Abstract
Systemic mastocytosis is a heterogeneous disease characterized by the accumulation of neoplastic mast cells in the bone marrow and other organ organs/tissues. Mutations in KIT, most frequently KIT D816V, are detected in over 80% of all systemic mastocytosis patients. While most systemic mastocytosis patients suffer from an indolent disease variant, some present with more aggressive variants, collectively called “advanced systemic mastocytosis”, which include aggressive systemic mastocytosis, systemic mastocytosis with an associated hematologic, clonal non mast cell-lineage disease, and mast cell leukemia. Whereas patients with indolent systemic mastocytosis have a near normal life expectancy, patients with advanced systemic mastocytosis have a reduced life expectancy. Although cladribine and interferon-alpha are of benefit in a group of patients with advanced systemic mastocytosis, no curative therapy is available for these patients except possible allogeneic hematopoietic stem cell transplantation. Recent studies have also revealed additional somatic defects (apart from mutations in KIT) in a majority of patients with advanced systemic mastocytosis. These include TET2, SRSF2, ASXL1, RUNX1, JAK2, and/or RAS mutations, which may adversely impact prognosis and survival in particular systemic mastocytosis with an associated hematological neoplasm. In addition, several additional signaling molecules involved in the abnormal proliferation of mast cells in systemic mastocytosis have been identified. These advances have led to a better understanding of the biology of advanced systemic mastocytosis and to the development of new targeted treatment concepts. Herein, we review the biology and pathogenesis of advanced systemic mastocytosis, with a special focus on novel molecular findings as well as current and evolving therapeutic options.Introduction
Mastocytosis comprises a pathomorphologically and clinically heterogeneous spectrum of localized or systemic disorders characterized by an abnormal accumulation of mast cells (MCs) in one or more organs.1 In children, the disease is mostly restricted to the skin (cutaneous mastocytosis: CM).32 By contrast, adult patients usually present with systemic mastocytosis (SM). In patients with SM, neoplastic MCs are almost always detectable in the bone marrow (BM), and usually also in other internal organs.741 The exact incidence of SM remains uncertain, but a prevalence of mastocytosis including all the subtypes is estimated to be approximately 1 in 10,000 people.8 A recent study from Denmark showed the incidence rate for all SM, including CM, was 0.89 per 100,000/year.9
The World Health Organization (WHO) classification has defined major categories and variants of SM (Online Supplementary Table S1).1061 Most adult patients present with indolent SM (ISM), which is mainly characterized by mediator-related symptoms, frequent skin involvement, no organ dysfunction and a nearly normal life expectancy.1 By contrast, in advanced variants of the disease (AdvSM), including SM with an associated clonal hematologic non-MC lineage disease (SM-AHNMD; recently updated to systemic mastocytosis with an associated hematological neoplasm (SM-AHN) by WHO),11 aggressive SM (ASM), and mast cell leukemia (MCL), the malignant expansion and accumulation of neoplastic MCs can lead to organ damage (“C-findings”, Online Supplementary Table S2).76 No skin lesions are found in some patients.12 Depending on the subtype, the survival of patients with AdvSM ranges from a few months to several years,14131 therefore cytoreductive therapy is indicated in most of these patients.15
Response criteria were developed (Online Supplementary Table S2),16 and updated and detailed17 for clinical trials by a consensus group.
Molecular defects found in advanced systemic mastocytosis KIT mutations and their sensitivity to tyrosine kinase inhibitors
KIT is a type III tyrosine kinase (TK) transmembrane receptor for stem cell factor (SCF), which is the major growth factor of MCs in humans (Figure 1).18 Interestingly, in most cases of SM (overall >80%, in typical ISM >90%, and in AdvSM >70%), an acquired point mutation in the gene coding for KIT (CD117) is found. Although KIT D816V, an activation loop mutation, is the most common mutation found, more than 20 other mutations in KIT have been described in SM.2019 The exact percentages vary, depending on disease subtypes (e.g. ISM vs. ASM) and cell source [e.g. BM vs. peripheral blood (PB)].18 The KIT D816V mutation is detected in AHN cells in the majority of cases, which reflects multilineage involvement.2321 There are, however, cases in which two independent (sub)clones exist and this might depend on the type of AHN.2524 KIT mutations often cause ligand-independent constitutive phosphorylation and activation of KIT, which transforms cell lines from factor-dependent growth to factor independence and tumorigenicity.2826 Longley et al. proposed to divide activating mutations of KIT into two types: “regulatory type” mutations affecting regulation of the kinase molecule, and “enzymatic pocket type” mutations, which change the amino acid sequence of the enzymatic site.29 These latter mutations induce stabilization of the activation loop in an active conformation and/or structural alteration at the ATP-binding site of KIT, resulting in a decreased affinity for type I TK inhibitors (TKI), such as imatinib, that recognize the active conformation of a kinase. The MCL-like cell line HMC-1 has developed two sub-clones: HMC-1.1 which harbors a juxtamembrane domain (JMD) regulatory type mutation, KITV560G, and HMC-1.2 expressing both KIT D816V and KITV560G.30 Imatinib inhibits only the regulatory type mutant affecting the juxtamembrane inhibitory helix, but does not significantly inhibit KIT D816V.31 However, even some JMD-type KIT mutations (e.g. KITV559I) can cause imatinib resistance by leading to structural changes of the JMD of KIT, which affects the structure of the kinase domain.32 Other TKIs, such as PKC412 (midostaurin) effectively suppress the activity of imatinib-resistant KIT mutants.3533 Of note, the allele burden of the KIT mutant, determined by highly sensitive techniques, such as allele specific quantitative PCR (ASO-qPCR), correlates with the burden of neoplastic MCs, and with survival and prognosis.373618 Finally, although the KIT D816V mutant is recurrently found in SM patients, a recent report has pointed to the possibility that such patients may present with concurrent mutations in other codons of the KIT gene.38 Indeed, out of 21 patients analyzed, the authors found 3 (15%) patients with KIT D816V and a concurrent mutation.38 Overall, these data suggest an advantage for double mutations that might contribute to the aggressiveness of SM.
Tyrosine kinase inhibitors (TKI)
Midostaurin (PKC412): Midostaurin (PKC412) is an oral multi-kinase inhibitor with activity against protein kinase C (PKC), FMS-related tyrosine kinase 3 (FLT3), PDGFRA/B, vascular endothelial growth factor receptor 2 (VEGFR-2), and KIT. Midostaurin was evaluated in a centrally adjudicated, phase II multi-center international study in 116 patients with ASM, of which 89 were evaluable for efficacy.39 Overall, 73 patients (82%) had ASM, 16 (18%) had MCL, and 63/89 patients (71%) had an AHN. Seventy-seven patients (87%) were positive for a codon 816 KIT mutation. After a median follow-up of 26 months (range 12–54 months), the overall response rate (ORR) was 60%. Most responses were major (75%), including decreases of >50% in serum tryptase and BM mast cell levels. These responses were durable: the median duration of response and median OS were 24.1 and 28.7 months, respectively. Median OS was 9.4 months in patients with MCL; however, responders in the MCL group did not reach a median OS. Midostaurin was tolerated fairly well with grade 1–2 gastrointestinal side effects being the most common adverse events (Table 1). Patient-reported outcomes, including symptoms and quality of life, measured by the Memorial Symptom Assessment Scale and the Short Form-12 Health Survey, respectively, significantly improved with midostaurin therapy. These results indicate that the drug has a favorable efficacy and safety profile with activity in AdvSM regardless of KIT mutation status. Although midostaurin has not been approved by major drug authorities in either Europe or the USA, it is available for patients with AdvSM within a compassionate use program sponsored by the drug company.
After the remarkable success of TKIs in chronic myeloid leukemia (CML), significant enthusiasm for TKI in the treatment of SM emerged in the early 2000s.19 However, imatinib is largely ineffective in patients with KIT D816V SM.40 On the other hand, some patients with SM may respond very well to imatinib, especially those with other KIT mutations such as K509I,41 F522C42 or KIT WT.43 In patients with FIP1L1-PDGFRA-positive myeloid neoplasms with eosinophilia, small doses of imatinib (100 mg/d) will effect durable hematologic and cytogenetic/molecular remission in almost all cases.4544 Although some of these patients may exhibit scattered/interstitial distributions of increased abnormal CD25 MCs in the BM, these cases are not considered a subtype of SM by the WHO because typical dense infiltrates of spindle-shaped mast cells are missing. In SM patients with KIT WT, imatinib may even induce CR with the disappearance of skin lesions and return of elevated serum tryptase levels to the normal range (<15 ng/mL).46 Imatinib (400 mg daily) is still the only TKI approved by the US Food and Drug Administration (FDA) for adult patients who have ASM either without the KIT D816V mutation or with unknown KIT mutational status.
Dasatinib, a multikinase inhibitor (e.g. BCR-ABL1, KIT, and PDGFRα),4847 has proven to be effective in vitro against KIT D816V+ neoplastic MCs.49 However, the half-life of the drug is very short, and no durable and meaningful clinical responses were observed in clinical studies in AdvSM (Table 1).50
Masitinib, which inhibits KIT WT and LYN,51 is an effective drug for canine MC tumors.52 However, in humans, the KIT D816V mutation introduces resistance against masitinib. In one study, masitinib was administered daily (3–6 mg orally) for 12 weeks in 25 patients diagnosed as having SM or CM with a related “handicap” (i.e. disabilities associated with flushes, depression, pruritus and quality of life).53 ORR was 56% by AFIRMM response criteria.54 Severe toxicities occurred in <10% of all patients. Currently, a larger study is being performed in patients with CM and ISM with a “handicap” (AdvSM was excluded).
In a phase II trial of 61 patients with SM (37 with AdvSM), nilotinib (400 mg twice a day) induced overall responses of 21.6% (including a decrease in serum tryptase and BM mast cells) and of 21% in ASM.55 All responders had the KIT D816V mutation. Nine AdvSM patients died during 34.7 months of follow-up. No active study is currently being performed with nilotinib.
Most of the data on these TKIs resulted from pre-clinical studies or case reports. Ponatinib, a multi-kinase blocker, inhibits the kinase activity of KITV560G and, less effectively, KIT D816V in HMC-1 cells.5635 Ponatinib induced dose-dependent growth inhibition and apoptosis in primary neoplastic MCs, HMC-1.1 cells, and HMC-1.2 cells.56 Ponatinib and midostaurin were found to exert synergistic growth-inhibitory effects against neoplastic MCs harboring the KIT D816V mutant.56 Other novel TKIs with potent TKI inhibiting properties (e.g. EXEL-0862)57 and thiazole amine derivatives inhibiting β-catenin signaling (e.g. semaxinib (SU5416) and compound 126332).5958 BLU-285, a selective KIT D816V inhibitor with encouraging pre-clinical activity and a narrow target profile, is expected to enter clinical trial testing in AdvSM in the near future.60
Progress in somatic mutations other than KIT in SM
Recent studies have reported the presence of additional, recurrent somatic mutations (apart from KIT mutations) in AdvSM, especially in SM-AHN, including mutations in TET2, SRSF2, ASXL1, RUNX1, JAK2, and/or RAS (Figure 2).6561
Mutations in TET2, also detected in healthy individuals,66 cause loss of function (i.e. regulating gene expression at the cellular level),67 and are associated with increased self-renewal capacity of hematopoietic stem cells.68 Recently, several investigators have identified TET2 mutations scattered across several of its 12 exons in 1 or both TET2 alleles, as an early event during the development of various malignancies.69 Patients with mutant TET2+ myeloid disorders show a decreased level of 5-hmC with hypomethylation or hypermethylation of DNA.70 Altogether these data show that TET2 plays a role in various hematologic malignancies. In line with these recently published data, TET2 mutations have been reported in 20–40% of KIT D816V-positive AdvSM patients.64626125 The cooperation between KIT D816V and loss of function of TET2 in MC results in transformation to a more aggressive disease phenotype in mice.71 It has also been suggested that TET2 mutations can occur before KIT D816V in ASM-AHN patients.72 Thus, the acquisition of KIT D816V might act as a phenotype modifier of ASM in these cases.72 Patients carrying a combination of TET2 and DNMT3A (a DNA methyltransferase) mutations have a poor prognosis compared to those with wild-type genes.61 In vitro, a combination of dasatinib and decitabine (a hypomethylating agent) was more effective at inducing apoptosis and cell death in HMC-1.2 cells harboring a TET2 mutant compared to each compound alone.71 This combination also had less effect in TET2 wild-type cells due to a lower efficacy of decitabine. The impact of TET2 mutations on overall survival remains uncertain.736261
The spliceosome machinery includes SRSF2, U2AF1, and SF3B1 proteins, and is involved in the removal of introns from a transcribed pre-mRNA.74 Mutations in the spliceosome machinery have recently been identified using whole exome/genome technologies in MDS and MPN.75 A mutation in the hotspot region of SRSF2 (codon P95) is found in approximately 1/3 of AdvSM patients6564 but is usually not detectable in patients with ISM or SSM.25 It is more common in ASM-AHN656425 and precedes KIT D816V in these patients.25 The frequency of SF3B1 mutations in AdvSM is low, ranging from 0 to 5%.6564 U2AF1 mutations are less frequently reported in SM.6564
The gene ASXL1 (additional sex combs–like 1) encodes for a protein of the polycomb group and trithorax complex family, which interacts with retinoic acid receptor and may be involved in chromatin remodeling.76 The presence of ASXL1 mutations has been reported in SM at various frequencies,73646125 and alone or with other mutations seems to be a poor prognostic factor for OS in patients.736461 RUNX1, and less frequently, JAK2 mutations, are found in AdvSM, but not in ISM or SSM.64 The frequency of RAS mutations (e.g. NRAS, KRAS or HRAS) in SM has been investigated, with KRAS and NRAS mutants being found in AdvSM at a relatively low frequency, and not usually detectable in patients with ISM.6463
The presence of additional genetic defects in KIT D816V+ AdvSM patients may confer adverse prognosis as compared with patients without such abnormalities.7264 In a recent study, Jawhar et al. have analyzed the impact of several additional defects on 70 multi-mutated KIT D816V+ patients with an AHN.77 In this study, the most frequently identified mutated genes were TET2 (n=33 of 70 patients), SRSF2 (n=30), ASXL1 (n=20), RUNX1 (n=16) and JAK2 (n=11).77 In multivariate analysis, SRSF2 and ASXL1 remained the most predictive adverse indicators concerning OS. Furthermore, the authors found that inferior OS and adverse clinical characteristics were significantly influenced by the number of mutated genes in the SRSF2/ASXL1/RUNX1 (S/A/R) panel (P<0.0001).77
It appears that, based on these findings, the inclusion of molecular markers should be considered in upcoming prognostic scoring systems for patients with SM. This might be particularly important for patients with SM-AHN given that most of these studies were done in patients with SM-AHN.7873 Although it is arguable that these mutations could be detected due to the copresence of an AHN, there are recent studies in pure SM showing these mutations as well.7972 In addition, it has been described in many previous reports that KIT mutations are not restricted to the mast cell disease components in SM-AHN.80 Although we are at an early stage in the understanding of the clinical and biological importance of these mutations in SM, most likely these mutations affect hematopoietic stem and progenitor cells, and the rate of multilineage involvement increases with the aggressiveness of SM.
In addition, recent investigations on mutational profiles of colonies grown from granulocyte-macrophage colony-forming progenitor cells (CFU-GM) and microdissected mature cells (tryptase or CD15 positive) revealed that these additional mutations develop prior to KIT D816V in almost all patients, indicating a multi-mutated stem cell disease with strong phenotype modification (i.e. the mastocytosis component) driven by KIT D816V.25
Critical intracellular pro-oncogenic pathways in neoplastic mast cells as novel potential therapeutic targets
Several studies have reported that the ability of wild-type and oncogenic mutant forms of KIT to induce signal transduction differs not only quantitatively but also qualitatively. These altered pathways, which are presented in Figure 3 together with potential targeted drugs, may have an effect on several properties of neoplastic MCs by reducing apoptosis and/or by inducing alterations in the cell cycle.
MCL-1, a BCL-2 family member with anti-apoptotic properties, is expressed in primary neoplastic MCs in SM as well as in the HMC-1.1 and HMC-1.2 cell lines.81 The targeting of MCL-1 by antisense oligonucleotides (ASOs) or MCL-1-specific siRNA resulted in reduced survival and increased apoptosis in these cell lines.81 Moreover, MCL-1 ASOs cooperated with various KIT-targeting TKIs in producing growth inhibition in neoplastic MC lines.81
BIM, a pro-apoptotic member of the BCL-2 family, has been identified as a tumor suppressor in neoplastic MCs.82 BIM is downregulated in neoplastic MCs by SCF as well as by KIT D816V.82 Midostaurin, bortezomib (a proteasome inhibitor), and obatoclax (a pan-BCL-2 family blocker) reportedly upregulate BIM expression in HMC-1 cells and may thereby promote apoptosis.8382 Obatoclax also increased apoptosis in these cells.83
Activated LYN and BTK are expressed in neoplastic MCs in a KIT-independent manner in patients with ASM and MCL, and may thus contribute to malignant transformation.49 LYN is a member of the SRC family involved in cellular signaling processes regulating growth, differentiation, and apoptosis. Activated LYN regulates BTK function and may influence the process of degranulation and cytokine production in MCs.8584 Dasatinib and bosutinib (SRC inhibitors) disrupt LYN and BTK activation and oncogenic signaling in neoplastic MCs.49 Bosutinib inhibits the growth of neoplastic MCs in vitro at relatively high concentrations, with no effect on KIT.8649 Bosutinib acts synergistically with midostaurin on HMC-1 cell proliferation.49 However, bosutinib is unable to induce any response in patients with AdvSM.87
Phosphoinositide 3-kinase (PI3-K), a lipid kinase, is important for the function of intracellular signaling molecules, like BTK, AKT and PDK1, by inducing phosphatidylinositol 3,4,5-trisphosphate (PIP3) that provides membrane docking sites for these signaling molecules.88 In both HMC-1 subclones (HMC-1.1 and HMC-1.2), mutated KIT leads to constitutive activation of PI3-K.89 Once activated, the PI3-K subsequently activates AKT,89 a key signaling molecule involved in KIT-dependent differentiation and growth of neoplastic MCs harboring oncogenic KIT mutants.90 Indeed, AKT was found to be phosphorylated in neoplastic MCs in patients with KIT D816V+ SM and in the HMC-1.2 cell line.9190
PI3-K and AKT are also important for the regulation of the mammalian target of rapamycin (mTOR), a serine/threonine kinase that interacts with 2 regulatory protein complexes called mTOR complex 1 (mTORC1) and complex 2 (mTORC2). PI3-K regulates the mTORC1 pathway via the activation of AKT which directly inactivates tuberin, the inhibitor of mTOR activation. Once activated, mTORC1 phosphorylates p70 ribosomal S6 kinase (p70S6K), resulting in increased gene transcription that regulates cell growth, survival, protein synthesis and metabolism. Smrz et al. showed that the expression and activation of mTORC1 and mTORC2 was increased in neoplastic human MC lines and in immature normal MCs, as compared with mature normal MCs.92 Interestingly, the authors demonstrated that mTORC1 might contribute to MC survival, while mTORC2 might only fulfill critical functions in the context of proliferating (dividing) neoplastic and immature MCs.92 Rapamycin, a specific inhibitor of mTORC1, has been shown to block FcεRI- and KIT-induced mTORC1-dependent p70S6K phosphorylation in normal MCs.88 Furthermore, BEZ235, a dual PI3-K/mTOR blocker, exerted strong growth-inhibitory effects on neoplastic MCs in vitro.93 Of note, BEZ235 was also found to reduce the engraftment and growth of HMC-1 cells in a xenotransplanted mouse model employing NMR1-Foxn1(nu) mice.93 Everolimus, another mTOR-blocker, was ineffective in patients with SM.94
Neoplastic MCs express cytoplasmic and nuclear phos-pho-STAT5 (pSTAT5).95 In an in vitro study,90 knockdown of STAT5 was followed by growth inhibition of neoplastic MCs. Furthermore, it has been shown that KIT D816V directly promotes STAT5-activation, and that pSTAT5 contributes to the growth of neoplastic MCs.95 This makes STAT5 an attractive target for therapy in AdvSM. However, until now, most drugs targeting STAT5 exert anti-neoplastic effects only at high, non-pharmacological concentrations in vitro. The inhibition of the JAK-STAT signaling pathway in vitro decreased KIT D816V-mediated cell growth.96 Ruxolitinib, a JAK1/2 inhibitor, has shown clinical benefit in patients with MPN regardless of JAK2 V617F-mutation.97 Ruxolitinib decreased spleen size and improved blood counts in a KIT-mutated but not JAK2-mutated patient with SM-MPN primary myelofibrosis.98 Therefore, JAK1/2 blockers can be considered in studies of patients with SM-MPN.
NF-κB, a dimeric transcription factor of the REL family, was found to be spontaneously activated in HMC-1 cells.99 IMD-0354 inhibited translocation of NF-κB to the nucleus, and thus led to decreased cyclin D3 expression and increased cell cycle arrest in HMC-1 cells in vitro.99 Another transcription factor of the REL family, nuclear factor of activated T cells (NFAT), has also been found constitutively activated in KIT-mutated neoplastic MCs.100 The combination of a KIT inhibitor and of a calcineurin phosphatase inhibitor (a NFAT regulator) exhibited a synergistic inhibitory effect on cell viability and survival in KIT-mutated MC lines.100
One promising class of targets within chromatin regulatory molecules and related antigens are the bromodomain (BRD)-containing proteins.103101 Indeed, inhibition of the epigenetic reader bromodomain-containing protein-4 (BRD4) by exposure to RNA interference or treatment with JQ1, a drug blocking the specific interactions between BRD4 and acetylated histones, resulted in major antileukemic effects in murine and human AML cells.102 More recently, BRD4 has been identified as a novel drug target in AdvSM.104 The authors showed that neoplastic MCs expressed substantial amounts of BRD4 in ASM and MCL, as assessed by immunohistochemistry and PCR.104 They also reported that the human MCL lines HMC-1 and ROSA also expressed BRD4, and that a BRD4-specific short hairpin RNA or the BRD4-targeting drug JQ1 induced dose-dependent growth inhibition and apoptosis in HMC-1 and ROSA cells, regardless of the presence or absence of the KIT D816V mutant.104 Moreover, the authors demonstrated that JQ1 suppressed the proliferation of primary neoplastic MCs obtained from patients with ASM or MCL. Finally, in drug combination experiments, midostaurin (PKC412) and all-trans retinoic acids were found by the authors to cooperate with JQ1 in producing synergistic effects on survival in HMC-1 and ROSA cells.104 Taken together, these data identified BRD4 as a promising drug target in advanced SM. However, whether JQ1 or other BET bromodomain inhibitors are effective in vivo in patients with AdvSM remains to be elucidated.
Antibody-mediated therapeutic approach to target neoplastic mast cells and stem cells
Based on recent knowledge on the phenotype of malignant MCs and their neoplastic progenitors, a number of cell surface antigens might be aberrantly expressed, including CD13, CD25, CD30, CD33, CD44, CD52, CD87, and CD117, and therefore might be considered also as potential targets of therapy in AdvSM.110105 For example, neoplastic MCs and their progenitors have been shown to respond in vitro to gemtuzumab ozogamicin (a monoclonal antibody targeting CD33 combined to a cytostatic agent).111 The CD52-targeting antibody alemtuzumab induces cell death in neoplastic MCs in vitro and in mice xenotransplanted with HMC-1 cells.108 CD30 is expressed on the surface of neoplastic MCs in a proportion of patients with AdvSM, but not on normal/reactive MCs, making this antigen an attractive target of specific therapy in these patients.113112107 A single-arm, open-label clinical trial applying brentuximab vedotin (SGN-35) to patients with CD30-positive AdvSM (clinicaltrials.gov identifier: 01807598) is ongoing in the US. Neoplastic (leukemic) stem cells (LSCs) have recently been identified in AdvSM. These cells reside within a CD34 cell fraction and co-express aminopeptidase N (CD13), leukosialin (CD43), Pgp-1 (CD44), the IL-3R α-chain (CD123), AC133 (CD133), CXCR4 (CD184), CD33, CD52 and CD117.115114 As observed in chronic myeloid leukemia, a part of these LSCs might be non-cycling and therefore probably resistant to treatment with TKIs. Thus, a combination of a TKI that targets KIT on neoplastic MCs and a mAb targeting a surface antigen, such as CD52 for instance, expressed on non-cycling LSCs, may help to achieve a minimal residual disease negative state in AdvSM.
Conventional therapies with anti-neoplastic drugs and allogeneic hematopoietic cell transplantation
Cytarabine, fludarabine, hydroxyurea (a drug of choice in palliative care)15 and interferon-alpha (IFN−α),118116 have been frequently used for cytoreduction in the treatment of AdvSM. Hydroxyurea is useful to control leukocyte counts in AdvSM, especially in SM-AHN (palliative therapy) and in patients with comorbidity. Cladribine (2-CDA) is the most effective and frequently used drug. Kluin-Nelemans et al., used 2-CdA in 10 patients with SM, most of them suffering from AdvSM (Table 1).119 All patients responded concerning clinical symptoms and MC burden as reflected in declining serum tryptase values and urinary histamine metabolite excretion. Although no patient achieved a complete remission (CR), clinically meaningful and some durable responses were seen, suggesting that 2-CdA may be a potentially effective treatment option for some patients with severe SM.119 These results have been supported by more recent studies.123120 For instance, in a study on 44 SM patients, the median duration of response was 20 months; however, none of the patients with SM-AHN responded.122 However, 2-CdA usually does not control the disease for prolonged periods of time in rapidly progressing ASM and MCL. For these patients, more intensive therapy, such as AML-like multi-agent chemotherapy, including fludarabine and cytarabine124 should be considered in induction therapy and then for allogeneic hematopoietic cell transplantation (HCT) for consolidation therapy.127125 Allogeneic HCT remains the only potentially curative treatment option for patients with AdvSM. We have recently reported data on the effect of allo-HCT in patients with AdvSM (Table 1).125 Most patients (the median age was 46) received a graft from HLA-identical siblings (n=34) or unrelated donors (URD) (n=17). Overall survival (OS) and SM progression-free survival (PFS) at 3 years for all patients were 57% and 51%, respectively. They were significantly affected, however, by the type of advanced SM: 74% and 63%, respectively, for SM-AHN; 43% and 43%, respectively, for ASM; and 17% and 17%, respectively, for MCL. Although the data presented are very encouraging, future prospective studies, perhaps per recommended consensus opinion to homogenously collect data,128 are required to confirm the safety129 and efficacy of this treatment approach in AdvSM.
Miscellaneous aspects of management in AdvSM
Patients with SM-AHN should be treated according to generally accepted guidelines: the SM component of the disease is treated as if no AHN was diagnosed, and the AHN component of the disease is treated as if no SM was diagnosed, with the recognition of potential drug interactions1510 deciding whether the SM or AHN component is primarily contributing to organ damage or other related clinical, laboratory concerns. However, admittedly it is often not possible to clearly delineate whether one or the other component is responsible for the clinical issues/organ damage.
As a supportive therapy, H1-receptor antagonists, such as the classical antihistamine hydroxyzine, or non-sedating antihistamines, such as loratadine or fexofenadine, can be administered for the alleviation of symptoms caused by the release of the mediators (e.g. pruritus and flushing).132130
Conclusion and perspectives
Advanced variants of SM share two major characteristics: i) the prognosis of the disease remains poor, and ii) other than allogeneic HCT no curative therapy is available. Only a few drugs have shown beneficial effects in AdvSM (2-CdA, interferon-alpha, and midostaurin). We propose a treatment algorithm with current therapy options (Figure 4). However, this is a subject to change in the future due to remarkable progress in the biology of AdvSM. Neoplastic cells in SM are usually driven by a canonical KIT-downstream pathway as well as by additional somatic mutations and KIT-independent pathways and molecules, including TET2, the spliceosome machinery, ASXL1, or RAS. We may better prognosticate AdvSM using these additional genetic defects. The PI3-kinase, AKT, STAT-5, BTK, FES, mTORC2, and BCL-2 family members as well as certain surface molecules and disease initiating (quiescent) neoplastic stem cells can be a target for therapies in the future. Potentially, studies will combine the most effective targeted drugs with one another and/or with conventional chemotherapy options to improve patient survival.
Acknowledgments
The authors thank Sabrina Porter for her assistance in the manuscript preparation. M. Arock is supported by Fondation de France. J. Gotlib is supported by the Charles and Ann Johnson Foundation. O. Hermine is supported by Agence nationale pour la recherche (ANR) and by Fondation pour la recherche médicale (FRM). P. Valent is supported by Austrian Science Funds (FWF) Projects SFB F4611 and SFB F4704-B20. A. Reiter is supported by research grants from the ‘Deutsche José Carreras Leukämie-Stiftung e.V. (H 11/03 and R 13/05)). D. D. Metcalfe is supported by the Division of Intramural Research, NIAID/NIH.
Footnotes
- Check the online version for the most updated information on this article, online supplements, and information on authorship & disclosures: www.haematologica.org/content/101/10/1133
- Received March 31, 2016.
- Accepted May 25, 2016.
References
- Valent P, Horny HP, Escribano L. Diagnostic criteria and classification of mastocytosis: a consensus proposal. Leuk Res. 2001; 25(7):603-625. PubMedhttps://doi.org/10.1016/S0145-2126(01)00038-8Google Scholar
- Hartmann K, Metcalfe DD. Pediatric mastocytosis. Hematol Oncol Clin North Am. 2000; 14(3):625-640. PubMedhttps://doi.org/10.1016/S0889-8588(05)70299-9Google Scholar
- Wiechers T, Rabenhorst A, Schick T. Large maculopapular cutaneous lesions are associated with favorable outcome in childhood-onset mastocytosis. J Allergy Clin Immunol. 2015; 136(6):1581-1590. https://doi.org/10.1016/j.jaci.2015.05.034Google Scholar
- Robyn J, Metcalfe DD. Systemic mastocytosis. Adv Immunol. 2006; 89:169-243. PubMedhttps://doi.org/10.1016/S0065-2776(05)89005-4Google Scholar
- Escribano L, Alvarez-Twose I, Sanchez-Munoz L. 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(3):514-521. PubMedhttps://doi.org/10.1016/j.jaci.2009.05.003Google Scholar
- Horny HP AC, Metcalfe DD, Escribano L, Bennett JM, Valent P, Bain BJ. World Health Organization (WHO) Classification of Tumours Pathology & Genetics Tumours of Haematopoietic and Lymphoid Tissues. IARC Press: Lyon; 2008. Google Scholar
- Gotlib J, Pardanani A, Akin C. 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(13):2393-2401. PubMedhttps://doi.org/10.1182/blood-2012-09-458521Google Scholar
- Brockow K. Epidemiology, prognosis, and risk factors in mastocytosis. Immunol Allergy Clin North Am. 2014; 34(2):283-295. PubMedhttps://doi.org/10.1016/j.iac.2014.01.003Google Scholar
- Cohen SS, Skovbo S, Vestergaard H. Epidemiology of systemic mastocytosis in Denmark. Br J Haematol. 2014; 166(4):521-528. PubMedhttps://doi.org/10.1111/bjh.12916Google Scholar
- Valent P, Akin C, Escribano L. Standards and standardization in mastocytosis: consensus statements on diagnostics, treatment recommendations and response criteria. Eur J Clin Invest. 2007; 37(6):435-453. PubMedhttps://doi.org/10.1111/j.1365-2362.2007.01807.xGoogle Scholar
- Arber DA, Orazi A, Hasserjian R. The 2016 revision to the World Health Organization (WHO) classification of myeloid neoplasms and acute leukemia. Blood. 2016. Google Scholar
- Hein MS, Hansen L. Aggressive systemic mastocytosis: a case report and brief review of the literature. S D J Med. 2005; 58(3):95-100. PubMedGoogle Scholar
- Lim KH, Tefferi A, Lasho TL. Systemic mastocytosis in 342 consecutive adults: survival studies and prognostic factors. Blood. 2009; 113(23):5727-5736. PubMedhttps://doi.org/10.1182/blood-2009-02-205237Google Scholar
- Pardanani A, Lim KH, Lasho TL. Prognostically relevant breakdown of 123 patients with systemic mastocytosis associated with other myeloid malignancies. Blood. 2009; 114(18):3769-3772. PubMedhttps://doi.org/10.1182/blood-2009-05-220145Google Scholar
- Valent P, Sperr WR, Akin C. How I treat patients with advanced systemic mastocytosis. Blood. 2010; 116(26):5812-5817. PubMedhttps://doi.org/10.1182/blood-2010-08-292144Google Scholar
- Valent P, Akin C, Sperr WR. Aggressive systemic mastocytosis and related mast cell disorders: current treatment options and proposed response criteria. Leuk Res. 2003; 27(7):635-641. PubMedhttps://doi.org/10.1016/S0145-2126(02)00168-6Google Scholar
- Gotlib J, Pardanani A, Akin C. 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(13):2393-2401. PubMedhttps://doi.org/10.1182/blood-2012-09-458521Google Scholar
- Arock M, Sotlar K, Akin C. KIT mutation analysis in mast cell neoplasms: recommendations of the European competence network on mastocytosis. Leukemia. 2015; 29(6):1223-1232. PubMedhttps://doi.org/10.1038/leu.2015.24Google Scholar
- Ustun C, DeRemer DL, Akin C. Tyrosine kinase inhibitors in the treatment of systemic mastocytosis. Leuk Res. 2011; 35(9):1143-1152. PubMedhttps://doi.org/10.1016/j.leukres.2011.05.006Google Scholar
- Bibi S, Langenfeld F, Jeanningros S. Molecular defects in mastocytosis: KIT and beyond KIT. Immunol Allergy Clin North Am. 2014; 34(2):239-262. PubMedhttps://doi.org/10.1016/j.iac.2014.01.009Google Scholar
- Pullarkat VA, Bueso-Ramos C, Lai R. Systemic mastocytosis with associated clonal hematological non-mast-cell lineage disease: analysis of clinicopathologic features and activating c-kit mutations. Am J Hematol. 2003; 73(1):12-17. PubMedhttps://doi.org/10.1002/ajh.10322Google Scholar
- Nagai S, Ichikawa M, Takahashi T. The origin of neoplastic mast cells in systemic mastocytosis with AML1/ETO-positive acute myeloid leukemia. Exp Hematol. 2007; 35(11):1747-1752. PubMedhttps://doi.org/10.1016/j.exphem.2007.08.016Google Scholar
- Ustun C, Corless CL, Savage N. Chemotherapy and dasatinib induce long-term hematologic and molecular remission in systemic mastocytosis with acute myeloid leukemia with KIT D816V. Leuk Res. 2009; 33(5):735-741. PubMedhttps://doi.org/10.1016/j.leukres.2008.09.027Google Scholar
- Sotlar K, Colak S, Bache A. Variable presence of KITD816V in clonal haematological non-mast cell lineage diseases associated with systemic mastocytosis (SM-AHNMD). J Pathol. 2010; 220(5):586-595. PubMedhttps://doi.org/10.1002/path.2677Google Scholar
- Jawhar M, Schwaab J, Schnittger S. Molecular profiling of myeloid progenitor cells in multi-mutated advanced systemic mastocytosis identifies KIT D816V as a distinct and late event. Leukemia. 2015; 29(5):1115-1122. PubMedhttps://doi.org/10.1038/leu.2015.4Google Scholar
- Hashimoto K, Tsujimura T, Moriyama Y. Transforming and differentiation-inducing potential of constitutively activated c-kit mutant genes in the IC-2 murine interleukin-3-dependent mast cell line. Am J Pathol. 1996; 148(1):189-200. PubMedGoogle Scholar
- Tsujimura T, Hashimoto K, Kitayama H. Activating mutation in the catalytic domain of c-kit elicits hematopoietic transformation by receptor self-association not at the ligand-induced dimerization site. Blood. 1999; 93(4):1319-1329. PubMedGoogle Scholar
- Saleh R, Wedeh G, Herrmann H. A new human mast cell line expressing a functional IgE receptor converts to factor-independence and tumorigenicity by KIT D816V-transfection. Blood. 2014; 124(1):111-120. PubMedhttps://doi.org/10.1182/blood-2013-10-534685Google Scholar
- Longley BJ, Reguera MJ, Ma Y. Classes of c-KIT activating mutations: proposed mechanisms of action and implications for disease classification and therapy. Leuk Res. 2001; 25(7):571-576. PubMedhttps://doi.org/10.1016/S0145-2126(01)00028-5Google Scholar
- Furitsu T, Tsujimura T, Tono T. 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(4):1736-1744. PubMedhttps://doi.org/10.1172/JCI116761Google Scholar
- Ma YS, Zeng S, Metcalfe DD. 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 regulatory-type mutations. Blood. 2002; 99(5):1741-1744. PubMedhttps://doi.org/10.1182/blood.V99.5.1741Google Scholar
- Nakagomi N, Hirota S. Juxtamembrane-type c-kit gene mutation found in aggressive systemic mastocytosis induces imatinib-resistant constitutive KIT activation. Lab Invest. 2007; 87(4):365-371. PubMedGoogle Scholar
- Gleixner KV, Mayerhofer M, Sonneck K. Synergistic growth-inhibitory effects of two tyrosine kinase inhibitors, dasatinib and PKC412, on neoplastic mast cells expressing the D816V-mutated oncogenic variant of KIT. Blood. 2006; 108(11):159a-159a. Google Scholar
- Gleixner KV, Mayerhofer M, Sonneck K. 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(11):1451-1459. PubMedhttps://doi.org/10.3324/haematol.11339Google Scholar
- Jin B, Ding K, Pan J. Ponatinib induces apoptosis in imatinib-resistant human mast cells by dephosphorylating mutant D816V KIT and silencing beta-catenin signaling. Mol Cancer Ther. 2014; 13(5):1217-1230. PubMedhttps://doi.org/10.1158/1535-7163.MCT-13-0397Google Scholar
- Erben P, Schwaab J, Metzgeroth G. The KIT D816V expressed allele burden for diagnosis and disease monitoring of systemic mastocytosis. Ann Hematol. 2014; 93(1):81-88. PubMedhttps://doi.org/10.1007/s00277-013-1964-1Google Scholar
- Hoermann G, Gleixner KV, Dinu GE. The KIT D816V allele burden predicts survival in patients with mastocytosis and correlates with the WHO type of the disease. Allergy. 2014; 69(6):810-813. PubMedhttps://doi.org/10.1111/all.12409Google Scholar
- Lasho T, Finke C, Zblewski D. Concurrent activating KIT mutations in systemic mastocytosis. Br J Haematol. 2016; 173(1):153-156. PubMedhttps://doi.org/10.1111/bjh.13560Google Scholar
- Gotlib J, Kluin-Nelemans HC, George TI. 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;636-636. Google Scholar
- Vega-Ruiz A, Cortes JE, Sever M. Phase II study of imatinib mesylate as therapy for patients with systemic mastocytosis. Leuk Res. 2009; 33(11):1481-1484. PubMedhttps://doi.org/10.1016/j.leukres.2008.12.020Google Scholar
- de Melo Campos P, Machado-Neto JA, Scopim-Ribeiro R. Familial systemic mastocytosis with germline KIT K509I mutation is sensitive to treatment with imatinib, dasatinib and PKC412. Leuk Res. 2014; 38(10):1245-1251. PubMedhttps://doi.org/10.1016/j.leukres.2014.07.010Google Scholar
- Akin C, Fumo G, Yavuz AS. A novel form of mastocytosis associated with a transmembrane c-kit mutation and response to imatinib. Blood. 2004; 103(8):3222-3225. PubMedhttps://doi.org/10.1182/blood-2003-11-3816Google Scholar
- Alvarez-Twose I, Gonzalez P, Morgado JM. Complete response after imatinib mesylate therapy in a patient with well-differentiated systemic mastocytosis. J Clin Oncol. 2012; 30(12):e126-129. PubMedhttps://doi.org/10.1200/JCO.2011.38.9973Google Scholar
- Tefferi A, Lasho TL, Brockman SR. FIP1L1-PDGFRA and c-kit D816V mutation-based clonality studies in systemic mast cell disease associated with eosinophilia. Haematologica. 2004; 89(7):871-873. PubMedGoogle Scholar
- Klion AD, Robyn J, Akin C. Molecular remission and reversal of myelofibrosis in response to imatinib mesylate treatment in patients with the myeloproliferative variant of hypereosinophilic syndrome. Blood. 2004; 103(2):473-478. PubMedhttps://doi.org/10.1182/blood-2003-08-2798Google Scholar
- Horny HP, Sotlar K, Valent P. Mastocytosis: immunophenotypical features of the transformed mast cells are unique among hematopoietic cells. Immunol Allergy Clin North Am. 2014; 34(2):315-321. PubMedhttps://doi.org/10.1016/j.iac.2014.01.005Google Scholar
- Shah NP, Lee FY, Luo R. Dasatinib (BMS-354825) inhibits KITD816V, an imatinib-resistant activating mutation that triggers neoplastic growth in most patients with systemic mastocytosis. Blood. 2006; 108(1):286-291. PubMedhttps://doi.org/10.1182/blood-2005-10-3969Google Scholar
- Olivieri A, Manzione L. Dasatinib: a new step in molecular target therapy. Ann Oncol. 2007; 18(Suppl 6):vi42-46. PubMedhttps://doi.org/10.1093/annonc/mdm223Google Scholar
- Gleixner KV, Mayerhofer M, Cerny-Reiterer S. KIT-D816V-independent oncogenic signaling in neoplastic cells in systemic mastocytosis: role of Lyn and Btk activation and disruption by dasatinib and bosutinib. Blood. 2011; 118(7):1885-1898. PubMedhttps://doi.org/10.1182/blood-2010-06-289959Google Scholar
- Verstovsek S, Tefferi A, Cortes J. Phase II study of dasatinib in Philadelphia chromosome-negative acute and chronic myeloid diseases, including systemic mastocytosis. Clin Cancer Res. 2008; 14(12):3906-3915. PubMedhttps://doi.org/10.1158/1078-0432.CCR-08-0366Google Scholar
- Dubreuil P, Letard S, Ciufolini M. Masitinib (AB1010), a Potent and Selective Tyrosine Kinase Inhibitor Targeting KIT. PloS One. 2009; 4(9)Google Scholar
- Hahn KA, Oglivie G, Rusk T. Masitinib is Safe and Effective for the Treatment of Canine Mast Cell Tumors. J Vet Intern Med. 2008; 22(6):1301-1309. PubMedhttps://doi.org/10.1111/j.1939-1676.2008.0190.xGoogle Scholar
- Paul C, Sans B, Suarez F. Masitinib for the treatment of systemic and cutaneous mastocytosis with handicap: A phase 2a study. Am J Hematol. 2010; 85(12):921-925. PubMedhttps://doi.org/10.1002/ajh.21894Google Scholar
- Hermine O, Lortholary O, Leventhal PS. Case-control cohort study of patients’ perceptions of disability in mastocytosis. PloS One. 2008; 3(5):e2266. PubMedhttps://doi.org/10.1371/journal.pone.0002266Google Scholar
- Hochhaus A, Baccarani M, Giles FJ. Nilotinib in patients with systemic mastocytosis: analysis of the phase 2, open-label, single-arm nilotinib registration study. J Cancer Res Clin Oncol. 2015; 141(11):2047-2060. PubMedhttps://doi.org/10.1007/s00432-015-1988-0Google Scholar
- Gleixner KV, Peter B, Blatt K. Synergistic growth-inhibitory effects of ponatinib and midostaurin (PKC412) on neoplastic mast cells carrying KIT D816V. Haematologica. 2013; 98(9):1450-1457. PubMedhttps://doi.org/10.3324/haematol.2012.079202Google Scholar
- Pan J, Quintas-Cardama A, Kantarjian HM. EXEL-0862, a novel tyrosine kinase inhibitor, induces apoptosis in vitro and ex vivo in human mast cells expressing the KIT D816V mutation. Blood. 2007; 109(1):315-322. PubMedhttps://doi.org/10.1182/blood-2006-04-013805Google Scholar
- Kosmider O, Denis N, Dubreuil P, Moreau-Gachelin F. Semaxinib (SU5416) as a therapeutic agent targeting oncogenic Kit mutants resistant to imatinib mesylate. Oncogene. 2007; 26(26):3904-3908. PubMedhttps://doi.org/10.1038/sj.onc.1210159Google Scholar
- Jin Y, Ding K, Wang D, Shen M, Pan J. Novel thiazole amine class tyrosine kinase inhibitors induce apoptosis in human mast cells expressing D816V KIT mutation. Cancer Lett. 2014; 353(1):115-123. PubMedhttps://doi.org/10.1016/j.canlet.2014.07.017Google Scholar
- Evans EK, Hodous BL, Gardino A. First Selective KIT D816V Inhibitor for Patients with Systemic Mastocytosis. Blood. 2014; 124(21):3217. Google Scholar
- Traina F, Visconte V, Jankowska AM. Single nucleotide polymorphism array lesions, TET2, DNMT3A, ASXL1 and CBL mutations are present in systemic mastocytosis. PloS One. 2012; 7(8):e43090. PubMedhttps://doi.org/10.1371/journal.pone.0043090Google Scholar
- Tefferi A, Levine RL, Lim KH. Frequent TET2 mutations in systemic mastocytosis: clinical, KITD816V and FIP1L1-PDGFRA correlates. Leukemia. 2009; 23(5):900-904. PubMedhttps://doi.org/10.1038/leu.2009.37Google Scholar
- Wilson TM, Maric I, Simakova O. Clonal analysis of NRAS activating mutations in KIT-D816V systemic mastocytosis. Haematologica. 2011; 96(3):459-463. PubMedhttps://doi.org/10.3324/haematol.2010.031690Google Scholar
- Schwaab J, Schnittger S, Sotlar K. Comprehensive mutational profiling in advanced systemic mastocytosis. Blood. 2013; 122(14):2460-2466. PubMedhttps://doi.org/10.1182/blood-2013-04-496448Google Scholar
- Hanssens K, Brenet F, Agopian J. SRSF2-p95 hotspot mutation is highly associated with advanced forms of mastocytosis and mutations in epigenetic regulator genes. Haematologica. 2014; 99(5):830-835. PubMedhttps://doi.org/10.3324/haematol.2013.095133Google Scholar
- Busque L, Patel JP, Figueroa ME. Recurrent somatic TET2 mutations in normal elderly individuals with clonal hematopoiesis. Nat Genet. 2012; 44(11):1179-1181. PubMedhttps://doi.org/10.1038/ng.2413Google Scholar
- Ito S, Shen L, Dai Q. Tet Proteins Can Convert 5-Methylcytosine to 5-Formylcytosine and 5-Carboxylcytosine. Science. 2011; 333(6047):1300-1303. PubMedhttps://doi.org/10.1126/science.1210597Google Scholar
- Moran-Crusio K, Reavie L, Shih A. Tet2 Loss Leads to Increased Hematopoietic Stem Cell Self-Renewal and Myeloid Transformation. Cancer Cell. 2011; 20(1):11-24. PubMedhttps://doi.org/10.1016/j.ccr.2011.06.001Google Scholar
- Holmfeldt L, Mullighan CG. The role of TET2 in hematologic neoplasms. Cancer Cell. 2011; 20(1):1-2. PubMedhttps://doi.org/10.1016/j.ccr.2011.06.025Google Scholar
- Ko M, Huang Y, Jankowska AM. Impaired hydroxylation of 5-methylcytosine in myeloid cancers with mutant TET2. Nature. 2010; 468(7325):839-843. PubMedhttps://doi.org/10.1038/nature09586Google Scholar
- De Vita S, Schneider RK, Garcia M. Loss of function of TET2 cooperates with constitutively active KIT in murine and human models of mastocytosis. PloS One. 2014; 9(5):e96209. PubMedhttps://doi.org/10.1371/journal.pone.0096209Google Scholar
- Jawhar M, Schwaab J, Schnittger S. Molecular profiling of myeloid progenitor cells in multi-mutated advanced systemic mastocytosis identifies KIT D816V as a distinct and late event. Leukemia. 2015; 29(5):1115-1122. PubMedhttps://doi.org/10.1038/leu.2015.4Google Scholar
- Damaj G, Joris M, Chandesris O. ASXL1 but not TET2 mutations adversely impact overall survival of patients suffering systemic mastocytosis with associated clonal hematologic non-mast-cell diseases. PloS one. 2014; 9(1):e85362. PubMedhttps://doi.org/10.1371/journal.pone.0085362Google Scholar
- Will CL, Lührmann R. Spliceosome Structure and Function. Cold Spring Harb Perspect Biol. 2011; 3(7)Google Scholar
- Visconte V, Makishima H, Maciejewski JP, Tiu RV. Emerging roles of the spliceosomal machinery in myelodysplastic syndromes and other hematological disorders. Leukemia. 2012; 26(12):2447-2454. PubMedhttps://doi.org/10.1038/leu.2012.130Google Scholar
- Katoh M. Functional proteomics of the epigenetic regulators ASXL1, ASXL2 and ASXL3: a convergence of proteomics and epigenetics for translational medicine. Expert Rev Proteomics. 2015; 12(3):317-328. PubMedhttps://doi.org/10.1586/14789450.2015.1033409Google Scholar
- Jawhar M, Schwaab J, Schnittger S. Additional mutations in SRSF2, ASXL1 and/or RUNX1 identify a high risk group of patients with KIT D816V+ advanced systemic mastocytosis. Leukemia. 2016; 30(1):136-143. PubMedhttps://doi.org/10.1038/leu.2015.284Google Scholar
- Jawhar M, Schwaab J, Schnittger S. Additional mutations in SRSF2, ASXL1 and/or RUNX1 identify a high-risk group of patients with KIT D816V(+) advanced systemic mastocytosis. Leukemia. 2016; 30(1):136-143. PubMedhttps://doi.org/10.1038/leu.2015.284Google Scholar
- Soverini S, De Benedittis C, Mancini M. Genome-Wide Molecular Portrait of Aggressive Systemic Mastocytosis and Mast Cell Leukemia Depicted By Whole Exome Sequencing and Copy Number Variation Analysis. Blood. 2015; 126(23)Google Scholar
- Yavuz AS, Lipsky PE, Yavuz S, Metcalfe DD, Akin C. 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(2):661-665. PubMedhttps://doi.org/10.1182/blood-2002-01-0203Google Scholar
- Aichberger KJ, Mayerhofer M, Gleixner KV. Identification of MCL1 as a novel target in neoplastic mast cells in systemic mastocytosis: inhibition of mast cell survival by MCL1 antisense oligonucleotides and synergism with PKC412. Blood. 2007; 109(7):3031-3041. PubMedhttps://doi.org/10.1182/blood-2006-07-032714Google Scholar
- Aichberger KJ, Gleixner KV, Mirkina I. Identification of proapoptotic Bim as a tumor suppressor in neoplastic mast cells: role of KIT D816V and effects of various targeted drugs. Blood. 2009; 114(26):5342-5351. PubMedhttps://doi.org/10.1182/blood-2008-08-175190Google Scholar
- Peter B, Cerny-Reiterer S, Hadzijusufovic E. The pan-Bcl-2 blocker obatoclax promotes the expression of Puma, Noxa, and Bim mRNA and induces apoptosis in neoplastic mast cells. J Leukocyte Biol. 2014; 95(1):95-104. PubMedhttps://doi.org/10.1189/jlb.1112609Google Scholar
- Kawakami Y, Kitaura J, Satterthwaite AB. Redundant and opposing functions of two tyrosine kinases, Btk and Lyn, in mast cell activation. J Immunol. 2000; 165(3):1210-1219. PubMedhttps://doi.org/10.4049/jimmunol.165.3.1210Google Scholar
- Alvarez-Errico D, Yamashita Y, Suzuki R. Functional analysis of Lyn kinase A and B isoforms reveals redundant and distinct roles in Fc epsilon RI-dependent mast cell activation. J Immunol. 2010; 184(9):5000-5008. PubMedhttps://doi.org/10.4049/jimmunol.0904064Google Scholar
- Gleixner KV, Mayerhofer M, Hormann G. Bosutinib Blocks Lyn and Btk Activation and Synergizes with the KIT D816V-Targeting Drug Midostaurin in Inducing Apoptosis in Neoplastic Human Mast Cells. ASH Annual Meeting Abstracts. 2009; 114(22):1717. Google Scholar
- Randall N, Courville EL, Baughn L, Afrin L, Ustun C. Bosutinib, a Lyn/Btk inhibiting tyrosine kinase inhibitor, is ineffective in advanced systemic mastocytosis. Am J Hematol. 2015; 90(4):E74-E74. PubMedhttps://doi.org/10.1002/ajh.23942Google Scholar
- Kim MS, Kuehn HS, Metcalfe DD, Gilfillan AM. Activation and function of the mTORC1 pathway in mast cells. J Immunol. 2008; 180(7):4586-4595. PubMedhttps://doi.org/10.4049/jimmunol.180.7.4586Google Scholar
- Sundstrom M, Vliagoftis H, Karlberg P. Functional and phenotypic studies of two variants of a human mast cell line with a distinct set of mutations in the c-kit proto-oncogene. Immunology. 2003; 108(1):89-97. PubMedhttps://doi.org/10.1046/j.1365-2567.2003.01559.xGoogle Scholar
- Harir N, Boudot C, Friedbichler K. Oncogenic Kit controls neoplastic mast cell growth through a Stat5/PI3-kinase signaling cascade. Blood. 2008; 112(6):2463-2473. PubMedhttps://doi.org/10.1182/blood-2007-09-115477Google Scholar
- Yang Y, Letard S, Borge L. Pediatric mastocytosis-associated KIT extracellular domain mutations exhibit different functional and signaling properties compared with KIT-phosphotransferase domain mutations. Blood. 2010; 116(7):1114-1123. PubMedhttps://doi.org/10.1182/blood-2009-06-226027Google Scholar
- Smrz D, Kim MS, Zhang S. mTORC1 and mTORC2 differentially regulate homeostasis of neoplastic and non-neoplastic human mast cells. Blood. 2011; 118(26):6803-6813. PubMedhttps://doi.org/10.1182/blood-2011-06-359984Google Scholar
- Blatt K, Herrmann H, Mirkina I. The PI3-kinase/mTOR-targeting drug NVP-BEZ235 inhibits growth and IgE-dependent activation of human mast cells and basophils. PloS one. 2012; 7(1):e29925. PubMedhttps://doi.org/10.1371/journal.pone.0029925Google Scholar
- Parikh SA, Kantarjian HM, Richie MA, Cortes JE, Verstovsek S. Experience with everolimus (RAD001), an oral mammalian target of rapamycin inhibitor, in patients with systemic mastocytosis. Leuk Lymphoma. 2010; 51(2):269-274. PubMedhttps://doi.org/10.3109/10428190903486220Google Scholar
- Baumgartner C, Cerny-Reiterer S, Sonneck K. Expression of activated STAT5 in neoplastic mast cells in systemic mastocytosis: subcellular distribution and role of the transforming oncoprotein KIT D816V. Am J Pathol. 2009; 175(6):2416-2429. PubMedhttps://doi.org/10.2353/ajpath.2009.080953Google Scholar
- Lasho T, Tefferi A, Pardanani A. Inhibition of JAK-STAT signaling by TG101348: a novel mechanism for inhibition of KITD816V-dependent growth in mast cell leukemia cells. Leukemia. 2010; 24(7):1378-1380. PubMedhttps://doi.org/10.1038/leu.2010.109Google Scholar
- Vannucchi AM, Kantarjian HM, Kiladjian JJ. A pooled analysis of overall survival in COMFORT-I and COMFORT-II, 2 randomized phase III trials of ruxolitinib for the treatment of myelofibrosis. Haematologica. 2015; 100(9):1139-1145. PubMedhttps://doi.org/10.3324/haematol.2014.119545Google Scholar
- Santos FPS, Helman R, Pereira WO. Activity Of a JAK1/JAK2 Inhibitor In a Patient With KIT-Mutated Systemic Mastocytosis (SM) Associated With Myelofibrosis. Blood. 2013; 122(21):5246. Google Scholar
- Tanaka A, Konno M, Muto S. A novel NF-kappa B inhibitor, IMD-0354, suppresses neoplastic proliferation of human mast cells with constitutively activated c-kit receptors. Blood. 2005; 105(6):2324-2331. PubMedhttps://doi.org/10.1182/blood-2004-08-3247Google Scholar
- Macleod AC, Klug LR, Patterson J. Combination therapy for KIT-mutant mast cells: targeting constitutive NFAT and KIT activity. Mol Cancer Ther. 2014; 13(12):2840-2851. PubMedhttps://doi.org/10.1158/1535-7163.MCT-13-0830Google Scholar
- Belkina AC, Denis GV. BET domain co-regulators in obesity, inflammation and cancer. Nat Rev Cancer. 2012; 12(7):465-477. PubMedhttps://doi.org/10.1038/nrc3256Google Scholar
- Zuber J, Shi J, Wang E. RNAi screen identifies Brd4 as a therapeutic target in acute myeloid leukaemia. Nature. 2011; 478(7370):524-528. PubMedhttps://doi.org/10.1038/nature10334Google Scholar
- Godley LA, Le Beau MM. The histone code and treatments for acute myeloid leukemia. N Engl J Med. 2012; 366(10):960-961. PubMedhttps://doi.org/10.1056/NEJMcibr1113401Google Scholar
- Wedeh G, Cerny-Reiterer S, Eisenwort G. Identification of bromodomain-containing protein-4 as a novel marker and epigenetic target in mast cell leukemia. Leukemia. 2015; 29(11):2230-2237. PubMedhttps://doi.org/10.1038/leu.2015.138Google Scholar
- Sotlar K, Horny HP, Simonitsch I. 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(10):1319-1325. PubMedhttps://doi.org/10.1097/01.pas.0000138181.89743.7bGoogle Scholar
- Valent P, Cerny-Reiterer S, Herrmann H. Phenotypic heterogeneity, novel diagnostic markers, and target expression profiles in normal and neoplastic human mast cells. Best Pract Res Clin Haematol. 2010; 23(3):369-378. PubMedhttps://doi.org/10.1016/j.beha.2010.07.003Google Scholar
- Valent P, Sotlar K, Horny HP. Aberrant expression of CD30 in aggressive systemic mastocytosis and mast cell leukemia: a differential diagnosis to consider in aggressive hematopoietic CD30-positive neoplasms. Leuk Lymphoma. 2011; 52(5):740-744. PubMedhttps://doi.org/10.3109/10428194.2010.550072Google Scholar
- Hoermann G, Blatt K, Greiner G. CD52 is a molecular target in advanced systemic mastocytosis. FASEB J. 2014; 28(8):3540-3551. PubMedhttps://doi.org/10.1096/fj.14-250894Google Scholar
- Teodosio C, Mayado A, Sanchez-Munoz L. The immunophenotype of mast cells and its utility in the diagnostic work-up of systemic mastocytosis. J Leukoc Biol. 2015; 97(1):49-59. PubMedhttps://doi.org/10.1189/jlb.5RU0614-296RGoogle Scholar
- Quintas-Cardama A, Kantarjian H, Verstovsek S. Treatment of systemic mastocytosis with denileukin diftitox. Am J Hematol. 2007; 82(12):1124. PubMedGoogle Scholar
- Krauth MT, Bohm A, Agis H. Effects of the CD33-targeted drug gemtuzumab ozogamicin (Mylotarg) on growth and mediator secretion in human mast cells and blood basophils. Exp Hematol. 2007; 35(1):108-116. PubMedhttps://doi.org/10.1016/j.exphem.2006.09.008Google Scholar
- Sotlar K, Cerny-Reiterer S, Petat-Dutter K. Aberrant expression of CD30 in neoplastic mast cells in high-grade mastocytosis. Modern Pathol. 2011; 24(4):585-595. PubMedhttps://doi.org/10.1038/modpathol.2010.224Google Scholar
- Blatt K, Cerny-Reiterer S, Schwaab J. Identification of the Ki-1 antigen (CD30) as a novel therapeutic target in systemic mastocytosis. Blood. 2015; 126(26):2832-2841. PubMedhttps://doi.org/10.1182/blood-2015-03-637728Google Scholar
- Florian S, Sonneck K, Hauswirth AW. Detection of molecular targets on the surface of CD34+/CD38– stem cells in various myeloid malignancies. Leuk Lymphoma. 2006; 47(2):207-222. PubMedhttps://doi.org/10.1080/10428190500272507Google Scholar
- Eisenwort G, Peter B, Blatt K. Identification of a Neoplastic Stem Cell in Human Mast Cell Leukemia. Blood. 2014; 124(21):817. Google Scholar
- Casassus P, Caillat-Vigneron N, Martin A. Treatment of adult systemic mastocytosis with interferon-alpha: results of a multicentre phase II trial on 20 patients. Br J Haematol. 2002; 119(4):1090-1097. PubMedhttps://doi.org/10.1046/j.1365-2141.2002.03944.xGoogle Scholar
- Hauswirth AW, Simonitsch-Klupp I, Uffmann M. 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(3):249-257. PubMedhttps://doi.org/10.1016/S0145-2126(03)00259-5Google Scholar
- Kluin-Nelemans HC, Jansen JH, Breukelman H. Response to interferon alfa-2b in a patient with systemic mastocytosis. N Engl J Med. 1992; 326(9):619-623. PubMedhttps://doi.org/10.1056/NEJM199202273260907Google Scholar
- Kluin-Nelemans HC, Oldhoff JM, Van Doormaal JJ. Cladribine therapy for systemic mastocytosis. Blood. 2003; 102(13):4270-4276. PubMedhttps://doi.org/10.1182/blood-2003-05-1699Google Scholar
- Lim KH, Pardanani A, Butterfield JH, Li CY, Tefferi A. 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(12):790-794. PubMedhttps://doi.org/10.1002/ajh.21561Google Scholar
- Bohm A, Sonneck K, Gleixner KV. 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(9):744-755. PubMedhttps://doi.org/10.1016/j.exphem.2010.05.006Google Scholar
- Hermine O, Hirsh I, Damaj G. Long Term Efficacy and Safety of Cladribine In Adult Systemic mastocytosis: a French Multicenter Study of 44 Patients. ASH Annual Meeting Abstracts. 2010; 116(21):1982. Google Scholar
- Barete S, Lortholary O, Damaj G. Long-term efficacy and safety of cladribine (2-CdA) in adult patients with mastocytosis. Blood. 2015; 126(8):1009-1016. PubMedhttps://doi.org/10.1182/blood-2014-12-614743Google Scholar
- Valent P, Blatt K, Eisenwort G. FLAG-induced remission in a patient with acute mast cell leukemia (MCL) exhibiting t(7;10)(q22;q26) and KIT D816H. Leuk Res Rep. 2014; 3(1):8-13. PubMedGoogle Scholar
- Ustun C, Reiter A, Scott BL. Hematopoietic stem-cell transplantation for advanced systemic mastocytosis. J Clin Oncol. 2014; 32(29):3264-3274. PubMedhttps://doi.org/10.1200/JCO.2014.55.2018Google Scholar
- Sperr WR, Drach J, Hauswirth AW. Myelomastocytic leukemia: evidence for the origin of mast cells from the leukemic clone and eradication by allogeneic stem cell transplantation. Clin Cancer Res. 2005; 11(19 Pt 1):6787-6792. PubMedhttps://doi.org/10.1158/1078-0432.CCR-05-1064Google Scholar
- Ustun C, Courville E. Resolution of osteosclerosis after alloHCT in systemic mastocytosis. Blood. 2016; 127(14):1836-1836. PubMedhttps://doi.org/10.1182/blood-2016-01-690669Google Scholar
- Ustun C, Gotlib J, Popat U. Consensus opinion on allogeneic hematopoietic cell transplantation in advanced systemic mastocytosis. Biol Blood Marrow Transplant. 2016. Google Scholar
- Ustun C, Smith A, Cayci Z. Allogeneic hematopoietic cell transplantation in systemic mastocytosis: Is there a high risk for veno-occlusive disease. Eur J Haematol. 2015. Google Scholar
- Akin C, Scott LM, Kocabas CN. Demonstration of an aberrant mast-cell population with clonal markers in a subset of patients with “idiopathic” anaphylaxis. Blood. 2007; 110(7):2331-2333. PubMedhttps://doi.org/10.1182/blood-2006-06-028100Google Scholar
- Akin C, Metcalfe DD. Systemic mastocytosis. Annu Rev Med. 2004; 55:419-432. PubMedhttps://doi.org/10.1146/annurev.med.55.091902.103822Google Scholar
- Theoharides TC, Valent P, Akin C. Mast Cells, Mastocytosis, and Related Disorders. N Engl J Med. 2015; 373(19):1885-1886. Google Scholar