Acquired Aplastic Anemia and Pure Red Cell Aplasia PDF

Loading...
Loading...
Loading...
Loading...
Loading...
Loading...
Loading...

Document Details

RealizableJoy2827

Uploaded by RealizableJoy2827

Akiko Shimamura and David A. Williams

Tags

aplastic anemia bone marrow failure medical hematology

Summary

This chapter discusses acquired aplastic anemia and pure red cell aplasia, focusing on their causes, pathophysiology, and management. It highlights the importance of distinguishing between inherited and acquired forms to guide treatment decisions. The chapter also reviews epidemiologic data on aplastic anemia.

Full Transcript

C H A P T E R 6 Acquired Aplastic Anemia and Pure Red Cell Aplasia...

C H A P T E R 6 Acquired Aplastic Anemia and Pure Red Cell Aplasia Akiko Shimamura and David A. Williams CHAPTER OUTLINE APLASTIC ANEMIA Management and Outcome Epidemiology PURE RED CELL APLASIA Causal Factors Primary Acquired Pure Red Cell Aplasia Pathophysiology Secondary Acquired Pure Red Cell Aplasia Clinical Evaluation Acquired and congenital bone marrow failure syndromes anemias are presented in Box 6-1. A careful medical are characterized by a reduction in the effective produc- history, physical examination, and laboratory evaluation tion of mature erythrocytes, granulocytes, and platelets are critical to discern inherited versus acquired causes of by the bone marrow. Bone marrow failure leads to various aplastic anemia. The distinction between inherited and peripheral blood cytopenias. In some conditions, only one acquired causes of aplastic anemia carries profound or two cell lines may be affected. In others such as aplastic implications for medical management and treatment, so anemia the result is pancytopenia. In this chapter, we deal a careful search for possible underlying inherited syn- only with the acquired bone marrow failure syndromes, dromes should be undertaken prior to initiation of though we use the term rather loosely because the acquired therapy. marrow failure syndromes may have a genetic basis. The Camitta and co-workers classified the severity of aplas- known inherited/congenital bone marrow failure syn- tic anemia in an effort to make possible the comparison dromes are described in detail in Chapter 7. The acquired of diverse groups of patients and different therapeutic marrow failure syndromes that affect only platelets or approaches.1 Diagnosis of severe aplastic anemia requires granulocytes are described in Chapters 22 and 34 respec- that the patient have at least two of the following anoma- tively. The acquired single lineage deficiency syndromes lies: a granulocyte count below 500/µL, a platelet count that involve red cell production (pure red cell aplasia) below 20,000/µL, and an absolute reticulocyte count less are also described in this chapter, except that the 5Q- than or equal to 40 × 109/L. In addition the bone marrow syndrome is discussed in Chapter 7 and in more detail biopsy must contain less than 25% of the normal cellular- Chapter 51. ity or less than 30% hematopoietic elements (Fig. 6-1). Very severe aplastic anemia is further defined by a granu- locyte count of less than 200/µL.2 Mild or moderate APLASTIC ANEMIA aplastic anemia, sometimes called hypoplastic anemia, is Decreased production of mature blood cells may result distinguished from the severe form by the presence of from a reduction in the number or function of their pro- mild or moderate cytopenias and more variable, but still genitors. Aplastic anemia is a descriptive term referring deficient, bone marrow cellularity. These distinctions are to a clinical state in which peripheral blood pancytopenia more than semantic; they are critical for the prediction results from reduced or absent production of blood cells of outcome and the choice of therapy. in the bone marrow. Aplastic anemia may arise in the setting of inherited/congenital syndromes associated with Epidemiology a predisposition to marrow failure (discussed in Chapter Epidemiologic studies performed in Europe estimate that 7), may develop secondary to marrow-toxic stressors in the annual incidence of aplastic anemia is 2 per million an otherwise seemingly normal host, or may lack any per year.3 By comparison, the incidence of acute leukemia apparent underlying cause. Classifications of the aplastic is about 50 per million per year. Higher figures for the 161 162 SECTION II BONE MARROW FAILURE Box 6-1 Classification of the Aplastic Anemias339 distinction between acquired versus inherited causes of aplastic anemia is crucial to guide clinical management ACQUIRED and treatment (see Chapter 8). Secondary Radiation Drugs Drugs and chemicals The incidence of drug- and chemical-related aplastic Direct toxicity: chemotherapy; benzene Idiosyncratic: chloramphenicol; antiinflammatory drugs; antiepi- anemia11 varies over time and from place to place. Many leptics; carbonic anhydrase inhibitors drugs and toxins have been implicated by inferential and Viruses circumstantial evidence; the magnitude of the risk is Epstein-Barr virus usually unknown (Box 6-2). Presence of an agent on this Hepatitis (non-A, B, C, E, or G) Human immunodeficiency virus list suggests caution regarding its use, but no drug on this Immune diseases list should be proscribed if there are strong clinical indica- Eosinophilic fasciitis tions for its use. From a public health perspective, even Hypoimmunoglobulinemia drugs associated with an increased risk of marrow failure Systemic lupus erythematosis (uncommon) do not cause large numbers of cases of aplastic anemia.3 Thymoma Graft-versus-host disease in immunodeficiency Note that even confirmed associations do not substan- Pregnancy tiate causality. Antibiotics felt to be causative may have Paroxysmal nocturnal hemoglobinuria been administered for the viral infection that led to the Myelodysplasia aplastic anemia or for symptoms from already established Idiopathic neutropenia in an undiagnosed case of aplastic anemia. INHERITED Bleeding may be precipitated in undiagnosed thrombocy- Fanconi anemia topenic patients who receive nonsteroidal antiinflamma- Dyskeratosis congenita tory drugs. As an example of known errors in associations, Shwachman-Diamond syndrome among six patients reported to have sniffed glue and Amegakaryocytic thrombocytopenia Diamond-Blackfan anemia Reticular dysgenesis GATA-2 syndromes Familial aplastic anemias Nonhematologic syndromes (e.g., Down, Dubowitz, and Seckel syndromes) incidence of aplastic anemia have been obtained from smaller studies in the United States and earlier surveys in Europe, but these figures may have been inflated by the inclusion of cases of myelodysplasia, a much more common syndrome. Aplastic anemia is more common in Asia (4 to 7 per million per year)4 than in the West. Chloramphenicol, a known cause of aplastic anemia, has been widely used in much of Asia because of its efficacy and low cost, but reductions in its use have not been A accompanied by reductions in aplastic anemia incidence in Japan5,6 or elsewhere,7,8 and no association was observed in the case control studies in Thailand.9 A recent case control study10 in Thailand spanning 1989-2002 described an association between aplastic anemia and exposure to benzene or pesticides. Although an increased risk of aplastic anemia was also associated with animal exposures and ingestion of nonbottled or nondistilled water, no significant associations with known infections or hepatitis were observed. The peak age of presentation of aplastic anemia is at 15 to 25 years or over 60 years. The male-to-female ratio in acquired aplastic anemia is approximately 1 : 1. Causal Factors When no causative factors are ascertained, the cases are B classified as idiopathic. A search for possible causative agents is warranted because some patients may improve Figure 6-1 Bone marrow examination in severe aplastic anemia. A, Note hypocellularity in biopsy. B, Residual cells seen in aspirate are following the removal of the offending agent. However, lymphocytes, stromal cells, plasma cells, and mast cells. (Courtesy of most cases of aplastic anemia remain idiopathic. The Dr. Gail Wolfe, Boston, Mass.) 6 Acquired Aplastic Anemia and Pure Red Cell Aplasia 163 Box 6-2 Classification of Drugs and Chemicals Associated the medication, sometimes weeks to months after its with Aplastic Anemia* administration is discontinued. This last category of patients may possess a genetic propensity for this phe- AGENTS THAT REGULARLY PRODUCE MARROW DEPRESSION nomenon or be due to a direct effect of the drug on Antibiotics: daunorubicin, doxorubicin hydrochloride (Adriamycin), antigen presentation of abnormal peptides (see below). chloramphenicol Antimetabolites: antifolic compounds, nucleotide analogues Chloramphenicol Antimitotics: vinblastine, colchicine Benzene and chemicals containing benzene: carbon tetrachloride, Chloramphenicol was considered to be the commonest chlorophenols, kerosene, Stoddard solvent cause of aplastic anemia at the peak of its use, which Cytotoxic cancer chemotherapy with alkylating drugs: busulfan, mel- began in 1949.13 A genetic predisposition may exist. The phalan, cyclophosphamide mechanism of the idiosyncratic aplasia remains unknown AGENTS POSSIBLY ASSOCIATED BUT WITH A LOW PROBABILITY RELATIVE despite extensive investigation. TO USE Chloramphenicol contains a nitrobenzene ring and thus Chloramphenicol resembles amidopyrine, a drug known to cause agranulo- Insecticides: chlordane, chlorophenothane (DDT), γ-benzene hexa- cytosis.14,15 Chloramphenicol is the prime example of a chloride (lindane), parathion Anticonvulsants: carbamazepine, hydantoins, phenacemide drug that causes both dose-related marrow suppression Nonsteroidal antiinflammatory agents: indomethacin, ibuprofen, oxy- through mechanisms that include mitochondrial inhibi- phenylbutazone, phenylbutazone, sulindac tion,16 and idiosyncratic aplastic anemia.17 Antihistamines: cimetidine, chlorpheniramine, ranitidine The signs of dose-related toxicity appear more rapidly Antiprotozoal drugs: quinacrine, chloroquine in patients with hepatic or renal disease because the drug Sulfonamides: some antibiotics, antidiabetics (chlorpropamide, tol- butamide), antithyroid drugs (methimazole, methylthiouracil, pro- must be inactivated by conjugation with glucuronide in pylthiouracil), carbonic anhydrase inhibitors (acetazolamide, the liver and excreted in the urine. High doses and high methazolamide) plasma levels correlate with the characteristic reversible Penicillamine erythroid depression. In vitro, chloramphenicol inhibits Metals: gold, arsenic, bismuth, mercury the growth of both colony-forming units granulocyte/ AGENTS MORE RARELY ASSOCIATED macrophage (CFU-GMs) and colony-forming units ery- Allopurinol (may potentiate marrow suppression by cytotoxic drugs) throid (CFU-Es)18,19,20,21,22 and also may inhibit the hema- Antibiotics: flucytosine, mebendazole, methicillin, sulfonamides, topoietic microenvironment (HM).20,23 streptomycin, tetracycline, trimethoprim/sulfamethoxazole Carbimazole Other Drugs Guanidine Lithium Nonsteroidal antiinflammatory drugs, which are used Methyldopa more extensively in adults than in children, are associated Potassium perchlorate with aplasia.3 Nonsteroidal antiinflammatory drugs asso- Quinidine Sedatives and tranquilizers: chlordiazepoxide, chlorpromazine, ciated by occasional case reports with aplastic anemia meprobamate, methyprylon, piperacetazine, prochlorperazine include aspirin,24 indomethacin,25,26,27,28 and ibuprofen.29 Thiocyanate Several large studies reported an increased risk of aplastic anemia with phenylbutazone30,31 and identified even Modified from Young NS, Alter BP: Aplastic anemia: acquired and higher probabilities with some of the other nonsteroidal inherited, Philadelphia, 1994, WB Saunders, p 104. *Agents are listed because they have been cited in the literature; antiinflammatory drugs.3 Cimetidine, another commonly inclusion in this list does not imply acceptance by the author of a used drug, is associated with a 2 per 100,000 user risk causal relationship. of cytopenias.32,33 Sulfa-containing compounds, which appear as risk factors in most case-control studies of drugs become aplastic, five had sickle cell anemia and aplastic and aplastic anemia, are used in a wide variety of clinical crises now known to be due to parvovirus infection.12 circumstances.34,35,36,37,38,39,40,41,42,43 Other drugs implicated The incidence of drug-related aplasia in pediatric cases in aplastic anemia that are commonly used in the pediatric is low, mainly because many of the drugs felt to be related population include anticonvulsants (hydantoins,44,45,46 to aplasia are not used in childhood, with the exception carbamazepine47,48,49,50,51) and carbonic anhydrase inhibi- of antiepileptic drugs, carbonic anhydrase inhibitors, tors52 (acetazolamide53,54 and methazolamide55,56). Many nonsteroidal antiinflammatory medications, and some of the drugs listed in Figure 6-1 also have been associated antibiotics. with agranulocytosis. In general only a minority of cases Drug-related aplasia may occur in several ways. Drugs of aplastic anemia can be assigned a drug association. may exert direct cytotoxic or suppressive effects on the The distinction between aplasia secondary to the medica- bone marrow. Myelosuppressive drugs, such as those tion versus aplasia arising from the underlying disorder used in cancer chemotherapy, lead to predictable and (or occult viral infection) requiring treatment can be dose-related marrow suppression. Benzene, too, can be difficult. demonstrated regularly to suppress the bone marrow in animals in a dose-linked manner, and most individuals Chemicals and Toxins exposed to sufficient amounts of benzene would probably suffer some type of marrow damage. In practice, most Benzene drug-related aplastic anemia is idiosyncratic and occurs Benzene is a particularly dangerous environmental con- unpredictably in only rare individuals who are prescribed taminant.57,58 It is found in organic solvents, coal tar 164 SECTION II BONE MARROW FAILURE derivatives, and petroleum products.59,60,61,62 Fatal aplas- tic anemia, leukemia, or both, have been reported years Hepatitis later in factory workers who had benzene exposure. Although hepatitis is frequently associated with mild Benzene is concentrated in bone marrow fat,63 forms depression of blood cell counts, aplasia is a rare sequela, water-soluble intermediates,64,65,66 and damages estimated to occur in fewer than 0.07% of the total 67,68,69 DNA. It decreases the numbers of progenitors and number of pediatric hepatitis cases73 and in fewer than damages stroma. The risk of cytopenias is likely related 2% of those with non-A, non-B hepatitis.74 Nonetheless to cumulative exposure. as an identifiable clinical event, a prior episode of hepa- titis is recognized in 2% to 5% of aplastic anemia patients Other Aromatic Hydrocarbons in a Western series.75 The prevalence of prior hepatitis is Toxicity thought to be due to other organic solvents may about twofold this proportion in the Far East.76 Among in some instances be caused by benzene contaminants. children with aplastic anemia in Taiwan, 24% had a Neither pure toluene nor xylene is a marrow toxin. history of recent acute hepatitis.77 Antecedent hepatitis Aplastic anemia has been linked by many case reports to may be subclinical, as about 50% of patients with aplas- insecticides, particularly γ-hexachlorobenzene (lindane) tic anemia may have elevated hepatic transaminases in children.70 Aromatic hydrocarbons are present in insec- before their first transfusion. In a report of 32 patients ticides and herbicides and may comprise the solvents for with liver transplantation for hepatic failure following these agents. Some organophosphate insecticides have non-A, non-B hepatitis, 28% developed aplastic anemia.78 been shown to inhibit in vitro hematopoietic colony for- While aplasia has been reported following both hepatitis mation, as has lindane.71 A and B virus infections,79, 80 the majority of cases of the hepatitis/aplasia syndrome are not associated with sero- Ionizing Radiation typable hepatitis virus.81, 82 Marrow aplasia may occur as an acute toxic sequela of A study of aplastic anemia patients reported to the irradiation due to nuclear bomb explosion, radioactive European Registry from 1990-2007 found that 5% of fallout, reactor accidents, and accidental exposure in patients with aplastic anemia had an antecedent serone- medicine and industry. Bone marrow cells may be affected gative hepatitis. Hepatitis-associated aplastic anemia was by high-energy γ-rays as well as by ingested or absorbed slightly more common in males and tended to present at lower-energy α particles. The radiation injury is to the a younger age. Actuarial survival after treatment with actively replicating pool of precursor and progenitor either immunosuppression or hematopoietic stem cell cells and also to stem cells, in which DNA damage may transplant was similar between patients with hepatitis- have a more severe effect. Nonetheless, radiation-related associated aplastic anemia versus other acquired aplastic marrow aplasia is infrequent. Even in a restricted episode, anemia.83 such as the Chernobyl reactor accident in 1986, most immediate deaths were due to skin burns and damage to Flaviviruses gastrointestinal and pulmonary systems. Flaviviruses cause arbovirus hemorrhagic fevers, dengue, Chronic radiation-induced aplasia is dose related. and other hematodepressive syndromes. Dengue can Patients who were irradiated for ankylosing spondylitis propagate in bone marrow cultures without direct cyto- had an increased risk of aplastic anemia, and American toxicity, and dengue antigens induce lymphocyte activa- radiologists have been reported to have an increased tion and the release of marrow suppressive cytokines.84 death rate from aplasia (for both groups, the pathologic distinction of aplasia and myelodysplasia was not made). Epstein-Barr Virus The incidence of late aplasia in atomic bomb victims Herpesviruses such as Epstein-Barr virus (EBV) are large, was not increased, nor was it increased in patients complex DNA viruses.85 EBV causes infectious mono- receiving radiation therapy for malignancies. Knospe nucleosis, which has pancytopenic complications in less and co-workers suggested that irradiation with an expo- than 1% of cases. More than 12 such cases have been sure greater than 4.4 Gy was required for the develop- reported, and one half of these cases had a fatal outcome. ment of aplasia; they also postulated that low doses In one study, EBV was demonstrated by immunologic and might damage only stem cells, whereas high doses would molecular methods in the bone marrow of six patients also damage the supportive hematopoietic stromal with aplastic anemia.86 Only two had a history of typical microenvironment.72 mononucleosis, although all six had serologic evidence, suggesting that EBV may be an unrecognized cause of Infectious Agents aplastic anemia. The EBV’s target is B cells, although T Patients with bacterial or viral illnesses frequently cells also may be infected. Because EBV is a common develop mild pancytopenia during or following the infec- infection, issues of ascertainment can render the causative tion (see Chapter 37). Patients with bacterial or viral determination of aplasia difficult. infections often receive antibiotics and other medica- tions, and it is frequently not clear whether an ensuing Cytomegalovirus and Human Herpesvirus 6 aplastic anemia was caused by the infection, by the drug, Infection with cytomegalovirus (CMV) may lead to graft or by the combination of the two, or even whether the failure in immunosuppressed bone marrow transplant infectious illness was the result and not the cause of the (BMT) recipients.87 CMV can infect marrow stromal cells pancytopenia. in vitro and can inhibit their ability to produce growth 6 Acquired Aplastic Anemia and Pure Red Cell Aplasia 165 factors;88 direct progenitor cell infection by some CMV lysis. Increased sensitivity and specificity are achieved strains also has been documented.88,89 Herpesvirus 6 is using flow cytometry assays for the absence of GPI- the cause of exanthem subitum90 and, like CMV, may be anchored proteins such as CD59 and CD55.110 More found in the marrow of patients with graft failure after sensitive tests utilize aerolysin, a channel-forming toxin transplantation,91 as well as in hematopoietic progenitors that binds GPI-anchored proteins to result in cell lysis but infected in vitro.92 As with other viruses, both of these leaves the GPI-deficient PNH cells intact.111 The use of a are ubiquitous infections making causality difficult to fluorescently labeled aerolysin variant that binds to GPI ascertain. but fails to lyse the cells offers increased sensitivity and specificity for the detection of the PNH clone.112 Human Immunodeficiency Virus The clinical significance of small populations of PNH Patients with acquired immunodeficiency syndrome clones is unclear, and most of the published data are (AIDS) often have cytopenias,93 but their marrow is much derived from adult patients. Small numbers of PNH cells more commonly cellular and dysplastic than empty. are detectable in healthy controls,113 although the PNH Colony formation by marrow from patients may be cells in this context are typically polyclonal. Despite the diminished.94,95,96,97 The action of human immunodefi- frequent finding of PNH clones in the bone marrows of ciency virus-1 (HIV-1), a lentivirus, on hematopoietic aplastic anemia patients, only 10% to 15% of patients cells remains a subject of controversy. HIV-1 infection of subsequently develop the clinical syndrome of PNH.114 CD34+ cells has been difficult to detect in vivo from In the majority of patients, such clones may persist or patient material96,98 or after tissue culture infection of disappear; however, some patients may develop large normal cells.96,99 The virus apparently directly infects symptomatic PNH clones requiring therapeutic interven- megakaryocytes, which bear the CD4 receptor present on tion.115,116 Patients with small asymptomatic PNH clones T cells.100 The virus also may affect stroma functions, at may respond to the same treatments as other patients least in vitro.101,102 HIV-1 can act indirectly on hemato- with aplastic anemia. Patients who develop large PNH poiesis through inhibitory lymphokine production: The clones are at risk for developing symptoms of hemolysis envelope glycoprotein can stimulate macrophages to or thrombosis.115 One recent retrospective study included produce tumor necrosis factor (TNF), which in turn 47 pediatric patients with aplastic anemia of whom 14 inhibits hematopoietic colony formation.103 Hematologic had a PNH clone greater than 1% at baseline (median suppression can also be due to opportunistic infections, clone size 16%). The median clone size remained small neoplasms, or marrow suppression from the drugs used for all patients, and none of these patients developed to treat AIDS and its complications. clinical symptoms of PNH.115 The etiology of marrow aplasia in PNH remains an Other Viruses area of active investigation. Although severe combined Blood count abnormalities, which are rarely severe, may immunodeficiency (SCID) mice infused with bone marrow be observed in the course of rubella, measles, mumps, from PNH patients show preferential engraftment with varicella, and influenza A.104 the PNH clones,117 studies of hematopoiesis in PNH patients have not detected any selective proliferative Paroxysmal Nocturnal Hemoglobinuria advantage of the PIG-A− hematopoietic clones.118 A Paroxysmal nocturnal hemoglobinuria (PNH) is a disease subsequent study comparing in vitro proliferation of characterized by variable combinations of mild to severe PIG-A− and PIG-A+ CD34 cells from PNH patients intravascular hemolysis, large venous thromboses, and found a selective growth deficiency in the PIG-A+ cell aplastic anemia.105 It is uncommon in adults and even population rather than an advantage for the PIG-A rarer in children (see Chapter 13). There is a clear asso- mutant cells: Fas expression was elevated on the wild ciation of PNH with aplastic anemia: Many patients with type compared with the GPI-deficient cells, suggesting PNH develop pancytopenia and marrow hypoplasia, and increased resistance to apoptosis as one potential mecha- PNH clones are detectable in up to 50% to 70% of nism to explain their findings.119 No proliferative advan- patients with acquired aplastic anemia.106,107,108,109 tage of PNH hematopoietic clones was observed in mice PNH is characterized by an inability to inactivate mosaic for the PIG-A gene.120,121 To evaluate the hypoth- complement on the erythrocyte cell surface, resulting in esis that autoreactive T cells might preferentially elimi- increased sensitivity to complement. Deficits were subse- nate PIG-A+ hematopoietic stem cells while sparing the quently identified in a family of membrane proteins, all PNH clones, the sensitivity of normal versus PNH EBV- of which were anchored to the cell membrane via glyco- transformed B-cell lines to autologous EBV-specific T-cell sylphosphatidylinositol (GPI). GPI binds covalently to lines was examined.122 The PNH cells were no less sensi- specific carboxyl terminal protein sequences and attaches tive to T-cell-mediated cytotoxicity than the non-PNH them to cell membrane phosphatidylinositol residues. cells; thus the GPI-linked cell surface molecules are not The genetic defect in PNH was localized to the X-linked required for killing by T cells. An abnormal distribution phosphatidylinositol glycan class A (PIG-A) gene, whose of expanded T-cell clones detected by size analysis of the product functions in the transfer of N-acetylglucosamine complementarity-determining region 3 (CDR3) in the to phosphatidylinositol as an early step in GPI anchor β-variable region (BV) mRNA of the T-cell receptor formation. Early tests for PNH, such as the Ham test or (TCR) has been noted in PNH patients,123 although the sucrose hemolysis test, relied on the demonstration of targets of such T-cell populations remain to be ascer- increased sensitivity to complement-mediated red cell tained. The mechanisms underlying the clonal expansion 166 SECTION II BONE MARROW FAILURE of the GPI-negative cells in PNH or aplastic anemia is I currently unclear.124 Despite the presence of clonal popu- Toxins medications lations of PNH cells, patients with PNH do not exhibit infections an increased incidence of leukemias, and the GPI-negative clones do not behave in a malignant fashion. Immunologic Diseases Normal HSC Ten percent of patients with the rare collagen vascular syndrome, eosinophilic fasciitis, have associated aplastic anemia.125 This condition is usually limited to adults. Thymoma associated with hematopoietic failure gener- II ally but not exclusively presents as pure red cell aplasia.126 Some of the adult cases had also received drugs such as ? Marrow chloramphenicol or sulfonamides, and the aplasia could stress? have been due to the drugs alone or to the combination. An iatrogenic aplasia can be induced by the transfusion of competent lymphocytes into immunodeficient hosts. In Abnormal HSC these cases, marrow failure arises from the transfusional graft-versus-host disease (GVHD).127 III Pathophysiology T T The purported mechanisms for failure of hematopoiesis T T are predicated on our current knowledge of normal hematopoiesis (see Chapter 7). Disease could result from decreased numbers or defective function of the cellular or soluble components required for blood cell production or from exogenous factors that result in damage or elimina- Immune attack tion of hematopoietic cells. Figure 6-2 depicts different potential mechanisms for the loss of hematopoietic cells in aplastic anemia. In model I, marrow toxins such as IV irradiation, drugs, or chemicals or direct invasion by viruses might cause hematopoietic stem and/or progeni- tor cell death. In model II, an underlying abnormality of the hematopoietic cells may result in a predisposition to hematopoietic stem cell damage, premature stem cell loss, or insufficient stem cell production (see Chapter 1). In model III, viruses, drugs, toxins, or immune dysregula- tion may incite a cellular or humoral immunologic reac- tion against a normal hematopoietic compartment. In Abnormal marrow microenvironment model IV, abnormalities of the marrow stromal environ- Figure 6-2 Models for the pathogenesis of aplastic anemia. I. Healthy ment may actively inhibit hematopoiesis. These possibili- hematopoietic stem cells and progenitors are damaged by exogenous ties are not mutually exclusive, and supportive data for agents such as toxins, medications, or infectious pathogens resulting in marrow aplasia. II. Abnormal marrow cells (e.g., inherited marrow each model have been described. Current data suggest failure syndromes) undergo premature attrition. Marrow aplasia might that aplastic anemia may represent the final seemingly be exacerbated by external factors. III. Immune-mediated attack (cel- common outcome of different pathogenic mechanisms, lular or humeral) eliminates hematopoietic stem cells and progenitors. with different underlying causes entailing different IV. Abnormal marrow microenvironment impairs hematopoiesis. HSC, medical management considerations. Hematopoietic stem cell. Hematopoietic Stem Cells and Clonality Patients with aplastic anemia have decreased numbers activated stem cells.130,131,132,133 CD34+ cells are reduced of blood and marrow committed progenitor cells, in aplastic anemia, and almost all patients show not only assayed as myeloid colony-forming cells (CFU-GMs); severe reduction in the numbers of these cells but also erythroid colony-forming cells (CFU-Es and burst- poor plating efficiency for colony formation from purified forming units erthyroid [BFU-Es]); megakaryocytic pro- CD34+ cells.134,135 genitors (CFU-megas); multipotent colony-forming cells Several lines of evidence point to primary abnormali- (CFU granulocyte, erythrocyte, monocyte, megakaryo- ties in the hematopoietic stem cell as a potential etiology cyte [CFU-GEMMs]); and long-term culture-initiating for aplastic anemia. Multiple studies have noted that cells (LTC-ICs).128,129 Hematopoietic cells can also be simple infusion of stem cells from an identical twin donor assessed phenotypically for the presence of the CD34 without prior conditioning was sufficient to treat the antigen, which defines a compartment of about 1% of aplasia in many cases.136,137,138,139 These results strongly marrow cells that includes progenitors and possibly support a pathogenic stem cell defect and argue against 6 Acquired Aplastic Anemia and Pure Red Cell Aplasia 167 permanent disorders of the bone marrow environment, marrow failure, though telomere lengths are particularly including immunologic mechanisms, as the cause of stem short in patients with dyskeratosis congenita, a disorder cell destruction. Low stem cell numbers persist even with of telomerase function, even in comparison to other hematologic recovery after medical therapy with antithy- marrow failure patients.150 Whether telomere shortening mocyte globulin (ATG) and cyclosporine (see following in marrow failure patients lacking telomerase mutations discussion), and blood cell parameters, such as macrocy- represents a primary defect in telomere maintenance or a tosis, do not return to normal in many cases, again secondary effect of increased cycling of the few remaining suggestive of an underlying stem cell abnormality. A hematopoietic stem cells remains to be clarified. Shorter primary stem cell defect likely also contributes to the telomere lengths have been associated with a higher risk observation of clonal hematopoiesis in aplastic anemia140 of relapse and clonal progression with decreased survival as well as late development of other hematologic diseases in patients with aplastic anemia.151 such as myelodysplasia and acute leukemia (see following discussion). Immune Destruction A subset of patients with seemingly idiopathic aplastic Several lines of data suggest that immune phenomena anemia may actually carry an inherited/congenital predis- may result in aplastic anemia.152 In some syngeneic (iden- position to marrow failure (see Chapter 7). Although the tical twin) BMTs, simple infusion of stem cells is not inherited/congenital marrow failure syndromes are often sufficient to reconstitute hematopoiesis. The requirement associated with characteristic physical findings, patients for prior conditioning of the host may reflect some opera- may present with isolated aplastic anemia as the sole tional immunologic process, though it is also consistent manifestation of their underlying inherited syndrome.141 with the presence of an underlying clonal abnormality In some patients lacking significant physical findings or that requires ablation.153 Histocompatibility studies con- cytopenias at the time of evaluation, an elevated mean ducted in Europe, Asia, and America have indicated an corpuscular volume (MCV) may be the only clue to an increased representation of HLA-DR2 among aplastic underlying inherited marrow failure syndrome. A careful anemia patients, consistent with a genetically determined family history for hematologic abnormalities, malignancy immune susceptibility to disease.153,154,155 Evidence for predisposition, or other characteristic clinical stigmata specific immune mechanisms is reviewed below. may be helpful in distinguishing an inherited marrow failure syndrome.142 For example a subset of patients Lymphocytes presenting with aplastic anemia without other clinical Lymphocyte infusions in mouse models result in marrow stigmata of an inherited marrow failure syndrome were destruction and aplastic anemia.156 Autologous recovery found to harbor mutations in the dyskeratosis congenita has been reported after either mismatched or matched genes hTERC,143,144 which encodes the RNA component transplants in patients prepared with antilymphocyte of telomerase, or hTERT,145,146,147 which encodes the cata- sera or cyclophosphamide.157,158,159 While the etiology of lytic subunit of the telomerase complex. Telomeres are aplastic anemia in children remains unclear, evidence sup- specialized structures that stabilize the ends of each chro- ports an immunologic mechanism of disease in the mosome to prevent excessive shortening with replication, absence of known environmental toxins, drugs, and con- to distinguish chromosome ends from internal DNA stitutional syndromes. Overriding these considerations breaks, and to prevent end-to-end fusions. Telomeres are the large proportion of responses in acquired aplastic consist of six–base pair repeated sequences (TTAGGG) anemia to immunosuppressive therapies, although the associated with a specific protein complex. Telomerase is antilymphocyte preparations contain heterogeneous mix- a ribonucleoprotein enzyme consisting of an H/ACA tures of antibodies to many cells, including lymphocytes, RNA component (TERC) complexed with a reverse tran- and are clearly immunosuppressive as well as generally scriptase (TERT) and additional H/ACA binding proteins cytotoxic. Cyclosporine exerts inhibitory T cells effects including dyskerin, GAR1, NHP2, and NOP10. When and inhibits transcription of genes for cytokines including telomerase levels are diminished or absent, telomeres pro- IL-2 and interferon-gamma (IFN-γ).160 It has many other gressively shorten with each cell division. At a critically toxic effects on several tissues, including brain and kidney. short telomere length, a checkpoint is triggered for cells The diverse antibodies in ATG react with a wide range to stop dividing and undergo senescence to prevent chro- of antigens, including signal transduction and adhesion mosomal rearrangements. molecules.161 Thus the attribution of immune dysregula- Shortened telomeres have been observed in the leuko- tion as causal in aplastic anemia based on therapy must cytes of aplastic anemia patients.148 Of five patients noted be tempered with caution because ATG and cyclosporine to have mean telomere lengths less than 5 kb, three had exert multiple effects in addition to immunosuppression. acquired cytogenetic abnormalities, suggesting that com- Of note, therapies with mixtures of monoclonal antibod- promise in the telomere’s probable role in stabilizing ies specific for human T cells have not been effective in chromosome ends may contribute to the increased inci- clinical trials to date162 (see following discussion). Fur- dence of myelodysplasias and acute leukemias. A study thermore ATG can increase colony growth in normal, by Brummendorf and colleagues reported relatively myelodysplastic syndrome (MDS), and aplastic anemia normal telomere lengths in patients who responded to CD34+ bone marrow cells in culture.163,164 ATG treat- immunosuppression, while untreated or unresponsive ment also reduces the expression of Fas-Ag on aplastic patients showed significant telomere shortening.149 Telo- anemia bone marrow CD34+ cells.165 Effective treatment mere shortening is a common feature associated with of myelodysplasia with immunosuppressive medications 168 SECTION II BONE MARROW FAILURE has been reported166 and may suggest that such agents recently demonstrated somatic alterations in 20% of might exert additional effects on the bone marrow.167 adult patients with acquired aplastic anemia utilizing Nonetheless multiple reports have demonstrated an next generation sequencing technology.176 Alterations association of aplastic anemia with alterations in lym- included mutations of DNMT3A and ASCL1, both asso- phocyte numbers or specific immunologic changes at ciated with MDS in adults, and mutations of β2- presentation and in some cases further changes with microglobulin, which is required for class I expression hematologic responses. Hoffman and colleagues showed and CD8 T-cell function. Thus previously unrecognized inhibition of erythroid growth in vitro utilizing coculture mutations may predispose to acquired aplastic anemia of peripheral blood lymphocytes from five out of seven via either mechanisms leading to a prodrome of myelo- aplastic anemia patients studied. 168 Zoumbos and col- dysplasia or by subtle alternations in immune functions. leagues noted increased circulating and bone marrow Limitations of these studies include the paucity of puri- levels of IFN-γ in aplastic anemia patients and a response fied cell populations required for sequencing and the lack of increased colony growth in vitro in response to inhibi- of functional data that addresses the biologic relevance tion with neutralizing antibody to this proinflammatory of these alterations with respect to stem cell function cytokine.169 Hinterberger and colleagues failed to confirm and hematopoiesis. Additional studies will undoubtedly the reported increased circulating levels of proinflamma- follow. tory cytokines in aplastic anemia patients, but did dem- onstrate increased TNF and IFN-γ production by Lymphokines peripheral blood mononuclear cells (PBMCs) from aplas- Whether overproduction or dysregulated production of tic anemia patients at baseline and after stimulation in inhibitory cytokines represents a primary etiology versus vitro.170 Risitano and colleagues reported a dominant a secondary effect of an underlying bone marrow abnor- clonal T-cell immune response characterized by flow mality remains unclear. IFN-γ, a soluble inhibitor pro- cytometry using antibodies against different TCR-β sub- duced by T cells, was found to be produced at high levels families and by sequencing the CDR3 region of the in cultures from aplastic anemia patients.177,178,169 IFN-γ TCR-β chain in 54 patients studied with aplastic is expressed in the marrow of most patients with aplastic anemia.171 The size of these clones and the total number anemia but not in normal people or in patients with other of T cells decreased following successful treatment with hematologic diseases who have undergone frequent trans- ATG and increased with relapse. This phenomenon was fusions.179,180 High pretreatment intracellular γ-interferon associated with HLA-DR1501 genotype and activated levels correlated with response to immunosuppressive cytotoxic T cells assayed in vitro. The association with therapy.181 Other inhibitory cytokines, such as TNF182 HLA type was not confirmed by a subsequent study of and macrophage inflammatory protein-1,183,184 also are Japanese patients.172 Solomou and colleagues reported overexpressed in aplastic marrow. Both interferon and reduced T regulatory cells (TRegs) (CD4+, CD25+, TNF directly and synergistically inhibit hematopoiesis in FoxP3+) in 20 patients with aplastic anemia that was vitro.185 In long-term bone marrow culture, constitutive consistent with increased IFN-γ and a TH1 polarization low-level expression of IFN-γ is sufficient to markedly phenotype of T cells.173 Six of seven patents showed reduce the output of committed progenitor cells and LTC- slightly increased numbers of TRegs after immunosup- ICs, the stem cell surrogate.186 Both interferon and TNF pression therapy (IST) during hematologic responses. In increase the potential for programmed cell death within a case report, Ikeda and colleagues noted marked altera- the CD34+ compartment by increasing Fas antigen tion of the CD4/CD8 T-cell ratio after hepatitis that was expression on target cells.187 Fas antigen is a cell surface subsequently followed by onset of aplastic anemia.174 IST molecule in the TNF receptor family; its activation signals was followed by normalization of lymphocyte subsets apoptosis. with hematologic recovery. These studies imply concomitant immune dysregula- Microenvironment tion with aplastic anemia and suggest a mechanism by In theory aplastic anemia could be due to a microenviron- which IST could be working therapeutically. However, ment that fails to support hematopoiesis: a lesion of no specific antigen has previously been implicated mech- “soil” rather than “seed.”72 The role of the HM in etiol- anistically in the evolution of aplasia. In a recent report, ogy of aplastic anemia or the response to treatment it was shown that a prodrug of the anti-HIV drug, aba- remains controversial. Ershler and colleagues showed cavir, which is associated with a hypersensitivity reaction decreased erythroid growth in whole bone explants from in treated patients, can bind to the HLA-B*57:01 mole- a single patient with constitutional red cell aplasia but no cule, resulting in altered peptide presentation.175 The effect from the patient’s plasma, mononuclear cells, or T altered peptides appear as foreign and lead to activation cells, implying an HM defect.188 This observation is remi- of CD8+ T cells. One can imagine such a scenario leading niscent of the HM defect seen in Steel mice,189 which lack to acquired aplastic anemia if the abnormal peptide pre- production of normal stem cell factor (SCF) in nonhema- sentation of “self-antigens” leads to CD8 activation topoietic cells, but it seems more likely that this was directed against hematopoietic stem cells in the bone simply a demonstration of a cell-intrinsic erythroid cell marrow. While such a finding is not reported yet in aplas- defect. Dexter and colleagues described an in vitro long- tic anemia patients, this could underlie the immunologic term marrow culture (LTMC) that consisted of an adher- mechanism long hypothesized as the etiology of this ent cell compartment made up of a number of different disease. Interestingly Weinstock and colleagues also cell types that supported hematopoiesis for weeks to 6 Acquired Aplastic Anemia and Pure Red Cell Aplasia 169 months ex vivo.190 Subsequently Marsh and colleagues have not examined long-term hematopoietic stem cells in examined LTMC from 32 adult aplastic anemia patients functional assays. Alterations of the sinusoidal endothe- and found 31 of the 32 had abnormal hematopoiesis in lium with antibodies to VE-cadherin leads to failure of these cultures.191 However, using cross-over cultures, only megakaryocytopoiesis in murine models.205 Some models 1 of 23 patients had defective adherent components. suggest that quiescent hematopoietic stem cells may Juneja and colleagues demonstrated reduced cobblestone migrate from the osteoblastic niche to the perivascular areas (containing active hematopoiesis) but normal adher- niche via β1 integrin function and chemokine/growth ent cell fractions in eight aplastic anemia patients com- factor stimulation. Overall, whether there are truly dis- pared with normal controls.192 Marsh and colleagues tinct functional differences between these putative hema- utilized human LTMC and assessed the HM in cross-over topoietic stem cell niches remains to be determined, and experiments.193 They demonstrated that stroma generated the role of specific growth factors, adhesion molecules, from aplastic anemia patients performed normally when and chemokines are incompletely understood. A long- inoculated with CD34+ cells from normal donors. Nissen term goal of investigators in this field is to recapitulate and colleagues compared short-term stroma and progeni- the HM in vitro such that true expansion of hematopoi- tor colony growth in aplastic anemia patients treated etic stem cells may occur in the laboratory setting. Success with or without IST and found a modest increase in both in this area would have a major impact in BMT and of these cell fractions in treated versus untreated regenerative medicine. patients.194 Chatterejee and colleagues used a cytotoxic Hematopoietic growth factor production and plasma drug animal model of aplasia to demonstrate failure of levels are usually increased rather than decreased in recovery of marrow stromal cells with time.195 While this patients with aplastic anemia.207 Circulating levels of study may be relevant to some isolated instances of erythropoietin, granulocyte colony-stimulating factor acquired aplastic anemia, it seems more relevant to (G-CSF), and granulocyte-macrophage colony-stimulating marrow damage in cancer or leukemia patients undergo- factor (GM-CSF), thrombopoietin, and flt-3 ligand are ing therapy. Shipounova and colleagues showed impaired elevated in patients with aplastic anemia. SCF levels have adipogenesis and osteogenic potential with increased size been reported to be low to normal. Levels of interleukin and number of fibroblast colonies from aplastic anemia 1 (IL-1), produced by monocytes, may be low in aplastic patients’ bone marrow.196 These authors also demon- anemia. Therapeutic trials with these factors have yielded strated increased stromal-derived cytokines, including divergent and incomplete responses,208 casting doubt on ANG1, VEGF, and VCAM-1. Overall these studies the pathophysiologic significance of deficiency of these suggest some alterations in the HM of aplastic anemia factors. The report of inhibitory effects of marrow adi- patients, but there is currently no definitive evidence in pocytes on hematopoiesis in mouse models is intriguing humans of a mechanistic link between defective marrow given the typical fatty replacement observed in aplastic microenvironment and development of acquired aplastic bone marrows.209 anemia. More recently interest has focused on defining specific Clinical Evaluation anatomic and/or biochemical spaces (niches) in the bone The differential diagnosis of pancytopenia is broad (Box marrow occupied by hematopoietic stem cells. Schofield 6-3). The evaluation and diagnostic work up of the first termed the phrase stem cell niche to define a sup- patient with presumed acquired aplastic anemia should porting cellular environment in which hematopoietic be directed at eliminating alternative diagnoses for which stem cells reside.197 Currently controversy exists about curative therapies are available or for which the therapies the overall relevance of the two best-studied stem cell for aplastic anemia would be inappropriate (Table 6-1). niches.198 Hematopoietic stem cells appear to reside in Because the inherited marrow failure syndromes entail close proximity to osteoblasts that line the cortical bone different medical management issues, a careful medical of the marrow space, and osteoblasts may be important history, family history, physical examination, and appro- in maintaining quiescence of the primitive hematopoietic priate laboratory evaluation to assess for underlying stem cells. This area of the marrow has thus been termed inherited bone marrow failure syndromes should be the osteoblastic niche. A subset of these cells, spindle- pursued. The diagnosis of an inherited marrow failure shaped N-cadherin+ CD45− osteoblasts (SNO cells) resid- syndrome also carries profound implications for hemato- ing in trabecular bone, may be derived from mesenchymal poietic stem cell donor selection from family members stem cells, and their number correlate with the content and may be important for family planning. Given the of long-term hematopoietic stem cells.199 Ablation of wide clinical variability of the inherited marrow failure osteoblasts leads to loss of HSC200 and failure of hema- syndromes even amongst members of a given family, all topoiesis, while manipulations that increase osteoblasts siblings of a patient with an inherited marrow failure have the opposite effect.201 disorder should be tested whether or not they appear to An anatomically distinct putative hematopoietic stem carry clinical stigmata of the disease. cell niche is made up in part by sinusoidal endothelial cells and has been termed the perivascular niche.202 Some History data suggest this niche may be critical for mobilization The chief presenting symptoms relate to low blood and homing of hematopoietic stem cells via the Rac counts. Bleeding, such as gum oozing, nosebleeds, easy GTPase pathway203,204 and/or for progenitor cell prolif- bruising with minimal trauma, or heavy menses may be eration and differentiation,205,206 although these studies seen. Chronic anemia may present with fatigue, decreased 170 SECTION II BONE MARROW FAILURE Box 6-3 Causes of Pancytopenia membranes. Neutropenia may be associated with oral ulcerations, bacterial infections, and fever; these signs are HYPOCELLULAR BONE MARROW rarely present early. Evidence of erythropoietic failure Acquired aplastic anemia characterized by pallor, fatigue, and tachycardia is often Inherited bone marrow failure syndrome late because the life span of the erythrocyte (120 days) (Fanconi anemia, amegakaryocytic thrombocytopenia, dyskeratosis far exceeds that of platelets (10 days) or white cells (vari- congenita, Shwachman–Diamond syndrome, GATA-2 syndromes) Paroxysmal nocturnal hemoglobinuria* able, but measured in hours for granulocytes). Mucous Hypoplastic myelodysplastic syndrome membranes and nail beds may be pale. Lymphadenopa- Virus-associated aplastic anemia thy, splenomegaly, and severe weight loss are uncommon CELLULAR BONE MARROW and may suggest other underlying disorders. Short stature, PRIMARY BONE MARROW DISEASE congenital anomalies (particularly of the thumbs and forearms), areas of hyper- or hypopigmentation, dystro- Myelodysplasia Paroxysmal nocturnal hemoglobinuria* phic nails, immunologic abnormalities, or pulmonary disease should alert the examiner to possible inherited SECONDARY TO SYSTEMIC DISEASE bone marrow failure disorders. Immunologic abnormali- Systemic lupus erythematosis, Sjögren syndrome ties or lymphedema raises consideration of inherited Hypersplenism Vitamin B12, folate deficiency GATA-2 syndromes (see Chapter 24). Infection Storage disease (Gaucher, Niemann-Pick) Laboratory Studies Alcoholism Evaluation should include a complete blood count Sarcoidosis and reticulocyte count. A careful evaluation of the periph- Infiltrative bone marrow disorders Acute myelogenous leukemia eral blood smear may suggest infection or dietary defi- Acute lymphoblastic leukemia ciency. Blood counts are most often uniformly depressed. Hemophagocytic lymphohistiocytosis The blood smear shows a paucity of platelets and leuko- Metastatic solid tumors cytes. The anemia is often macrocytic and particularly Osteopetrosis Myelofibrosis useful in pointing to a possible constitutional syndrome, though if erythropoiesis is entirely arrested, it may be normocytic. The red cell distribution width (RDW), a numeric measure of anisocytosis, is most often normal. Platelet size is not increased, as it is in most cases of activity, or exercise intolerance, though the gradual onset immune thrombocytopenias. The absolute reticulocyte of severe anemia can be surprisingly well compensated in count is decreased. Granulocyte numbers are clearly young children. Serious infection is not a frequent pre- diminished, as may be those of monocytes and lympho- senting symptom early in the course of aplastic anemia. cytes.211,212 Increases in fetal hemoglobin (Hb F) and red Many patients feel well with few symptoms on initial cell i antigen are manifestations of the fetal-like erythro- presentation. A history of steatorrhea or fatty food intol- poiesis seen in stress hematopoiesis213 but occur irregu- erance may be a manifestation of Shwachman–Diamond larly in aplastic anemia patients, have no prognostic syndrome, though the absence of these symptoms does value, and can persist in recovered patients. not rule out this diagnosis.210 Gastrointestinal disorders Laboratory evaluation for possible infectious etiolo- may also be associated with other inherited marrow fail- gies should be obtained as clinically indicated. Additional ures syndromes such as dyskeratosis congenita or Fanconi testing for possible immunologic or autoimmune diseases anemia. A careful family history for blood disorders, may be considered. Blood tests to rule out Fanconi anemia malignancies (particularly ovarian and breast cancer or and PNH are important, as these diagnoses would entail hepatocellular carcinoma), premature graying, hepatitis, alternative treatments. Telomere length measurements pulmonary fibrosis, or congenital anomalies that might may be useful as a screen for possible underlying telomere suggest an inherited bone marrow syndrome should be disorders. Very short telomeres in long-lived lymphocyte sought. Patients with a history suggestive of a familial populations should prompt consideration of additional marrow failure syndrome or familial MDS should be genetic workup for dyskeratosis congenita. Data on the evaluated further with genetic and laboratory studies. A use of telomere measurements in isolation to guide clini- developmental history can provide a helpful screen for cal treatment decisions in this setting are sparse. A potential congenital, metabolic, or storage diseases. A growing array of genetic testing is available to assist in detailed medication, environmental, and infectious the diagnosis of many of the inherited marrow failure history, particularly for the period 1 to 12 months prior syndromes (see Chapter 7). Patients with stigmata of col- to presentation, should be obtained. lagen vascular disease should be appropriately evaluated. Early HLA typing of the patient and family members is Physical Examination helpful in guiding future therapies. Baseline renal and Clinical appearance is related to the severity and duration hepatic laboratory evaluations are also indicated prior to of the underlying pancytopenia. Hemorrhagic manifesta- the initiation of potentially nephrotoxic or hepatotoxic tions from thrombocytopenia occur early and include therapies. petechiae (particularly on the border of the hard and soft Bone marrow examination must be done by both aspi- palate), ecchymoses, epistaxis, and bleeding from mucous ration and biopsy so that cellularity can be evaluated 6 Acquired Aplastic Anemia and Pure Red Cell Aplasia 171 TABLE 6-1 Clinical Evaluation340 Clinical History Rationale Recent illnesses Infectious etiology Medications Marrow suppression Toxic exposures Marrow suppression Family history of blood disorders, malignancy, congenital anomalies Assess for inherited marrow failure Physical Examination Vital signs Clinical sequelae of cytopenias Height and weight Assess for inherited marrow failure Congenital anomalies, stigmata Assess for inherited marrow failure Assess for other systemic diseases Laboratory Tests Complete blood count and differential Defines severity Morphology Malignant versus benign Vitamin B12 deficiency Storage disease Reticulocyte count Defines severity Differentiate production versus destruction Bone marrow biopsy Assess cellularity Assess architecture (granuloma, fibrosis, hemophagocytosis, infiltrative or metastatic disease) Bone marrow aspirate Morphology Malignant versus benign Storage disease Hemophagocytosis Congenital disorder Cytogenetics Myelodysplasia Culture Infectious agent (e.g., tuberculosis, virus) Other DNA/antigen-based viral tests Peripheral blood Chemistry AST, ALT, GGT, Bilirubin, LDH Hepatitis Coombs test Autoimmune cytopenias BUN, creatinine, electrolytes Chronic renal failure Serologic testing/PCR Hepatitis, EBV, HIV, other virus Flow cytometry for CD55/CD59 Paroxysmal nocturnal hemoglobinura Diepoxybutane or mitomycin C chromosomal breakage Fanconi anemia Telomere length, dyskeratosis congenita Autoimmune disease evaluation Evidence of collagen vascular disease Histocompatibility testing of patient and family Establish potential donor pool ALT, Alanine aminotransferase; AST, aspartate aminotransferase; BUN, blood urea nitrogen; GGT, γ-glutamyl transferase; LDH, lactate dehydrogenase. both qualitatively and quantitatively. The bone marrow asynchrony, may be seen. Aspirates alone may appear in aplastic anemia is hypocellular, with empty spicules, hypocellular owing to dilution with peripheral blood, or and increased fat, reticulum cells, plasma cells, and they may look hypercellular because of areas of focal mast cells. In young children there may be residual residual hematopoiesis. Biopsies provide more reliable lymphocytes that appear prominent due to the lack of specimens for assessment of cellularity. Biopsies also myeloid elements in the marrow cavity. Some dyseryth- provide critical information regarding bone marrow ropoiesis, with megaloblastosis and nuclear-cytoplasmic architecture such as fibrosis or granulomas. Bone marrow 172 SECTION II BONE MARROW FAILURE aspirates should also be sent for karyotype analysis and never be from the patient’s family for the same reason. fluorescence in situ hybridization (FISH) to assess for Complete red cell phenotyping should be considered clonal cytogenetic abnormalities associated with myelo- where feasible to minimize the potential risk of erythro- dysplasia or malignancy. While FISH studies are more cyte antibodies in patients who may receive long-term sensitive, they can only detect the specific chromosomal blood product support. All blood products should be abnormality tested. Bone marrow may also be sent for irradiated because standard treatments are associated culture or DNA-based antigen detection of infectious with profound immunosuppression. Leukoreduction agents such as viruses, though interpretation is tempered appears prudent to reduce the potential risk of infection by the frequent persistence of viral DNA long after the transmission and allosensitization because aplastic anemia active infection has passed. patients may require a hematopoietic stem cell transplant. Exposure to potentially hazardous drugs or toxins should be eliminated. Treatment with single hematopoietic Management and Outcome growth factors, such as granulocyte-colony stimulating Prognosis factor (G-CSF) or erythropoietin should be avoided in Outcome depends on the types of treatments offered, the part due to the resulting delay in definitive therapy.222 causes of the aplasias, and the eras and countries being analyzed. In a large series conducted before 1957, Wolff Bleeding found a survival rate of only 3%.214 In other series ending Platelet (and red cell) support has probably had the great- no later than 1965, complete recoveries were reported in est impact on survival in aplastic anemia and has changed 10% to 35% of patients. In a series of 40 pediatric the cause of death from bleeding to infection. Platelets patients seen before 1958, Shahidi and Diamond found can be provided from several individual units of blood that only two patients recovered, one of whom later had or, preferably, by platelet pheresis from a single donor to recurrence,215 though these studies may have included reduce antigenic exposure (see Chapter 36). patients with moderate disease for whom spontaneous The main role of prophylactic platelet support is recovery is more common.216,217 Long-term prognosis is reduction of the risk of intracranial hemorrhage, which adversely affected by delayed complications such as clonal is a rare but potentially devastating event. The general disease. The clinical course of 24 pediatric patients with threshold platelet count has traditionally been 20,000 moderate aplastic anemia was followed for a median of cells/µL; however, this figure and the entire practice of 66 months (range 10 to 293 months).218 Moderate aplas- prophylactic administration of platelet transfusions have tic anemia was defined as marrow hypoplasia of less than recently been questioned.223,224,225 Platelet transfusion at 50%, at least two cytopenias such as an absolute neutro- platelet counts below 10,000/µL in stable outpatients phil count (ANC) less than 1500/mm3, platelet count less with chronic severe aplastic anemia was feasible and than 100,000/mm3, or reticulocytopenic anemia lasting safe.226 Chronic platelet transfusions for aplastic anemia at least 6 weeks. Sixteen patients (67%) progressed to are generally given when there are symptoms of bleeding severe aplastic anemia, five patients (21%) remained in or if the patient is at increased bleeding risk (e.g., toddlers the moderate range, and three patients (12%) resolved. learning to walk, patients with hypertension while on It is currently unclear whether early institution of therapy cyclosporine, patients with fever and infection). In some for moderate aplastic anemia confers improved outcome. institutions a higher platelet transfusion trigger point of Because current therapies are associated with significant 20,000/µL is used during initiation of IST. side effects, their institution for children with moderate Other measures to reduce bleeding include mainte- aplastic anemia, for whom the marrow aplasia may spon- nance of good dental hygiene, use of a soft toothbrush, taneously resolve, warrants careful consideration. stool softeners, and avoidance of trauma. Drugs that may interfere with platelet function, such as aspirin or nonste- Spontaneous Recovery roidal antiinflammatory drugs, should be avoided in Spontaneous recovery has been reported anecdotally, thrombocytopenia. Antifibrinolytic agents such as amino- although most cases were probably of moderate rather caproic acid and tranexamic acid may decrease bleeding, than of severe aplastic anemia. Good supportive care (see particularly from the gums and nasal mucosa. following discussion) may contribute to this; in one series, 14 of 33 children recovered,219 and children with moder- Infections. There are few reported studies of infection in ate aplastic anemia have been found to have an excellent aplastic anemia.227,228 Severe granulocytopenia may last prognosis even without specific treatment.220 A study of for years, and infection remains the leading cause of immunosuppressive therapy in which 21 patients were death in aplastic anemia patients, particularly bacterial randomized (some to receive only supportive care) found and fungal infections.229,230 However, the immune systems no improvement in the control group after 3 months.221 of granulocytopenic patients remain intact; this is in con- Spontaneous recovery is sufficiently rare in severe aplastic trast to the situation of patients with malignancies who anemia that all modalities of current therapy should be receive chemotherapy and who experience simultaneous initiated. neutropenia and immunosuppression (as during IST for aplastic anemia). Neutropenia (and perhaps monocyto- Supportive Care penia) increases the risk of bacterial infection in aplastic Blood product support should be used judiciously to anemia.211 Because neutropenia precludes the develop- avoid sensitization, and blood product donors should ment of an inflammatory response, identification of an 6 Acquired Aplastic Anemia and Pure Red Cell Aplasia 173 infected locus is often difficult. The bacterial organisms indicated in the persistently febrile patient who is unre- may be gram-negative bacilli such as Escherichia coli, sponsive to antibiotics or in the appropriate clinical Klebsiella pneumoniae, and Pseudomonas aeruginosa, as setting. well as gram-positive cocci, including Staphylococcus aureus and S. epidermidis and streptococci. Use of G-CSF Anemia. Red cells should be provided as clinically indi- has been studied in a few prospective analysis and other cated. The hemoglobin should be maintained at a level randomized trials, and these studies have been examined consistent with normal activities if routine transfusions by meta-analysis. Gluckman and colleagues randomized are to be used. Chronic anemia can be well tolerated once aplastic patients receiving IST to prophylactic G-CSF adaptation has occurred, and some children who can treatment or not.231 These investigators reported enhanced sustain a hemoglobin level greater than 6 to 7 g/dL recovery of neutrophils in the G-CSF group but no effect without transfusions (i.e., a child who is not bleeding) on overall survival. More recently, Tichelli and colleagues may be carefully observed. Long-term transfusions lead randomized 192 aplastic patients to ATG/cyclosporine to iron overload, with accumulation in critical organs. +/− G-CSF and reported significantly fewer infectious Permanent damage to heart, liver, and endocrine glands episodes and hospitalization days in the G-CSF group from iron overload may be prevented by iron chelation but no differences in overall survival or event-free sur- therapy.234 vival between groups.230 Interestingly they found that the lack of neutrophil response at day 30 post initiation of Treatment treatment was significantly associated with a lower Hematopoietic Stem Cell Transplantation. Hematopoi- response rate overall and a lower survival rate. They etic stem-cell transplant offers curative therapy for aplas- postulated that if confirmed by additional studies, the tic anemia. The general topic of BMT is described in lack of neutrophil recovery at 30 days posttreatment Chapter 8. The role of BMT in aplastic anemia treatment could identify a group of patients in which alternative is discussed in Chapter 8. treatment might begin earlier than in the past. This study did not confirm a prior study by Teramura and col- Immunosuppression leagues that reported a lower incidence of relapse in Antithymocyte Globulin. Antilymphocyte globulin patients treated with G-CSF.232 Meta-analysis of a large (ALG) and ATG (the sera produced by immunization of number of studies has also reported that in aggregrate horses or rabbits with human thoracic duct lymphocytes these studies showed no effect of G-CSF on overall sur- or thymocytes) are highly cytotoxic reagents with activity vival, hematologic responses, or specific infections and against all blood and marrow cells, including progenitors. no association of the use of G-CSF with MDS, AML or These reagents were initially used to decrease the rate of PNH.233 rejection of HLA-matched bone marrow. ALG and/or Immunosuppression due to preparation for BMT or as cyclophosphamide were also employed to permit at least primary therapy for the aplastic anemia may lead to temporary engraftment with mismatched marrow in unusual bacterial, fungal, viral, and protozoan infections. aplastic patients without a matched donor.157,158,159,235,236 Recommendations for specific antibiotics and other anti- Autologous recoveries occurred in some of these patients; infection agents are beyond the scope of this chapter, and they encouraged several groups to examine the use of regimens are changing rapidly as new generations of ATG or ALG with and without haploidentical marrow. treatments are developed. Treatment with ALG was superior to controls in both The use of routine prophylactic antibiotics has no response rate and 3-year survival.221 The addition of addi- demonstrated role in the “well” patient with aplastic tional agents to ALG therapy can further enhance thera- anemia. In many institutions, guidelines are derived from peutic effect. experience in neutropenic cancer and post–BMT patients ATG or ALG/ALS is often given to a total cumulative because prospective studies in pediatric aplastic anemia dose of 100 to 160 mg/kg, typically divided over 4 days. patients are rare.229 In the opinion of many, even without Immediate allergic reactions to ALG are rare and can be supporting outcomes data, aplastic anemia patients with predicted by skin testing followed by desensitization in ANC less than 500 should receive antibacterial and anti- those who are allergic.237 The patient begins to make fungal prophylaxis. Patients undergoing IST are typically antibodies to horse protein about 1 week after first expo- given prophylaxis for Pneumocystis carinii, although sure; at this point, equine antibodies are rapidly cleared data supporting the need for this treatment are sparse or from the circulation. This reduces the effective dose and nonexistent. In the context of fever and neutropenia, enhances immune complex formation. Serum sickness complete evaluation and cultures of all possible sites due to immune complex deposition may manifest around should be followed by the administration of broad- 11 days after initiation of treatment as fever, a serpigi- spectrum parenteral antibiotics. Fungal infections occur nous rash at the volar–dorsal border of the hands and frequently in neutropenic patients who have received feet, arthralgia, myalgia, lassitude, and changes in urine repeated or extended courses of antibiotics; candidiasis sediment. Bronchospasm or liver chemistry abnormalities and especially aspergillosis, which lead to sinusitis, lung may also occur. A short course of corticosteroids is disease, or disseminated infection, are distressingly fre- usually given (prednisone, 1 mg/kg) beginning at the start quent causes of death in aplastic anemia.228 Antecedent of ATG treatment and for 10 to 14 days to reduce ATG- or concurrent administration of steroids increases this related side effects. ATG binds to determinants on all cell risk. Aggressive introduction of antifungal treatment is types in blood and marrow and reduces platelet and 174 SECTION II BONE MARROW FAILURE granulocyte counts in addition to the lymphocyte counts, at the NIH reported a response rate of 67% at 43 months and it may lead to a positive Coombs test. Thrombocy- and a 5-year actuarial survival rate of 70% (86% for topenia may require intensive blood product support responders and 16% for nonresponders).253 The Gruppo during this time.238 Italiano Trapianti di Midolio Osseo (GITMO)/European Responses to ATG given as a single agent usually occur Group for Blood and Marrow Transplantation (EBMT) within the first 3-months, if at all, and blood count study of 100 patients with severe aplastic anemia includ- improvement by 3 months correlates with survival.239 The ing children (median age 16 years) treated with ALG, earliest response to therapy may be heralded by the cyclosporine, prednisolone, and G-CSF found trilineage appearance of a few granulocytes and nucleated red cells hematopoietic response in 77% after one or more courses in the circulation. Red-cell size distribution histograms of ALG.254 Several trials in children have reported the typically show a macrocytic shoulder.240 Reticulocytes efficacy of combined ATG and cyclosporine in the pedi- appear, transfusions decline, and the hemoglobin level atric population with an overall response rate of close to increases slowly. The white cell count rises next. The last 80%255,256,250,251,257 (Table 6-2). Although greater lympho- cell line to return is often the platelets, whose numbers cyte depletion is achieved with rabbit ATG as compared may remain low for months or even years. The recovering to that with horse ATG, a randomized trial comparing red cells are not normal; in addition to macrocytosis, Hb horse ATG versus rabbit ATG in the upfront treatment F level is increased, and fetal membrane antigens may of aplastic anemia demonstrated that superior hemato- remain present.241 Long-term survivors (except those with logic response (68% versus 37%) and survival (96% successful transplants) often have residual abnormalities, versus 76%) was achieved with horse ATG.258 such as thrombocytopenia, red cell macrocytosis, and ele- ATG is typically given over a 4-day course in combina- vated Hb F levels.242 Clearly we do not understand either tion with a 6 to 12 month course of cyclosporine. The the mechanism of action of this poorly characterized drug, lack of a unified definition for response complicates com- the ultimate course of patients with ATG-induced remis- parison of different studies, but generally a complete sions, or the basis of their partial recovery. response (CR) refers to normal or functionally normal Cyclosporine. Cyclosporine (formerly cyclosporin A) blood counts, a partial response (PR) denotes hemoglobin is a fungal cyclic undecapeptide and an effective immu- and platelet counts adequate to avoid the need for trans- nosuppressive agent.243 Among its many effects, cyclospo- fusion, and neutrophil counts typically greater than rine can inhibit T cells, preventing production of IL-2 and 500/µL, and no response commonly refers to continued IFN-γ.160 About one half of patients who had failed ATG severe aplastic anemia. Improvements in blood counts are or ALG treatment subsequently experienced remission delayed and may not be evident for many weeks. In a with cyclosporine.244,245 Treatment with cyclosporine study including both children and adults, response rates, alone yielded significantly lower response rates and higher defined as at least a PR in two blood cell lineages, were rates of disease progression as compared with combined 67% at 3 months and 71% at 6 months following initia- therapy.246,247 tion of combined ATG and cyclosporine therapy.252 Oral cyclosporine should be administered twice daily Cyclosporine should be tapered gradually with close to maintain blood trough levels between 100 and 250 ng/ monitoring of the blood counts prior to each decrease in mL as measured by radioimmunoassay, though the target dosage. A persistent drop in the blood counts can often levels may vary depending on laboratory methods used be treated with either an increase in cyclosporine dosage to determine cyclosporine levels. Hematologic responses or reinitiating the full dose. Some patients require con- can take weeks to months, and an initial trial period of tinuous cyclosporine, and these patients should be main- 3 to 6 months is generally recommended. tained on the lowest effective dose. Relapse risk was Toxic effects from cyclosporine use are not insignifi- reduced when cyclosporine was tapered slowly.259 In a cant and include hypertension, hypertensive encepha­ review of ongoing trials by the German multicenter group lopathy, azotemia, hirsutism, gingival hypertrophy, and the NIH, 29% and 14% of initial responders, respec- coarsening of facial features, tremor, immunodeficiency, tively, were found to require continuous treatment.253 The increase in serum creatinine levels, transient or irrevers- reoccurrence of frank pancytopenia generally requires a ible nephrotoxicity, hepatotoxicity, seizures, and P. carinii second course of ATG followed by full-dose cyclosporine. pneumonia.243 As with ATG, in vitro tests of the effect The relapse rate after combined IST varies widely from of cyclosporine on progenitor cells do not predict study to study. Most studies do not include only children. responses.248 Kamio and colleagues recently reported a relapse rate of 16% in 264 responding aplastic patients260 (which com- Combination Immunosuppressive Therapy. The combi- pared to 33% reported in an NIH study).261 Relapse rates nation of ATG with cyclosporine has resulted in higher were even lower in a Japanese study.232 Patients treated rates of hematologic recovery than either drug with IST often continue to have subnormal counts and alone.246,247,249 Importantly, intensive IST has greatly may remain macrocytic. While the majority of patients improved the results of therapy in two groups commonly respond within 6 months, delayed responses beyond that refractory to ALG or ATG alone: children and patients time may be seen for some patients. with very severe disease.250,251 In the National Institutes Around 20% of patients in pediatric studies fail to of Health (NIH) Clinical Center trial,252 survival curves respond to ATG and cyclosporine therapy (Table 6-2), of those older and younger than 35 years of age were while a recent study of mostly adult patients reported similar. A large study including both children and adults failure of response in around 40% of patients.262 In 6 Acquired Aplastic Anemia and Pure Red Cell Aplasia 175 TABLE 6-2 Pediatric Studies of Immunosuppressive Therapy for Aplastic Anemia Patient Follow-Up Clonal Study Years Number Treatment (years) Response Survival Relapse Abnormality Fuher 1993-1997 86 (49vSAA, ALG, 4 80% (OR) 87% 15% 3% clonal et al255 30SAA, CSA, abnormalities 7MAA) G-CSF* 4.7% AML Kojima 1992-1997 119 (50 ATG, 4 71% (OR) 83% vSAA 13% vSAA 3% et al250 vSAA, 36 CSA, vSAA 91-93% 29% SAA, 33 DAN, 61%(OR) MAA+SAA MAA/ MAA) +/-G- SAA+G- SAA CSF CSF, 83% +G-CSF (OR) 64% SAA-G- MAA/ CSF SAA -G-CSF Kojima 1992-1997 113 ATG, Median 5.3 30% CR 87% NR 13.7% et al278 (39vSAA, CSA, Range 31% PR cumulative 47SAA, DAN, 3.75-8.9 42% NR incidence 27MAA) +/- at 6 MDS at 8 G-CSF months years, 21.9% cumulative incidence clonal cytogenetic abnormalities at 8 years Goldenberg 1990-2003 14 ATG, CSA Median 4.4 93% (OR) 93% 0 0 et al257 +/-G- Range CSF 0.8-13.3 Fuher 1993-2001 97 vSAA ATG CSA Median 4.1 69% (CR) 93% vSAA 13% vSAA NR et al251 G-CSF vSAA 49 SAA 44% (CR) 81% SAA 14% SAA NR SAA CR, Complete response; CSA, cyclosporine A; DAN, danazol; MAA, moderate aplastic anemia; NR, not reported; OR, overall response; PR, partial response; SAA, severe aplastic anemia; vSAA, very severe aplastic anemia. *G-CSF added for SAA and vSAA. most studies, failure of response is typically defined as reported no difference in response rate or survival in persistence of severe aplastic anemia. In adult patients patients with moderate to severe aplastic anemia.268 failing the first treatment with horse ATG, 63% of patients Studies by Kaltwasser and colleagues269 and the EBMT270 receiving a second course of horse ATG responded,263 found significantly higher response rates after ALG plus while 77% (23 out of 30) of patients initially treated with androgens (73% and 56%) than in patients receiving horse ATG and cyclosporine responded to a second ALG alone (31% and 40%). A recent retrospective study course with rabbit ATG and cyclosporine.264 In some in adults reported an overall response rate of 77% and small studies in children, the response rate to second overall survival rate of 78% at 5 years following treat- immunosuppressoin has been less,265 and treatment with ment with ATG plus androgens.271 At least one report rabbit ATG as a second therapy was associated with only showed increased relapse after ATG and cyclosporine 27% response rate.266 In a large NIH study that included when danazol was included in the treatment.260 Because 77 children less than 18 years of age, of 14 relapsed some patients with inherited causes of aplastic anemia patients, 6 were retreated with ATG and were alive at may experience hematologic improvement following report, but the hematologic response of these patients androgen therapy,272 it is important to rule out an under- was not reported.261 Less acute reactions were noted lying inherited marrow failure syndrome. Androgen side when the source of ATG was varied between the first and effects may be unacceptable to some patients, and their second treatments. The time frame to response following use in acquired aplastic anemia in children is generally a second course of immunosuppression was similar to limited. that observed following initial treatment. Patients failing Relapse. Relapse after immunosuppressive therapy to respond to two courses of immunosuppressive therapy has been reported in around one third of patients in are unlikely to respond to a third treatment course.267 studies including both children and adults,262,273 although Because androgens are effective in treating some lower rates of relapse may be attained with slow tapering patients with aplastic anemia (see following discussion), of cyclosporine and continued maintenance doses at the the effect of combining androgen treatment with ATG lowest required cyclosporine dose for some patients. Lack was examined in several trials. Champlin and colleagues of standardized definitions of relapse hamper comparison 176 SECTION II BONE MARROW FAILURE between studies. An EBMT study including both children clonal transformation of stem cells may underlie aplas

Use Quizgecko on...
Browser
Browser