Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-nmvwc Total loading time: 0 Render date: 2024-06-20T21:38:20.818Z Has data issue: false hasContentIssue false

Chapter 18 - Bone Marrow Changes Following Therapy and Immunosuppression

Published online by Cambridge University Press:  12 November 2020

Jon van der Walt
Affiliation:
St Thomas’ Hospital, London
Attilio Orazi
Affiliation:
Texas Tech University
Daniel A. Arber
Affiliation:
University of Chicago
Get access

Summary

Bone marrow (BM) trephine biopsy (BMB) is a frequent and routine diagnostic investigation, as nicely described in the previous chapters, and is also widely used for follow-up of haematological disorders to judge the effectiveness of therapeutic interventions [1]. In addition, several drugs applied primarily for the treatment of non-haematological disorders may cause serious haematological side effects, such as pancytopaenia, agranulocytosis or anaemia. On occasion, the causative link between the haematological symptoms and the previous drug exposure is not perceived and it is not uncommon for BMB to be obtained in such circumstances without any information of a history of previous drug exposure being made available to the haematopathologist [2].

Type
Chapter
Information
Publisher: Cambridge University Press
Print publication year: 2021

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Dirnhofer, S, Went, P, Tichelli, A. Diagnostic problems in follow-up bone marrow biopsies of patients treated for acute and chronic leukaemias and MDS. Pathobiology. 2007;74:115–20.CrossRefGoogle ScholarPubMed
Hoeller, S, Tzankov, A. Consideration of exposure to pharmacological agents helps to avoid diagnostic pitfalls in bone marrow histopathology. Magazine of European Medical Oncology. 2010;3:132–5.Google Scholar
Foucar, K, Reichard, K, Czuchlewski, D. Bone Marrow Pathology. 3rd edn. Chicago: ASCP Press; 2010.Google Scholar
Ho, MS, Medcalf, RL, Livesey, SA, Traianedes, K. The dynamics of adult haematopoiesis in the bone and bone marrow environment. Br J Haematol. 2015;170:472–86.Google Scholar
Johnston, A, Uetrecht, J. Current understanding of the mechanisms of idiosyncratic drug-induced agranulocytosis. Expert Opin Drug Metab Toxicol. 2015;11:243–57.Google Scholar
Riether, C, Schürch, CM, Ochsenbein, AF. Regulation of hematopoietic and leukemic stem cells by the immune system. Cell Death Differ. 2015;22:187–98.Google Scholar
Salvador, C, Steurer, M, Tzankov, A, et al. Naturally occuring CD4+CD25+ regulatory T-cells modulate the hematopoietic potential of human stem cells in vitro. Br J Haematol. 2008;142:845–7.Google Scholar
Goldstein, M, Kastan, MB. The DNA damage response: implications for tumor responses to radiation and chemotherapy. Annu Rev Med. 2015;66:129–43.CrossRefGoogle ScholarPubMed
Percival, ME, Lai, C, Estey, E, Hourigan, CS. Bone marrow evaluation for diagnosis and monitoring of acute myeloid leukemia. Blood Rev. 2017;31:185–92.Google Scholar
Hurwitz, N. Bone marrow trephine biopsy changes following chemotherapy and/or bone marrow transplantation. Curr Diagn Pathol. 1997;196–202.CrossRefGoogle Scholar
Khan, R, Schmidt-Mende, J, Karimi, M, et al. Hypomethylation and apoptosis in 5-azacytidine-treated myeloid cells. Exp Hematol. 2008;36:149–57.Google Scholar
Ogata, K, Della Porta, MG, Malcovati, L, et al. Diagnostic utility of flow cytometry in low-grade myelodysplastic syndromes: a prospective validation study. Haematologica. 2009;94:1066–74.Google Scholar
Bartels, S, Schipper, E, Hasemeier, B, Kreipe, H, Lehmann, U. Routine clinical mutation profiling using next generation sequencing and a customized gene panel improves diagnostic precision in myeloid neoplasms. Oncotarget. 2016;7:30,084–93.Google Scholar
Vardiman, JW, Arber, DA, Brunning, RD, et al. Therapy-related myeloid neoplasms. In Swerdlow, SH, Campo, E, Harris, NL, et al. (eds.). WHO Classification of Tumours of the Haematopoietic and Lymphoid Tissues. Lyon: IARC Press; 2017.Google Scholar
McNerney, ME, Godley, LA, Le Beau, MM. Therapy-related myeloid neoplasms: when genetics and environment collide. Nat Rev Cancer. 2017;17:513–27.Google Scholar
Klco, JM, Miller, CA, Griffith, M, et al. Association between mutation clearance after induction therapy and outcomes in acute myeloid leukemia. JAMA. 2015;314:811–22.CrossRefGoogle ScholarPubMed
Jongen-Lavrencic, M, Grob, T, Hanekamp, D, et al. Molecular minimal residual disease in acute myeloid leukemia. N Engl J Med. 2018;378:1189–99.CrossRefGoogle ScholarPubMed
Ivey, A, Hills, RK, Simpson, MA, et al. Assessment of minimal residual disease in standard-risk AML. N Engl J Med. 2016;374:422–33.Google Scholar
Cheson, BD, Bennett, JM, Kopecky, KJ, et al. Revised recommendations of the International Working Group for Diagnosis, Standardization of Response Criteria, Treatment Outcomes, and Reporting Standards for Therapeutic Trials in Acute Myeloid Leukemia. J Clin Oncol. 2003;21:4642–9.Google Scholar
Bloomfield, CD, Estey, E, Pleyer, L, et al. Time to repeal and replace response criteria for acute myeloid leukemia? Blood Rev. 2018;32:416–25.Google Scholar
Dohner, H, Estey, E, Grimwade, D, et al. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood. 2017;129:424–47.CrossRefGoogle ScholarPubMed
Sweet, K, Lancet, J. State of the art update and next questions: acute myeloid leukemia. Clin Lymphoma Myeloma Leuk. 2017;17:703–9.Google Scholar
Kato, M, Manabe, A. Treatment and biology of pediatric acute lymphoblastic leukemia. Pediatr Int. 2018;60:412.CrossRefGoogle ScholarPubMed
Lauten, M, Möricke, A, Beier, R, et al. Prediction of outcome by early bone marrow response in childhood acute lymphoblastic leukemia treated in the ALL-BFM 95 trial: differential effects in precursor B-cell and T-cell leukemia. Haematologica. 2012;97:1048–56.Google Scholar
Chantepie, SP, Cornet, E, Salaün, V, Reman, O. Hematogones: an overview. Leuk Res. 2013;37:1404–11.CrossRefGoogle ScholarPubMed
Rimsza, LM, Larson, RS, Winter, SS, et al. Benign hematogone-rich lymphoid proliferations can be distinguished from B-lineage acute lymphoblastic leukemia by integration of morphology, immunophenotype, adhesion molecule expression, and architectural features. Am J Clin Pathol. 2000;114:6675.CrossRefGoogle ScholarPubMed
Chopade, P, Akard, LP. Improving outcomes in chronic myeloid leukemia over time in the era of tyrosine kinase inhibitors. Clin Lymphoma Myeloma Leuk. 2018;18:710–23.CrossRefGoogle ScholarPubMed
Mahon, FX. Discontinuation of TKI therapy and ‘functional’ cure for CML. Best Pract Res Clin Haematol. 2016;29:308–13.CrossRefGoogle ScholarPubMed
Baccarani, M, Saglio, G, Goldman, J, et al. Evolving concepts in the management of chronic myeloid leukemia: recommendations from an expert panel on behalf of the European LeukemiaNet. Blood. 2006;108:1809–20.Google Scholar
Lugli, A, Ebnoether, M, Cogliatti, SB, et al. Proposal of a morphologic bone marrow response score for imatinib mesylate treatment in chronic myelogenous leukemia. Hum Pathol. 2005;36:91100.CrossRefGoogle ScholarPubMed
Tefferi, A. Myeloproliferative neoplasms: a decade of discoveries and treatment advances. Am J Hematol. 2016;91:50–8.CrossRefGoogle ScholarPubMed
Tefferi, A, Barosi, G, Mesa, RA, et al. International Working Group (IWG) consensus criteria for treatment response in myelofibrosis with myeloid metaplasia, for the IWG for Myelofibrosis Research and Treatment (IWG-MRT). Blood. 2006;108:1497–503.Google Scholar
Jamieson, C, Hasserjian, R, Gotlib, J, et al. Effect of treatment with a JAK2-selective inhibitor, fedratinib, on bone marrow fibrosis in patients with myelofibrosis. J Transl Med. 2015;13:294.Google Scholar
Wilkins, BS, Radia, D, Woodley, C, et al. Resolution of bone marrow fibrosis in a patient receiving JAK1/JAK2 inhibitor treatment with ruxolitinib. Haematologica. 2013;98:1872–6.CrossRefGoogle Scholar
Thiele, J, Kvasnicka, HM, Facchetti, F, et al. European consensus on grading bone marrow fibrosis and assessment of cellularity. Haematologica. 2005;90:1128–32.Google Scholar
Kvasnicka, HM, Beham-Schmid, C, Bob, R, et al. Problems and pitfalls in grading of bone marrow fibrosis, collagen deposition and osteosclerosis: a consensus-based study. Histopathology. 2016;68:905–15.Google Scholar
Horn, T, Kremer, M, Dechow, T, et al. Detection of the activating JAK2 V617F mutation in paraffin-embedded trephine bone marrow biopsies of patients with chronic myeloproliferative diseases. J Mol Diagn. 2006;8:299304.CrossRefGoogle ScholarPubMed
Stein, H, Bob, R, Dürkop, H, et al. A new monoclonal antibody (CAL2) detects CALRETICULIN mutations in formalin-fixed and paraffin-embedded bone marrow biopsies. Leukemia. 2016;30:131–5.CrossRefGoogle ScholarPubMed
Aboudola, S, Murugesan, G, Szpurka, H, et al. Bone marrow phospho-STAT5 expression in non-CML chronic myeloproliferative disorders correlates with JAK2 V617F mutation and provides evidence of in vivo JAK2 activation. Am J Surg Pathol. 2007;31:233–9.CrossRefGoogle ScholarPubMed
Gibson, SE, Schade, AE, Szpurka, H, et al. Phospho-STAT5 expression pattern with the MPL W515L mutation is similar to that seen in chronic myeloproliferative disorders with JAK2 V617F. Hum Pathol. 2008;39:1111–14.Google Scholar
Santini, V. Society of Hematologic Oncology (SOHO) state of the art updates and next questions: myelodysplastic syndromes. Clin Lymphoma Myeloma Leuk. 2018;18:495500.Google Scholar
Valent, P, Horny, HP, Bennett, JM, et al. Definitions and standards in the diagnosis and treatment of the myelodysplastic syndromes: consensus statements and report from a working conference. Leuk Res. 2007;31:727–36.CrossRefGoogle ScholarPubMed
Cheson, BD, Greenberg, PL, Bennett, JM, et al. Clinical application and proposal for modification of the International Working Group (IWG) response criteria in myelodysplasia. Blood. 2006;108:419–25.CrossRefGoogle Scholar
Onida, F, Barosi, G, Leone, G, et al. Management recommendations for chronic myelomonocytic leukemia: consensus statements from the SIE, SIES, GITMO groups. Haematologica. 2013;98:1344–52.Google Scholar
Pich, A, Godio, L, Davico Bonino, L. p53 protein expression in patients with myelodysplasia treated with allogeneic bone marrow transplantation. Mol Clin Oncol. 2017;6:876–80.Google Scholar
Saft, L, Karimi, M, Ghaderi, M, et al. p53 protein expression independently predicts outcome in patients with lower-risk myelodysplastic syndromes with del(5q). Haematologica. 2014;99:1041–9.CrossRefGoogle ScholarPubMed
Durie, BG, Harousseau, JL, Miguel, JS, et al. International uniform response criteria for multiple myeloma. Leukemia. 2006;20:2220–6.Google Scholar
Kumar, S, Paiva, B, Anderson, KC, et al. International Myeloma Working Group consensus criteria for response and minimal residual disease assessment in multiple myeloma. Lancet Oncol. 2016;17:e328e346.CrossRefGoogle Scholar
Hallek, M, Cheson, BD, Catovsky, D, et al. Guidelines for the diagnosis and treatment of chronic lymphocytic leukemia: a report from the International Workshop on Chronic Lymphocytic Leukemia updating the National Cancer Institute-Working Group 1996 guidelines. Blood. 2008;111:5446–56.Google Scholar
Byrd, JC, Furman, RR, Coutre, SE, et al. Targeting BTK with ibrutinib in relapsed chronic lymphocytic leukemia. N Engl J Med. 2013;369:3242.Google Scholar
Sah, SP, Matutes, E, Wotherspoon, AC, Morilla, R, Catovsky, D. A comparison of flow cytometry, bone marrow biopsy, and bone marrow aspirates in the detection of lymphoid infiltration in B cell disorders. J Clin Pathol. 2003;56:129–32.Google Scholar
Schmidt, B, Kremer, M, Gotze, K, et al. Bone marrow involvement in follicular lymphoma: comparison of histology and flow cytometry as staging procedures. Leuk Lymphoma. 2006;47:1857–62.CrossRefGoogle ScholarPubMed
Fend, F, Tzankov, A, Bink, K, et al. Modern techniques for the diagnostic evaluation of the trephine bone marrow biopsy: methodological aspects and applications. Prog Histochem Cytochem. 2008;42:203–52.Google Scholar
Harris, AC, Todd, WM, Hackney, MH, Ben-Ezra, J. Bone marrow changes associated with recombinant granulocyte-macrophage and granulocyte colony-stimulating factors. Discrimination of granulocytic regeneration. Arch Pathol Lab Med. 1994;118:624–9.Google Scholar
Liu, CZ, Persad, R, Inghirami, G, et al. Transient atypical monocytosis mimic acute myelomonocytic leukemia in post-chemotherapy patients receiving G-CSF: report of two cases. Clin Lab Haematol. 2004;26:359–62.Google Scholar
Glasser, L, Legolvan, M, Horwitz, HM. Florid histiocytic hemophagocytosis following therapy with long acting G-CSF (pegfilgrastim). Am J Hematol. 2007;82:753–7.Google Scholar
Osuorji, I, Goldman, L. G-CSF-associated bone marrow necrosis in AML after induction chemotherapy. Case Rep Hematol. 2012;2012:314278.Google Scholar
Dorn, C, Bugl, S, Malenke, E, et al. Paraneoplastic granulocyte colony-stimulating factor secretion in soft tissue sarcoma mimicking myeloproliferative neoplasia: a case report. BMC Res Notes. 2014;7:313.Google Scholar
Rosenberg, PS, Zeidler, C, Bolyard, AA, et al. Stable long-term risk of leukaemia in patients with severe congenital neutropenia maintained on G-CSF therapy. Br J Haematol. 2010;150:196–9.Google Scholar
Beekman, R, Valkhof, MG, Sanders, MA, et al. Sequential gain of mutations in severe congenital neutropenia progressing to acute myeloid leukemia. Blood. 2012;119:5071–7.Google Scholar
Martino, M, Fedele, R, Massara, E, et al. Long-term safety of granulocyte colony-stimulating factor in normal donors: is it all clear? Expert Opin Biol Ther. 2012;12:609–21.Google Scholar
Douglas, VK, Tallman, MS, Cripe, LD, Peterson, LC. Thrombopoietin administered during induction chemotherapy to patients with acute myeloid leukemia induces transient morphologic changes that may resemble chronic myeloproliferative disorders. Am J Clin Pathol. 2002;117:844–50.Google Scholar
Kuter, DJ, Mufti, GJ, Bain, BJ, et al. Evaluation of bone marrow reticulin formation in chronic immune thrombocytopenia patients treated with romiplostim. Blood. 2009;114:3748–56.CrossRefGoogle ScholarPubMed
Stone, RL, Nick, AM, McNeish, IA, et al. Paraneoplastic thrombocytosis in ovarian cancer. N Engl J Med. 2012;366:610–18.Google Scholar
Biljanović-Paunović, L, Djukanović, L, Lezaić, V, et al. In vivo effects of recombinant human erythropoietin on bone marrow hematopoiesis in patients with chronic renal failure. Eur J Med Res. 1998;3:564–70.Google Scholar
Rosado, FG, Oliveira, JL, Sohani, AR, et al. Bone marrow findings of the newly described TEMPI syndrome: when erythrocytosis and plasma cell dyscrasia coexist. Mod Pathol. 2015;28:367–72.Google Scholar
Vainchenker, W, Leroy, E, Gilles, L, et al. JAK inhibitors for the treatment of myeloproliferative neoplasms and other disorders. F1000Res. 2018;7:82.Google Scholar
Saeed, I, McLornan, D, Harrison, CN. Managing side effects of JAK inhibitors for myelofibrosis in clinical practice. Expert Rev Hematol. 2017;10:617–25.Google Scholar
Jonasch, E, Haluska, FG. Interferon in oncological practice: review of interferon biology, clinical applications, and toxicities. Oncologist. 2001;6:3455.CrossRefGoogle ScholarPubMed
Brunner, A, Kantner, J, Tzankov, A. Granulomatous reactions cause symptoms or clinically imitate treatment resistance in small lymphocytic lymphoma/chronic lymphocytic leukaemia more frequently than in other non-Hodgkin lymphomas. J Clin Pathol. 2005;58:815–19.Google Scholar
Syed, YY. Lenalidomide: a review in newly diagnosed multiple myeloma as maintenance therapy after ASCT. Drugs. 2017;77:1473–80.Google Scholar
Talati, C, Sallman, D, List, A. Lenalidomide: myelodysplastic syndromes with del(5q) and beyond. Semin Hematol. 2017;54:159–66.CrossRefGoogle ScholarPubMed
Dada, R. Lenalidomide maintenance after R-CHOP therapy in diffuse large B-cell lymphoma: can it be a standard of care? Acta Haematol. 2017;138:216–20.Google Scholar
Pierpont, TM, Limper, CB, Richards, KL. Past, present, and future of rituximab: the world’s first oncology monoclonal antibody therapy. Front Oncol. 2018;8:163.Google Scholar
Raynaud, P, Caulet-Maugendre, S, Foussard, C, et al. T-cell lymphoid aggregates in bone marrow after rituximab therapy for B-cell follicular lymphoma: a marker of therapeutic efficacy? Hum Pathol. 2008;39:194200.CrossRefGoogle ScholarPubMed
Jilani, I, O’Brien, S, Manshuri, T, et al. Transient down-modulation of CD20 by rituximab in patients with chronic lymphocytic leukemia. Blood. 2003;102:3514–20.Google Scholar
Maeshima, AM, Taniguchi, H, Fukuhara, S, et al. Follow-up data of 10 patients with B-cell non-Hodgkin lymphoma with a CD20-negative phenotypic change after rituximab-containing therapy. Am J Surg Pathol. 2013;37:563–70.CrossRefGoogle ScholarPubMed
Grant, C, Wilson, WH, Dunleavy, K. Neutropenia associated with rituximab therapy. Curr Opin Hematol. 2011;18:4954.Google Scholar
Tesfa, D, Ajeganova, S, Hägglund, H, et al. Late-onset neutropenia following rituximab therapy in rheumatic diseases: association with B lymphocyte depletion and infections. Arthritis Rheum. 2011;63:2209–14.Google Scholar
Kotsianidis, I, Goutzouvelidis, A, Anastasiades, A, et al. Severe thrombocytopenia and fibrinolysis mimicking disseminated intravascular coagulation after rituximab infusion. Am J Hematol. 2010;85:146.Google Scholar
Donato, EM, Fernández-Zarzoso, M, Hueso, JA, de la Rubia, J. Brentuximab vedotin in Hodgkin lymphoma and anaplastic large-cell lymphoma: an evidence-based review. Onco Targets Ther. 2018;11:4583–90.Google Scholar
Oak, E, Bartlett, NL. A safety evaluation of brentuximab vedotin for the treatment of Hodgkin lymphoma. Expert Opin Drug Saf. 2016;15:875–82.Google Scholar
Plesner, T, Krejcik, J. Daratumumab for the treatment of multiple myeloma. Front Immunol. 2018;9:1228.Google Scholar
Minarik, J, Novak, M, Flodr, P, et al. CD38-negative relapse in multiple myeloma after daratumumab-based chemotherapy. Eur J Haematol. 2017;99:186–9.Google Scholar
Riera, R, Porfírio, GJ, Torloni, MR. Alemtuzumab for multiple sclerosis. Cochrane Database Syst Rev. 2016;4:CD011203.Google Scholar
Laribi, K, Lemaire, P, Sandrini, J, Baugier de Materre, A. Advances in the understanding and management of T-cell prolymphocytic leukemia. Oncotarget. 2017;8:104,664–86.Google Scholar
Monteleone, F, Buccisano, F Boffa, L, et al. Reversible hyporegenerative anemia during natalizumab treatment. Mult Scler. 2015;21:257–8.Google Scholar
Simone, AM, Ferraro, D, Vitetta, F, et al. Severe anemia in a patient with multiple sclerosis treated with natalizumab. Neurology. 2014;83:374–5.Google Scholar
García-Sanz, R, Jiménez, C, González De La Calle, V, Sarasquete, ME. A safety profile of medications used to treat Waldenström’s macroglobulinemia. Expert Opin Drug Saf. 2018;17:609–21.Google Scholar
Blaker, PA, Arenas-Hernandez, M, Smith, MA, et al. Mechanism of allopurinol induced TPMT inhibition. Biochem Pharmacol. 2013;86:539–47.Google Scholar
Zaza, G, Cheok, M, Krynetskaia, N, et al. Thiopurine pathway. Pharmacogenet Genomics. 2010;20:573–4.Google Scholar
McGrath, BP, Ibels, LS, Raik, E, Hargrave, MJ. Erythroid toxicity of azathioprin. Macrocytosis and selective marrow hypoplasis. Q J Med. 1975;44:5763.Google Scholar
Katara, P, Kuntal, H. TPMT polymorphism: when shield becomes weakness. Interdiscip Sci. 2016;8:150–5.Google Scholar
Yunis, JJ, Corzo, D, Salazar, M, et al. HLA associations in clozapine-induced agranulocytosis. Blood. 1995;86:1177–83.Google Scholar
Turbay, D, Lieberman, J, Alper, CA, et al. Tumor necrosis factor constellation polymorphism and clozapine-induced agranulocytosis in two different ethnic groups. Blood. 1997;89:4167–74.CrossRefGoogle ScholarPubMed
Grann, VR, Bowman, N, Joseph, C, et al. Neutropenia in 6 ethnic groups from the Caribbean and the U.S. Cancer. 2008;113:854–60.Google Scholar
Mosyagin, I, Dettling, M, Roots, I, Mueller-Oerlinghausen, B, Cascorbi, I. Impact of myeloperoxidase and NADPH-oxidase polymorphisms in drug-induced agranulocytosis. J Clin Psychopharmacol. 2004;24:613–17.Google Scholar
van der Weide, K, Loovers, H, Pondman, K, et al. Genetic risk factors for clozapine-induced neutropenia and agranulocytosis in a Dutch psychiatric population. Pharmacogenomics J. 2017;17:471–8.Google Scholar
Tesfa, D, Keisu, M, Palmblad, J. Idiosyncratic drug-induced agranulocytosis: possible mechanisms and management. Am J Hematol. 2009;84:428–34.Google Scholar
Loeffler, S, Fehsel, K, Henning, U, et al. Increased apoptosis of neutrophils in a case of clozapine-induced agranulocytosis: a case report. Pharmacopsychiatry. 2003;36:3741.Google Scholar
Krech, R, Thiele, J. Histopathology of the bone marrow in toxic myelopathy. A study of drug induced lesions in 57 patients. Virchows Arch A Pathol Anat Histopathol. 1985;405:225–35.Google Scholar
Weinzierl, EP, Arber, DA. The differential diagnosis and bone marrow evaluation of new-onset pancytopenia. Am J Clin Pathol. 2013;139:929.Google Scholar
Young, NS, Kaufman, DW The epidemiology of acquired aplastic anemia. Haematologica. 2008;93:489–92.CrossRefGoogle ScholarPubMed
Bubp, J, Jen, M, Matuszewski, K. Caring for glucose-6-phosphate dehydrogenase (G6PD)-deficient patients: implications for pharmacy. PT. 2015;40:572–4.Google Scholar
Garratty, G, Arndt, PA. Drugs that have been shown to cause drug-induced immune hemolytic anemia or positive direct antiglobulin tests: some interesting findings since 2007. Immunohematology. 2014;30:6679.Google Scholar
Garratty, G. Immune hemolytic anemia caused by drugs. Expert Opin Drug Saf. 2012;11:635–42.Google Scholar
Garbe, E, Andersohn, F, Bronder, E, et al. Drug-induced immune thrombocytopenia: results from the Berlin Case-Control Surveillance Study. Eur J Clin Pharmacol. 2012;68:821–32.Google Scholar
Moulis, G, Sommet, A, Sailler, L, Lapeyre-Mestre, M, Montastruc, JL. French Association of Regional Pharmacovigilance Centers. Drug-induced immune thrombocytopenia: a descriptive survey in the French Pharmacovigilance database. Platelets. 2012;23:490–4.Google Scholar
Criado, PR, Criado, RF, Avancini, JM, Santi, CG. Drug reaction with Eosinophilia and Systemic Symptoms (DRESS)/Drug-induced Hypersensitivity Syndrome (DIHS): a review of current concepts. An Bras Dermatol. 2012;87:435–49.Google Scholar
Husain, Z, Reddy, BY, Schwartz, RA. DRESS syndrome: Part I. Clinical perspectives. J Am Acad Dermatol. 2013;68:693,e114.Google Scholar
Shalom, R, Rimbroth, S, Rozenman, D, Markel, A. Allopurinol-induced recurrent DRESS syndrome: pathophysiology and treatment. Ren Fail. 2008;30:327–9.Google Scholar
Kardaun, SH, Sekula, P, Valeyrie-Allanore, L, et al. Drug reaction with eosinophilia and systemic symptoms (DRESS): an original multisystem adverse drug reaction. Results from the prospective RegiSCAR study. Br J Dermatol. 2013;169:1071–80.Google Scholar
Katzung, BG. Basic and Clinical Pharmacology, 14th edn. New York: McGraw-Hill; 2018.Google Scholar
Toyokawa, Y, Kingetsu, I, Yasuda, C, et al. Pancytopenia, including macrocytic anemia, associated with leflunomide in a rheumatoid arthritis patient. Mod Rheumatol. 2007;17:436–40.CrossRefGoogle Scholar
McEwen, J, Purcell, PM, Hill, RL, Calcino, LJ, Riley, CG. The incidence of pancytopenia in patients taking leflunomide alone or with methotrexate. Pharmacoepidemiol Drug Saf. 2007;16:6573.Google Scholar
Weyrich, AS, Denis, MM, Schwertz, H, et al. mTOR-dependent synthesis of Bcl-3 controls the retraction of fibrin clots by activated human platelets. Blood. 2007;109:1975–83.Google Scholar
Kuitunen, T, Malmström, J, Palva, E, Pettersson, T. Pancytopenia induced by low-dose methotrexate. A study of the cases reported to the Finnish Adverse Drug Reaction Register from 1991 to 1999. Scand J Rheumatol. 2005;34:238–41.Google Scholar
Kolte, B, Baer, AN, Sait, SN, et al. Acute myeloid leukemia in the setting of low dose weekly methotrexate therapy for rheumatoid arthritis. Leuk Lymphoma. 2001;42:371–8.Google Scholar
Desai, SB, Furst, DE. Problems encountered during anti-tumour necrosis factor therapy. Best Pract Res Clin Rheumatol. 2006;20:757–90.Google Scholar
Bessissow, T, Renard, M, Hoffman, I, et al. Review article: non-malignant haematological complications of anti-tumour necrosis factor alpha therapy. Aliment Pharmacol Ther. 2012;36:312–23.CrossRefGoogle ScholarPubMed
Lamy, T, Moignet, A, Loughran, TP Jr. LGL leukemia: from pathogenesis to treatment. Blood. 2017;129:1082–94.Google Scholar
Vishnu, P, Aboulafia, DM. Haematological manifestations of human immune deficiency virus infection. Br J Haematol. 2015;171:695709.Google Scholar
Bozzi, A, Brisdelli, F, D’Alessandro, AM, et al. Effects of AZT on cellular iron homeostasis. Biometals. 2004;17:443–50.Google Scholar
Romanelli, F, Empey, K, Pomeroy, C. Macrocytosis as an indicator of medication (zidovudine) adherence in patients with HIV infection. AIDS Patient Care STDS. 2002;16:405–11.Google Scholar
Opie, J. Haematological complications of HIV infection. S Afr Med J. 2012;102:465–8.CrossRefGoogle ScholarPubMed
Linke-Serinsöz, E, Fend, F, Quintanilla-Martinez, L. Human immunodeficiency virus (HIV) and Epstein–Barr virus (EBV) related lymphomas, pathology view point. Semin Diagn Pathol. 2017;34:352–63.Google Scholar
Scaradavou, A. HIV-related thrombocytopenia. Blood Rev. 2002;16:73–6.Google Scholar
Jaffe, ES, Arber, DA, Campo, E, Harris, NL, Quintanilla-Martinez, L. Hematopathology, 2nd edn. Philadelphia: Elsevier; 2017.Google Scholar
Gyurkocza, B, Rezvani, A, Storb, RF. Allogeneic hematopoietic cell transplantation: the state of the art. Expert Rev Hematol. 2010;3:285–99.Google Scholar
Rousselet, MC, Kerjean, A, Guyétant, S, et al. Histopathology of bone marrow after allogeneic bone marrow transplantation for chronic myeloid leukaemia. Pathol Res Pract. 1996;192:790–5.Google Scholar
Rowley, SD, Donato, ML, Bhattacharyya, P. Red blood cell-incompatible allogeneic hematopoietic progenitor cell transplantation. Bone Marrow Transplant. 2011;46:1167–85.Google Scholar
Kraft, S, Bollinger, N, Bodenmann, B, et al. High mortality in hematopoietic stem cell transplant-associated thrombotic microangiopathy with and without concomitant acute graft-versus-host disease. Bone Marrow Transplant. 2018; doi: 10.1038/s41409-018-0293-3.Google Scholar
Gheysens, O, Thielemans, S, Morscio, J, et al. Detection of bone marrow involvement in newly diagnosed post-transplant lymphoproliferative disorder: (18)F-fluorodeoxyglucose positron emission tomography/computed tomography versus bone marrow biopsy. Leuk Lymphoma. 2016;57:2382–8.Google Scholar

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×