investigation of disease pathogenesis will lead to identi- CD94 1A transcripts characterize lymphoblastic lym-
phoma/leukemia of immature natural killer cell origin with
fy molecular targets and discovery of more efficacious distinct clinical features. Blood 2005;106:3567-74.
and less toxic treatments. The elucidation of mecha- 17. Oshimi K. Progress in undestanding and managing natural
nisms by which EBV transforms NK cells might help to killer cell-malignancies. Br J Haematol 2007;139:532-44.
18. Liang X, Graam GK. Natural Killer cell neoplasms. Cancer
identify new molecular targets. Furthermore, the mech- 2008;112:1425-36.
anisms by which NK cell tumours are resistant to ther- 19. Petrella T, Comeau MR, Maynadie M, Coullault G, De
apy should be explored, and molecules and pathways Muret A, et al. “Agranular CD4+ CD56+ hematodermic neo-
responsible for apoptosis of cells should be character- plasm” (blastic NK-cell lymphoma) originated from a popu-
lation of CD56+ precursor cells related to plasmacytoid
ized. Finally, to extend the understanding of NK neo- monocytes. Am J Surg Pathol 2002;26:852-62.
plasm pathophysiology and development of novel ther- 20. Petrella T, Bagot M, Willemze R, Beylot-Barry M, Vergier B,
apeutics, physicians should be encouraged to assess et al. Blastic NK-cell lymphomas (agranular CD4+CD56+
hematodermic neoplasms). Am J Clin Pathol 2005;123:662-
National Registries and stimulate patients to participate. 75.
21. Scott AA, Head DR, Kopecky KJ, Appelbaum FR, Theil KS,
Funding et al. HLA-DR-, CD33+, CD56+, CD16- myeloid/natural
This work was supported by A.I.R.C. (Milan), by killer cell acute leukemia: a previously unrecognized form of
acute leukemia potentially misdiagnosed as French-
Fondazione CARIPARO e CARIVERONA, and by a grant American-British acute myeloid leukemia-M3. Blood 1994;
from Fondazione Berlucchi per la Ricerca sul Cancro and by 84:244-55.
Regione Veneto. 22. Liang X, Greffe B, Garrington T, Graham DK. Precursor nat-
ural killer cell leukemia. Pediar Blood Cancer 2008;50:876-8.
23. Dubois SG, Etzell JE, Matthay KK, et al. Pediatric acute blas-
tic natural killer cell leukemia. Leuk Lymphoma 2002;43:901-
References 6.
24. Hyakuna N, Toguchi S, Higa T, et al. Childhood blastic NK
1. Spits H, Lanier LL, Phillips JH. Development of human T and cell leukemia successfully treated with L-asparaginase and
natural killer cells. Blood 1995;85:2654-70. allogeneic bone marrow transplantation. Pediatr Blood
2. Trinchieri G. Biology of natural killer cells. Adv Immunol Cancer 2004;42:631-4.
1989;47:187-376. 25. Matano S, Nakamura S, Nakamura S, Annen Y, Hattori N,
3. Caligiuri M. Human natural killer cells. Blood 2008;112:461- Kobayashi K, et al. Monomorphic agranular natural killer cell
9. lymphoma/leukemia with no Epstein-Barr virus association.
4. Lanier LL, Spits H, Phillips JH. The developmental relation- Acta Haematol 1999;101:206-8.
ship between NK cells and T cells. Immunol Today 1992;13: 26. Chang C, Rodriguez A, Carreterro M, López-Botet M,
392-5. Phillips JH, Lanier LL. Molecular characterization of human
5. Sanchez MJ, Muench MO, Roncarolo MG, et al. Identifi- CD94: a type II membrane glycoprotein related to the C-
cation of a common T/natural killer cell progenitor in human type lectin superfamily. Eur J Immunol 1995;9:2433-7.
fetal thymus. J Exp Med 1994;180:569-76. 27. López-Botet M, Pérez-Villar JJ, Carretero M, Rodríguez A,
6. Huntinton ND, Vosshenrich CAJ, Di Santo JP. Development Melero I, Bellón T, et al. Structure and function of the CD94
pathways that generate natural-killer-cell diversity in mice C-type lectin receptor complex involved in recognition of
and humans. Nat Rev Cancer 2007;7:703-14. HLA class I molecules. Immmunol Rev 1997;155:165-74.
7. Moretta A, Locatelli F, Moretta L. Human NK cells: from 28. Cerwenka A, Lanier LL. Ligands for natural killer cells recep-
HLA class I-specific killer Ig-like receptors to the therapy of tors: redundanct or specificity. Immunol Rev 2001;1:41-9.
acute leukemia. Immunol Rev 2008;224:58-69. 29. Kwong YL. Natural killer-cell malignancies: diagnosis and
8. Borowitz MJ, Béné MC, Harris NL, Porwit A, Matutes E. treatment. Leukemia 2005;19:2186-94.
Acute leukemias of ambigous lineage. In Swerdlow SH, 30. Suzumiya J, Takeshita M, Kimura N, Kikuchi M, Uchida T,
Campo E, Harris NL, Jaffe EJ, Pileri SA, Stein H, Thiele J, Hisano S, et al. Expression of adult and fetal natural killer cell
Vardiman J. editors. WHO Classification of Tumours of markers in sinunasal lymphomas. Blood 1994;83 2255-60.
Haematopoietic and Lymphoid Tissues. IARD Lyon; 2008. p. 31. Cheung MM, Chan JK, Lau WH, Foo W, Chan PT, Ng CS, et
155. al. Primary non-Hodgkin’s lymphoma of the nose and
9. Villamor N, Morice WG, Chan WC, Foucar K. Chronic lym- nasopharynx: clinical features, tumor immunophenotype,
phoproliferative disorders of NK cells. In Swerdlow SH, and treatment outcome in 113 patients. J Clin Oncol
Campo E, Harris NL, Jaffe EJ, Pileri SA, Stein H, Thiele J, 1998;16:70-7.
Vardiman J. editors. WHO Classification of Tumours of Hae- 32. Kim GE, Cho JH, Yang WI, Chung EJ, Suh CO, Park KR, et
matopoietic and Lymphoid Tissues. IARD Lyon; 2008. p. al. Angiocentric lymphoma of the head and neck: patterns of
274-5. systemic failure after radiation treatment. J Clin Oncol 2000;
10. Chan JKC, Jaffe ES, Ralfkiaer E, Ko YK. Aggressive NK- 18:54-63.
leukemia. In: Swerdlow SH, Campo E, Harris NL, Jaffe EJ, 33. Siu LLP, Chan JKC, Kwong YL. Natural killer cell malignan-
Pileri SA, Stein H, Thiele J, Vardiman J. editors. WHO cies: clinicopathologic and molecular features. Histol
Classification of Tumours of Haematopoietic and Lymphoid Histopathol 2002;17:539-54.
Tissues. IARD Lyon, 2008, p. 276-7. 34. Chan JK, Sin VC, Wong KF, Ng CS, Tsang WY, Chan CH, et
11. Chan JKC, Quintanilla-Martinez NL, Ferry JA, Peh SC. al. Nonnasal lymphoma expressing the natural killer cell
Extranodal NK/T-cell lymphoma, nasal type. In Swerdlow marker CD56: a clinicopathologic study of 49 cases of an
SH, Campo E, Harris NL, Jaffe EJ, Pileri SA, Stein H, Thiele J, uncommon aggressive neoplasm. Blood 1997;89:4501-14.
Vardiman J. editors. WHO Classification of Tumours of 35. Harabuchi Y, Yamanaka N, Kataura A, Imai S, Kinoshita T,
Haematopoietic and Lymphoid Tissues. IARD Lyon, 2008, p. Mizuno F, et al. Epstein-Barr virus in nasal T-cell lymphomas
285-8. in patients with lethal midline granuloma. Lancet 1990;
12. Jaffe ES. Classification of natural killer (NK) cell and NK-like 335:128-30.
T cell malignancies. Blood 1996;87:1207-10. 36. Medeiros LJ, Peiper SC, Elwood L, Yano T, Raffeld M, Jaffe
13. Jaffe ES, Chan JK, Su IJ, Frizzera G, Mori S, Feller AC, et al. ES. Angiocentric immunoproliferative lesions: a molecular
Report of the workshop on nasal and related extranodal analysis of 8 cases. Hum Pathol 1991;22:1150-7.
angiocentric T/natural killer cell lymphomas. Am J Surg 37. Minarovits J, Hu LF, Imai S, Harabuchi Y, Kataura A, Mina-
Pathol 1996;20:103-11. rovits-Kormuta S, et al. Clonality, expression and methyla-
14. Sheibani K, Winberg CD, Burke JS, Nathwani BN, Blayney tion patterns of the EBV genomes in lethal midline granulo-
DW, Van de Velde S, et al. Lymphoblastic lymphoma mas classified as peripheral angiocentric T-cell lymphomas. J
expressing natural killer cell-associated antigens: a clinico- Gen Virol 1994;75:77-84.
pathologic study of 6 cases. Leuk Res 1987;11:371-7. 38. Fernandez LA, Pope B, Lee C, Zayed E. Aggressive natural
15. Oshimi K. Leukemia and lymphoma of natural killer lineage killer cell leukemia in an adult with establishment of an NK
cells. Int J Hematol 2003;78:18-23. line. Blood 1986;67:925-30.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 299 |
14th Congress of the European Hematology Association

39. Mori N, Yamashita Y, Tsuzuki T, Nakayama A, Nakazawa ral cytotoxicity receptors in NK-type lymphoproliferative
M, Hasegawa Y, et al. Lymphomatous features of aggressive disease of granular lymphocytes. Blood 2003;102:1797-805.
NK cell leukaemia/lymphoma with massive necrosis, 61. Zambello R, Trentin L, Ciccone E, Bulian P, Agostini C,
haemophagocytosis and EB virus infection. Histopathology Moretta A, et al. Phenotypic diversityof natural killer (NK)
2000;37: 363-71. populations in patients with NK-type lymphoproliferative
40. Quintanilla-Martinez L, Jaffe ES. Aggressive NK cell lym- disease of granular lymphocytes. Blood 1993;81:2381-5.
phomas: insights into the spectrum of NK cell derived malig- 62. Pascal V, Schleinitz N, Brunet C, Ravet S, Bonnet E, Lafarge
nancies. Histopathology 2000;37:372-4. X, et al. Comparative analysis of NK cell subset distribution
41. Oshima K, Haraoka S, Ishihara S. Analysis of chrosome 6q in normal and lymphoproliferative disease of granular lym-
deletion in EBV-associated NK cell leukemia/lymphoma. phocyte conditions. Eur J Immunol 2004;34:2930-40.
Leuk Lymphoma 2002;43:293-300. 63. Morice WG, Kurtin PJ, Leibson PJ, Tefferi A, Hanson CA.
42. Wong KF, Chan JK, Kwong YL. Identification of del Demonstration of aberrant T-cell and Natural Killer-cell anti-
(6)(q21q25) as a recurring chromosomal abnormality in puta- gen expression in all cases of granular lymphocytic
tive NK cell lymphoma/leukemia. Br J Haematol 1997;98: leukaemia. Br J Haematol 2003;120:1026-36.
922-6. 64. Hoffman T, De Libero G, Colonna M, Wodnar-Filipowicz A,
43. Nakashima Y, Tagawa H, Suzuki R, Karman S, Karube K, Passweg J, Favre G, et al. Natural killer-type receptors for
Ohshima K, et al. Genoma-wide array-based comparative HLA class I antigens are clonally expressed in lymphoprolif-
genomic hybridization of natural killer cell lymph- erative disorders of natural killer and T-cell type. Br J
oma/leukemia: different genomic alteration pattern of Haematol 2000; 110:525-36.
aggressive NK-leukemia and extranodal NK/T cell lym- 65. Epling-Burnette PK, Sokol L, Chen X, Bai F, Zhou J, Bla-
phoma, nasal type. Genes Chromosomes Cancer 2005;44: skovich MA, et al. Clinical improvement by farnesyltrans-
247-55. ferase inhibition in NK large granular lymphocyte leukemia
44. Loughran TP, Clonal diseases of large granular lymphocytes. associated with imbalanced NK receptor signaling. Blood
Blood 1993;82:1-14. 2008;112:4694-8.
45. Zambello R, Semenzato G. Large granular lymphocytosis. 66. Zambello R, Semenzato G. Natural killer receptors in
Haematologica 1998;83:936-42. patients with lymphoproliferative diseases of granular lym-
46. Tefferi A, Li CY, Witzig TE, Dhodapkar MV, Okuno SH, phocytes. Sem Hematol 2003;40:201-12.
Phyliky RL. Chronic natural killer cell lymphocytosis: a 67. Semenzato G, Pandolfi F, Chisesi T, De Rossi G, Pizzolo G,
descriptive clinical study. Blood 1994;84:2721-5. Zambello R, et al. The lymphoproliferative disease of granu-
47. Semenzato G, Zambello R, Starkebaum G, Oshimi K, lar lymphocytes. A heterogeneous disorder ranging from
Loughran TP. The lymphoproliferative disease of granular indolent to aggressive conditions. Cancer 1987;60:2971-8.
lymphocytes: updated criteria for diagnosis. Blood 1997;89: 68. Kim DH, Kamel-Reid S, Chang H, Sutherland R, Jung CW,
256-60. Kim HJ, et al. Natural Killer or natural killer/T cell lineage
48. Oshimi K. Lymphoproliferative disorders of natural killer large granular lymphocytosis associated with dasatinib ther-
cells. Int J Hematol 1996;63:279-90. apy for Philadelphia chromosome positive leukemia.
49. Morice WG, Leibson PJ, Tefferi A. Natural killer cells and the Haematologica 2009;94:135-9.
syndrome of chronic natural killer cell lymphocytosis. Leuk 69. Oshimi K, Yamada O, Kaneko T, Nishinarita S, Iizuka Y,
Lymphoma 2004;41:277-84. Urabe A, et al. Laboratory findings and clinical courses of
50. Lamy T, Loughran TP. Clinical features of large granular patients with granular lymphocyte-proliferative disorders.
lymphocyte leukemia. Semin Hematol 2003;40:185-95. Leukemia 1993;7:782-8.
51. Lima M, Almeida J, Montero AG, Teixeira Mdos A, Queirós 70. Kawa-Ha K, Ishihara S, Ninomiya T, Yumura-Yagi K, Hara J,
ML, Santos AH, et al. Clinicobiological, immunophenotypic Murayama F, et al CD3-negative lymphoproliferative disease
and molecular characteristics of CD56-/+ dim chronic natu- of granular lymphocytes containing Epstein-Barr viral DNA.
ral killer cells large granular lymphocytosis. Am J Pathol J Clin Invest 1989;84:51-5.
2004; 165:1117-27. 71. Oshimi K, Kawa K, Nakamura S, Suzuki R, Suzumiya J,
52. Loughran TP Jr, Zambello R, Ashley R, Guderian J, Pellenz Yamaguchi M, et al. NK-cell neoplasms in Japan. NK-cell
M, Semenzato G, et al. Failure to detect Epstein-Barr virus Tumor Study Group. Hematology 2005;10:237-45.
DNA in peripheral blood mononuclear cells in most patients 72. Zambello R, Loughran TP Jr, Trentin L, Rassu M, Facco M,
with large granular lymphocyte leukemia. Blood 1993;81: Bortolin M, et al. Spontaneous resolution of p58/EB6 antigen
2723-7. restricted NK-type lymphoprolifertative disease of granular
53. Rabbani GR, Phyliky RL, Tefferi A. A long-term study of lymphocytes: role of Epstein Barr virus infection. Br J
patients with chronic natural killer cells lymphocytosis. Br J Haematol 1997;99:215-21.
Haematol 1999;106:960-6. 73. Kelly A, Richards SJ, Sivakumaran M, Shiach C, Stewart AD,
54. Zambello R, Loughran TP Jr, Trentin L, Pontisso P, Battistella Roberts BE, et al. Clonality of CD3 negative large granular
L, Raimondi R, et al. Serologic and molecular evidence for a lymphocyte proliferation determined by PCR based X-inac-
possible pathogenetic role of viral infection in CD3-negative tivation studies. J Clin Pathol 1994;47:99-404.
natural killer-type lymphoproliferative diseases of granular 74. Nash R, McWeeney P, Zambello R, Semenzato G, Loughran
lymphocytes. Leukemia 1995;9:1207-11. TPJr. Clonal studies of CD3- lymphoproliferative disease of
55. Loughran TP Jr, Hadlock KG, Yang Q, Perzova R, Zambello granular lymphocytes. Blood 1993;81:2363-8.
R, Semenzato G, et al. Seroreactivity to an envelope protein 75. Hart DN, Baker BW, Inglis MJ, Nimmo JC, Starling GC,
of human T-cell leukemia/lymphoma virus in patients with Deacon E, et al. Epstein-Barr viral DNA in acute large granu-
CD3- (natural killer) lymphoproliferative disease of granular lar lymphocyte (natural killer) leukemic cells. Blood 1992;
lymphocytes. Blood 1997;90:1977-81. 72:2116-23.
56. Zambello R, Berno T, Cannas G, Baesso I, Binotto G, Bonoldi 76. Swerdlow SH, Webber SA, Chadburn A, Ferry JA. Post-
E, et al. Phenotypic and functional analyses of dendritic cells transplant lymphoproliferative disorders. In: Swerdlow SH,
in patients with lymphoproliferative disease of granular lym- Campo E, Harris NL, Jaffe EJ, Pileri SA, Stein H, Thiele J,
phocytes (LDGL). Blood 2003;102:1797-805. Vardiman J. editors. WHO Classification of Tumours of
57. Zambello R, Facco M, Trentin L, Sancetta R, Tassinari C, Haematopoietic and Lymphoid Tissues. IARD Lyon, 2008, p.
Perin A, et al. Interleukin-15 triggers the proliferation and 349.
cytotoxicity of granular lymphocytes in patients with lym- 77. Sakata K, Hareyama M, Ohuchi A, Sido M, Nagakura H,
phoproliferative disease of granular lymphocytes. Blood Morita K, et al. Treatment of lethal midline granuloma type
1997;89:201-11. nasal T-cell lymphoma. Acta Oncol 1997;36:307-11.
58. Epling-Burnette PK, Painter JS, Chaurasia P, Bai F, Wei S, 78. Robbins KT, Fuller LM, Vlasak M, Osborne B, Jing BS,
Djeu JY, et al. Dysregulated NK receptor expression in Velasquez WS, et al. Primary lymphomas of the nasal cavity
patients with lymphoproliferative disease of granular lym- and paranasal sinuses. Cancer 1985;56:814-9.
phocytes. Blood 2004;103:3431-9. 79. Cheung MM, Chan JK, Lau WH, Ngan RK, Foo WW. Early
59. Scquizzato E, Teramo A, Miorin M, Facco M, Piazza F, stage nasal NK/T-cell lymphoma: clinical outcome, prognos-
Noventa F, et al. Genotypic evaluation of killer immunoglob- tic factors, and the effect of treatment modality. Int J Radiat
ulin-like receptors in NK-type lymphoproliferative disease of Oncol Biol Phys 2002;54:182-90.
granular lymphocytes. Leukemia 2007;21:1060-9. 80. You JY, Chi KH, Yang MH, Chen CC, Ho CH, Chau WK, et
60. Zambello R, Facco M, Della Chiesa M, Trentin L, Carollo D, al. Radiation therapy versus chemotherapy as initial treat-
Castriconi R, et al. Expression and function of KIR and natu- ment for localized nasal natural killer (NK)/T-cell lymphoma:

| 300 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

a single institute survey in Taiwan. Ann Oncol 2004;15:618- 86. Shen L, Au WY, Wong KY, Shimizu N, Tsuchiyama J, Kwong
25. YL, et al. Cell death by bortezomib-induced mitotic catastro-
81. Ribrag V, Ell Hajj M, Janot F, Girinsky T, Domenge C, phe in natural killer lymphoma cell. Mol Cancer Ther 2008;
Schwaab G, et al. Early locoregional high-dose radiotherapy 7:3807-15.
is associated with long-term disease control in localized pri- 87. Au WY, Weisenburger DD, Intragumtornchai T, Nakamura
mary angiocentric lymphoma of the nose and nasopharynx. S, Kim WS, Sng I, et al. Clinical differences between nasal
Leukemia 2001;15:123-6. and extranasal NK/T-cell lymphoma: a study of 136 cases
82. Kwong YL, Chan AC, Liang R, Chiang AK, Chim CS, Chan from the International Peripheral T-cell Lymphoma Project.
TK, et al. CD56+ NK lymphomas: clinicopathological fea- Blood 2008; [Epub ahead of print].
tures and prognosis. Br J Haematol 1997;97:821-29. 88. Greer JP. Therapy of peripheral T/NK neoplasms.
83. Jaccard A, Petit B, Girault S, Suarez F, Gressin R, Zini JM, et Hematology Am Soc Hematol Educ Program 2006;331-7.
al. L-asparaginase-based treatment of 15 western patients 89. Duvic M, Talpur R, Ni X, Zhang C, Hazarika P, Kelly C, et al.
with extranodal NK/T-cell lymphoma and leukemia and Phase 2 trial of oral vorinostat (suberoylanilide hydroxamic
review of literature. Ann Oncol 2009;20:110-6. acid, SAHA) for refractory cutaneous T-cell lymphoma
84. Jaccard A, Gachard N, Coppo P, Suarez F, Gressin R, Zini JM, (CTCL). Blood 2007;109:31-9.
et al. A prospective phase II trial of an L-Asparaginase con- 90. Shen L, Au WY, Guo T, Wong KY, Wong ML, Tsuchiyama J,
taining regimen in patients with refractory or relapsing extra et al. Proteasome inhibitor bortezomib-induced apoptosis in
nodal NK/T cell lymphoma. Blood 2009;20:110-6. natural killer(NK)-cell leukemia and lymphoma: an in vitro
85. Lee J, Suh C, Kang HJ, Ryoo BY, Huh J, Ko YH, et al. Phase and in vivo preclinical evaluation. Blood 2007;110:469-70.
I study of proteosoma inhibitor bortezomib plus CHOP in 91. Zola H, Swart B, Nicholson I, Aasted B, Bensussan A,
patients with advanced, aggressive T-cell or NK/T-cell lym- Boumsell L, et al. CD molecules 2005: human cell differenti-
phoma. Ann Oncol 2008;19:2079-83. ation molecules. J Immunol 2005;106:3123-6.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 301 |
Uncommon lymphoproliferative disorders

Immunological and clinical features of T cell large


granular lymphocyte leukemia

E. Matutes A B S T R A C T

T-cell large granular lymphocytes (LGL) disorders comprise a spectrum of conditions, ranging from
Haemato-Oncology Unit, Royal polyclonal to clonal asymptomatic and/or frank leukemic LGL proliferations. We will focus on the clon-
Marsden Hospital and Institute of
Cancer Research, London, UK al LGL proliferations designated T-cell LGL leukemia. Its main features are cytopenias, splenomegaly
and autoimmune phenomena. Morphology, immunophenotype and molecular analysis are key diag-
nostic investigations. Most cases represent clonal expansions of TCRα/β+ LGL, displaying a CD8+ CD4-
or rarely a CD4+ CD8-/+dim phenotype with expression of cytotoxic T-cell antigens (CD57, CD16, TIA-1,
Hematology Education: perforin and granzyme B); a minority are clonal expansions of TCRγ/δ+ LGL. The etiology is unknown
the education program for the
but over the last years, there have been advances in understanding its pathogenesis. There is evidence
annual congress of the European
Hematology Association that T-cell LGL leukemia results from an expansion of a terminal memory activated cytotoxic lympho-
cyte. This is due to a persistent stimulation by an antigen with failure to undergo activated induced
2009;3:302-307 cell death because of an impaired apoptotic pathway. Gene profiling has shown deregulation of genes
involved in the apoptosis. In the rare form with a CD4+ CD8-/+dim phenotype, cytomegalovirus is the
agent involved in triggering this response on individuals with a genetic predisposition. The course is
usually chronic and transformation is rare. Treatment strategies comprise immunomodulatory agents,
purine analogs and antibody therapy.

Introduction and NK-cell derived diseases but the World


Proliferations of large granular lympho- Health Organization (WHO) considers the
cytes (LGL) comprise a spectrum of condi- latter as a separate group designated chron-
tions, ranging from polyclonal and self-lim- ic lymphoid disorders of NK cells.5 The
ited LGL expansions to asymptomatic majority of cases with a T-cell LGL
clonal LGL and frank leukemic sympto- leukemia represent the clonal expansion of
matic disease. They are defined by the a memory activated cytotoxic T-cell bear-
polyclonal, oligoclonal and/or clonal ing the T-cell receptor (TCR) α/β and are
expansion in the peripheral blood and bone CD8+ CD4–, rarely CD4+ CD8-/+dim; only a
marrow, or rarely in other lymphoid and minority are expansions of TCR γ/δ+ cells.
non-lymphoid tissues of a lymphocyte We will focus on the clinical features,
with a distinct morphology and for this diagnosis, biology and molecular character-
designated LGL. Polyclonal expansions of istics of the clonal T-cell LGL proliferations
LGL are usually transient and due to a viral as expansions of NK cells are the topic of
infection, such as Epstein Barr virus (EBV), another presentation.
solid tumors, connective tissue diseases,
and so on,1 or develop post-splenectomy. Etiology
This is in contrast to clonal LGL prolifera- The etiology of T-cell LGL leukemia is
tions that persist along time whether the largely unknown. However, over the last
patients are or not symptomatic. Oligo- years, there have been major advances in
clonal and clonal expansions of T-cell LGL the understanding of the pathogenesis of
have been documented in different settings this disease. A number of reports strongly
such as in patients with B-cell neoplasms,2 support the role of a chronic/persistent anti-
following allogeneic bone marrow trans- genic stimulation by an auto- or foreign
plant3 or Rituximab treatment for a B-cell infective antigen as an initial event. This
neoplasm possibly as a result from a pro- would lead to the expansion of a memory
found B-cell depletion.4 It has been suggest- effector cytotoxic LGL, which is not elimi-
ed that a disturbance of the immunity with nated from the system due to an impair-
mechanisms similar to those in T-cell LGL ment on the apoptotic pathway.6,7 No single
leukemia occurs in these patients. infective agent has been identified in the
In normal individuals, a minority of cir- most common form of T-cell LGL leukemia
culating blood lymphocytes display the (CD8+ CD4–). Early data suggesting the
morphological characteristics of LGL. The pathogenic role of EBV or human T-cell
majority of these lymphocytes have a cyto- leukemia viruses has not been confirmed. T-
toxic T-cell phenotype (CD3+ CD8+ CD4–), cell LGL leukemia is more common in
while only a minority corresponds to natu- patients with autoimmune disorders, such
ral killer (NK) cells (CD3– CD56+). Clonal as rheumatoid arthritis, Sjogrens syndrome,
expansions of LGL may comprise T-cell Hashimoto’s thyroiditis and lupus erythe-

| 302 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

matosus, as well as in patients with bone marrow fail- usually requires the integration of morphology,
ure due to myelodysplasia; thus, reinforcing that the immunophenotyping, and TCR rearrangement studies
immune system plays a role in its pathogenesis.8-10 A by Southern-blot or the polymerase chain reaction
history of rheumatoid arthritis is documented in about (PCR). Suggested criteria for diagnosis comprise: (i) per-
25% of patients, and it has been shown that LGL sistent and sustained expansions of LGL; (ii) demon-
leukemia patients have the HLADR4 haplotype, as it stration of a characteristic immunophenotype; (iii) con-
occurs in patients with rheumatoid arthritis, with a firmation of clonality by molecular studies; (iv) integra-
higher frequency than in the normal population. This tion of all these data with clinical features. Updated cri-
suggests a similar immunogenetic basis for these two teria by Semenzato et al. stresses the fact that not all
conditions.11 Unlike in CD8+ CD4- T-cell LGL leukemia, these criteria may be present in some patients with low
in the rare form with a CD4+, CD8–/CD8+dim phenotype LGL counts in whom an expansion of an LGL clone is
a persistent infection by cytomegalovirus (CMV), detected by molecular analysis.13
appears to play a key role as an initiating event in On blood films, LGLs have a nucleus with con-
patients with a genetic predisposition.12 densed chromatin, no visible nucleoli, and abundant
cytoplasm that contains coarse granules (Figure 1).
Clinical features The bone marrow cellularity is variable and involve-
The most common form of T-cell LGL leukemia ment by LGL may be inconspicuous; there may be a
preferentially affects adult patients and is rare in child- good hemopoietic reserve even with increase in the
hood. It is more common in Eastern than in Western myeloid precursors with left shift despite of the neu-
countries. Approximately one-third of patients are tropenia. The trephine biopsy shows interstitial or
asymptomatic at diagnosis, and the disease is discov- patchy lymphoid infiltrates and/or intrasinusoidal
ered on a blood count that shows lymphocytosis, per- infiltration; there may be lymphoid nodules but these
sistent mild neutropenia or both.1,13 These asympto- are shown to be reactive by immunohistochemistry.17
matic patients may have associated other conditions, Immunohistochemistry allows us to better estimate
such as myelofibrosis, eosinophilia, monoclonal gam- the degree of involvement by highlighting the infil-
mopathy, and so on. The term of T-cell clonopathy of trates. In a few patients, there is a picture of pure red
unknown significance (TLUS) has been suggested to cell aplasia or reticulin fibrosis. Immunohisto-
be more appropriate to designate these asymptomatic chemistry shows the cells to be CD3+CD8+ CD4- TIA-
patients that represent the benign end of the clonal T- 1+. The reactive nodules are composed by B-cells
cell LGL proliferations.14 Symptomatic patients fre- (CD20+) and non clonal CD4+ T-cells.17 Immuno-
quently manifest with fever due to infections or histochemistry in the marrow trephine parallel find-
mouth ulcers, often related to neutropenia, arthralgias ings of flow cytometry in the blood and marrow aspi-
and tiredness; the latter not attributable to the ane- rates. Spleen histology shows red pulp involvement
mia. A few patients may manifest with immune or no with preservation of the white pulp or a reactive ger-
immune thrombocytopenia and pulmonary artery minal centre hyperplasia with mantle zone expansion;
hypertension.8,15 Up to a half of the patients have unlike in T-cell prolymphocytic leukemia there is no
splenomegaly, around 20% skin lesions and, a minor- invasion of the white pulp. The neoplastic LGL
ity have hepatomegaly; lymphadenopathy, is rare. In express TIA-1 and perforin but show a CD5–,
some cases, computed tomography shows that the CD45RO– phenotype unlike the normal splenic red
spleen is enlarged. Patients with the CD4+ CD8-/+dim pulp cells which are CD5+, CD45RO+ .18
phenotype rarely present with cytopenias and In most cases, immunophenotyping by flow cytom-
autoimmune phenomena whilst its association with etry shows a CD2, CD3, CD8, CD16, CD57 positive
neoplasms is frequent.16 phenotype (Figure 2A). CD5 and CD7 are weakly
The leukocyte count may be normal or slightly expressed or negative; CD56 is rarely positive and it
raised but in most patients, there is an increase in cir- has been suggested to be associated with a more
culating LGL, even without having an absolute lym- aggressive course.19 A small proportion of cases have a
phocytosis; a few patients are lymphopenic but many CD4+ CD8-/+dim (Figure 2B) or have a double
develop lymphocytosis after splenectomy. Neutro- CD4/CD8 negative phenotype. It has been suggested
penia is the most frequent cytopenia and it is unrelat- that cases with a CD4+ CD8–/+dim phenotype are a dis-
ed to the degree of marrow infiltration or hyper- tinct subgroup with a preferential Vb13.1 usage (44%
splenism. Anemia and thrombocytopenia are less fre- of cases) and associated with neoplasias.16 These cells
quent and are present in around a third of patients. have a cytotoxic (CD56+, CD57+, granzyme B+) pheno-
Liver function tests may be impaired and the autoim- type corresponding to that of a memory/activated
mune screen reveals abnormalities, such as the pres- cell. Most cases represent expansions of T cells bear-
ence of rheumatoid factor, antinuclear antibodies, cir- ing the TCR αβ and a very minority is derived from
culating immunocomplexes, and polyclonal hyper- TCRγδ+ cells, which have a Vγ9/δ2 or a Vnonγ9/δ1
gammaglobulinemia; a few patients may have genophenotypic profile equally restricted compared to
hypogammaglobulinemia that relates to the suppres- normal TCRγδ+ T-cells. It is uncertain as to whether
sor function of the LGL upon the B lymphocytes. cases with TCRγδ+ T-cell LGL leukemia have different
clinical manifestations compared to the most common
Diagnosis form of TCRαβ+ T-cell LGL leukemia; it seems that
Although the diagnosis of T-cell LGL can be suspect- there is a higher proportion of cases in this cohort that
ed in patients with a persistent increase in LGL (greater have a double CD4/CD8 negative phenotype.20 The
than 6 months and with >2000/µL circulating LGL), it availability of a repertoire of Monoclonal antibodies

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 303 |
14th Congress of the European Hematology Association

Figure 1. Circulating lymphocyte dysplaying an eccentric


nucleus and abundant cytoplasm with coarse granules,
feaures typical of LGL.

(McAb) against NK cell antigens has proved to be


valuable for the diagnosis of LGL leukemia. These
McAb recognize receptors that belong to the
immunoglobulin (Ig) superfamily and comprise killer-
cell Ig-like receptors (KIRs) clustered under CD158a,
CD158b, CD158e and CD158i and C-type lectin
receptors (CD94, CD161). In healthy individuals, T-
cells do not express KIRs and a small proportion is
CD94+ and/or CD161+. By contrast, CD94, like CD57,
is expressed in clonal LGL, and some cases, may
express one or more KIR.21,22 Over-expression of KIRs
in LGL is aberrant and potentially diagnostic. CD52
has been documented to be expressed in cells from all
T-cell LGL leukemias investigated and there is a
potential for the use of Campath-1H in this disease.23
Flow cytometry analysis with McAb against the Figure 2. (A) Flow cytometry dot plots from a case with a
various Vβ regions of the TCR allows establishing CD8+ T-cell leukemia showing expression on the leukemic
clonality of the LGLs by showing the preferential use cells of CD2, CD5, CD45RA, CD8, CD11c, CD16, CD57,
of one or two TCR-Vβ segments. There is a correla- CD94, granzyme B, perforin and HLA-Dr whilst are negative
for CD7, CD45RO and CD27. A residual normal T-cell pop-
tion between the PCR and flow cytometry findings, ulation with a different phenotype can be also identified.
with flow cytometry able to predict clonality with a (B) Flow cytometry dot plots from a case with a CD4+ T-cell
sensitivity of 93% and a specificity of 80%.24 Unlike leukemia showing expression of CD2, CD4, CD5, CD45RA,
CD45RO, CD56, granzyme B, perforin and partial expres-
in CD4+ CD8–/+dim T-cell LGL leukemias, there is no sion of CD57 whilst cells are negative with CD16, CD161,
preferential use of a Vβ segment as the pattern of dis- CD158a and CD27.
tribution is similar to that found in normal blood.
The diagnosis of T-cell LGL leukemia should be
confirmed by documenting clonal TCR rearrangement
of the β or γ chain genes by Southern blot or PCR abnormalities include those involving rearrangements
(Figure 3). These techniques should be applied to all of the TCR loci genes at 7p14-p15 and 14q11, inver-
suspected cases and are useful in those patients in sion of 12p, 5q deletion and trisomy 3, 8 and 14.5,25
which the absolute LGL count is not significantly It is now becoming evident that T-cell LGL
increased.13 leukemia with a CD8+ CD4– phenotype results from
The differential diagnosis arises with self-limited an expansion and accumulation of a CD8+ terminal
polyclonal expansions of LGL and other post-thymic effector memory cytotoxic cell. This is due to a per-
T-cell disorders, namely prolymphocytic leukemia sistent stimulation by an auto or foreign antigen with
and γ/δ T-cell lymphoma, myelofibrosis and a failure to undergo activate induced cell death
myelodysplasia. (AICD) consequent to a profound impaired apoptotic
pathway. Elimination of potentially harmful antigen-
Pathogenesis and molecular features stimulated effector memory T-cells after a successful
Cytogenetic studies in T-cell LGL leukemia are immune response physiologically occurs by triggering
scant; clonal abnormalities are infrequently detected, Fas-mediated apoptosis through induction of a death-
without a recurrent genetic marker identified.1,5 Clonal inducing signaling complex (DISC). This allows main-

| 304 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

genes in the sphingolipid metabolism and signaling


which are up-regulated in LGL, include acid
ceraminidase and S1P5 receptor. Up-regulation of
ceramidase is found in many neoplasms and it has
been postulated to also be involved in the apoptosis
failure in LGL.
TCRα/β+ CD4+ CD8-/+dim LGL leukemias is an infre-
quent form. There is compelling evidence that chron-
ic stimulation of T-cells by CMV leads to a persistent
clonal expansion of CD4+/CD8-/+dim LGL, with a pre-
dominance of TCR VB13.1 usage in individuals with a
HLADRB1*0701 haplotype. Recent reports have
shown that the expanded T-cell clones have a similar
TCR with a high degree of homology in the comple-
mentary determinant region (CDR3) and similar com-
binatorial gene rearrangements.29 In vitro studies have
demonstrated that the expanded CD4+ LGL clones
have a characteristic and unique response to a CMV
Figure 3. PCR for the TCRβ and TCRγ chain genes showing peptide.12 The gene expression signature of these cells
multiple bands in a control (polyclonal pattern) and a sin-
gle TCRβ (monoallelic) and two TCRγ bands (bi-allelic) on a is different from that of normal memory CD4+ LGL
patient with T-cell LGL leukemia. with outstanding abnormalities in CD4+ leukemic
LGL encompassing a variety of genes that function in
cell cycle, resistance to apoptosis and genetic instabil-
ity. All these data strongly supports that a persistent
taining the T-cell homeostasis. Globally, leukemic CMV stimulation is the responsible for triggering and
LGL are characterized by a profound deregulation of maintaining the activated/memory LGL clone in
the apoptosis and do not undergo to AICD. Although patients with a genetic predisposition. Beside the
clonal LGL constitutively express high levels of both pathogenesis of the expansion of LGL, the underlying
Fas (CD95) and Fas ligand (Fas-L), and do not harbor mechanisms of the cytopenias may be multiple, such
mutations of the Fas receptor, they are resistant to as lack of erythroid precursors (PRCA), autoimmune
Fas-induced apoptosis due to a defective Fas apoptot- mediated or inhibition of the burst-forming and ery-
ic pathway.26 Fas resistance can occur in cells with throid colony-forming units by the release of
over-expression of the DISC inhibitory protein c-FLIP. cytokines by the LGL.30
c-FLIP is constituvely expressed in leukemic LGL pre-
venting DISC formation and Fas mediated apoptosis.27 Prognosis and survival
Besides this, it is likely that several genes are involved T-cell LGL leukemia usually runs a chronic course in
in such apoptotic failure as revealed by gene microar- most patients with mortalities of 10 to 28% at 4 years
ray expression as discussed in the following para- and a median survival greater than 10 years.1,31 Most
graph. Gene expression profiling has revealed that deaths are due to sepsis and rarely occur due to dis-
leukemic LGLs have a different signature from that of ease progression; transformation to large-cell lym-
normal naïve and activated memory T-cells.28 Major phoma is exceedingly rare.32
differences are found on the expression of genes
belonging to the categories of apoptosis, regulation of Therapy
TCR signaling and immune response. This supports Up to a third of patients may will never require
the notion that an uncoupling of activation and apop- treatment as they are asymptomatic and cytopenias
totic pathways is responsible for a failure of AICD. are mild; a watch and wait strategy is appropriate.1
One of the main findings is the deregulation in the Indications for treatment include severe neutropenia,
expression of genes involved in the apoptotic path- symptomatic anemia or thrombocytopenia and pro-
way. Genes which are up-regulated in leukemic LGL gressive disease, (i.e., organomegaly and/or rapidly
have anti-apoptotic functions whilst those which are raising counts). The aim of treatment is to correct the
down-regulated are pro-apoptotic. Among the apop- cytopenias, and this can be achieved without signifi-
totic related genes, the gene that shows the highest cant reduction of the clone with immunomodulatory
up-regulation compared to normal activated CD8+ drugs, such as methotrexate (10 mg/m2/weekly),
lymphocytes is the tumour necrosis factor α induced cyclosporine A (5-10 mg/kg/day) or low dose cyclo-
protein 3 (TNFAIP3).28 This is a NF-Kb inducible gene phosphamide (50 to 100 mg/day).31,33 Corticosteroids
that protects T-lymphocytes from undergoing TNF may be useful as part of the initial treatment to accel-
induced apoptosis and, in physiological conditions is, erate response; growth factors are of value as they
unlike in leukemic LGL, down-regulated in T-cells fol- may act synergistically with the immunosuppressive
lowing activation. Genes belonging to the BCL-2 fam- therapy. There have not been randomized trials to
ily, such as the BCL-2 related X gene (BAX) are down- compare the efficacy of these agents. While all are
regulated, whilst the myeloid cell factor (MCL-1) is effective in improving cytopenias in up to two thirds
up-regulated. Recent data indicates that the sphin- of patients, the malignant clone persists. Adverse
golipid-mediated signaling pathway plays a role on events are not severe and are more common with
the apoptotic deregulation in leukemic LGL. Key cyclosporine A, particularly in elderly patients.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 305 |
14th Congress of the European Hematology Association

Second malignancies are rare but close vigilance is rec- Immunogenetic similarities between patients with Felty’s
syndrome and those with clonal expansion of large granular
ommended, particularly in cases with prolonged lymphocytes in rheumatoid arthritis. Arthritis Rheum 1997;
immunosuppression. Splenectomy may be considered 40:624-6.
as an adjuvant in patients with gross splenomegaly 12. Rodriguez-Caballero A, Garcia-Montero AC, Barcena P,
and refractory cytopenias.34 Almeida J., Ruiz-Cabello F, Tabernero MD, et al. Expanded
cells in monoclonal TCR-αβ+/CD4+/Nkα+/CD8-/+dim T-
Purine analogs have been only used in few patients LGL lymphocytes recognize hCMV antigens. Blood 2008;
with response rates of 40 to 67% for pentostatin,35 flu- 112:4609-16.
darabine alone36 or in combination.37 The use of these 13. Semenzato G, Zambello R, Starkebaum G, Oshimi K,
Loughran TP. The lymphoproliferative disease of granular
agents should be contemplated in young patients as lymphocytes: updated criteria for diagnosis. Blood 1997; 89:
they allow achieving good remissions, including 256-60.
reduction of bone marrow infiltration. 14. Sabnani I,Tsang P. Are clonal T-cell large granular lympho-
cytes to blame for unexplained haematological abnormali-
Newer agents such as the McAb anti-CD52 ties? Br J Haematol 2006;136:30-7.
(Campath-1H), anti-CD122 and anti-CD2 are being 15. Chen X, Bai F, Sokol L, Zhou J, Ren A, Painter JS. et al. A crit-
incorporated in the therapeutic scenario.38 There have ical role for DAP10 and DAP12 in CD8+ T-cell mediated tis-
sue damage in large granular lymphocyte leukemia. Blood
been anecdotal reports of successful treatment with 2009 (In press).
anti-CD52 antibody in refractory disease. In light of 16. Lima M, Almeida J, Dos Anjos Teixeira M, Alguero Md
the deregulation of the MAPK/Ras pathway in the Mdel C, Santos AH, Balanzategui A, et al. TCR-
leukemic NK and T-cell LGL, the use of the farnesyl- alphabeta+/CD4+ large granular lymphocytosis: a new clon-
al T-cell lymphoproliferative disorder. Am J Pathol 2003;
transferase inhibitor tipifarnib may offer therapeutic 163:763-71.
utility.39 There are few ongoing clinical trials for T-cell 17. Osuji N, Beiske K, Randen U, Matutes E, Tjonnfjord G,
LGL leukemia, such as a phase II ECOG study of Catovsky D, et al. Characteristic appearances of the bone
marrow in T-cell large granular lymphocyte leukemia.
methotrexate 10 mg/m2 weekly, with or without Histopathology 2007;50:547-54.
prednisolone 30 mg/day with tapering doses, crossing 18. Osuji N, Matutes E, Catovsky D, Lampert I, Wotherspoon
over to low-dose cyclophosphamide 100 mg daily for A. Histopathology of the spleen in T-cell large granular lym-
phocyte leukemia and T-cell prolymphocytic leukemia: a
non-responders, a phase II study of tipifarnib39 and a comparative review. Am J Surg Pathol 2005; 29:935-41.
phase I study of the anti-CD2 antibody siplizumab. 19. Alekshun TJ, Tao J, Sokol L. Aggressive T-cell large granular
In current practice, the approach to treatment in T- lymphocyte leukemia: a case report and review of the liter-
ature. Am J Hematol 2007;82:481-5.
cell LGL leukemia is to start with an immunosuppres- 20. Bourgault-Rouxel AS, Loughran TP, Zambello R, Epling-
sive agent, such as methotrexate or cyclosporine A, Burnette PK, Semenzato G, Donadieu J, et al. Clinical spec-
with or without prednisolone or growth factors. trum of γδ+ T cell LGL leukemia: analysis of 20 cases. Leuk
Purine analogs, McAb and/or experimental drugs may Res 2008;32:45-8.
21. Morice WG, Kurtin PJ, Leibson PJ, Tefferi A, Hanson CA.
be considered in selected patients with the aim of Demonstration of aberrant T-cell and natural killer-cell anti-
achieving a complete and durable remission. gen expression in all cases of granular lymphocytic
leukaemia. Br J Haematol 2003;120:1026-36.
22. Fischer L, Hummel M, Burmeister T, Schwartz S, Thiel E.
Skewed expression of natural-killer (NK)-associated anti-
References gens on lymphoproliferations of large granular lymphocytes
(LGL). Hematol Oncol 2006;24:78-85.
1. Loughran TP. Clonal diseases of large granular lymphocytes. 23. Osuji N, Del Giudice I, Matutes E, Morilla A, Owusu-
Blood 1993;82:1-14. Ankomah K, Morilla R, et al. CD52 expression in T-cell large
2. Martinez A, Pitaluga S, Villamor N, Colomer D., Rozman M, granular lymphocyte leukemia: implications for treatment
Raffeld M, et al. Clonal T-cell populations and increased risk with alemtuzumab. Leuk Lymphoma 2005;46:723-7.
for cytotoxic T-cell lymphoma in B-CLL patients: clinico- 24. Lima M, Almeida J, Santos AH, dos Anjos Texeira M,
pathologic observations and molecular analysis. Am J Surg Alguero MC, Queros ML, et al. Immunophenotypic analysis
Pathol 2004;28:849-58. of the TCR-Vβ repertoire in 98 persistent expansions of
3. Au WY, Lam CC, Lie AK, Pang A, Kwong YL. T-cell large CD3(+)/TCR-αβ(+) large granular lymphocytes:utility in
granular lymphocyte leukemia of donor origin after allo- assessing clonality and insights into the pathogenesis of the
geneic bone marrow transplantation. Am J Clin Pathol 2003; disease. Am J Pathol 2001;159:1861-8.
120:626-30. 25. Wong KF, Chan JC, Liu HS, Man C, Kwong YL.
4. Stamatopoulos K, Papadaki T, Pontikoglou C, Athanasiadou Chromosomal abnormalities in T-cell large granular lym-
I, Stavroyianni N, Bux J, et al. Lymphocyte subpopulation phocyte leukemia: report of two cases and review of the lit-
imbalances, bone marrow hematopoiesis and histopatholo- erature. Br J Haematol 2002;116:598-600.
gy in rituximab treated lymphoma patients with late onset 26. Lamy T, Liu JH, Landowski TH, Dalton WS, Loughran TP.
neutropenia. Leukemia 2008;22:1446-9. Dysregulation of CD95/CD95 ligand-apoptotic pathway in
5. Chan WC, Foucar KM, Morice WG, Catovsky D. T-cell large CD3(+) large granular lymphocyte leukemia. Blood 1998;
granular lymphocytic leukaemia. In: Swerdlow SH, Campo 92: 4771-7.
E, Harris NL, Jaffe ES, Pileri SA, Stein H, et al. editors. WHO 27. Yang J, Epling-Burnette PK, Painter JS, Zou J, Bai F, Wei S,
Classification of Tumours of Haematopoietic and Lymphoid Loughran TP. Antigen activation and impaired Fas-induced
Tissues. Lyon: IARC Press, 2008. p. 272-3. death-inducing signaling complex formation in T-large gran-
6. Epling-Burnette PK, Loughran TP Jr. Survival signals in ular lymphocyte leukemia. Blood 2008;111:1610-16.
leukemic large granular lymphocytes. Semin Hematol 2003; 28. Shah MV, Zhang R, Irby R, Kothapalli R, Liu X, Arrington T,
40:213-20. et al. Molecular profiling of LGL leukemia reveals role of
7. Wlodarski MW, O’Keefe C, Howe EC, Risitano AM, sphingolipid signaling in survival of cytotoxic lymphocytes.
Rodriguez A, Warshawsky I, et al. Pathologic clonal cyto- Blood 2008;112:770-81.
toxic T-cell responses: nonrandom nature of the T-cell- 29. Garrido P, Ruiz-Cabello F, Barcena P, Sandberg Y, Canton J,
receptor restriction in large granular lymphocyte leukemia. Lima M, et al. Monoclonal TCR-Vβ 13.1+/CD4+/NKa+/
Blood 2005;106:2769-80. CD8-/+dim T-LGL lymphocytosis: evidence for an antigen-
8. Sokol L, Loughran TP. Large granular lymphocyte leukemia. driven chronic T-cell stimulation origin. Blood 2007;109:
Oncologist 2006;11:263-73. 4890-8.
9. O’Malley DP. T-cell large granular lymphocyte leukaemia 30. Handgretinger R, Geiselhart A, Moris A, Grau R, Teuffel O,
and related proliferations. Am J Clin Path 2007;127:850-9. Bethge W, et al. Pure red-cell aplasia associated with clonal
10. Starkebaum G. Chronic neutropenia associated with expansion of granular lymphocytes expressing killer-cell
autoimmune disease. Semin Hematol 2002;39:121-7. inhibitory receptors. N Engl J Med 1999;340:278-84.
11. Starkebaum G, Loughran TP, Gaur LK, Davis P, Nepon BS. 31. Osuji N, Matutes E, Tjonnfjord G, Grech H, Del Giudice I,

| 306 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

Wotherspoon A, et al. T-cell large granular lymphocyte 2003;27:865-7.


leukemia: A report on the treatment of 29 patients and a 36. Sternberg A, Eagleton H, Pillai N, Leyden K, Turner S,
review of the literature. Cancer 2006;107:570-8. Pearson D, et al. Neutropenia and anaemia associated with
32. Matutes E, Wotherspoon AC, Parker NE, Osuji N, Isaacson T-cell large granular lymphocyte leukemia responds to flu-
PG, Catovsky D. Transformation of T-cell large granular darabine with minimal toxicity. Br J Haematol 2003;120:
lymphocyte leukemia into a high-grade large T-cell lym- 699-701.
phoma. Br J Haematol 2001;115:801-6. 37. Tse E, Chan JC, Pang A, Au WY, Leung AY, Lam CC, et al.
33. Loughran TP, Kidd PG, Starkebaum G. Treatment of large Fludarabine, mitoxantrone and dexamethasone as first-line
granular lymphocyte leukemia with oral low-dose metho- treatment for T-cell large granular lymphocyte leukemia.
trexate. Blood 1994;84:2164-70. Leukemia 2007;21:2225-56.
34. Subbiah V, Viny AD, Rosenblatt S, Pohlman B, Lichtin A, 38. Alekshun TJ, Sokol L. Diseases of large granular lympho-
Maciejewski JP. Outcomes of splenectomy in T-cell large cytes. Cancer Control 2007;14:141-50.
granular lymphocyte leukemia with splenomegaly and 39. Epling-Burnette PK, Sokol L, Chen X, Bai F, Zhou J,
cytopenias. Exp Hematol 2008;36:1078-83. Blaskovich MA, et al. Clinical improvement by farnesyl-
35. Tsirigotis P, Venetis E, Kapsimali V, Rontogianni D, transferase inhibition in NK large granular lymphocyte
Varvitsioti E, Pappa V, et al. 2-deoxycoformycin in the treat- leukemia associated with imbalanced NK receptor signaling.
ment of T-large granular lymphocyte leukemia. Leuk Res Blood 2008;112:4694-8.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 307 |
Uncommon lymphoproliferative disorders

The treatment of hairy cell leukemia

D. Catovsky A B S T R A C T
M. Else
There has been significant progress in our understanding of the nature of hairy cell leukemia (HCL)
since the first description of the disease fifty years ago. In contrast to other B-cell lymphoprolifera-
Section of Haemato-Oncology tive disorders, progress in HCL treatment has been remarkable, with several agents, mainly the purine
The Institute of Cancer Research, analogs pentostatin and cladribine, able to achieve a high proportion of complete responses, which
Sutton, UK are of long duration. We will illustrate this from our experience with these two agents from a series
of 233 patients treated over the last 22 years, with a median follow-up of 16 years. For the propor-
tion of relapsing patients (42% at ten years and 47% at fifteen years), as well as those initial non-
Hematology Education: responders, there are now new treatment modalities in the form of monoclonal antibodies which,
the education program for the either alone or in combination with either purine analog, could improve outcome. We will show that
annual congress of the European rituximab (anti-CD20), combined with either pentostatin or cladribine, could achieve better respons-
Hematology Association es in relapsing patients than when used alone. This is not surprising, as the density of expression of
the CD20 antigen is the highest of all the B-cell malignancies. Anti-CD22, an antibody linked to an
2009;3:308-313 immunotoxin, also offers new hope for the minority of relapsing/refractory patients.

Introduction
Exactly fifty years ago, a seminal paper by lating villous (or hairy-looking) lymphocytes
Bouroncle et al.1 put hairy cell leukemia can present a diagnostic problem. There are
(HCL) on the clinico-pathological map. The three conditions: (i) splenic marginal zone
report described clearly all the features we lymphoma (formerly described as splenic
now recognize in this distinct disease entity. lymphoma with circulating villous lympho-
The term leukemic reticuloendotheliosis was cytes); (ii) HCL-variant, which is a rare con-
coined and the benefits of splenctomy, with dition and now included in the WHO classi-
long-term remissions in some patients, were fication as a provisional entity. It bears no
noted. It is of historical interest that biological connection with typical HCL; (iii)
Rosenthal described a similar entity a few a recently described condition with some
years earlier (1952) under the term reticular similarities to splenic marginal zone lym-
cell leukemia but, because of the vagaries of phoma and HCL-variant, described as
the peer review system, the paper was splenic diffuse red-pulp B-cell lymphoma.
rejected, and only published in 1969. Schrek This is also a provisional entity in the new
and Donnelly2 introduced the term hairy cell WHO classification.7 The latter two condi-
later, when they compared hairy cells to tions have the involvement of the splenic red
some flagellated cells seen in lymph nodes, pulp and the presence of circulating lympho-
using phase contrast microscopy. Other dis- cytes with villous projections in common
tinct features of HCL were noted: the bone with HCL.
marrow fibrosis in the presence of lympho- The key to HCL diagnosis is the bone mar-
cytes, the rather unique tartrate resistant row histology. Monoclonal antibodies
acid phosphatase (TRAP),3 the ribosome (MoAb) can facilitate the diagnosis. This can
lamella complex seen first by electron be flow cytometry and/or immunohisto-
microscopy and later by light microscopy4 chemistry in bone marrow sections.
and, finally, the distinct pattern of the bone Characteristic MoAb used by flow cytome-
marrow histology.5 try are CD25, CD103, CD123, in addition to
In the 1970s, hairy cells were shown to be markers seen in other B-cells, CD11c, CD19,
of B-cell origin; a fact confirmed in the 1980s CD20, FMC7, and monotypic membrane
by showing clonal rearrangement of the Igl.6,8 By immunohistochemistry, specific
immunoglobulin (Igl) genes. markers are Annexin A1, TRAP (demonstrat-
ed by means of a MoAb), DBA44, CD20 and
Diagnosis CD11c (a formalin resistant epitope).9 The
HCL is now a well-recognized entity in importance of immunohistochemistry is not
the World Health Organization (WHO) clas- just for diagnosis but also for its power to
sification.6 Although the diagnosis is relative- identify minimal residual disease after thera-
ly straightforward in the presence of cytope- py. Without this methodology, it is very dif-
nias in middle-aged male patients with a pal- ficult to identify small clusters of residual
pable spleen, it is important to be aware that hairy cells in the bone marrow by conven-
other splenic B-cell lymphomas with circu- tional hemotoxylin and eosin staining. The

| 308 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

antibody to Annexin A1, a very specific marker of HCL


(as distinct from other B-cell disorders), is less useful for
detecting residual disease because it also reacts with
myeloid cells.6

Clinical features and indications for treatment


In contrast to chronic lymphocytic leukemia (CLL),
most patients with HCL are diagnosed because of clini-
cal problems at presentation: cytopenias, infections
caused by neutropenia and/or monocytopenia,
splenomegaly. A chance diagnosis is rare. Lack of clini-
cal progression (in the absence of treatment) is also very
rare. In our experience, around 3% of patients may
remain stable without the need for treatment for long
periods of time.
Although treatment is seldom an emergency, infec-
tion is one of the main clinical indications, as well as
progressive splenomegaly, anemia or thrombocytope- Figure 1. Relapse-free survival by hemoglobin and platelet
nia. A period of watch and wait, as in CLL, is possible counts. Patients with hemoglobin <10 g/dL and/or
platelets <100×109/L had shorter relapse-free survival
but very often the manifestations of the disease show than patients with higher counts, p<0.0001. (Royal
progressive features requiring an urgent initiation of Marsden Hospital/Institute of Cancer Research series of
therapy. 233 patients with hairy cell leukemia, updated 2009).52
A few studies are looking at the prognostic features at
presentation in HCL. A large French series of 238
patients10 identified a hemoglobin level less than 10 g/dL
and a white blood count less than 2.0×109/L as factors patients, it should probably be a rule in relapsing cases,
influencing the achievement of a complete response and particularly in those who presented with large
(CR). Both factors were also associated with reduced spleens. Treating such patients without checking for
survival.10 In our study of 233 patients, we also docu- evidence of abdominal nodes may result in inadequate
mented that hemoglobin less than 10 g/dL and platelets clinical responses because of giving insufficient treat-
less than 100×109/L at the start of treatment were, when ment. It was of interest that out of our four patients
combined, associated with a shorter relapse-free sur- with abdominal lymphadenopathy who were deemed
vival (9 years) versus patients without these features to have become refractory to pentostatin, we docu-
(20+ years).52 Figure 1 illustrates this. Therefore, as in mented a CR in two and a partial response (PR) in
CLL, a poor bone marrow function, reflected in low another, using cladribine.13 This provided some evi-
hemoglobin and platelet levels, seems to indicate a dence for a lack of cross-resistance between these two
worse prognosis. Such features may need to be re- commonly used purine analogs.
examined prospectively, since patients with poor bone
marrow function may require special attention for their Treatments for hairy cell leukemia
treatment plan. There are several treatment modalities, which can be
In the era before effective therapy was available, it used in HCL although, for the last 20 years, the main
was common to see patients with mycobacterial infec- agents have been the nucleoside analogs pentostatin (2’-
tions, both classic tuberculosis and atypical mycobacte- deoxycoformycin) and cladribine (2-chlorodeoxyadeno-
ria, such as mycobacteria Kansasai. In patients present- sine) because of their ability to induce high response
ing with sweating and/or unexplained fever, it is always rates and long remissions. A summary of some of the
important to investigate such infections, sometimes large published series with both reagents is shown in
requiring a liver biopsy to establish the cause. Table 1.
The issue of lymphadenopathy in HCL is of particu- Splenectomy, first mentioned by Bouroncle et al.,1 can
lar interest. Peripheral enlarged lymph nodes are not a still achieve spectacular results, with rapid improve-
feature of the disease, as they are in CLL. In an earlier ment of the cytopenias in close to 90% of cases. A
study, we noticed a group of patients with advanced minority of such patients may not require any further
HCL who had large abdominal masses of lymph therapy for very long periods (e.g., 15-20 years) and
nodes.11 A study by Hakimian et al.12 reported a similar enjoy excellent quality of life. We have treated one such
finding in 25% of previously treated HCL cases, but patient whose clinical remission lasted 28 years before
none in 19 newly-diagnosed patients screened by CT he relapsed and achieved a complete response with two
scan. Maloisel et al.10 identified 8% of patients with sig- courses of cladribine. Splenectomy is not a treatment of
nificant lymphadenopathy at diagnosis, and 7% previ- choice in HCL, but it may benefit patients presenting
ously treated. Large abdominal masses seem, in our with massive splenomegaly (e.g., >10 cm below the
experience and that of others10,12 to be associated with costal margin) and little or moderate bone marrow
bulky disease, also manifested by a large spleen (which involvement. Rarely, a splenectomy may be useful to
has often been removed). In our series13 and that of the establish a diagnosis and to distinguish HCL from some
French group,10 patients with abdominal nodes have a of the other forms of splenic B-cell lymphoma men-
worse outcome. Although it is not common practice to tioned above. The first major advance in the treatment
perform abdominal CT scanning in newly diagnosed of HCL was interferon-α (IFN-α), first published by

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 309 |
14th Congress of the European Hematology Association

Table 1. Responses to pentostatin and cladribine in HCL.

Ref. No. pts. % ORR % CR

Pentostatin
Cassileth et al. 1991 (14) 50 84 64
Golomb et al. 1994 (15) 85 84 42
Ribeiro et al. 1999 (16) 50 96 44
Rafel et al. 2000 (17) 78 88 72
Grever et al. 1995 (18) 154 79 76
Johnston et al. 2000 (19) 28 100 89
Maloisel et al. 2003 (10) 238 96 79
Else et al. 2005 (20) 185 96 81

Cladribine
Estey et al. 1992 (21) 46 89 78
Tallman et al. 1996 (22) 50 100 86
Hoffman et al. 1997 (23) 49 100 76
Cheson et al. 1998 (24) 861 87 50 Figure 2. Relapse-free survival by response to treatment.
Patients who achieved a complete response to treatment
Goodman et al. 2003 (25) 207 100 95 had longer relapse-free survival than patients who
Jehn et al. 2004 (26) 44 100 98 attained a partial response, p<0.0001. (Royal Marsden
Zinzani et al. 2004 (27) 37 100 81 Hospital/Institute of Cancer Research series of 233
Chadha et al. 2005 (28) 86 99 79 patients with hairy cell leukemia, updated 2009).52
Else et al. 2005 (20) 34 100 82
Robak et al. 2007 (29) 132 93 74
CR: complete response; ORR: overall response rate (CR + partial response).
tions. The number of injections was variable (range 2-
27; median 10) as it was guided by the target of achiev-
ing CR. CR is the aim of treatment in HCL, as it is
Quesada in 1984, with 3 out of 7 patients achieving associated with long disease-free survival, regardless
CR.30 In fact, IFN-α does not induce a high CR rate, but of the therapy used (as shown in Figure 2, which com-
the overall response rate in some series is 91%.31 The bines our data with both pentostatin and cladribine).
responses to IFN-α are rapid and are not associated The third major landmark in the treatment of HCL
with the myeloid and lymphoid toxicities seen with was cladribine, introduced by Piro et al.36 in 1990.
the purine analogs. There has been only one random- Cladribine has become very well established in the
ized trial comparing IFN-α with pentostatin18 and that USA, where it is the treatment of choice.
showed a very superior disease-free interval after ces- One important aspect of treatment with cladribine is
sation of therapy with pentostatin. Clearly, IFN-α that many physicians thought that one single course of
needs continuous treatment in order to maintain nor- treatment was sufficient and failed to look at the bone
mal counts, as HCL is still present in the bone marrow. marrow to assess response. We repeated a bone mar-
Some authors have tested the possibility of long-term row examination after 4 to 6 months and, if there was
maintenance, which prolongs progression-free sur- only a PR, a further course was given. This was neces-
vival.31 In fact, two such studies have shown that it is sary in 18% of patients and the majority of them
possible to apply such a program in 50-60% of switched from PR to CR after the second course.
patients.31,32 Because the CR rate with IFN-α is low and Cladribine is an easier agent to produce than pento-
there is discomfort associated with the side effects of statin and can be given in different modalities or routes
continuous maintenance, this agent is no longer used of administration. We have used the original dose of
as first line in HCL. One possible additional advantage 0.1 mg/kg/day by continuous infusion over 7 days.36
of using IFN-α for a short term (circa 3 months), prior Others have used short two-hour infusions at 0.12-
to giving a nucleoside analog, is that it may result in 0.15 mg/kg for 5 or 7 days, with apparently similar
fewer early infections;33 an observation that coincides response rates.37 More recently, Robak et al.29 compared
with our early experience.34 Whether there is still a role the daily short infusions for five days with weekly
for IFN-α in the treatment of HCL, for example, in infusions over six weeks, with no differences in
patients non-responsive to cladribine or pentostatin, is response rate or toxicities. A preparation for bolus sub-
an open question. The advent of MoAb with therapeu- cutaneous injections is now available and this has also
tic activity in this disease (see below) suggests that this been used with equal success with respect to the
is unlikely to be the case. response rate. What is missing in many of these stud-
The first nucleoside analog with activity in HCL was ies, which compare the various modalities for admin-
pentostatin. Spiers et al.35 reported a CR in 16 out of 27 istering cladribine, is data on whether the quality of
patients. This high CR rate was confirmed in several the response translates into a long disease-free interval.
other studies (Table 1), including our own, which now To this end, it is critical always to examine the bone
has a median follow-up of 16 years.52 One feature of marrow after the treatment is completed.
the way we have administered pentostatin is to use 4 We compared pentostatin and cladribine in our
mg/m2 as an intravenous push every two weeks until series of 233 patients and found no differences in
maximum response, followed by two additional injec- response rates, disease-free survival and overall sur-

| 310 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

Table 2. Responses to rituximab in HCL, as a single agent malignancies.47 Also, adding this antibody to either
and combined with pentostatin or cladribine. nucleoside analog does not increase toxicity and allows
the achievement of better responses in the
Ref. No. pts. % ORR % CR relapse/refractory setting than with either cladribine or
pentostatin alone. Although it is not clear whether it is
Single agent better to use rituximab concurrently or sequentially
Zinzani et al. 2000 (39) 24 25 13
(after the initial response to the nucleoside analog), 9
Lauria et al. 2001 (40) 10 50 10
Hagberg & Lundholm 2001 (41) 11 64 54 out of our 12 patients were given the MoAb concur-
Nieva et al. 2003 (42) 24 25 13 rently. By analogy with data in CLL,48 concurrently may
Thomas et al. 2003* (43) 15 80 53 achieve a better synergistic effect. The issue also arises
Zenhausern et al. 2008 (44) 26 80 32 as to the number of rituximab infusions and whether
this antibody should move to being used as part of the
Combination treatment first line therapy, as used by Ravandi et al.45 Because
Ravandi et al. 2006* (45) 13 100 100 best results may be achieved with six to eight ritux-
Else et al. 2007 (38) 8 100 87 imab infusions, plus the lack of added toxicity, it is like-
Cervetti et al. 2008 (46) 27 100 89 ly that this number, rather than the conventional four
infusions, should be used for optimal effect. Now, we
*8 doses rituximab (previous standard: 4)
would consider this combined therapy – nucleoside
analog plus rituximab – for the 42-47% who experience
a relapse within the first 10 -15 years after first line
vival.20 Although this was not a randomized trial; nev- therapy. The choice of nucleoside analog will depend
ertheless, the characteristics of the patients were not on which of the two was previously used. The exis-
very different between the treatment groups and our tence of possible lack of cross-resistance mentioned
conclusion is that there is no major difference above suggests that the alternative agent, if available,
between these two purine analogs. The choice will should be used with rituximab in such indications. The
depend on cost, ease of administration and, ultimate- issue of cost may determine whether such modalities
ly, quality of life and duration of remission. Survival at become the treatment of choice for all patients.
ten years was 96% and 100% with pentostatin and
cladribine, respectively. For patients achieving CR and Immunotoxins in the treatment of hairy cell leukemia
still in CR at five years, the risk of relapse by 15 years
Kreitman et al.49 reported promising results with a
was only 25%.
recombinant immunotoxin containing anti-CD22 vari-
From our data, at 16 years’ median follow-up from
able domain fused to truncated Pseudomonas exotox-
diagnosis, only one fifth of patients have died, and this
in (BL22) in a series of patients resistant to cladribine.
is equivalent to the age/sex-matched death rate for the
A recent update of this series50 reported a CR rate of
general population.52 The incidence of HCL-related
deaths in our series was less than 4%. Damasio et al.31 61% and an overall response rate of 81%, with a medi-
reached a similar conclusion with the maintenance an duration of remission of 36 months. Although
schedule of IFN-α. Thus, in HCL the long-term sur- refractory HCL is not common, this modality could
vival is not a major issue. The main measures to assess also be considered in relapsing patients, not necessari-
the benefits of any therapy are the duration and quali- ly refractory, and a clinical trial could perhaps be envis-
ty of remissions, as assessed by disease-free survival. aged to compare BL22 with the combination of a
purine analog plus rituximab. Problems with BL22 are
Treatment of relapsed/refractory disease hemolytic uremic syndrome in some patients, liver
We have also investigated the effect of therapy with dysfunction and hypoalbuminemia. Again, issues of
either nucleoside analog after first and subsequent safety and cost will be paramount. Patients in whom
relapses and in non-responding patients.52 The CR rate these new modalities may be used include: (i) non-
after first line was 80%, after second line was 69%, responders to first line pentostatin or cladribine (<5%);
and after third line was 50%, clearly suggesting that (ii) partial responders to either drug (15-20%); (iii)
achieving a new response in relapsing patients complete responders but who tend to relapse in the
becomes more difficult. Nevertheless, when we first 5-10 years (circa 40%).
looked at disease-free survival in those achieving CR,
the duration of these remissions was equally long, Second malignancies in hairy cell leukemia
whether after first, second or third-line therapy. This There are uneven data in the literature as to whether
introduces the concept that new modalities, perhaps the rate of second malignancies in HCL may be
adding MoAb to chemotherapy, may be used to increased, either by the disease itself or by the use of
improve the CR rate in such relapsing/refractory purine analogs and/or IFN-α. In our series, 31 patients
patients. Indeed, in a preliminary analysis, now updat- developed second malignancies (excluding non-
ed to 12 patients relapsing after 2, 3, 4 or 6 lines of melanoma skin cancers).52 This was equivalent to the
therapy, we recorded a CR rate of 92% when ritux- incidence of cancer in the general population matched
imab (Mabthera) was used in combination with either by age and sex. Similar conclusions were reached by
cladribine or pentostatin.38 two other large series of 238 patients10 and 241
Table 2 shows the results of various small series using patients;51 therefore, it seems unlikely that either the
this anti-CD20 MoAb. There is evidence that the den- disease or its treatment increases the likelihood of
sity of CD20 in hairy cells is the highest of all the B-cell other malignancies.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 311 |
14th Congress of the European Hematology Association

Conclusions hairy cell leukemia: an intergroup study. J Clin Oncol 1995;13:


974-82.
The treatment of HCL has been remarkably success- 19. Johnston JB, Eisenhauer E, Wainman N, Corbett WE, Zaentz
ful and new possibilities have appeared on the horizon, SD, et al. Long-term outcome following treatment of hairy cell
in the form of MoAb for therapeutic use, which will leukemia with pentostatin (Nipent): a National Cancer
improve treatment further. For a relatively rare disease, Institute of Canada study. Semin Oncol 2000;27 2 Suppl 5:32-
6.
it is a major success story, achieved without random- 20. Else M, Ruchlemer R, Osuji N, Del Giudice I, Matutes E,
ized clinical trials. The outlook for patients with respect Woodman A, et al. Long remissions in hairy cell leukemia
to disease-free interval and overall survival has with purine analogs: a report of 219 patients with a median
follow-up of 12.5 years. Cancer 2005;104:2442-8.
improved dramatically since the disease was first 21. Estey EH, Kurzrock R, Kantarjian HM, O'Brien SM, McCredie
described fifty years ago.1 KB, Beran M, et al. Treatment of hairy cell leukemia with 2-
chlorodeoxyadenosine (2-CdA). Blood 1992;79:882-7.
22. Tallman MS, Hakimian D, Rademaker AW, Zanzig C, Wollins
E, Rose E, et al. Relapse of hairy cell leukemia after 2-chloro-
References deoxyadenosine: long-term follow-up of the Northwestern
University experience. Blood 1996;88:1954-9.
1. Bouroncle BA, Wiseman, BK, Doan CA. Leukemic reticuloen- 23. Hoffman MA, Janson D, Rose E, Rai KR. Treatment of hairy-
dotheliosis. Blood 1958;13:609-30. cell leukemia with cladribine: response, toxicity, and long-
2. Schrek R, Donnelly WJ. “Hairy” cells in blood in lymphoretic- term follow-up. J Clin Oncol 1997;15:1138-42.
ular neoplastic disease and “flagellated” cells of normal lymph 24. Cheson BD, Sorensen JM, Vena DA, Montello MJ, Barrett JA,
nodes. Blood 1966;27:199-211. Damasio E, et al. Treatment of hairy cell leukemia with 2-
3. Yam LT, Li CY, Lam KW. Tartrate-resistant acid phosphatase chlorodeoxyadenosine via the Group C protocol mechanism
isoenzyme in the reticulum cells of leukemic reticuloendothe- of the National Cancer Institute: a report of 979 patients. J Clin
liosis. N Engl J Med 1971;284:357-60. Oncol 1998;16:3007-15.
4. Katayama I, Li CY, Yam LT. Ultrastructural characteristics of 25. Goodman GR, Burian C, Koziol JA, Saven A. Extended follow-
the “hairy cells” of leukemic reticuloendotheliosis. Am J Pathol up of patients with hairy cell leukemia after treatment with
1972;67:361-70. cladribine. J Clin Oncol 2003;21:891-6.
5. Burke JS, Byrne GE Jr, Rappaport H. Hairy cell leukemia 26. Jehn U, Bartl R, Dietzfelbinger H, Haferlach T, Heinemann V.
(leukemic reticuloendotheliosis). I. A clinical pathologic study An update: 12-year follow-up of patients with hairy cell
of 21 patients. Cancer 1974;33:1399-410. leukemia following treatment with 2-chlorodeoxyadenosine.
6. Foucar K, Falini B, Catovsky D, Stein H. Hairy Cell Leukaemia. Leukemia 2004;18:1476-81.
In: Swerdlow SH, Campo E, Harris NL, et al. editors. WHO 27. Zinzani PL, Tani M, Marchi E, Stefoni V, Alinari L, Musuraca
Classification of Tumours of Haematopoietic and Lymphoid G, et al. Long-term follow-up of front-line treatment of hairy
Tissues. Lyon: IARC Press; 2008. p. 188-90. cell leukemia with 2-chlorodeoxyadenosine. Haematologica
7. Piris M, Foucar K, Mollejo M, Campo E, Falini B. Splenic B-cell 2004;89:309-13.
lymphoma/leukaemia unclassifiable. In: Swerdlow SH, 28. Chadha P, Rademaker AW, Mendiratta P, Kim B, Evanchuk
Campo E, Harris NL, et al. editors. WHO Classification of DM, Hakimian D, et al. Treatment of hairy cell leukemia with
Tumours of Haematopoietic and Lymphoid Tissues. Lyon: 2-chlorodeoxyadenosine (2-CdA): long-term follow-up of the
IARC Press; 2008, p. 191-3. Northwestern University experience. Blood 2005;106:241-6.
8. Del Giudice I, Matutes E, Morilla R, Morilla A, Owusu- 29. Robak T, Jamroziak K, Gora-Tybor J, Blonski JZ, Kasznicki M,
Ankomah K, Rafiq F, et al. The diagnostic value of CD123 in Dwilewicz-Trojaczek J, et al. Cladribine in a weekly versus
B-cell disorders with hairy or villous lymphocytes. Haemato- daily schedule for untreated active hairy cell leukemia: final
logica 2004;89:303-8. report from the Polish Adult Leukemia Group (PALG) of a
9. Johrens K, Happerfield LC, Brown JP, Erber WN, Stein H, prospective, randomized, multicenter trial. Blood 2007;109:
Anagnostopoulos I. A novel CD11c monoclonal antibody 3672-5.
effective in formalin-fixed tissue for the diagnosis of hairy cell 30. Quesada JR, Reuben J, Manning JT, Hersh EM, Gutterman JU.
leukemia. Pathobiology 2008;75:252-6. α interferon for induction of remission in hairy-cell leukemia.
10. Maloisel F , Benboubker L, Gardembas M, Coiffier B, Divine N Engl J Med 1984;310:15-8.
M, Sebban C, et al. Long-term outcome with pentostatin treat- 31. Damasio EE, Clavio M, Masoudi B, Isaza A, Spriano M, Rossi
ment in hairy cell leukemia patients. A French retrospective E, et al. α-interferon as induction and maintenance therapy in
study of 238 patients. Leukemia 2003;17:45-51. hairy cell leukemia: a long-term follow-up analysis. Eur J
11. Mercieca J, Puga M, Matutes E, Moskovic E, Salim S, Ca- Haematol 2000;64:47-52.
tovsky D. Incidence and significance of abdominal lym- 32. Benz R, Siciliano D, Stussi G, Fehr J. Long-term follow-up of
phadenopathy in hairy cell leukaemia. Leuk Lymphoma 1994; interferon-α induction and low-dose maintenance therapy in
(14 Suppl 1):79-83. hairy cell leukemia. Eur J Haematol 2009;82:194-200.
12. Hakimian D, Tallman MS, Kiley C, Peterson L. Detection of 33. Habermann TM, Andersen JW, Cassileth PA, Bennett JM,
minimal residual disease by immunostaining of bone marrow Oken MM. Sequential administration of recombinant interfer-
biopsies after 2-chlorodeoxyadenosine for hairy cell leukemia. on α and deoxycoformycin in the treatment of hairy cell
Blood 1993;82:1798-802. leukaemia. Br J Haematol 1992;80:466-71.
13. Mercieca J, Matutes E, Emmett E, Coles H, Catovsky D. 2- 34. Catovsky D, Matutes E, Talavera JG. Long term results with
Chlorodeoxy-adenosine in the treatment of hairy cell 2’deoxycoformycin in hairy cell leukemia. Leuk Lymphoma
leukaemia: differences in response in patients with and with- 1994; 14:109-13.
out abdominal lymphadenopathy. Br J Haematol 1996; 93:409- 35. Spiers AS, Parekh SJ, Bishop MB. Hairy-cell leukemia: induc-
11. tion of complete remission with pentostatin (2'-deoxyco-
14. Cassileth PA, Cheuvart B, Spiers AS, et al. Pentostatin induces formycin). J Clin Oncol 1984;2:1336-42.
durable remissions in hairy cell leukemia. J Clin Oncol 1991; 36. Piro LD, Carrera CJ, Carson DA, Beutler E. Lasting remissions
9:243-46. in hairy-cell leukemia induced by a single infusion of 2-
15. Golomb HM, Dodge R, Mick R, Budman D, Hutchison R, chlorodeoxyadenosine. N Engl J Med 1990;322:1117-21.
Horning SJ, et al. Pentostatin treatment for hairy cell leukemia 37. Robak T. Current treatment options in hairy cell leukemia and
patients who failed initial therapy with recombinant alpha- hairy cell leukaemia variant. Cancer Treat Rev 2006;32:365-76.
interferon: a report of CALGB study 8515. Leukemia 1994; 38. Else M, Osuji N, Forconi F, et al. The role of rituximab in com-
8:2037-40. bination with pentostatin or cladribine for the treatment of
16. Ribeiro P, Bouaffia F, Peaud PY, Blanc M, Salles B, Salles G, et relapsed/refractory hairy cell leukaemia. Cancer 2007;110:
al. Long term outcome of patients with hairy cell leukemia 2240-47.
treated with pentostatin. Cancer 1999;85:65-71. 39. Zinzani PL, Ascani S, Piccaluga PP, Bendandi M, Pileri S, Tura
17. Rafel M, Cervantes F, Beltrán JM, Zuazu F, Hernández Nieto L, S. Efficacy of rituximab in hairy cell leukemia treatment. J Clin
Rayón C, et al. Deoxycoformycin in the treatment of patients Oncol 2000;18:3875-7.
with hairy cell leukemia: results of a Spanish collaborative 40. Lauria F, Lenoci M, Annino L, Raspadori D, Marotta G,
study of 80 patients. Cancer 2000;88:352-57. Bocchia M, et al. Efficacy of anti-CD20 monoclonal antibodies
18. Grever M, Kopecky K, Foucar MK, Head D, Bennett JM, (Mabthera) in patients with progressed hairy cell leukemia.
Hutchison RE, et al. Randomized comparison of pentostatin Haematologica 2001;86:1046-50.
versus interferon α-2a in previously untreated patients with 41. Hagberg H, Lundholm L. Rituximab, a chimaeric anti-CD20

| 312 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1)
Berlin, Germany, June 4-7, 2009

monoclonal antibody, in the treatment of hairy cell leukaemia. 48. Byrd JC, Peterson BL, Morrison VA, Park K, Jacobson R, Hoke
Br J Haematol 2001;115:609-11. E, et al. Randomized phase 2 study of fludarabine with con-
42. Nieva J, Bethel K, Saven A. Phase 2 study of rituximab in the current versus sequential treatment with rituximab in sympto-
treatment of cladribine-failed patients with hairy cell matic, untreated patients with B-cell chronic lymphocytic
leukemia. Blood 2003;102:810-3. leukemia: results from Cancer and Leukemia Group B 9712
43. Thomas DA, O'Brien S, Bueso-Ramos C, Faderl S, Keating MJ, (CALGB 9712). Blood 2003;101:6-14.
Giles FJ, et al. Rituximab in relapsed or refractory hairy cell 49. Kreitman RJ, Wilson WH, Bergeron K, Raggio M, Stetler-
leukemia. Blood 2003; 102:3906-11. Stevenson M, FitzGerald DJ, et al. Efficacy of the anti-CD22
44. Zenhausern R, Von Rohr A, Rufibach K, Solenthaler M, recombinant immunotoxin BL22 in chemotherapy-resistant
Meyer-Monard S, Gratwohl A, et al. Low dose 2-chlorode- hairy-cell leukemia. N Engl J Med 2001;345:241-7.
oxyadenosine given as a single subcutaneous injection in 50. Kreitman RJ, Pastan I. Immunotoxins in the treatment of
patients with hairy cell leukemia - a multicentre trial SAKK
32/95. Leuk Lymphoma 2009;16:1-4. refractory hairy cell leukemia. Hematol Oncol Clin North Am
45. Ravandi F, Jorgensen JL, O'Brien SM, Verstovsek S, Koller CA, 2006;20:1137-51.
Faderl S, et al. Eradication of minimal residual disease in hairy 51. Flinn IW, Kopecky KJ, Foucar MK, Head D, Bennett JM,
cell leukemia. Blood 2006; 107:4658-62. Hutchison R, et al. Long-term follow-up of remission duration,
46. Cervetti G, Galimberti S, Andreazzoli F, Fazzi R, Cecconi N, mortality, and second malignancies in hairy cell leukemia
Caracciolo F, et al. Rituximab as treatment for minimal resid- patients treated with pentostatin. Blood 2000;96:2981-6.
ual disease in hairy cell leukaemia: extended follow-up. Br J 52. Else M, Dearden CE, Matutes E, Garcia-Talavera J, Rohatiner
Haematol 2008;143:296-8. AZ, Johnson SA, et al. Long-term follow-up of 233 patients
47. Ginaldi L, De Martinis M, Matutes E, Farahat N, Morilla R, with hairy cell leukaemia, treated initially with pentostatin or
Catovsky D. Levels of expression of CD19 and CD20 in cladribine, at a median of 16 years from diagnosis. Br J
chronic B cell leukaemias. J Clin Pathol 1998;51:364-9. Haematol 2009 [Epub ahead of print].

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 313 |
Berlin, Germany, June 4-7, 2009

Index of authors

Armstrong S.A. 8 Gratwohl A. 256 Mullighan C.G. 1


Gudgin E. 17 Munshi N.C. 161
Baglin T. 266
Baron F. 242 Hartmann E. 106 Niemeyer C.M. 208
Barrett J. 250 Hillmen P. 68 Nimer S.D. 177
Bladé J. 166 Hochhaus A. 78
Boyd K. 155 Hoffman R. 192 Pamphilon D.H. 282
Brunet S. 30 Huntly B. 17,83 Putti M.C. 221
Büller H.R. 271
Izraeli S. 13 Randi M.L. 221
Catovsky D. 308 Raza A. 172
Cazzola M. 181 Kantarjian H. 83 Reitsma P.H. 261
Chapuy B. 112 Kaspers G.J.L. 214 Rosenquist R. 61
Cilloni D. 24 Kiladjian J.-J. 200 Rosenwald A. 106
Cortes J. 83 Klein U. 55 Rosiñol L. 166
Kleinjan A. 271
De Meyer S.F. 38 Kralovics R. 188 Saglio G. 24
Deininger M.W. 72 Sandmaier B. 242
Dzierzak E. 123 Landman-Parker J. 140 Semenzato G. 294
Lapid K. 133 Shipp M. 112
Efficace F. 89 Lapidot T. 133 Sierra J. 30
Eichenauer D.A. 151 Luban N.L.C. 288 Simon R. 96
Else M. 308 Sprangers M.A. 89
Engert A. 151 Malcovati L. 181 Steinberg M.H. 233
Estey E. 101 Manz M.G. 128
Mascarenhas J. 192 Takizawa H. 128
Federici A.B. 50,89 Matutes E. 302
McMullin M.F. 238 Vanhoorelbeke K. 38
Galili N. 172 Messa E. 24
Gallamini A. 144 Mohandas N. 227 Zambello R. 294
Gisselbrecht C. 118 Morgan G. 155
Goodeve A. 44,78 Moulds J.M. 276

Hematology Education: the education program for the annual congress of the European Hematology Association | 2009; 3(1) | 314 |

You might also like