You are on page 1of 7

Aplastic Anemia

http://emedicine.medscape.com/article/198759-overview

View updated Privacy Policy, effective January 16, 2012

Aplastic Anemia
Author: Sameer Bakhshi, MD; Chief Editor: Emmanuel C Besa, MD more... Updated: Nov 1, 2011

Background
Aplastic anemia is a syndrome of bone marrow failure characterized by peripheral pancytopenia and marrow hypoplasia. Mild macrocytosis is observed in association with stress erythropoiesis and elevated fetal hemoglobin levels. Paul Ehrlich introduced the concept of aplastic anemia in 1888 when he studied the case of a pregnant woman who died of bone marrow failure. However, it was not until 1904 that Anatole Chauffard named this disorder aplastic anemia. (See Etiology.) Go to Anemia, Chronic Anemia, Megaloblastic Anemia, Myelophthisic Anemia, Hemolytic Anemia, and Sideroblastic Anemias for complete information on these topics.

Complications
Complications of aplastic anemia include infections and bleeding. (See Prognosis.) Complications of bone marrow transplantation (BMT), used in the treatment of aplastic anemia, include graft versus host disease (GVHD) and graft failure. (See Treatment.)

Staging
Staging of aplastic anemia is based on the criteria of the International Aplastic Anemia Study Group, as follows: Blood - Neutrophils less than 0.5 X 109/L; platelets less than 20 X 109/L; reticulocytes less than 1% corrected (percentage of actual hematocrit [Hct] to normal Hct) Marrow - Severe hypocellularity; Moderate hypocellularity, with hematopoietic cells representing less than 30% of residual cells Severe aplasia is defined as including any 2 or 3 peripheral blood criteria and either marrow criterion. A further subclassification developed after the recognition that individuals with neutrophil counts lower than 0.2 X 109/L had very severe aplastic anemia (VSAA). This group is less likely than others to respond to immunosuppressive therapy.

Patient education
Patients should maintain hygiene to reduce the risks of infection. Clinicians must stress the need for compliance with therapy. For patient education information, see the Blood and Lymphatic System Center, as well as Anemia.

Etiology
The theoretical basis for marrow failure includes primary defects in or damage to the stem cell or the marrow microenvironment.[1, 2, 3] The distinction between acquired and inherited disease may present a clinical challenge, but more than 80% of cases are acquired. In acquired aplastic anemia, clinical and laboratory observations suggest that this is an autoimmune disease. On morphologic evaluation, the bone marrow is devoid of hematopoietic elements, showing largely fat cells. Flow

1 of 7

12/26/2011 9:09 AM

Aplastic Anemia

http://emedicine.medscape.com/article/198759-overview

cytometry shows that the CD34 cell population, which contains the stem cells and the early committed progenitors, is substantially reduced.[2, 4] Data from in vitro colony-culture assays suggest profound functional loss of the hematopoietic progenitors, so much so that they are unresponsive even to high levels of hematopoietic growth factors. It was hypothesized that aplastic anemia may be due to a defect at various levels such as an intrinsic defect of hematopoietic cells, external injury to hematopoietic cells, and defective stroma, which is critical for normal proliferation and functioning of hematopoietic cells. Thus, theoretically, all of these mechanisms could be responsible for aplastic anemia. This theory was the basis of many in vitro stem cell culture experiments using a cross-over design in which stem cells from patients with aplastic anemia were cultured with normal stroma and vice-versa. The conclusions from these studies led to the understanding that stem cell defect is the central mechanism in the majority of patients with aplastic anemia.[5, 6] In patients with severe aplastic anemia (SAA), stromal cells have normal function, including growth factor production. Adequate stromal function is implicit in the success of BMT in aplastic anemia because the stromal elements are frequently of host origin. The role of an immune dysfunction was suggested in 1970, when autologous recovery was documented in a patient with aplastic anemia in whom engrafting failed after BMT. Mathe proposed that the immunosuppressive regimen used for conditioning promoted the return of normal marrow function. Since then, numerous studies have shown that, in approximately 70% of patients with acquired aplastic anemia, immunosuppressive therapy improves marrow function.[3, 7, 8, 9, 10] Immunity is genetically regulated (by immune response genes), and it is also influenced by environment (eg, nutrition, aging, previous exposure).[11, 12] Although the inciting antigens that breach immune tolerance with subsequent autoimmunity are unknown, human leukocyte antigen (HLA)-DR2 is overrepresented among European and United States patients with aplastic anemia, suggesting a role for antigen recognition, and its presence is predictive of a better response to cyclosporine. Suppression of hematopoiesis is likely mediated by an expanded population of the cytotoxic T lymphocytes (CTLs) CD8 and HLA-DR+, which are detectable in the blood and bone marrow of patients with aplastic anemia. These cells produce inhibitory cytokines, such as gamma-interferon and tumor necrosis factor, which can suppress progenitor cell growth. Polymorphisms in these cytokine genes, associated with an increased immune response, are more prevalent in patients with aplastic anemia. These cytokines suppress hematopoiesis by affecting the mitotic cycle and cell killing by inducing Fas-mediated apoptosis. In addition, these cytokines induce nitric oxide synthase and nitric oxide production by marrow cells, which contributes to immune-mediated cytotoxicity and the elimination of hematopoietic cells. Constitutive expression of Tbet, a transcriptional regulator that is critical to Th1 polarization, occurs in a majority of aplastic anemia patients.[7] Perforin is a cytolytic protein expressed mainly in activated cytotoxic lymphocytes and natural-killer cells. Mutations in the perforin gene are responsible for some cases of familial hemophagocytosis [13] ; mutations in SAP, a gene encoding for a small modulator protein that inhibits undefined-interferon production, underlie X-linked lymphoproliferation, a fatal illness associated with an aberrant immune response to herpesviruses and aplastic anemia. Perforin and SAP protein levels are markedly diminished in a majority of acquired aplastic anemia cases. Apart from immunological, toxin/drug-related, and infectious etiopathologies, around 10-15% of patients with apparently acquired aplastic anemia may have shortened telomeres in blood lymphocytes. This was initially presumed to reflect stressed hematopoiesis, but, subsequently, telomerase gene complex mutations have been demonstrated in such individuals as well as their healthy family members. These apparently healthy family members were subsequently tested and found to have normal or near-normal blood counts, along with hypocellular marrow fragments.[14]

Congenital or inherited causes


Congenital or inherited causes of aplastic anemia (20%) include the following: Patients usually have dysmorphic features or physical stigmata; on occasion, marrow failure may be the initial presenting feature. Fanconi anemia Dyskeratosis congenita Cartilage-hair hypoplasia Pearson syndrome Amegakaryocytic thrombocytopenia (thrombocytopenia-absent radius [TAR] syndrome)

2 of 7

12/26/2011 9:09 AM

Aplastic Anemia

http://emedicine.medscape.com/article/198759-overview

Shwachman-Diamond syndrome Dubowitz syndrome Diamond-Blackfan syndrome Familial aplastic anemia

Acquired causes
Acquired causes of aplastic anemia (80%) include the following: Idiopathic factors Infectious causes, such as hepatitis viruses, Epstein-Barr virus (EBV), human immunodeficiency virus (HIV), parvovirus, and mycobacteria Toxic exposure to radiation and chemicals, such as benzene Transfusional GVHD Orthotopic liver transplantation for fulminant hepatitis Pregnancy Eosinophilic fasciitis Drugs and elements, such as chloramphenicol, phenylbutazone, and gold, may cause aplasia of the marrow. The immune mechanism does not account for the marrow failure in idiosyncratic drug reactions. In such cases, direct toxicity may occur, perhaps due to genetically determined differences in metabolic detoxification pathways. For example, the null phenotype of certain glutathione transferases is overrepresented among patients with aplastic anemia. Paroxysmal nocturnal hemoglobinuria (PNH) is caused by an acquired genetic defect limited to the stem-cell compartment affecting the PIGA gene. Mutations in the PIGA gene render cells of hematopoietic origin sensitive to increased complement lysis. Approximately 20% of patients with aplastic anemia have evidence of PNH at presentation, as detected by means of flow cytometry. Furthermore, patients whose disease responds after immunosuppressive therapy frequently recover with clonal hematopiesis and PNH.

Epidemiology
Occurrence in the United States
No accurate prospective data are available regarding the incidence of aplastic anemia in the United States. Findings from several retrospective studies suggest that the incidence is 0.6-6.1 cases per million population; this rate was largely based on data from retrospective reviews of death registries.

International occurrence
The annual incidence of aplastic anemia in Europe, as detailed in large, formal epidemiologic studies, is 2 cases per million population. Aplastic anemia is thought to be more common in Asia than in the West. The incidence was accurately determined to be 4 cases per million population in Bangkok, but it may be closer to 6 cases per million population in the rural areas of Thailand and as high as 14 cases per million population in Japan, based on prospective studies. This increased incidence may be related to environmental factors, such as increased exposure to toxic chemicals, rather than to genetic factors, because this increase is not observed in people of Asian ancestry who are living in the United States.

Race-, sex-, and age-related demographics


No racial predisposition is reported in the United States. However, the prevalence is increased in the Far East. The male-to-female ratio for acquired aplastic anemia is approximately 1:1, although there are data to suggest that a male preponderance may be observed in the Far East. Aplastic anemia occurs in all age groups. A small peak in the incidence is observed in childhood because of the inclusion of inherited marrow-failure syndromes. The incidence of aplastic anemia peaks in people aged 20-25 years, and a subsequent peak is observed in people older than age 60 years. The latter peak may be due to the inclusion of myelodysplastic syndromes (MDSs), which are syndromes of stem-cell failure unrelated to aplastic anemia. These syndromes must be considered in the differential diagnosis of any marrow-failure syndrome.

3 of 7

12/26/2011 9:09 AM

Aplastic Anemia

http://emedicine.medscape.com/article/198759-overview

Prognosis
The outcome of patients with aplastic anemia has substantially improved because of improved supportive care. The natural history of aplastic anemia suggests that as many as one fifth of patients may spontaneously recover with supportive care; however, observational and/or supportive care therapy alone is rarely indicated. The estimated 5-year survival rate for the typical patient receiving immunosuppression is 75%. The rate for those receiving a BMT from a matched sibling donor is greater than 90%. However, in cases of immunosuppression, relapse and late clonal disease are risks. In a single institution analysis of 183 patients who received immunosuppressive treatments for severe aplastic anemia, the telomere length of peripheral blood leukocytes was unrelated to treatment response. In a multivariate analysis, however, telomere length was associated with risk of relapse, clonal evolution, and overall survival. Additional studies are needed to validate these findings and to determine how this information might be incorporated into treatment algorithms.[15]

Mortality/morbidity
The major causes of morbidity and mortality from aplastic anemia include infection and bleeding. Patients who undergo BMT have additional issues related to toxicity from the conditioning regimen and GVHD.[12, 16, 17, 18, 19, 20] With immunosuppression, aplastic anemia in approximately one third of patients does not respond. For the responders, relapse and late-onset clonal disease, such as PNH, MDS, and leukemia, are risks.[8, 21, 22, 23, 24]

Contributor Information and Disclosures


Author Sameer Bakhshi, MD Additional Professor of Pediatric Oncology, Department of Medical Oncology, Dr BRA Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, India Disclosure: Nothing to disclose. Specialty Editor Board David Aboulafia, MD Medical Director, Bailey-Boushay House, Clinical Professor, Department of Medicine, Division of Hematology, Attending Physician, Section of Hematology/Oncology, Virginia Mason Clinic; Investigator, Virginia Mason Community Clinic Oncology Program/SWOG David Aboulafia, MD is a member of the following medical societies: American College of Physicians, American Medical Association, American Medical Directors Association, American Society of Hematology, Infectious Diseases Society of America, and Phi Beta Kappa Disclosure: Nothing to disclose. Francisco Talavera, PharmD, PhD Adjunct Assistant Professor, University of Nebraska Medical Center College of Pharmacy; Editor-in-Chief, Medscape Drug Reference Disclosure: Medscape Salary Employment Chief Editor Emmanuel C Besa, MD Professor, Department of Medicine, Division of Hematologic Malignancies, Kimmel Cancer Center, Jefferson Medical College of Thomas Jefferson University Emmanuel C Besa, MD is a member of the following medical societies: American Association for Cancer Education, American College of Clinical Pharmacology, American Federation for Medical Research, American Society of Clinical Oncology, American Society of Hematology, and New York Academy of Sciences Disclosure: Nothing to disclose. Additional Contributors The authors and editors of Medscape Reference gratefully acknowledge the contributions of previous authors RoyBaynes, MB, BCh, PhD, FACP, andEsteban Abella, MD,to the development and writing of the source article.

References
4 of 7

12/26/2011 9:09 AM

Aplastic Anemia

http://emedicine.medscape.com/article/198759-overview

1. Wu Y, Yu J, Zhang L, Luo Q, Xiao JW, Liu XM, et al. [Hematopoiesis support of mesenchymal stem cells in children with aplastic anemia]. Zhongguo Dang Dai Er Ke Za Zhi. Aug 2008;10(4):455-9. [Medline]. 2. Scopes J, Daly S, Atkinson R, Ball SE, Gordon-Smith EC, Gibson FM. Aplastic anemia: evidence for dysfunctional bone marrow progenitor cells and the corrective effect of granulocyte colony-stimulating factor in vitro. Blood. Apr 15 1996;87(8):3179-85. [Medline]. 3. Young NS. Pathophysiologic mechanisms in acquired aplastic anemia. Hematology Am Soc Hematol Educ Program. 2006;72-7. [Medline]. 4. Liu H, Mihara K, Kimura A, Tanaka K, Kamada N. Induction of apoptosis in CD34+ cells by sera from patients with aplastic anemia. Hiroshima J Med Sci. Jun 1999;48(2):57-63. [Medline]. 5. Marsh JC. Long-term bone marrow cultures in aplastic anaemia. Eur J Haematol Suppl. 1996;60:75-9. [Medline]. 6. Marsh JC, Chang J, Testa NG, Hows JM, Dexter TM. The hematopoietic defect in aplastic anemia assessed by long-term marrow culture. Blood. Nov 1 1990;76(9):1748-57. [Medline]. 7. Solomou EE, Keyvanfar K, Young NS. T-bet, a Th1 transcription factor, is up-regulated in T cells from patients with aplastic anemia. Blood. May 15 2006;107(10):3983-91. [Medline]. [Full Text]. 8. Soci G, Rosenfeld S, Frickhofen N, Gluckman E, Tichelli A. Late clonal diseases of treated aplastic anemia. Semin Hematol. Jan 2000;37(1):91-101. [Medline]. 9. Nakao S. Immune mechanism of aplastic anemia. Int J Hematol. Aug 1997;66(2):127-34. [Medline]. 10. Frickhofen N, Kaltwasser JP, Schrezenmeier H, Raghavachar A, Vogt HG, Herrmann F, et al. Treatment of aplastic anemia with antilymphocyte globulin and methylprednisolone with or without cyclosporine. The German Aplastic Anemia Study Group. N Engl J Med. May 9 1991;324(19):1297-304. [Medline]. 11. Passweg JR, Prez WS, Eapen M, Camitta BM, Gluckman E, Hinterberger W, et al. Bone marrow transplants from mismatched related and unrelated donors for severe aplastic anemia. Bone Marrow Transplant. Apr 2006;37(7):641-9. [Medline]. 12. Bacigalupo A, Brand R, Oneto R, Bruno B, Soci G, Passweg J, et al. Treatment of acquired severe aplastic anemia: bone marrow transplantation compared with immunosuppressive therapy--The European Group for Blood and Marrow Transplantation experience. Semin Hematol. Jan 2000;37(1):69-80. [Medline]. 13. Solomou EE, Gibellini F, Stewart B, Malide D, Berg M, Visconte V, et al. Perforin gene mutations in patients with acquired aplastic anemia. Blood. Jun 15 2007;109(12):5234-7. [Medline]. [Full Text]. 14. Yamaguchi H, Calado RT, Ly H, et al. Mutations in TERT, the gene for telomerase reverse transcriptase, in aplastic anemia. N Engl J Med. Apr 7 2005;352(14):1413-24. [Medline]. 15. Scheinberg P, Cooper JN, Sloand EM, Wu CO, Calado RT, Young NS. Association of telomere length of peripheral blood leukocytes with hematopoietic relapse, malignant transformation, and survival in severe aplastic anemia. JAMA. Sep 22 2010;304(12):1358-64. [Medline]. 16. Chan KW, McDonald L, Lim D, Grimley MS, Grayson G, Wall DA. Unrelated cord blood transplantation in children with idiopathic severe aplastic anemia. Bone Marrow Transplant. Nov 2008;42(9):589-95. [Medline]. 17. Bunin N, Aplenc R, Iannone R, Leahey A, Grupp S, Monos D, et al. Unrelated donor bone marrow transplantation for children with severe aplastic anemia: minimal GVHD and durable engraftment with partial T cell depletion. Bone Marrow Transplant. Feb 2005;35(4):369-73. [Medline]. 18. Kang HJ, Shin HY, Choi HS, Ahn HS. Fludarabine, cyclophosphamide plus thymoglobulin conditioning regimen for unrelated bone marrow transplantation in severe aplastic anemia. Bone Marrow Transplant. Dec 2004;34(11):939-43. [Medline]. 19. Brodsky RA, Sensenbrenner LL, Smith BD, Dorr D, Seaman PJ, Lee SM, et al. Durable treatment-free remission after high-dose cyclophosphamide therapy for previously untreated severe aplastic anemia. Ann Intern Med. Oct 2 2001;135(7):477-83. [Medline].

5 of 7

12/26/2011 9:09 AM

Aplastic Anemia

http://emedicine.medscape.com/article/198759-overview

20. Horowitz MM. Current status of allogeneic bone marrow transplantation in acquired aplastic anemia. Semin Hematol. Jan 2000;37(1):30-42. [Medline]. 21. Kaito K, Kobayashi M, Katayama T, Masuoka H, Shimada T, Nishiwaki K, et al. Long-term administration of G-CSF for aplastic anaemia is closely related to the early evolution of monosomy 7 MDS in adults. Br J Haematol. Nov 1998;103(2):297-303. [Medline]. 22. Piaggio G, Podest M, Pitto A, Sessarego M, Figari O, Fugazza G, et al. Coexistence of normal and clonal haemopoiesis in aplastic anaemia patients treated with immunosuppressive therapy. Br J Haematol. Dec 1999;107(3):505-11. [Medline]. 23. Rosti V. The molecular basis of paroxysmal nocturnal hemoglobinuria. Haematologica. Jan 2000;85(1):82-7. [Medline]. 24. Orazi A, Czader MB. Myelodysplastic syndromes. Am J Clin Pathol. Aug 2009;132(2):290-305. [Medline]. 25. Breakey VR, Meyn S, Ng V, Allen C, Dokal I, Lansdorp PM, et al. Hepatitis-associated aplastic anemia presenting as a familial bone marrow failure syndrome. J Pediatr Hematol Oncol. Nov 2009;31(11):884-7. [Medline]. 26. Gonzalez-Casas R, Garcia-Buey L, Jones EA, Gisbert JP, Moreno-Otero R. Systematic review: hepatitisassociated aplastic anaemia--a syndrome associated with abnormal immunological function. Aliment Pharmacol Ther. Sep 1 2009;30(5):436-43. [Medline]. 27. Valdez JM, Scheinberg P, Young NS, Walsh TJ. Infections in patients with aplastic anemia. Semin Hematol. Jul 2009;46(3):269-76. [Medline]. 28. Jancel T, Penzak SR. Antiviral therapy in patients with hematologic malignancies, transplantation, and aplastic anemia. Semin Hematol. Jul 2009;46(3):230-47. [Medline]. 29. Di Bona E, Rodeghiero F, Bruno B, Gabbas A, Foa P, Locasciulli A, et al. Rabbit antithymocyte globulin (r-ATG) plus cyclosporine and granulocyte colony stimulating factor is an effective treatment for aplastic anaemia patients unresponsive to a first course of intensive immunosuppressive therapy. Gruppo Italiano Trapianto di Midollo Osseo (GITMO). Br J Haematol. Nov 1999;107(2):330-4. [Medline]. 30. Bacigalupo A, Broccia G, Corda G, Arcese W, Carotenuto M, Gallamini A, et al. Antilymphocyte globulin, cyclosporin, and granulocyte colony-stimulating factor in patients with acquired severe aplastic anemia (SAA): a pilot study of the EBMT SAA Working Party. Blood. Mar 1 1995;85(5):1348-53. [Medline]. 31. Dufour C, Ferretti E, Bagnasco F, Burlando O, Lanciotti M, Ramenghi U, et al. Changes in cytokine profile pre- and post-immunosuppression in acquired aplastic anemia. Haematologica. Dec 2009;94(12):1743-7. [Medline]. [Full Text]. 32. Storb R, Longton G, Anasetti C, et al. Changing trends in marrow transplantation for aplastic anemia. Bone Marrow Transplant. 1992;10 Suppl 1:45-52. [Medline]. 33. McCann SR, Bacigalupo A, Gluckman E, et al. Graft rejection and second bone marrow transplants for acquired aplastic anaemia: a report from the Aplastic Anaemia Working Party of the European Bone Marrow Transplant Group. Bone Marrow Transplant. Mar 1994;13(3):233-7. [Medline]. 34. Storb R, Deeg HJ. Failure of allogeneic canine marrow grafts after total-body irradiation. Allogeneic "resistance" versus transfusion-induced sensitization. Transplantation. Dec 1986;42(6):571-80. [Medline]. 35. Storb R, Thomas ED, Buckner CD, et al. Marrow transplantation in thirty "untransfused" patients with severe aplastic anemia. Ann Intern Med. Jan 1980;92(1):30-6. [Medline]. 36. Schuening F, Bean MA, Deeg HJ, Storb R. Prevention of graft failure in patients with aplastic anemia. Bone Marrow Transplant. 1993;12 Suppl 3:S48-9. [Medline]. 37. Locatelli F, Bruno B, Zecca M, Van-Lint MT, McCann S, Arcese W, et al. Cyclosporin A and short-term methotrexate versus cyclosporin A as graft versus host disease prophylaxis in patients with severe aplastic anemia given allogeneic bone marrow transplantation from an HLA-identical sibling: results of a GITMO/EBMT randomized trial. Blood. Sep 1 2000;96(5):1690-7. [Medline]. 38. George B, Mathews V, Viswabandya A, Kavitha ML, Srivastava A, Chandy M. Fludarabine and

6 of 7

12/26/2011 9:09 AM

Aplastic Anemia

http://emedicine.medscape.com/article/198759-overview

cyclophosphamide based reduced intensity conditioning (RIC) regimens reduce rejection and improve outcome in Indian patients undergoing allogeneic stem cell transplantation for severe aplastic anemia. Bone Marrow Transplant. Jul 2007;40(1):13-8. [Medline]. 39. Yagasaki H, Takahashi Y, Hama A, et al. Comparison of matched-sibling donor BMT and unrelated donor BMT in children and adolescent with acquired severe aplastic anemia. Bone Marrow Transplant. Oct 2010;45(10):1508-13. [Medline]. 40. Maury S, Balre-Appert ML, Chir Z, Boiron JM, Galambrun C, Yakouben K, et al. Unrelated stem cell transplantation for severe acquired aplastic anemia: improved outcome in the era of high-resolution HLA matching between donor and recipient. Haematologica. May 2007;92(5):589-96. [Medline]. 41. Kojima S, Matsuyama T, Kato S, Kigasawa H, Kobayashi R, Kikuta A, et al. Outcome of 154 patients with severe aplastic anemia who received transplants from unrelated donors: the Japan Marrow Donor Program. Blood. Aug 1 2002;100(3):799-803. [Medline]. 42. MacMillan ML, Walters MC, Gluckman E. Transplant outcomes in bone marrow failure syndromes and hemoglobinopathies. Semin Hematol. Jan 2010;47(1):37-45. [Medline]. 43. Chan KW, McDonald L, Lim D, Grimley MS, Grayson G, Wall DA. Unrelated cord blood transplantation in children with idiopathic severe aplastic anemia. Bone Marrow Transplant. Nov 2008;42(9):589-95. [Medline]. 44. Yoshimi A, Kojima S, Taniguchi S, et al. Unrelated cord blood transplantation for severe aplastic anemia. Biol Blood Marrow Transplant. Sep 2008;14(9):1057-63. [Medline]. 45. Scheinberg P, Nunez O, Weinstein B, et al. Horse versus rabbit antithymocyte globulin in acquired aplastic anemia. N Engl J Med. Aug 4 2011;365(5):430-8. [Medline]. 46. Maschan A, Bogatcheva N, Kryjanovskii O, Shneider M, Litvinov D, Mitiushkina T, et al. Results at a single centre of immunosuppression with cyclosporine A in 66 children with aplastic anaemia. Br J Haematol. Sep 1999;106(4):967-70. [Medline]. 47. Brodsky RA, Chen AR, Dorr D, et al. High-dose cyclophosphamide for severe aplastic anemia: long-term follow-up. Blood. Mar 18 2010;115(11):2136-41. [Medline]. [Full Text]. 48. Tisdale JF, Dunn DE, Geller N, et al. High-dose cyclophosphamide in severe aplastic anaemia: a randomised trial. Lancet. Nov 4 2000;356(9241):1554-9. [Medline]. Medscape Reference 2011 WebMD, LLC

7 of 7

12/26/2011 9:09 AM

You might also like