A JH CME Information: Systemic mastocytosis in adults: 2015 update on diagnosis, risk-stratification, and management Author: Animesh Pardanani M.B.B.S, Ph.D CME Editor: Ayalew Tefferi M.D.

If you wish to receive credit for this activity, please refer to the website: www.wileyhealthlearning.com

䊏 Accreditation and Designation Statement: Blackwell Futura Media Services is accredited by the Accreditation Council for Continuing Medical Education to provide continuing medical education for physicians. Blackwell Futura Media Services designates this journal-based CME for a maximum of 1 AMA PRA Category 1 CreditTM. Physicians should only claim credit commensurate with the extent of their participation in the activity.

䊏 Educational Objectives Upon completion of this educational activity, participants will be better able to: 1. Identify the current pathogenetic concepts, classification, diagnosis and risk stratification of systemic mastocytosis. 2. Identify the current management of systemic mastocytosis, including updates in investigational therapies.

䊏 Activity Disclosures No commercial support has been accepted related to the development or publication of this activity. CME Editor: Ayalew Tefferi, M.D., has no relevant financial relationships to disclose. Author: Animesh Pardanani, M.B.B.S, Ph.D, has no relevant financial relationships to disclose. This activity underwent peer review in line with the standards of editorial integrity and publication ethics maintained by American Journal of Hematology. The peer reviewers have no conflicts of interest to disclose. The peer review process for American Journal of Hematology is single blinded. As such, the identities of the reviewers are not disclosed in line with the standard accepted practices of medical journal peer review. Conflicts of interest have been identified and resolved in accordance with Blackwell Futura Media Services’s Policy on Activity Disclosure and Conflict of Interest. The primary resolution method used was peer review and review by a non-conflicted expert.

䊏 Instructions on Receiving Credit This activity is intended for physicians. For information on applicability and acceptance of continuing medical education credit for this activity, please consult your professional licensing board. This activity is designed to be completed within one hour; physicians should claim only those credits that reflect the time actually spent in the activity. To successfully earn credit, participants must complete the activity during the valid credit period, which is up to two years from initial publication. Additionally, up to 3 attempts and a score of 70% or better is needed to pass the post test. Follow these steps to earn credit:       

Log on to www.wileyhealthlearning.com Read the target audience, educational objectives, and activity disclosures. Read the activity contents in print or online format. Reflect on the activity contents. Access the CME Exam, and choose the best answer to each question. Complete the required evaluation component of the activity. Claim your Certificate.

This activity will be available for CME credit for twelve months following its launch date. At that time, it will be reviewed and potentially updated and extended for an additional twelve months.

C 2015 Wiley Periodicals, Inc. V

250

American Journal of Hematology, Vol. 90, No. 3, March 2015

doi:10.1002/ajh.00044

ANNUAL CLINICAL UPDATES IN HEMATOLOGICAL MALIGNANCIES

AJH Educational Material

A JH

Systemic mastocytosis in adults: 2015 update on diagnosis, risk stratification, and management Animesh Pardanani1,2* Disease overview: Systemic mastocytosis (SM) results from a clonal proliferation of abnormal mast cells (MC) in one or more extracutaneous organs. Diagnosis: The major criterion is presence of multifocal clusters of morphologically abnormal MC in the bone marrow. Minor diagnostic criteria include elevated serum tryptase level, abnormal MC expression of CD25 and/or CD2, and presence of KITD816V. Risk stratification: The 2008 World Health Organization classification of SM has been shown to be prognostically relevant. Classification of SM patients into indolent SM (ISM), aggressive SM (ASM), SM associated with a clonal non-MC lineage disease (SM-AHNMD), and mast cell leukemia (MCL) subgroups is a useful first step in establishing prognosis. Management: SM treatment is generally palliative. ISM patients have a normal life expectancy and receive symptom-directed therapy; infrequently, cytoreductive therapy may be indicated for refractory symptoms. ASM patients have disease-related organ dysfunction; interferon-a (1/2corticosteroids) can control dermatological, hematological, gastrointestinal, skeletal, and mediator-release symptoms, but is hampered by poor tolerability. Similarly, cladribine has broad therapeutic activity, with particular utility when rapid MC debulking is indicated; the main toxicity is myelosuppression. Imatinib has a therapeutic role in the presence of an imatinib-sensitive KIT mutation or in KITD816-unmutated patients. Treatment of SM-AHNMD is governed primarily by the non-MC neoplasm; hydroxyurea has modest utility in this setting; there is a role for allogeneic stem cell transplantation in select cases. Investigational Drugs: Recent data confirms midostaurin’s significant anti-MC activity in patients with advanced SM. C 2015 Wiley Periodicals, Inc. Am. J. Hematol. 90:251–262, 2015. V

䊏 Disease Overview and Pathogenesis Mastocytosis is one of eight subcategories of myeloproliferative neoplasms (MPN) per the 2008 World Health Organization (WHO) classification of tumors of hematopoietic and lymphoid tissues [1]. It results from a clonal, neoplastic proliferation of morphologically, and immunophenotypically abnormal mast cells (MC) that accumulate in one or more organ systems. The sine qua non of mastocytosis is the presence of multifocal clusters of abnormal MC, which in contrast to normal MC are variable in appearance, ranging from round to fusiform variants with long, polar cytoplasmic processes, and may display cytoplasmic hypogranularity with uneven distribution of fine granules, as well as atypical nuclei with monocytoid appearance [2–4]. The clinical presentation of mastocytosis is heterogeneous, ranging from skin-limited disease (cutaneous mastocytosis, CM), particularly in pediatric cases where the majority have disease-onset within the first 2 years of life and commonly experience spontaneous regression of skin lesions [5–8], to a more aggressive variant with extracutaneous involvement (systemic mastocytosis, SM) that may be associated with multiorgan dysfunction/failure and shortened survival, that is, generally seen in adult patients (Fig. 1) [9]. The WHO document distinguishes the usually KIT-mutated SM from a Philadelphia chromosome-negative MPN with hematological features of chronic eosinophilic leukemia associated with splenomegaly, marked elevation of serum vitamin B12, elevation of serum tryptase and increased bone marrow (BM) MC commonly in scattered or noncohesive clusters [10]. The latter entity is commonly associated with rearrangement of PDGFRA (i.e., FIP1L1-PDGFR) and less commonly, PDGFRB (e.g., PRKG2-PDGFRB), and is sensitive to treatment with imatinib [11–18]. WHOdefined SM is sometimes associated with a clonally related second myeloid neoplasm [19–22], which is not surprising considering its origin as a stem cell disease with multilineage clonal involvement [23–25]. Conversely, an otherwise well-defined myeloid malignancy, such as myelodysplastic syndrome (MDS) or a non-mast cell disease MPN, might also harbor neoplastic mast cells [26]. Mastocytosis is frequently associated with somatic gain-of-function point mutations within KIT. KIT (CD117) is a Type III receptor tyrosine kinase that is expressed by MC, hematopoietic progenitor cells, germ cells, melanocytes, and interstitial cells of Cajal in the gastrointestinal tract

Additional Supporting Information may be found in the online version of this article. 1

Division of Hematology, Mayo Clinic, Rochester, Minnesota; 2Department of Medicine, Mayo Clinic, Rochester, Minnesota

Conflict of interest: Nothing to report *Correspondence to: Animesh Pardanani, MBBS, PhD, Division of Hematology, Department of Medicine, Mayo Clinic, 200 First St. SW, Rochester, MN 55905, USA. E-mail: [email protected] Received for publication: 23 December 2014; Accepted: 24 December 2014 Am. J. Hematol. 90:251–262, 2015. Published online: in Wiley Online Library (wileyonlinelibrary.com). DOI: 10.1002/ajh.23931 C 2015 Wiley Periodicals, Inc. V

doi:10.1002/ajh.23931

American Journal of Hematology, Vol. 90, No. 3, March 2015

251

ANNUAL CLINICAL UPDATES IN HEMATOLOGICAL MALIGNANCIES

Pardanani

Figure 1. Clinical spectrum of patients with clonal mast cell disorders. Please refer to Tables I and II for the WHO classification of mastocytosis. “Prediagnostic” SM refers to an abnormal clonal BM mast cell infiltrate that falls short of the diagnostic threshold for SM (generally satisfies 1–2 minor criteria only). [Color figure can be viewed in the online issue, which is available at wileyonlinelibrary.com.]

Figure 2. Diagnostic algorithm for SM.

and is therefore functionally relevant for normal mast cell development, hematopoiesis, gametogenesis, melanogenesis, and regulation of slow gastric waves [27]. KIT expression is down regulated upon differentiation of hematopoietic progenitors into mature cells of all lineages, except mast cells, which retain high levels of cell surface KIT expression. The interaction between KIT and its ligand, stem cell factor, plays a key role in regulating mast cell proliferation, maturation, adhesion, chemotaxis, and survival [28]. Gain-of-function somatic mutations in the KIT tyrosine kinase domain, particularly the D816V mutation, have been found to occur 252

American Journal of Hematology, Vol. 90, No. 3, March 2015

in a majority of cases of adult SM, irrespective of WHO SM subtype [9,29]. Other less common (25% of the mast cells in the infiltrate are spindle-shaped or have atypical morphology or, of all mast cells in BM aspirate smears, >25% are immature or atypical b. Detection of an activating point mutation at codon 816 of KIT in BM, blood or other extracutaneous organ c. Mast cells in BM, blood or other extracutaneous organ express CD2 and/or CD25 in addition to normal mast cell markersa d. Serum total tryptase persistently exceeds 20 ng/mL (unless there is an associated clonal myeloid disorder, in which case this parameter is not valid) “B” findings 1. BM biopsy showing >30% infiltration by MC (focal, dense aggregates) and/or serum total tryptase level >200 ng/mL 2. Signs of dysplasia or myeloproliferation, in non-MC lineage(s), but insufficient criteria for definitive diagnosis of a hematopoietic neoplasm (AHNMD), with normal or slightly abnormal blood counts. 3. Hepatomegaly without impairment of liver function, and/or palpable splenomegaly without hypersplenism, and/or lymphadenopathy on palpation or imaging. “C” findings 1. BM dysfunction manifested by one or more cytopenia(s) (ANC MPN, AML > lymphoid neoplasms) [67]. Attempts at validating the WHO diagnostic criteria reveal that 20% of ISM patients lack mast cell clusters in the BM and 30% exhibit a serum tryptase level lower than 20 ng/mL [68]. In contrast, the sensitivity for detecting morphologic atypia, aberrant CD25 and/ or CD2 expression, or KITD816V in BM mast cells exceeds 90% when sensitive assays are used, thereby illustrating the increasing importance of these specific minor criteria in diagnosing SM [68,69]. Patients who have mast cell degranulation symptoms with clonal mast cells (i.e., mutated KIT gene and/or CD25 expression), but who do not meet criteria for SM (only 1 or 2 minor criteria satisfied, and no skin involvement), may have “prediagnostic ISM” or “monoclonal mast cell activation syndrome” (Fig. 3); these patients generally have normal or slightly elevated baseline serum tryptase level [70]. While the clinical characteristics of this entity may be indistinguishable from ISM, its true natural history remains to be defined. Rare SM cases may exhibit a well-differentiated phenotype (i.e., relatively normal BM MC morphology; absence of aberrant MC CD25/CD2 expression); these cases are associated with non-D816V KIT mutations (e.g., germline F522C or somatic I817V) and, in the case of KITF522C-associated SM, has been shown to be sensitive to imatinib therapy [29,45]. In the presence of blood eosinophilia, screening for FIP1L1PDGFRA, using either FISH or RT-PCR, is warranted [16]. In contrast, conventional cytogenetics analysis generally permits identification of cases of BMM associated with a PDGFRB rearrangement (i.e., chromosomal translocations involving 5q31-32) [18]. These cases with PDGFRA/PDGFRB-rearranged MPN with BM MC hyperplasia are appropriately classified as “Myeloid or lymphoid neoplasms with eosinophilia and abnormalities of PDGFRA, PDGFRB, or FGFR1” per the WHO classification [10].

䊏 Risk Stratification This section focuses on adult SM patients; their life expectancy, when considered as a group, appears to be shorter as compared to doi:10.1002/ajh.23931

age- and gender-matched controls, with the excess deaths in this group occurring within the first 3–5 years after diagnosis (Fig. 4A) [9,71]. The categorization of adult SM patients per the 2008 WHO classification system remains the most practical first step in risk stratifying newly diagnosed patients [1]. The WHO classification recognizes seven mastocytosis categories (Table I) SM is subclassified into four subcategories: ISM (no evidence of extracutaneous organ dysfunction), ASM (presence of extracutaneous organ dysfunction), SM associated with another clonal hematological non-MC lineage disease (SM-AHNMD), and MCL. A recent study of 342 adult patients has validated the prognostic value of the WHO classification for SM [9].

Indolent SM It comprised the largest subgroup (n 5 159; 46%) [9]. Compared with patients with ASM and SM-AHNMD, ISM patients were significantly younger at presentation (median age 49 years) and had a higher prevalence (66–75%) of UP-like skin lesions, MCMRS and gastrointestinal symptoms; ISM patients were significantly less likely however to exhibit constitutional symptoms or hepatosplenomegaly ( val in microdissected pooled single mast cells and leukemic cells in a patient with systemic mastocytosis and concomitant chronic myelomonocytic leukemia. Leuk Res 2002;26:979–984. 21. Travis WD, Li CY, Yam LT, et al. Significance of systemic mast cell disease with associated hematologic disorders. Cancer 1988;62:965–972.

doi:10.1002/ajh.23931

ANNUAL CLINICAL UPDATES IN HEMATOLOGICAL MALIGNANCIES 22. Pardanani A, Lim KH, Lasho TL, et al. Prognostically relevant breakdown of 123 patients with systemic mastocytosis associated with other myeloid malignancies. Blood 2009;114:3769– 3772. 23. Pardanani A, Reeder T, Li CY, et al. Eosinophils are derived from the neoplastic clone in patients with systemic mastocytosis and eosinophilia. Leuk Res 2003;27:883–885. 24. Taylor ML, Sehgal D, Raffeld M, et al. Demonstration that mast cells, T cells, and B cells bearing the activating kit mutation D816V occur in clusters within the marrow of patients with mastocytosis. J Mol Diagn 2004;6:335–342. 25. Tefferi A, Lasho TL, Brockman SR, et al. FIP1L1-PDGFRA and c-kit D816V mutationbased clonality studies in systemic mast cell disease associated with eosinophilia. Haematologica 2004;89:871–873. 26. Dunphy CH. Evaluation of mast cells in myeloproliferative disorders and myelodysplastic syndromes. Arch Pathol Lab Med 2005;129:219– 222. 27. Miettinen M, Lasota J. KIT (CD117): A review on expression in normal and neoplastic tissues, and mutations and their clinicopathologic correlation. Appl Immunohistochem Mol Morphol 2005;13:205–220. 28. Valent P, Spanblochl E, Sperr WR, et al. Induction of differentiation of human mast cells from bone marrow and peripheral blood mononuclear cells by recombinant human stem cell factor/kitligand in long-term culture. Blood 1992;80: 2237–2245. 29. Garcia-Montero AC, Jara-Acevedo M, Teodosio C, et al. KIT mutation in mast cells and other bone marrow hematopoietic cell lineages in systemic mast cell disorders: A prospective study of the spanish network on mastocytosis (REMA) in a series of 113 patients. Blood 2006;108: 2366–2372. 30. Buttner C, Henz BM, Welker P, et al. Identification of activating c-kit mutations in adult-, but not in childhood-onset indolent mastocytosis: A possible explanation for divergent clinical behavior. J Invest Dermatol 1998;111:1227– 1231. 31. Furitsu T, Tsujimura T, Tono T, et al. Identification of mutations in the coding sequence of the proto-oncogene c-kit in a human mast cell leukemia cell line causing ligand-independent activation of c-kit product. J Clin Invest 1993; 92:1736–1744. 32. Sotlar K, Escribano L, Landt O, et al. One-step detection of c-kit point mutations using peptide nucleic acid-mediated polymerase chain reaction clamping and hybridization probes. Am J Pathol 2003;162:737–746. 33. Beghini A, Cairoli R, Morra E, et al. In vivo differentiation of mast cells from acute myeloid leukemia blasts carrying a novel activating ligand-independent C-kit mutation. Blood Cells Mol Dis 1998;24:262–270. 34. Longley BJJ, Metcalfe DD, Tharp M, et al. Activating and dominant inactivating c-KIT catalytic domain mutations in distinct clinical forms of human mastocytosis. Proc Natl Acad Sci USA 1999;96:1609–1614. 35. Pullarkat VA, Pullarkat ST, Calverley DC, et al. Mast cell disease associated with acute myeloid leukemia: Detection of a new c-kit mutation Asp816His. Am J Hematol 2000;65:307–309. 36. Pignon JM, Giraudier S, Duquesnoy P, et al. A new c-kit mutation in a case of aggressive mast cell disease. Br J Haematol 1997;96:374–376. 37. Bodemer C, Hermine O, Palmerini F, et al. Pediatric mastocytosis is a clonal disease associated with D816V and other activating c-KIT mutations. J Invest Dermatol 2010;130:804–815. 38. Yanagihori H, Oyama N, Nakamura K, et al. ckit mutations in patients with childhood-onset mastocytosis and genotype-phenotype correlation. J Mol Diagn 2005;7:252–257. 39. Verzijl A, Heide R, Oranje AP, et al. C-kit Asp816-val mutation analysis in patients with mastocytosis. Dermatology 2007; 214:15–20.

doi:10.1002/ajh.23931

40. Gari M, Goodeve A, Wilson G, et al. c-kit proto-oncogene exon 8 in-frame deletion plus insertion mutations in acute myeloid leukaemia. Br J Haematol 1999;105:894–900. 41. Goemans BF, Zwaan CM, Miller M, et al. Mutations in KIT and RAS are frequent events in pediatric core-binding factor acute myeloid leukemia. Leukemia 2005;19:1536–1542. 42. Hartmann K, Wardelmann E, Ma Y, et al. Novel germline mutation of KIT associated with familial gastrointestinal stromal tumors and mastocytosis. Gastroenterology 2005;129:1042– 1046. 43. Zhang LY, Smith ML, Schultheis B, et al. A novel K509I mutation of KIT identified in familial mastocytosis-in vitro and in vivo responsiveness to imatinib therapy. Leuk Res 2006;30:373– 378. 44. Lux ML, Rubin BP, Biase TL, et al. KIT extracellular and kinase domain mutations in gastrointestinal stromal tumors. Am J Pathol 2000; 156:791–795. 45. Akin C, Fumo G, Yavuz AS, et al. A novel form of mastocytosis associated with a transmembrane c-kit mutation and response to imatinib. Blood 2004;103:3222–3225. 46. Tang X, Boxer M, Drummond A, et al. A germline mutation in KIT in familial diffuse cutaneous mastocytosis. J Med Genet 2004;41:e88 47. Zappulla JP, Dubreuil P, Desbois S, et al. Mastocytosis in mice expressing human kit receptor with the activating Asp816Val mutation. J Exp Med 2005;202:1635–1641. 48. Gerbaulet A, Wickenhauser C, Scholten J, et al. Mast cell hyperplasia, B cell malignancy, and intestinal inflammation in mice with conditional expression of a constitutively active kit. Blood 2011;117:2012–2021. 49. Tefferi A, Levine RL, Lim KH, et al. Frequent TET2 mutations in systemic mastocytosis: Clinical, KITD816V and FIP1L1-PDGFRA correlates. Leukemia 2009;23:900–904. 50. Wilson TM, Maric I, Simakova O, et al. Clonal analysis of NRAS activating mutations in KITD816V systemic mastocytosis. Haematologica 2011;96:459–463. 51. Soucie E, Hanssens K, Mercher T, et al. In aggressive forms of mastocytosis, TET2 loss cooperates with c-KITD816V to transform mast cells. Blood 2012;120:4846–4849. 52. Guo X, Schrader KA, Xu Y, et al. Expression of a constitutively active mutant of M-ras in normal bone marrow is sufficient for induction of a malignant mastocytosis/mast cell leukemia, distinct from the histiocytosis/monocytic leukemia induced by expression of activated H-ras. Oncogene 2005;24:2330–2342. 53. Pardanani A. How I treat patients with indolent and smoldering mastocytosis (rare conditions but difficult to manage). Blood, 2013;121:3085– 3094. 54. Pardanani A, Lim KH, Lasho TL, et al. WHO subvariants of indolent mastocytosis: Clinical details and prognostic evaluation in 159 consecutive adults. Blood 2010;115:150–151. 55. Horny HP, Sillaber C, Menke D, et al. Diagnostic value of immunostaining for tryptase in patients with mastocytosis. Am J Surg Pathol 1998;22:1132–1140. 56. Horny HP, Valent P. Histopathological and immunohistochemical aspects of mastocytosis. Int Arch Allergy Immunol 2002;127:115–117. 57. Sotlar K, Horny HP, Simonitsch I, et al. CD25 indicates the neoplastic phenotype of mast cells: A novel immunohistochemical marker for the diagnosis of systemic mastocytosis (SM) in routinely processed bone marrow biopsy specimens. Am J Surg Pathol 2004;28:1319–1325. 58. Jordan JH, Walchshofer S, Jurecka W, et al. Immunohistochemical properties of bone marrow mast cells in systemic mastocytosis: Evidence for expression of CD2, CD117/kit, and bcl-x(L). Hum Pathol 2001;32:545–552. 59. Sotlar K, Cerny-Reiterer S, Petat-Dutter K, et al. Aberrant expression of CD30 in neoplastic mast cells in high-grade mastocytosis. Mod Pathol 2011;24:585–595.

SM in adults 60. Morgado JM, Perbellini O, Johnson RC, et al. CD30 expression by bone marrow mast cells from different diagnostic variants of systemic mastocytosis. Histopathology 2013;63:780–787. 61. Escribano L, Garcia Montero AC, Nunez R, et al. Flow cytometric analysis of normal and neoplastic mast cells: Role in diagnosis and follow-up of mast cell disease. Immunol Allergy Clin North Am 2006;26:535–547. 62. Pardanani A, Kimlinger T, Reeder T, et al. Bone marrow mast cell immunophenotyping in adults with mast cell disease: A prospective study of 33 patients. Leuk Res 2004;28:777–783. 63. Galli SJ, Kalesnikoff J, Grimbaldeston MA, et al. Mast cells as “tunable” effector and immunoregulatory cells: Recent advances. Annu Rev Immunol 2005;23:749–786. 64. Sperr WR, El-Samahi A, Kundi M, et al. Elevated tryptase levels selectively cluster in myeloid neoplasms: A novel diagnostic approach and screen marker in clinical haematology. Eur J Clin Invest 2009;39:914–923. 65. Akin C. Molecular diagnosis of mast cell disorders: A paper from the 2005 william beaumont hospital symposium on molecular pathology. J Mol Diagn 2006;8:412–419. 66. Yavuz AS, Lipsky PE, Yavuz S, et al. Evidence for the involvement of a hematopoietic progenitor cell in systemic mastocytosis from single-cell analysis of mutations in the c-kit gene. Blood 2002;100:661–665. 67. Sotlar K, Colak S, Bache A, et al. Variable presence of KITD816V in clonal haematological non-mast cell lineage diseases associated with systemic mastocytosis (SM-AHNMD). J Pathol 2010;220:586–595. 68. Sanchez-Munoz L, Alvarez-Twose I, GarciaMontero AC, et al. Evaluation of the WHO criteria for the classification of patients with mastocytosis. Mod Pathol 2011;24:1157–1168. 69. Morgado JM, Sanchez-Munoz L, Teodosio CG, et al. Immunophenotyping in systemic mastocytosis diagnosis: ‘CD25 positive’ alone is more informative than the ‘CD25 and/or CD2’ WHO criterion. Mod Pathol 2012;25:516–521. 70. Valent P, Akin C, Arock M, et al. Definitions, criteria and global classification of mast cell disorders with special reference to mast cell activation syndromes: A consensus proposal. Int Arch Allergy Immunol 2012;157:215–225. 71. Travis WD, Li CY, Bergstralh EJ, et al. Systemic mast cell disease. Analysis of 58 cases and literature review. Medicine (Baltimore) 1988;67:345– 368. 72. Escribano L, Alvarez-Twose I, Sanchez-Munoz L, et al. Prognosis in adult indolent systemic mastocytosis: A long-term study of the spanish network on mastocytosis in a series of 145 patients. J Allergy Clin Immunol 2009;124:514– 521. 73. Pardanani A, Finke C, Abdelrahman RA, et al. Increased circulating IL-2Ralpha (CD25) predicts poor outcome in both indolent and aggressive forms of mastocytosis: A comprehensive cytokine-phenotype study. Leukemia 2013;26: 1430–1433. 74. Teodosio C, Garcia-Montero AC, Jara-Acevedo M, et al. An immature immunophenotype of bone marrow mast cells predicts for multilineage D816V KIT mutation in systemic mastocytosis. Leukemia 2011;26:951–958. 75. Teodosio C, Garcia-Montero AC, Jara-Acevedo M, et al. Mast cells from different molecular and prognostic subtypes of systemic mastocytosis display distinct immunophenotypes. J Allergy Clin Immunol 2010;125:719–726, 726 e711-726 e714. 76. Nakamura R, Chakrabarti S, Akin C, et al. A pilot study of nonmyeloablative allogeneic hematopoietic stem cell transplant for advanced systemic mastocytosis. Bone Marrow Transplant 2006;37:353–358. 77. Ustun C, Reiter A, Scott BL, et al. Hematopoietic stem-cell transplantation for advanced systemic mastocytosis. J Clin Oncol 2014;32:3264– 3274.

American Journal of Hematology, Vol. 90, No. 3, March 2015

261

ANNUAL CLINICAL UPDATES IN HEMATOLOGICAL MALIGNANCIES

Pardanani 78. Gotlib J, Pardanani A, Akin C, et al. International working Group-myeloproliferative neoplasms research and treatment (IWG-MRT) & European competence network on mastocytosis (ECNM) consensus response criteria in advanced systemic mastocytosis. Blood 2013; 121:2393–2401. 79. Kluin-Nelemans HC, Jansen JH, Breukelman H, et al. Response to interferon alfa-2b in a patient with systemic mastocytosis. N Engl J Med 1992; 326:619–623. 80. Butterfield JH. Response of severe systemic mastocytosis to interferon alpha. Br J Dermatol 1998;138:489–495. 81. Butterfield JH, Tefferi A, Kozuh GF. Successful treatment of systemic mastocytosis with highdose interferon-alfa: Long-term follow-up of a case. Leuk Res 2005;29:131–134. 82. Kolde G, Sunderkotter C, Luger TA. Treatment of urticaria pigmentosa using interferon alpha. Br J Dermatol 1995;133:91–94. 83. Lehmann T, Beyeler C, Lammle B, et al. Severe osteoporosis due to systemic mast cell disease: Successful treatment with interferon alpha-2B. Br J Rheumatol 1996;35:898–900. 84. Lehmann T, Lammle B. IFNalpha treatment in systemic mastocytosis. Ann Hematol 1999;78: 483–484. 85. Takasaki Y, Tsukasaki K, Jubashi T, et al. Systemic mastocytosis with extensive polypoid lesions in the intestines; successful treatment with interferon-alpha. Intern Med 1998;37:484– 488. 86. Weide R, Ehlenz K, Lorenz W, et al. Successful treatment of osteoporosis in systemic mastocytosis with interferon alpha-2b. Ann Hematol 1996;72:41–43. 87. Hauswirth AW, Simonitsch-Klupp I, Uffmann M, et al. Response to therapy with interferon alpha-2b and prednisolone in aggressive systemic mastocytosis: Report of five cases and review of the literature. Leuk Res 2004;28:249– 257. 88. Lippert U, Henz BM. Long-term effect of interferon alpha treatment in mastocytosis. Br J Dermatol 1996;134:1164–1165. 89. Petit A, Pulik M, Gaulier A, et al. Systemic mastocytosis associated with chronic myelomonocytic leukemia: Clinical features and response to interferon alfa therapy. J Am Acad Dermatol 1995;32:850–853. 90. Worobec AS, Kirshenbaum AS, Schwartz LB, et al. Treatment of three patients with systemic mastocytosis with interferon alpha-2b. Leuk Lymphoma 1996;22:501–508. 91. Brunel V, Tadrist Z, Cailleres S, et al. [Interferon alpha and pamidronate: A useful combination in the treatment of osteoporosis and systemic mastocytosis]. Presse Med 1998;27:64 92. Czarnetzki BM, Algermissen B, Jeep S, et al. Interferon treatment of patients with chronic urticaria and mastocytosis. J Am Acad Dermatol 1994;30:500–501. 93. Hennessy B, Giles F, Cortes J, et al. Management of patients with systemic mastocytosis: Review of M. D. Anderson cancer center experience. Am J Hematol 2004;77:209–214. 94. Delaporte E, Pierard E, Wolthers BG, et al. Interferon-alpha in combination with corticosteroids improves systemic mast cell disease. Br J Dermatol 1995;132:479–482. 95. Lippert U, Henz BM. Long-term effect of interferon alpha treatment in mastocytosis.[comment]. Br J Dermatol 1996;134:1164–1165.

262

96. Lim KH, Pardanani A, Butterfield JH, et al. Cytoreductive therapy in 108 adults with systemic mastocytosis: Outcome analysis and response prediction during treatment with interferon-alpha, hydroxyurea, imatinib mesylate or 2-chlorodeoxyadenosine. Am J Hematol 2009;84:790–794. 97. Simon J, Lortholary O, Caillat-Vigneron N, et al. Interest of interferon alpha in systemic mastocytosis. The french experience and review of the literature. Pathol Biol (Paris) 2004;52: 294–299. 98. Bohm A, Sonneck K, Gleixner KV, et al. In vitro and in vivo growth-inhibitory effects of cladribine on neoplastic mast cells exhibiting the imatinib-resistant KIT mutation D816V. Exp Hematol 2010;38:744–755. 99. Gleixner KV, Mayerhofer M, Aichberger KJ, et al. PKC412 inhibits in vitro growth of neoplastic human mast cells expressing the D816Vmutated variant of KIT: Comparison with AMN107, imatinib, and cladribine (2CdA) and evaluation of cooperative drug effects. Blood 2006;107:752–759. 100. Penack O, Sotlar K, Noack F, et al. Cladribine therapy in a patient with an aleukemic subvariant of mast cell leukemia. Ann Hematol 2005; 84:692–693. 101. Tefferi A, Li CY, Butterfield JH, et al. Treatment of systemic mast-cell disease with cladribine. N Engl J Med 2001;344:307–309. 102. Kluin-Nelemans HC, Oldhoff JM, Van Doormaal JJ, et al. Cladribine therapy for systemic mastocytosis. Blood 2003;102:4270–4276. 103. Pardanani A, Hoffbr AV, Butterfield JH, et al. Treatment of systemic mast cell disease with 2chlorodeoxyadenosine. Leuk Res 2004;28:127– 131. 104. Hermine O, Hirsh I, Damaj G, et al. Long term efficacy and safety of cladribine in adult systemic mastocytosis: A French Multicenter Study of 44 Patients. Blood ASH Annual Meeting Abstracts 2010;116:1982. 105. Zermati Y, De Sepulveda P, Feger F, et al. Effect of tyrosine kinase inhibitor STI571 on the kinase activity of wild-type and various mutated c-kit receptors found in mast cell neoplasms. Oncogene 2003;22:660–664. 106. Akin C, Brockow K, D’Ambrosio C, et al. Effects of tyrosine kinase inhibitor STI571 on human mast cells bearing wild-type or mutated c-kit. Exp Hematol 2003;31:686–692. 107. Ma Y, Zeng S, Metcalfe DD, et al. The c-KIT mutation causing human mastocytosis is resistant to STI571 and other KIT kinase inhibitors; kinases with enzymatic site mutations show different inhibitor sensitivity profiles than wild-type kinases and those with regulatorytype mutations. Blood 2002;99:1741–1744. 108. Nakagomi N, Hirota S. Juxtamembrane-type ckit gene mutation found in aggressive systemic mastocytosis induces imatinib-resistant constitutive KIT activation. Lab Invest 2007;87:365– 371. 109. Droogendijk HJ, Kluin-Nelemans HJ, van Doormaal JJ, et al. Imatinib mesylate in the treatment of systemic mastocytosis: A phase II trial. Cancer 2006;107:345–351. 110. Vega-Ruiz A, Cortes JE, Sever M, et al. Phase II study of imatinib mesylate as therapy for patients with systemic mastocytosis. Leuk Res 2009;33:1481–1484. 111. Pagano L, Valentini CG, Caira M, et al. Advanced mast cell disease: An Italian hemato-

American Journal of Hematology, Vol. 90, No. 3, March 2015

112.

113.

114.

115.

116.

117.

118. 119.

120.

121.

122.

123.

124.

logical multicenter experience. Int J Hematol 2008;88:483–488. Schittenhelm MM, Shiraga S, Schroeder A, et al. Dasatinib (BMS-354825), a dual SRC/ ABL kinase inhibitor, inhibits the kinase activity of wild-type, juxtamembrane, and activation loop mutant KIT isoforms associated with human malignancies. Cancer Res 2006;66:473– 481. Shah NP, Lee FY, Luo R, et al. Dasatinib (BMS-354825) inhibits KITD816V, an imatinib-resistant activating mutation that triggers neoplastic growth in most patients with systemic mastocytosis. Blood 2006;108:286– 291. Gleixner KV, Mayerhofer M, Sonneck K. et al. Synergistic growth-inhibitory effects of two tyrosine kinase inhibitors, dasatinib and PKC412, on neoplastic mast cells expressing the D816V-mutated oncogenic variant of KIT. Haematologica 2007;92:1451–1459. Aichberger KJ, Sperr WR, Gleixner KV, et al. Treatment responses to cladribine and dasatinib in rapidly progressing aggressive mastocytosis. Eur J Clin Invest 2008;38:869–873. Ustun C, Corless CL, Savage N, et al. Chemotherapy and dasatinib induce long-term hematologic and molecular remission in systemic mastocytosis with acute myeloid leukemia with KIT D816V. Leuk Res 2009;33:735–741. Verstovsek S, Tefferi A, Cortes J, et al. Phase II study of dasatinib in philadelphia chromosome-negative acute and chronic myeloid diseases, including systemic mastocytosis. Clin Cancer Res 2008;14:3906–3915. Purtill D, Cooney J, Sinniah R, et al. Dasatinib therapy for systemic mastocytosis: Four cases. Eur J Haematol 2008;80:456–458. Growney JD, Clark JJ, Adelsperger J, et al. Activation mutations of human c-KIT resistant to imatinib mesylate are sensitive to the tyrosine kinase inhibitor PKC412. Blood 2005;106: 721–724. Gotlib J, Berube C, Growney JD, et al. Activity of the tyrosine kinase inhibitor PKC412 in a patient with mast cell leukemia with the D816V KIT mutation. Blood 2005;106:2865– 2870. Gotlib J, Kluin-Nelemans H, George T, et al. Midostaurin (PKC412) Demonstrates a high rate of durable responses in patients with advanced systemic mastocytosis: Results from the fully accrued global phase 2 CPKC412D2201 trial. Blood 2014;124:Abstract 636. Dubreuil P, Letard S, Ciufolini M, et al. Masitinib (AB1010), a potent and selective tyrosine kinase inhibitor targeting KIT. PLoS One 2009; 4:e7258 Paul C, Sans B, Suarez F, et al. Masitinib for the treatment of systemic and cutaneous mastocytosis with handicap: A phase 2a study. Am J Hematol 2010;85:921–925. Moura DS, Sultan S, Georgin-Lavialle S, et al. Depression in patients with mastocytosis: Prevalence, features and effects of masitinib therapy. PLoS One 2011;6:e26375

doi:10.1002/ajh.23931

Systemic mastocytosis in adults: 2015 update on diagnosis, risk stratification, and management.

Systemic mastocytosis (SM) results from a clonal proliferation of abnormal mast cells (MC) in one or more extracutaneous organs...
1MB Sizes 0 Downloads 7 Views