Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Mohamad Jawhar is active.

Publication


Featured researches published by Mohamad Jawhar.


Blood | 2013

Comprehensive mutational profiling in advanced systemic mastocytosis

Juliana Schwaab; Susanne Schnittger; Karl Sotlar; Christoph Walz; Alice Fabarius; Markus Pfirrmann; Alexander Kohlmann; Vera Grossmann; Manja Meggendorfer; Hans-Peter Horny; Peter Valent; Mohamad Jawhar; Martina Teichmann; Georgia Metzgeroth; Philipp Erben; Thomas Ernst; Andreas Hochhaus; Torsten Haferlach; Wolf-Karsten Hofmann; Nicholas C.P. Cross; Andreas Reiter

To explore mechanisms contributing to the clinical heterogeneity of systemic mastocytosis (SM) and to suboptimal responses to diverse therapies, we analyzed 39 KIT D816V mutated patients with indolent SM (n = 10), smoldering SM (n = 2), SM with associated clonal hematologic nonmast cell lineage disorder (SM-AHNMD, n = 5), and aggressive SM (n = 15) or mast cell leukemia (n = 7) with (n = 18) or without (n = 4) AHNMD for additional molecular aberrations. We applied next-generation sequencing to investigate ASXL1, CBL, IDH1/2, JAK2, KRAS, MLL-PTD, NPM1, NRAS, TP53, SRSF2, SF3B1, SETBP1, U2AF1 at mutational hotspot regions, and analyzed complete coding regions of EZH2, ETV6, RUNX1, and TET2. We identified additional molecular aberrations in 24/27 (89%) patients with advanced SM (SM-AHNMD, 5/5; aggressive SM/mast cell leukemia, 19/22) whereas only 3/12 (25%) indolent SM/smoldering SM patients carried one additional mutation each (U2AF1, SETBP1, CBL) (P < .001). Most frequently affected genes were TET2, SRSF2, ASXL1, CBL, and RUNX1. In advanced SM, 21/27 patients (78%) carried ≥3 mutations, and 11/27 patients (41%) exhibited ≥5 mutations. Overall survival was significantly shorter in patients with additional aberrations as compared to those with KIT D816V only (P = .019). We conclude that biology and prognosis in SM are related to the pattern of mutated genes that are acquired during disease evolution.


Leukemia | 2016

Additional mutations in SRSF2, ASXL1 and/or RUNX1 identify a high-risk group of patients with KIT D816V(+) advanced systemic mastocytosis

Mohamad Jawhar; Juliana Schwaab; S Schnittger; Manja Meggendorfer; Markus Pfirrmann; Karl Sotlar; H.-P. Horny; Georgia Metzgeroth; Sebastian Kluger; Nicole Naumann; Claudia Haferlach; T Haferlach; Peter Valent; Wolf-Karsten Hofmann; A. Fabarius; Nicholas C.P. Cross; Andreas Reiter

Most patients with KIT D816V+ advanced systemic mastocytosis (SM) are characterized by somatic mutations in additional genes. We sought to clarify the prognostic impact of such mutations. Genotype and clinical characteristics of 70 multi-mutated KIT D816V+ advanced SM patients were included in univariate and multivariate analyses. 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). In univariate analysis, overall survival (OS) was adversely influenced by mutations in SRSF2 (P<0.0001), ASXL1 (P=0.002) and RUNX1 (P=0.03), but was not influenced by mutations in TET2 or JAK2. In multivariate analysis, SRSF2 and ASXL1 remained the most predictive adverse indicators concerning OS. Furthermore, we 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). In conclusion, the presence and number of mutated genes within the S/A/R panel are adversely associated with advanced disease and poor survival in KIT D816V+ SM. On the basis of these findings, inclusion of molecular markers should be considered in upcoming prognostic scoring systems for patients with SM.


Leukemia | 2015

Molecular profiling of myeloid progenitor cells in multi-mutated advanced systemic mastocytosis identifies KIT D816V as a distinct and late event

Mohamad Jawhar; Juliana Schwaab; S Schnittger; Karl Sotlar; H.-P. Horny; Georgia Metzgeroth; Nadine Müller; Sven Schneider; Nicole Naumann; Christoph Walz; T Haferlach; Peter Valent; Wolf-Karsten Hofmann; Nicholas C.P. Cross; A. Fabarius; Andreas Reiter

To explore the molecular profile and its prognostic implication in systemic mastocytosis (SM), we analyzed the mutation status of granulocyte–macrophage colony-forming progenitor cells (CFU-GM) in patients with KIT D816V+ indolent SM (ISM, n=4), smoldering SM (SSM, n=2), aggressive SM (ASM, n=1), SM with associated clonal hematologic non-mast cell lineage disorder (SM-AHNMD, n=5) and ASM-AHNMD (n=7). All patients with (A)SM-AHNMD (n=12) carried 1–4 (median 3) additional mutations in 11 genes tested, most frequently TET2, SRSF2, ASXL1, CBL and EZH2. In multi-mutated (A)SM-AHNMD, KIT D816V+ single-cell-derived CFU-GM colonies were identified in 8/12 patients (median 60%, range 0–95). Additional mutations were identified in CFU-GM colonies in all patients, and logical hierarchy analysis indicated that mutations in TET2, SRSF2 and ASXL1 preceded KIT D816V. In ISM/SSM, no additional mutations were detected and CFU-GM colonies were exclusively KIT D816V−. These data indicate that (a) (A)SM-AHNMD is a multi-mutated neoplasm, (b) mutations in TET2, SRSF2 or ASXL1 precede KIT D816V in ASM-AHNMD, (c) KIT D816V is thus a phenotype modifier toward SM and (d) KIT D816V or other mutations are rare in CFU-GM colonies of ISM/SSM patients, which might explain at least in part their better prognosis.


American Journal of Hematology | 2015

KIT D816V and JAK2 V617F mutations are seen recurrently in hypereosinophilia of unknown significance

Juliana Schwaab; Roland Umbach; Georgia Metzgeroth; Nicole Naumann; Mohamad Jawhar; Karl Sotlar; Hans-Peter Horny; Timo Gaiser; Wolf-Karsten Hofmann; Susanne Schnittger; Nicholas C.P. Cross; Alice Fabarius; Andreas Reiter

Myeloproliferative neoplasms with eosinophilia are commonly characterized by a normal karyotype and remain poorly defined at the molecular level. We therefore investigated 426 samples from patients with hypereosinophilia of unknown significance initially referred for screening of the FIP1L1‐PDGFRA (FP) fusion gene also for KIT D816V and JAK2 V617F mutations. Overall, 86 (20%) patients tested positive: FP+ in 55 (12%), KIT D816V+ in 14 (3%), and JAK2 V617F+ in 17 (4%) patients, respectively. To gain better insight into clinical characteristics, we compared these cases with 31 additional and well‐characterized KIT D816V+ eosinophilia‐associated systemic mastocytosis (SM‐eo) patients enrolled within the “German Registry on Disorders of Eosinophils and Mast cells.” Significant differences included younger age, male predominance, and higher eosinophil counts for FP+ cases while abdominal lymphadenopathy, ascites, and serum tryptase levels >100 μg/l were characteristic for those with KIT D816V. Leukocytes, hemoglobin, and splenomegaly did not differ significantly. A median of three additional mutations, most frequently TET2 and SRSF2, were identified in 12/13 KIT D816V+ SM‐eo patients with available material indicating a more complex molecular pathogenesis. Median survival was not reached for FP+ cases but was only 26 and 41 months for KIT D816V+ SM and JAK2 V617F+ MPN‐eo, respectively. Eosinophilia of ≥2 × 109/l was identified as discriminator for inferior survival in KIT D816V+ and/or JAK2 V617F+ patients (median survival 20 months vs. not reached, P = 0.002). Thus, there is a clear prognostic and therapeutic rationale for detection of KIT D816V and JAK2 V617F in the diagnostic work up of eosinophilia. Am. J. Hematol. 90:774–777, 2015.


Haematologica | 2017

The clinical and molecular diversity of mast cell leukemia with or without associated hematologic neoplasm.

Mohamad Jawhar; Juliana Schwaab; Manja Meggendorfer; Nicole Naumann; Hans-Peter Horny; Karl Sotlar; Torsten Haferlach; Karla Schmitt; Alice Fabarius; Peter Valent; Wolf-Karsten Hofmann; Nicholas C.P. Cross; Georgia Metzgeroth; Andreas Reiter

Mast cell leukemia is a rare variant of advanced systemic mastocytosis characterized by at least 20% of mast cells in a bone marrow smear. We evaluated clinical and molecular characteristics of 28 patients with (n=20, 71%) or without an associated hematologic neoplasm. De novo mast cell leukemia was diagnosed in 16 of 28 (57%) patients and secondary mast cell leukemia evolving from other advanced systemic mastocytosis subtypes in 12 of 28 (43%) patients, of which 7 patients progressed while on cytoreductive treatment. Median bone marrow mast cell infiltration was 65% and median serum tryptase was 520 μg/L. C-findings were identified in 26 of 28 (93%) patients. Mutations in KIT (D816V, n=19; D816H/Y, n=5; F522C, n=1) were detected in 25 of 28 (89%) patients and prognostically relevant additional mutations in SRSF2, ASXL1 or RUNX1 (S/A/Rpos) in 13 of 25 (52%) patients. Overall response rate in 18 treatment-naïve patients was 5 of 12 (42%) on midostaurin and 1 of 6 (17%) on cladribine, and after switch 1 of 4 (25%) on midostaurin and 0 of 3 on cladribine, respectively. S/A/Rpos adversely affected response to treatment and progression to secondary mast cell leukemia (n=6) or acute myeloid leukemia (n=3) while on treatment (P<0.05). The median overall survival from mast cell leukemia diagnosis was 17 months as compared to 44 months in a control group of 124 patients with advanced systemic mastocytosis but without mast cell leukemia (P=0.03). In multivariate analyses, S/A/Rpos remained the only independent poor prognostic variable predicting overall survival (P=0.007). In conclusion, the molecular signature should be determined in all patients with mast cell leukemia because of its significant clinical and prognostic relevance.


Blood | 2017

Response and progression on midostaurin in advanced systemic mastocytosis: KIT D816V and other molecular markers.

Mohamad Jawhar; Juliana Schwaab; Nicole Naumann; Hans-Peter Horny; Karl Sotlar; Torsten Haferlach; Georgia Metzgeroth; Alice Fabarius; Peter Valent; Wolf-Karsten Hofmann; Nicholas C.P. Cross; Manja Meggendorfer; Andreas Reiter

In advanced systemic mastocytosis (advSM), disease evolution is often triggered by KIT mutations (D816V in >80% of cases) and by additional mutations (eg, in SRSF2, ASXL1, and/or RUNX1 [S/A/Rpos in >60% of cases]). In a recently reported phase 2 study, midostaurin, a multikinase/KIT inhibitor, demonstrated an overall response rate (ORR) of 60% in advSM but biomarkers predictive of response are lacking. We evaluated the impact of molecular markers at baseline and during follow-up in 38 midostaurin-treated advSM patients. The median overall survival (OS) was 30 months (95% confidence interval, 6-54) from start of midostaurin. ORR and OS were significantly different between S/A/Rneg (n = 12) and S/A/Rpos (n = 23) patients (ORR: 75% vs 39%, P = .04; OS: P = .01, HR 4.5 [1.3-16.2]). Depending on the relative reduction of the KIT D816V expressed allele burden (EAB) at month 6, patients were classified as KIT responders (≥25%, n = 17) or KIT nonresponders (<25%, n = 11). In univariate analyses at month 6, reduction of KIT D816V EAB ≥25%, tryptase ≥50%, and alkaline phosphatase ≥50% were significantly associated with improved OS. In multivariate analysis, only KIT D816V EAB reduction ≥25% remained an independent on-treatment marker for improved OS (P = .004, HR 6.8 [1.8-25.3]). Serial next-generation sequencing analysis of 28 genes in 16 patients revealed acquisition of additional mutations or increasing variant allele frequency in K/NRAS, RUNX1, IDH2, or NPM1 associated with progression in 7 patients. In midostaurin-treated advSM patients, the complexity and dynamics of mutational profiles significantly affect response, progression, and prognosis.


Genes, Chromosomes and Cancer | 2015

Fusion of PDGFRB to MPRIP, CPSF6, and GOLGB1 in three patients with eosinophilia-associated myeloproliferative neoplasms

Nicole Naumann; Juliana Schwaab; Georgia Metzgeroth; Mohamad Jawhar; Claudia Haferlach; Gudrun Göhring; Brigitte Schlegelberger; Christian Dietz; Susanne Schnittger; Sina Lotfi; Michael Gärtner; Tu-Anh Dang; Wolf-Karsten Hofmann; Nicholas C.P. Cross; Andreas Reiter; Alice Fabarius

In eosinophilia‐associated myeloproliferative neoplasms (MPN‐eo), constitutive activation of protein tyrosine kinases (TK) as consequence of translocations, inversions, or insertions and creation of TK fusion genes is recurrently observed. The most commonly involved TK and their potential TK inhibitors include PDGFRA at 4q12 or PDGFRB at 5q33 (imatinib), FGFR1 at 8p11 (ponatinib), and JAK2 at 9p24 (ruxolitinib). We here report the identification of three new PDGFRB fusion genes in three male MPN‐eo patients: MPRIP‐PDGFRB in a case with t(5;17)(q33;p11), CPSF6‐PDGFRB in a case with t(5;12)(q33;q15), and GOLGB1‐PDGFRB in a case with t(3;5)(q13;q33). The fusion proteins identified by 5′‐rapid amplification of cDNA ends polymerase chain reaction (PCR) or DNA‐based long distance inverse PCR are predicted to contain the TK domain of PDGFRB. The partner genes contain domains like coiled‐coil structures, which are likely to cause dimerization and activation of the TK. In all patients, imatinib induced rapid and durable complete remissions.


Leukemia | 2017

Cytogenetically cryptic ZMYM2-FLT3 and DIAPH1-PDGFRB gene fusions in myeloid neoplasms with eosinophilia.

Mohamad Jawhar; Nicole Naumann; Marcin Knut; Joannah Score; M Ghazzawi; B Schneider; K-A Kreuzer; Michael Hallek; Hans G. Drexler; J. Chacko; Louise Wallis; A. Fabarius; Georgia Metzgeroth; W-K Hofmann; Andrew Chase; William Tapper; Andreas Reiter; Nicholas C.P. Cross

Cytogenetically cryptic ZMYM2-FLT3 and DIAPH1-PDGFRB gene fusions in myeloid neoplasms with eosinophilia


Leukemia | 2018

Drug-induced inhibition of phosphorylation of STAT5 overrides drug resistance in neoplastic mast cells

Barbara Peter; S Bibi; Gregor Eisenwort; B Wingelhofer; Daniela Berger; Gabriele Stefanzl; Katharina Blatt; Harald Herrmann; Emir Hadzijusufovic; Gregor Hoermann; Thomas Hoffmann; Juliana Schwaab; Mohamad Jawhar; M Willmann; Wolfgang R. Sperr; Johannes Zuber; Karl Sotlar; H-P Horny; Richard Moriggl; Andreas Reiter; Michel Arock; Peter Valent

Systemic mastocytosis (SM) is a mast cell (MC) neoplasm with complex pathology and a variable clinical course. In aggressive SM (ASM) and MC leukemia (MCL), responses to conventional drugs are poor and the prognosis is dismal. R763 is a multi-kinase inhibitor that blocks the activity of Aurora-kinase-A/B, ABL1, AKT and FLT3. We examined the effects of R763 on proliferation and survival of neoplastic MC. R763 produced dose-dependent inhibition of proliferation in the human MC lines HMC-1.1 (IC50 5–50 nM), HMC-1.2 (IC50 1–10 nM), ROSAKIT WT (IC50 1–10 nM), ROSAKIT D816V (IC50 50–500 nM) and MCPV-1.1 (IC50 100–1000 nM). Moreover, R763 induced growth inhibition in primary neoplastic MC in patients with ASM and MCL. Growth-inhibitory effects of R763 were accompanied by signs of apoptosis and a G2/M cell cycle arrest. R763 also inhibited phosphorylation of KIT, BTK, AKT and STAT5 in neoplastic MC. The most sensitive target appeared to be STAT5. In fact, tyrosine phosphorylation of STAT5 was inhibited by R763 at 10 nM. At this low concentration, R763 produced synergistic growth-inhibitory effects on neoplastic MC when combined with midostaurin or dasatinib. Together, R763 is a novel promising multi-kinase inhibitor that blocks STAT5 activation and thereby overrides drug-resistance in neoplastic MC.


Genes, Chromosomes and Cancer | 2018

Incidence and prognostic impact of cytogenetic aberrations in patients with systemic mastocytosis

Nicole Naumann; Mohamad Jawhar; Juliana Schwaab; Sebastian Kluger; Johannes Lübke; Georgia Metzgeroth; Henning D. Popp; Nada Khaled; Hans-Peter Horny; Karl Sotlar; Peter Valent; Claudia Haferlach; Gudrun Göhring; Brigitte Schlegelberger; Manja Meggendorfer; Wolf-Karsten Hofmann; Nicholas C.P. Cross; Andreas Reiter; Alice Fabarius

The clinical behavior of systemic mastocytosis (SM) is strongly associated with activating mutations in KIT (D816V in >80% of cases), with the severity of the phenotype influenced by additional somatic mutations, for example, in SRSF2, ASXL1, or RUNX1. Complex molecular profiles are frequently associated with the presence of an associated hematologic neoplasm (AHN) and an unfavorable clinical outcome. However, little is known about the incidence and prognostic impact of cytogenetic aberrations. We analyzed cytogenetic and molecular characteristics of 109 patients (KIT D816V+, n = 102, 94%) with indolent (ISM, n = 26) and advanced SM (n = 83) with (n = 73, 88%) or without AHN. An aberrant karyotype was identified in SM‐AHN (16/73, 22%) patients only. In patients with an aberrant karyotype, additional somatic mutations were identified in 12/16 (75%) patients. Seven of 10 (70%) patients with a poor‐risk karyotype, for example, monosomy 7 or complex karyotype, and 1/6 (17%) patients with a good‐risk karyotype progressed to secondary acute myeloid leukemia (n = 7) or mast cell leukemia (n = 1) within a median of 40 months (range 2‐190, P = .04). In advanced SM, the median overall survival (OS) of poor‐risk karyotype patients was significantly shorter than in good‐risk/normal karyotype patients (4 vs 39 months; hazard ratio 11.7, 95% CI 5.0‐27.3; P < .0001). Additionally, the shortened OS in patients with poor‐risk karyotype was independent from the mutation status. In summary, a poor‐risk karyotype is an independent prognostic variable in advanced SM. Cytogenetic and molecular analyses should be routinely performed in all patients with advanced SM ± AHN because these investigations greatly support prognostication and treatment decisions.

Collaboration


Dive into the Mohamad Jawhar's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Peter Valent

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge