You are on page 1of 11

Vaccines for haematological tumours

Review

Part II: Vaccines for haematological malignant


disorders

Simone Mocellin, Gianpietro Semenzato, Susanna Mandruzzato, and Carlo Riccardo Rossi

The search for new therapeutic approaches to haemato-


logical malignant disease involves exploitation of the
antitumour potential of adaptive immunity—the most
specific killing system against cancer currently known. Bone-marrow Donor-lymphocyte
Here, we summarise immunological strategies behind transplantation infusion
active specific immunotherapy, and describe the clinical
and immunological results from trials published to date.
Available data in humans support the hypothesis that
various vaccination regimens can polarise adaptive Chemotherapy Monoclonal
and radiotherapy Leukaemias antibodies
immunity towards effective control of cancer-cell growth.
and
However, the exploratory nature of clinical studies done lymphomas
thus far does not allow any cancer vaccine to be used as
standard treatment for haematological malignant
disorders. Because the cause of disease recurrence is the Kinase Active specific
presence of minimal residual disease after conventional inhibitors immunotherapy?

treatments, the adjuvant setting might be the most Others (eg,


appropriate therapeutic strategy for active specific interferon alfa
and tretinoin)
immunotherapy, when the immunosuppressive effects of
bulky disease are virtually absent and when the effector-
target ratio is favourable. In the near future, completion of
randomised phase III trials as well as clinical
Figure 1. The arsenal of therapeutic weapons against leukaemias and
implementation of the most recent insights into tumour lymphomas is constantly growing.
immunology that aim to overcome immune tolerance
towards malignant cells should allow investigators to destroy chemotherapy-resistant cell lines from chronic
define the actual role of vaccines in the management of myeloid leukaemia and multiple myeloma,7,8 have prompted
haematological tumours. development of immunotherapeutic strategies against
haematological cancers.9 Among these approaches, active
Lancet Oncol 2004; 5: 727–37 specific immunisation or vaccination is emerging as a
valuable tool to polarise the adaptive immune system
The past two decades have seen substantial advances in against malignant cells.10 As is the case for infectious
treatment of haematological malignant disorders (figure 1). diseases, anticancer vaccination is based on the assumption
Present use of intensified radiotherapy and chemotherapy that the immune system can recognise tumour-associated
protocols can lead to first remission in most patients, and antigens and destroy malignant cells that express them
bone-marrow transplantation or stem-cell transplantation (figure 2). Identification of leukaemia-associated or
are effective therapeutic options for those who have disease lymphoma-associated antigens that are specifically targeted
recurrence.1,2 However, a substantial number of patients will by T-cell or B-cell responses has spurred development of
ultimately die of their disease.3 Therefore, new non-cross- different strategies of active specific immunisation for
resistant treatment strategies that might improve outlook the treatment of haematological malignant disorders
for patients are awaited. (table 1).11
Emerging preclinical and clinical data suggest that
immune-cell mediators can recognise and kill malignant S Mocellin is a postdoctorate fellow, S Mandruzzato is an
cells in patients with haematological malignant disorders. immunologist, and CRR is Professor of Surgery; all at the
The lower rates of relapse in the setting of allogeneic Department of Oncological and Surgical Sciences, University of
transplantation compared with those in autologous bone- Padova, Italy. GS is Professor of Haematology in the Department of
Clinical and Experimental Medicine, University of Padova, Italy.
marrow transplantation;4 the striking clinical benefit of
Correspondence: Dr Simone Mocellin, Dipartimento di Scienze
donor-lymphocyte infusions;5 and the clinical effectiveness Oncologiche e Chirurgiche, Sezione di Clinica Chirurgica, Via
of antibody-based therapies for treatment of non-Hodgkin Giustiniani, 2, 35128 Padova, Italy. Tel: +39 049 8211851.
lymphomas6 as well as the finding that human T cells can Fax: +39 049 651891. Email: mocellins@hotmail.com

Oncology Vol 5 December 2004 http://oncology.thelancet.com 727


Review Vaccines for haematological tumours

Cytokines (eg, interleukin 2 and inter-


Naive helper
Naive cytotoxic T lymphocyte
leukin 12) can also cause paracrine
T-cell receptor
T lymphocyte costimulation. In the absence of such
secondary signals, T cells are rendered
anergic to the presented antigen.12 As
HLA class I and II,
and peptides
leukaemia or lymphoma cells generally
express high amounts of HLA class I
CD8 molecules, the decreased T-cell stimu-
CD4 latory activity of these cells is mainly
CD80 t(9;22) because of deficient costimulation. Active
CD86 t(8;14)
t(14;18) CD20
specific immunisation aims to compen-
sate for the decreased immunogenicity of
FAS Peptides
Tumour-
tumour cells. By comparison with solid
associated Haematological tumours, haematological malignant dis-
antigen tumour cell orders have favourable features that make
Proteasome them ideal targets for vaccine-based thera-
Ig
peutic interventions. Ease of tumour
Perforin accessibility, achievement of a state of
Granzyme CD28
FAS ligand CTLA4 minimal residual disease by current treat-
Th1-type cytokines ments, and the antigen-presenting-cell
(eg, interleukin 2
and interferon γ) properties of most lymphoid13 and
myeloid14–17 tumours facilitate an effective
Effector helper immunotherapeutic strategy. These fea-
T lymphocyte tures, coupled with the non-cross-reactive
Th2-type cytokines
nature of active specific immunotherapy
(eg, interleukin 4 and chemotherapy, have led to the
Effector cytotoxic and interleukin 10) integration of different immunothera-
T lymphocyte peutic strategies with current treatment
regimens in the clinical setting.

Antigen-specific vaccines
Antibodies Idiotype vaccines
B cell (IgM, IgG)
The specific antigenic determinants of Ig
variable regions, called the idiotype, are
produced by a single B-cell clone and are
Figure 2. Expression of unique tumour-specific antigens (eg, Ig idiotype, translocation-derived therefore unique tumour-associated
fusion proteins, or CD20) recognised by effector cells of adaptive immunity (ie, cytotoxic T antigens for B-cell malignant diseases. In
lymphocytes and B cells) make most haematological tumour cells ideal targets for active specific
animal studies of antitumour vaccination,
immunotherapy. In addition to conventional cross presentation of tumour-associated antigens
by professional antigen-presenting cells such as dendritic cells, several haematological these determinants serve as tumour-
malignant cells have antigen-presenting capabilities, which at the same time make them targets, associated antigens,18 and idiotypes have
and mediators, of the immune response after vaccination. been tested in non-randomised clinical
trials in patients with B-cell non-Hodgkin
Although results of active specific immunisation in lymphoma and multiple myeloma (table 2); randomised
humans for treatment of haematological cancers is phase III trials are also under way (table 3). Similar to other
substantially smaller than those recorded with solid tumours, vaccine formulations (eg, vaccines based on dendritic cells,
currently available results are encouraging and support heat-shock proteins, or on genetically engineered whole-cell
further investigation of the anticancer potential of the vaccines), they need to be custom-made for each patient.
immune system. We summarise current vaccination
strategies, and discuss clinical and immunological findings Multiple myeloma
from clinical trials. Tumour-specific idiotype protein secreted by multiple
myeloma cells can be followed easily in the blood of patients,
Vaccination strategies concentrations of which correlate with disease status. Unlike
Two signals are thought to be needed for effective T-cell B-cell non-Hodgkin lymphoma, the yield of idiotype protein
stimulation.10 The first signal is generated when the correct from patient’s serum is usually enough to prepare the
T-cell receptor recognises a peptide derived from tumour- vaccine.
associated antigen that is complexed with a HLA class I Although immune responses can be generated against
molecule. The second signal is delivered by costimulatory idiotype protein in multiple myeloma, the primary target of
molecules such as CD80 or CD86, which are expressed by such responses could be the circulating idiotype protein and
professional antigen-presenting cells such as dendritic cells. not the tumour cell. Moreover, animal studies have showed

728 Oncology Vol 5 December 2004 http://oncology.thelancet.com


Vaccines for haematological tumours
Review

that free idiotype protein might inhibit idiotype-specific Table 1. Tumour-associated antigens expressed in
immunity.45 However, these concerns are more relevant to haematological malignant disease
antibodies, which can bind circulating idiotype and thus
might not reach the target tumour cells in sufficient Category Tumour-associated Tumour
concentration to be effective. Because T cells do not recognise antigen
intact protein, cell-mediated adaptive immune responses Unique BCR-ABL1* Chronic myeloid leukaemia
DEK-NUP214 Acute myeloid leukaemia
might be unaffected by circulating idiotype and could be AML1-CBFA2T1 Acute myeloid leukaemia
generated to recognise idiotype-specific peptides present on RAR␣ Promyelocytic leukaemia
the surface of plasma cells or their precursors. Ig idiotype* B-cell non-Hodgkin lymphoma
However, CD4-positive T-cell anergy has been described Complementarity B-cell non-Hodgkin lymphoma
as a consequence of high-dose administration of idiotype in determining region 3*
T-cell receptor idiotype T-cell lymphomas
animals.46 Although the effect on the cytotoxic T-cell response
P53 Miscellaneous
towards malignant cells in humans remains to be defined, RAS Miscellaneous
peripheral T-cell tolerance to high concentrations of idiotype Shared Melanoma antigen Multiple myeloma
could be a tumour-escape mechanism in patients with B-cell family
malignant disorders. Thus, idiotype vaccination should be B Melanoma antigen Multiple myeloma
family
reserved for eradication of minimal residual disease after
Cancer/testis antigen Multiple myeloma
high-dose chemotherapy. G antigen family Multiple myeloma
Bergenbrant and colleagues19 gave five patients injections Preferentially Acute myeloid leukaemia
of paraprotein taken from the patients, which had been expressed antigen
emulsified in aluminum phosphate. Despite some evidence of melanoma
for a transient increase in cellular and humoral immune Overexpressed Myeloblastin precursor Acute myeloid leukaemia
protein and chronic myeloid
responses and despite an association between B-cell response leukaemia
and decreased CD19-positive peripheral B cells, this regimen WT1* Acute myeloid leukaemia,
was insufficient to generate a sustained and clinically active chronic myeloid leukaemia,
anti-myeloma immunity. and acute lymphoblastic
leukaemia
In a further trial published by the same researchers,20 five MUC1 Multiple myeloma
patients were treated with idiotype combined with Viral LMP1, LMP2 (Epstein- Burkitt’s lymphoma
granulocyte-monocyte colony-stimulating factor (GM-CSF) Barr virus)
in the same site on subsequent days. All patients developed a Antigens from human Hodgkin’s lymphoma and
cellular immune response characterised in vitro mainly by an T-cell lymphotropic acute T-cell leukaemia
idiotype-specific increase in T cells that secreted interferon ␣ virus type I

and interleukin 2. This response was present in CD4-positive *Antigens used in the clinical setting.

and CD8-positive T-cell subsets, and could be inhibited by


the blockade of MHC class I molecules. Furthermore,
production of idiotype-specific IgM was induced in vivo. method of real-time allele-specific oligonucleotide PCR. Four
However, delayed-type hypersensitivity to idiotype protein of six patients had a significant substantial decrease in
was not found, and only one patient had a decrease in serum peripheral tumour burden. Furthermore, one patient had
paraprotein concentration. complete molecular remission in the blood and three patients
Massaia and colleagues23 described a vaccine trial in which had an idiotype-specific T-cell response. In the two patients
idiotype was conjugated with the carrier protein keyhole who did not respond to vaccination, one mounted a T-cell
limpet haemocyanin (KLH) and administered with response.
interleukin 2 or GM-CSF.23 12 patients who had previously
undergone autologous stem-cell transplantation and who B-cell non-Hodgkin lymphoma
were in complete clinical remission were subsequently The first trial of idiotype vaccination in patients with
vaccinated at various times after transplantation. Nine of 11 lymphoma was done by Kwak and colleagues.47 Patients
patients tested had evidence of cellular response to KLH, with follicular non-Hodgkin lymphoma were vaccinated with
whereas only two of 11 patients had a substantial T-cell idiotype–KLH conjugate while in clinical remission after
response to idiotype protein. Interestingly, no humoral anti- chemotherapy. To obtain sufficient amounts of idiotype
idiotype response was detected; however, development of protein for vaccination, a somatic-cell hybrid was generated
positive delayed-type hypersensitivity of the skin was by fusion of each patient’s tumour cells with a hybridoma cell
reported in eight of ten patients. line, and the secreted idiotype was purified from bulk
Rasmussen and co-workers21 reported on a study in which supernatant. Complete tumour regression was recorded in
six patients with IgG-producing multiple myeloma were two patients who had measurable lymphoma lesions.
immunised with autologous purified M component, together Subsequent analyses showed an increased frequency of T-cells
with interleukin 12 alone or combined with GM-CSF. This that had specific cytotoxic effects against primary lymphoma
active specific immunisation was the only treatment given to cells in 11 of 16 vaccinated patients25—an increasing immune
these patients. The effect of idiotype vaccination on response that correlated with disease-free survival. A final
circulating clonal tumour B cells was monitored by the analysis of 41 patients treated with this approach found that

Oncology Vol 5 December 2004 http://oncology.thelancet.com 729


Review Vaccines for haematological tumours

Table 2. Vaccine trials for haematological malignant diseases

Cancer Clinical Vaccine Study design Toxic Response Immunological Ref


setting effects* findings
Multiple Stage I–III Idiotype Phase I Few OR, two of five T-cell response: 19
myeloma Immunological adjuvant: n=5 (decreased CD19- cytokine release, three
aluminium hydroxide positive B cells) of five
Route: intradermal OR better in immune B-cell response, three of
responders five
Multiple Stage II Idiotype Phase I Not OR, one of five (50% T-cell response: prolif- 20
myeloma Immunological adjuvant: n=5 reported decrease in serum eration, one of five;
aluminium hydroxide and paraprotein) cytokine release,
GM-CSF five of five
Route: intradermal B-cell response: five of five
Multiple Stage I Idiotype Phase I Few OR, five of six - T-cell response, 21
myeloma Immunological adjuvant: n=6 (molecular remission) four of six
aluminium hydroxide and
interleukin 12, with or
without GM-CSF
Route: intradermal
Multiple First remission Idiotype Phase II Few OR, none of 15- T-cell response: 22,23
myeloma Immunological n=15 (molecular remission) delayed-type hyper-
adjuvant: KLH and OS similar to sensitivity, 12 of 15
GM-CSF historical controls B-cell response,
Route: subcutaneous three of 15
B-cell Measurable Idiotype Phase II Few CR, two of 20 T-cell response: 24,25
non-Hodgkin disease (n=20) Immunological adjuvant: n=41 OS better in immune proliferation, seven
lymphoma or KLH and Thr-MDP responders of 41; frequency†,
(follicular) clinically Route: subcutaneous 11 of 16
complete B-cell response,
remission 17 of 41
B-cell Clinically Idiotype Phase II Few OR, eight of 11 T-cell response: 26
non-Hodgkin complete Immunological adjuvant: n=20 (molecular remission) cytokine release, 19
lymphoma remission KLH and GM-CSF of 20; cytotoxicity, six
(follicular) Route: subcutaneous of six
B-cell response, 15 of 20
B-cell Measurable Idiotype Phase I–II Few CR, two of four; B-cell response, eight 27
non-Hodgkin disease (n=4) Immunological adjuvant: n=9 molecular remission, of nine
lymphoma or clinical KLH three of five
(follicular) complete remission Route: subcutaneous
B-cell Residual DNA plasmid encoding Phase I–II Few CR, one of 12 T-cell response: delayed- 28
non-Hodgkin disease after idiotype dose escalation type hypersensitivity and
lymphoma conventional Immunological adjuvant: n=12 proliferation, six of 12
treatment none
Route: intramuscular with or
without intradermal injection
B-cell Stage IV Peptide: complementartity Pilot study Few OR, 0% T-cell response: prolif- 29
non-Hodgkin determining region 3 n=1 eration, cytokine release,
lymphoma Immunological adjuvant: and cytotoxicity, one
(follicular) KLH and GM-CSF of one
Route: subcutaneous
Chronic Partial or Peptide: BCR-ABL1 Phase I dose Few OR not reported T-cell response: delayed- 30
myeloid complete Immunological adjuvant: escalation type hypersensitivity, two
leukaemia remission QS-21 n=12 of 12; proliferation, three
Route: subcutaneous Restriction: HLA-A3, of 12; cytotoxicity, none of
HLA-A11, HLA-B8, 12
and HLA-DR
Chronic Chronic phase, Peptide: BCR-ABL1 Phase II Few OR, five of 14 T-cell response: CD4+ 31
myeloid measurable Immunological adjuvant: n=14 (molecular remission), T-cell proliferation and
leukaemia disease QS-21 Restriction: HLA delayed-type hyper-
Route: subcutaneous class I and class II sensitivity, 14 of 14; CD4+
T-cell cytokine release,
11 of 14; CD8+ T-cell
cytokine release four of 14
Acute Measurable Peptide: WT1 Pilot study Few CR, one of one T-cell response: 32
myeloid disease Immunological adjuvant: n=1 frequency and
leukaemia KLH and GM-CSF CK release, one of
Restriction: HLA-A2 one
Route intradermal and
subcutaneous Continued . . .

730 Oncology Vol 5 December 2004 http://oncology.thelancet.com


Vaccines for haematological tumours
Review

Table 2. Vaccine trials for haematological malignant diseases (continued)

Cancer Clinical Vaccine Study design Toxic Response Immunological Ref


setting effects* findings
B-cell chronic Progressive, Autologous tumour Phase I–II study Few OR, nine of 11 T-cell response: cytokine 33
lymphocytic intermediate, (engineered CD40 ligand) dose escalation (decreased release and proliferation,
leukaemia or high-risk Immunological adjuvant: n=11 blood-lymphocyte three of three
disease none count)
Route: intravenous
Multiple After high-dose Autologous tumour Phase I study Few OR, none of one T-cell response: cytokine 34
myeloma chemotherapy (engineered interleukin 2) n=8 (decreased serum release and cytotoxicity,
Immunological adjuvant: paraprotein) 0%
none
Route: subcutaneous
Low-grade Measurable Heat-shock protein (GP96) Phase II study Few PR, one of ten Not reported 35
non-Hodgkin disease Immunological adjuvant: n=10
lymphoma none
Route: intradermal
Multiple Advanced, Dendritic cells pulsed with Pilot study Few OR, transient minor T-cell response: 36
myeloma refractory idiotype n=1 decrease in proliferation,
disease Immunological adjuvant: serum paraprotein cytokine release and
none cytotoxicity, one of one
Route: intravenous
Multiple Advanced, Dendritic cells pulsed with Pilot study Few OR not reported T-cell response: 37
myeloma refractory idiotype n=2 proliferation and cytokine
disease Immunological adjuvant: release, two of two;
KLH and GM-CSF cytotoxicity, none of two
Route: intravenous B-cell response, one of two
Multiple IgG multiple Dendritic cells pulsed with Phase I Few OR, one of six T-cell response: prolifer- 38
myeloma myeloma idiotype n=6 (decreased serum ation, five of six; cytokine
Immunological adjuvant: paraprotein) release, two of six;
none cytotoxicity, three of six
Route: intravenous B-cell response, four of
five
Multiple Measurable Dendritic cells pulsed with Phase I–II Few OR, one of 12 T-cell response: 39
myeloma disease (n=10) idiotype (with subsequent n=12 (decreased serum proliferation, two of 12;
or clinically idiotype and KLH) paraprotein) cytotoxicity, one of three
complete Immunological adjuvant: Immune response
remission none correlated with clinical
Route: intravenous outcome
Multiple Advanced Dendritic cells pulsed with Phase I–II Few OR, one of 11 T-cell response: 40
myeloma disease idiotype (with subsequent n=11 (decreased bone- cytokine release,
idiotype and GM-CSF) marrow plasma cells) four of ten
Route: intravenous B-cell response, three of
ten
B-cell Measurable Dendritic cells pulsed with Phase II Few CR, four of 28 B-cell or T-cell response: 41,42
non-Hodgkin disease idiotype (with subsequent n=35 PR, six of 28 proliferation, 23 of 33
lymphoma (n=28) or idioytpe and KLH)
(follicular) clinically Immunological adjuvant:
complete none
remission Route: intravenous
Cutaneous Measurable Dendritic cells pulsed with Phase I–II Few CR, one of ten T-cell response: 43
T-cell non- refractory tumour n=10 PR, four of ten delayed-type
Hodgkin disease Immunological adjuvant: hypersensitivity, three of
lymphoma none eight; proliferation and
Route: intranodal cytokine release, three of
five
Chronic Interferon alfa- Autologous chronic Pilot study Skin OR, 0% T-cell response: delayed- 44
myeloid resistant myeloid leukaemic cells n=3 ulceration type hypersensitivity,
leukaemia disease and dendritic cells No HLA-matched (n=1) cytokine release, two of
Immunological adjuvant: donors available three; cytotoxicity, none
KLH of three
Route: intradermal
*Few toxic effects refers to no grade III–IV toxic effects. †Frequency refers to concentration of tumour-specific T cells in peripheral blood of patients. OR, overall response; CR,
complete response; PR, partial response; GM-CSF, granulocyte-monocyte colony-stimulating factor; KLH, keyhole limpet haemocyanin; Thr-MDP, threonyl-muramyl dipeptide.

Oncology Vol 5 December 2004 http://oncology.thelancet.com 731


Review Vaccines for haematological tumours

20 patients developed an idiotype-specific humoral or cellular immunoglobulin,and there was one new responder. No other
immune response.24 Effective immunisation significantly patients showed idiotype-specific humoral or T-cell
correlated with improved freedom from disease progression reactivity.
compared with historical controls or with those who were
vaccinated but did not mount a detectable immune response. Peptide vaccines
Thus, these results suggest that patients might benefit from Peptide-based vaccines need knowledge, and matching, of a
such active specific immunisation if the vaccine is sufficiently patient’s HLA haplotype and expression profile of tumour-
immunogenic to induce a measurable humoral or cellular associated antigens for appropriate vaccination of each
immune response. individual. Most known tumour-associated antigens are
presented in association with class I MHC molecules and are
DNA vaccines recognised by the patient’s tumour-specific CD8-positive T
DNA plasmids consist of a gene for a tumour-associated cells; a few tumour-associated-antigen epitopes are presented
antigen that is regulated by a promoter with constitutive in association with class II MHC molecules and are
activity. Because of the striking advances in genetic recognised by CD4-positive T cells. Although many T-cell-
engineering, such vaccine formulation is especially suitable defined epitopes of tumour-associated antigens are now
for large-scale production. The protein antigen produced by available for potential clinical application as vaccines, most
the target cells (usually myocytes or fibroblasts depending on are expressed by melanoma cells and few epitopes have been
the injection route) is taken up by host antigen-presenting characterised in other tumours, especially those that are
cells, processed, and cross-presented to the immune system haematological.50 Although case reports of peptide-based
in draining lymph nodes, although direct transfection of rare immunisation have been reported for other haematological
antigen-presenting cells that reside at the injection site has malignant disorders (table 2), the largest (although still
also been shown.48 A limitation of DNA vaccines common to limited) clinical experience has been described in patients
all antigen-specific immunotherapy is its restricted use in the with chronic myeloid leukaemia. One possible tumour-
few tumours with molecularly defined tumour-associated associated antigen for this disease is the BCR–ABL1 chimeric
antigens. Although results from animal studies of fusion protein, which is uniquely expressed by chronic
haematological malignant disease have been encouraging,49 myeloid leukaemic cells. Preclinical studies51,52 found that this
clinical implementation of such active specific immunisation protein has immunogenic peptides, and two trials have been
is still in its early stages. Because idiotype protein is laborious done in humans (table 2), with others under way (table 3).
and time-consuming to produce, DNA vaccination is an Other tumour-associated antigens specific to leukaemia are
attractive alternative for delivery of idiotype vaccines, because under assessment as a source of immunogenic peptides. For
its DNA can be isolated rapidly by PCR. example, myeloblastin precursor protein (previously called
leucocyte proteinase 3) is a myeloid-tissue-restricted 26 kDa
Non-Hodgkin lymphoma serine protease that is abundantly expressed in azurophil
Only one clinical trial on a DNA vaccine for the treatment of granules in healthy myeloid cells, and is overexpressed by
haematological malignant disorders has been reported to between two fold and five fold in some leukaemia cells where
date. Timmerman and co-workers28 did a phase I–II clinical it could be important for maintenance of leukaemic
trial to study the safety and immunogenicity of naked DNA phenotype. Cytotoxic-T-lymphocyte cell lines that recognise
idiotype vaccines in 12 patients with follicular B-cell a HLA-A2.1-restricted nonapeptide derived from
lymphoma.28 The DNA encoded a chimeric Ig molecule that myeloblastin precursor preferentially lyses progenitor and
had variable heavy-chain and light-chain sequences derived malignant cells from chronic myeloid leukaemia over healthy
from each patient’s tumour, which were linked to the IgG2a bone-marrow cells.53 Active specific immunisation with
and ␬ mouse Ig heavy-chain and light-chain constant regions, myeloblastin precursor protein is under assessment in a
respectively. Patients received intramuscular injections of clinical trial (table 3).
DNA according to a dose-escalation regimen. After
vaccination, seven of 12 patients mounted either a humoral Chronic myeloid leukaemia
or T-cell proliferative response to the mouse component of Pinilla-Ibarz and colleagues30 did a dose-escalation study of a
the vaccine, and one patient had a measurable T-cell response vaccine made from multivalent peptides (ie, five peptides
specific to autologous idiotype. Antibodies against the restricted by HLA class I and II) that spanned the b3a2
idiotype were not detected in any patient. Patients then breakpoint of the BCR–ABL1 fusion protein.30 The
received a second series of vaccinations to deliver the researchers found peptide-specific delayed-type-
maximum DNA dose of the escalation series both hypersensitivity reactions in vivo and T-cell proliferation in
intramuscularly and intradermally. Nine of 12 patients had a vitro, but no peptide-specific cytotoxic-T-cell responses in
humoral or T-cell response to mouse Ig, and six of 12 patients vitro.
showed an idiotype-specific humoral or T-cell response.
Subsequently, a third series of vaccinations mixed idiotype Polyvalent vaccines
DNA with DNA from GM-CSF. The number of patients who A limitation of antigen-specific vaccines is that immune
responded to mouse Ig was about the same (eight of 12). responses will be restricted to the single tumour-associated
Seven of the nine patients who previously responded antigen targeted by the vaccine. Consequently, although this
continued to show humoral or T-cell reactivity toward mouse strategy might initially eliminate tumours that express the

732 Oncology Vol 5 December 2004 http://oncology.thelancet.com


Vaccines for haematological tumours
Review

Table 3. Vaccine trials in progress for haematological malignant diseases

Trial Cancer Clinical setting Vaccine Study design


UNMC-260-00 B-cell non-Hodgkin After autologous transplantation Idiotype Phase II
lymphoma (follicular) Immunological adjuvant: n=20
KLH and GM-CSF
NCI-00-C-0050 B-cell non-Hodgkin Stage III–IV responsive to conventional Idiotype Phase III
lymphoma (follicular) treatment Immunological adjuvant: randomised trial
KLH and GM-CSF n=563
CUMC-0101-142 B-cell non-Hodgkin Stages III and IV responsive Idiotype Phase III
lymphoma (follicular) to conventional treatment Immunological adjuvant: randomised trial
KLH and GM-CSF n=360
NCI-00-C-0133 Mantle-cell non-Hodgkin All stages of de novo disease Idiotype Phase II
lymphoma Immunological adjuvant: n=26
KLH and GM-CSF
FAV-ID-01 Low-grade non-Hodgkin All stages Idiotype Phase II
lymphoma Immunological adjuvant: n=25
KLH and GM-CSF
NCI-00-C-0201 Multiple myeloma Stage II–III after allogeneic stem-cell Idiotype Phase I–II
transplantation Immunological adjuvant: n=22
KLH and GM-CSF
DM99-412 Chronic lymphocytic Binet stage A DNA plasmid encoding Phase I–II
leukaemia idiotype
NCI-01-C-0069 B-cell non-Hodgkin Stages III and IV de novo disease or recurrent Autologous tumour Phase II
lymphoma (follicular) disease Immunological adjuvant: n=20
interleukin 2
JHOC-JO115, Multiple myeloma De novo disease Autologous tumour Phase I–II
K0007 Immunological adjuvant: n=15
GM-CSF (secreted by
bystander cell line)
JHOC-JO115, Acute myeloid leukaemia De novo disease Autologous tumour Phase I–II
K0009 Immunological adjuvant: n=20
GM-CSF (secreted by
bystander cell line)
H11541 Chronic lymphocytic Untreated disease or complete remission Autologous tumour Phase I dose
leukaemia engineered with escalation
interleukin 2 and CD40
ligand
MDA-DM-97325 Acute myeloid leukaemia, Accelerated phase Peptide PR-1 Phase I–II
chronic myeloid leukaemia HLA-A2 restriction Immunological adjuvant: n=60
Not eligible for conventional therapy incomplete Freund’s adjuvant
MSKCC-99012 Chronic myeloid leukaemia Chronic phase BCR-ABL1 peptide Phase II
t(9;22) and b3a3 breakpoint-positive disease Immunological adjuvant: n=24
QS-21
KLH, keyhole limpet haemocyanin; GM-CSF, granulocyte-monocyte colony-stimulating factor.

tumour-associated antigen, the patient will ultimately relapse outcome.


with a tumour that no longer expresses the tumour-
associated antigen against which they were vaccinated. Whole-cell polyvalent vaccines
Furthermore, with few exceptions, it is still unclear whether A vaccine formulation that contains the tumour cell has a
tumour-associated antigens identified so far are broad range of tumour-associated antigens that could serve as
immunodominant proteins to which effective immune targets for the immune system. Because of advances over the
responses can be generated. Because polyvalent vaccines are past decade in gene-transfer techniques various tumour cells
made of a pool of several tumour-associated antigens, they have been genetically modified to either secrete cytokines (eg,
are thought to overcome the limitations of interleukin 2 and GM-CSF), or to express components of the
immunotherapeutic strategies based on single tumour- cell membrane such as adhesion molecules or costimulatory
associated antigens. molecules.54 This approach increases the immunogenicity of
However, most tumour-associated antigens of polyvalent malignant cells either by enhanced presentation of tumour
vaccines are molecularly unknown and immunological antigens, or by enhanced costimulatory signals to the T-cell
monitoring of patients given polyvalent vaccination is either arm of the immune system.
non-specific (eg, measurement of delayed-type However, development of this vaccination strategy is
hypersensitivity) or is limited to the tumour-associated hindered by the time needed for labour-intensive
antigen known to be present in the vaccine formulation, preparation of the vaccine and by the variability in the
making it difficult to correlate immune response with clinical cytokine production of each patient’s vaccine formulation.

Oncology Vol 5 December 2004 http://oncology.thelancet.com 733


Review Vaccines for haematological tumours

To overcome such drawbacks, investigators have developed Heat-shock proteins


an allogeneic bystander cell line (called K562) that secretes These intracellular molecules act as chaperones for
large and stable amounts of GM-CSF.55 This cell line can be peptides,58 and complexes of heat-shock proteins and
grown easily in suspension and has no detectable expression peptides can be isolated—a fairly easy way to obtain a
of HLA class I or class II molecules, and thus minimises the molecularly undefined cocktail of epitopes derived from
likelihood of anti-bystander allogeneic responses with tumour-associated antigens. When extracted from tumour
multiple vaccinations. This strategy of universal bystander samples, heat-shock proteins can be administered as
vaccine obviates the need for gene modification for each vaccines to humans; generation of a cellular immune
individual tumour source and ensures uniform cytokine response will need cross-presentation of the peptides
production, thereby eliminating intra-patient and inter- associated with these proteins. Dendritic cells have a specific
patient variability with each vaccination. This means of receptor for heat-shock proteins called low-density
active specific immunisation is presently being tested in two lipoprotein receptor-related protein 1, engagement of
trials (table 3). which leads to maturation of dendritic cells.59 Thus, heat-
shock proteins function both as multiple-peptide carriers
Chronic lymphocytic leukaemia and as natural immunological adjuvants.
Wierda and co-workers56 transduced the tumour cells of For solid cancers, some trials of anticancer vaccination
11 patients with CLL by use of an adenoviral expression based on heat-shock proteins have been published and
vector encoding the mouse gene Tnfsf5 (tumour necrosis some randomised studies are under way;60 however, only
factor ligand superfamily member 5, previously called CD40 one trial enrolling patients with haematological malignant
ligand). This study was based on the preclinical observation disorders has been published. In this study, Younes35 treated
that CLL cells transduced with Tnfsf5 became highly ten patients who had indolent non-Hodgkin lymphoma
proficient at antigen presentation and induced autologous with autologous heat-shock proteins and recorded partial
T-cell responses that generated specific cytotoxic tumour regression in one patient (table 2).
T-lymphocytes. Each patient received a single infusion of
the genetically modified CLL cells in a phase I dose- Dendritic-cell-based vaccines
escalation trial. 4 weeks after treatment, an in vitro assay A pivotal factor for an effective antitumour immune
showed an increase in T cells that secreted interferon ␥ in response is the optimum processing and presentation of
response to CLL cells activated by TNFRSF5 (tumour tumour-associated-antigen epitopes to T cells in the
necrosis factor receptor superfamily member 5). 6 months appropriate HLA context. Dendritic cells are the most
after therapy, T cells maintained vigorous proliferation and potent antigen-presenting cells that initiate an effective
higher interferon-␥ release than baseline values. When a T-cell response. The enhanced priming of T cells by
sizeable number (ie, more than 39%) of the infused cells dendritic cells, as well as new techniques for obtaining large
expressed the Tnfsf5 transgene, there was a decrease in numbers of human dendritic cells, has made possible the
circulating tumour-cell load as well as regressions of lymph- use of these cells for therapeutic vaccination.61,62 However,
node enlargements. However, the design of the study meant vaccine preparation remains time consuming and labour
it was not possible to define whether the immunological intensive, which is probably why no randomised trials have
and clinical effects of treatment were attributable to yet been done for any type of cancer.63 Two general
expression of Tnfsf5 or to the adenovirus vector backbone. strategies have been used to obtain human dendritic cells
for clinical studies: first, purification of immature
Multiple myeloma dendritic-cell precursors from peripheral blood; and
A phase I assessment of vaccination with interleukin 2 and second, differentiation in vitro of dendritic cells from
virally transduced autologous plasma cells by Trudel and peripheral-blood CD14-positive monocytes or from CD34-
colleagues34 found that vaccine was effective in seven of positive haemopoietic stem cells.
eight patients. 2 months after high-dose therapy, six Although blood dendritic cells might not need extensive
patients received between one and five injections of the culture, they must be activated through molecularly defined
vaccine. Moreover, injection with tumour cells induced a triggers such as toll-like receptors and TNFSF5 before
local inflammatory response consisting mainly of CD8- reinfusion, especially since non-activated (or improperly
positive T cells and TIA1-positive T cells, a finding also activated) dendritic cells can cause tolerance instead of
reported in patients with melanoma metastases who immunisation.64 Culturing of dendritic cells from CD14-
responded to peptide-based active specific immunisation.57 positive monocytes or CD-34 positive haemopoietic stem-
The multiple myeloma-specific immune response was not cell cultures occurs in two steps. First, immature dendritic
enhanced after vaccination, but only one patient was cells are generated initially in the presence of GM-CSF and
assessed. Similarly, there was no clinical response in the one interleukin 4 for 5–7 days. These cells have high expression
patient who had measurable disease at time of vaccination. of MHC, adhesion, and costimulatory molecules, and can
These results support the feasibility that generation of process and present antigens to T cells. Second, further
plasma-cell vaccines modified by an adenovirus vector is culture with tumour-necrosis factor ␣, TNFSF5, or a
technically feasible and can be administered safely after monocyte-conditioned medium results in stable maturation
transplantation. Further studies of immunological and of the dendritic cells.
clinical effectiveness of plasma-cell vaccines are needed.

734 Oncology Vol 5 December 2004 http://oncology.thelancet.com


Vaccines for haematological tumours
Review

Multiple myeloma that continuation of vaccination with new tumour lysate


In a pilot study by Lim and co-workers,38 six patients with reinduced regression of progressive disease in two patients.
IgG multiple myeloma were vaccinated with intravenous
infusions of dendritic cells derived from peripheral-blood Chronic myeloid leukaemia
mononuclear cells pulsed with autologous idiotype.38 Fusion of tumour cells with dendritic cells (ie, a
Although both a B-cell and a T-cell immune response were tumour–dendritic cell hybrid) has been proposed for the
found in most cases tumour responses were only minor, preparation of vaccines for solid cancers.65 Leukaemic-cell
which consisted of a modest (25%) but consistent decrease progenitors have the unique feature of acting as professional
in serum concentration of idiotype in one patient. In two antigen-presenting cells that express the entire repertoire of
other clinical trials on dendritic-cell-based active specific tumour-associated antigens, and are thus an enticing tool
immunisation, investigators combined idiotype-pulsed for development of autologous vaccine formulation in the
infusions of dendritic cells with idiotype-booster presence of the necessary immunological costimulation.66
immunisations. Reichardt and colleagues39 reported on Several groups have reported the generation of leukaemic-
12 patients who had undergone autologous peripheral derived dendritic cells obtained by the culturing of
stem-cell transplantation and who were then given a series leukaemic blasts in the presence of various combinations of
of monthly immunisations of two intravenous infusions of GM-CSF, interleukin 4, tumour necrosis factor ␣, and
idiotype-pulsed autologous dendritic cells followed by TNFSF5.14–17
boost immunisations with subcutaneous idiotype-KLH.39 Ossenkoppele and colleagues44 reported on the clinical
This strategy was tolerated well: patients had only minor implementation of such an immunisation strategy. Accrual
side-effects. Furthermore, two of 12 patients developed was prematurely interrupted because of the availability of
idiotype-specific cellular proliferative immune response, imatinib for chronic myeloid leukaemia resistant to
and one of three patients developed an idiotype-specific treatment with interferon alfa.67 Although no haemato-
cytotoxic-T-cell responses despite recent high-dose therapy. logical or cytogenetic responses were achieved, two of three
Specificity was proven in this assay by artificial expression patients developed strong delayed-type hypersensitivity
of idiotype genes in autologous fibroblasts by use of against leukaemic dendritic cells and uncultured chronic
adenoviral gene transfer. The researchers concluded that myeloid leukaemic cells. Because many patients with
dendritic-cell-based idiotype vaccination is feasible after chronic myeloid leukaemia who have complete cytogenetic
peripheral-blood stem-cell transplantation and can elicit response after imatinib are still BCR–ABL1 positive at PCR
idiotype-specific T-cell responses in patients with multiple analysis and because resistance to imatinib might be
myeloma. The lower rate of induction of immune responses acquired, the researchers conclude that the potential of
in this group of patients compared with patients with non- active specific immunisation should be investigated further
secreting lymphomas who were vaccinated on a similar in the adjuvant setting.
schedule might be a result of the inhibitory effect of
circulating paraprotein, or caused by immunosuppressive Conclusion
effects of the previous high-dose chemotherapy. Although Conventional approaches to most haematological
no firm evidence for clinical benefit was found, two patients malignant disorders can achieve clinically evident tumour
who developed a cellular idiotype-specific immune responses in a substantial number of patients. However,
response remained in complete remission. these treatments lack tumour specificity and have been
Finally, Titzer and colleagues40 showed the feasibility of associated with high systemic toxic effects. Although early
vaccination with CD34-positive stem-cell-derived dendritic clinical studies of cancer vaccines showed that specificity
cells incubated ex vivo with autologous idiotype. Infusions could be achieved without high toxic effects, so far they
were followed by vaccination with idiotype-GM-CSF or have shown a limited effect in terms of clinical benefits.
idiotype-pulsed dendritic cells. Treatment evoked Reasons for this are multifactorial and still largely
increasing anti-idiotype titres in three of ten vaccinated unknown. A central issue is the difficulty to overcome the
patients, and in four patients an increase in the frequency of intrinsically poor immunogenicity of tumour-associated
idiotype-specific T cells was measured by ELISPOT assay for antigens so far identified and used in the clinical setting.
interferon ␥. One patient showed a significant decrease in Several efforts have been made to increase the
plasma-cell infiltration of the bone marrow. immunogenicity of tumour-associated antigens, but clinical
Maier and co-workers43 have used dendritic-cell-based implementation of recent technologies and immunological
vaccination for treatment of patients with cutaneous T-cell insights (eg, vaccines based on dendritic cells and
lymphoma. Patients were given weekly intranodal injections genetically engineered vaccines) is still in its infancy.
of mature monocyte-derived dendritic cells pulsed with However, after the sequencing of the human genome, the
autologous tumour lysate admixed with KLH. Tumour- rapid diffusion of high-throughput technologies such as
specific immune response was recorded in three of eight DNA array, proteomics should accelerate the discovery of
patients (delayed-type hypersensitivity) and in three of five new and more immunogenic tumour-associated antigens
patients (proliferation and cytokine production). Tumour suitable for anticancer vaccination.68,69 The immune
regression was seen in five of ten patients, with one responses seen in several studies together with low rates of
complete response and four partial responses. As in the tumour regression suggest that current methods of
study by Titzer and colleagues,40 these researchers showed immunological follow-up might be inadequate to show an

Oncology Vol 5 December 2004 http://oncology.thelancet.com 735


Review Vaccines for haematological tumours

2 Dreger P, Montserrat E. Autologous and allogeneic stem cell


Search strategy and selection criteria transplantation for chronic lymphocytic leukemia. Leukemia 2002;
Data for this review were identified by PubMed using the 16: 985–92.
searches terms “cancer vaccines”, “haematological 3 Voliotis D, Diehl V. Challenges in treating hematologic
malignancies. Semin Oncol 2002; 29: 30–39.
malignancies”, “leukaemia”, “lymphoma”, “multiple myeloma”, 4 Burnett AK, Goldstone AH, Stevens RM, et al. Randomised
“clinical trials” and the types of cancer vaccination (eg, comparison of addition of autologous bone-marrow
“idiotype vaccine”). Continuing clinical trials were also transplantation to intensive chemotherapy for acute myeloid
searched at National Cancer Institute (NCI) website (http:// leukaemia in first remission: results of MRC AML 10 trial.
Lancet 1998; 351: 700–08.
www.cancer.gov/clinicaltrials). Only papers published in
5 Roush KS, Hillyer CD. Donor lymphocyte infusion therapy.
English were reviewed. Transfus Med Rev 2002; 16: 161–76.
6 Campbell P, Marcus R. Monoclonal antibody therapy for
lymphoma. Blood Rev 2003; 17: 143–52.
effective immunisation status.70 Alternatively, excessive 7 Yotnda P, Firat H, Garcia-Pons F, et al. Cytotoxic T cell response
against the chimeric p210 BCR-ABL protein in patients with
tumour burdens might overwhelm the immune system, chronic myelogenous leukemia. J Clin Invest 1998; 101: 2290–96.
even in the presence of tumour-specific lymphocytes. 8 Li Y, Bendandi M, Deng Y, et al. Tumour-specific recognition of
Although complete haematological or cytogenetic human myeloma cells by idiotype-induced CD8(+) T cells.
Blood 2000; 96: 2828–33.
remissions are often achieved in several haematological 9 Costello RT, Fauriat C, Rey J, et al. Immunobiology of haemat-
tumours after conventional treatments, persistence of ological malignant disorders: the basis for novel immunotherapy
minimal residual disease is likely to underlie the lack of protocols. Lancet Oncol 2004; 5: 47–55.
survival benefit often reported in these patients. Therefore, 10 Gilboa E. Opinion: the promise of cancer vaccines. Nat Rev Cancer
2004; 4: 401–11.
the adjuvant setting seems to be the most suitable therapeutic 11 Padua RA, Larghero J, Robin M, et al. PML-RARA-targeted DNA
scenario for active specific immunisation, where the vaccine induces protective immunity in a mouse model of leukemia.
immunosuppressive effects of bulky disease are almost Nat Med 2003; 9: 1413–17.
12 Schwartz R. T cell anergy. Annu Rev Immunol 2003; 21: 305–34.
absent and the effector-target ratio is favourable. These 13 Trentin L, Perin A, Siviero M, et al. B7 costimulatory molecules
theoretical advantages might be counterbalanced by the from malignant cells in patients with B-cell chronic
global immune suppression that accompanies the early lymphoproliferative disorders trigger T-cell proliferation. Cancer
2000; 89: 1259–68.
period after transplantation. However, studies in animals 14 Harrison BD, Adams JA, Briggs M, et al. Stimulation of autologous
have shown the presence of an autologous graft-versus-host proliferative and cytotoxic T-cell responses by leukemic dendritic
effect early after transplantation, which is normally transient cells derived from blast cells in acute myeloid leukemia. Blood 2001;
97: 2764–71.
but might be extended after vaccination.71,72 15 Clark RE, Dodi IA, Hill SC, et al. Direct evidence that leukemic cells
Overall, the exceptional complexity of the immune present HLA-associated immunogenic peptides derived from the
network and of the interactions between the tumour and the BCR-ABL b3a2 fusion protein. Blood 2001; 98: 2887–93.
immune system make the task of identifying the optimum 16 Choudhury BA, Liang JC, Thomas EK, et al. Dendritic cells derived
in vitro from acute myelogenous leukemia cells stimulate
vaccine formulation (eg, tumour-associated antigen, autologous, antileukemic T-cell responses. Blood 1999; 93: 780–86.
immunological adjuvant, and vector) and regimen (eg, dose, 17 Nabarro S, Thrasher AJ, Kempski H, et al. Generation of
route, and schedule) highly challenging, so too will be the immunostimulatory dendritic cells from the malignant clone in
patients with juvenile myelomonocytic leukemia. Leukemia 2003;
task of identifying the clinical setting needed 17: 1910–12.
to achieve the greatest clinical benefit.73 Available data in 18 McCarthy H, Ottensmeier CH, Hamblin TJ, Stevenson FK.
humans do not allow any definitive conclusion to be drawn Anti-idiotype vaccines. Br J Haematol 2003; 123: 770–81.
19 Bergenbrant S, Yi Q, Osterborg A, et al. Modulation of anti-
on the role of active specific immunisation as part of idiotypic immune response by immunisation with the autologous
standard treatment for any haematological malignant M-component protein in multiple myeloma patients. Br J Haematol
disease; however, they do support the hypothesis that 1996; 92: 840–46.
20 Osterborg A, Yi Q, Henriksson L, et al. Idiotype immunization
appropriate therapeutic interventions might redirect combined with granulocyte-macrophage colony-stimulating factor
adaptive immunity against malignant cells. In the near in myeloma patients induced type I, major histocompatibility
future, completion of randomised phase III trials of cancer complex-restricted, CD8- and CD4-specific T-cell responses. Blood
1998; 91: 2459–66.
vaccines, as well as clinical implementation of the most 21 Rasmussen T, Hansson L, Osterborg A, et al. Idiotype vaccination in
recent insights into tumour immunology that aim to break multiple myeloma induced a reduction of circulating clonal tumour
immune tolerance towards malignant cells, should allow B cells. Blood 2003; 101: 4607–10.
22 Coscia M, Mariani S, Battaglio S, et al. Long-term follow-up of
investigators to define the actual role of active specific idiotype vaccination in human myeloma as a maintenance therapy
immunisation in the management of haematological cancers. after high-dose chemotherapy. Leukemia 2004; 18: 139–45.
23 Massaia M, Borrione P, Battaglio S, et al. Idiotype vaccination in
Conflict of interest human myeloma: generation of tumour-specific immune responses
after high-dose chemotherapy. Blood 1999; 94: 673–83.
We declare no conflicts of interest.
24 Hsu FJ, Caspar CB, Czerwinski D, et al. Tumour-specific idiotype
vaccines in the treatment of patients with B-cell lymphoma: long-
Acknowledgemnts term results of a clinical trial. Blood 1997; 89: 3129–35.
This work was supported in part by grants from the Italian Association 25 Nelson EL, Li X, Hsu FJ, et al. Tumour-specific, cytotoxic
for Cancer Research and from the Italian Ministry of Health 2002. T-lymphocyte response after idiotype vaccination for B-cell,
non-Hodgkin’s lymphoma. Blood 1996; 88: 580–89.
References 26 Bendandi M, Gocke CD, Kobrin CB, et al. Complete molecular
1 Bacigalupo A, Frassoni F, Van Lint MT. Bone marrow or peripheral remissions induced by patient-specific vaccination plus
blood as a source of stem cells for allogeneic transplantation. granulocyte-monocyte colony-stimulating factor against
Haematologica 2002; 87: 4–8. lymphoma. Nat Med 1999; 5: 1171–77.

736 Oncology Vol 5 December 2004 http://oncology.thelancet.com


Vaccines for haematological tumours
Review

27 Barrios Y, Cabrera R, Yanez R, et al. Anti-idiotypic vaccination in 50 Parmiani G, Castelli C, Dalerba P, et al. Cancer immunotherapy
the treatment of low-grade B-cell lymphoma. Haematologica 2002; with peptide-based vaccines: what have we achieved? Where are we
87: 400–07. going? J Natl Cancer Inst 2002; 94: 805–18.
28 Timmerman JM, Singh G, Hermanson G, et al. Immunogenicity of 51 Berke Z, Andersen MH, Pedersen M, et al. Peptides spanning the
a plasmid DNA vaccine encoding chimeric idiotype in patients with junctional region of both the ABL/BCR and the BCR/ABL fusion
B-cell lymphoma. Cancer Res 2002; 62: 5845–52. proteins bind common HLA class I molecules. Leukemia 2000;
29 Wen YJ, Lim SH. In-vivo immune responses to idiotypic VH 14: 419–26.
complementarity-determining region 3 peptide vaccination in B- 52 Wagner WM, Ouyang Q, Pawelec G. The ABL/BCR gene product as
cell non-Hodgkin’s lymphoma. Br J Haematol 1998; 103: 663–68. a novel leukemia-specific antigen: peptides spanning the
30 Pinilla-Ibarz J, Cathcart K, Korontsvit T, et al. Vaccination of fusion region of abl/bcr can be recognized by both CD4+ and
patients with chronic myelogenous leukemia with BCR-ABL CD8+ T lymphocytes. Cancer Immunol Immunother 2003; 52:
oncogene breakpoint fusion peptides generates specific immune 89–96.
responses. Blood 2000; 95: 1781–87. 53 Molldrem JJ, Clave E, Jiang YZ, et al. Cytotoxic T lymphocytes
31 Cathcart K, Pinilla-Ibarz J, Korontsvit T, et al. A multivalent BCR- specific for a nonpolymorphic proteinase 3 peptide preferentially
ABL fusion peptide vaccination trial in patients with chronic inhibit chronic myeloid leukemia colony-forming units. Blood 1997;
myeloid leukemia. Blood 2004; 103: 1037–42. 90: 2529–34.
32 Mailander V, Scheibenbogen C, Thiel E, et al. Complete remission 54 Nawrocki S, Wysocki PJ, Mackiewicz A. Genetically modified
in a patient with recurrent acute myeloid leukemia induced by tumour vaccines: an obstacle race to break host tolerance to cancer.
vaccination with WT1 peptide in the absence of hematological or Expert Opin Biol Ther 2001; 1: 193–204.
renal toxicity. Leukemia 2004; 18: 165–66. 55 Borrello I, Sotomayor EM, Cooke S, Levitsky HI. A universal
33 Wierda WG, Cantwell MJ, Woods SJ, et al. CD40-ligand (CD154) granulocyte-macrophage colony-stimulating factor-producing
gene therapy for chronic lymphocytic leukemia. Blood 2000; bystander cell line for use in the formulation of autologous tumour
96: 2917–24. cell-based vaccines. Hum Gene Ther 1999; 10: 1983–91.
34 Trudel S, Li Z, Dodgson C, et al. Adenovector engineered 56 Wierda WG,Cantwell MJ, Woods SJ, et al. CD40-ligand (CD154)
interleukin-2 expressing autologous plasma cell vaccination after gene therapy for chronic lymphocytic leukaemia. Blood 2000;
high-dose chemotherapy for multiple myeloma: a phase 1 study. 96: 2917–24.
Leukemia 2001; 15: 846–54. 57 Mocellin S, Rossi C, Nitti D, et al. Dissecting tumour
35 Younes A. A phase II study of heat shock protein-peptide complex- responsiveness to immunotherapy: the experience of peptide-based
96 vaccine therapy in patients with indolent non-Hodgkin’s melanoma vaccines. Biochim Biophys Acta Rev Cancer 2003; 1653:
lymphoma. Clin Lymphoma 2003; 4: 183–85. 61–71.
36 Wen YJ, Ling M, Bailey-Wood R, Lim SH. Idiotypic protein-pulsed 58 Srivastava PK, Menoret A, Basu S, et al. Heat shock proteins come
adherent peripheral blood mononuclear cell-derived dendritic cells of age: primitive functions acquire new roles in an adaptive world.
prime immune system in multiple myeloma. Clin Cancer Res 1998; Immunity 1998; 8: 657–65.
4: 957–62. 59 Binder RJ, Han DK, Srivastava PK. CD91: a receptor for heat shock
37 Cull G, Durrant L, Stainer C, et al. Generation of anti-idiotype protein gp96. Nat Immunol 2000; 1: 151–55.
immune responses following vaccination with idiotype-protein 60 Castelli C, Rivoltini L, Rini F, et al. Heat shock proteins: biological
pulsed dendritic cells in myeloma. Br J Haematol 1999; 107: 648–55. functions and clinical application as personalized vaccines for
38 Lim SH, Bailey-Wood R. Idiotypic protein-pulsed dendritic cell human cancer. Cancer Immunol Immunother 2004; 53: 227–33.
vaccination in multiple myeloma. Int J Cancer 1999; 83: 215–22. 61 Jefford M, Maraskovsky E, Cebon J, Davis ID. The use of dendritic
39 Reichardt VL, Okada CY, Liso A, et al. Idiotype vaccination using cells in cancer therapy. Lancet Oncol 2001; 2: 343–53.
dendritic cells after autologous peripheral blood stem cell 62 Fong L, Engleman EG. Dendritic cells in cancer immunotherapy.
transplantation for multiple myeloma: a feasibility study. Blood Annu Rev Immunol 2000; 18: 245–73.
1999; 93: 2411–19.
63 Figdor CG, De Vries IJ, Lesterhuis WJ, Melief CJ. Dendritic cell
40 Titzer S, Christensen O, Manzke O, et al. Vaccination of multiple
myeloma patients with idiotype-pulsed dendritic cells: immunotherapy: mapping the way. Nat Med 2004; 10: 475–80.
immunological and clinical aspects. Br J Haematol 2000; 108: 64 Steinman RM, Hawiger D, Nussenzweig MC. Tolerogenic dendritic
805–16. cells. Annu Rev Immunol 2003; 21: 685–711.
41 Hsu FJ, Benike C, Fagnoni F, et al. Vaccination of patients with 65 Phan V, Errington F, Cheong SC, et al. A new genetic method to
B-cell lymphoma using autologous antigen- pulsed dendritic cells. generate and isolate small, short-lived but highly potent dendritic
Nat Med 1996; 2: 52–58. cell-tumour cell hybrid vaccines. Nat Med 2003; 9: 1215–19.
42 Timmerman JM, Czerwinski DK, Davis TA, et al. Idiotype-pulsed 66 Stripecke R, Levine AM, Pullarkat V, Cardoso AA. Immunotherapy
dendritic cell vaccination for B-cell lymphoma: clinical and immune with acute leukemia cells modified into antigen-presenting cells: ex
responses in 35 patients. Blood 2002; 99: 1517–26. vivo culture and gene transfer methods. Leukemia 2002; 16:
43 Maier T, Tun-Kyi A, Tassis A, et al. Vaccination of patients with 1974–83.
cutaneous T-cell lymphoma using intranodal injection of 67 Druker BJ, Talpaz M, Resta DJ, et al. Efficacy and safety of a specific
autologous tumour-lysate-pulsed dendritic cells. Blood 2003; inhibitor of the BCR-ABL tyrosine kinase in chronic myeloid
102: 2338–44. leukemia. N Engl J Med 2001; 344: 1031–37.
44 Ossenkoppele GJ, Stam AG, Westers TM, et al. Vaccination of 68 Mocellin S, Riccardo Rossi C, et al. Molecular oncology in the post-
chronic myeloid leukemia patients with autologous in vitro cultured genomic era: the challenge of proteomics. Trends Mol Med 2004;
leukemic dendritic cells. Leukemia 2003; 17: 1424–26. 10: 24–32.
45 Kaminski MS, Kitamura K, Maloney DG, Levy R. Idiotype 69 Mocellin S, Wang E, Panelli M, et al. DNA array-based gene
vaccination against murine B cell lymphoma. Inhibition of tumour profiling in tumour immunology. Clin Cancer Res (in press).
immunity by free idiotype protein. J Immunol 1987; 138: 1289–96. 70 Keilholz U, Weber J, Finke JH, et al. Immunologic monitoring of
46 Bogen B, Schenck K, Munthe LA, Dembic Z. Deletion of idiotype cancer vaccine therapy: results of a workshop sponsored by the
(Id)-specific T cells in multiple myeloma. Acta Oncol 2000; 39: Society for Biological Therapy. J Immunother 2002; 25: 97–138.
783–88. 71 Borrello I, Sotomayor EM, Rattis FM, et al. Sustaining the
47 Kwak LW, Campbell MJ, Czerwinski DK, et al. Induction of graft-versus-tumour effect through posttransplant immunization
immune responses in patients with B-cell lymphoma against the with granulocyte-macrophage colony-stimulating factor
surface-immunoglobulin idiotype expressed by their tumours. (GM-CSF)-producing tumour vaccines. Blood 2000; 95:
N Engl J Med 1992; 327: 1209–15. 3011–19.
48 Shedlock DJ, Weiner DB. DNA vaccination: antigen presentation 72 Anderson LD Jr, Savary CA, Mullen CA. Immunization of
and the induction of immunity. J Leukoc Biol 2000; 68: 793–806. allogeneic bone marrow transplant recipients with tumour cell
49 King CA, Spellerberg MB, Zhu D, et al. DNA vaccines with single- vaccines enhances graft-versus-tumour activity without
chain Fv fused to fragment C of tetanus toxin induce protective exacerbating graft-versus-host disease. Blood 2000; 95: 2426–33.
immunity against lymphoma and myeloma. Nat Med 1998; 73 Finn OJ. Cancer vaccines: between the idea and the reality.
4: 1281–86. Nat Rev Immunol 2003; 3: 630–41.

Oncology Vol 5 December 2004 http://oncology.thelancet.com 737

You might also like