You are on page 1of 6

doi:10.1111/jpc.

14996

REVIEW ARTICLE

Aplastic anaemia: Current concepts in diagnosis and management


1
Eliska Furlong and Tina Carter1,2,3
1
Department of Paediatric and Adolescent Haematology, Oncology, Blood and Marrow Transplantation, Perth Children’s Hospital, 2Division of Paediatrics,
School of Medicine, University of Western Australia and 3Paediatric and Adolescent Haematology Service, PathWest Laboratory Medicine WA, Perth,
Western Australia, Australia

Aplastic anaemia is a rare, previously fatal condition with a significantly improved survival rate owing to advances in understanding of the
pathophysiology and improved treatment strategies including haematopoietic stem cell transplantation. Although a rare condition, aplastic anaemia
continues to present a high burden for affected patients, their families and the health system due to the prolonged course of disease
often associated with high morbidity and the uncertainty regarding clinical outcome. Modern molecular and genetic techniques including
next-generation sequencing have contributed to a better understanding of this heterogeneous group of conditions, albeit at a cost of increased com-
plexity of clinical decision-making regarding prognosis and choice of treatment for individual patients. Here we present a concise and comprehensive
review of aplastic anaemia and closely related conditions based on extensive literature review and long-standing clinical experience. The review takes
the reader across the complex pathophysiology consisting of three main causative mechanisms of bone marrow destruction resulting in aplastic
anaemia: direct injury, immune mediated and bone marrow failure related including inherited and clonal disorders. A comprehensive diagnostic algo-
rithm is presented and an up-to-date therapeutic approach to acquired immune aplastic anaemia, the most represented type of aplastic anaemia, is
described. Overall, the aim of the review is to provide paediatricians with an update of this rare, heterogeneous and continuously evolving condition.

Key words: aplastic anaemia; haematopoietic stem cell transplantation; inherited bone marrow failure syndrome; myelodysplasia.

Aplastic anaemia results from a destruction of haematopoietic cells aplastique’ was first used in 1904 by Louis Henri Vaquez, a French
by a variety of mechanisms ranging between direct environmental physicist and remained somewhat misleading terminology for a
insults, dysregulated immune system or an underlying inherited or condition mostly presenting with pancytopenia.
acquired bone marrow failure predisposition. The term ‘anaemia

Definition
Key Points
1 Recent advances in understanding and management of aplastic Aplastic anaemia (AA) is a descriptive term used for a combination
anaemia have resulted in significantly increased survival rate of of hypoplastic or aplastic bone marrow and a variable degree of
once fatal disease. Nonetheless, the medical and psycho-social paucity in at least two of the three main cell lines: red cells, white
burden remains high due to the prolonged course of the disease cells and/or platelets. The severity of AA is based on the Camitta
associated with high morbidity and outcome uncertainty. criteria, postulated more than 40 years ago1 (Table 1).
2 Modern molecular and genetic methods including next-
generation sequencing have become a powerful tool in under-
standing and diagnosing the condition, albeit at a cost of Epidemiology
increase complexity of clinical decision-making.
AA is a rare condition, with an estimated incidence rate of 2–2.3
3 Aplastic anaemia is a rare heterogeneous group of conditions
cases/million per year. The incidence in Asia is two to threefolds
necessitating multidisciplinary evidence-based clinical
higher reaching up to 7.4 cases/million per year.2 In almost all
approach supported by ongoing research, clinical trials and
centralisation of data in national and international registries. population-based studies the sex ratio is close to 1:1. There seem
to be two main peaks of age incidence, one among young adults
(20–25 years) and a second in the elderly. The median age at
diagnosis among children and adolescents is 8–9 years.3
Correspondence: Dr Eliska Furlong, Department of Paediatric and Ado-
lescent Haematology, Oncology, Blood and Marrow Transplantation, Perth
Children’s Hospital, 15 Hospital Avenue, Nedlands, WA 6009, Australia. Pathophysiology
Fax: +08 6456 2360; email: eliska.furlong@health.wa.gov.au
There are three main mechanisms that can lead to the develop-
Conflict of interest: None declared. ment of AA: (i) direct injury; (ii) immune mediated; and
(iii) bone marrow failure related that can be inherited or acquired
Accepted for publication 21 May 2020. (Table 2).

Journal of Paediatrics and Child Health (2020) 1


© 2020 Paediatrics and Child Health Division (The Royal Australasian College of Physicians)
Aplastic anaemia E Furlong and T Carter

Table 1 Definition and severity of aplastic anaemia based on the Table 2 Overview of pathophysiological mechanisms leading to
Camitta criteria. aplastic anaemia

Classification Criteria 1. Direct injury


Drugs
Severe Bone marrow (BM) cellularity <25% (or <50% if <30% Chemotherapy agents
of BM are haematopoietic cells) Anticonvulsants
And two or more of the following: Carbamazepine, phenytoin
Peripheral blood neutrophil count <0.5 × 109/L Antibiotics
Peripheral blood platelet count <20 × 109/L Sulphonamides, chloramphenicol
Peripheral blood reticulocyte count <20 × 109/L Anti-thyroid
Very severe As above, but peripheral blood neutrophil count Methimazole
must be <0.2 × 109/L Non-steroidal anti-inflammatory drugs
Non-severe Hypocellular BM with peripheral blood values not Indomethacin, phenylbutazone
meeting criteria for severe aplastic anaemia Other anti-inflammatory
Gold
Chemicals
Benzene, solvents, glue vapours, pesticides
Radiation
Direct injury Infection
Sepsis
Damage to the bone marrow can be iatrogenic and dose depen-
Viral
dent, as with chemotherapy and radiotherapy, resulting in a
Hepatitis A, B and C, parvovirus B19, HIV, Epstein-Barr virus,
predictable temporary state of reduced haematopoiesis. Some cytomegalovirus
drugs can have an idiosyncratic and possibly permanent effect, Infiltration
sometime occurring many weeks after the cessation of the Malignant disease
treatment.4–6 Haematological
Hepatitis-associated AA (haAA) usually develops 2–3 months Metastatic
post an episode of acute hepatitis, is often seronegative and Non-malignant
accounts for up to 10% of all cases of AA.7 An expansion of nar- Hemophagocytic lymphohistiocytosis
row spectrum T-cell receptor lymphocytes during the acute phase Depletion
Haematinic and other vitamin deficiency
of hepatitis-associated AA and its diminishing with immunosup-
Vitamin B12, folic acid, ferritin, vitamin D
pressive treatment suggests an immune-mediated mechanism in
Anorexia nervosa
this type of AA.8 2. Immune mediated
Acquired immune aplastic anaemia
Systemic auto-immune disease
Immune mediated Systemic lupus erythematosus
Eosinophilic fasciitis
Most cases of AA do not have a clear cause and have been Thymoma
labelled idiopathic. The immune mechanism of impaired Constitutional immune dysregulation
haematopoiesis in the so-called acquired idiopathic or immune CTLA-4 deficiency
DADA2
AA (iAA) has long been suspected based on observational clinical
GATA2 deficiency
and laboratory findings as well as animal studies.9 It seems that
3. Bone marrow failure related
cytotoxic T cells are at the centre of the dysregulated immune
Inherited bone marrow failure syndromes
system, either directly (activation of apoptosis via Fas/FasL path- Fanconi anaemia
way) or indirectly (increased INF-γ, TNF-α production) destroying Dyskeratosis congenita
haematopoietic stem cells.10 Regulatory T-cells are reduced in Shwachman–Diamond syndrome
patients with iAA and increase with effective treatment.11 Diamond–Blackfan anaemia
iAA is associated with specific histocompatibility antigens Congenital amegakaryocytic thrombocytopenia
(HLA).12 A clonal expansion of cells within the bone marrow car- Pearson syndrome
rying an acquired loss of heterozygosity at the HLA site of chro- Severe congenital neutropenia
Thrombocytopenia absent radii
mosome 6 can be found in up to 15% of patients with iAA, likely
Ataxia-pancytopenia syndrome
driven by selective advantage of sustained haematopoiesis.13 An
MECOM-associated syndrome
immune drive has also been hypothesised in the evolution of
Clonal disorders
glycosylphosphoinositol (GPI)-anchored protein deficient clones Paroxysmal nocturnal haemoglobinuria
characteristic for paroxysmal nocturnal haemoglobinuria (PNH), Myelodysplasia
a rare form of clonal haematopoietic stem cell disorder clinically Refractory cytopenia of childhood
manifesting as haemolytic anaemia and thrombosis. GPI-deficient
clones can be found in up to 50% of patients with iAA. Unlike in CTLA-4, cytotoxic T-lymphocyte associated antigen; DADA2, deficiency
patients with PNH, these tend to be small and rarely cause clinical of adenosine deaminase 2.
manifestations in patients with iAA.14

2 Journal of Paediatrics and Child Health (2020)


© 2020 Paediatrics and Child Health Division (The Royal Australasian College of Physicians)
E Furlong and T Carter Aplastic anaemia

Peripheral blood
amenable to prompt diagnosis. Nonetheless, these can be very sub-
(pan)cytopenia tle or even absent resulting in delayed diagnosis if not considered.
+/- low reticulocyte count

Bone marrow trephine Clonal evolution and myelodysplasia


Morphology AA often co-exists with an appearance of distinctive cell clones
hypoplasia/aplasia
dysplasia
arising from the bone marrow. These presumably emerge under
a selective immune pressure as suggested in the case of GPI-
deficient PNH clones and clones with loss of heterozygosity in 6p.
Other haematopoietic clones bear somatic mutations in genes
commonly associated with myelodysplastic syndrome (MDS) and
acute myeloid leukaemia (AML). These typically increase with
age and can be found in up to one third of adult patients with
Bone marrow aspirate
Cytogenetics/FISH AA.20 Favourable prognosis has been associated with more stable
chromosomal abnormalities clones with somatic mutations in PIGA (PNH clones), BCOR
Molecular studies
IBMFS associated mutations
and BCORL1. Other mutations such as DNMT3A and ASXL1 are
MDS/AML associated mutations associated with faster progression to AML and unfavourable
prognosis.21
Peripheral blood
UE+C, LFT Evidence of clonal expansion is reflected in cytogenetic abnor-
CRP, ESR
ANA, anti-ds DNA, RF, C3, C4
malities, present in up to 11% of patients with AA at the time of
CT chest IgG, IgA, IgM diagnosis. Some of these are typically associated with MDS/AML
CT/US abdomen TFT
lymphoproliferative Hepatitis, HIV, EBV, CMV, Parvovirus B19 and bear poor prognosis, such as −7/del(7q) and −5/del(5q);
disease LDH, Uric acid
Ferritin others are more benign and might be amenable to immunosup-
ECG
ECHO
Vitamin B12, folic acid, Vitamin D pressive therapy (IST) (see below), such as +Y and +8.22
Flow cytometry
constitutional disease
detection of PNH clones MDS among children and adolescents is very rare, with an
lymphocyte subsets annual incidence of 1–4 cases/million. The most common type of
Chromosomal fragility test
Fanconi anaemia paediatric MDS, refractory cytopenia of childhood (RCC), pre-
Telomere length studies sents with thrombocytopenia and/or neutropenia, and hypoplas-
Dyskeratosis congenita
tic bone marrow and as such needs to be considered in the
Immune aplastic anaemia differential diagnosis of patients with AA.23 Contrary to iAA,
Diagnosis of exclusion
HLA typing
RCC is typically associated with elevated mean corpuscular vol-
ume, moderately increased haemoglobin F and evidence of line-
Fig 1 Investigations included in the diagnostic algorithm of a patients age dysplasia, such as presence of micromegakaryocytes in the
with suspected aplastic anaemia. marrow.24 Most paediatric patients with RCC do not have associ-
ated chromosomal abnormalities with the exception of mono-
somy 7 found in about 11% of those who frequently present
with normocellular or hypercellular marrow.25
Some primary immunodeficiencies, such as cytotoxic T-lymphocyte
associated antigen (CTLA-4) and deficiency of adenosine deaminase
2 (DADA2) deficiencies, have been associated with the development of Diagnosis and Clinical Presentation
iAA as part of complex immune dysregulation comprising of autoim-
Patients typically present with non-specific symptoms resulting
munity and persistent infections with unusual organisms.15,16 GATA2
from associated cytopenia such as low energy levels, pallor and
deficiency, characterised by defective haematopoietic cell differentia-
headaches with anaemia; mucosal bleeding, bruising/petechiae,
tion and self-renewal, is associated with AA as well as clonal evolution
menorrhagia with thrombocytopenia and fever with or without
to myeloid malignancies alongside an increased susceptibility to
evidence of an infection with neutropenia (Fig. 1).
mycobacteria and papilloma viruses.17 Up to 70% of patients with
Detailed history and clinical examination are particularly perti-
GATA2 deficiency are found to have monosomy 7, a poor prognostic
nent in establishing a possible environmental or genetic cause.
marker associated with aggressive myeloid malignancies.18
Family history of unexplained foetal deaths, poor growth, develop-
mental delay and phenotypic abnormalities (i.e. absent thumbs,
Bone marrow failure related abnormal nails) should alert the clinician to the possibility of an
IBMFS.
Inherited bone marrow failure syndromes Bone marrow examination, including both the aspirate and
Inherited bone marrow failure syndromes (IBMFS) are a group of trephine, is mandatory for establishing the diagnosis. A finding of
inherited conditions that, similarly to the earlier described GATA2 hypoplastic or aplastic marrow supports the diagnosis of AA but
deficiency, are associated with an increased risk of progressing to might be difficult to distinguish from IBMFS or hypoplastic MDS.
AA and clonal myeloid dysplasia. Examples of such conditions are Cytogenetic studies including fluorescence in situ hybridisation
Fanconi anaemia and dyskeratosis congenita, resulting from loss- (FISH) can assist in elucidating the diagnosis. Next-generation
of-function germline mutations affecting the ability to repair DNA sequencing (NGS) panels have become increasingly important in
and telomeres, respectively.19 These congenital syndromes are establishing the diagnosis and are available in most tertiary
typically associated with abnormal physical features making them centres.

Journal of Paediatrics and Child Health (2020) 3


© 2020 Paediatrics and Child Health Division (The Royal Australasian College of Physicians)
Aplastic anaemia E Furlong and T Carter

Peripheral blood investigations aimed at excluding alternative Immunosuppressive therapy


causes of cytopenia such as viral infection, auto-immune disease,
vitamin deficiencies and malignant disease should be performed. In the absence of an MSD or alternatively a promptly identified
Lymphocyte subsets and immunoglobulin levels can be abnormal and available MUD, IST with horse anti-thymocyte globuline and
in the setting of a primary immunodeficiency. PNH clones can be cyclosporine A remains the first-line choice of treatment for iAA.
easily detected by flow cytometry. Increased chromosomal break- The long-term survival rate in children who respond to IST is
age in patients with Fanconi anaemia can be detected by per- comparable to those who have received an up-front MSD
forming the diepoxybutane test. Telomere length studies are HSCT.36 The overall response rate at 6 months post IST is 74%.
available in a limited number of centres for the detection of short Relapse occurs in approximately 30% of patients but usually
telomeres, a hallmark of dyskeratosis congenita. responds to further immunosuppression, although not sustain-
able long term.37 Patients who have failed a first course of IST
should proceed to a MUD or umbilical cord transplantation when
available.
Treatment Interestingly, in the absence of a suitable HSCT donor, IST has
Treatment strategies for iAA, the most represented type of AA, been used in the treatment of hypocellular RCC with a similar
are described. It is beyond the scope of this review to describe rate of response at 6 months but a higher relapse rate of nearly
treatment of IBMFS-associated AA and myelodysplasia. Gener- 50%.38 This is in keeping with previous findings suggestive of an
ally, allogeneic haematopoietic stem cell transplantation (HSCT) overlap between iAA and RCC.39,40
is the only curative option for patients with an IBMFS-associated Many efforts have been made to improve the efficacy of the
AA and patients with MDS. anti-thymocyte globuline based immunosuppressive regimen.
Severe iAA (sAA) requires prompt treatment aiming at re- The addition of granulocyte-colony stimulating factor failed to
establishing bone marrow function. Patients with non-sAA improve overall response rates or long-term outcomes and so
require regular and long-term follow-up as up to two thirds of have agents such as mycophenolate mofetil or sirolimus.41
them will progress to sAA.26 There are two main ways of Contrary to that, a synthetic analogue of thrombopoietin,
treating iAA. eltrombopag, has been shown to significantly improve recov-
ery and response rates, and has now become an IST additional
therapeutic agent.42

Haematopoietic stem cell transplantation


Supportive Care
HSCT is the preferred and curative treatment option for patients
with iAA considering late clonal disorders such as MDS and AML Transfusions
occurring in up to 15% of patients initially treated with IST.27
Excellent survival rates of more than 90% in young children and Most patients with sAA require regular red blood cell and platelet
more than 80% in adolescents have been achieved with HLA- transfusions. Transfusion thresholds are usually set by institu-
matched sibling donor (MSD) HSCTs, involving a low rate of tional guidelines and guided by international policies. Special
short- and long-term complications such as graft-versus-host dis- attention should be paid to patients awaiting HSCT where
ease (GVHD).28,29 repeated transfusions can lead to allo-immunisation and an
Early identification of a fully HLA-matched unrelated donor increased risk of graft failure.43 The use of leukocyte-depleted
(MUD) using international registries might prompt an up-front blood products has significantly decreased, although not
MUD HSCT, especially in paediatric patients where comparable completely eliminated this risk.44 HLA-antibodies should be mea-
result to MSD HSCT have been achieved, including a low inci- sured in case of inadequate increments post platelet transfusions
dence of GVHD.30,31 and HLA-matched single donor apheresis concentrates should be
Bone marrow is the preferred source of progenitor cells for used if HLA-antibodies have been detected.45 All blood products
both the MSD and MUD HSCT over peripheral blood stem cells for patients with AA should be irradiated to reduce the risk of
due to considerably lower occurrence of acute and chronic transfusion-associated GVHD and allo-sensitisation. Generally,
GVHD.32 Umbilical cord transplantation has been successfully there is no need for cytomegalovirus (CMV)-negative products as
used in the treatment of iAA, particularly in children due to the leukocyte-depleted blood products are considered to be suitable
lower absolute progenitor cell count required relative to weight, for haematological patients including patients undergoing HSCT.
with survival rates averaging 90%.33 The major disadvantage has Exceptions are CMV-negative patients undergoing HSCT with a
been delayed engraftment and prolonged neutropenia, increasing CMV-negative donor, where most institutions choose to use
the risk of severe and potentially fatal infections. CMV-negative blood products.46
Recent advances in transplant conditioning and donor cell
manipulations using T-cell depletion strategies have made half-
Prevention and treatment of infections
matched (haploidentical) donors a potential source for bone mar-
row transplantation (HAPLO HSCT). Although the survival rates Patients with sAA are at high risk of developing invasive fungal
are encouraging, the rate of GVHD and possible late effects infections due to prolonged periods of severe neutropenia.47 Pro-
remain high.34,35 HAPLO HSCT should only be considered for phylactic anti-fungals should be used in all patients with sAA,
patients who failed at least one course of IST and lack suitable posaconazole being considered more effective than fluconazole
matched unrelated or cord blood donors. for its extended mould species cover.48 Febrile neutropenia is a

4 Journal of Paediatrics and Child Health (2020)


© 2020 Paediatrics and Child Health Division (The Royal Australasian College of Physicians)
E Furlong and T Carter Aplastic anaemia

haematological emergency and is managed as per institutional well-established clinical trials and centralisation of data in
guidelines with broad spectrum antibiotics. Prolonged fever and national and international registries.
neutropenia should prompt investigations for a suspected fungal
infection such as galactomannan testing and computed tomogra- Acknowledgements
phy scans, as well as broadening anti-fungal agents.
Although using myeloid growth factors has proven to be The authors express their gratitude to Professor Catherine Cole
ineffective in the overall treatment of AA, granulocyte-colony for her long-standing dedication to patients with AA and shared
stimulating factor is often used during episodes of prolonged knowledge; and to Dr Shanti Ramachandran who kindly
neutropenia and fever where a fungal infection is suspected reviewed the summary of latest approach to AA and the role of
as it may temporarily increase the neutrophil count. In life- haematopoietic stem cell transplantation.
threatening infections, the use of granulocyte transfusions
may be considered as an adjunctive therapy taking in consid-
eration limited evidence and potentially significant side References
effects.49
1 Camitta BM, Rappeport JM, Parkman R, Nathan DG. Selection of
There are no general recommendations for prophylaxis against
patients for bone marrow transplantation in severe aplastic anemia.
Pneumocystis jirovecii. Most centres use P. jirovecii prophylaxis post Blood 1975; 45: 355–63.
IST as well as routinely in patients undergoing HSCT. Similarly, 2 Young NS, Kaufman DW. The epidemiology of acquired aplastic ane-
many centres choose to use antiviral prophylaxis against CMV mia. Haematologica 2008; 93: 489–92.
and Epstein-Barr virus in patients post IST as per standard HSCT 3 Saracco P, Quarello P, Iori AP et al. Cyclosporin A response and
guidelines.50 dependence in children with acquired aplastic anaemia: A multicentre
retrospective study with long-term observation follow-up. Br.
J. Haematol. 2008; 140: 197–205.
Psycho-social support 4 Wallerstein RO, Condit PK, Kasper CK, Brown JW, Morrison FR. State-
wide study of chloramphenicol therapy and fatal aplastic anemia.
Despite significant advances in the understanding and manage-
JAMA 1969; 208: 2045–50.
ment of AA, the course of the condition remains difficult. Fre-
5 Kay AG. Myelotoxicity of gold. Br. Med. J. 1976; 1: 1266–8.
quent transfusions and hospital checks are required during the 6 Handoko KB, Souverein PC, van Staa TP et al. Risk of aplastic anemia
time of establishing diagnosis and searching for suitable HSCT in patients using antiepileptic drugs. Epilepsia 2006; 47: 1232–6.
donors as well as in recovery phase. Patients undergoing HSCT 7 Locasciulli A, Bacigalupo A, Bruno B et al. Hepatitis-associated
might develop GVHD or other associated complications. Patients aplastic anaemia: Epidemiology and treatment results obtained in
post IST are at risk of not responding to the treatment or later Europe. A report of the EBMT aplastic anaemia working party. Br.
relapsing. Prolonged periods of neutropenia put patients at risk of J. Haematol. 2010; 149: 890–5.
severe fungal infections requiring lengthy hospitalisations and 8 Ikawa Y, Nishimura R, Kuroda R et al. Expansion of a liver-infiltrating
cytotoxic T-lymphocyte clone in concert with the development of
further provoking anxiety about the outcome.
hepatitis-associated aplastic anaemia. Br. J. Haematol. 2013; 161:
Ongoing psycho-social support for patients with AA and their
599–602.
families is an indispensable part of the disease management
9 Scheinberg P, Chen J. Aplastic anemia: What have we learned from ani-
within a multidisciplinary specialist centre facilitating communi- mal models and from the clinic. Semin. Hematol. 2013; 50: 156–64.
cation, reassurance and comfort during the course of the disease 10 Young NS, Calado RT, Scheinberg P. Current concepts in the pathophysi-
and long-term follow-up. ology and treatment of aplastic anemia. Blood 2006; 108: 2509–19.
11 Kordasti S, Costantini B, Seidl T et al. Deep phenotyping of Tregs iden-
tifies an immune signature for idiopathic aplastic anemia and predicts
Summary response to treatment. Blood 2016; 128: 1193–205.
Recent advances in understanding the pathophysiology and 12 Deng XZ, Du M, Peng J et al. Associations between the HLA-A/B/DRB1
polymorphisms and aplastic anemia: Evidence from 17 case-control
improved treatment strategies including HSCT have contributed
studies. Hematology 2018; 23: 154–62.
to a significantly increased survival of patients with AA, once an
13 Imi T, Katagiri T, Hosomichi K et al. Sustained clonal hematopoiesis by
inevitably fatal disease. Despite the progress, the medical and HLA-lacking hematopoietic stem cells without driver mutations in
psycho-social burden for patients with AA, their families and the aplastic anemia. Blood Adv. 2018; 2: 1000–12.
health system remain high due to the nature of the disease asso- 14 Luzzatto L, Notaro R. The ‘escape’ model: A versatile mechanism for
ciated with prolonged periods of hospitalisation, requirement for clonal expansion. Br. J. Haematol. 2019; 184: 465–6.
ongoing supportive care and the possibility of treatment failure. 15 Kuehn HS, Ouyang W, Lo B et al. Immune dysregulation in human
Modern molecular and genetic methods including next- subjects with heterozygous germline mutations in CTLA4. Science
generation sequencing have undoubtedly improved our under- 2014; 345: 1623–7.
standing of this heterogeneous group of conditions, and become a 16 Michniacki TF, Hannibal M, Ross CW et al. Hematologic manifestations
of deficiency of adenosine deaminase 2 (DADA2) and response to
powerful tool in establishing the diagnosis. Increased identification
tumor necrosis factor inhibition in DADA2-associated bone marrow
of rare genetic mutations in patients with AA has brought a new
failure. J. Clin. Immunol. 2018; 38: 166–73.
dimension to the complex decision-making regarding prognosis and
17 Wlodarski MW, Collin M, Horwitz MS. GATA2 deficiency and related
appropriate treatment for individual patients supported by limited myeloid neoplasms. Semin. Hematol. 2017; 54: 81–6.
long-term experience of these very rare disorders. 18 Wlodarski MW, Hirabayashi S, Pastor V et al. Prevalence, clinical charac-
The rarity and heterogeneity of AA highlights the necessity teristics, and prognosis of GATA2-related myelodysplastic syndromes in
of multidisciplinary approach supported by ongoing research, children and adolescents. Blood 2016; 127: 1387–97.

Journal of Paediatrics and Child Health (2020) 5


© 2020 Paediatrics and Child Health Division (The Royal Australasian College of Physicians)
Aplastic anaemia E Furlong and T Carter

19 Savage SA, Dufour C. Classical inherited bone marrow failure syn- European society for blood and marrow transplantation. Biol. Blood
dromes with high risk for myelodysplastic syndrome and acute mye- Marrow Transplant. 2017; 23: 1939–48.
logenous leukemia. Semin. Hematol. 2017; 54: 105–14. 34 Clay J, Kulasekararaj AG, Potter V et al. Nonmyeloablative peripheral
20 Yoshizato T, Dumitriu B, Hosokawa K et al. Somatic mutations and blood haploidentical stem cell transplantation for refractory severe
clonal hematopoiesis in aplastic anemia. N. Engl. J. Med. 2015; 373: aplastic anemia. Biol. Blood Marrow Transplant. 2014; 20: 1711–6.
35–47. 35 Esteves I, Bonfim C, Pasquini R et al. Haploidentical BMT and post-
21 Ogawa S. Clonal hematopoiesis in acquired aplastic anemia. Blood transplant Cy for severe aplastic anemia: A multicenter retrospective
2016; 128: 337–47. study. Bone Marrow Transplant. 2015; 50: 685–9.
22 Maciejewski JP, Risitano A, Sloand EM, Nunez O, Young NS. Distinct 36 Bacigalupo A. How I treat acquired aplastic anemia. Blood 2017; 129:
clinical outcomes for cytogenetic abnormalities evolving from aplastic 1428–36.
anemia. Blood 2002; 99: 3129–35. 37 Scheinberg P, Wu CO, Nunez O, Young NS. Long-term outcome of
23 Kardos G, Baumann I, Passmore SJ et al. Refractory anemia in child- pediatric patients with severe aplastic anemia treated with ant-
hood: A retrospective analysis of 67 patients with particular refer- ithymocyte globulin and cyclosporine. J. Pediatr. 2008; 153: 814–9.
ence to monosomy 7. Blood 2003; 102: 1997–2003. 38 Locatelli F, Strahm B. How I treat myelodysplastic syndromes of child-
24 Baumann I, Fuhrer M, Behrendt S et al. Morphological differentiation hood. Blood 2018; 131: 1406–14.
of severe aplastic anaemia from hypocellular refractory cytopenia of 39 de Vries AC, Langerak AW, Verhaaf B et al. T-cell receptor Vbeta
childhood: Reproducibility of histopathological diagnostic criteria. His- CDR3 oligoclonality frequently occurs in childhood refractory
topathology 2012; 61: 10–7. cytopenia (MDS-RC) and severe aplastic anemia. Leukemia 2008; 22:
25 Niemeyer CM, Baumann I. Classification of childhood aplastic anemia 1170–4.
and myelodysplastic syndrome. Hematology Am. Soc. Hematol. Educ. 40 Sloand EM, Wu CO, Greenberg P, Young N, Barrett J. Factors affecting
Program 2011; 2011: 84–9. response and survival in patients with myelodysplasia treated with
26 Howard SC, Naidu PE, Hu XJ et al. Natural history of moderate immunosuppressive therapy. J. Clin. Oncol. 2008; 26: 2505–11.
aplastic anemia in children. Pediatr. Blood Cancer 2004; 43: 545–51. 41 Scheinberg P, Young NS. How I treat acquired aplastic anemia. Blood
27 Tichelli A, Schrezenmeier H, Socie G et al. A randomized controlled 2012; 120: 1185–96.
study in patients with newly diagnosed severe aplastic anemia receiv- 42 Townsley DM, Scheinberg P, Winkler T et al. Eltrombopag added to
ing antithymocyte globulin (ATG), cyclosporine, with or without G- standard immunosuppression for aplastic anemia. N. Engl. J. Med.
CSF: A study of the SAA working party of the European group for 2017; 376: 1540–50.
blood and marrow transplantation. Blood 2011; 117: 4434–41. 43 Champlin RE, Horowitz MM, van Bekkum DW et al. Graft failure follow-
28 Dufour C, Pillon M, Socie G et al. Outcome of aplastic anaemia in chil- ing bone marrow transplantation for severe aplastic anemia: Risk fac-
dren. A study by the severe aplastic anaemia and paediatric disease tors and treatment results. Blood 1989; 73: 606–13.
working parties of the European group blood and bone marrow 44 Desmarets M, Cadwell CM, Peterson KR, Neades R, Zimring JC. Minor
transplant. Br. J. Haematol. 2015; 169: 565–73. histocompatibility antigens on transfused leukoreduced units of red
29 Dufour C, Pillon M, Passweg J et al. Outcome of aplastic anemia in blood cells induce bone marrow transplant rejection in a mouse
adolescence: A survey of the severe aplastic anemia working party of model. Blood 2009; 114: 2315–22.
the European group for blood and marrow transplantation. 45 Laundy GJ, Bradley BA, Rees BM, Younie M, Hows JM. Incidence and
Haematologica 2014; 99: 1574–81. specificity of HLA antibodies in multitransfused patients with acquired
30 Dufour C, Veys P, Carraro E et al. Similar outcome of upfront-unrelated aplastic anemia. Transfusion 2004; 44: 814–25.
and matched sibling stem cell transplantation in idiopathic paediatric 46 Australian and New Zealand Society of Blood Transfusion. Guidelines
aplastic anaemia. A study on behalf of the UK paediatric BMT working for Transfusion and Immunohaematology Laboratory Practice. Syd-
party, paediatric diseases working party and severe aplastic anaemia ney: The Society; 2016.
working party of EBMT. Br. J. Haematol. 2015; 171: 585–94. 47 Valdez JM, Scheinberg P, Young NS, Walsh TJ. Infections in patients
31 Samarasinghe S, Veys P, Vora A, Wynn R. Paediatric amendment to adult with aplastic anemia. Semin. Hematol. 2009; 46: 269–76.
BSH guidelines for aplastic anaemia. Br. J. Haematol. 2018; 180: 201–5. 48 Fleming S, Yannakou CK, Haeusler GM et al. Consensus guidelines for
32 Schrezenmeier H, Passweg JR, Marsh JC et al. Worse outcome and antifungal prophylaxis in haematological malignancy and haemopoietic
more chronic GVHD with peripheral blood progenitor cells than bone stem cell transplantation, 2014. Intern. Med. J. 2014; 44: 1283–97.
marrow in HLA-matched sibling donor transplants for young patients 49 Quillen K, Wong E, Scheinberg P et al. Granulocyte transfusions in
with severe acquired aplastic anemia. Blood 2007; 110: 1397–400. severe aplastic anemia: An eleven-year experience. Haematologica
33 Pagliuca S, Peffault de Latour R, Volt F et al. Long-term outcomes of 2009; 94: 1661–8.
cord blood transplantation from an HLA-identical sibling for patients 50 Höchsmann B, Moicean A, Risitano A, Ljungman P, Schrezenmeier H.
with bone marrow failure syndromes: A report from Eurocord, cord Supportive care in severe and very severe aplastic anemia. Bone Mar-
blood committee and severe aplastic anemia working party of the row Transplant. 2012; 48: 168.

6 Journal of Paediatrics and Child Health (2020)


© 2020 Paediatrics and Child Health Division (The Royal Australasian College of Physicians)

You might also like