You are on page 1of 22

463

CHAPTER 34 ACQUIRED APLASTIC ANEMIA


DEFINITION AND HISTORY
APLASTIC ANEMIA: Aplastic anemia is a clinical syndrome that results from a marked diminu-
tion of marrow blood cell production. The latter results in reticulocytope-
ACQUIRED AND nia, anemia, granulocytopenia, monocytopenia, and thrombocytopenia.
The diagnosis usually requires the presence of pancytopenia with a neutro-
INHERITED phil count fewer than 1500/L (1.5 109/L), a platelet count fewer than
50,000/L (50 109/L), a hemoglobin concentration less than 10 g/dL (100
g/L), and an absolute reticulocyte count fewer than 40,000/L (40 109/L),
George B. Segel and Marshall A. Lichtman accompanied by a hypocellular marrow without abnormal or malignant
cells or fibrosis.1 For the purpose of therapeutic decision making, compara-
tive clinical trials, and international sharing of data, the disease has been
stratified into moderately severe, severe, and very severe acquired aplastic
S U M M AR Y anemia based on the blood counts (especially the neutrophil count) and
Acquired aplastic anemia is a clinical syndrome in which there is a deficiency the degree of marrow hypocellularity (Table 341). Most cases of aplastic
of red cells, neutrophils, monocytes, and platelets in the blood, and fatty anemia are acquired; fewer cases are the result of an inherited disorder,
replacement of the marrow with a near absence of hematopoietic precursor such as Fanconi anemia, Shwachman-Diamond syndrome, and others (see
cells. Reticulocytopenia, neutropenia, monocytopenia, and thrombocytopenia, Hereditary Aplastic Anemia below).
when severe, are life-threatening because of the risk of infection and bleeding, Aplastic anemia was first recognized by Ehrlich in 1888.2 He
complicated by severe anemia. Most cases occur without an evident precipitating described a young, pregnant woman who died of severe anemia and
cause and are the result of the expression of autoreactive cytotoxic T lympho- neutropenia. Autopsy examination revealed a fatty marrow with essen-
cytes that suppress or destroy primitive CD34+ multipotential hematopoietic tially no hematopoiesis. The name aplastic anemia was subsequently
cells. The disorder also can occur after (1) prolonged high-dose exposure to applied to this disease by Chauffard, a French hematologist, in 1904,3
certain toxic chemicals (e.g., benzene), (2) after specific viral infections (e.g., and although an anachronistic term because the morbidity is the result
Epstein-Barr virus), (3) as an idiosyncratic response to certain pharmaceuticals of pancytopenia, especially neutropenia and thrombocytopenia, the
(e.g., ticlopidine, chloramphenicol), (4) as a feature of a connective tissue or designation is entrenched in medical usage. For the next 40 years, many
autoimmune disorder (e.g., lupus erythematosus), or, (5) rarely, in association conditions that caused pancytopenia were confused with aplastic ane-
with pregnancy. The final common pathway may be through cytotoxic T-cell mia based on incomplete or inadequate histologic study of the patients
autoreactivity, whether idiopathic or associated with an inciting agent since marrow.4 The development of improved instruments for percutaneous
they all respond in a similar fashion to immunosuppressive therapy. The differ- marrow biopsy in the last half of the 20th century improved diagnostic
ential diagnosis of acquired aplastic anemia includes the hypoplastic marrow precision. In 1972, Thomas and his colleagues established that marrow
that can accompany paroxysmal nocturnal hemoglobinuria or hypoplastic oli- transplantation from a histocompatible sibling could cure the disease.5
goblastic (myelodysplastic syndrome) or polyblastic myelogenous leukemia. The disease initially was thought to result from an atrophy or chemical
Allogeneic hematopoietic stem cell transplantation is curative in approximately injury of primitive marrow hematopoietic cells. The unexpected recov-
80 percent of younger patients with high-resolution human leukocyte antigen ery of marrow recipients who were given immunosuppressive condi-
(HLA)-matched sibling donors although the posttransplant period may be tioning therapy but who did not engraft with donor stem cells raised
severely complicated by graft-versus-host disease. The disease may be signifi- the possibility that the disease may not be intrinsic to primitive hema-
cantly ameliorated or occasionally cured by immunotherapy, especially a regi- topoietic cells but the result of a suppression of hematopoietic cells by
men coupling antithymocyte globulin with cyclosporine. However, after immune cells, notably T lymphocytes.6 The requirement to treat the
successful treatment with immunosuppressive agents, the disease may relapse recipient of a marrow transplant from an identical twin with immuno-
or evolve into a clonal myeloid disorder, such as paroxysmal nocturnal hemo- suppressive conditioning therapy for optimal results of transplant, but-
globinuria, a clonal cytopenia, or oligoblastic or polyblastic myelogenous leu- tressed this suspicion.7 This supposition was confirmed by a clinical
kemia. Several uncommon inherited disorders, including Fanconi anemia, trial that established antilymphocyte globulin (ALG) capable of amelio-
Shwachman-Diamond syndrome, dyskeratosis congenita and others have as a rating the disease in the majority of patients.8 Since that time, compel-
primary manifestation aplastic hematopoiesis. ling evidence for a cellular autoimmune mechanism has accumulated
(see the main section Etiology and Pathogenesis below).

Acronyms and abbreviations that appear in this chapter include: A, adenine; EPIDEMIOLOGY
ALG, antilymphocyte globulin; ALL, acute lymphocytic leukemia; AML, The International Aplastic Anemia and Agranulocytosis Study and a
acute myelogenous leukemia; ATG, antithymocyte globulin; ATR, ataxia- French study found the incidence rate of acquired aplastic anemia to be
telangiectasia mutated and rad3-related kinase; BFUE, erythroid burst-forming about 2 per 1,000,000 persons per year.1,9 This approximate annual
units; CD, cluster of differentiation; CFU-GM, colony-forming unitgranulocyte- incidence rate has been confirmed in studies in Spain (Barcelona),10
macrophage; CMV, cytomegalovirus; EBV, Epstein-Barr virus; G, guanine; G-CSF, Brazil (State of Parana),11 and Canada (British Columbia).12 The high-
granulocyte colony-stimulating factor; HHV, human herpes virus; HIV, human est frequency of aplastic anemia occurs in persons between the ages of
immunodeficiency virus; HLA, human leukocyte antigen; IL, interleukin; LDH, 15 and 25 years; a second peak occurs between the ages of 65 and 69.1
lactic dehydrogenase; NMRI, nuclear magnetic resonance imaging; PCP, Aplastic anemia is more prevalent in the Far East where the incidence is
pentachlorophenol; PNH, paroxysmal nocturnal hemoglobinuria; SCF, stem approximately 7 per 1,000,000 in parts of China,13 approximately 4 per
cell factor; T, thymine; TNF, tumor necrosis factor; TNT, trinitrotoluene; TPO, 1,000,000 in sections of Thailand,14 approximately 5 per 1,000,000 in
thrombopoietin. areas of Malaysia,15 and approximately 7 per 1,000,000 among children
of Asian descent living in a province of Canada.12 The explanation for a
464 PART VI: The Erythrocyte

TABLE 341. Degree of Severity of Acquired Aplastic Anemia


Diagnostic Reticulocyte Neutrophil Platelet
Categories Hemoglobin Concentration Count Count Marrow Biopsy Comments
Moderately severe <100 g/L <40 109/L <1.5 109/L <50 109/L Marked decrease of hemato- At the time of diagnosis at least 2 of 3
poietic cells. blood counts should meet these criteria.
Severe <90 g/L <30.0 109/L <0.5 109/L <30.0 109/L Marked decrease or absence Search for a histocompatible sibling
of hematopoietic cells. should be made if age permits.
Very Severe <80 g/L <20.0 109/L <0.2 109/L <20.0 109/L Marked decrease or absence Search for a histocompatible sibling
of hematopoietic cells. should be made if age permits.

NOTE: These values are approximations and must be considered in the context of an individual patients situation. (In some clinical trials, the blood count thresholds for moderately severe aplas-
tic anemia are higher, e.g., platelet count <100 109/L and absolute reticulocyte count <60,000 109/L.) The marrow biopsy may contain the usual number of lymphocytes and plasma cells;
hot spots, focal areas of erythroid cells, may be seen. No fibrosis, abnormal cells, or malignant cells should be evident in the marrow. Dysmorphic features of blood or marrow cells are not
features of acquired aplastic anemia. Ethnic differences in the lower limit of the absolute neutrophil count should be considered. (See Chap. 65.)

twofold or greater incidence in the Orient compared to the Occident very early CD34+ hematopoietic multipotential progenitor or stem
may be multifactorial,16 but a predisposition gene or genes is a likely cells.24 A small fraction of cases is initiated by a toxic exposure, drug
component.12,17 Studies have not established the use of chlorampheni- exposure, or viral infection, but in these cases the pathogenesis also
col in Asia as a cause. Poorly regulated exposure of workers to benzene
is a factor,18 but the attributable risk from benzene and other toxic
exposures does not explain the magnitude of the difference in the inci-
dence in Asia compared to that in Europe and South America.16,17 A
relationship to impure water use in Thailand has led to speculation of
TABLE 342. Etiologic Classification of Aplastic Anemia
an infectious etiology, although no agent, including seronegative hepa-
titis, a known association with the onset of acquired aplastic anemia,16 Acquired
has been identified. Seronegative viral hepatitis is a forerunner of Autoimmune
approximately 7 percent of cases of acquired aplastic anemia.17 The Drugs
male-to-female incidence ratio of aplastic anemia in most studies is See Table 343
approximately one.17
Toxins
Benzene
ETIOLOGY AND PATHOGENESIS
Chlorinated hydrocarbons
Table 342 lists the potential causes for aplastic anemia.
Organophosphates
The final common pathway to the clinical disease is a decrease in
blood cell formation in the marrow. The number of marrow CD34+ Viruses
cells (multipotential hematopoietic progenitors) and their derivative Epstein-Barr virus
colony-forming unitgranulocyte-macrophage (CFU-GM) and burst- Non-A, -B, -C, -D, -E, or -G hepatitis virus
forming uniterythroid (BFUE) are reduced markedly in patients Human immunodeficiency virus (HIV)
with aplastic anemia.1922 Long-term culture-initiating cells, an in vitro
Paroxysmal nocturnal hemoglobinuria
surrogate assay for hematopoietic stem cells, also are reduced to
approximately 1 percent of normal values.22 Potential mechanisms Autoimmune/connective tissue disorders
responsible for acquired marrow cell failure include (1) direct toxicity Eosinophilic fasciitis
to hematopoietic multipotential cells, (2) a defect in the stromal Immune thyroid disease (Graves disease, Hashimoto thyroiditis)
microenvironment of the marrow required for hematopoietic cell Rheumatoid arthritis
development, (3) impaired production or release of essential multilin-
Systemic lupus erythematosus
eage hematopoietic growth factors, (4) cellular or humoral immune
suppression of the marrow multipotential cells, and (5) progressive ero- Thymoma
sion of chromosome telomeres. There is little experimental evidence Pregnancy
for a stromal microenvironmental defect or a deficit of critical hemato- Iatrogenic
poietic growth factors, and the role of telomerase mutations with con- Radiation
sequent telomere shortening is unclear, although present in as much as Cytotoxic drug therapy
40 percent of patients.23 Deficiencies in telomere repair could predis-
Hereditary
pose to aplastic anemia by affecting the size of the multipotential hema-
topoietic cell compartment and by decreasing the multipotential cells Fanconi anemia
response to a marrow injury, and could play a role in the evolution of Dyskeratosis congenita
aplastic anemia to a clonal myeloid disease by contributing to genome Shwachman-Diamond syndrome
instability.23 Thus, reduced hematopoiesis in most cases of aplastic ane- Other rare syndromes (see Table 348)
mia results from cytotoxic T-cell-mediated immune suppression of
CHAPTER 34: Aplastic Anemia: Acquired and Inherited 465

FIGURE 341. Immune pathogenesis of apoptosis of CD34 multipotential hematopoietic cells in acquired aplastic anemia. Antigens are presented to T lymphoc ytes by antigen-
presenting cells (APCs). This triggers T cells to activate and proliferate. T-bet, a transcription factor, binds to the interferon- (IFN-) promoter region and induces gene expression.
SLAM-associated protein (SAP) binds to Fyn and modulates the signaling lymphocyte activation molecule (SLAM) activity on IFN- expression, diminishing gene transcription. Pa-
tients with aplastic anemia show constitutive T-bet expression and low SAP levels. IFN- and tumor necrosis factor- (TNF-) upregulate both the T cells cellular receptors and the
Fas receptor. Increased production of interleukin-2 leads to polyclonal expansion of T cells. Activation of the Fas receptor by the Fas ligand leads to apoptosis of target cells. Some ef-
fects of IFN- are mediated through interferon regulatory factor 1 (IRF-1), which inhibits the transcription of cellular genes and entry into the cell cycle. IFN- is a potent inducer of
many cellular genes, including inducible nitric oxide synthase (NOS), and production of nitric oxide (NO) may diffuse its cytotoxic effects. These events ultimately lead to reduced
cell cycling and cell death by apoptosis. (Reproduced from Young NS, Calado RT, Scheinberg P: Current concepts in the pathophysiology and treatment of aplastic anemia. Blood
108:2509, 2006, with permission of The American Society of Hematology.)

may relate to autoimmunity as there is evidence of immune dysfunc- gerated amounts of interferon-, markedly reduced the frequency of
tion in seronegative hepatitis, after benzene exposure, and many such long-term culture-initiating cells.24 These observations indicate that
patients respond to antiT-cell therapy.24 acquired aplastic anemia is the result of cellular immune-induced
apoptosis of primitive CD34+ multipotential hematopoietic progeni-
Autoreactive Cytotoxic T Lymphocytes tors, mediated by cytotoxic T lymphocytes, in part, through the expres-
In vitro and clinical observations have resulted in the identification of a sion of T-helper type 1 (Th1) inhibitory cytokines, interferon-, and
cytotoxic T-cell-mediated attack on multipotential hematopoietic cells TNF- (Fig. 341).32 The secretion of interferon- is a result of the
in the CD34+ cellular compartment as the basis for acquired aplastic upregulation of transcription regulatory factor T-bet,33 and apoptosis
anemia.25 Cellular immune injury to the marrow after drug-, viral-, or of CD34+ cells is, in part, mediated through a FAS-dependent path-
toxin-initiated marrow aplasia could result from the induction of way.24 Because HLA-DR2 is more prevalent in patients with aplastic
neoantigens that provoke a secondary T-cell-mediated attack on hema- anemia, antigen recognition may be a factor in those patients. A variety
topoietic cells. This mechanism could explain the response to immuno- of other potential factors have been found in some patients, including
suppressive treatment in cases that follow exposure to an exogenous nucleotide polymorphisms in cytokine genes, overexpression of per-
agent. Spontaneous or mitogen-induced increases in mononuclear cell forin in marrow cells, and decreased expression of SAP, a modulator
production of interferon-,26,27 interleukin (IL)-2,27 and tumor necrosis protein that inhibits interferon- secretion.24
factor- (TNF-)28,29 occur. Elevated serum levels of interferon- are A decrease in regulatory T cells contributes to the expansion of an
present in 30 percent of patients with aplastic anemia, and interferon- autoreactive CD8+CD28 T-cell population, which induces apoptosis
expression has been detected in the marrow of most patients with of autologous hematopoietic multipotential hematopoietic cells.34 One
acquired aplastic anemia.30 Addition of antibodies to interferon- mouse model of immune-related marrow failure, induced by infusion
enhances in vitro colony growth of marrow cells from affected of parental lymph node cells into F1 hybrid recipients, caused a fatal
patients.31 Long-term marrow cultures manipulated to elaborate exag- aplastic anemia. The aplasia could be prevented by immunotherapy or
466 PART VI: The Erythrocyte

with monoclonal antibodies to interferon- and TNF-.24 Another There appears to be little difference in the age distribution, gender,
mouse model of aplastic anemia induced by the infusion of lymph node response to immunotherapy, marrow transplantation, or survival whether
cells histoincompatible for the minor H antigen, H60, resulted from the or not a drug exposure preceded the onset of the marrow aplasia.
expansion of H60-specific CD8 T cells in recipient mice. The result was
severe marrow aplasia. The effect of the CD8 T cells could be abrogated Toxic Chemicals
by either immunosuppressive agents or administration of CD4+CD25+
Benzene was the first chemical linked to aplastic anemia, based on stud-
regulatory T cells,35 providing additional experimental evidence for the
ies in factory workers before the 20th century.5459 Benzene is used as a
role of regulatory T cells in the prevention of aplastic anemia.
solvent and is employed in the manufacture of chemicals, drugs, dyes,
Several putative target antigens on affected hematopoietic cells have
and explosives. It has been a vital chemical in the manufacture of rubber
been identified. Autoantibodies to one putative antigen, kinectin, have
and leather goods and has been used widely in the shoe industry, leading
been found in patients with aplastic anemia. T cells, responsive to
to an increased risk for aplastic anemia (and acute myelogenous leuke-
kinectin-derived peptides, suppress granulocyte-monocyte colony
mia) in workers exposed to a poorly regulated environment.56 In studies
growth in vitro. However, in these studies cytotoxic T lymphocytes with
in China, aplastic anemia among workers was sixfold higher than in the
that specificity were not isolated from patients.36
general population.18
The U.S. Occupational Safety and Health Administration has low-
Drugs ered the permissible atmospheric exposure limit of benzene to 1 part
Chloramphenicol is the most notorious drug documented to cause per million (ppm). Previous to that regulatory change, the frequency of
aplastic anemia. Although this drug is directly myelosuppressive at very aplastic anemia in workers exposed to greater than 100 ppm benzene
high dose because of its effect on mitochondrial DNA, the occurrence was approximately 1 in 100 workers, which decreased to 1 in 1000
of aplastic anemia appears to be idiosyncratic, perhaps related to an workers at 10 to 20 ppm exposure.55
inherited sensitivity to the nitroso-containing toxic intermediates.37 Organochlorine and organophosphate pesticide compounds have
This sensitivity may produce immunologic marrow suppression, as a been suspected in the onset of aplastic anemia57,58 and several studies
substantial proportion of affected patients respond to treatment with have indicated an increased relative risk, especially for agricultural
immunosuppressive therapy.38 The risk of developing aplastic anemia exposures11,16,59,60 and household11,60 exposures. These relationships
in patients treated with chloramphenicol is approximately 1 in 20,000, are suspect because dosedisease relationships and other important
or 25 times that of the general population.39 Although its use as an anti- factors have not been delineated, and several studies have not found an
biotic has been largely abandoned in industrialized countries, global association with environmental exposures.12,61 DDT (dichlorodiphenyl-
reports of fatal aplastic anemia continue to appear with topical or sys- trichloroethane), lindane, and chlordane are insecticides that have also
temic use of the drug.50,51 been associated with cases of aplastic anemia.16,58 Occasional cases still
Epidemiologic evidence established that quinacrine (Atabrine) occur following heavy exposure at industrial plants or after its use as a
increased the risk of aplastic anemia.40 This drug was administered to pesticide.62 Lindane is metabolized in part to pentachlorophenol
all U.S. troops in the South Pacific and Asiatic theaters of operations as (PCP), another potentially toxic chlorinated hydrocarbon that is manu-
prophylaxis for malaria during 1943 and 1944. The incidence of aplas- factured for use as a wood preservative. Many cases of aplastic anemia
tic anemia was 7 to 28 cases per 1,000,000 personnel per year in the and related blood disorders have been attributed to PCP over the past
prophylaxis zones, whereas untreated soldiers had 1 to 2 cases per 25 years.58,63 Prolonged exposures to petroleum distillates in the form
1,000,000 personnel per year. The aplasia occurred during administra- of Stoddard solvent64 and acute exposure to toluene through the prac-
tion of the offending agent and was preceded by a characteristic rash in tice of glue sniffing65,66 also have been reported to cause marrow apla-
nearly half the cases. Many other drugs have been reported to increase sia. Trinitrotoluene (TNT), an explosive used extensively during World
the risk of aplastic anemia, but owing to incomplete reporting of infor- Wars I and II, is absorbed readily by inhalation and through the skin.67
mation and the infrequency of the association, the spectrum of drug- Fatal cases of aplastic anemia were observed in munitions workers
induced aplastic anemia may not be fully appreciated. Table 343 is a exposed to TNT in Great Britain68 from 1940 to 1946. In most cases,
partial list of drugs that have been implicated.4151 these conclusions have not been derived from specific studies but from
Many of these drugs are known to also induce selective cytopenias, accumulation of case reports or from patient histories, making conclu-
such as agranulocytosis, which usually are reversible after discontinua- sions provisional, although the argument for minimizing exposures to
tion of the offending agent. These reversible reactions are not correlated potential toxins is logical in any case.
with the risk of aplastic anemia, casting doubt on the effectiveness of rou-
tine monitoring of blood counts as a strategy to avoid aplastic anemia. Viruses
Because aplastic anemia is a rare event with drug use, it may occur
because of an underlying metabolic or immunologic predisposition Non-A, -B, -C, -D, -E, -G Hepatitis Virus A relationship between hepa-
(gene polymorphism) in susceptible individuals. In the case of phe- titis and the subsequent development of aplastic anemia has been the
nylbutazone-associated marrow aplasia, there is delayed oxidation and subject of a number of case reports, and this association was empha-
clearance of a related compound, acetanilide, as compared to either sized by two major reviews in the 1970s.69,70 In the aggregate, these
normal controls or those with aplastic anemia from other causes. This reports summarized findings in more than 200 cases. In many
finding suggests excess accumulation of the drug as a potential mecha- instances, the hepatitis was improving or had resolved when the
nism for the aplasia. In some cases, drug interactions or synergy may be aplastic anemia was noted 4 to 12 weeks later. Approximately 10 per-
required to induce marrow aplasia. Cimetidine, a histamine H2-receptor cent of cases occurred more than 1 year after the initial diagnosis of
antagonist, is occasionally implicated in the onset of cytopenias and hepatitis. Most patients were young (ages 18 to 20 years); two-thirds
aplastic anemia, perhaps owing to a direct effect on early hematopoietic were male, and their survival was short (10 weeks). Although hepati-
progenitor cells.52 This drug accentuates the marrow-suppressive tis A and B have been implicated in aplastic anemia in a small num-
effects of the chemotherapy drug carmustine.53 In several instances, it ber of cases, most cases are related to non-A, non-B, non-C
has been reported as a possible cause of marrow aplasia when given hepatitis.7173 Severe aplastic anemia developed in 9 of 31 patients
with chloramphenicol. who underwent liver transplantation for non-A, non-B, non-C hepatitis,
CHAPTER 34: Aplastic Anemia: Acquired and Inherited 467

TABLE 343. Drugs Associated with Aplastic Anemia


Category High Risk Intermediate Risk Low Risk
Analgesic Phenacetin, aspirin, salicylamide
Antiarrhythmic Quinidine, tocainide
Antiarthritic Gold salts Colchicine
Anticonvulsant Carbamazepine, hydantoins, Ethosuximide, phenacemide, primidone,
felbamate trimethadione, sodium valproate
Antihistamine Chlorpheniramine, pyrilamine, tripelennamine
Antihypertensive Captopril, methyldopa
Antiinflammatory Penicillamine, phenylbutazone, Diclofenac, ibuprofen, indomethacin, naproxen,
oxyphenbutazone sulindac
Antimicrobial
Antibacterial Chloramphenicol Dapsone, methicillin, penicillin, streptomycin,
-lactam antibiotics
Antifungal Amphotericin, flucytosine
Antiprotozoal Quinacrine Chloroquine, mepacrine, pyrimethamine
Antineoplastic drugs
Alkylating agent Busulfan, cyclophosphamide, melphalan,
nitrogen mustard
Antimetabolite Fluorouracil, mercaptopurine, methotrexate
Cytotoxic antibiotic Daunorubicin, doxorubicin, mitoxantrone
Antiplatelet Ticlopidine
Antithyroid Carbimazole, methimazole, methylthiouracil,
potassium perchlorate, propylthiouracil, sodium
thiocyanate
Sedative and tranquilizer Chlordiazepoxide, chlorpromazine (and other
phenothiazines), lithium, meprobamate, methyprylon
Sulfa derivative Sulfonamides
Antibacterial Numerous sulfonamides
Diuretic Acetazolamide Chlorothiazide, furosemide
Hypoglycemic Chlorpropamide, tolbutamide
Miscellaneous Allopurinol, interferon, pentoxifylline, penicillamine

NOTE: Drugs that invariably cause marrow aplasia with high doses are termed high risk; drugs with 30 or more reported cases are listed as moderate risk; others are less often associated with
aplastic anemia (low risk).
SOURCE: This list was compiled from the AMA Registry,41 publications of the International Agranulocytosis and Aplastic Anemia Study,4246 other reviews and studies,24,4750 previous compila-
tions of offending agents,51 and selected reports. An additional comprehensive source for potentially offending drugs can be found in The Drug Etiology of Agranulocytosis and Aplastic Ane-
mia, Oxford, UK: Oxford University Press, 1991.

but in none of 1463 patients transplanted for other indications.75 Sev- therapy as does idiopathic aplastic anemia (see TREATMENT,
eral lines of evidence indicate there is no causal association with Combination Immunotherapy).
hepatitis C virus, suggesting that an unknown viral agent is
Epstein-Barr Virus Epstein-Barr virus (EBV) has been implicated in the
involved.16,75,76 Hepatitis virus B or C can be a secondary infection, if
pathogenesis of aplastic anemia.80,81 The onset usually occurs within 4 to
carefully screened blood products are not used for transfusion. In 15
6 weeks of infection. In some cases, infectious mononucleosis is subclini-
patients with posthepatitic aplastic anemia, no evidence was found
cal, with a finding of reactive lymphocytes in the blood film and serolog-
for hepatitis A, B, C, D, E, or G, transfusion-transmitted virus, or
ical results consistent with a recent infection (see Chap. 84). EBV has
parvovirus B19.76 Several reports suggest a relationship of parvovirus
been detected in marrow cells,81 but it is uncertain whether marrow apla-
B19 to aplastic anemia,77,78 whereas others have not.79 This relation-
sia results from a direct effect or an immunologic response by the host.
ship has not been established (see Chap. 35). The effect of seronega-
Patients have recovered following therapy with antithymocyte globulin.81
tive hepatitis may be mediated through an autoimmune T-cell effect
because of evidence of T-cell activation and cytokine elaboration.24 Other Viruses Human immunodeficiency virus (HIV) infection fre-
These patients also have a similar response to combined immuno- quently is associated with varying degrees of cytopenia. The marrow is
468 PART VI: The Erythrocyte

often cellular, but occasional cases of aplastic anemia have been who were given thorium dioxide (Thorotrast) as an intravenous con-
noted.8284 In these patients, marrow hypoplasia may result from viral trast medium suffered numerous late complications, including malig-
suppression and from the drugs used to control viral replication in this nant liver tumors, acute leukemia, and aplastic anemia.113 Chronic
disorder. Human herpes virus (HHV)-6 has caused severe marrow radium poisoning with osteitis of the jaw, osteogenic sarcoma, and
aplasia subsequent to marrow transplantation for other disorders.85 aplastic anemia was seen in workers who painted watch dials with
luminous paint when they moistened the brushes orally.114
Autoimmune Diseases Acute exposures to large doses of radiation are associated with the
The incidence of severe aplastic anemia was sevenfold greater than development of marrow aplasia and a gastrointestinal syndrome.115,116
expected in patients with rheumatoid arthritis.47 It is uncertain whether Total body exposure to between 1 and 2.5 Gy leads to gastrointestinal
the aplastic anemia is related directly to rheumatoid arthritis or to the symptoms and depression of leukocyte counts, but most patients
various drugs used to treat the condition (gold salts, D-penicillamine, recover. A dose of 4.5 Gy leads to death in half the individuals (LD50)
and nonsteroidal antiinflammatory agents). Occasional cases of aplastic owing to marrow failure. Higher doses in the range of 10 Gy are univer-
anemia are seen in conjunction with systemic lupus erythematosus.86 sally fatal unless the patient receives extensive supportive care followed
In vitro studies found either the presence of an antibody87 or suppressor by marrow transplantation. Aplastic anemia associated with nuclear
cell88,89 directed against hematopoietic progenitor cells. Patients have accidents was seen after the disaster that occurred at the Chernobyl
recovered after plasmapheresis,87 glucocorticoids,89 or cyclophospha- nuclear power station in the Ukraine in 1986.117
mide therapy,88,90 which is compatible with an immune etiology. Antineoplastic drugs such as alkylating agents, antimetabolites, and
Eosinophilic fasciitis, an uncommon connective tissue disorder with certain cytotoxic antibiotics have the potential for producing marrow
painful swelling and induration of the skin and subcutaneous tissue, has aplasia. In general, this is transient, is an extension of their pharmaco-
been associated with aplastic anemia.91,92 Although it may be antibody- logic action, and resolves within several weeks of completing chemo-
mediated in some cases, it has been largely unresponsive to therapy.91 therapy. Although unusual, severe marrow aplasia can follow use of the
Nevertheless, (1) stem cell transplantation, (2) immunosuppressive ther- alkylating agent, busulfan, and may persist indefinitely. Patients may
apy using cyclosporine,91 (3) immunosuppressive therapy using anti- develop marrow aplasia 2 to 5 years after discontinuation of alkylating
thymocyte globulin (ATG), or (4) immunosuppressive therapy with agent therapy. These cases often evolve into hypoplastic myelodysplas-
ATG and cyclosporine cures or significantly ameliorates the disease tic syndromes.
in a few patients.92
Severe aplastic anemia also has been reported coincident with Stromal Microenvironment and Growth Factors
immune thyroid disease (Graves disease)9397 and the aplasia has been Short-term clonal assays for marrow stromal cells have shown variable
reversed with treatment of the hyperthyroidism. Aplastic anemia has defects in stromal cell function. Serum levels of stem cell factor (SCF)
occurred in association with thymoma.98103 Autoimmune renal dis- have been either moderately low or normal in several studies of aplastic
ease and aplastic anemia have occurred concurrently. The underlying anemia.118,119 Although SCF augments the growth of hematopoietic
relationship may be the role of cytotoxic T lymphocytes in the patho- colonies from aplastic anemia patients marrows, its use in patients has
genesis of several autoimmune diseases and in aplastic anemia.104 not led to clinical remissions. Another early acting growth factor, FLT-3
ligand, is 30- to 100-fold elevated in the serum of patients with aplastic
Pregnancy anemia.120 Fibroblasts grown from patients with severe aplastic anemia
There are a number of reports of pregnancy-associated aplastic anemia, have subnormal cytokine production. However, serum levels of granu-
but the relationship between the two conditions is not always clear.105110 locyte colony-stimulating factor,121 erythropoietin,122 and thrombopoi-
In some patients, preexisting aplastic anemia is exacerbated with preg- etin (TPO)123 are usually high. Synthesis of IL-1, an early stimulator of
nancy, only to improve following termination of the pregnancy.105,106 In hematopoiesis, is decreased in mononuclear cells from patients with
other cases, the aplasia develops during pregnancy with recurrences aplastic anemia.124 Studies of the microenvironment have shown rela-
during subsequent pregnancies.106,107 Termination of pregnancy or tively normal stromal cell proliferation and growth factor produc-
delivery may improve the marrow function, but the disease may tion.125 These findings, coupled with the limited response of patients
progress to a fatal outcome even after delivery.105107 Therapy may with aplastic anemia to growth factors, suggest that cytokine deficiency
include elective termination of early pregnancy, supportive care, immu- is not the etiologic problem in most cases. The most compelling argu-
nosuppressive therapy, or marrow transplantation after delivery. Preg- ment is that most patients transplanted for aplastic anemia are cured
nancy in women previously treated with immunosuppression for with allogeneic donor stem cells and autologous stroma.126
aplastic anemia can result in the birth of a normal newborn.110 In this A rare exception is the homozygous or mixed heterozygous mutation of
latter study of 36 pregnancies, 22 were uncomplicated, 7 were compli- the TPO receptor gene, MPL, which can cause amegakaryocytic thrombo-
cated by a relapse of the marrow aplasia, and 5 without marrow aplasia cytopenia that evolves, later, into aplastic anemia (see Chap. 119).
required red cell transfusion during delivery.110 One death occurred
from cerebral thrombosis in a patient with paroxysmal nocturnal CLINICAL FEATURES
hemoglobinuria (PNH) and marrow aplasia. The onset of symptoms of aplastic anemia may be gradual with pallor,
weakness, dyspnea, and fatigue as a result of the anemia. Dependent pete-
Iatrogenic Causes chiae, bruising, epistaxis, vaginal bleeding, and unexpected bleeding at
Although marrow toxicity from cytotoxic chemotherapy or radiation other sites secondary to thrombocytopenia are frequent presenting signs of
produces direct damage to stem cells and more mature cells, resulting the underlying marrow disorder. Rarely, it may be more dramatic with
in marrow aplasia, most patients with acquired aplastic anemia cannot fever, chills, and pharyngitis or other sites of infection resulting from neu-
relate an exposure that would be responsible for marrow damage. tropenia and monocytopenia. Physical examination generally is unreveal-
Chronic exposure to low doses of radiation or use of spinal radiation ing except for evidence of anemia (e.g., conjunctival and cutaneous pallor,
for ankylosing spondylitis is associated with an increased, but delayed, resting tachycardia) or cutaneous bleeding (e.g., ecchymoses and pete-
risk of developing aplastic anemia and acute leukemia.111,112 Patients chiae), gingival bleeding and intraoral purpura. Lymphadenopathy and
CHAPTER 34: Aplastic Anemia: Acquired and Inherited 469

neutrophil count below 200/L (0.2 109/L) denotes very severe dis-
ease (see Table 341). Lymphocyte production is thought to be normal,
but patients may have mild lymphopenia. Platelets function normally.
TABLE 344. Approach to Diagnosis Significant qualitative changes of red cell, leukocyte, or platelet mor-
History and Physical Examination phology are not features of classical acquired aplastic anemia. On occa-
sion, only one cell line is depressed initially, which may lead to an early
Complete blood counts, reticulocyte count, and examination of the blood film
diagnosis of red cell aplasia or amegakaryocytic thrombocytopenia. In
Marrow aspiration and biopsy such patients, other cell lines will fail shortly thereafter (days to weeks)
Marrow cell cytogenetics to evaluate clonal myeloid disease and permit a definitive diagnosis. Table 344 is a plan for initial labora-
Fetal hemoglobin level and DNA stability test as markers of Fanconi anemia tory investigation.
Immunophenotyping of red and white cells, especially for CD55, CD59 to
exclude PNH Plasma Findings
Direct and indirect Coombs test to rule out immune cytopenia The plasma contains high levels of hematopoietic growth factors, includ-
Serum lactate dehydrogenase (LDH) and uric acid that if increased may ing erythropoietin, thrombopoietin, and myeloid colony-stimulating fac-
reflect neoplastic cell turnover tors. Plasma iron values are usually high, and 59Fe clearance is
Liver function tests to assess evidence of any recent hepatitis virus exposure prolonged, with decreased incorporation into red cells.
Screening tests for hepatitis viruses A, B, and C
Screening tests for EBV, cytomegalovirus (CMV), and HIV Marrow Findings
Serum B12 and red cell folic acid levels to rule out megaloblastic Morphology The marrow aspirate typically contains numerous spicules
pancytopenia with empty, fat-filled spaces, and relatively few hematopoietic cells.
Serum iron, iron-binding capacity, and ferritin as a baseline prior to chronic Lymphocytes, plasma cells, macrophages, and mast cells may be
transfusion therapy present. On occasion, some spicules are cellular or even hypercellular
(hot spots), but megakaryocytes usually are reduced. These focal
areas of residual hematopoiesis do not appear to be of prognostic sig-
splenomegaly are not features of aplastic anemia; such findings suggest an nificance. Residual granulocytic cells generally appear normal, but it is
alternative diagnosis such as a clonal myeloid or lymphoid disease. not unusual to see mild macronormoblastic erythropoiesis, presumably
as a result of the high levels of erythropoietin. Marrow biopsy is essen-
tial to confirm the overall hypocellularity (Fig. 342), as a poor yield of
LABORATORY FEATURES spicules and cells occurs in marrow aspirates in other disorders, espe-
Blood Findings cially if fibrosis is present.
In severe aplastic anemia, as defined by the International Aplastic
Patients with aplastic anemia have varying degrees of pancytopenia.
Anemia Study Group, less than 25 percent cellularity or less than 50
Anemia is associated with a low reticulocyte index. The relative reticu-
percent cellularity with less than 30 percent hematopoietic cells is seen
locyte count is usually less than 1 percent and may be zero despite the
in the marrow.
high levels of erythropoietin. Absolute reticulocyte counts are usually
fewer than 40,000/L (40 109/L). Macrocytes may be present. The Progenitor Cell Growth In vitro CFU-GM and BFUE colony assays
absolute neutrophil and monocyte count are low. An absolute neutro- reveal a marked reduction in progenitor cells.1922
phil count fewer than 500/L (0.5 109/L) along with a platelet count Cytogenetic Studies Cytogenetic analysis may be difficult to perform
fewer than 30,000/L (30 109/L) is indicative of severe disease and a owing to low cellularity; thus, multiple aspirates may be required to

A B

FIGURE 342. Marrow biopsy in aplastic anemia. A. A normal marrow biopsy section of a young adult. B. The marrow biopsy section of a young adult with very severe aplastic
anemia. The specimen is devoid of hematopoietic cells and contains only scattered lymphocytes and stromal cells. The hematopoietic space is replaced by reticular cells (pre-adipocytic
fibroblasts) converted to adipocytes.
470 PART VI: The Erythrocyte

provide sufficient cells for study. The results are normal in aplastic presence of hairy cells. Other clinical features may be distinctive (see
anemia. Clonal cytogenetic abnormalities in otherwise apparent aplas- Chap. 95). Organomegaly such as lymphadenopathy, hepatomegaly, or
tic anemia is indicative of an underlying hypoaccumulative clonal splenomegaly are inconsistent with the atrophic (hypoproliferative) fea-
myeloid disease.127 tures of aplastic anemia. Large granular lymphocytic leukemia has also
Imaging Studies Magnetic resonance imaging can be used to distin- been associated with aplastic anemia. Rare cases of typical acquired
guish between marrow fat and hematopoietic cells.128 This may be a aplastic anemia have been followed by t(9;22)-positive acute lympho-
more useful overall estimate of marrow hematopoietic cell density than cytic leukemia (ALL), or chronic myelogenous leukemia (CML).131
morphologic techniques and may help differentiate hypoplastic
myelogenous leukemia from aplastic anemia.128
RELATIONSHIP AMONG APLASTIC ANEMIA,
PNH, AND CLONAL MYELOID DISEASES
DIFFERENTIAL DIAGNOSIS In addition to the diagnostic difficulties occasionally presented by
Any disease that can present with pancytopenia may mimic aplastic patients with hypoplastic myelodysplastic syndromes, hypoplastic
anemia if only the blood counts are considered. Measurement of the acute myelogenous leukemia (AML), or PNH with hypocellular mar-
reticulocyte count and an examination of the blood film and marrow rows, there may be a more fundamental relationship among these three
biopsy are essential early steps to arrive at a diagnosis. A reticulocyte diseases and aplastic anemia. The development of clonal cytogenetic
percentage of 0.5 percent to zero is strongly indicative of aplastic eryth- abnormalities such as monosomy 7 or trisomy 8 in a patient with aplas-
ropoiesis, and when coupled with leukopenia and thrombocytopenia, tic anemia portends the evolution of a myelodysplastic syndrome or
points to aplastic anemia. Absence of qualitative abnormalities of cells acute leukemia. Occasionally, these cytogenetic markers have been
on the blood film and a markedly hypocellular marrow are characteris- transient, and in cases with disappearance of monosomy 7, hematologic
tic of acquired aplastic anemia. The disorders most commonly con- improvement has occurred as well.132 Persistent monosomy 7 carries a
fused with severe aplastic anemia include the approximately 5 to 10 poor prognosis as compared to trisomy 8.133,134
percent of patients with myelodysplastic syndromes who present with a As many as 15 to 20 percent of patients with aplastic anemia have a
hypoplastic rather than a hypercellular marrow. Myelodysplasia should 5-year probability of developing myelodysplasia.132 If one excludes
be considered if there is abnormal blood film morphology consistent any transformation to a clonal myeloid disorder that occurs up to 6
with myelodysplasia (e.g., poikilocytosis, basophilic stippling, neutro- months after treatment to avoid misdiagnosis among the hypoplastic
phils with the pseudo-Pelger-Het anomaly). Marrow erythroid pre- clonal myeloid diseases, the frequency of a clonal disorder was nearly
cursors in myelodysplasia may have dysmorphic features. Pathologic 15 times greater in patients treated with immunosuppression as com-
sideroblasts are inconsistent with aplastic anemia and a frequent feature pared to those treated with marrow transplantation after 39 months of
of myelodysplasia. Granulocyte precursors may have reduced or abnor- observation.135 This finding suggests either that immune suppression
mal granulation. Megakaryocytes may have abnormal nuclear lobula- by antiT-cell therapy enhances the evolution of a neoplastic clone or
tion (e.g., unilobular micromegakaryocytes; see Chap. 88). If clonal that it does not suppress the intrinsic tendency of aplastic anemia to
cytogenetic abnormalities are found, a clonal myeloid disorder, espe- evolve to a clonal disease, but provides the increased longevity of the
cially myelodysplastic syndrome or hypocellular myelogenous leuke- patient required to express that potential. The latter interpretation is
mia is likely. Magnetic resonance imaging (MRI) studies of bone may more likely as patients successfully treated solely with androgens
be useful in differentiating severe aplastic anemia from clonal myeloid develop clonal disease as frequently as those treated with immuno-
syndromes. The former gives a fatty signal and the latter a diffuse cellu- suppression.136 Transplantation may reduce the potential to clonal
lar pattern. evolution in patients with aplastic anemia by reestablishing robust
A hypocellular marrow frequently is associated with PNH. PNH is lymphohematopoiesis.
characterized by an acquired mutation in the PIG-A gene that encodes Telomere shortening also may play a pathogenetic role in the evolution
an enzyme that is required to synthesize mannolipids. The latter defi- of aplastic anemia into myelodysplasia. Patients with aplastic anemia
ciency prevents the synthesis of the glycosyl-phosphatidylinositol have shorter telomere lengths than matched controls, and patients with
anchor precursor. This moiety anchors several proteins, including aplastic anemia with persistent cytopenias had greater telomere shorten-
inhibitors of the complement pathway to blood cell membranes, and its ing over time than matched controls. Three of five patients with telomere
absence accounts for the complement-mediated hemolysis in PNH. As lengths less than 5 kb developed clonal cytogenetic changes, whereas
many as 50 percent of patients with otherwise typical aplastic anemia patients with longer telomeres did not develop such diseases.23,137
have evidence of glycosyl-phosphatidylinositol molecule defects and The relationship of PNH to aplastic anemia remains enigmatic.
diminished phosphatidylinositol-anchored protein on leukocytes and Because hematopoietic stem cells lacking the phosphatidylinositol-
red cells as judged by flow cytometry, analogous to that seen in PNH.129 anchored proteins are present in many or all normal persons in very
The decrease or absence of these membrane proteins may make the small numbers,138 it is not surprising that more than 50 percent of
PNH clone of cells resistant to the acquired immune attack on normal patients with aplastic anemia may have a PNH cell population as
marrow components, or the phosphatidylinositol-anchored protein(s) detected by immunophenotyping.129 The probability of patients with
on normal cells provides an epitope that initiates an aberrant T-cell aplastic anemia developing a clinical syndrome consistent with PNH is
attack, leaving the PNH clone relatively resistant (see Chap. 40).24 10 to 20 percent, and this is not a consequence of immunosuppressive
Occasionally, apparent aplastic anemia may be the prodrome to treatment.132 Patients also may present with the hemolytic anemia of
childhood130 or, less commonly, adult131 acute lymphoblastic leukemia. PNH and later develop progressive marrow failure so that any pathoge-
Sometimes, careful examination of marrow cells by light microscopy or netic explanation should consider both types of development of aplastic
flow cytometry will uncover a population of leukemic lymphoblasts. In marrows in PNH. The PIG-A mutation may confer either a proliferative
other cases, the acute leukemia may appear later. Hairy-cell leukemia, or survival advantage to PNH cells.139 A survival advantage could result
Hodgkin disease, or another lymphoma subtype, rarely, may be pre- if the anchor protein or one of its ligands served as an epitope for the T-
ceded by a period of marrow hypoplasia. Immunophenotyping of mar- lymphocyte cytotoxicity inducing the marrow aplasia. In this case, the
row and blood cells by flow cytometry for CD25 may uncover the presenting event could either reflect cytopenias or the sensitivity of red
CHAPTER 34: Aplastic Anemia: Acquired and Inherited 471

aplastic anemia, as their use has markedly reduced the problem of


graft rejection.140
Cytomegalovirus (CMV)-reduced risk red cells and platelets should
TABLE 345. Initial Management of Aplastic Anemia be given to a potential transplant recipient to minimize problems with
Discontinue any potential offending drug and use an alternative class of CMV infections after transplantation. Once a patient is shown to be
agents if essential. CMV-positive, this restriction is no longer necessary. Leukocyte-deple-
Anemia: transfusion of leukocyte-depleted, irradiated red cells as required tion filters or CMV serotesting are equivalent methods of decreasing
for very severe anemia. the risk of transmitting CMV.
Very severe thrombocytopenia or thrombocytopenic bleeding: consider Red Cell Transfusion Packed red cells to alleviate symptoms of anemia
-aminocaproic acid; transfusion of platelets as required. usually are indicated at hemoglobin values below 8 g/dL (80 g/L),
Severe neutropenia; use infection precautions. unless comorbid medical conditions require a higher hemoglobin con-
centration. These products should be leukocyte-depleted to lessen leu-
Fever (suspected infection): microbial cultures; broad-spectrum antibiotics
kocyte and platelet sensitization and to reduce subsequent transfusion
if specific organism not identified, granulocyte colony-stimulating factor
reactions and radiated to reduce the potential for a graft-versus-host
(G-CSF) in dire cases. If child or small adult with profound infection
reaction. It is important not to transfuse patients with red cells (or
(e.g., gram-negative bacteria, fungus, persistent positive blood cultures) can
platelets) from family members if transplantation within the family is
consider neutrophil transfusion from a G-CSF pretreated donor.
remotely possible, as this approach may sensitize patients to minor his-
Immediate assessment for allogeneic stem cell transplantation: Histocom- tocompatibility antigens, increasing the risk of graft rejection after
patibility testing of patient, parents, and siblings. Search databases for unre- marrow transplantation. Following a marrow transplant, or in those
lated donor, if appropriate. individuals in whom transplantation is not a consideration, family
members may be ideal donors for platelet products. Because each unit
of red cells adds approximately 200 to 250 mg of iron to the total body
iron, over the long-term transfusion-induced iron overload may occur.
cells to complement lysis and hemolysis, depending on the intrinsic
This is not a major problem in patients who respond to transplantation
proliferative potential of the PNH clone.
or immunosuppressive therapy, but it is an issue in nonresponders who
Within our current state of knowledge, aplastic anemia is an autoim-
require continued transfusion support. In the latter case, consideration
mune process, and any residual hematopoiesis is presumably polyclo-
should be given to iron chelation therapy. Newer oral agents will make
nal. This is a critical distinction from hypoplastic leukemia and PNH,
this procedure easier to effect (see Chap. 47).141
which are clonal (neoplastic) diseases. The environment of the aplastic
marrow, however, may favor the eventual evolution of a mutant (malig- Platelet Transfusion It is important to assess the risk of bleeding in each
nant) clone, especially if immunotherapy is used, whereas hematopoi- patient. Most patients tolerate platelet counts of 10,000/L (10 109/L)
etic stem cell transplantation may either ablate threatening minor without undue bruising or bleeding, unless a systemic infection is
clones or establish more robust hematopoiesis, an environment less present.142,143 A traumatic injury or surgery requires transfusion to
conducive to clonal evolution. >50,000/L or >100,000/L, respectively. Administration of -ami-
nocaproic acid, 100 mg/kg per dose every 4 hours (maximum dose 5 g)
orally or intravenously, may reduce the bleeding tendency.144 Pooled
TREATMENT random-donor platelets may be used until sensitization ensues,
Approach to Therapy although it is preferable to use single-donor platelets from the onset to
minimize sensitization to HLA or platelet antigens. Subsequently,
Severe anemia, bleeding from thrombocytopenia, and, rarely at the time
single-donor apheresis products or HLA-matched platelets may be
of diagnosis, infection secondary to granulocytopenia and monocytope-
required.
nia require prompt attention to remove potential life-threatening con-
Platelet refractoriness is a major problem with long-term transfusion
ditions and improve patient comfort (Table 345). More specific
support.145 This may occur transiently, with fever or infection, or as a
treatment of the marrow aplasia involves two principal options: (1) syn-
chronic problem secondary to HLA sensitization. In the past, this
geneic or allogeneic hematopoietic stem cell transplantation or (2) com-
occurred in approximately 50 percent of patients after 8 to 10 weeks of
bination immunosuppressive therapy with ATG and cyclosporine. The
transfusion support. Filtration of blood and platelet concentrates to
selection of the specific mode of treatment depends on several factors,
remove leukocytes reduces this problem to approximately 15 percent of
including the patients age and condition and the availability of a suitable
patients receiving chronic transfusions.145,146 Patients should also get
allele-level HLA-matched hematopoietic stem cell donor. In general,
ABO-identical platelets because this enhances platelet survival and fur-
transplantation is the preferred treatment for children and most other-
ther decreases refractoriness to platelet transfusion. Single-donor HLA-
wise healthy younger adults. Early histocompatibility testing of siblings
matched apheresis-harvested platelets may be necessary in previously
is of particular importance because it establishes whether there is an
pregnant or transfused patients who are already allosensitized or who
optimal donor available to the patient for transplantation. The preferred
become so after treatment with leukoreduced platelets. The frequency
stem cell source is a histocompatible sibling matched at the HLA-A, B,
of either of these events is less than 10 percent. Approaches to chronic
C, and DR loci.
platelet transfusion are discussed in Chap. 141. The utility of TPO
receptor agonists in the thrombocytopenia of aplastic anemia is yet to
Supportive Care be determined.
The Use of Blood Products Although it was recommended that red Management of Neutropenia Neutropenic precautions should be applied
cell and platelet transfusions be used sparingly in potential transplant to hospitalized patients with a severe depression of the neutrophil count.
recipients to minimize sensitization to histocompatibility antigens, The level of neutrophils requiring precautions is fewer than 500/L
this has become less important since ATG and cyclophosphamide (0.5 109/L). One approach is to use private rooms, with requirements
have been used as the preparative regimen for transplantation in for face masks and handwashing with antiseptic soap. Unwashed fresh
472 PART VI: The Erythrocyte

fruits and vegetables should be avoided as they are 100 100


sources of bacterial contamination. It is uncom-
90 90
mon for patients with aplastic anemia to present HLA-id sib, 20y (N = 915)
with a significant infection. When patients with 80 80
aplastic anemia become febrile, cultures should be HLA-id sib, >20y (N = 822)
70 70

Percent survival
obtained from the throat, sputum (if any), blood,
urine, stool, and any suspicious lesions. Broad- 60 Unrelated, 20y (N = 388) 60
spectrum bacteriocidal antibiotics should be initi- 50 50
ated promptly, without awaiting culture results.
The choice of antibiotics depends on the preva- 40 Unrelated, >20y (N = 212) 40
lence of organisms and their antibiotic sensitivity 30 30
in the local setting. Organisms of concern usually
20 20
include Staphylococcus aureus (notably methicillin-
and oxacillin-resistant strains), Staphylococcus epi- 10 10
P < 0.001
dermidis (in patients with venous access devices),
0 0
and gram-negative organisms. Patients with persis- 0 1 2 3 4 5 6
tent culture-negative fevers should be considered Years
for antifungal treatment (see Chap. 22).
In the past, leukocyte transfusions were used on a FIGURE 343. Probability of survival after hematopoietic stem cell transplantation for severe aplastic anemia by do-
daily basis to reduce the short-term mortality from nor type and age, 19982004. Patients receiving marrow from a matched sibling had better outcomes if they were
infections. It was unusual to detect more than 100 to equal to or less than 20 years of age, compared to those older than 20 years of age. Patients receiving marrow from
200 neutrophils per microliter for more than a few a matched sibling fared better than those who received marrow from a matched unrelated donor, at any age. Patients
hours after transfusion. The yield of neutrophils can younger than 20 or older than 20 years of age did not have a significant difference in outcome if they received mar-
be increased by administering granulocyte colony- row from an unrelated matched donor. Id, identical; sib, sibling. (Figure reproduced from the Center for Interna-
stimulating factor (G-CSF) to the donor,147 but most tional Transplant Research data on the National Marrow Donor Program website. http://www.marrow.org/
physicians avoid using white cell products because PATIENT/Undrstnd_Disease_Treat/Lrn_about_Disease/Aplastic_Anemia/AA_Tx_Outcomes/index.html. Last ac-
present-day antibiotics are usually sufficient to treat cessed February 2009.)
a patient for an episode of sepsis. Notable exceptions
include documented invasive aspergillosis unre-
sponsive to amphotericin (particularly in the posttransplant setting), gram or similar organizations in other countries.152 Umbilical cord
infections with organisms resistant to all known antibiotics, and if blood blood is an alternative source of stem cells from unrelated donors (or,
cultures remain positive in spite of antibiotic treatment. Leukocyte trans- rarely, siblings) for transplantation in children. The use of high-resolution,
fusion is more effective in children and adults with smaller body size, as HLA typing of a matched, unrelated donor markedly improves the
transfused leukocytes have a smaller distribution space, which results in prognosis for transplantation.153 High-resolution DNA matching at
higher blood and tissue concentrations. HLA-A, -B, -C, and -DRB1 (8 of 8 allele) is considered the lowest level
Hematopoietic Stem Cell Transplantation Prompt therapy usually is of matching consistent with the highest level of survival. If there is an
indicated for patients with severe aplastic anemia. The major curative HLA mismatch at one or more loci, especially HLA-A or -DRB1, the
approach is hematopoietic stem cell transplantation from a histocom- outcome is compromised,153 and immunosuppression with combined
patible sibling.148150 This treatment modality is described in Chap. 21. therapy may be preferred initially, depending on patient age, cytomega-
Only 20 to 30 percent of patients in the United States have compatible lovirus status, and disease severity. The use of hematopoietic stem cell
sibling donors (related to average family size). In the unusual case of an transplantation can be considered for patients who do not respond or
identical twin donor, conditioning is required to obliterate the immune who no longer respond to immunotherapy.151 If the patient in question
disease in the recipient, but it can be limited to cyclophosphamide. In is a candidate for stem cell transplantation based on all relevant factors,
this setting, an 80 to 90 percent survival is expected. Marrow stem cells transplantation could be considered at any age for a patient with a syn-
seem to perform better than blood stem cells when used as a source for geneic donor; transplantation could be considered as a first-choice
patients with aplastic anemia, although this is under continued study. therapy up to age 50 years for a patient with an HLA allele-level
The results of transplantation are best in patients younger than age 20 matched sibling donor; and transplantation could be considered a first-
years (80 to 90 percent long-term survival) but decrease every decade choice therapy if an allele-level HLA-matched unrelated donor is avail-
of age thereafter. Posttransplant mortality is increased and survival able for patients younger than age 20 years.151
decreased with increasing age (Fig. 343). In patients older than age 40
years, survival in matched sibling transplant is reduced to approxi-
Components of AntiT-Lymphocyte (Immunosuppressive) Therapy
mately 50 percent.151 There are still uncertainties about the optimal Antilymphocyte Serum and Antithymocyte Globulin ATG and ALG act
conditioning program in younger and older patients. ATG, cyclophos- principally by reducing cytotoxic T cells. This involves ATG-induced
phamide, total-body radiation, and fludarabine are among the agents apoptosis through both FAS and TNF pathways.154 Cathepsin B also
being studied.148,150,151 The longer the delay between diagnosis and plays a role in T-cell cytotoxicity at clinical concentrations of ATG, but
transplant, the less salutary the outcome, probably as a result of a may involve an independent apoptosis pathway.155 ATG and ALG also
greater number of transfusions and a higher likelihood of pretransplant release hematopoietic growth factors from T cells.156,157 Horse and rab-
infection. Acute and chronic graft-versus-host disease are serious com- bit ATG are licensed in the United States. Skin tests against horse serum
plications, and therapy to prevent or ameliorate them is a standard part should be performed prior to administration.158 If positive, the patient
of posttransplant treatment.148,151 Transplants have been performed may be desensitized. ATG therapy is given daily for 4 to 10 days with
using stem cells from partially matched siblings or unrelated, histo- doses of 15 to 40 mg/kg. Fever and chills are common during the first
compatible donors recruited through the National Marrow Donor Pro- day of treatment. Concomitant treatment with glucocorticoids, such as
CHAPTER 34: Aplastic Anemia: Acquired and Inherited 473

methylprednisolone or dexamethasone lessens the reaction to ATG. (4) in varying combinations with other modes of therapy.177 Cyclospor-
Studies are under way to compare equine to rabbit ATG in the immu- ine is administered orally at 10 to 12 mg/kg per day for at least 4 to 6
notherapy of aplastic anemia. months. Dosage adjustments may be required to maintain trough
ATG treatment may accelerate platelet destruction, reduce the abso- blood levels of 200 to 400 ng/mL. Renal impairment is common and
lute neutrophil count, and cause a positive direct antiglobulin test. This may require increased hydration or dose adjustments to keep creatinine
effect may lead to an increase in transfusion requirements during the 4- values below 2 mg/dL. Cyclosporine also may cause moderate hyper-
to 10-day treatment interval. Serum sickness, characterized by spiking tension, a variety of neurological manifestations, and other side effects.
fevers, skin rashes, and arthralgias, occurs commonly 7 to 10 days from Several drug classes interact with cyclosporine to either increase (e.g.,
the first dose. The clinical manifestations of serum sickness can be some antibiotics and antifungals) or decrease (e.g., some anticonvul-
diminished by increasing the glucocorticoid dose from day 10 to day 17 sants) blood levels. Responses usually are seen by 3 months and may
after treatment. Approximately one-third of patients no longer require range from achieving transfusion independence to complete remission.
transfusion support after treatment with ATG alone.159161 Approximately 25 percent of patients respond to this agent when used
Of 358 patients responding to immunosuppressive therapy, princi- alone, but the response rate has ranged from 0 to 80 percent in various
pally ATG alone, 74 (21%) relapsed after a mean of 2.1 years. The actu- reports.177
arial incidence of relapse was 35 percent at 10 years.162 Similar results Although immunosuppression with ALG or ATG has been used the
were observed when 227 patients were treated with immunosuppres- longest and has a seemingly better response rate, there are certain
sion, primarily ATG alone.163 The actuarial survival at 15 years was 38 advantages to cyclosporine. This drug does not require hospitalization
percent following immunosuppression.162 However, a combination of or use of central venous catheters. Fewer platelet transfusions are
immunosuppressive agents provides more effective therapy than ATG required during the first few weeks of therapy compared to treatment
alone (see Combination Immunotherapy below). with ALG or ATG. A French cooperative trial showed equal effective-
Twenty-eight (22%) of 129 patients treated with ALG developed ness of ATG plus prednisone compared to cyclosporine.178 In this
myelodysplasia, leukemia, paroxysmal nocturnal hemoglobinuria, or crossover study of newly diagnosed patients, survival of approximately
combined disorders.164 This tendency to relapse and to develop clonal 65 percent was observed 12 months after diagnosis.
hematologic disorders was reviewed by the European Cooperative Combination Immunotherapy Combination treatment of severe aplas-
Group for Bone Marrow Transplantation in 468 patients, most of tic anemia usually includes, for example, ATG, 40 mg/kg per day, for 4
whom received ATG.165 The risk of a hematologic complication days; cyclosporine, 10 to 12 mg/kg per day, for 6 months and methyl-
increased continuously and reached 57 percent at 8 years after prednisolone, 1 mg/kg per day, for 2 weeks.179 The dose of cyclosporine
immunosuppressive therapy. A further survey found 42 (5%) malig- is adjusted to maintain a trough level of 200400 ng/mL. Prophylaxis
nancies in 860 patients treated with immunosuppression, whereas only for Pneumocystis carinii with daily trimethoprim-sulfamethoxazole or
9 (1%) malignancies were seen in 748 patients who received marrow with monthly pentamidine inhalations should be considered for these
transplants.166 patients as they receive immunosuppressive therapy.
Cyclosporine Administration of cyclosporine, a cyclic polypeptide that The addition of cyclosporine to the combination of ALG and gluco-
inhibits IL-2 production by T lymphocytes and prevents expansion of corticoids improves response rates to approximately 70 percent of
cytotoxic T cells in response to IL-2, is another approach to immunother- patients (Table 346).180,181 G-CSF added to the combined immuno-
apy. After the initial report of its ability to induce remission in 1984,167 suppressive therapy does not increase response rate or survival.183
several groups have used cyclosporine as either (1) primary treat- Response is usually defined as a significant improvement in red cells,
ment,168171 (2) in patients refractory to ATG or glucocorticoids,169174 white cells, and platelets to eliminate risk of infection and bleeding and
(3) in combination with granulocyte colony-stimulating factors,175,176 or the requirement for red cell transfusions.

TABLE 346. Response to Immunotherapy in Patients with Severe Aplastic Anemia


Significant Survival at Relapse at
Year of No. Pts (Age- Response 5/10 Years 5 Years
Publication Principal Drugs Used range, yrs) No. (%) (%) (Cum%) Comments Reference
2008 ATG + CYA 77 (<18) 57 (74) 83/80 25 8.5% evolved to clonal myeloid disease 180
2007 ATG + CYA 44 (NR) 31 (70) NR/88 NR All cases were associated with hepatitis 181
2007 ATG + CYA 47 (1975) 31 (66) 80/NR 45 No late clonal diseases at 5 years 182
2007 ATG + CYA + G-CSF 48 (1974) 37 (77) 90/NR 15 No late clonal diseases at 5 years 182
2006 ATG + CYA 47 (871) 37 (79) 80/75 NR No late clonal diseases at 10 years 183
2006 ATG + CYA + G-CSF 30 (568) 22 (73) 80/75 NR One patient developed clonal myeloid 183
+ rhuEPO disease

ATG, antithymocyte globulin; Cum%, cumulative percent; CYA, cyclosporine A, G-CSF, granulocyte colony-stimulating factor; No. Pts, number of patients; NR, not reported; rhuEPO, recombi-
nant human erythropoietin.
NOTE: In some cases response, survival, and relapse percentages are very close approximations, read off the published graphs. Significant response combines complete and partial remissions,
which usually means the platelet and red cell count are high enough to avoid transfusions and neutrophil count over a critical level. Some protocols used short periods of glucocorticoid treat-
ment to ameliorate reactions to ATG. There is a significant frequency of relapses or progression to clonal myeloid disease after 5 to 10 years postimmunotherapy.
474 PART VI: The Erythrocyte

The 5-year survival after completion of combination immunosup- as a third drug in an immunotherapy regimen. The role of B lympho-
pressive therapy may approximate that after stem cell transplanta- cytes in the pathogenesis of aplastic anemia has not been defined.
tion.184 Forty-eight children treated between 1983 and 1992 had a 10- Androgens Randomized trials have not shown efficacy when andro-
year survival of approximately 75 percent for marrow transplantation gens were used as primary therapy for severe or moderately severe
and approximately 75 percent for combined immunosuppressive ther- aplastic anemia.199,200
apy, although there were only half the number of severely affected Androgens stimulate the production of erythropoietin, and their
patients in the immunosuppressive therapy group.185 Thus, immuno- metabolites stimulate erythropoiesis when added to marrow cultures in
suppression may be preferable for patients who are older than 30 years vitro. High doses of androgens were beneficial in some patients with
of age and in those who may experience a delay in finding a suitable moderately severe aplasia.199 Series of patients were reported in which
donor. Marrow transplants are, however, curative for aplastic anemia, survival seemed improved as compared with historical controls, but
whereas more frequent sequelae have been found after immunosup- this could have resulted from improved supportive care.136 Masculin-
pressive therapy,186188 notably a substantial rate of evolution to a ization and other androgen side effects can be severe. Long-term survi-
myelodysplastic syndrome or acute myelogenous leukemia. vors after androgen therapy have essentially the same progression to
A recent National Institutes of Health protocol was designed to clonal hematologic disorders as patients treated with immunosuppres-
increase immune tolerance by specific deletion of activated T lympho- sive agents.136 These agents have been replaced by immunosuppression
cytes that target primitive hematopoietic progenitor cells.24 Concurrent or allogeneic hematopoietic stem cell transplantation.
administration of cyclosporine with ATG may diminish the ATG effect
Cytokines Despite their effectiveness in accelerating recovery from che-
so that in this program cyclosporine is introduced at a later time. The
motherapy, these agents have been far less effective in achieving long-
addition of new immunosuppressive agents, such as mycophenolate
term benefits in patients with severe aplastic anemia. Daily treatment
mofetil, rapamycin, or monoclonal antibodies, to the IL-2 receptor may
with G-CSF201,202 has improved marrow cellularity and increased neutro-
be more effective in decreasing cytotoxic T cells, sparing the targeted
phil counts approximately 1.5- to 10-fold. Unfortunately, in nearly all
hematopoietic stem cells.24
patients, the blood counts return to baseline within several days of cessa-
For the 30 to 40 percent of patients who relapse after immunother-
tion of therapy. Although occasional patients show evidence of trilineage
apy, retreatment with ATG and cyclosporine is effective in 50 to 60 per-
marrow recovery with long-term therapy, the vast majority do not
cent of them.189,190
respond. Therapy with myeloid growth factors is probably best reserved
High-Dose Glucocorticoid Treatment Marrow recovery can occur after for episodes of severe infection or as a preventive measure prior to dental
very high doses of glucocorticoids.191,192 Methylprednisolone in the work or other procedures that would compromise mucosal barriers in
range of 500 to 1000 mg daily for 3 to 14 days has been successful, but patients who have not responded to stem cell transplant or immunother-
the side effects, which include marked hyperglycemia and glycosuria, apy. Prophylactic use of growth factors is not warranted. G-CSF in a dose
electrolyte disturbances, gastric irritation, psychosis, increased infec- of 5 g/kg by subcutaneous injection is easiest to administer and seems
tions, and aseptic necrosis of the hips, can be severe. Glucocorticoids at to be associated with the fewest side effects. The drug can be given daily
lower doses commonly are used only as a component of combination or fewer times per week depending on the response. Newer pegylated
therapy for aplastic anemia to ameliorate the toxic effects of ATG and preparations have greater longevity and usually are administered at less
in providing additional lymphocyte suppression. frequent, every-other-week intervals.
High-Dose Cyclophosphamide Therapy High-dose cyclophosphamide has IL-1, a potent stimulator of marrow stromal cell production of other
been used as a form of immunosuppression.193 Although it would seem cytokines, and IL-3 have been ineffective in small numbers of patients
inappropriate to administer high doses of chemotherapy to patients with severe aplastic anemia.204,205 These disappointing results with
with severe marrow aplasia, this approach was based on observations of cytokines are not unexpected, as previous work has found high serum
autologous recovery after preparative therapy for allogeneic trans- levels of growth factors in patients with aplastic anemia. Moreover, the
plants.6 Ten patients received cyclophosphamide at 45 mg/kg per day majority of patients have suppression of very primitive progenitors,
intravenously for 4 days with or without cyclosporine for an additional which may be unresponsive to individual factors that act on more
100 days. Gradual neutrophil and platelet recovery ensued over 3 mature progenitor cells.
months. Seven patients responded completely and remained in remis- Splenectomy Removal of the spleen does not increase hematopoiesis but
sion 11 years after treatment. High-dose cyclophosphamide treatment may increase neutrophil and platelet counts two- to threefold and
may spare hematopoietic stem cells, which have high levels of aldehyde improve survival of transfused red cells or platelets in highly sensitized
dehydrogenase and are relatively resistant to cyclophosphamide.194,195 individuals.206 The surgical morbidity and mortality in patients with few
Thus, cyclophosphamide in this situation may be more immunosup- platelets and white cells makes this a questionable therapeutic procedure.
pressive than myelotoxic. The most extensive trial of high-dose cyclo- Because there are more successful methods of therapy that attack the fun-
phosphamide resulted in 65 percent of patients responding completely damental problem, this approach would not be used today.
at 50 months.196 However, the role of this regimen as initial therapy is Other Therapy High doses of intravenous gamma globulin have been
not clear because of early toxicity that may exceed that of the ATG- given to small numbers of patients with severe aplastic anemia207,208
cyclosporine combination.197 The probability of a durable remission because of its success in treating certain cases of antibody-mediated
may be superior, but there are insufficient data (comparative clinical pure red cell aplasia. Some improvement was noted in 4 of 6 patients
trials) to conclude whether high-dose cyclophosphamide provides bet- treated. Another treatment that is occasionally successful is lympho-
ter long-term results than ATG and cyclosporine. The latter approach is cytapheresis to deplete T cells.209,210
favored at this time.
Rituximab A case report of the successful use of the anti-CD20 human- Course and Prognosis
ized mouse antibody rituximab has provided preliminary evidence for At diagnosis, the prognosis is largely related to the absolute neutrophil
its potential effectiveness in treating aplastic anemia.198 Clinical trials and platelet count. The absolute neutrophil count is the most important
should examine its efficacy compared to standard immunotherapy prognostic feature, with a count of fewer than 500/L (0.5 109/L) con-
(ATG and cyclosporine), in patients refractory to standard therapy, or sidered severe aplastic anemia and a count of fewer than 200/L (0.2
CHAPTER 34: Aplastic Anemia: Acquired and Inherited 475

109/L) very severe aplastic anemia, the latter associated with a poor complementation group is a genetic subgroup. Identifying a comple-
response to immunotherapy and usually a dire prognosis if early suc- mentation group requires adding a gene to the genome of a cell to cor-
cessful allogeneic transplant is not available. In the past, the prognosis rect (complement) the genetic defect. This procedure can be done by
appeared worse when the disease followed hepatitis.69,70 But more com- cell fusion studies. After fusing two cells together, thereby joining their
prehensive results with immunosuppression191 or hematopoietic stem genetic material, one can test the cells for the genetic defect. In the case
cell transplantation211 show an equivalent response to that seen with of Fanconi anemia, this would be with the diepoxybutane test. Hybrids
idiopathic or drug-induced cases. in which the hypersensitivity to diepoxybutane is corrected (comple-
Before marrow transplantation and immunosuppressive therapy, more mented) can be assumed to result from the fusion of cells from differ-
than 25 percent of the patients with severe aplastic anemia died within 4 ent genetic subgroups (complementation groups), whereas hybrids that
months of diagnosis; half succumbed within 1 year.212,213 Marrow trans- still show the sensitivity are the result of fusion of cells from the same
plantation is curative for approximately 80 to 90 percent of patients subgroup. Because one can determine the complementation group
younger than 20 years of age, approximately 70 percent if between the without knowing the gene involved, this approach is the first step in
ages of 20 and 40 years, and approximately 50 percent if older than age 40 understanding the genetic basis of a disease. Once the genes are known,
years.151,214 Unfortunately, as many as 40 percent of transplant survivors one does not need to use cell fusion studies; rather, retroviral vectors
suffer the deleterious consequences of chronic graft-versus-host dis- can be used to insert corrected genes into the cells.
ease,151 and the risk of subsequent cancer can be as high as 10 percent in The complementation groups have been designated FANCA, B, C,
older patients or after immunotherapy prior to hematopoietic stem cell D1, D2, E, F, G, I, J, L, M, and N. Table 347 lists the gene mutations cor-
transplantation.215 The best outcomes occur in those patients who have responding to these complementation groups. The great majority of
an allele-based HLA-matched sibling, have not been exposed to immuno- patients have mutations of FANCA, FANCC, or FANCG.222 It has been
suppressive therapy prior to transplantation, have not been exposed and proposed that the A and C gene products, which are cytoplasmic pro-
sensitized to blood cell products, have had marrow rather than a blood teins, form an FA core complex with the products of genes B, E, F, G,
stem cell donor product, and have not been subjected to high-dose radia- L, and M, which are adaptors or phosphorylators.222,223 The complex
tion in the conditioning regimen for transplantation.151,215217 translocates to the nucleus, where it is required for the ubiquitylation of
Combination immunosuppressive therapy with ATG and cyclosporine FANCD2 and protects the cell from DNA cross-linking and participates
leads to a marked improvement in approximately 70 percent of the in DNA repair (Fig. 344). DNA damage initiates activation of the
patients. Although some patients have normal blood counts, many con- FA/BRCA pathway and ubiquitylation of FANCD2, which is targeted to
tinue with moderate anemia or thrombocytopenia. In as many as 40 per- the altered DNA and facilitates repair by interacting with DNA repair
cent of patients initially responding to immunosuppressive therapy, their proteins, BRCA1, FANCD1/BRCA2, FANCN/PALB2, and RAD51. In
disease may relapse or progress over 10 years to paroxysmal nocturnal the presence of a mutant gene product, normal function is disturbed
hemoglobinuria, a myelodysplastic syndrome, or acute myelogenous leu- leading to damaging effects in sensitive tissues, including hematopoietic
kemia.162169,195197 Moreover, the beneficial effects of immunotherapy cells. In addition to the genetic defects leading to DNA instability and an
are often lost 10 years after treatment. In 168 transplanted patients the inability to repair DNA, TNF- and - are overexpressed in the marrow
actuarial survival at 15 years was 69 percent, and in 227 patients receiving of Fanconi anemia patients.224 The excess TNF- may play a role in the
immunosuppressive therapy it was 38 percent.162 suppression of erythropoiesis in these patients.
Treatment with high-dose cyclophosphamide produces early results
similar to that seen with the combination of ATG and cyclosporine.218 Clinical Features
However, cyclophosphamide has greater early toxicity and slower Growth retardation results in short stature, and skeletal anomalies are
hematologic recovery, but may generate more durable remissions. Its common. Absent, misshapen, or supernumerary thumbs and dysplastic
use has been too limited to reach a firm conclusion on its relative mer- radii occur in half the patients. Hip and vertebral abnormalities also
its and it is rarely used as first choice immunotherapy. may occur. Septal heart defects, eye abnormalities, and absent, mis-
shapen, or fused kidneys may be present. Learning disability is fre-
quent, and microcephaly and mental retardation may be a feature.
HEREDITARY APLASTIC ANEMIA Hypogonadism also may be evident. The skin may be generally hyper-
pigmented or may have areas of abnormal skin pigmentation referred
FANCONI ANEMIA to as caf-au-lait spots, which are flat, light brown, and from 1 to 12
centimeters in diameter. Hepatosplenomegaly is not a feature of the
Definition and History disease. Some patients have no or minor phenotypic abnormalities and
Fanconi anemia is the most common form of constitutional aplastic may be diagnosed as a result of the onset of marrow failure or a cancer
anemia and was initially described in three brothers by Fanconi in involving any of many sites as late as the fifth decade of life.
1927.219 It is inherited as an autosomal recessive condition that results The onset of marrow failure is gradual and usually is evident during
from defects in genes that modulate the stability of DNA. the last half of the first decade of life. The manifestations of anemia,
including weakness, fatigue, and dyspnea on exertion, and of thrombo-
Epidemiology cytopenia with epistaxis, purpura, or other unexpected bleeding, are
the principal findings. Hematologic and visceral manifestations are
Fanconi Anemia is an uncommon disorder and is estimated to be combined eventually in more than a third of patients, but some may
present in 1 in 1 million individuals. It is far more frequent in Afrikaners have cytopenias and inconspicuous somatic changes, whereas others
of European descent.220 This unusually high frequency has been attrib- may have somatic anomalies with no or a nominal disorder of blood
uted to a founder effect. cell formation for months or years. Some who carry the gene may be
virtually unaffected.225227 In a review of the more than 1300 patients in
Etiology and Pathogenesis the literature, 100 patients or fewer than 7 percent without anomalies
Thirteen complementation groups, defined by somatic cell hybridiza- were identified by chromosome breakage studies (see Laboratory Fea-
tion, are associated with the development of Fanconi anemia.221 A tures below) because of affected siblings.227 In the past, children in
476 PART VI: The Erythrocyte

TABLE 347. Gene Mutations Found in Fanconi Anemia


Gene Chromosome Location % of Patients Inheritance Protein Function
FANCA 16q24.3 ~65* AR FA Core Complex
FANCB Xp22.31 rare XLR FA Core Complex
FANCC 9q22.3 ~10 AR FA Core Complex
FANCDI (BRCA2) 13q12.3 rare AR RAD51 Recruitment
FANCD2 3p25.3 rare AR Monoubiquitinated protein
FANCE 6p21.3 ~10 AR FA Core Complex
FANCF 11p15 rare AR FA Core Complex
FANCG (XRCC9) 9p13 ~10 AR FA Core Complex
FANCI (KIAA1794) 15q2526 rare AR Monoubiquitination of FANCD2
FANCJ (BACH1/BRIP1) 17q22.3 rare AR 5 to 3 DNA helicase/ATPase
FANCL (PHF9/ POG) 2q16.1 rare AR FA Core Complex, E3 Ubiquitin ligase
FANCM (Hef) 14q21.3 rare AR FA Core Complex, ATPase/translocase, DNA helicase motifs
FANCN (PALB2) 16q12.1 rare AR Regulation of BRCA2 localization
To be identified ~5

AR, autosomal recessive; ATPase, adenosine triphosphatase; XLR, X-linked recessive.


*There are more than 100 mutant FANCA alleles, approximately 40% of which are large intragenic deletions. This table was made using material from references 221223.

Fanconi families with an onset of aplastic anemia without congenital phocytes show sensitivity to diepoxybutane, are considered to have
somatic abnormalities were thought to have a different disorder termed Fanconi anemia without skeletal abnormalities.
Estren-Dameshek syndrome.228 However, these children, whose lym-
Laboratory Features
Ub Blood counts and marrow cellularity are often normal until 5
DNA Ub to 10 years of age, when pancytopenia develops over an
damage AGBL D2 extended interval. Macrocytosis with anisocytosis and poikilo-
CF I
cytosis may be present before any cytopenia occurs. Thrombo-
ME Ub Ub
ATR cytopenia may precede the development of granulocytopenia
activation D2 I
and anemia. The marrow becomes hypocellular, and in vitro
D2
BRCAI colony assays reveal a decrease in CFU-GM and BFUE.227
I BRCA2
Stalled ATM Random chromatid breaks are present in myeloid cells,
(D1)
replication RAD51
BRIPI (J) lymphocytes, and chorionic villus biopsy samples. This chro-
PALB2 (N)
fork mosome damage is intensified after exposure to DNA cross-
P I linking agents such as mitomycin C or diepoxybutane. The
D2 P
hypersensitivity of the chromosomes of marrow cells or lym-
DNA repair phocytes to the latter agent is used as a diagnostic test for this
Checkpoint condition. Cell-cycle progression is prolonged at the G2 to M
response transition, and the cells are more susceptible to oxygen toxicity
Genomic stability when cultured in vitro. It is important to test the lymphocytes
from pediatric patients with aplastic anemia for sensitivity to
diepoxybutane, because therapy for Fanconi anemia differs
from that used for acquired aplastic anemia.
In the near future, clinical laboratories will be able to geno-
FIGURE 344. Representation of the FA/BRCA pathway. Following DNA damage when a replica-
type suspected patients. Determining the specific gene muta-
tion fork encounters a DNA cross-link, ATR (ataxia telangiectasia and rad3-related protein) is activated.
tion responsible in a patient (see Table 347) is important
This leads to the activation of the FA pathway as well as cell-cycle checkpoint activation via the ATM
because it confirms the diagnosis, identifies the genotype
(Ataxia Telangiectasia Mutated) protein. Activation of the FA pathway leads to the formation of the FA
linked to BRCA2 that may predispose to a cancer (e.g. breast,
core complex (consisting of the FA proteins A, B, C, E, F, G, L, and M). This activated FA core complex
ovary), and permits carrier detection.229
leads to the monoubiquitination of FANCD2 (FANCD2-Ub) and FANCI (I-Ub). The I-Ub/FANCD2-Ub
complex is then targeted to the chromatin containing the cross link where it interacts with BRCA2 and
possibly other DNA repair proteins (e.g., RAD51, J, N) leading to the repair of the DNA damage. Pro- Differential Diagnosis
teins mutated in the different FA subtypes are shown in yellow. (Reproduced from reference 221 with The differential diagnosis of Fanconi anemia includes other
permission of Elsevier.) causes of aplastic anemia, particularly those familial syndromes
CHAPTER 34: Aplastic Anemia: Acquired and Inherited 477

TABLE 348. Other Rare Syndromes Associated with Aplastic Anemia


Disorder Findings Inheritance Mutated Gene References
Ataxia-pancytopenia Cerebellar atrophy and ataxia; aplastic pancytopenia; AD Unknown 256258
(myelocerebellar disorder) monosomy 7; increased risk of AML
Congenital amegakaryocytic Thrombocytopenia; absent or markedly decreased marrow AR (compound MPL 259, 260
thrombocytopenia megakaryocytes; hemorrhagic propensity; elevated thrombopoietin; heterozygotes)
propensity to progress to aplastic pancytopenia; propensity to evolve
to clonal myeloid disease
DNA ligase IV deficiency Pre- and postnatal growth delay; dysmorphic facies; aplastic AR (compound LIG4 261263
pancytopenia heterozygotes)
Dubowitz syndrome Intrauterine and post-partum growth failure; short stature; AR Unknown 264, 265
microcephaly; mental retardation; distinct dysmorphic facies;
aplastic pancytopenia; increased risk of AML and ALL
Nijmegen breakage syndrome Microcephaly; dystrophic facies; short stature; immunodeficiency; AR NBS1 266, 267
radiation sensitivity; aplastic pancytopenia; predisposition to
lymphoid malignancy
Reticular dysgenesis (type of severe Lymphopenia; anemia and neutropenia; corrected by hematopoietic XLR Unknown 268, 269
immunodeficiency syndrome) stem cell transplantation
Seckel syndrome Intrauterine and post- partum growth failure; microcephaly; AR ATR (and 270273
characteristic dysmorphic facies (bird-headed profile); aplastic RAD3-related
pancytopenia; ? increased risk of AML gene); PCNT
WT syndrome Radial/ulnar abnormalities; aplastic pancytopenia; increased risk of AML AD Unknown 274

AD, autosomal dominant; ALL, acute lymphocytic leukemia; AML, acute myelogenous leukemia; AR, autosomal recessive; XLR, X-linked recessive.
NOTE:The listed clinical findings in each syndrome are not comprehensive. The designated clinical findings may not be present in all cases of the syndrome. Isolated cases of familial aplastic
anemia with or without associated anomalies that are not consistent with Fanconi anemia or other defined syndromes have been reported.227

associated with skeletal anomalies and other dysmorphic features. exams in females, hepatic ultrasonography to detect adenomas, and
Other familial types of aplastic anemia have been reported with or careful oropharyngeal examinations. Therapy of cancer in patients with
without associated anomalies. In those instances in which no sensitivity Fanconi anemia needs to consider the marked sensitivity of their cells
to DNA damaging agents is observed, the syndrome does not represent to DNA cross-linking agents and radiotherapy.
Fanconi anemia. Several uncommon syndromes of this type are Normal cDNA has been transferred into cells from patients with res-
described below and are tabulated in Table 348. toration of resistance to DNA damaging agents.236,237 Difficulties in this
approach include the paucity of stem cells in these patients, as well as
Therapy and Course the potential toxicity of the gene transfer methodology.
Most patients with Fanconi anemia do not respond to ATG or cyclo-
sporine but do improve with androgen preparations, often for as long
as several years. Cytokines may provide some improvement in blood
DYSKERATOSIS CONGENITA
counts, but their effect may wane. Studies in a mouse model also sug- Definition
gest that cytokine effects may not be sustained.230 The cumulative
This inherited disorder is characterized by cutaneous and mucous mem-
median survival is about 20 years from progressive marrow failure,
brane abnormalities, progressive marrow insufficiency, and a predisposi-
conversion to myelodysplastic syndrome, acute myelogenous leukemia
tion to malignant transformation. It is much more common in males
(approximately 10 percent of patients), or the development of a variety
than in females, and occurs in about 1 per 1 million population.221,238
of other cancers such as genitourinary system, digestive system (espe-
cially liver), head and neck.231 Multiple cancers in an individual patient
also occur. Cancers may occur as late as the fifth decade of life and pre- Pathogenesis
cede the diagnosis of Fanconi anemia in 25 percent of patients.231 The Dyskeratosis usually is inherited as a recessive X-chromosomelinked
presence of a clonal cytogenetic abnormality or marrow morphology disorder although rare cases can have autosomal dominant or autoso-
consistent with myelodysplasia markedly reduces the 5-year survival.232 mal recessive inheritance (Table 349). The disease is a reflection of
Allogeneic hematopoietic stem cell transplantation is curative for the telomere complex dysfunction,239,240 and it results from defective telomer-
marrow manifestations of Fanconi anemia.232235 A marked reduction ase activity resulting from mutations in the telomerase-related genes (Fig.
in dosage of the marrow-conditioning regimen of cyclophosphamide 345).240 The telomerase complex maintains the length of telomeres,
and radiation is necessary owing to the undue sensitivity of the tissues which are nucleotide tandem repeat structures residing at the termini of
to DNA-damaging exposures. The risk of cancer is so high that, where eukaryotic chromosomes (e.g., 5-TTAGGG-3). Telomerase restores the
practical, surveillance should be used, for example, frequent pelvic G-rich telomere repeats that are lost as a result of end-processing during
478 PART VI: The Erythrocyte

TABLE 349. Gene Mutations in Dyskeratosis Congenita


Gene Chromosome Location % of Patients Inheritance Protein Function
DKC1 Xq28 30 XLR Essential part of snoRNPs and telomerase
TERC 3q26 <5 AD RNA 3 end processing and stability
TERT 5p15.33 <6 AD, AR Reverse transcriptase component of telomerase
NOP10 (NOLA3) 15q14-q15 <1 AR RNA binding
TINF2 14q11.2 11 AD ? Binds to TRF1 to regulate telomere length
To be identified ~50%

AD, autosomal dominant; AR, autosomal recessive; XLR, X-linked recessive.


NOTE: Table prepared from data in references 221, 223, and 273. Percent of patients is approximate because of continuing identification of mutations.

normal cell division. Combined with protein, located at the ends of sites of DNA damage, preventing their otherwise inappropriate process-
chromosomes, they maintain chromosome integrity by preventing end- ing. Recessive mutations in NOP10, which encodes small ribonucleopro-
to-end chromosome fusion, preventing chromosome degradation, and teins associated with the telomerase complex, have been described in a
preventing chromosome instability. consanguineous family.241 Homozygous recessive mutations in the telo-
In dyskeratosis congenita, the telomeres are markedly shortened merase reverse transcriptase (TERT) produce a severe variant, referred to
resulting in genomic instability and cell (including marrow cell) apopto- as the Hyeraal-Hreidarsson syndrome.275
sis. Rapidly proliferating cells are at highest risk for dysfunction. Muta-
tions of the DKC1 gene are responsible for the X-linked recessive form. Clinical Findings
DKC1 encodes dyskerin, which is a conserved multifunctional protein The cutaneous findings usually appear after 5 years of age and include
component of the telomerase complex. Mutations of the TERT, TERC, reticulated, tan to gray, hyperpigmented and hypopigmented cutaneous
and TINF2 genes are the principal abnormalities in the autosomal domi- macules; alopecia of scalp, eyelashes, and eyebrows; adermatoglyphia
nant form. TERC is the RNA component of the telomerase reverse tran- (loss of dermal ridges on fingers and toes); hyperkeratosis of palms and
scriptase that TERT, the reverse transcriptase, uses to synthesize the 6-bp soles; mucosal leukoplakia in 75 percent of patients; and dystrophic
repeats on the 3 end of telomeric DNA. TINF2 is a component of the nails in more than 85 percent of patients.221,238,239 Other mucosal sites,
shelterin complex. The latter permits the distinction of telomeres from such as conjunctiva, lacrimal duct, esophagus, urethra, vagina, and
anus, can be involved, sometimes with stenosis and,
for example, dysphagia or dysuria. Pulmonary vascu-
lar involvement occurs in a significant minority of
TERT: heterozygous TERC: heterozygous affected children. Aplastic anemia usually develops in
mutations in AA, a mutations in ADDC, AA, late childhood or early adulthood and is evident in the
disease resembling MDS, PNH, and PF classical blood and marrow findings described under
ADDC and PF
acquired aplastic anemia, above. Female carriers of X-
Homozygous
linked dyskeratosis congenital may have slight abnor-
Dyskerin: hemizygous malities such as a dystrophic nail, a single area of
mutations in classical
mutations in X-linked DC hypopigmentation, or slight leukoplakia.238
ARDC and ARHH
and X-linked HH

Diagnosis
The dyskerin complex:
also involved in The diagnosis results from the combination of pheno-
GAR1 processing ribosomal typic findings and blood cell deficiencies. Genetic
and small nuclear RNAs analysis for telomerase complex gene mutations
should be used to confirm the clinical conclusion.
NHP2 NOP10: homozygous Shortened telomere length in leukocytes also can be
mutation in ARDC assessed by flow cytometric fluorescence in situ
hybridization studies.242
FIGURE 345. Representation of the interaction between dyskerin and the other molecules (GAR1, NHP2,
NOP10, TERC, and TERT) of the telomerase complex (and their association with different disease categories).
Telomerase is an RNA-protein complex because TERC is an RNA molecule that is never translated. The other Management
molecules (dyskerin, GAR1, NHP2, NOP10 and TERT) are proteins. The minimal active telomerase enzyme is Stem cell hematopoietic transplantation has had
composed of two molecules each of TERT, TERC, and dyskerin. Dyskerin, GAR1, NHP2, and NOP10 are impor- inconsistent results because of frequent and severe
tant for the stability of the telomerase complex. AA, aplastic anemia; ADDC, autosomal dominant dyskerato- posttransplantation complications.243 Nonmyeloabla-
sis congenita; ARDC, autosomal recessive dyskeratosis congenita; ARHH, autosomal recessive Hyeraal- tive transplantation might improve results.244,245 Trans-
Hreidarsson syndrome; MDS, myelodysplasia; PNH, paroxysmal nocturnal hemoglobinuria; PF, pulmonary plantation might improve the cytopenias but not the
fibrosis; X-linked DC, X-linked dyskeratosis congenita; X-linked HH, X-linked Hyeraal-Hreidarsson syndrome. abnormalities of other organs or the frequency of sec-
(Reprinted from reference 221 with permission of Elsevier.) ondary nonhematopoietic cancer.
CHAPTER 34: Aplastic Anemia: Acquired and Inherited 479

Course and Prognosis and cytopenias can be corrected with allogeneic hematopoietic stem
The incidence of squamous cell carcinoma of mucosal sites is increased cell transplantation.253
and they often originate in sites of leukoplakia in the skin, gastrointesti-
nal, or genitourinary tracts. These usually develop between the ages of Course and Prognosis
20 and 30 years. Mortality from neutropenic infection or thrombocyto- Death from overwhelming sepsis is common. These patients, especially
penic hemorrhage occurs in about two-thirds of patients with aplastic males, have a significant risk of progression to a myelodysplastic syn-
anemia. Median survival is about 30 years. drome or acute myelogenous leukemia.248,254,255 Survival is a function
of the severity of the cytopenias. If the cytopenias are mild, survival is
not uncommon into the fourth or fifth decade of life. If symptomatic
SHWACHMAN-DIAMOND SYNDROME pancytopenia, especially neutropenia, is present, median survival is
Definition about 20 to 30 years.248,255
An uncommon inherited disorder that is estimated to occur once in
every 100,000 births, manifesting exocrine pancreatic insufficiency OTHER INHERITED APLASTIC ANEMIAS
with secondary steatorrhea, blood cell deficiencies, and skeletal abnor-
Several other rare syndromes are associated with aplastic pancytopenia,
malities. It was first described in 1964.247,249
and these are described in Table 348. Congenital amegakaryocytic
thrombocytopenia results from mutations in the thrombopoietin
Pathogenesis receptor gene, MPL.259,260 Reticular dysgenesis results from a pluripo-
Shwachman-Diamond syndrome results from mutations in the SBDS tential stem cell defect as both lymphoid and myeloid progenitors are
gene on chromosome 7q11, which induces accelerated cellular apoptosis affected.268,269 The Seckel syndrome results from mutations in the ATR
via the FAS pathway.249 The resulting hyperproliferation may account for gene, and marrow cells exhibit heightened sister chromatid exchange.270273
the abnormal telomere shortening that has been documented in the leu- The ataxia-telangiectasia mutated and rad3-related (ATR) kinase
kocytes in this condition.250 The pathogenetic mechanism that (1) pre- orchestrates cellular responses to DNA damage and replication stress.
vents development of pancreatic acinar cells, (2) results in abnormal bone The genetic basis of marrow failure in these four syndromes that
morphogenesis, and (3) causes marrow impairment of blood cell produc- involve aplastic pancytopenia is not yet known, but may be related to
tion is not understood. SBDS knockdown in experimental animals affects defects in the ATR-dependent DNA damage-repair pathway.259 Most of
expression of genes involved in brain, bone, and marrow development, these syndromes can be treated by marrow transplantation, but this
and may be the result of the genes role in RNA processing.251,252 The step, if successful, does not correct somatic abnormalities, only the
mutations also result in abnormalities in neutrophil motility and chemo- hematopoietic defect. The restoration of robust hematopoiesis by trans-
taxis, but pus formation in vivo seems adequate. plantation may decrease their propensity to undergo clonal evolution to
a clonal myeloid or, in some cases, lymphoid disorder.
Clinical Findings
Pancreatic insufficiency, steatorrhea, and neutropenia are present in
most patients at the time of diagnosis.247,248 Pallor may reflect anemia REFERENCES
and easy bruising; epistaxis or bleeding from other sites reflect throm- 1. The International Agranulocytosis and Aplastic Anemia Study. Incidence of aplastic
bocytopenia. Neutropenia occurs in approximately 95 percent, ane- anemia: The relevant diagnostic criteria. Blood 70:1718, 1987.
mia in approximately 50 percent, and thrombocytopenia in 2. Ehrlich P: ber einen Fall von Anamie mit Bemerkungen ber regenerative Veran-
derungen des Knochenmarks. Charite Ann 13:300, 1888.
approximately 35 percent of patients.249 Thus, a substantial plurality 3. Chauffard M: Un cas danmie pernicieuse aplastique. Bull Soc Med Hop Paris
of patients has bicytopenia or tricytopenia with an hypoplastic mar- 21:313, 1904.
row. Fetal hemoglobin levels are elevated in approximately 75 percent 4. Scott JL, Cartwright GE, Wintrobe MM: Acquired aplastic anemia: an analysis of
thirty-nine cases and review of the pertinent literature. Medicine (Baltimore) 38:119,
of the patients, perhaps secondary to erythroid hypoplasia. Cytoge- 1959.
netic abnormalities involving chromosomes 7 and 20 have been 5. Thomas ED, Storb R, Fefer A, et al: Aplastic anemia treated by bone marrow trans-
described in marrow cells. Nutritional inadequacies related to intesti- plantation. Lancet 1: 284, 1972.
6. Thomas ED, Storb R, Giblett B et al: Recovery from aplastic anemia following
nal malabsorption result in a failure to thrive. Short stature is charac-
attempted marrow transplantation. Exp Hematol 4:97, 1976.
teristic. Skeletal abnormalities are present in most patients, notably 7. Champlin RE, Feig SA, Sparkes RS, Gale RP: Bone marrow transplantation from
osteopenia, but also syndactyly, supernumerary metatarsals, coax vera identical twins in the treatment of aplastic anaemia: Implication for the pathogenesis
deformity, and dental enamel defects and caries. Delayed puberty is of the disease. Br J Haematol 56:455, 1984.
8. Speck B, Gluckman E: Treatment of aplastic anemia by antilymphocyte globulin with
common. The neutropenia and chemotactic abnormality may result and without allogeneic bone marrow infusions. Lancet II:11451148, 1977.
in recurrent infections, including sinusitis, otitis, pneumonia, osteo- 9. Mary JY, Baumelou E, Guiguet M: Epidemiology of aplastic anemia in France: A pro-
myelitis, and others. Pancreatic cell lipase production improves with spective multicenter study. Blood 75:1646, 1990.
10. Montan E, Ibez L, Vidal X, et al: Epidemiology of aplastic anemia: A prospective
age, and as many as half the patients may have improvement in lipid multicenter study. Haematologica 93:518, 2008.
absorption in the small bowel with time. 11. Maluf EM, Pasquini R, Eluf JN, et al: Aplastic anemia in Brazil: Incidence and risk
factors. Am J Hematol 71:268, 2002.
12. McCahon E, Tang K, Rogers PC, McBride ML, Schultz KR: The impact of Asian
Management descent on the incidence of acquired severe aplastic anaemia in children. Br J Hae-
Supportive care, particularly with supplemental pancreatic enzymes, to matol 121:170, 2003.
13. Chongli Y and Ziaobo Z: Incidence survey of aplastic anemia in China. Chin Med Sci
provide proper nutrition, and appropriate and prompt treatment of J 6:203, 1991.
bacterial infections with antibiotics is important. Many agents, includ- 14. Issaragrisil S: Epidemiology of aplastic anemia in Thailand. Thai Aplastic Anemia
ing G-CSF, glucocorticoids, pancreatic extract, vitamins, have been Study Group. Int J Hematol 70:137, 1999.
15. Yong AS, Goh AS, Rahman M, et al: Epidemiology of aplastic anemia in the state of
tried to improve the neutropenia with erratic results. Some agents have
Sabah, Malaysia. Med J Malaysia 53:59, 1998.
potential risks, such as G-CSF fostering clonal evolution and glucocor- 16. Issaragrisil S, Kaufman DW, Anderson T, et al: The epidemiology of aplastic anemia
ticoids fostering immunodeficiency. Severe hematopoietic dysfunction in Thailand. Blood 107:1299, 2006.
480 PART VI: The Erythrocyte

17. Young NS, Kaufman DW: The epidemiology of acquired aplastic anemia. Haemato- 51. Williams DM: Pancytopenia, aplastic anemia and pure red cell aplasia, in Wintrobes
logica 93:489, 2008. Clinical Hematology, 10th ed, edited by GR Lee, J Foerster, J Lukens, et al, pp 1452
18. Yin SN, Hayes RB, Linet MS, et al: A cohort study of cancer among benzene-exposed 1459. Williams & Wilkins, Baltimore, 1999.
workers in China: overall results. Am J Ind Med 29:227, 1996. 52. Tonkonow B, Hoffman R: Aplastic anemia and cimetidine. Arch Intern Med
19. Kagan WA, Ascensao J, Pahwa R, et al: Aplastic anemia: presence in human bone 140:1123, 1980.
marrow of cells that suppress myelopoiesis. Proc Natl Acad Sci U S A 73:2890, 1976. 53. Volkin RL, Shadduck RK, Winkelstein A, et al: Potentiation of carmustine-cranial-irra-
20. Maciejewski JP, Anderson S, Katevas P, Young NS: Phenotypic and functional analy- diation-induced myelosuppression by cimetidine. Arch Intern Med 142:243, 1982.
sis of bone marrow progenitor cell compartment in bone marrow failure. Br J Hae- 54. Khan HA: Benzene toxicity: A consolidated short review of human and animal stud-
matol 87:227, 1994. ies. Hum Exp Toxicol 26:677, 2007.
21. Scopes J, Bagnara M, Gordon-Smith EC, et al: Haemopoietic progenitor cells are 55. Smith MT: Overview of benzene-induced aplastic anemia. Eur J Haematol 60:107,
reduced in aplastic anaemia. Br J Haematol 86:427, 1994. 1996.
22. Maciejewski JP, Selleri C, Sato T, et al: A severe and consistent deficit in marrow and 56. Snyder R: Benzene and leukemia. Crit Rev Toxicol 32:155, 2002.
circulating primitive hematopoietic cells (long-term culture-initiating cells) in 57. Fleming LE, Timmeny MA: Aplastic anemia and pesticides. An etiologic associa-
acquired aplastic anemia. Blood 88:1983, 1996. tion? J Occup Med 35:1106, 1993.
23. Young NS, Scheinberg P, Calado RT: Aplastic anemia. Curr Opin Hematol 15:162, 58. Rugman FP, Cosstick R: Aplastic anaemia associated with organochlorine pesticide:
2008. Case reports and review of evidence. J Clin Pathol 43:98, 1990.
24. Young NS, Calado RT, Scheinberg P: Current concepts in the pathophysiology and 59. Muir KR, Chilvers CE, Harriss C, et al: The role of occupational and environmental
treatment of aplastic anemia. Blood 108:2509, 2006. exposures in the aetiology of acquired severe aplastic anaemia: A case control inves-
25. Young NS, Maciejewski J: Mechanisms of disease: The pathophysiology of acquired tigation. Br J Haematol 123:906, 2003.
aplastic anemia. N Engl J Med 336:1365, 1997. 60. Valdez Salas B, Garcia Duran EI, Wiener MS: Impact of pesticides use on human
26. Laver J, Castro-Malaspina H, Kernan NA, et al: In vitro interferon-gamma produc- health in Mexico: A review. Rev Environ Health 15:399, 2000.
tion by cultured T-cells in severe aplastic anaemia: Correlation with granulomonop- 61. Ahamed M, Anand M, Kumar A, Siddiqui MK: Childhood aplastic anaemia in Luc-
oietic inhibition in patients who respond to anti-thymocyte globulin. Br J Haematol know, India: Incidence, organochlorines in the blood and review of case reports fol-
69:545, 1988. lowing exposure to pesticides. Clin Biochem 39:762, 2006.
27. Gascon P, Zoumbos NC, Scala G, et al: Lymphokine abnormalities in aplastic ane- 62. Rauch AE, Kowalsky SF, Lesar TS, et al: Lindane (Kwell)-induced aplastic anemia.
mia: implications for the mechanism of action of antithymocyte globulin. Blood Arch Intern Med 150:2393, 1990.
65:407, 1985. 63. Roberts HJ: Pentachlorophenol-associated aplastic anemia, red cell aplasia, leukemia
28. Hinterberger W, Adolf G, Bettelheim P, et al: Lymphokine overproduction in severe and other blood disorders. J Fla Med Assoc 77:86, 1990.
aplastic anemia is not related to blood transfusions. Blood 74:2713, 1989. 64. Prager D, Peters C: Development of aplastic anemia and the exposure to Stoddard
29. Shinohara K, Ayame H, Tanaka M, et al: Increased production of tumor necrosis fac- solvent. Blood 35:286, 1970.
tor alpha by peripheral blood mononuclear cells in the patients with aplastic anemia. 65. Powers D: Aplastic anemia secondary to glue sniffing. N Engl J Med 273:700, 1965.
Am J Hematol 37:75, 1991. 66. Kirtadze I, Zurabashvili D: Study of chemical composition of glue RAZI used by
30. Nistico, A, Young, NS: Gamma-interferon gene expression in the bone marrow of solvent abusers in Tbilisi. Georgian Med News 133:65, 2006.
patients with aplastic anemia. Ann Intern Med 120:463, 1994. 67. Sabbioni G, Sepai O, Norppa H, et al: Comparison of biomarkers in workers exposed
31. Zoumbos N, Gascon P, Djeu J, Young NS: Interferon is a mediator of hematopoietic to 2,4,6-trinitrotoluene. Biomarkers 12:21, 2007.
suppression in aplastic anemia in vitro and possibly in vivo. Proc Natl Acad Sci U S A 68. Crawford MAD: Aplastic anaemia due to trinitrotoluene intoxication. Br Med J
82:188, 1985. 2:430, 1954.
32. Sloand E, Kim S, Maciejewski JP, et al: Intracellular interferon-gamma in circulating 69. Ajlouni K, Doeblin TD: The syndrome of hepatitis and aplastic anaemia. Br J Hae-
and marrow T cells detected by flow cytometry and the response to immunosup- matol 27:345, 1974.
pressive therapy in patients with aplastic anemia. Blood 100:1185, 2002. 70. Hagler L, Pastore RA, Bergin JJ: Aplastic anemia following viral hepatitis: Report of 2
33. Solomou EE, Keyvanfar K, Young NS: T-bet, a Th1 transcription factor, is up-regu- fatal cases and literature review. Medicine (Baltimore) 54:139, 1975.
lated in T cells from patients with aplastic anemia. Blood 107:3983, 2006. 71. Pol S, Driss F, Devergie A, et al: Is hepatitis C virus involved in hepatitis-associated
34. Risitano AM, Maciejewski JP, Green S, et al: In vivo dominant immune responses in aplastic anemia? Ann Intern Med 113:435, 1990.
aplastic anaemia: Molecular tracking of putatively pathogenetic T-cell clones by TCR 72. Hibbs JR, Frickhofen N, Rosenfeld SJ, et al: Aplastic anemia and viral hepatitis: Non-
beta-CDR3 sequencing. Lancet 364:355, 2004. A, non-B, non-C? JAMA 267:2051, 1992.
35. Chen J, Ellison FM, Eckhaus MA, et al: Minor antigen h60-mediated aplastic anemia 73. Honkaniemi E, Gustafsson B, Fischler B, et al: Acquired aplastic anaemia in seven
is ameliorated by immunosuppression and the infusion of regulatory T cells. J Immu- children with severe hepatitis with or without liver failure. Acta Paediatr 96:1660,
nol 178:4159, 2007. 2007.
36. Hirano N, Butler MO, Von Bergwelt-Baildon MS, et al: Autoantibodies frequently 74. Tzakis AG, Arditi M, Whitington PF, et al: Aplastic anemia complicating orthotopic
detected in patients with aplastic anemia. Blood 102:4567, 2003. liver transplantation for non-A, non-B hepatitis. N Engl J Med 319:393, 1988.
37. Smick K, Condit PK, Proctor RL, Sutcher V: Fatal aplastic anemia: An epidemiologi- 75. Brown KE, Tisdale J, Barrett AJ, Dunbar CE, Young NS: Hepatitis-associated aplastic
cal study of its relationship to the drug chloramphenicol. J Chronic Dis 17:899, 1964. anemia. N Engl J Med 336:1059, 1997.
38. Modan B, Segal S, Shani M, Sheba C: Aplastic anemia in Israel: Evaluation of the eti- 76. Safadi R, Or R, Ilan Y et al: Lack of known hepatitis virus in hepatitis-associated
ological role of chloramphenicol on a community-wide basis. Am J Med Sci 270:441, aplastic anemia and outcome after bone marrow transplantation. Bone Marrow
1975. Transplant 27:183, 2001.
39. Yunis AA Chloramphenicol toxicity: 25 years of research. Am J Med 87:44N, 1989. 77. Mishra B, Malhotra P, Ratho RK, et al: Human parvovirus B19 in patients with aplas-
40. Custer RP: Aplastic anemia in soldiers treated with Atabrine (quinacrine). Am J Med tic anemia. Am J Hematol 79:166, 2005.
Sci 212:211, 1946. 78. Yetgin S, Cetin M, Ozyrek E, et al: Parvovirus B19 infection associated with severe
41. Best WR: Drug-associated blood dyscrasias. JAMA 185:286, 1963. aplastic anemia in an immunocompetent patient. Pediatr Hematol Oncol 21:223,
42. The International Agranulocytosis and Aplastic Anemia Study: Risks of agranulocy- 2004.
tosis and aplastic anemia: A first report of their relation to drug use with special ref- 79. Wong S, Young NS, Brown KE: Prevalence of parvovirus B19 in liver tissue: No asso-
erence to analgesics. JAMA 256:1749, 1986. ciation with fulminant hepatitis or hepatitis-associated aplastic anemia. J Infect Dis
43. Retsagi G, Kelly JP, Kaufman DW: Risk of agranulocytosis and aplastic anaemia in 187:1581, 2003.
relation to use of antithyroid drugs: International Agranulocytosis and Aplastic 80. Lazarus KH, Baehner RL: Aplastic anemia complicating infectious mononucleosis: A
Anaemia Study. BMJ 297:262, 1988. case report and review of the literature. Pediatrics 67:907, 1981.
44. International Agranulocytosis and Aplastic Anemia Study: Anti-infective drug use 81. Baranski B, Armstrong G, Truman JT, et al: Epstein-Barr virus in the bone marrow
in relation to the risk of agranulocytosis and aplastic anemia. Arch Intern Med of patients with aplastic anemia. Ann Intern Med 109:695, 1988.
149:1036, 1989. 82. Vinters HV, Mah V, Mohrmann R, Wiley CA: Evidence for human immunodefi-
45. Kelly JP, Kaufman DW, Shapiro S: Risks of agranulocytosis and aplastic anemia in ciency virus (HIV) infection of the brain in a patient with aplastic anemia. Acta Neu-
relation to the use of cardiovascular drugs: The International Agranulocytosis and ropathol 76:311, 1988.
Aplastic Anemia Study. Clin Pharmacol Ther 49:330, 1991. 83. Samuel D, Castaing D, Adam R, et al: Fatal acute HIV infection with aplastic
46. Kaufmann DW, Kelly JP, Jurgelon JM, et al: Drugs in the aetiology of agranulocytosis anaemia, transmitted by liver graft. Lancet 1:1221, 1988.
and aplastic anaemia. Eur J Haematol 57(Suppl):23, 1996. 84. Morales CE, Sriram I, Baumann MA: Myelodysplastic syndrome occurring as possi-
47. Baumelou E, Guiguet M, Mary JY, et al: Epidemiology of aplastic anemia in France: ble first manifestation of human immunodeficiency virus infection with subsequent
A case control study. I. Medical history and medication use. Blood 81:1471, 1993. progression to aplastic anaemia. Int J STD AIDS 1:55, 1990.
48. Bithell TC, Wintrobe MM: Drug-induced aplastic anemia. Semin Hematol 4:194, 85. Rosenfeld CS, Rybka WB, Weinbaum D, et al: Late graft failure due to dual bone
1967. marrow infection with variants A and B of human Herpesvirus-6. Exp Hematol
49. Williams DM, Lynch RE, Cartwright GE: Drug-induced aplastic anemia. Semin 23:626, 1995.
Hematol 10:195, 1973. 86. Pavithran K, Raji NL, Thomas M: Aplastic anemia complicating lupus erythemato-
50. Heimpel H, Heit W: Drug-induced aplastic anaemia. Clin Haematol 9:641, 1980. sisReport of a case and review of the literature. Rheumatol Int 22:253, 2002.
CHAPTER 34: Aplastic Anemia: Acquired and Inherited 481

87. Bailey FA, Lilly M, Bertoli LF, Ball GV: An antibody that inhibits in vitro bone mar- 121. Kojima S, Matsuyama T, Kodera Y, et al: Measurement of endogenous plasma granu-
row proliferation in a patient with systemic lupus erythematosus and aplastic ane- locyte colony-stimulating factor in patients with acquired aplastic anemia by a sensi-
mia. Arthritis Rheum 31:901, 1989. tive chemiluminescent immunoassay. Blood 87:1303, 1996.
88. Roffe C, Cahill MR, Samanta A, et al: Aplastic anaemia in systemic lupus erythema- 122. Kojima S, Matsuyama T, Kodera Y: Circulating erythropoietin in patients with
tosus: A cellular immune mechanism? Br J Rheumatol 30:301, 1991. acquired aplastic anaemia. Acta Haematol 94:117, 1995.
89. Sumimoto S, Kawai M, Kasajima Y, Hamamoto T: Aplastic anemia associated with 123. Emmons RVD, Reid DM, Cohen RL, et al: Human thrombopoietin levels are high
systemic lupus erythematosus. Am J Hematol 38:329, 1991. when thrombocytopenia is due to megakaryocyte deficiency and low when due to
90. Winkler A, Jackson RW, Kay DS, et al: High-dose intravenous cyclophosphamide increased platelet destruction. Blood 87:4068, 1996.
treatment of systemic lupus erythematosus-associated aplastic anemia [letter]. 124. Nakao S, Matsushima K, Young N: Deficient interleukin I production by aplastic
Arthritis Rheum 31:693, 1988. anaemia monocytes. Br J Haematol 71:431, 1989.
91. Kim SW, Rice L, Champlin R, Udden MM. Aplastic Anemia in eosinophilic fasciitis: 125. Holmberg LA, Seidel K, Leisenring W, Torok-Storb B: Aplastic anemia: Analysis of
Responses to immunosuppression and marrow transplantation. Haematologica stromal cell function in long-term marrow cultures. Blood 84:3685, 1994.
28:131, 1997. 126. Stute N, Fehse B, Schroder J et al: Human mesenchymal stem cells are not of donor
92. Debusscher L, Bitar N, DeMaubeuge J, et al: Eosinophilic fasciitis and severe aplastic origin in patients with severe aplastic anemia who underwent sex-mismatched allo-
anemia: Favorable response to either antithymocyte globulin or cyclosporin A in geneic bone marrow transplant. J Hematother Stem Cell Res 11:977, 2002.
blood and skin disorders. Transplant Proc 20:310, 1988. 127. Applebaum FR, Barrall J, Storb R, et al: Clonal cytogenetic abnormalities in patients
93. Kumar M and Goldman J: Severe aplastic anemia and Graves disease in a paediatric with otherwise typical aplastic anemia. Exp Hematol 15:1134, 1987.
patient. Br J Haematol 118:327, 2002. 128. Negendank W, Weissman D, Bey TM, et al: Evidence for clonal disease by magnetic res-
94. Tomonari A, Tojo A, Iseki T, et al: Severe aplastic anemia with autoimmune thyroidi- onance imaging in patients with hypoplastic marrow disorders. Blood 78:2872, 1991.
tis showing no hematological response to intensive immunosuppressive therapy. 129. Schrezenmeier H, Hertenstein B, Wagner B, et al: A pathogenetic link between aplas-
Acta Haematol 109:90, 2003. tic anemia and paroxysmal nocturnal hemoglobinuria is suggested by a high fre-
95. Aydin Y, Berker D, Ustn I, et al: A very rare cause of aplastic anemia: Graves dis- quency of aplastic anemia patients with a deficiency of phosphatidylinositol glycan
ease. South Med J 101:666, 2008. anchored proteins. Exp Hematol 23:81, 1995.
96. Lima CS, Zantut Wittmann DE, Castro V, et al: Pancytopenia in untreated patients 130. Horsley SW, Colman S, McKinley M, et al: Genetic lesions in a preleukemic aplasia
with Graves disease. Thyroid 16:403, 2006. phase in a child with acute lymphoblastic leukemia. Genes Chromosomes Cancer
97. Das PK, Wherrett D, Dror Y: Remission of aplastic anemia induced by treatment for 47:333, 2008.
Graves disease in a pediatric patient. Pediatr Blood Cancer 49:210, 2007. 131. Suzan F, Terr C, Garcia I, et al: Three cases of typical aplastic anaemia associated
98. Dincol G, Saka B, Aktan M, et al: Very severe aplastic anemia following resection of with a Philadelphia chromosome. Br J Haematol 112:385, 2001.
lymphocytic thymoma: effectiveness of antilymphocyte globulin, cyclosporine A and 132. Socie G, Rosenfeld S, Frickhofen N, et al: Late clonal diseases of aplastic anemia.
granulocyte-colony stimulating factor. Am J Hematol 64:78, 2000. Semin Hematol 37:91101 2000.
99. Ritchie DS, Underhill C, Grigg AP. Aplastic anemia as a late complication of thy- 133. Gordon-Smith EC, Marsh JC, Gibson FM: Views on the pathophysiology of aplastic
moma in remission. Eur J Haematol 68:389, 2002. anemia. Int J Hematol 76(Suppl 2):163, 2002.
100. Gaglia A, Bobota A, Pectasides E, et al: Successful treatment with cyclosporine of 134. Maciejewski JP, Risitano A, Sloand EM, et al: Distinct clinical outcomes for cytoge-
thymoma-related aplastic anemia. Anticancer Res 27:3025, 2007. netic abnormalities evolving from aplastic anemia. Blood 99:3129, 2002.
101. Trisal V, Nademanee A, Lau SK, Grannis FW Jr: Thymoma-associated severe aplastic 135. Socie G, Henryamar M, Bacigalupo A, et al: Malignant tumors occurring after treat-
anemia treated with surgical resection followed by allogeneic stem-cell transplanta- ment of aplastic anemia. N Engl J Med 329:1152, 1993.
tion. J Clin Oncol 25:3374, 2007. 136. Najean Y, Haguenauer O: Long-term (520 years) evolution of non-grafted aplastic
102. Arcasoy MO, Gockerman JP: Aplastic anaemia as an autoimmune complication of anemias. Blood 76:2222, 1990.
thymoma. Br J Haematol 137:272, 2007. 137. Ball SE, Gibson FM, Rizzo S: Progressive telomere shortening in aplastic anemia.
103. Park CY, Kim HJ, Kim YJ, et al: Very severe aplastic anemia appearing after thymec- Blood 91:3582, 1998.
tomy. Korean J Intern Med 18:61, 2003. 138. Rosse WF: New insights into paroxysmal nocturnal hemoglobinuria. Curr Opin
104. Abrams EM, Gibson IW, Blydt-Hansen TD: The concurrent presentation of minimal Hematol 8:61, 2001.
change nephrotic syndrome and aplastic anemia. Pediatr Nephrol 24:407, 2009. 139. Nakakuma H, Kawaguchi T: Pathogenesis of selective expansion of PNH clones. Int J
105. Aitchison RGM, Marsh JCW, Hows JM, et al: Pregnancy associated aplastic anaemia: Hematol 77:121, 2003.
A report of 5 cases and review of current management. Br J Haematol 73:541, 1989. 140. Storb R, Blume KG, ODonnell MR, et al: Cyclophosphamide and antithymocyte
106. Pajor A, Kelemen E, Szakacs Z, Lehoczky D: Pregnancy in idiopathic aplastic anemia globulin to condition patients with aplastic anemia for allogeneic marrow transplan-
(report of 10 patients). Eur J Obstet Gynecol Reprod Biol 45:19, 1992. tation: the experience in four centers. Biol Blood Marrow Transplant 7:39, 2001.
107. Bourantas K, Makrydimas G, Georgiou I, et al: Aplastic anemia: Report of a case 141. Metzgeroth G, Dinter D, Schultheis B, et al: Deferasirox in MDS patients with trans-
with recurrent episodes in consecutive pregnancies. J Reprod Med 42:672, 1997. fusion-caused iron overload-a phase-II study. Ann Hematol 88:301, 2009.
108. Kwon JY, Lee Y, Shin JC, et al: Supportive management of pregnancy-associated 142. Sagmeister M, Oec L, Gmur J: A restrictive platelet transfusion policy allowing long-
aplastic anemia. Int J Gynaecol Obstet 95:115, 2006. term support of outpatients with severe aplastic anemia. Blood 93:3124, 1999.
109. Thakral B, Saluja K, Sharma RR, et al: Successful management of pregnancy-associ- 143. Lawrence JB, Yomtovian RA, Hammons T, et al: Lowering the prophylactic platelet
ated severe aplastic anemia. Eur J Obstet Gynecol Reprod Biol 131:244, 2007. transfusion threshold: a prospective analysis. Leuk Lymphoma 41:67, 2001.
110. Tichelli A, Socie G, Marsh J et al: Outcome of pregnancy and disease course among 144. Zeigler ZR: Effects of epsilon aminocaproic acid on primary haemostasis. Haemosta-
women with aplastic anemia treated with immunosuppression. Ann Intern Med sis 21:313, 1991.
137:164, 2002. 145. Hod E, Schwartz J: Platelet transfusion refractoriness. Br J Haematol 142:348, 2008.
111. Court-Brown WM, Doll R: Leukaemia and aplastic anaemia in patients irradiated 146. Slichter SJ, Davis K, Enright H, et al: Factors affecting posttransfusion platelet incre-
for ankylosing spondylitis. 1957. J Radiol Prot 27:B15-B154, 2007. ments, platelet refractoriness, and platelet transfusion intervals in thrombocytopenic
112. Darby SC, Doll R, Gill SK, Smith PG: Long term mortality after a single treatment patients. Blood 105:4106, 2005.
course with x-rays in patients treated with ankylosing spondylitis. Br J Cancer 147. Drewniak A, Boelens JJ, Vrielink H, et al: Granulocyte concentrates: prolonged func-
55:179, 1987. tional capacity during storage in the presence of phenotypic changes. Haematologica
113. Johnson SAN, Bateman CJT, Beard MEJ, et al: Long-term haematological complica- 93:1058, 2008.
tions of Thorotrast. Q J Med 182:259, 1977. 148. Armand P, Antin JH: Allogeneic stem cell transplantation for aplastic anemia. Biol
114. Martland HS: The occurrence of malignancy in radioactive persons: A general Blood Marrow Transplant 13:505, 2007.
review of data gathered in the study of the radium dial painters, with special refer- 149. Georges GE, Storb R. Stem cell transplantation for aplastic anemia. Int J Hematol
ence to the occurrence of osteogeneic sarcoma and the inter-relationship of certain 75:141, 2002.
blood diseases. Am J Cancer 15:2435, 1931. 150. Champlin RE, Perez WS, Passweg JR, et al: Bone marrow transplantation for severe
115. Cronkite EP, Haley TJ: Clinical aspects of acute radiation injury, in Manual on Radi- aplastic anemia: A randomized controlled study of conditioning regimens. Blood
ation Haematology, pp 169173. International Atomic Energy Agency, Vienna, 1971. 109:4582, 2007.
116. Mettler FA Jr, Moseley RD Jr: Medical Effects of Ionizing Irradiation, pp 1185. Grune 151. Locasciulli A, Oneto R, Bacigalupo A, et al: Outcome of patients with acquired aplas-
and Stratton, New York, 1985. tic anemia given first line bone marrow transplantation or immunosuppressive treat-
117. Gale RP: USSR: Follow-up after Chernobyl. Lancet 1:401, 1990. ment in the last decade: A report from the European Group for Blood and Marrow
118. Nimer SD, Leung DHY, Wolin MJ, Golde DW: Serum stem cell factor levels in Transplantation (EBMT). Haematologica 92:11, 2007.
patients with aplastic anemia. Int J Hematol 60:185, 1994. 152. Viollier R, Soci G, Tichelli A, et al: Recent improvement in outcome of unrelated
119. Kojima S, Matsuyama T, Kodera Y: Plasma levels and production of soluble stem cell donor transplantation for aplastic anemia. Bone Marrow Transplant 41:45, 2008.
factor by marrow stromal cells in patients with aplastic anaemia. Br J Haematol 153. Lee SJ, Klein J, Haagenson M, Baxter-Lowe LA, et al: High-resolution donor-recipi-
99:440, 1997. ent HLA matching contributes to the success of unrelated donor marrow transplan-
120. Lyman SD, Seaberg M, Hanna R, et al: Plasma/serum levels of flt3 ligand are low in tation. Blood 110:4576, 2007.
normal individuals and highly elevated in patients with Fanconi anemia and 154. Dubey S and Nityanand S: Involvement of Fas and TNF pathways in the induction of
acquired aplastic anemia. Blood 86:4091, 1995. apoptosis of T cells by antithymocyte globulin. Ann Hematol 82:496, 2003.
482 PART VI: The Erythrocyte

155. Michallet M-C, Saltel F, Preville X, et al: Cathepsin-B-dependent apoptosis triggered 185. Gillio AP, Boulad F, Small TN, et al: Comparison of long-term outcome of children
by antithymocyte globulins: A novel mechanism of T-cell depletion. Blood 102:3719, with severe aplastic anemia treated with immunosuppression versus bone marrow
2003. transplantation. Biol Blood Marrow Transplant 3:18, 1997.
156. Mangan KF, DAlessandro L, Mullaney MT: Action of antithymocyte globulin on 186. De Planque MM, Kluin-Nelemans HC, Van Krieken HJM, et al: Evolution of
normal human erythroid progenitor cell proliferation in vitro: Erythropoietic acquired severe aplastic anaemia to myelodysplasia and subsequent leukaemia in
growth-enhancing factors are released from marrow accessory cells. J Lab Clin Med adults. Br J Haematol 70:55, 1988.
107:353, 1986. 187. Tichelli A, Gratwohl A, Wrsch A, et al: Late haematological complications in severe
157. Kawano Y, Nissen C, Gratwohl A, Speck B: Immunostimulatory effects of different aplastic anaemia. Br J Haematol 69:413, 1988.
antilymphocyte globulin preparations: A possible clue to their clinical effect. Br J 188. Moore MAS, Castro-Malaspina H: Immunosuppression in aplastic anemiaPost-
Haematol 68:115, 1988. poning the inevitable? N Engl J Med 314:1358, 1991.
158. Bielory L, Wright R, Nienhuis AW, et al: Antithymocyte globulin hypersensitivity in 189. Tichelli A, Passweg J, Nissen C, et al: Repeated treatment with horse antilymphocyte
bone marrow failure patients. JAMA 260:3164, 1988. globulin for severe aplastic anaemia. Br J Haematol 100:393, 1998.
159. Camitta B, OReilly RJ, Sensenbrenner L: Antithoracic duct lymphocyte globulin 190. Scheinberg P, Nunez O, Young NS: Retreatment with rabbit anti-thymocyte globulin
therapy of severe aplastic anemia. Blood 62:883, 1983. and ciclosporin for patients with relapsed or refractory severe aplastic anaemia. Br J
160. Champlin R, Ho W, Gale RP: Antithymocyte globulin treatment in patients with Haematol 133:622, 2006.
aplastic anemia: a prospective randomized trial. N Engl J Med 308:113, 1983. 191. Bacigalupo A, Van Lint MT, Cerri R, et al: Treatment of severe aplastic anemia with
161. Young N, Griffin P, Brittain E, et al: A multicenter trial of antithymocyte globulin in bolus 6-methylprednisolone and antilymphocyte globulin. Blut 41:168, 1980.
aplastic anemia and related diseases. Blood 72:1861, 1988. 192. Issaragrisil S, Tangnai-Trisorana Y, Siriseriwan T, et al: Methylprednisolone therapy
162. Schrezenmeier H, Marin P, Raghavachar A, et al: Relapse of aplastic anaemia after in aplastic anaemia: Correlation of in vitro tests and lymphocyte subsets with clinical
immunosuppressive treatment: A report from the European Bone Marrow Trans- response. Eur J Haematol 40:343, 1988.
plantation Group SAA Working Party. Br J Haematol 85:371, 1993. 193. Brodsky RA, Sensenbrenner LL, Jones RJ: Complete remission in severe aplastic ane-
163. Doney K, Leisenring W, Storb R, Appelbaum FR: Primary treatment of acquired mia after high-dose cyclophosphamide without bone marrow transplantation. Blood
aplastic anemia: Outcomes with bone marrow transplantation and immunosuppres- 87:491, 1996.
sive therapy. Ann Intern Med 126:107, 1997. 194. Jones RJ, Barber JP, Vala MS et al: Assessment of aldehyde dehydrogenase in viable
164. Tichelli A, Gratwohl A, Nissen C, Speck B: Late clonal complications in severe aplas- cells. Blood 85:2742, 1995.
tic anemia. Leuk Lymphoma 12:167, 1994. 195. Kastan MB, Schlaffer I, Russo JE, et al: Direct demonstration of aldehyde dehydro-
165. De Planque MM, Bacigalupo A, Wrsch A, et al: Long-term follow-up of severe genase in human hematopoietic progenitor cells Blood 75:1947, 1990.
aplastic anaemia patients treated with antithymocyte globulin. Br J Haematol 73:121, 196. Brodsky RA, Sensenbrenner LL, Smith BD, et al: Durable treatment-free remission
1989. following high-dose cyclophosphamide for previously untreated severe aplastic ane-
166. Soci G, Henry-Amar M, Bacigalupo A, et al: Malignant tumors occurring after mia. Ann Intern Med 135:477, 2001.
treatment of aplastic anemia. N Engl J Med 319:1152, 1993. 197. Brodsky RA: High-dose cyclophosphamide for aplastic anemia and autoimmunity.
167. Stryckmans PA, Dumont JP, Velu T, Debusscher L: Cyclosporine in refractory severe Curr Opin Oncol 14:143, 2002.
aplastic anemia [letter]. N Engl J Med 310:655, 1984. 198. Hansen PB, Lauritzen AM: Aplastic anemia successfully treated with rituximab. Am
168. Lazzarino M, Morra E, Canevari A, et al: Cyclosporine in the treatment of aplastic J Hematol 80:292, 2005.
anaemia and pure red-cell aplasia. Bone Marrow Transplant 4(Suppl 4):165, 1989. 199. French Cooperative Group for the Study of Aplastic and Refractory Anemias:
169. Hinterberger-Fischer M, Hcker P, Lechner K, et al: Oral cyclosporin-A is effective Androgen therapy in aplastic anemia: A comparative study of high and low doses of
treatment for untreated and also for previously immunosuppressed patients with 4 different androgens. Scand J Haematol 36:346, 1986.
severe bone marrow failure. Eur J Haematol 43:136, 1989. 200. Champlin RE, Ho WG, Feig SA, et al: Do androgens enhance the response to anti-
170. Ttterman TH, Hglund M, Bengtsson M, et al: Treatment of pure red-cell aplasia thymocyte globulin in patients with aplastic anemia? A prospective randomized
and aplastic anaemia with cyclosporin: Long-term clinical effects. Eur J Haematol trial. Blood 66:184, 1985.
42:126, 1989. 201. Sonoda Y, Ohno Y, Fujii H, et al: Multilineage response in aplastic anemia patients
171. Leeksma OC, Thomas LLM, van der Lelie J, et al: Effectiveness of low dose cyclospor- following long-term administration of filgrastim (recombinant human granulocyte
ine in acquired aplastic anaemia with severe neutropenia. Neth J Med 41:143, 1992. colony stimulating factor). Stem Cells 11:543, 1993.
172. Leonard EM, Raefsky E, Griffith P, et al: Cyclosporine therapy of aplastic anaemia, 202. Bessho M, Jinnai I, Hirashima K, et al: Trilineage recovery by combination therapy
congenital and acquired red-cell aplasia. Br J Haematol 72:278, 1989. with recombinant human granulocyte colony-stimulating factor and erythropoietin
173. Tong J, Bacigalupo A, Piaggio G, et al: Severe aplastic anemia (SAA): Response to in patients with aplastic anemia and refractory anemia. Stem Cells 12:604, 1994.
cyclosporin A (CyA) in vivo and in vitro. Eur J Haematol 46:212, 1991. 203. Socie G, Mary JY, Schrezenmeier H, et al: Granulocyte-stimulating factor and severe
174. Nakao S, Yamaguchi M, Shiobara S, et al: Interferon-g gene expression in unstimu- aplastic anemia: A survey by the European Group for Blood and Marrow Transplan-
lated bone marrow mononuclear cells predicts a good response to cyclosporine ther- tation (EBMT). Blood 109:2794, 2007.
apy in aplastic anemia. Blood 79:2531, 1992. 204. Ganser A, Lindemann A, Siepelt G, et al: Effects of recombinant human interleukin-
175. Kojima S, Fukada M, Miyajima Y, Matsuyama T: Cyclosporine and recombinant 3 in aplastic anemia. Blood 76:1287, 1990.
granulocyte colony-stimulating factor in severe aplastic anemia [letter]. N Engl J Med 205. Walsh CE, Liu JM, Anderson SM, et al: A trial of recombinant human interleukin-1
313:920, 1990. in patients with severe refractory aplastic anaemia. Br J Haematol 80:106, 1992.
176. Bertrand Y, Amri F, Capdeville R, et al: The successful treatment of two cases of 206. Speck B, Tichelli A, Widmer E, et al: Splenectomy as an adjuvant measure in the
severe aplastic anaemia with granulocyte colony-stimulating factor and cyclosporine treatment of severe aplastic anaemia. Br J Haematol 92:818, 1996.
A [case report]. Br J Haematol 79:648, 1991. 207. Sadowitz PD, Dubowy RL: Intravenous immunoglobulin in the treatment of aplastic
177. Schrezenmeier H, Schlander M, Raghavachar A: Cyclosporin A in aplastic anemia anemia. Am J Pediatr Hematol Oncol 12:198, 1990.
Report of a workshop. Ann Hematol 65:33, 1992. 208. Bodenstein H: Successful treatment of aplastic anemia with high-dose immunoglob-
178. Gluckman E, Esperou-Bourdeau H, Baruchel A, et al: Multicenter randomized study ulin [letter]. N Engl J Med 314:1368, 1991.
comparing cyclosporine-A alone and antithymocyte globulin with prednisone for 209. Ito T, Haraiwa M, Ishikawa Y, et al: Lymphocytapheresis in a patient with severe
treatment of severe aplastic anemia. Blood 79:2540, 1992. aplastic anaemia. Acta Haematol 80:167, 1988.
179. Rosenfeld S, Follmann D, Nunez O, et al: Antithymocyte globulin and cyclosporine 210. Morales-Polanco MR, Sanchez-Valle E, Guerrero-Rivera S, et al: Treatment results of
for severe aplastic anemia: Association between hematologic response and long-term 23 cases of severe aplastic anemia with lymphocytapheresis. Arch Med Res 28:85,
outcome. JAMA 289:1130, 2003. 1997.
180. Scheinberg P, Wu CO, Nunez O, et al: Long-term outcome of pediatric patients with 211. Kiem HP, McDonald GB, Myerson D, et al: Marrow transplantation for hepatitis-
severe aplastic anemia treated with antithymocyte globulin and cyclosporine. J Pedi- associated aplastic anemia: A follow-up of long-term survivors. Biol Blood Marrow
atr 153:814, 2008. Transplant 2:93, 1996.
181. Osugi Y, Yagasaki H, Sako M, et al: Antithymocyte globulin and cyclosporine for 212. Lewis SM: Course and prognosis in aplastic anemia. Br Med J 1:1027, 1965.
treatment of 44 children with hepatitis associated aplastic anemia. Haematologica 213. Lynch RE, Williams DM, Reading JC, Cartwright GE: The prognosis in aplastic ane-
92:1687, 2007. mia. Blood 45:517, 1975.
182. Teramura M, Kimura A, Iwase S, et al: Treatment of severe aplastic anemia with anti- 214. Horowitz MM: Current status of allogeneic bone marrow transplantation in
thymocyte globulin and cyclosporin A with or without G-CSF in adults: A multi- acquired aplastic anemia. Semin Hematol 37:30, 2000.
center randomized study in Japan. Blood 110:1756, 2007. 215. Ades L, Mary J-Y, Robin M, et al: Long-term outcome after bone marrow transplan-
183. Zheng Y, Liu Y, Chu Y: Immunosuppressive therapy for acquired severe aplastic ane- tation for severe aplastic anemia. Blood 103:2490, 2004.
mia (SAA): A prospective comparison of four different regimens. Exp Hematol 216. Schrezenmeier H, Passweg JR, Marsh JC, et al: Worse outcome and more chronic
34:826, 2006. GVHD with peripheral blood progenitor cells than bone marrow in HLA-matched
184. Bacigalupo A, Brand R, Oneto R, et al: Treatment of acquired severe aplastic anemia: sibling donor transplants for young patients with severe acquired aplastic anemia.
Bone marrow transplantation compared with immunosuppressive therapyThe Blood 110:1397, 2007.
European Group for Blood and Marrow Transplantation experience. Semin Hematol 217. Locasciulli A: Acquired aplastic anemia in children: incidence, prognosis and treat-
37:69, 2000. ment options. Paediatr Drugs 4:761, 2002.
CHAPTER 34: Aplastic Anemia: Acquired and Inherited 483

218. Tisdale JF, Dunn DE, Maciejewski J: Cyclophosphamide and other new agents for 247. Ginzberg H, Shin J, Ellis L, et al: Shwachman syndrome: Phenotypic manifestations
the treatment of severe aplastic anemia. Semin Hematol 37:102109 2000. of sibling sets and isolated cases in a large patient cohort are similar. J Pediatr 135:81,
219. Fanconi G: Familire infantile perniziosaartige anmie (pernizises blutbild und 1999.
konstitution). Jahrbuch Kinderheil 117:257, 1927. 248. Shimamura A: Shwachman-Diamond syndrome. Semin Hematol 43:178, 2006.
220. Rosendorff J, Bernstein R, Macdougall L, Jenkins T: Fanconi anemia: another disease 249. Rujkijyanont P, Watanabe K, Ambekar C, et al: SBDS-Deficient cells undergo accel-
of unusually high prevalence in the Afrikaans population of South Africa. Am J Med erated apoptosis through the FAS pathway. Haematologica 93:363, 2008.
Genet 27:793, 1987. 250. Thornley I, Dror Y, Sung L, et al: Abnormal telomere shortening in leucocytes of
221. Dokal I, Vulliamy T: Inherited aplastic anaemias/bone marrow failure syndromes. children with Shwachman-Diamond syndrome. Br J Haematol 117:189, 2002.
Blood Rev 22:141, 2008. 251. Ganapathi KA, Shimamura A: Ribosomal dysfunction and inherited marrow failure.
222. Jacquemont C, Taniguchi T: The Fanconi anemia pathway and ubiquitin. BMC Bio- Br J Haematol 141(3):376, 2008.
chem 22(8 Suppl 1):S10, 2007. 252. Ganapathi KA, Austin KM, Lee CS, et al: The human Shwachman-Diamond syn-
223. Alter BP, Giri N, Savage SA, et al: Update on inherited bone marrow failure syn- drome protein, SBDS, associates with ribosomal RNA. Blood 110:1458, 2007.
dromes (IBMFS). IBMFS Newsletter of the Clinical Genetics Branch, National Cancer 253. Bhatla D, Davies SM, Shenoy S, et al: Reduced-intensity conditioning is effective and
Institute. P.1, Summer 2008. safe for transplantation of patients with Shwachman-Diamond syndrome. Bone Mar-
224. Dufour C, Corcione A, Svahn J, et al: TNF- and TNF- are overexpressed in the row Transplant 42:159, 2008.
bone marrow of Fanconi anemia patients and TNF- suppresses erythropoiesis in 254. Dokal I, Rule S, Chen F, et al: Adult onset of acute myeloid leukaemia (M6) in
vitro. Blood 102:2053, 2003. patients with Shwachman-Diamond syndrome. Br J Haematol 99:171, 1997.
225. DApolito M, Zelante L, Savoia A: Molecular basis of Fanconi anemia. Haematologica 255. Dror Y, Squire J, Durie P, Freedman MH: Malignant myeloid transformation with
83:533, 1998. isochromosome 7q in Shwachman-Diamond syndrome. Leukemia 12:1591, 1998.
226. Garcia-Higuera I, Kuang Y, DAndrea AD: The molecular and cellular biology of 256. Li FP, Hecht F, Kaiser-McCaw B et al: Ataxia-pancytopenia: syndrome of cerebellar
Fanconi anemia. Curr Opin Hematol 6:83, 1999. ataxia, hypoplastic anemia, monosomy 7 and acute myelogenous leukemia. Cancer
227. Young NA, Alter BP: Aplastic Anemia: Acquired and Inherited. W.B. Saunders, Phila- Genet Cytogenet 4:189, 1981.
delphia, 1994. 257. Mahmood F, King MD, Smyth OO, et al: Familial cerebellar hypoplasia and pancy-
228. Estren S, Damshek W: Familial hypoplastic anemia of childhood: Report of 8 cases topenia without chromosomal breakages. Neuropediatrie 29:302, 1998.
in 2 families with beneficial effects of splenectomy in 1 case. Am J Dis Child 73:671, 258. Gonzlez-del AA, Cervera M, Gomez L, et al: Ataxia-pancytopenia syndrome. Am J
1947. Med Genet 90:252, 2000.
229. Swhimamura A, DAndrea AD: Subtyping of Fanconi anemia patients: Implications 259. Geddis AE: Inherited thrombocytopenia: Congenital amegakaryocytic thrombocy-
for clinical management. Blood 102:3459, 2003. topenia and thrombocytopenia with absent radii. Semin Hematol 43:196, 2006.
230. Carreau M, Liu L, Gan OI, et al: Short-term granulocyte colony-stimulating factor 260. Germeshausen M, Ballmaier M, Welte K: MPL mutations in 23 patients suffering
and erythropoietin treatment enhances hematopoiesis and survival in the mitomy- from congenital amegakaryocytic thrombocytopenia: The type of mutation predicts
cin C-conditioned Fancc(-/-) mouse model, while long-term treatment is ineffective. the course of the disease. Hum Mutat 27:296, 2006.
Blood 100:1499, 2002. 261. Pierce AJ, Jasin M: NHEJ deficiency and disease. Mol Cell 8:1160, 2001.
231. Alter BP: Cancer in Fanconi anemia. Cancer 97:425, 2003. 262. ODriscoll M, Gennery AR, Seidel J, et al: An overview of three new disorders associ-
232. Alter BP, Caruso JP, Drachtman RA, et al: Fanconi anemia: Myelodysplasia as a pre- ated with genetic instability: LIG4 syndrome, RS-SCID and ATR-Seckel syndrome.
dictor of outcome. Cancer Genet Cytogenet 117:125, 2000. DNA Repair (Amst) 3:1227, 2004.
233. Gluckman E, Wagner JE: Hematopoietic stem cell transplantation in childhood 263. ODriscoll M, Jeggo PA: CSA can induce DNA double-strand breaks: Implications
inherited bone marrow failure syndrome. Bone Marrow Transplant 41:127, 2008. for BMT regimens particularly for individuals with defective DNA repair. Bone Mar-
234. Huck K, Hanenberg H, Nrnberger W, et al: Favourable long-term outcome after row Transplant 41:983, 2008.
matched sibling transplantation for Fanconi-anemia (FA) and in vivo T-cell deple- 264. Walters TR, Desposito F: Aplastic anemia in Dubowitz syndrome. J Pediatr 106:622,
tion. Klin Padiatr 220:147, 2008. 1985.
235. Ayas M, Al-Jefri A, Al-Seraihi A, et al: Second stem cell transplantation in patients 265. Berthold F, Fuhrmann W, Lampert F: Fatal aplastic anemia in a patient with Dubow-
with Fanconi anemia using antithymocyte globulin alone for conditioning. Biol itz syndrome. Eur J Pediatr 146:605, 1987.
Blood Marrow Transplant 14:445, 2008. 266. Gennery AR, Slatter MA, Bhattacharya A, et al: The clinical and biological overlap
236. Kelly PF, Radtke S, von Kalle C, et al: Stem cell collection and gene transfer in Fan- between Nijmegen Breakage Syndrome and Fanconi anemia. Clin Immunol 113:214,
coni anemia. Mol Ther 15:211, 2007. 2004.
237. Dufour C, Svahn J: Fanconi anaemia: New strategies. Bone Marrow Transplant 267. Gadkowska-Dura M, Dzieranowska-Fangrat K, Dura W, et al: Unique morphologi-
41(Suppl 2):S90, 2008. cal spectrum of lymphomas in Nijmegen breakage syndrome (NBS) patients with
238. Dokal I: Dyskeratosis congenita in all its forms. Br J Haematol 110:768, 2000. high frequency of consecutive lymphoma formation. J Pathol 216:337, 2008.
239. Vulliamy TJ, Dokal I: Dyskeratosis congenita: the diverse clinical presentation of 268. Stephan JL, Vlekova V, Le Deist F, et al: Severe combined immunodeficiency: A
mutations in the telomerase complex. Biochimie 90:122, 2008. retrospective single-center study of clinical presentation and outcome in 117
240. Savage SA, Alter BP: The role of telomere biology in bone marrow failure and other patients. J Pediatr 123:564, 1993.
disorders. Mech Ageing Dev 129(12):35, 2008. 269. Bertrand Y, Muller SM, Casanova JL, et al: Reticular dysgenesis: HLA non-identical
241. Walne AJ, Vulliamy T, Marrone A, et al: Genetic heterogeneity in autosomal reces- bone marrow transplants in a series of 10 patients. Bone Marrow Transplant 29:759,
sive dyskeratosis congenita with one subtype due to mutations in the telomerase- 2002.
associated protein NOP10. Hum Mol Genet 16:1619, 2007. 270. Esperou-Bourdeau H, Leblanc T, Schaison G, et al: Aplastic anemia associated with
242. Alter BP, Baerlocher GM, Savage SA, et al: Very short telomere length by flow fluo- bird-headed dwarfism (Seckel syndrome). Nouv Rev Fr Hematol 35:99, 1993.
rescence in situ hybridization identifies patients with dyskeratosis congenita. Blood 271. ODriscoll M, Ruiz-Perez VL, Woods CG, et al: A splicing mutation affecting expres-
110:1439, 2007. sion of ataxia-telangiectasia and RAD3-related protein (ATR) results in Seckel syn-
243. Ghavamzadeh A, Alimoghadam K, Nasseri P, et al: Correction of bone marrow fail- drome. Nat Genet 33:497, 2003.
ure in dyskeratosis congenita by bone marrow transplantation. Bone Marrow Trans- 272. Griffith E, Walker S, Martin CA, et al: Mutations in pericentrin cause Seckel syn-
plant 23:299, 1999. drome with defective ATR-dependent DNA damage signaling. Nat Genet 40:232,
244. Cesaro S, Oneto R, Messina C, et al: Haematopoietic stem cell transplantation for 2008.
Shwachman-Diamond disease: A study from the European Group for Blood and 273. Hayani A, Suarez CR, Molnar Z, et al: Acute myeloid leukemia in a patient with
Marrow Transplantation. Br J Haematol 131:231, 2005. Seckel syndrome. J Med Genet 31:148, 1994.
245. Gngr T, Corbacioglu S, Storb R, Seger RA: Nonmyeloablative allogeneic hemato- 274. Gonzalez CH, Durkin-Stamm MV, Geimer NF, et al: The WT syndromeA new
poietic stem cell transplantation for treatment of dyskeratosis congenita. Bone Mar- autosomal dominant pleiotropic trait of radial/ulnar hypoplasia with high risk of
row Transplant 31:407, 2003. bone marrow failure and/or leukemia. Birth Defects Orig Artic Ser 13:31, 1977.
246. Dror Y, Freedman MH, Leaker M, et al: Low-intensity hematopoietic stem-cell 275. Walne AJ, Vulliamy TJ, Beswick R, et al: TINF2 mutations result in very short telo-
transplantation across human leucocyte antigen barriers in dyskeratosis congenita. meres: Analysis of a large cohort of patients with dyskeratosis congenita and related
Bone Marrow Transplant 31:847, 2003. bone marrow failure syndromes. Blood 112:3594, 2008.

You might also like