AUY922, a Heat Shock Protein 90 (Hsp90) Inhibitor, Demonstrates Activity in Patients with Myeloproliferative Neoplasms (MPNs)

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 4075-4075 ◽  
Author(s):  
Gabriella Hobbs ◽  
Rivka Litvin ◽  
Jihae Ahn ◽  
Anna Sophia Mckenney ◽  
Michael J. Mauro ◽  
...  

Abstract The introduction of JAK inhibitors into clinical practice has produced significant clinical benefits for patients with MPNs, including reduction of splenomegaly and improvement in symptom burden. However, JAK inhibitors have demonstrated limited clinical ability to alter the natural history and biology of disease (such as reversal of fibrosis) in MPNs, and have produced only modest reductions in JAK2 V617F allele burden. This has prompted the evaluation of other therapeutic strategies in MPN patients. Heat Shock Protein 90 (Hsp90) inhibitors have recently emerged as promising potential treatment for MPN patients. Hsp90 physically interacts with JAK2, and inhibition of Hsp90 induces degradation of JAK2. Hsp90 is also a client chaperone for a variety of other proteins involved in pathways known to be critical for cancer cell differentiation, proliferation and survival. Preclinical studies have demonstrated that Hsp90 inhibitor therapy results in dose-dependent degradation of JAK2 and inhibition of downstream signaling pathways including STAT5, STAT3 and MAPK, as well as inhibition of growth of cells expressing mutant JAK2. In vivo treatment with Hsp90 inhibitors in MPN retroviral murine models led to normalized of peripheral blood counts, reduction in organomegaly, and improvement in overall survival. Based on these promising preclinical data, we conducted an open label phase II trial to assess the efficacy and confirm the safety of a novel Hsp90 inhibitor, AUY922 (Novartis) in patients with primary myelofibrosis (PMF), post-polycythemia vera and post-essential thrombocythemia myelofibrosis (IPSS-2 or higher) and in patients with polycythemia vera or essential thrombocythemia, who were refractory to, intolerant of, or ineligible for conventional therapy. The primary objective of the study was to determine the efficacy of AUY922 in this patient population. Secondary objectives included confirmation of safety and tolerability along with exploration of how treatment modified the biology of the disease. From 2012-2014, 6 patients were treated, 4 female, median age 55 (53-72), with MPNs; 4 with primary-MF, 1 with PV and 1 with ET, 4 patients were JAK2 V617F positive. Prior treatments included hydroxyurea in 5, anagrelide in 3, interferon in 1, azacitadine and pamidronate in 1. Median length of treatment was 39.5 months (1-145). 3 patients with MF experienced stable disease (including two patients with blast-phase disease), and one experienced an anemia response as measured by the European LeukemiaNet (ELN) response criteria. The patient with ET experienced stable disease as measured by the Revised International Working Group for Myeloproliferative Neoplasms Research and Treatment (IWG-MRT). Notable (greater than grade 2) adverse events included diarrhea in 5 patients, night blindness in 4, alkaline phosphatase elevation in 2, nausea in 2, emesis in 2, blurred vision in 1 and CPK elevation in 1 patient Serious Adverse Events (SAEs) included nausea, vomiting, diarrhea and altered mental status. As well, three patients experienced gastrointestinal bleeding as an SAE (grade 2 and 3). Two of these events were associated with an ileocecal ulcer. Two events were deemed to be possibly related to exposure to the study drug. The trial was terminated for this reason. Evaluation of the impact on JAK-STAT signaling is underway with evidence of reduction in total JAK2 protein following infusion (figure 1). Further evaluation of JAK-STAT signaling, impact on cytokine production, and JAK2 allele burden is under evaluation and results will be presented. In summary, treatment with AUY922 in patients with has demonstrated response including stable disease (including in blast-phase myelofibrosis patients) as well as anemia response and clinical improvement in one myelofibrosis patient. These data indicate that Hsp90 inhibition has clinical activity in MPN patients, and that further clinical therapeutic efforts targeting Hsp90 warrant investigation in this patient population. Figure 1. Responses to treatment with AUY922 per Revised International Working Group for Myeloproliferative Neoplasms Research and Treatment (IWG-MRT) and European LeukemiaNet criteria (ELN). Figure 1. Responses to treatment with AUY922 per Revised International Working Group for Myeloproliferative Neoplasms Research and Treatment (IWG-MRT) and European LeukemiaNet criteria (ELN). Figure 2. Western Blot analysis of total JAK2 from peripheral blood of patient at baseline, cycle 1 day 2, and end of treatment. Figure 2. Western Blot analysis of total JAK2 from peripheral blood of patient at baseline, cycle 1 day 2, and end of treatment. Disclosures Mauro: Pfizer: Consultancy; Novartis Pharmaceutical Corporation: Consultancy, Research Funding; Bristol-Myers Squibb: Consultancy; Ariad: Consultancy. Levine:Foundation Medicine: Consultancy; CTI BioPharma: Membership on an entity's Board of Directors or advisory committees; Loxo Oncology: Membership on an entity's Board of Directors or advisory committees.

2012 ◽  
Vol 209 (2) ◽  
pp. 259-273 ◽  
Author(s):  
Oliver Weigert ◽  
Andrew A. Lane ◽  
Liat Bird ◽  
Nadja Kopp ◽  
Bjoern Chapuy ◽  
...  

Enzymatic inhibitors of Janus kinase 2 (JAK2) are in clinical development for the treatment of myeloproliferative neoplasms (MPNs), B cell acute lymphoblastic leukemia (B-ALL) with rearrangements of the cytokine receptor subunit cytokine receptor–like factor 2 (CRLF2), and other tumors with constitutive JAK2 signaling. In this study, we identify G935R, Y931C, and E864K mutations within the JAK2 kinase domain that confer resistance across a panel of JAK inhibitors, whether present in cis with JAK2 V617F (observed in MPNs) or JAK2 R683G (observed in B-ALL). G935R, Y931C, and E864K do not reduce the sensitivity of JAK2-dependent cells to inhibitors of heat shock protein 90 (HSP90), which promote the degradation of both wild-type and mutant JAK2. HSP90 inhibitors were 100–1,000-fold more potent against CRLF2-rearranged B-ALL cells, which correlated with JAK2 degradation and more extensive blockade of JAK2/STAT5, MAP kinase, and AKT signaling. In addition, the HSP90 inhibitor AUY922 prolonged survival of mice xenografted with primary human CRLF2-rearranged B-ALL further than an enzymatic JAK2 inhibitor. Thus, HSP90 is a promising therapeutic target in JAK2-driven cancers, including those with genetic resistance to JAK enzymatic inhibitors.


Haematologica ◽  
2016 ◽  
Vol 102 (1) ◽  
pp. 85-93 ◽  
Author(s):  
Holly L. Geyer ◽  
Heidi Kosiorek ◽  
Amylou C. Dueck ◽  
Robyn Scherber ◽  
Stefanie Slot ◽  
...  

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 2971-2971
Author(s):  
Taylor B. Collins ◽  
Stephanie A. Luff ◽  
Luis F.Z. Batista ◽  
Christopher M. Sturgeon ◽  
Stephen T. Oh

JAK2 V617F is the most frequent mutation found in myeloproliferative neoplasms (MPNs), with 50-60% of myelofibrosis (MF) patients harboring this mutation. Mutations in ASXL1 often co-occur with JAK2 V617F and are associated with decreased survival and increased risk of transformation to secondary acute myeloid leukemia. How mutant ASXL1 contributes to the MPN disease phenotype and confers poor prognosis is not fully understood. Controversy remains as to whether ASXL1 mutations found in patients confer a loss of function, gain of function, and/or dominant negative phenotype. Additionally, Asxl1 mutation knock in mouse models present with a relatively modest phenotype, following a very long latency period. A human model system has the potential to provide a useful tool to understand how these mutations affect ASXL1 function. This project therefore seeks to uncover how expression of mutant JAK2 in conjunction with mutant ASXL1 influences hematopoietic output and proliferation, utilizing human pluripotent stem cells. Two complementary approaches have been utilized to study how ASXL1 mutations influence MPN pathogenicity. CD34+ cells from an MF patient harboring JAK2 V617F and an ASXL1 mutation (P920Tfs*4) were FACS-sorted and reprogrammed to generate distinct iPSC clones. Because the majority of the patient-derived iPSC clones were double mutant, CRISPR gene editing was utilized to revert the JAK2 and ASXL1 mutations to generate all four possible genotypes. In parallel, two different ASXL1 mutations (the most common G646Wfs*12 mutation as well as the P920Tfs*4 mutation) were introduced into H1 human embryonic stem cells, each in isolation and in combination with JAK2 V617F, resulting in the generation of single and double mutant lines. These genetically engineered pluripotent stem cell lines were then differentiated specifically into definitive hematopoietic lineages, through a stepwise program regulated by Wnt signaling. This allowed us to analyze the role of ASXL1 and JAK2 mutations during the development of the various lineages of the hematopoietic system known to be dysregulated in MPNs, such as the myeloid and erythroid lineages. Following hematopoietic differentiation, colony forming assays were performed. The JAK2 mutant line produced four-fold more colonies than WT line, with a significant bias towards the generation of erythroid colonies. In contrast, the ASXL1 mutant line produced substantially fewer colonies than the WT line, with the majority of these colonies resembling myeloid colonies. The double mutant line (ASXL1/JAK2) generated more colonies than the WT and ASXL1 mutant lines, but fewer colonies as compared to the JAK2 mutant line. Of note, the JAK2 V617F mutation generated much larger erythroid colonies compared to the WT H1 control line or the double mutant line. Our in vitro differentiation experiments also allowed us to sort equal numbers of CD45+CD34+ cells from the different genotypes, and plate them in methylcellulose. These experiments allow the ability to assess how mutations impact differentiation on a per cell basis. Similar trends were observed (i.e. more erythroid colonies with JAK2 and fewer and mostly myeloid colonies with ASXL1) with this approach, suggesting that the phenotypes observed can be attributed at least in part to progenitor cell output on a per cell basis and not solely reflecting the total number of progenitors generated through the pluripotent stem cell differentiation protocol. In summary, with his human pluripotent stem cell model system, we have observed that JAK2 V617F induced an increase in total colony production with a marked expansion of the erythroid lineage, while mutant ASXL1 impaired colony production overall with substantial myeloid skewing. Cells expressing both mutations presented with an intermediate phenotype. Ongoing efforts include gene expression analysis to understand how JAK2 and ASXL1 mutations direct these observed functional differences. Disclosures Oh: Incyte: Membership on an entity's Board of Directors or advisory committees; Blueprint Medicines: Membership on an entity's Board of Directors or advisory committees; Novartis: Consultancy.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 5383-5383
Author(s):  
Murtadha Al-Khabori ◽  
Shoaib Al-Zadjali ◽  
Iman Al Noumani ◽  
Khalil Al Farsi ◽  
Salam Al-Kindi ◽  
...  

Objectives: Mutations in additional sex combs-like transcriptional regulator 1 (ASXL1) have been previously described in myeloid neoplasms (21% in non-Myeloproliferative [MPN; Tefferi A, Leukemia, 2010) and have been associated with a more aggressive disease [Rocquain J et al, BMC Cacer, 2010]. They can also be found in patients with JAK2 positive MPN [Abdel-Wahab O et al, Cancer Research, 2010). Disruption of ASXL1 gene leads to MPN phenotype in zebrafish model (Gjini E, Dis Model Mech, 2019). The co-expression and the prognostic significance of ASXL1 in patients with JAK2 positive MPN are not yet fully defined. We therefore planned to define the prognostic impact of ASXL1 mutations on the Overall Survival (OS) of patients with JAK2 positive MPN. Methods: We included patients with JAK2 V617F positive MPN diagnosed according to the World Health Organization 2016 criteria and treated at the three largest hematology centers in Oman. The entire coding region of ASXL1 gene was sequenced using Next Generation Sequencing (NGS; Ion PGM Sequencer; Thermo Fisher Scientific®). The library was constructed and the templates were prepared using the PGM tool and the variants were annotated using the ClinVar database and the prediction from the Scale-Invariant Feature Transform (SIFT) and or Polymorphism Phenotyping (Polyphen) algorithms. The NGS analysis was done on the frozen diagnostic bone marrow samples. The survival probability was estimated using Kaplan-Meier estimator and Cox regression was used to assess the impact of predictors on the OS outcome. An alpha threshold of 0.05 was used. The R program (version 3.1.2) was used for all statistical analyses. Results: A total of 58 patients with JAK2 V617F positive MPN were included. All of these patients were found to have mutated ASXL1 using the NGS (ASXL1 p.Leu815Pro was found in all patients). The median age of this cohort was 62 years (InterQuartile Range [IQR]: 44 - 70) and female to male ratio was 25:33. The median hemoglobin, hematocrit, white blood cell count and platelet count was 14.7 g/dL, 58%, 11.5 x109/L and 518 x109/L respectively. Out of the 58 patients included, 28 had polycythemia vera, 20 had essential thrombocythemia, 8 had myelofibrosis and 2 had MPN-Unclassified. The median time from diagnosis to last follow up or death was 13 months (IQR: 3-39). During this period, 5 patients died. The probability of OS at 3 years was 88%. The median OS was not reached. In the univariable analysis, age was a statistically significant predictor of OS (p = 0.0355) but not gender (p = 0.434) and MPN subtype (p = 0.7). In the multivariable analysis model of the previous three factors, age remained statistically significant (Hazard ratio = 1.13, p = 0.041). Conclusions: ASXL1 is mutated in high proportion of patients with JAK2 positive MPN. Despite the negative impact of ASXL1 in patients with non-MPN myeloid neoplasms, the patients with combined positivity of JAK2 and ASXL1 in this study had a very good OS probability. Age was a predictor of OS in the univariable and multivariable models. We recommend the development and the assessment of ASXL1 inhibitors as therapeutic strategies in patients with MPN. Disclosures Al-Khabori: Novartis: Honoraria, Membership on an entity's Board of Directors or advisory committees; NovoNardisk: Membership on an entity's Board of Directors or advisory committees; Servier: Membership on an entity's Board of Directors or advisory committees; Shire (Takeda): Membership on an entity's Board of Directors or advisory committees; SOBI: Honoraria; AstraZeneca: Honoraria; Abbvie: Membership on an entity's Board of Directors or advisory committees; Roche: Membership on an entity's Board of Directors or advisory committees; Amgen: Membership on an entity's Board of Directors or advisory committees.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3862-3862
Author(s):  
Irum Khan ◽  
Zan Huang ◽  
Qiang Jeremy Wen ◽  
Priya Koppikar ◽  
Ross L Levine ◽  
...  

Abstract Abstract 3862 Somatic mutations in JAK2 and MPL are associated with the BCR-ABL negative myeloproliferative neoplasms (MPNs) including polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF). While oral JAK2 inhibitors improve peripheral blood counts and splenomegaly, these drugs show varying effects on JAK2 mutant allele burden and do not eliminate the malignant clone in humans or in animal models of MPN. Although much of the research to date has focused on JAK/STAT signaling, other pathways downstream of the class I cytokine receptors, including PI3K/AKT and ERK are also activated in MPNs. Our hypothesis is that persistent activation of these signaling pathways contributes to the progression of myeloproliferative neoplasms (MPNs). Multiple studies have shown that overexpression of activated JAK2 and MPL mutants (e.g. JAK2 V617F or MPL W515L) in primary murine bone marrow cells causes robust AKT, STAT3, and STAT5, JNK, ERK phosphorylation. To determine the extent to which these signaling pathways are involved in the disease, we cultured JAK2 V617F and MPL W515L expressing clones of the G1ME and 6133 megakaryocytic cell lines with a panel of small molecule kinase inhibitors. We discovered that inhibition of the PI3K/AKT and MAPK/JNK signaling pathways with triciribine and SP600125 respectively, potently suppressed growth of these cells by inducing G1 arrest and apoptosis. In contrast, inhibitors of Ras, ERK, Src family tyrosine kinase, and PKC failed to significantly inhibit proliferation of the JAK2 or MPL mutant expressing cells. Murine bone marrow cells transduced with MPL W515L show a dramatic expansion of megakaryocytes (CFU-MKs) and this expansion was abrogated in the presence of PI3K/AKT inhibitors suggesting a requirement for this pathway in aberrant megakaryocyte expansion. Since AKT inhibition showed the strongest effect, we assayed the activity of MK-2206, a potent and selective allosteric AKT inhibitor, on multiple models of MPNs. We discovered that MK-2206 induced proliferative arrest and apoptosis accompanied by suppression of PI3K/AKT signaling in G1ME and 6133 cells expressing MPLW515L. MK-2206 also potently inhibited liquid culture growth and colony formation of PMF patient CD34+ cells in vitro. Finally, in preliminary murine transplants experiments with MPLW515L expressing bone marrow progenitors, treatment with MK-2206 led to significant reductions in peripheral blood leukocytosis and extramedullary hematopoiesis. Together, these findings demonstrate that the PI3K/AKT axis represents a rational target for therapy in human myeloproliferative neoplasms. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 4995-4995
Author(s):  
Pu Chen ◽  
Boting Wu ◽  
Xia Shao ◽  
Chanjuan Liu ◽  
Zhenglin Yu ◽  
...  

Abstract Background Myeloproliferative neoplasms (MPNs) are characterized by marker somatic mutations involving JAK2, MPL, and CALR, which lead to constitutive activation of tyrosine kinase signaling cascades, subsequently dysregulated proliferative of myeloid linages, and eventually myelofibrosis or leukemic transformation. Recently, it has been argued that proinflammatory processes are crucial to the pathogenesis and progression of MPNs. A number of proinflammatory cytokines including lipocalin, IL-1, IL-2R, IL-6, IL-8, IL-12, IL-15, and IP-10 have been found elevated in the peripheral blood (PB) of MPN patients. However, there has been limited data on the levels of proinflammatory cytokines in the bone marrow (BM) of MPN patients. The present study determined and compared 40 proinflammatory cytokine levels in the PB and BM plasma of MPN patients with unequivocal molecular background, thus intending to illustrate the proinflammatory features of BM microenvironment as well as to evaluate the credibility of PB cytokine profiles. Methods Newly diagnosed MPN patients (n=12, 8 had JAK2 V617F mutation, 4 had CALR mutations) were included in the present study. PB samples were taken within 48 hours of BM samples. Paired PB and BM plasma cytokine profiles were measured as well as 10 health control PB plasma samples in a single procedure by Quantibody Human Inflammatory Array 3 (RayBiotech, Norcross, GA) which permitted detection of 40 inflammation-associated cytokines. Results Among 12 MPN patients, 8 had JAK2 V617 mutation (6 males, median age 61.5 years), and 4 had CALR mutations (3 males, median age 53.5 years). Positive linear correlations between PB and BM levels were found in 12 proinflammatory cytokines including BLC (r=0.613, p=0.034), I-309 (r=0.872, p<0.001), IL-1α (r=0.666, p=0.018), IL-1β (r=0.929, p<0.001), IL-12p40 (r=0.642, p=0.024), IL-15 (r=0.608, p=0.036), M-CSF (r=0.906, p<0.001), MIG (r=0.596, p=0.041), MIP-1α (r=0.787, p=0.002), MIP-1δ (r=0.648, p=0.023), sTNFRI (r=0.827, p=0.001), and sTNFRII (r=0.644, p=0.024). Compared to health controls, BM levels of G-CSF, IL-2, IL-4, IL-6R, IL-7, IL-8, IL-10, IL-13, MIP-1β, PDGF-BB, RANTES, and TIMP-1 were markedly elevated (all p<0.01), among which IL-2, IL-4, and IL-7 levels were even higher in patients with CALR mutations than those with classic JAK2 V617F mutation (all p<0.05). Conclusions Optimal linear correlation could only be found in limited species of proinflammatory cytokines, especially I-309, IL-1β, M-CSF, and sTNFRI, between PB and BM plasma of MPN patients. Therefore, caution should be recommended during attempts to illustrate the status of inflammation in MPN patients by circulating cytokine markers. A stronger inflammatory component might exist in MPN patients with CALR mutations than those with classic JAK2 V617F mutation. Disclosures No relevant conflicts of interest to declare.


2021 ◽  
Vol 156 (Supplement_1) ◽  
pp. S136-S136
Author(s):  
T Lynn ◽  
A Campbell ◽  
Y Ding

Abstract Introduction/Objective In patients with suspicion of Myeloproliferative neoplasm (MPN) and negative for BCR-ABL1, NCCN guideline currently recommends two molecular workup pathways in peripheral blood: 1) a multi-step reflex mutation testing algorithm including JAK2 V617F, CALR, MPL, JAK2 exon12 or 2) a multigene Next Generation Sequencing (NGS) panel that includes at least JAK2, CALR and MPL genes. Here we report the clinical utilization and impact of a NGS based MPN diagnosis assay. Methods/Case Report Total of 690 consecutive cases at Geisinger between 2019 and 2021 were included in this study. Patient’s CBC showed chronic cytosis in either single or multi-lineage myelopoiesis and was clinically suspicious for MPNs. For BCR-ABL1 negative cases, NGS based MPN diagnostic assay was performed, which include the four disease defining genes recommended by NCCN guideline: JAK2, CALR, MPL, CSF3R as well as three additional genes NRAS, PPM1D and TP53. Variants are classified in to four tiers based on their level of clinical significance. Results (if a Case Study enter NA) Among all cases tested, 25 out of 690 cases (3.6%) were positive for BCR-ABL1 transcript. 20.9% (139 out of 665 BCR-ABL1 negative cases) had at least one variant detected, which included 73 variants in Tier I category (11.0%), 6 variants in Tier II (0.9%), 57 variants in Tier III (8.6%) and 3 variants in Tier IV(0.5%). Among all disease defining mutations, JAK2 V617F was the most commonly detected mutation (59 cases and 8.8%), followed by CALR indel mutations (13 cases and 2.0%). In addition, double variants were detected in total 7 cases (1.0%). Conclusion Comparing to the conventional multi-step sequential workup for MPN diagnosis, the multi-gene NGS panel provides a cost-effective and time- saving solution. When detected concurrently with the disease defining mutations, NRAS, PPM1D and TP53 could provide not only additional prognostic information but also may suggest pending progression in myeloproliferative neoplasms.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1777-1777
Author(s):  
Massimo Breccia ◽  
Roberto Latagliata ◽  
Laura Cannella ◽  
Ida Carmosino ◽  
Enrico Gottardi ◽  
...  

Abstract Abstract 1777 Poster Board I-803 Introduction The proteasome, a multicatalytic enzyme complex, plays an important role in the activation of nuclear factor kappa B (NF-kB), through degradation of its suppressor IkB. NF-kB can intervene in oncogenesis with its capacity to regulate the expression of genes that modulate apoptosis, cell survival as well as proliferation, inflammation, angiogenesis and tumour metastasis. Bortezomib (Velcade, formerly PS-341) is a proteasome inhibitor with documented antitumor activity in multiple myeloma and other lymphoid malignancies and has also been investigated in myeloid malignancies, such as acute leukemias (AL) and myelodysplastic syndromes (MDS), indicating some therapeutic activities. We performed a Phase II study (EudraCT number 2004-002935-23) to investigate the efficacy and safety of bortezomib in patients with MDS. Patients and methods: Ten patients were enrolled in the study: there were 6 females and 4 males, median age was 64 years (range 44-70), median duration of MDS phase was 12 months. All patients but one received prior erythropoietin therapy without efficacy. 8/10 patients required transfusional therapy at the time of enrolment (median 4 units per month). Results According to WHO classification, 5 patients were refractory cytopenia with multilineage dysplasia (RCMD), 1 patient was pure refractory anemia (RA), 1 patient was RCMD with ringed sideroblasts (RCMD-RS), 3 patients were refractory anemia with excess of blasts type 1 (RAEB-1). According to IPSS stratification, 5 patients were classified as low risk, 3 patients as intermediate-1 and 2 patients as intermediate-2 risk. Six patients presented cytogenetic aberrations at baseline (isolated del(5q), 2 patients; del(5q) plus t(3;12), 1 patient; del(5q) and monosomy 7, 1 patient, isolated trisomy 8, 1 patient; trisomy 8 and trisomy 11, 1 patient). WT1 gene expression was tested in all cases: 9/10 patients displayed elevated WT1 expression level in bone marrow (median 219 copies, range 20-2682) and 7/10 also in peripheral blood (median 31 copies, range 11-2247). Bortezomib was administered at the dose of 1.3 mg/mq with a 1, 4, 8, 11 day schedule, every 28 days, for a maximum of 8 cycles. Four patients discontinued therapy after 1, 3, 3 and 4 cycles respectively, due to vascular disease not related to the drug in the first two cases and to disease progression in the other two (both patients with intermediate-2 risk), whereas 6 patients performed all planned eight cycles. As to safety, grade 3/4 neutropenia was recorded in 4 patients and grade 3/4 thrombocytopenia in 6 patients. Non-hematological side effects were recorded in 7 patients: diarrhea in 1 patient, fever in 3 patients, skin rash in 2 patients and pneumonia in 1 patient during neutropenic phase. As to response, 3 patients (50% of evaluable cases) achieved a minor erythroid response (according to IWG criteria) and 3 patients had a stable disease. We did not reveal modifications as to bone marrow blasts and cytogenetic changes with respect to pre-treatment findings. As to outcome, 7/10 patients are still alive at a median follow-up of 24 months. WT1 decrease was recorded in the 3 patients with minor erythroid response, both in bone marrow (from median 109 copies to median 14 copies at the end of treatment) and in peripheral blood (from median 70 copies to median 9 copies at the end of treatment), and in 1 patient with stable disease. In the remaining patients with stable disease, median bone marrow WT1 copies increased from 659 copies at baseline to 890 copies at the end of treatment, and to median 736 copies at baseline to 788 copies at the end of treatment in peripheral blood. Conclusions Present results indicate that bortezomib as single agent in MDS, seems to have some efficacy in terms of hematological improvement and to also affect the WT1 gene expression. Further studies on larger series of MDS patients are needed to confirm our observation. The combined use of this drug with other agents might potentiate its therapeutic activity. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 2106-2106
Author(s):  
Brady Stein ◽  
Jerry L. Spivak ◽  
Alison R Moliterno

Introduction: Host modifiers contribute to variation in disease pathogenesis and clinical features in the myeloproliferative neoplasms (MPN), essentialthrombocytosis (ET), polycythemia vera (PV), and myelofibrosis (MF).Gender modifies the JAK2 V617F allele burden (women had lower levels and lower annual increases), influences PV gene expression (men had twice as many differentially-regulated genes as women) and influences thrombotic events (a predominance of abdominal vein thrombosis in women). The MPN symptom burden also appears to differ by gender. In this analysis, we describe gender differences in disease presentation and outcome in relation tomutational status. Methods: We analyzed a prospective cohort of 612 PV, ET and MF patients enrolled between 2005 and 2013 to study genomic modifiers of the MPN and disease outcomes. We stratified disease evolution and survival by both gender and mutational status. Results: In this cohort, 26% were newly diagnosed, and 61% were female. The current median follow-up is 8 years (> 4800 patient-years) and 110 have died. Prevalence of Molecular lesions JAK2 V617F was identified in 468/612 MPN patients (76%; 61% female), and comprised the majority of ET (62%; 70% female), PV (99.5%; 59% female), MF (62%; 39% female) and post MPN AML (80.7%; 38% female) patients. CALR mutations were identified in 81/612 MPN patients (13%; 58% female), representing 23% of all ET (59% female), and 21 % of all MF (52% female) and 3.8% of post MPN AML. MPL mutations were identified in 9 MPN patients (1.5%): 7 females with ET, and 2 males with MF. Disease presentation Among JAK2 V617F positive patients, more females presented with ET compared to males (43% vs 29%; p=0.002) and more males presented with MF (21% vs 9%; p=0.001). Gender differences in presentation were less apparent in those with CALR -mutated ET (59% women) and MF (53% women). Disease transformation By gender, among 197 women with an original diagnosis of ET, 62% retained their phenotype, whereas 20%, 17%, and 1.5% evolved to PV, MF, and AML, respectively. Among men with ET, 63% retained their phenotype, while 16%, 20%, and 1.1% evolved to PV, MF, and AML. Among MF patients, regardless of mutational status, 1 (3%) women and 5 (10%) men evolved to AML (p=0.39). With regard to transformations by gender, rates of MF (16% in both) and AML (2.6% and 5.5%p=0.083) were similar between women and men. Among 123 JAK2V617F- positive ET women, during the follow-up period, 66 (54%) retained an ET phenotype, while 32%, 14%, and 0.8% evolved to PV, MF, and AML, respectively. Among 53 JAK2V617F-positive ET men, 54% retained an ET phenotype; 25%, 17%, and 4% evolved to PV, MF, and AML, respectively. Among 141 JAK2V617F-positive PV females, 109 (77%) retained PV, 26 (18%) evolved to MF, and 6 (4%) evolved to AML (1 via MF, 5 PV to AML); among 100 JAK2V617F-positive males, 73% retained a PV phenotype, 20% evolved to MF, and 7% to AML (3 PV to AML, 4 via MF phase). Among JAK2V617F -positive patients with an original MF diagnosis, 1/20 (5%) and 3/31 (10%) women and men evolved to AML (p=NS); 22% vs. 18% of JAK2V617F -positive men versus women evolved to MF or AML (p=0.28). No CALR -mutated ET patient evolved to PV. Of 38 CALR- ET women, 27 (71%) retained their phenotype, 10 evolved to MF (26%), and 1 to AML (3%). Among the 26 CALR -mutated ET men, 81% retained their phenotype, while 19% evolved to MF (no AML transformations). Survival The proportion of deaths differed by gender; of the 612 patients, despite the predominance of females in the cohort, at the time of last follow-up, there were 110 deaths (18%): 50/375 women died, compared to 60/237 men (13% vs. 25%; p value=0.0002). A larger proportion of male deaths were due to MF and MF to AML, but this was not statistically different (male deaths in MF/AML phase: 54/60; (90%); women 38/50 (75%); p=0.0692)). Conclusion: We identified a gender influence on disease distribution at presentation, particularly in the JAK2V617F-positive subset with females presenting more commonly with ET and males more commonly with MF. Despite the predominance of females in this MPN cohort, the distribution of females at presentation and evolution into more indolent phenotypes compared to males and a trend toward lower rates of AML evolution may have accounted for longer disease duration and fewer deaths in females compared to males. This trend in MPN evolution complements a theme of gender differences in symptom burden, clinical consequences, and genomic changes. Disclosures Stein: Incyte Corporation: Consultancy, Membership on an entity's Board of Directors or advisory committees. Spivak:Incyte: Membership on an entity's Board of Directors or advisory committees. Moliterno:Incyte: Membership on an entity's Board of Directors or advisory committees.


2010 ◽  
Vol 01 (04) ◽  
pp. 161-162
Author(s):  
Günter Springer

Patienten mit Myelodysplastischem Syndrom (MDS) und Krankheitsstabilisierung (SD) als erstes Ansprechen haben unter kontinuierlicher Behandlung mit Azacitidin (Vida-za®) gute Aussichten, noch ein Ansprechen gemäß Kriterien der International Working Group (IWG) und dadurch einen Überlebensvorteil zu erreichen. Zu diesem Ergebnis kam eine auf dem Kongress der American Society of Oncology (ASCO) vorgestellte Analyse der Zulassungsstudie AZA-001.


Sign in / Sign up

Export Citation Format

Share Document