61 results on '"Kiladjian JJ"'
Search Results
2. Prediction of thrombosis in post-polycythemia vera and post-essential thrombocythemia myelofibrosis: a study on 1258 patients.
- Author
-
Mora B, Guglielmelli P, Kuykendall A, Rumi E, Maffioli M, Palandri F, De Stefano V, Caramella M, Salmoiraghi S, Kiladjian JJ, Gotlib J, Iurlo A, Cervantes F, Ruggeri M, Silver RT, Albano F, Benevolo G, Ross DM, Della Porta MG, Devos T, Rotunno G, Komrokji RS, Casetti IC, Merli M, Brociner M, Caramazza D, Auteri G, Barbui T, Cattaneo D, Bertù L, Arcaini L, Vannucchi AM, and Passamonti F
- Subjects
- Humans, Hydroxyurea therapeutic use, Janus Kinase Inhibitors, Polycythemia Vera complications, Polycythemia Vera therapy, Primary Myelofibrosis etiology, Primary Myelofibrosis therapy, Thrombocythemia, Essential complications, Thrombosis etiology
- Abstract
Patients with Philadelphia-negative myeloproliferative neoplasms are at high risk of thrombotic events (TEs). Predisposing factors have been identified in essential thrombocythemia (ET), polycythemia vera (PV) and primary myelofibrosis (primary MF, PMF), while yet not recognized in post PV/ET-MF (known as secondary MF, SMF). Within the 1258 SMF of the MYSEC (MYelofibrosis SECondary to PV and ET) dataset, 135 (10.7%) developed a TE at a median follow-up of 3.5 years (range, 1-21.4), with an incidence of 2.3% patients per year. Venous events accounted for two-thirds of the total. Cox multivariable analysis, supported by Fine-Gray models with death as competitive risk, showed that being on cytoreductive therapy at time of SMF evolution is associated with an absolute risk reduction of thrombosis equal to 3.3% within 3 years. Considering individually cytoreductive therapies, univariate regression model found that both conventional cytoreduction, mainly hydroxyurea, (HR 0.41, 95% CI: 0.26-0.65, p = 0.0001) and JAK inhibitors, mostly ruxolitinib, (HR 0.50, 95% CI: 0.24-1.02, p = 0.05) were associated with fewer thrombosis. Our study informs treating physicians of a non-low incidence of TEs in post PV/ET-MF and of the potential protective role of cytoreductive therapy in terms of thrombotic events., (© 2022. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2022
- Full Text
- View/download PDF
3. Real-world study of children and young adults with myeloproliferative neoplasms: identifying risks and unmet needs.
- Author
-
Sobas M, Kiladjian JJ, Beauverd Y, Curto-Garcia N, Sadjadian P, Shih LY, Devos T, Krochmalczyk D, Galli S, Bieniaszewska M, Seferynska I, McMullin MF, Armatys A, Spalek A, Waclaw J, Zdrenghea M, Legros L, Girodon F, Lewandowski K, Angona Figueras A, Samuelsson J, Abuin Blanco A, Cony-Makhoul P, Collins A, James C, Kusec R, Lauermannova M, Noya MS, Skowronek M, Szukalski L, Szmigielska-Kaplon A, Wondergem M, Dudchenko I, Gora Tybor J, Laribi K, Kulikowska de Nalecz A, Demory JL, Le Du K, Zweegman S, Besses Raebel C, Skoda R, Giraudier S, Griesshammer M, Harrison CN, and Ianotto JC
- Subjects
- Adult, Child, Humans, Prospective Studies, Young Adult, Myeloproliferative Disorders complications, Myeloproliferative Disorders diagnosis, Myeloproliferative Disorders epidemiology, Polycythemia Vera complications, Primary Myelofibrosis genetics, Thrombocythemia, Essential, Thrombosis etiology
- Abstract
Myeloproliferative neoplasms (MPNs) are uncommon in children/young adults. Here, we present data on unselected patients diagnosed before 25 years of age included from 38 centers in 15 countries. Sequential patients were included. We identified 444 patients, with median follow-up 9.7 years (0-47.8). Forty-nine (11.1%) had a history of thrombosis at diagnosis, 49 new thrombotic events were recorded (1.16% patient per year [pt/y]), perihepatic vein thromboses were most frequent (47.6% venous events), and logistic regression identified JAK2V617F mutation (P = .016) and hyperviscosity symptoms (visual disturbances, dizziness, vertigo, headache) as risk factors (P = .040). New hemorrhagic events occurred in 44 patients (9.9%, 1.04% pt/y). Disease transformation occurred in 48 patients (10.9%, 1.13% pt/y), usually to myelofibrosis (7.5%) with splenomegaly as a novel risk factor for transformation in essential thrombocythemia (ET) (P= .000) in logistical regression. Eight deaths (1.8%) were recorded, 3 after allogeneic stem cell transplantation. Concerning conventional risk scores: International Prognostic Score for Essential Thrombocythemia-Thrombosis and new International Prognostic Score for Essential Thrombocythemia-Thrombosis differentiated ET patients in terms of thrombotic risk. Both scores identified high-risk patients with the same median thrombosis-free survival of 28.5 years. No contemporary scores were able to predict survival for young ET or polycythemia vera patients. Our data represents the largest real-world study of MPN patients age < 25 years at diagnosis. Rates of thrombotic events and transformation were higher than expected compared with the previous literature. Our study provides new and reliable information as a basis for prospective studies, trials, and development of harmonized international guidelines for the specific management of young patients with MPN., (© 2022 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
4. An inherited gain-of-function risk allele in EPOR predisposes to familial JAK2 V617F myeloproliferative neoplasms.
- Author
-
Rabadan Moraes G, Pasquier F, Marzac C, Deconinck E, Damanti CC, Leroy G, El-Khoury M, El Nemer W, Kiladjian JJ, Raslova H, Najman A, Vainchenker W, Marty C, Bellanné-Chantelot C, and Plo I
- Subjects
- Alleles, Gain of Function Mutation, Humans, Janus Kinase 2 genetics, Janus Kinase 2 metabolism, Mutation, Myeloproliferative Disorders diagnosis, Myeloproliferative Disorders genetics, Polycythemia Vera genetics, Receptors, Erythropoietin genetics, Thrombocythemia, Essential genetics
- Abstract
Myeloproliferative neoplasms (MPN) are mainly sporadic but inherited variants have been associated with higher risk development. Here, we identified an EPOR variant (EPOR
P488S ) in a large family diagnosed with JAK2V617F -positive polycythaemia vera (PV) or essential thrombocytosis (ET). We investigated its functional impact on JAK2V617F clonal amplification in patients and found that the variant allele fraction (VAF) was low in PV progenitors but increase strongly in mature cells. Moreover, we observed that EPORP488S alone induced a constitutive phosphorylation of STAT5 in cell lines or primary cells. Overall, this study points for searching inherited-risk alleles affecting the JAK2/STAT pathway in MPN., (© 2022 British Society for Haematology and John Wiley & Sons Ltd.)- Published
- 2022
- Full Text
- View/download PDF
5. A randomized phase 3 trial of interferon-α vs hydroxyurea in polycythemia vera and essential thrombocythemia.
- Author
-
Mascarenhas J, Kosiorek HE, Prchal JT, Rambaldi A, Berenzon D, Yacoub A, Harrison CN, McMullin MF, Vannucchi AM, Ewing J, O'Connell CL, Kiladjian JJ, Mead AJ, Winton EF, Leibowitz DS, De Stefano V, Arcasoy MO, Kessler CM, Catchatourian R, Rondelli D, Silver RT, Bacigalupo A, Nagler A, Kremyanskaya M, Levine MF, Arango Ossa JE, McGovern E, Sandy L, Salama ME, Najfeld V, Tripodi J, Farnoud N, Penson AV, Weinberg RS, Price L, Goldberg JD, Barbui T, Marchioli R, Tognoni G, Rampal RK, Mesa RA, Dueck AC, and Hoffman R
- Subjects
- Disease Progression, Humans, Hydroxyurea adverse effects, Interferon-alpha adverse effects, Polycythemia Vera drug therapy, Polycythemia Vera genetics, Thrombocythemia, Essential drug therapy, Thrombocythemia, Essential genetics, Thrombosis chemically induced, Thrombosis prevention & control
- Abstract
The goal of therapy for patients with essential thrombocythemia (ET) and polycythemia vera (PV) is to reduce thrombotic events by normalizing blood counts. Hydroxyurea (HU) and interferon-α (IFN-α) are the most frequently used cytoreductive options for patients with ET and PV at high risk for vascular complications. Myeloproliferative Disorders Research Consortium 112 was an investigator-initiated, phase 3 trial comparing HU to pegylated IFN-α (PEG) in treatment-naïve, high-risk patients with ET/PV. The primary endpoint was complete response (CR) rate at 12 months. A total of 168 patients were treated for a median of 81.0 weeks. CR for HU was 37% and 35% for PEG (P = .80) at 12 months. At 24 to 36 months, CR was 20% to 17% for HU and 29% to 33% for PEG. PEG led to a greater reduction in JAK2V617F at 24 months, but histopathologic responses were more frequent with HU. Thrombotic events and disease progression were infrequent in both arms, whereas grade 3/4 adverse events were more frequent with PEG (46% vs 28%). At 12 months of treatment, there was no significant difference in CR rates between HU and PEG. This study indicates that PEG and HU are both effective treatments for PV and ET. With longer treatment, PEG was more effective in normalizing blood counts and reducing driver mutation burden, whereas HU produced more histopathologic responses. Despite these differences, both agents did not differ in limiting thrombotic events and disease progression in high-risk patients with ET/PV. This trial was registered at www.clinicaltrials.gov as #NCT01259856., (© 2022 by The American Society of Hematology.)
- Published
- 2022
- Full Text
- View/download PDF
6. Long-term outcomes of polycythemia vera patients treated with ropeginterferon Alfa-2b.
- Author
-
Kiladjian JJ, Klade C, Georgiev P, Krochmalczyk D, Gercheva-Kyuchukova L, Egyed M, Dulicek P, Illes A, Pylypenko H, Sivcheva L, Mayer J, Yablokova V, Krejcy K, Empson V, Hasselbalch HC, Kralovics R, and Gisslinger H
- Subjects
- Humans, Polycythemia Vera drug therapy
- Published
- 2022
- Full Text
- View/download PDF
7. Appropriate management of polycythaemia vera with cytoreductive drug therapy: European LeukemiaNet 2021 recommendations.
- Author
-
Marchetti M, Vannucchi AM, Griesshammer M, Harrison C, Koschmieder S, Gisslinger H, Álvarez-Larrán A, De Stefano V, Guglielmelli P, Palandri F, Passamonti F, Barosi G, Silver RT, Hehlmann R, Kiladjian JJ, and Barbui T
- Subjects
- Cytoreduction Surgical Procedures, Humans, Hydroxyurea therapeutic use, Quality of Life, Splenomegaly drug therapy, Polycythemia Vera drug therapy
- Abstract
Polycythaemia vera is associated with a reduced quality of life, a high rate of vascular events, and an intrinsic risk of disease evolution. The results of several randomised trials for the treatment of this disorder are now available, and both a new ropegylated formulation of interferon alfa-2b (ropeginterferon alfa-2b; 2018) and ruxolitinib (2015) have been approved in Europe. European LeukemiaNet (ELN) investigators have therefore deemed it appropriate to provide recommendations for the use of these drugs in clinical practice. An expert panel of 14 senior haematologists from ELN centres that had actively participated in previous ELN projects or relevant randomised trials, chaired by a member of the ELN Steering Committee, developed a list of clinical questions, and a methodologist established three patient, intervention, comparator, outcome (PICO) questions and systematically reviewed the evidence. Recommendations were approved by six Delphi consensus rounds and two virtual meetings (on Jan 26, 2021, and June 24, 2021). The expert panel recommended that patients with polycythaemia vera who are younger than 60 years and have not had previous thrombotic events should start cytoreductive drug therapy if at least one of the following criteria are fulfilled: strictly defined intolerance to phlebotomy, symptomatic progressive splenomegaly, persistent leukocytosis (>15 × 10
9 white blood cells per L), progressive leukocytosis (at least 100% increase if baseline count is <10 × 109 cells per L or at least 50% increase if baseline count is >10 × 109 cells per L), extreme thrombocytosis (>1500 × 109 platelets per L), inadequate haematocrit control requiring phlebotomies, persistently high cardiovascular risk, and persistently high symptom burden. Recombinant interferon alfa, either in the form of ropeginterferon alfa-2b or pegylated interferon alfa-2a, is the recommended cytoreductive treatment for these patients. The expert panel suggested that either interferon alfa or ruxolitinib should be considered for patients who are being treated with hydroxyurea but require a therapy change., Competing Interests: Declaration of interests MM has received consulting fees from Gilead Sciences, speaker fees from Amgen, support for attending meetings from Takeda, and is a member of the guideline committee of the Italian Society of Hematology. AMV has received speaker fees from Novartis, AOP Health, Incyte, AbbVie, GlaxoSmithKline (GSK), and Bristol Myers Squibb (BMS); and has participated on the advisory boards of Novartis, Incyte, AOP Orphan Pharmaceuticals, AbbVie, GSK, BMS, and Roche. MG has received consulting and speaker fees, support for attending meetings, and participated on advisory boards for AOP Health, Novartis, Celgene, Amgen, AstraZeneca, CTI BioPharma, Shire, Pfizer, Roche, Janssen Pharmaceuticals, and Gilead Sciences. CH has received grants or contracts with payment to her institution from Novartis, Celgene, and Constellation Pharmaceuticals; consulting fees from Keros Therapeutics, Galecto Biotech, and Roche; speaker fees from Novartis, Celgene, CTI BioPharma, AbbVie, Gilead Sciences, Janssen Pharmaceuticals, Promedior, and Geron Corporation; support for attending meetings from Novartis and Celgene, and has participated on advisory boards for Roche, CTI Biopharma, Geron Corporation, Promedior, AbbVie, AOP Orphan Pharmaceuticals, and Galecto. SK has received research grants from AOP Health, Novartis, Janssen Pharmaceuticals, and Imago BioScience; consulting fees from Pfizer, CTI BioPharma, Sanofi, Novartis, BMS/Celgene, Incyte/ARIAD, Roche, AOP Orphan Pharmaceuticals, and Janssen Pharmaceuticals; speaker fees from Pfizer, CTI BioPharma, Sanofi, Novartis, BMS/Celgene, Incyte/ARIAD, Roche, AOP Orphan Pharmaceuticals, Janssen Pharmaceuticals, Kartos Therapeutics, and Imago BioScience; support for attending meetings from Alexion Pharmaceuticals, Novartis, BMS/Celgene, Incyte/ARIAD, AOP Orphan Pharmaceuticals, CTI BioPharma, Pfizer, Sanofi, Janssen Pharmaceuticals, Geron Corporation, Kartos Therapeutics, Sierra Oncology, Imago BioScience; has participated on advisory boards for Pfizer, CTI BioPharma, Sanofi, Novartis, BMS/Celgene, Incyte/ARIAD, Roche, AOP Orphan Pharmaceuticals, Kartos Therapeutics, and Imago BioScience; has patents planned, issued, or pending from Rheinisch-Westfalische Technische Hochschule Aachen; and has a leadership role in Deutsche Gesellschaft fur Hamatologie und Medizinische Onkologie. HG has received research grants and support for attending meetings from AOP Orphan Pharmaceuticals and Novartis; consulting fees from AOP Orphan Pharmaceuticals, Novartis, and BMS/Celgene; and speaker fees from AOP Orphan Pharmaceuticals, Novartis, BMS/Celgene, and Janssen. AÁ-L has participated on advisory boards and has received speaker fees from Novartis, AOP Orphan Pharmaceuticals, and Celgene; and has received support for attending meetings from Novartis and Pfizer. VDS has received speaker fees from AbbVie and Novartis, and participated on advisory boards of AOP Health and Novartis. PG has received speaker fees from AbbVie and Novartis, and support for attending meetings from Sanofi. FPal has received speaker fees from Novartis and Incyte; consulting fees from AOP Health, CTI BioPharma, Novartis, and Celgene/BMS; and support for attending meetings from Celgene/BMS. FPas has received consulting fees from AbbVie, Novartis, BMS, and Amomed Pharma; and speaker fees from AbbVie, Janssen Pharmaceuticals, Novartis, and BMS. RTS has received research grants from the Cancer Research & Treatment Fund and Johns Family Fund, received speaker fees and payment for expert testimony from PharmaEssentia, participated on an advisory board for PharmaEssentia, and has a leadership role (Vice-President and Medical Director) at the Cancer Research & Treatment Fund. J-JK received consulting fees from AbbVie and Novartis; speaker fees from AOP Orphan Pharmaceuticals, Novartis, and BMS; and has participated on advisory boards for Incyte. TB has received research grants and speaker fees from AOP Orphan Pharmaceuticals and Novartis, and consultancy fees from AOP Orphan Pharmaceuticals. GB and RH declare no competing interests., (Copyright © 2022 Elsevier Ltd. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
8. Revisiting Diagnostic performances of serum erythropoïetin level and JAK2 mutation for polycythemias: analysis of a cohort of 1090 patients with red cell mass measurement.
- Author
-
Maslah N, Ravdan O, Drevon L, Verger E, Belkhodja C, Chomienne C, Cassinat B, Kiladjian JJ, Giraudier S, and Schlageter MH
- Subjects
- Alleles, Amino Acid Substitution, Clinical Decision-Making, Disease Management, Erythrocyte Indices, Erythrocyte Volume, Gene Frequency, Genetic Association Studies, Genetic Predisposition to Disease, Humans, Plasma Volume, Polycythemia Vera diagnosis, Polycythemia Vera epidemiology, Sensitivity and Specificity, Erythropoietin blood, Janus Kinase 2 genetics, Mutation, Polycythemia Vera blood, Polycythemia Vera genetics
- Abstract
We assessed the diagnostic performances of erythropoietin and JAK2 mutations in 1,090 patients with suspected polycythemia who were referred for red cell mass (RCM) measurement. In patients with a high haematocrit and/or haemoglobin level, a low erythropoietin level (<=3·3 mUI/ml) and JAK2 mutation showed comparable positive predictive value (PPV) for true polycythemia (RCM>=125%), 92·1% and 90% respectively. A very-low erythropoietin level (<=1·99 mUI/ml) had a PPV of 100% for polycythemia vera (PV) diagnosis. We confirmed the correlations between RCM, erythropoietin and JAK2 variant allelic frequency in PV patients. This study prompts the need to revisit the role of EPO in PV diagnostic criteria., (© 2021 British Society for Haematology and John Wiley & Sons Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
9. Altered Ca 2+ Homeostasis in Red Blood Cells of Polycythemia Vera Patients Following Disturbed Organelle Sorting during Terminal Erythropoiesis.
- Author
-
Buks R, Dagher T, Rotordam MG, Monedero Alonso D, Cochet S, Gautier EF, Chafey P, Cassinat B, Kiladjian JJ, Becker N, Plo I, Egée S, and El Nemer W
- Subjects
- Animals, Cell Size, Erythroblasts metabolism, Erythroid Cells metabolism, Erythroid Cells pathology, Humans, Intermediate-Conductance Calcium-Activated Potassium Channels metabolism, Intracellular Space metabolism, Janus Kinase 2 genetics, Mice, Inbred C57BL, Mutation genetics, Proteome metabolism, Reticulocytes metabolism, Ribosomes metabolism, Thrombocytosis blood, Mice, Calcium metabolism, Erythrocytes metabolism, Erythropoiesis, Homeostasis, Organelles metabolism, Polycythemia Vera blood, Polycythemia Vera metabolism
- Abstract
Over 95% of Polycythemia Vera (PV) patients carry the V617F mutation in the tyrosine kinase Janus kinase 2 (JAK2), resulting in uncontrolled erythroid proliferation and a high risk of thrombosis. Using mass spectrometry, we analyzed the RBC membrane proteome and showed elevated levels of multiple Ca
2+ binding proteins as well as endoplasmic-reticulum-residing proteins in PV RBC membranes compared with RBC membranes from healthy individuals. In this study, we investigated the impact of JAK2V617F on (1) calcium homeostasis and RBC ion channel activity and (2) protein expression and sorting during terminal erythroid differentiation. Our data from automated patch-clamp show modified calcium homeostasis in PV RBCs and cell lines expressing JAK2V617F , with a functional impact on the activity of the Gárdos channel that could contribute to cellular dehydration. We show that JAK2V617F could play a role in organelle retention during the enucleation step of erythroid differentiation, resulting in modified whole cell proteome in reticulocytes and RBCs in PV patients. Given the central role that calcium plays in the regulation of signaling pathways, our study opens new perspectives to exploring the relationship between JAK2V617F , calcium homeostasis, and cellular abnormalities in myeloproliferative neoplasms, including cellular interactions in the bloodstream in relation to thrombotic events.- Published
- 2021
- Full Text
- View/download PDF
10. ABCG2 Is Overexpressed on Red Blood Cells in Ph-Negative Myeloproliferative Neoplasms and Potentiates Ruxolitinib-Induced Apoptosis.
- Author
-
Buks R, Brusson M, Cochet S, Galochkina T, Cassinat B, Nemazanyy I, Peyrard T, Kiladjian JJ, de Brevern AG, Azouzi S, and El Nemer W
- Subjects
- ATP Binding Cassette Transporter, Subfamily G, Member 2 antagonists & inhibitors, ATP Binding Cassette Transporter, Subfamily G, Member 2 chemistry, Apoptosis drug effects, Binding Sites, Cell Differentiation drug effects, Cell Line, Computer Simulation, Diketopiperazines pharmacology, Erythrocytes drug effects, Erythroid Cells drug effects, Heterocyclic Compounds, 4 or More Rings pharmacology, Humans, Hydroxyurea metabolism, Hydroxyurea pharmacology, Interferon-alpha pharmacology, K562 Cells, Myeloproliferative Disorders blood, Myeloproliferative Disorders drug therapy, Myeloproliferative Disorders pathology, Neoplasm Proteins antagonists & inhibitors, Neoplasm Proteins chemistry, Nitriles, Phosphatidylserines metabolism, Polycythemia Vera blood, Polycythemia Vera pathology, Pyrazoles chemistry, Pyrazoles metabolism, Pyrazoles pharmacokinetics, Pyrimidines, ATP Binding Cassette Transporter, Subfamily G, Member 2 metabolism, Erythrocytes metabolism, Neoplasm Proteins metabolism, Polycythemia Vera drug therapy, Pyrazoles pharmacology
- Abstract
Myeloproliferative neoplasms (MPNs) are a group of disorders characterized by clonal expansion of abnormal hematopoietic stem cells leading to hyperproliferation of one or more myeloid lineages. The main complications in MPNs are high risk of thrombosis and progression to myelofibrosis and leukemia. MPN patients with high risk scores are treated by hydroxyurea (HU), interferon-α, or ruxolitinib, a tyrosine kinase inhibitor. Polycythemia vera (PV) is an MPN characterized by overproduction of red blood cells (RBCs). ABCG2 is a member of the ATP-binding cassette superfamily transporters known to play a crucial role in multidrug resistance development. Proteome analysis showed higher ABCG2 levels in PV RBCs compared to RBCs from healthy controls and an additional increase of these levels in PV patients treated with HU, suggesting that ABCG2 might play a role in multidrug resistance in MPNs. In this work, we explored the role of ABCG2 in the transport of ruxolitinib and HU using human cell lines, RBCs, and in vitro differentiated erythroid progenitors. Using stopped-flow analysis, we showed that HU is not a substrate for ABCG2. Using transfected K562 cells expressing three different levels of recombinant ABCG2, MPN RBCs, and cultured erythroblasts, we showed that ABCG2 potentiates ruxolitinib-induced cytotoxicity that was blocked by the ABCG2-specific inhibitor KO143 suggesting ruxolitinib intracellular import by ABCG2. In silico modeling analysis identified possible ruxolitinib-binding site locations within the cavities of ABCG2. Our study opens new perspectives in ruxolitinib efficacy research targeting cell types depending on ABCG2 expression and polymorphisms among patients.
- Published
- 2021
- Full Text
- View/download PDF
11. Long-term follow-up of JAK2 exon 12 polycythemia vera: a French Intergroup of Myeloproliferative Neoplasms (FIM) study.
- Author
-
Tondeur S, Paul F, Riou J, Mansier O, Ranta D, Le Clech L, Lippert E, Tavitian S, Chaoui D, Mercier M, De Renzis B, Cottin L, Cassinat B, Chrétien JM, Ianotto JC, Allangba O, Marzac C, Voillat L, Boyer F, Orvain C, Hunault-Berger M, Girodon F, Kiladjian JJ, Ugo V, and Luque Paz D
- Subjects
- Adult, Aged, Cohort Studies, Female, Follow-Up Studies, France, Humans, Male, Middle Aged, Myeloproliferative Disorders genetics, Polycythemia Vera genetics, Prognosis, Survival Rate, Biomarkers, Tumor genetics, Exons, Janus Kinase 2 genetics, Mutation, Myeloproliferative Disorders pathology, Polycythemia Vera pathology
- Published
- 2021
- Full Text
- View/download PDF
12. Transient expansion of TP53 mutated clones in polycythemia vera patients treated with idasanutlin.
- Author
-
Marcellino BK, Farnoud N, Cassinat B, Lu M, Verger E, McGovern E, Patel M, Medina-Martinez J, Levine MF, Arango Ossa JE, Zhou Y, Kosiorek H, Mehrotra M, Houldsworth J, Dueck A, Rossi M, Mascarenhas J, Kiladjian JJ, Rampal RK, and Hoffman R
- Subjects
- Clone Cells metabolism, Humans, Proto-Oncogene Proteins c-mdm2 genetics, Proto-Oncogene Proteins c-mdm2 metabolism, Pyrrolidines, Tumor Suppressor Protein p53 genetics, para-Aminobenzoates, Polycythemia Vera drug therapy, Polycythemia Vera genetics
- Abstract
Activation of the P53 pathway through inhibition of MDM2 using nutlins has shown clinical promise in the treatment of solid tumors and hematologic malignancies. There is concern, however, that nutlin therapy might stimulate the emergence or expansion of TP53-mutated subclones. We recently published the results of a phase 1 trial of idasanutlin in patients with polycythemia vera (PV) that revealed tolerability and clinical activity. Here, we present data indicating that idasanutlin therapy is associated with expansion of TP53 mutant subclones. End-of-study sequencing of patients found that 5 patients in this trial harbored 12 TP53 mutations; however, only 1 patient had been previously identified as having a TP53 mutation at baseline. To identify the origin of these mutations, further analysis of raw sequencing data of baseline samples was performed and revealed that a subset of these mutations was present at baseline and expanded during treatment with idasanutlin. Follow-up samples were obtained from 4 of 5 patients in this cohort, and we observed that after cessation of idasanutlin, the variant allele frequency (VAF) of 8 of 9 TP53 mutations decreased. Furthermore, disease progression to myelofibrosis or myeloproliferative neoplasm blast phase was not observed in any of these patients after 19- to 32-month observation. These data suggest that idasanutlin treatment may promote transient TP53 mutant clonal expansion. A larger study geared toward high-resolution detection of low VAF mutations is required to explore whether patients acquire de novo TP53 mutations after idasanutlin therapy., (© 2020 by The American Society of Hematology.)
- Published
- 2020
- Full Text
- View/download PDF
13. From leeches to interferon: should cytoreduction be prescribed for all patients with polycythemia vera?
- Author
-
Kiladjian JJ and Barbui T
- Subjects
- Animals, Clinical Decision-Making, Clinical Trials as Topic, Disease Management, Humans, Interferons administration & dosage, Interferons adverse effects, Polycythemia Vera complications, Polycythemia Vera diagnosis, Polycythemia Vera etiology, Thrombosis etiology, Thrombosis prevention & control, Treatment Outcome, Interferons therapeutic use, Leeches, Leeching methods, Phlebotomy methods, Polycythemia Vera therapy
- Published
- 2020
- Full Text
- View/download PDF
14. Molecular profiling and risk classification of patients with myeloproliferative neoplasms and splanchnic vein thromboses.
- Author
-
Debureaux PE, Cassinat B, Soret-Dulphy J, Mora B, Verger E, Maslah N, Plessier A, Rautou PE, Ollivier-Hourman I, De Ledinghen V, Goria O, Bureau C, Siracusa C, Valla D, Giraudier S, Passamonti F, and Kiladjian JJ
- Subjects
- Adult, Female, Humans, Male, Retrospective Studies, Myeloproliferative Disorders diagnosis, Myeloproliferative Disorders genetics, Polycythemia Vera, Primary Myelofibrosis, Venous Thrombosis
- Abstract
Myeloproliferative neoplasms (MPNs) are the most frequent underlying causes of splanchnic vein thromboses (SVTs). MPN patients with SVTs (MPN-SVT) often have a unique presentation including younger age, female predominance, and low Janus kinase 2 (JAK2) mutation allele burden. This study aimed at identifying risk factors for adverse hematologic outcomes in MPN-SVT patients. We performed a retrospective study of a fully characterized cohort of MPN-SVT patients. The primary outcome was the incidence of evolution to myelofibrosis, acute leukemia, or death. Eighty patients were included in the testing cohort. Median follow-up was 11 years. Most of the patients were women with a mean age of 42 years and a diagnosis of polycythemia vera. The primary outcome was met in 13% of the patients and was associated with a JAK2V617F allele burden ≥50% (odds ratio [OR], 14.7) and presence of additional mutations in genes affecting chromatin/spliceosome (OR, 9). We identified high-risk patients (29% of the cohort) as those harboring at least 1 molecular risk factor: JAK2-mutant allele burden ≥50%, presence of chromatin/spliceosome/TP53 mutation. High-risk patients had worse event-free survival (81% vs 100%; P = .001) and overall survival at 10 years (89% vs 100%; P = .01) than low-risk patients. These results were confirmed in an independent validation cohort of 30 MPN-SVT patients. In conclusion, molecular profiling identified MPN-SVT patients with dismal outcome. In this high-risk population, a disease-modifying therapy should be taken into consideration to minimize the probability of transformation., (© 2020 by The American Society of Hematology.)
- Published
- 2020
- Full Text
- View/download PDF
15. Long-term efficacy and safety of ruxolitinib in polycythaemia vera - Authors' reply.
- Author
-
Kiladjian JJ
- Subjects
- Antilymphocyte Serum, Humans, Nitriles, Pyrazoles, Pyrimidines, Unrelated Donors, Graft vs Host Disease, Hematologic Neoplasms, Polycythemia Vera
- Published
- 2020
- Full Text
- View/download PDF
16. Ropeginterferon alfa-2b versus standard therapy for polycythaemia vera (PROUD-PV and CONTINUATION-PV): a randomised, non-inferiority, phase 3 trial and its extension study.
- Author
-
Gisslinger H, Klade C, Georgiev P, Krochmalczyk D, Gercheva-Kyuchukova L, Egyed M, Rossiev V, Dulicek P, Illes A, Pylypenko H, Sivcheva L, Mayer J, Yablokova V, Krejcy K, Grohmann-Izay B, Hasselbalch HC, Kralovics R, and Kiladjian JJ
- Subjects
- Aged, Equivalence Trials as Topic, Female, Follow-Up Studies, Humans, Male, Middle Aged, Polycythemia Vera pathology, Prognosis, Recombinant Proteins therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Antiviral Agents therapeutic use, Interferon alpha-2 therapeutic use, Interferon-alpha therapeutic use, Polycythemia Vera drug therapy, Polyethylene Glycols therapeutic use
- Abstract
Background: The PROUD-PV and CONTINUATION-PV trials aimed to compare the novel monopegylated interferon ropeginterferon alfa-2b with hydroxyurea, the standard therapy for patients with polycythaemia vera, over 3 years of treatment., Methods: PROUD-PV and its extension study, CONTINUATION-PV, were phase 3, randomised, controlled, open-label, trials done in 48 clinics in Europe. Patients were eligible if 18 years or older with early stage polycythaemia vera (no history of cytoreductive treatment or less than 3 years of previous hydroxyurea treatment) diagnosed by WHO's 2008 criteria. Patients were randomly assigned 1:1 to ropeginterferon alfa-2b (subcutaneously every 2 weeks, starting at 100 μg) or hydroxyurea (orally starting at 500 mg/day). After 1 year, patients could opt to enter the extension part of the trial, CONTINUATION-PV. The primary endpoint in PROUD-PV was non-inferiority of ropeginterferon alfa-2b versus hydroxyurea regarding complete haematological response with normal spleen size (longitudinal diameter of ≤12 cm for women and ≤13 cm for men) at 12 months; in CONTINUATION-PV, the coprimary endpoints were complete haematological response with normalisation of spleen size and with improved disease burden (ie, splenomegaly, microvascular disturbances, pruritus, and headache). We present the final results of PROUD-PV and an interim analysis at 36 months of the CONTINUATION-PV study (per statistical analysis plan). Analyses for safety and efficacy were per-protocol. The trials were registered on EudraCT, 2012-005259-18 (PROUD-PV) and 2014-001357-17 (CONTINUATION-PV, which is ongoing)., Findings: Patients were recruited from Sept 17, 2013 to March 13, 2015 with 306 enrolled. 257 patients were randomly assigned, 127 were treated in each group (three patients withdrew consent in the hydroxyurea group), and 171 rolled over to the CONTINUATION-PV trial. Median follow-up was 182·1 weeks (IQR 166·3-201·7) in the ropeginterferon alfa-2b and 164·5 weeks (144·4-169·3) in the standard therapy group. In PROUD-PV, 26 (21%) of 122 patients in the ropeginterferon alfa-2b group and 34 (28%) of 123 patients in the standard therapy group met the composite primary endpoint of complete haematological response with normal spleen size. In CONTINUATION-PV, complete haematological response with improved disease burden was met in 50 (53%) of 95 patients in the ropeginterferon alfa-2b group versus 28 (38%) of 74 patients in the hydroxyurea group, p=0·044 at 36 months. Complete haematological response without the spleen criterion in the ropeginterferon alfa-2b group versus standard therapy group were: 53 (43%) of 123 patients versus 57 (46%) of 125 patients, p=0·63 at 12 months (PROUD-PV), and 67 (71%) of 95 patients versus 38 (51%) of 74 patients, p=0·012 at 36 months (CONTINUATION-PV). The most frequently reported grade 3 and grade 4 treatment-related adverse events were increased γ-glutamyltransferase (seven [6%] of 127 patients) and increased alanine aminotransferase (four [3%] of 127 patients) in the ropeginterferon alfa-2b group, and leucopenia (six [5%] of 127 patients) and thrombocytopenia (five [4%] of 127 patients) in the standard therapy group. Treatment-related serious adverse events occurred in three (2%) of 127 patients in the ropeginterferon alfa-2b group and five (4%) of 127 patients in the hydroxyurea group. One treatment-related death was reported in the standard therapy group (acute leukaemia)., Interpretation: In patients with early polycythaemia vera, who predominantly presented without splenomegaly, ropeginterferon alfa-2b was effective in inducing haematological responses; non-inferiority to hydroxyurea regarding haematological response and normal spleen size was not shown at 12 months. However, response to ropeginterferon alfa-2b continued to increase over time with improved responses compared with hydroxyurea at 36 months. Considering the high and durable haematological and molecular responses and its good tolerability, ropeginterferon alfa-2b offers a valuable and safe long-term treatment option with features distinct from hydroxyurea., Funding: AOP Orphan Pharmaceuticals AG., (Copyright © 2020 Elsevier Ltd. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
17. Masked polycythemia vera: analysis of a single center cohort of 2480 red cell masses.
- Author
-
Maslah N, Soret J, Dosquet C, Vercellino L, Belkhodja C, Schlageter MH, Cassinat B, Kiladjian JJ, Chomienne C, and Giraudier S
- Subjects
- Cohort Studies, Erythrocyte Volume, Humans, Polycythemia Vera diagnosis, Polycythemia Vera genetics
- Published
- 2020
- Full Text
- View/download PDF
18. Long-term efficacy and safety of ruxolitinib versus best available therapy in polycythaemia vera (RESPONSE): 5-year follow up of a phase 3 study.
- Author
-
Kiladjian JJ, Zachee P, Hino M, Pane F, Masszi T, Harrison CN, Mesa R, Miller CB, Passamonti F, Durrant S, Griesshammer M, Kirito K, Besses C, Moiraghi B, Rumi E, Rosti V, Blau IW, Francillard N, Dong T, Wroclawska M, Vannucchi AM, and Verstovsek S
- Subjects
- Antiviral Agents therapeutic use, Drug Therapy, Combination, Fibrinolytic Agents therapeutic use, Follow-Up Studies, Humans, Hydroxyurea administration & dosage, Nitriles, Pipobroman administration & dosage, Prognosis, Pyrimidines, Recombinant Proteins therapeutic use, Survival Rate, Time Factors, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Interferon alpha-2 therapeutic use, Interferon-alpha therapeutic use, Polycythemia Vera drug therapy, Polyethylene Glycols therapeutic use, Pyrazoles therapeutic use, Quinazolines therapeutic use
- Abstract
Background: Polycythaemia vera is a myeloproliferative neoplasm characterised by excessive proliferation of erythroid, myeloid, and megakaryocytic components in the bone marrow due to mutations in the Janus kinase 2 (JAK2) gene. Ruxolitinib, a JAK 1 and JAK 2 inhibitor, showed superiority over best available therapy in a phase 2 study in patients with polycythaemia vera who were resistant to or intolerant of hydroxyurea. We aimed to compare the long-term safety and efficacy of ruxolitinib with best available therapy in patients with polycythaemia vera who were resistant to or intolerant of hydroxyurea., Methods: We report the 5-year results for a randomised, open-label, phase 3 study (RESPONSE) that enrolled patients at 109 sites across North America, South America, Europe, and the Asia-Pacific region. Patients (18 years or older) with polycythaemia vera who were resistant to or intolerant of hydroxyurea were randomly assigned 1:1 to receive either ruxolitinib or best available therapy. Patients randomly assigned to the ruxolitinib group received the drug orally at a starting dose of 10 mg twice a day. Single-agent best available therapy comprised hydroxyurea, interferon or pegylated interferon, pipobroman, anagrelide, approved immunomodulators, or observation without pharmacological treatment. The primary endpoint, composite response (patients who achieved both haematocrit control without phlebotomy and 35% or more reduction from baseline in spleen volume) at 32 weeeks was previously reported. Patients receiving best available therapy could cross over to ruxolitinib after week 32. We assessed the durability of primary composite response, complete haematological remission, overall clinicohaematological response, overall survival, patient-reported outcomes, and safety after 5-years of follow-up. This study is registered with ClinicalTrials.gov, NCT01243944., Findings: We enrolled patients between Oct 27, 2010, and Feb 13, 2013, and the study concluded on Feb 9, 2018. Of 342 individuals screened for eligibility, 222 patients were randomly assigned to receive ruxolitinib (n=110, 50%) or best available therapy (n=112, 50%). The median time since polycythaemia vera diagnosis was 8·2 years (IQR 3·9-12·3) in the ruxolitinib group and 9·3 years (4·9-13·8) in the best available therapy group. 98 (88%) of 112 patients initially randomly assigned to best available therapy crossed over to receive ruxolitinib and no patient remained on best available therapy after 80 weeks of study. Among 25 primary responders in the ruxolitinib group, six had progressed at the time of final analysis. At 5 years, the probability of maintaining primary composite response was 74% (95% CI 51-88). The probability of maintaining complete haematological remission was 55% (95% CI 32-73) and the probability of maintaining overall clinicohaematological responses was 67% (54-77). In the intention-to-treat analysis not accounting for crossover, the probability of survival at 5 years was 91·9% (84·4-95·9) with ruxolitinib therapy and 91·0% (82·8-95·4) with best available therapy. Anaemia was the most common adverse event in patients receiving ruxolitinib (rates per 100 patient-years of exposure were 8·9 for ruxolitinib and 8·8 for the crossover population), though most anaemia events were mild to moderate in severity (grade 1 or 2 anaemia rates per 100 patient-years of exposure were 8·0 for ruxolitinib and 8·2 for the crossover population). Non-haematological adverse events were generally lower with long-term ruxolitinib treatment than with best available therapy. Thromboembolic events were lower in the ruxolitinib group than the best available therapy group. There were two on-treatment deaths in the ruxolitinib group. One of these deaths was due to gastric adenocarcinoma, which was assessed by the investigator as related to ruxolitinib treatment., Interpretation: We showed that ruxolitinib is a safe and effective long-term treatment option for patients with polycythaemia vera who are resistant to or intolerant of hydroxyurea. Taken together, ruxolitinib treatment offers the first widely approved therapeutic alternative for this post-hydroxyurea patient population., Funding: Novartis Pharmaceuticals Corporation., (Copyright © 2020 Elsevier Ltd. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
19. Impact of bone marrow fibrosis grade in post-polycythemia vera and post-essential thrombocythemia myelofibrosis: A study of the MYSEC group.
- Author
-
Mora B, Guglielmelli P, Rumi E, Maffioli M, Barraco D, Rambaldi A, Caramella M, Komrokji RS, Kiladjian JJ, Gotlib J, Iurlo A, Cervantes F, Devos T, Palandri F, De Stefano V, Ruggeri M, Silver RT, Albano F, Benevolo G, Cavalloni C, Uccella S, Accetta R, Siracusa C, Agnoli S, Merli M, Barbui T, Bertù L, Cazzola M, Vannucchi AM, and Passamonti F
- Subjects
- Female, Humans, Male, Middle Aged, Polycythemia Vera, Primary Myelofibrosis pathology, Thrombocythemia, Essential
- Published
- 2020
- Full Text
- View/download PDF
20. Safety and efficacy findings from the open-label, multicenter, phase 3b, expanded treatment protocol study of ruxolitinib for treatment of patients with polycythemia vera who are resistant/intolerant to hydroxyurea and for whom no alternative treatments are available.
- Author
-
Foltz L, Pica GM, Zerazhi H, Van Droogenbroeck J, Visanica S, Báez de la Fuente E, Leber B, de Almeida AM, Ranta D, Kiladjian JJ, Chrit L, Kandra A, Morando J, and Devos T
- Subjects
- Adult, Aged, Aged, 80 and over, Female, Follow-Up Studies, Humans, Male, Middle Aged, Nitriles, Polycythemia Vera pathology, Prognosis, Pyrimidines, Survival Rate, Antineoplastic Agents pharmacology, Drug Resistance, Neoplasm drug effects, Hydroxyurea pharmacology, Polycythemia Vera drug therapy, Pyrazoles therapeutic use, Salvage Therapy
- Abstract
Ruxolitinib was recently approved for the treatment of patients with polycythemia vera who are resistant/intolerant to hydroxyurea based on data from the RESPONSE studies. This phase 3b, Expanded Treatment Protocol study (NCT02292446) of ruxolitinib for hydroxyurea-resistant/intolerant patients with polycythemia vera ( N = 161: median exposure = 25.1 weeks) further evaluated the safety of ruxolitinib. Adverse events (AEs) led to dose adjustment/interruption in 37.9% of patients and study drug discontinuation in 8.7% of patients. The most common hematologic AEs included anemia and thrombocytosis; while headache and diarrhea were the most frequent nonhematologic AEs. At week 24, 45.3% of patients achieved hematocrit control; hematologic remission was seen in 18% of patients. At least, 50% of reduction in spleen length was achieved in 86.7% of patients from baseline at any time. The observed safety profile of ruxolitinib was consistent and the efficacy results were similar to the observed values in the RESPONSE studies.
- Published
- 2019
- Full Text
- View/download PDF
21. Characteristics and outcomes of patients with essential thrombocythemia or polycythemia vera diagnosed before 20 years of age: a systematic review.
- Author
-
Ianotto JC, Curto-Garcia N, Lauermanova M, Radia D, Kiladjian JJ, and Harrison CN
- Subjects
- Adolescent, Asymptomatic Diseases, Child, Cytotoxins therapeutic use, Early Diagnosis, Fibrinolytic Agents therapeutic use, Gene Expression, Hemorrhage drug therapy, Hemorrhage genetics, Hemorrhage pathology, Humans, Janus Kinase 1 genetics, Janus Kinase 2 genetics, Mutation, Polycythemia Vera drug therapy, Polycythemia Vera genetics, Polycythemia Vera pathology, Prognosis, Splenomegaly drug therapy, Splenomegaly genetics, Splenomegaly pathology, Thrombocythemia, Essential drug therapy, Thrombocythemia, Essential genetics, Thrombocythemia, Essential pathology, Thrombosis drug therapy, Thrombosis genetics, Thrombosis pathology, Young Adult, Hemorrhage diagnosis, Polycythemia Vera diagnosis, Splenomegaly diagnosis, Thrombocythemia, Essential diagnosis, Thrombosis diagnosis
- Abstract
Although it is well known that myeloproliferative neoplasms occur in younger patients, few large cohorts of such patients have been reported. Thus, our knowledge about circumstances of diagnosis, outcome and treatment is limited, especially for children and young adults. We therefore performed a systematic review of cases, published since 2005, concerning patients aged below 20 years at the time of diagnosis of essential thrombocythemia or polycythemia vera. We identified 396 cases of essential thrombocythemia and 75 of polycythemia vera. The median age at diagnosis was 9.3 and 12 years, respectively, and females constituted 57.6% and 45% of the groups, respectively. Half of the patients were asymptomatic at diagnosis. The proportion of so-called triple negativity was high: 57% in essential thrombocythemia and 73% in polycythemia vera. The incidence of thrombosis during the follow-up was 9.3% in patients with polycythemia vera and less, 3.8%, in those with essential thrombocythemia. Venous events were predominant (84.2%), with hemorrhagic episodes being rarer (<5%). The risk of evolution also seemed low (2% to myelofibrosis and no reports of acute leukemia), but the median follow-up was only 50 months. Survival curves were not available. Half of the patients received an antithrombotic drug and 40.5% received a cytoreductive drug. All data should be analyzed with care because of the proportion of missing data (10.7% to 74.7%). This review highlights interesting points concerning this population of young patients with myeloproliferative neoplasms, including that such patients were identified as negative for all common driver mutations, but also shows the need for larger contemporary cohorts with longer follow-up to assess the true prognosis of these patients., (Copyright© 2019 Ferrata Storti Foundation.)
- Published
- 2019
- Full Text
- View/download PDF
22. Second primary malignancies in postpolycythemia vera and postessential thrombocythemia myelofibrosis: A study on 2233 patients.
- Author
-
Mora B, Rumi E, Guglielmelli P, Barraco D, Maffioli M, Rambaldi A, Caramella M, Komrokji R, Gotlib J, Kiladjian JJ, Cervantes F, Devos T, Palandri F, De Stefano V, Ruggeri M, Silver RT, Benevolo G, Albano F, Cavalloni C, Pietra D, Barbui T, Rotunno G, Cazzola M, Vannucchi AM, Giorgino T, and Passamonti F
- Subjects
- Cohort Studies, Female, Humans, Incidence, Janus Kinase Inhibitors therapeutic use, Male, Neoplasms, Second Primary drug therapy, Polycythemia Vera drug therapy, Primary Myelofibrosis drug therapy, Treatment Outcome, Neoplasms, Second Primary epidemiology, Polycythemia Vera epidemiology, Primary Myelofibrosis epidemiology, Skin Neoplasms epidemiology, Thrombocythemia, Essential epidemiology
- Abstract
Patients with myeloproliferative neoplasms (MPN) are known to have higher incidence of nonhematological second primary malignancies (SPM) compared to general population. In the MYSEC study on 781 secondary myelofibrosis (SMF) patients, the incidence of SPM after SMF diagnosis resulted 0.98/100 patient-years. When including non-melanoma skin cancers (NMSC), the incidence arose to 1.56/100 patient-years. In SMF, JAK inhibitor treatment was associated only with NMSC occurrence. Then, we merged the MYSEC cohort with a large dataset of PV and ET not evolving into SMF. In this subanalysis, we did not find any correlation between SPM and SMF occurrence. These findings highlight the need of studies aimed at identifying MPN patients at higher risk of SPM., (© 2019 The Authors. Cancer Medicine published by John Wiley & Sons Ltd.)
- Published
- 2019
- Full Text
- View/download PDF
23. Thromboembolic events in polycythemia vera.
- Author
-
Griesshammer M, Kiladjian JJ, and Besses C
- Subjects
- Age Factors, Anticoagulants therapeutic use, Aspirin therapeutic use, Humans, Hydroxyurea adverse effects, Hydroxyurea therapeutic use, Interferons therapeutic use, Nitriles, Phlebotomy, Pyrazoles therapeutic use, Pyrimidines, Risk Factors, Thrombosis diagnosis, Thrombosis etiology, Thrombosis therapy, Polycythemia Vera complications, Polycythemia Vera diagnosis, Polycythemia Vera therapy, Thromboembolism diagnosis, Thromboembolism etiology, Thromboembolism therapy
- Abstract
Thromboembolic events and cardiovascular disease are the most prevalent complications in patients with polycythemia vera (PV) compared with other myeloproliferative disorders and are the major cause of morbidity and mortality in this population. Moreover, a vascular complication such as arterial or venous thrombosis often leads to the diagnosis of PV. The highest rates of thrombosis typically occur shortly before or at diagnosis and decrease over time, probably due to the effects of treatment. Important risk factors include age (≥ 60 years old) and a history of thrombosis; elevated hematocrit and leukocytosis are also associated with an increased risk of thrombosis. The goal of therapy is to reduce the risk of thrombosis by controlling hematocrit to < 45%, a target associated with reduced rates of cardiovascular death and major thrombosis. Low-risk patients (< 60 years old with no history of thrombosis) are managed with phlebotomy and low-dose aspirin, whereas high-risk patients (≥ 60 years old and/or with a history of thrombosis) should be treated with cytoreductive agents. Interferon and ruxolitinib are considered second-line therapies for patients who are intolerant of or have an inadequate response to hydroxyurea, which is typically used as first-line therapy. In this review, we discuss factors associated with thrombosis and recent data on current treatments, including anticoagulation, highlighting the need for more controlled studies to determine the most effective cytoreductive therapies for reducing the risk of thrombosis in patients with PV.
- Published
- 2019
- Full Text
- View/download PDF
24. Ropeginterferon alpha-2b targets JAK2V617F-positive polycythemia vera cells in vitro and in vivo.
- Author
-
Verger E, Soret-Dulphy J, Maslah N, Roy L, Rey J, Ghrieb Z, Kralovics R, Gisslinger H, Grohmann-Izay B, Klade C, Chomienne C, Giraudier S, Cassinat B, and Kiladjian JJ
- Subjects
- Cell Line, Tumor, Cell Proliferation drug effects, Dose-Response Relationship, Drug, Gene Frequency, Genotype, Humans, Interferon alpha-2 therapeutic use, Interferon-alpha therapeutic use, Polycythemia Vera diagnosis, Polycythemia Vera drug therapy, Polyethylene Glycols therapeutic use, Recombinant Proteins pharmacology, Recombinant Proteins therapeutic use, Alleles, Amino Acid Substitution, Interferon alpha-2 pharmacology, Interferon-alpha pharmacology, Janus Kinase 2, Mutation, Polycythemia Vera genetics, Polyethylene Glycols pharmacology
- Abstract
Polycythemia vera is characterized by the acquisition of the JAK2V617F mutation. Recommended treatments include hydroxyurea and interferon-alpha. Several groups have reported a reduction in the JAK2 mutant allele burden in interferon-treated patients, but significance of this observation is questioned. We characterized the activity of ropeginterferon alpha-2b, a novel form of interferon-alpha recently shown to be safe and efficacious in polycythemia vera. Ropeginterferon was able to inhibit the proliferation of the HEL, UKE-1, and UT-7 JAK2-mutant cell lines while sparing JAK2-wild-type UT-7 and normal CD34+ cells growth. In vitro treatment of erythroid progenitors derived from PV patients showed that ropeginterferon could considerably inhibit the growth of endogenous erythroid colonies, a hallmark of polycythemia vera. Finally, we could study in sequential samples the clonal architecture of erythroid progenitors derived from patients included in a randomized study comparing hydroxyurea to ropeginterferon. After 1 year of treatment with ropeginterferon, the ratio of JAK2-mutated to wild-type colonies grown from bone marrow progenitors was reduced by 64%, compared to 25% in patients receiving hydroxyurea. This study shows that ropeginterferon has a potent targeted activity against JAK2-mutant cells and is able to drastically reduce the proportion of malignant progenitors in patients treated with this drug.
- Published
- 2018
- Full Text
- View/download PDF
25. Gender effect on phenotype and genotype in patients with post-polycythemia vera and post-essential thrombocythemia myelofibrosis: results from the MYSEC project.
- Author
-
Barraco D, Mora B, Guglielmelli P, Rumi E, Maffioli M, Rambaldi A, Caramella M, Komrokji R, Gotlib J, Kiladjian JJ, Cervantes F, Devos T, Palandri F, De Stefano V, Ruggeri M, Silver RT, Benevolo G, Albano F, Merli M, Pietra D, Barbui T, Rotunno G, Cazzola M, Giorgino T, Vannucchi AM, and Passamonti F
- Subjects
- Adult, Aged, Aged, 80 and over, Biomarkers, Female, Genetic Association Studies, Genetic Predisposition to Disease, Humans, Male, Middle Aged, Polycythemia Vera mortality, Primary Myelofibrosis mortality, Prognosis, Severity of Illness Index, Sex Factors, Thrombocythemia, Essential mortality, Genotype, Phenotype, Polycythemia Vera diagnosis, Polycythemia Vera genetics, Primary Myelofibrosis diagnosis, Primary Myelofibrosis genetics, Thrombocythemia, Essential diagnosis, Thrombocythemia, Essential genetics
- Published
- 2018
- Full Text
- View/download PDF
26. Value of cytogenetic abnormalities in post-polycythemia vera and post-essential thrombocythemia myelofibrosis: a study of the MYSEC project.
- Author
-
Mora B, Giorgino T, Guglielmelli P, Rumi E, Maffioli M, Rambaldi A, Caramella M, Komrokji R, Gotlib J, Kiladjian JJ, Cervantes F, Devos T, Palandri F, De Stefano V, Ruggeri M, Silver RT, Benevolo G, Albano F, Cavalloni C, Barraco D, Merli M, Pietra D, Casalone R, Barbui T, Rotunno G, Cazzola M, Vannucchi AM, and Passamonti F
- Subjects
- Adult, Aged, Aged, 80 and over, Biomarkers, Biopsy, Bone Marrow metabolism, Bone Marrow pathology, Disease Progression, Female, Humans, Male, Middle Aged, Polycythemia Vera mortality, Polycythemia Vera pathology, Primary Myelofibrosis mortality, Primary Myelofibrosis pathology, Prognosis, Thrombocythemia, Essential mortality, Thrombocythemia, Essential pathology, Chromosome Aberrations, Polycythemia Vera genetics, Primary Myelofibrosis genetics, Thrombocythemia, Essential genetics
- Published
- 2018
- Full Text
- View/download PDF
27. Phase 2 study of gandotinib (LY2784544) in patients with myeloproliferative neoplasms.
- Author
-
Berdeja J, Palandri F, Baer MR, Quick D, Kiladjian JJ, Martinelli G, Verma A, Hamid O, Walgren R, Pitou C, Li PL, and Gerds AT
- Subjects
- Adult, Aged, Aged, 80 and over, Female, Humans, Janus Kinase 2 genetics, Male, Middle Aged, Mutation, Myeloproliferative Disorders drug therapy, Myeloproliferative Disorders genetics, Polycythemia Vera genetics, Thrombocythemia, Essential genetics, Antineoplastic Agents therapeutic use, Imidazoles therapeutic use, Polycythemia Vera drug therapy, Primary Myelofibrosis drug therapy, Pyrazoles therapeutic use, Pyridazines therapeutic use, Thrombocythemia, Essential drug therapy
- Abstract
Background: The Philadelphia chromosome-negative myeloproliferative neoplasms (MPNs) are associated with increases in janus kinase 2 (JAK2) signaling, often resulting from the JAK2 V617F mutation. LY2784544 (gandotinib) is a potent, selective, small-molecule inhibitor of JAK2 that has potential dose-dependent selectivity for the JAK2 V617F mutation and may inhibit additional JAK2 mutant isoforms in nonclinical testing., Methods: A multicenter, single-arm, outpatient phase 2 study evaluated the efficacy, safety, and pharmacokinetics (PK) of gandotinib administered to patients (120 mg once daily) with MPNs, including polycythemia vera (PV), essential thrombocythemia (ET), and myelofibrosis (MF). Between May 2012 and March 2015, 138 patients received at least one dose of study drug., Findings: Most frequent Grade 3 or 4 treatment-emergent adverse events that were considered study-drug related were anemia (11.6%), hyperuricemia (3.2%), fatigue (2.9%), diarrhea (2.2%), and thrombocytopenia (2.2%). Overall response rates (ORRs) in patients with JAK2 V617F-mutated PV, ET, and MF were 95%, 90.5%, and 9.1%, respectively, while patients with ET and MF without the JAK2 V617F mutations had ORRs of 43.7% and 0%, respectively., Interpretations: LY2784544 demonstrated efficacy in JAK2 V617F-mutated MPNs, including in patients previously on ruxolitinib therapy, who had an ORR of 3.3%. At the 1-year visit, 44% of patients experienced a ≥50% improvement in the MPN-Symptom Assessment Form Total Symptom Score, and 26% of patients had a 50% reduction in Brief Fatigue Inventory score., (Copyright © 2018 Elsevier Ltd. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
28. Comprehensive haematological control with ruxolitinib in patients with polycythaemia vera resistant to or intolerant of hydroxycarbamide.
- Author
-
Harrison CN, Griesshammer M, Miller C, Masszi T, Passamonti F, Zachee P, Durrant S, Pane F, Guglielmelli P, Verstovsek S, Jones MM, Hunter DS, Sun W, Li J, Khan M, Habr D, and Kiladjian JJ
- Subjects
- Adult, Aged, Aged, 80 and over, Drug Resistance, Neoplasm, Humans, Hydroxyurea therapeutic use, Middle Aged, Nitriles, Pyrimidines, Treatment Outcome, Antineoplastic Agents therapeutic use, Polycythemia Vera drug therapy, Pyrazoles therapeutic use
- Published
- 2018
- Full Text
- View/download PDF
29. Phenotype variability of patients with post polycythemia vera and post essential thrombocythemia myelofibrosis is associated with the time to progression from polycythemia vera and essential thrombocythemia.
- Author
-
Mora B, Giorgino T, Guglielmelli P, Rumi E, Maffioli M, Rambaldi A, Caramella M, Komrokji R, Gotlib J, Kiladjian JJ, Cervantes F, Devos T, Palandri F, De Stefano V, Ruggeri M, Silver RT, Benevolo G, Albano F, Cavalloni C, Barraco D, Pietra D, Barbui T, Rotunno G, Vannucchi AM, and Passamonti F
- Subjects
- Disease Progression, Humans, Phenotype, Polycythemia Vera blood, Polycythemia Vera complications, Primary Myelofibrosis complications, Retrospective Studies, Thrombocythemia, Essential blood, Thrombocythemia, Essential complications, Polycythemia Vera pathology, Primary Myelofibrosis pathology, Thrombocythemia, Essential pathology
- Published
- 2018
- Full Text
- View/download PDF
30. Impact of hydroxycarbamide and interferon-α on red cell adhesion and membrane protein expression in polycythemia vera.
- Author
-
Brusson M, De Grandis M, Cochet S, Bigot S, Marin M, Leduc M, Guillonneau F, Mayeux P, Peyrard T, Chomienne C, Le Van Kim C, Cassinat B, Kiladjian JJ, and El Nemer W
- Subjects
- Alleles, Biomarkers, Cell Adhesion drug effects, Cell Adhesion genetics, Cell Adhesion Molecules metabolism, Erythrocyte Membrane metabolism, Erythrocytes pathology, Female, Humans, Janus Kinase 2 genetics, Male, Membrane Proteins metabolism, Middle Aged, Mutation, Polycythemia Vera blood, Polycythemia Vera diagnosis, Cell Adhesion Molecules genetics, Erythrocytes drug effects, Erythrocytes metabolism, Gene Expression Regulation drug effects, Hydroxyurea pharmacology, Membrane Proteins genetics, Polycythemia Vera genetics
- Abstract
Polycythemia vera is a chronic myeloproliferative neoplasm characterized by the JAK2V617F mutation, elevated blood cell counts and a high risk of thrombosis. Although the red cell lineage is primarily affected by JAK2V617F, the impact of mutated JAK2 on circulating red blood cells is poorly documented. Recently, we showed that in polycythemia vera, erythrocytes had abnormal expression of several proteins including Lu/BCAM adhesion molecule and proteins from the endoplasmic reticulum, mainly calreticulin and calnexin. Here we investigated the effects of hydroxycarbamide and interferon-α treatments on the expression of erythroid membrane proteins in a cohort of 53 patients. Surprisingly, while both drugs tended to normalize calreticulin expression, proteomics analysis showed that hydroxycarbamide deregulated the expression of 53 proteins in red cell ghosts, with overexpression and downregulation of 37 and 16 proteins, respectively. Within over-expressed proteins, hydroxycarbamide was found to enhance the expression of adhesion molecules such as Lu/BCAM and CD147, while interferon-α did not. In addition, we found that hydroxycarbamide increased Lu/BCAM phosphorylation and exacerbated red cell adhesion to its ligand laminin. Our study reveals unexpected adverse effects of hydroxycarbamide on red cell physiology in polycythemia vera and provides new insights into the effects of this molecule on gene regulation and protein recycling or maturation during erythroid differentiation. Furthermore, our study shows deregulation of Lu/BCAM and CD147 that are two ubiquitously expressed proteins linked to progression of solid tumors, paving the way for future studies to address the role of hydroxycarbamide in tissues other than blood cells in myeloproliferative neoplasms., (Copyright © 2018 Ferrata Storti Foundation.)
- Published
- 2018
- Full Text
- View/download PDF
31. Efficacy and safety of ruxolitinib after and versus interferon use in the RESPONSE studies.
- Author
-
Kiladjian JJ, Guglielmelli P, Griesshammer M, Saydam G, Masszi T, Durrant S, Passamonti F, Jones M, Zhen H, Li J, Gadbaw B, Perez Ronco J, Khan M, and Verstovsek S
- Subjects
- Adult, Aged, Antineoplastic Agents adverse effects, Bloodletting adverse effects, Combined Modality Therapy adverse effects, Cross-Over Studies, Drug Monitoring, Drug Resistance, Multiple, Drug Resistance, Neoplasm, Female, Humans, Hydroxyurea adverse effects, Hydroxyurea therapeutic use, Interferons adverse effects, Janus Kinases metabolism, Male, Middle Aged, Nitriles, Polycythemia Vera metabolism, Polycythemia Vera physiopathology, Polycythemia Vera therapy, Practice Patterns, Physicians', Protein Kinase Inhibitors adverse effects, Pyrazoles adverse effects, Pyrimidines, Reproducibility of Results, Splenomegaly etiology, Splenomegaly prevention & control, Antineoplastic Agents therapeutic use, Interferons therapeutic use, Janus Kinases antagonists & inhibitors, Polycythemia Vera drug therapy, Protein Kinase Inhibitors therapeutic use, Pyrazoles therapeutic use
- Abstract
Ruxolitinib was well tolerated and superior to best available therapy (including interferon [IFN]) in controlling hematocrit without phlebotomy eligibility, normalizing blood counts, and improving polycythemia vera-related symptoms in the Study of Efficacy and Safety in Polycythemia Vera Subjects Who Are Resistant to or Intolerant of Hydroxyurea: JAK Inhibitor INC424 (INCB018424) Tablets Versus Best Available Care (RESPONSE) studies. This ad hoc analysis focuses on ruxolitinib in relation to IFN in the RESPONSE studies, with attention on the following: (1) safety and efficacy of ruxolitinib and best available therapy in patients who received IFN before study randomization, (2) safety and efficacy of IFN during randomized treatment in best available therapy arm, and (3) use of ruxolitinib after crossover from best available therapy in IFN-treated patients. IFN exposure before randomization had little effect on the efficacy or safety of ruxolitinib. In the randomized treatment arms, ruxolitinib was superior to IFN in efficacy [hematocrit control (RESPONSE = 60% of ruxolitinib vs 23% of IFN patients; RESPONSE-2 = 62% of ruxolitinib vs 15% of IFN patients)] and was tolerated better in hydroxyurea-resistant or hydroxyurea-intolerant patients. After crossing over to receive ruxolitinib, patients who had initially received IFN and did not respond had improved hematologic and spleen responses (62% of patients at any time after crossover) and an overall reduction in phlebotomy procedures. Rates and incidences of the most common adverse events decreased after crossover to ruxolitinib, except for infections (primarily grade 1 or 2). These data suggest that ruxolitinib is efficacious and well tolerated in patients who were previously treated with IFN. The RESPONSE (NCT01243944) and RESPONSE-2 (NCT02038036) studies were registered at clinicaltrials.gov .
- Published
- 2018
- Full Text
- View/download PDF
32. A clinical-molecular prognostic model to predict survival in patients with post polycythemia vera and post essential thrombocythemia myelofibrosis.
- Author
-
Passamonti F, Giorgino T, Mora B, Guglielmelli P, Rumi E, Maffioli M, Rambaldi A, Caramella M, Komrokji R, Gotlib J, Kiladjian JJ, Cervantes F, Devos T, Palandri F, De Stefano V, Ruggeri M, Silver RT, Benevolo G, Albano F, Caramazza D, Merli M, Pietra D, Casalone R, Rotunno G, Barbui T, Cazzola M, and Vannucchi AM
- Subjects
- Adult, Aged, Aged, 80 and over, Biomarkers, Female, Follow-Up Studies, Humans, Male, Middle Aged, Mutation, Polycythemia Vera diagnosis, Primary Myelofibrosis diagnosis, Prognosis, Risk Factors, Survival Analysis, Thrombocythemia, Essential diagnosis, Polycythemia Vera genetics, Polycythemia Vera mortality, Primary Myelofibrosis genetics, Primary Myelofibrosis mortality, Thrombocythemia, Essential genetics, Thrombocythemia, Essential mortality
- Abstract
Polycythemia vera (PV) and essential thrombocythemia (ET) are myeloproliferative neoplasms with variable risk of evolution into post-PV and post-ET myelofibrosis, from now on referred to as secondary myelofibrosis (SMF). No specific tools have been defined for risk stratification in SMF. To develop a prognostic model for predicting survival, we studied 685 JAK2, CALR, and MPL annotated patients with SMF. Median survival of the whole cohort was 9.3 years (95% CI: 8-not reached-NR-). Through penalized Cox regressions we identified negative predictors of survival and according to beta risk coefficients we assigned 2 points to hemoglobin level <11 g/dl, to circulating blasts ⩾3%, and to CALR-unmutated genotype, 1 point to platelet count <150 × 10
9 /l and to constitutional symptoms, and 0.15 points to any year of age. Myelofibrosis Secondary to PV and ET-Prognostic Model (MYSEC-PM) allocated SMF patients into four risk categories with different survival (P<0.0001): low (median survival NR; 133 patients), intermediate-1 (9.3 years, 95% CI: 8.1-NR; 245 patients), intermediate-2 (4.4 years, 95% CI: 3.2-7.9; 126 patients), and high risk (2 years, 95% CI: 1.7-3.9; 75 patients). Finally, we found that the MYSEC-PM represents the most appropriate tool for SMF decision-making to be used in clinical and trial settings.- Published
- 2017
- Full Text
- View/download PDF
33. European LeukemiaNet study on the reproducibility of bone marrow features in masked polycythemia vera and differentiation from essential thrombocythemia.
- Author
-
Kvasnicka HM, Orazi A, Thiele J, Barosi G, Bueso-Ramos CE, Vannucchi AM, Hasserjian RP, Kiladjian JJ, Gianelli U, Silver R, Mughal TI, and Barbui T
- Subjects
- Diagnosis, Differential, Europe, Female, Hemoglobins analysis, Humans, Janus Kinase 2 genetics, Male, Mutation, Polycythemia Vera blood, Polycythemia Vera genetics, Reproducibility of Results, Thrombocythemia, Essential blood, Thrombocythemia, Essential genetics, World Health Organization, Bone Marrow pathology, Polycythemia Vera pathology, Thrombocythemia, Essential pathology
- Abstract
The purpose of the study was to assess consensus and interobserver agreement among an international panel of six hematopathologists regarding characterization and reproducibility of bone marrow (BM) histologic features used to diagnose early stage myeloproliferative neoplasms, in particular differentiation of so-called masked/prodromal polycythemia vera (mPV) from JAK2-mutated essential thrombocythemia (ET). The six members of the hematopathology panel evaluated 98 BM specimens independently and in a blinded fashion without knowledge of clinical data. The specimens included 48 cases of mPV according to the originally published hemoglobin threshold values for this entity (male: 16.0-18.4 g/dL, female: 15.0-16.4 g/dL), 31 cases with overt PV according to the updated 2016 WHO criteria, and 19 control cases. The latter group included cases of JAK2-mutated ET, primary myelofibrosis, myelodysplastic syndrome, and various reactive conditions. Inter-rater agreement between the panelists was very high (overall agreement 92.6%, kappa 0.812), particularly with respect to separating mPV from ET. Virtually all cases of mPV were correctly classified as PV according to their BM morphology. In conclusion, a central blinded review of histology slides by six hematopathologists demonstrated that highly reproducible specific histological pattern characterize PV and confirmed the notion that there are no significant differences between mPV and overt PV in relation to BM morphology., (© 2017 Wiley Periodicals, Inc.)
- Published
- 2017
- Full Text
- View/download PDF
34. Enhanced calreticulin expression in red cells of polycythemia vera patients harboring the JAK2 V617F mutation.
- Author
-
Brusson M, Cochet S, Leduc M, Guillonneau F, Mayeux P, Peyrard T, Chomienne C, Le Van Kim C, Cassinat B, Kiladjian JJ, and El Nemer W
- Subjects
- Alleles, Amino Acid Substitution, Biomarkers, Calreticulin metabolism, Case-Control Studies, Erythrocyte Membrane metabolism, Female, Humans, Janus Kinase 2 metabolism, Male, Polycythemia Vera metabolism, Calreticulin genetics, Erythrocytes metabolism, Gene Expression Regulation, Janus Kinase 2 genetics, Mutation, Polycythemia Vera genetics
- Published
- 2017
- Full Text
- View/download PDF
35. Ruxolitinib reduces JAK2 p.V617F allele burden in patients with polycythemia vera enrolled in the RESPONSE study.
- Author
-
Vannucchi AM, Verstovsek S, Guglielmelli P, Griesshammer M, Burn TC, Naim A, Paranagama D, Marker M, Gadbaw B, and Kiladjian JJ
- Subjects
- Adult, Aged, Aged, 80 and over, Alleles, Cross-Over Studies, Female, Gene Frequency, Humans, Janus Kinase 2 antagonists & inhibitors, Janus Kinase 2 metabolism, Male, Middle Aged, Nitriles, Polycythemia Vera enzymology, Polycythemia Vera genetics, Pyrimidines, Time Factors, Treatment Outcome, Janus Kinase 2 genetics, Mutation, Missense, Polycythemia Vera drug therapy, Pyrazoles therapeutic use
- Abstract
In patients with polycythemia vera (PV), an elevated JAK2 p.V617F allele burden is associated with indicators of more severe disease (e.g., leukocytosis, splenomegaly, and increased thrombosis risk); however, correlations between allele burden reductions and clinical benefit in patients with PV have not been extensively evaluated in a randomized trial. This exploratory analysis from the multicenter, open-label, phase 3 Randomized Study of Efficacy and Safety in Polycythemia Vera With JAK Inhibitor INCB018424 Versus Best Supportive Care trial evaluated the long-term effect of ruxolitinib treatment on JAK2 p.V617F allele burden in patients with PV. Evaluable JAK2 p.V617F-positive patients randomized to ruxolitinib (n = 107) or best available therapy (BAT) who crossed over to ruxolitinib at week 32 (n = 97) had consistent JAK2 p.V617F allele burden reductions throughout the study. At all time points measured (up to weeks 208 [ruxolitinib-randomized] and 176 [ruxolitinib crossover]), mean changes from baseline over time in JAK2 p.V617F allele burden ranged from -12.2 to -40.0% (ruxolitinib-randomized) and -6.3 to -17.8% (ruxolitinib crossover). Complete or partial molecular response was observed in 3 patients (ruxolitinib-randomized, n = 2; ruxolitinib crossover, n = 1) and 54 patients (ruxolitinib-randomized, n = 33; ruxolitinib crossover, n = 20; BAT, n = 1), respectively. Among patients treated with interferon as BAT (n = 13), the mean maximal reduction in allele burden from baseline was 25.6% after crossover to ruxolitinib versus 6.6% before crossover. Collectively, the data from this exploratory analysis suggest that ruxolitinib treatment for up to 4 years provides progressive reductions in JAK2 p.V617F allele burden in patients with PV who are resistant to or intolerant of hydroxyurea. The relationship between allele burden changes and clinical outcomes in patients with PV remains unclear.
- Published
- 2017
- Full Text
- View/download PDF
36. Markers of iron deficiency in patients with polycythemia vera receiving ruxolitinib or best available therapy.
- Author
-
Verstovsek S, Harrison CN, Kiladjian JJ, Miller C, Naim AB, Paranagama DC, Habr D, and Vannucchi AM
- Subjects
- Biomarkers blood, Female, Humans, Male, Middle Aged, Nitriles, Prospective Studies, Pyrazoles pharmacology, Pyrimidines, Treatment Outcome, Iron Deficiencies, Polycythemia Vera drug therapy, Pyrazoles therapeutic use
- Abstract
Polycythemia vera (PV) is characterized by erythropoiesis and JAK2-activating mutations, with increased risks of morbidity and mortality. Most patients with PV are iron deficient, and treatment often includes hematocrit control with phlebotomy, which may exacerbate iron deficiency-associated complications. The phase 3 RESPONSE trial evaluated the JAK1/JAK2 inhibitor ruxolitinib (n=110) versus best available therapy (BAT; n=112) in patients with PV who were hydroxyurea-resistant/intolerant. Ruxolitinib was superior to BAT for hematocrit control, reduction in splenomegaly, and blood count normalization. This exploratory analysis, the first to evaluate iron status in a prospective study of patients with PV, investigated ruxolitinib effects on 7 serum iron markers and iron deficiency-related patient-reported outcomes (PRO). Among patients with evidence of baseline iron deficiency, ruxolitinib was associated with normalization of iron marker levels, compared with lesser improvement with BAT. Iron levels remained stable in ruxolitinib patients with normal iron levels at baseline. Regardless of baseline iron status, treatment with ruxolitinib was associated with improvements in concentration problems, cognitive function, dizziness, fatigue, headaches, and inactivity, although improvements were generally greater among patients with baseline iron deficiency. The improvements in iron deficiency markers and PROs observed with ruxolitinib are suggestive of clinical benefits that warrant further exploration., (Copyright © 2017 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
37. Changes in quality of life and disease-related symptoms in patients with polycythemia vera receiving ruxolitinib or standard therapy.
- Author
-
Mesa R, Verstovsek S, Kiladjian JJ, Griesshammer M, Masszi T, Durrant S, Passamonti F, Harrison CN, Pane F, Zachee P, Zhen H, Jones MM, Parasuraman S, Li J, Côté I, Habr D, and Vannucchi AM
- Subjects
- Adult, Aged, Female, Humans, Male, Middle Aged, Nitriles, Patient Reported Outcome Measures, Polycythemia Vera diagnosis, Protein Kinase Inhibitors administration & dosage, Protein Kinase Inhibitors adverse effects, Pyrazoles administration & dosage, Pyrazoles adverse effects, Pyrimidines, Treatment Outcome, Polycythemia Vera epidemiology, Polycythemia Vera therapy, Protein Kinase Inhibitors therapeutic use, Pyrazoles therapeutic use, Quality of Life, Standard of Care
- Abstract
Objectives: Polycythemia vera (PV)-related symptoms may not be adequately controlled with conventional therapy. This current analysis of the RESPONSE trial evaluated the effects of ruxolitinib compared with standard therapy on quality of life (QoL) and symptoms in patients with PV who were hydroxyurea resistant/intolerant., Methods: In the previously reported primary analysis, ruxolitinib achieved the primary composite endpoint of hematocrit control and ≥35% reduction in spleen volume at Week 32. The current analysis evaluated patient-reported outcomes using the European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire-Core 30 (EORTC QLQ-C30), the Myeloproliferative Neoplasm Symptom Assessment Form (MPN-SAF), the Pruritus Symptom Impact Scale (PSIS), and the Patient Global Impression of Change (PGIC)., Results: Compared with standard therapy, ruxolitinib was associated with greater improvements in global health status/QoL, functional subscales, and individual symptom scores of the EORTC QLQ-C30. At Week 32, more patients in the ruxolitinib arm (44%) achieved a ≥10-point improvement in global health status/QoL vs. standard therapy (9%). Improvements in MPN-SAF symptom scores were consistent with improvements in EORTC QLQ-C30, PSIS, and PGIC scores., Conclusions: Ruxolitinib provides clinically relevant improvements in QoL and ameliorates symptom burden in patients with PV who are hydroxyurea resistant/intolerant., (© 2016 The Authors. European Journal of Haematology Published by John Wiley & Sons Ltd.)
- Published
- 2016
- Full Text
- View/download PDF
38. Ruxolitinib versus best available therapy in patients with polycythemia vera: 80-week follow-up from the RESPONSE trial.
- Author
-
Verstovsek S, Vannucchi AM, Griesshammer M, Masszi T, Durrant S, Passamonti F, Harrison CN, Pane F, Zachee P, Kirito K, Besses C, Hino M, Moiraghi B, Miller CB, Cazzola M, Rosti V, Blau I, Mesa R, Jones MM, Zhen H, Li J, Francillard N, Habr D, and Kiladjian JJ
- Subjects
- Adult, Aged, Alleles, Combined Modality Therapy, Female, Follow-Up Studies, Gene Frequency, Hematocrit, Humans, Janus Kinases antagonists & inhibitors, Janus Kinases genetics, Male, Middle Aged, Molecular Targeted Therapy, Mutation, Nitriles, Polycythemia Vera diagnosis, Polycythemia Vera genetics, Protein Kinase Inhibitors administration & dosage, Protein Kinase Inhibitors adverse effects, Pyrazoles administration & dosage, Pyrazoles adverse effects, Pyrimidines, Treatment Outcome, Polycythemia Vera drug therapy, Protein Kinase Inhibitors therapeutic use, Pyrazoles therapeutic use
- Abstract
RESPONSE is an open-label phase 3 study evaluating the Janus kinase 1/Janus kinase 2 inhibitor ruxolitinib versus best available therapy for efficacy/safety in hydroxyurea-resistant or intolerant patients with polycythemia vera. This preplanned analysis occurred when all patients completed the Week 80 visit or discontinued. Objectives included evaluating the durability of the primary response (Week 32 phlebotomy-independent hematocrit control plus ≥35% spleen volume reduction), its components, and that of complete hematologic remission; and long-term safety. Median exposure was 111 weeks; 91/110 (82.7%) patients randomized to ruxolitinib remained on treatment. No patients continued best available therapy (98/112 [87.5%] crossed over to ruxolitinib, most at/soon after Week 32). At Week 32, primary response was achieved by 22.7% vs. 0.9% of patients randomized to ruxolitinib and best available therapy, respectively (hematocrit control, 60.0% vs. 18.8%; spleen response, 40.0% vs. 0.9%). The probability of maintaining primary and hematocrit responses for ≥80 weeks was 92% and 89%, respectively; 43/44 spleen responses were maintained until Week 80. Complete hematologic remission at Week 32 was achieved in 23.6% of ruxolitinib-randomized patients; the probability of maintaining complete hematologic remission for ≥80 weeks was 69%. Among ruxolitinib crossover patients, 79.2% were not phlebotomized, and 18.8% achieved a ≥35% reduction from baseline in spleen volume after 32 weeks of treatment. New or worsening hematologic laboratory abnormalities in ruxolitinib-treated patients were primarily grade 1/2 decreases in hemoglobin, lymphocytes, and platelets. The thromboembolic event rate per 100 patient-years was 1.8 with randomized ruxolitinib treatment vs. 8.2 with best available therapy. These data support ruxolitinib as an effective long-term treatment option for hydroxyurea-resistant or intolerant patients with polycythemia vera. This trial was registered at clinicaltrials.gov identifier: 01243944., (Copyright© Ferrata Storti Foundation.)
- Published
- 2016
- Full Text
- View/download PDF
39. Symptomatic Profiles of Patients With Polycythemia Vera: Implications of Inadequately Controlled Disease.
- Author
-
Geyer H, Scherber R, Kosiorek H, Dueck AC, Kiladjian JJ, Xiao Z, Slot S, Zweegman S, Sackmann F, Fuentes AK, Hernández-Maraver D, Döhner K, Harrison CN, Radia D, Muxi P, Besses C, Cervantes F, Johansson PL, Andreasson B, Rambaldi A, Barbui T, Bonatz K, Reiter A, Boyer F, Etienne G, Ianotto JC, Ranta D, Roy L, Cahn JY, Maldonado N, Barosi G, Ferrari ML, Gale RP, Birgegard G, Xu Z, Zhang Y, Sun X, Xu J, Zhang P, te Boekhorst PA, Commandeur S, Schouten H, Pahl HL, Griesshammer M, Stegelmann F, Lehmann T, Senyak Z, Vannucchi AM, Passamonti F, Samuelsson J, and Mesa RA
- Subjects
- Adult, Aged, Aged, 80 and over, Antineoplastic Agents administration & dosage, Fatigue etiology, Female, Fever etiology, Humans, Hydroxyurea administration & dosage, Janus Kinases antagonists & inhibitors, Janus Kinases genetics, Male, Middle Aged, Pain etiology, Palpation, Prognosis, Prospective Studies, Pruritus etiology, Severity of Illness Index, Sweating, Weight Loss, Antineoplastic Agents adverse effects, Hydroxyurea adverse effects, Phlebotomy, Polycythemia Vera complications, Polycythemia Vera drug therapy, Splenomegaly etiology
- Abstract
Purpose: Polycythemia vera (PV) is a myeloproliferative neoplasm (MPN) associated with disabling symptoms and a heightened risk of life-threatening complications. Recent studies have demonstrated the effectiveness of JAK inhibitor therapy in patients with PV patients who have a history of prior hydroxyurea (HU) use (including resistance or intolerance), phlebotomy requirements, and palpable splenomegaly. We aimed to determine how these features contribute alone and in aggregate to the PV symptom burden., Patients and Methods: Through prospective evaluation of 1,334 patients with PV who had characterized symptom burden, we assessed patient demographics, laboratory data, and the presence of splenomegaly by disease feature (ie, known HU use, known phlebotomy requirements, splenomegaly)., Results: The presence of each feature in itself is associated with a moderately high symptom burden (MPN symptom assessment form [SAF] total symptom score [TSS] range, 27.7 to 29.2) that persists independent of PV risk category. In addition, symptoms incrementally increase in severity with the addition of other features. Patients with PV who had all three features (PV-HUPS) faced the highest total score (MPN-SAF TSS, 32.5) but had similar individual symptom scores to patients with known HU use (PV-HU), known phlebotomy (PV-P), and splenomegaly (PV-S)., Conclusion: The results of this study suggest that patients with PV who have any one of the features in question (known HU use, known phlebotomy, or splenomegaly) have significant PV-associated symptoms. Furthermore, it demonstrates that many PV symptoms remain severe independent of the number of features present., (© 2015 by American Society of Clinical Oncology.)
- Published
- 2016
- Full Text
- View/download PDF
40. Ruxolitinib for the treatment of patients with polycythemia vera.
- Author
-
Kiladjian JJ, Winton EF, Talpaz M, and Verstovsek S
- Subjects
- Clinical Trials, Phase I as Topic, Clinical Trials, Phase II as Topic, Clinical Trials, Phase III as Topic, Humans, Janus Kinases antagonists & inhibitors, Nitriles, Polycythemia Vera metabolism, Pyrimidines, Treatment Outcome, Polycythemia Vera drug therapy, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Pyrazoles pharmacology, Pyrazoles therapeutic use
- Abstract
Polycythemia vera (PV) is a hematopoietic proliferative disorder associated with Janus-associated kinase/signal transducer and activator of transcription pathway dysregulation resulting in erythrocytosis and, possibly, leukocytosis and thrombocytosis. Patients diagnosed with PV experience a broad range of symptoms associated with a reduced quality of life, often develop splenomegaly, and have an increased risk of death compared with age-matched subjects without PV. Current treatment options, notably hydroxyurea, help with disease management; however, insufficient efficacy or progressive resistance occurs in some patients, highlighting the need for new treatment options. Ruxolitinib is an oral JAK1/JAK2 inhibitor that has been evaluated in Phase II and III clinical trials in patients with PV, who are intolerant of or resistant to hydroxyurea. In this setting, ruxolitinib treatment has demonstrated normalization of blood cell counts, reduction in splenomegaly and improvements in PV-related symptom burden.
- Published
- 2015
- Full Text
- View/download PDF
41. Lu/BCAM-mediated cell adhesion as biological marker of JAK2V617F activity in erythrocytes of polycythemia vera patients.
- Author
-
De Grandis M, Cassinat B, Kiladjian JJ, Chomienne C, and El Nemer W
- Subjects
- Alleles, Biomarkers analysis, Cell Adhesion, Cells, Cultured, Erythrocytes metabolism, Humans, Laminin chemistry, Mutation, Polycythemia Vera pathology, Polymorphism, Genetic, Cell Adhesion Molecules genetics, Erythrocytes pathology, Janus Kinase 2 genetics, Lutheran Blood-Group System genetics, Polycythemia Vera diagnosis, Polycythemia Vera genetics
- Published
- 2015
- Full Text
- View/download PDF
42. Ruxolitinib versus standard therapy for the treatment of polycythemia vera.
- Author
-
Vannucchi AM, Kiladjian JJ, Griesshammer M, Masszi T, Durrant S, Passamonti F, Harrison CN, Pane F, Zachee P, Mesa R, He S, Jones MM, Garrett W, Li J, Pirron U, Habr D, and Verstovsek S
- Subjects
- Adult, Aged, Aged, 80 and over, Antineoplastic Agents adverse effects, Blood Cell Count, Female, Humans, Hydroxyurea adverse effects, Hydroxyurea therapeutic use, Kaplan-Meier Estimate, Male, Middle Aged, Nitriles, Organ Size drug effects, Pyrazoles adverse effects, Pyrazoles pharmacology, Pyrimidines, Remission Induction, Spleen drug effects, Spleen pathology, Thrombocytopenia chemically induced, Antineoplastic Agents therapeutic use, Janus Kinases antagonists & inhibitors, Polycythemia Vera drug therapy, Pyrazoles therapeutic use
- Abstract
Background: Ruxolitinib, a Janus kinase (JAK) 1 and 2 inhibitor, was shown to have a clinical benefit in patients with polycythemia vera in a phase 2 study. We conducted a phase 3 open-label study to evaluate the efficacy and safety of ruxolitinib versus standard therapy in patients with polycythemia vera who had an inadequate response to or had unacceptable side effects from hydroxyurea., Methods: We randomly assigned phlebotomy-dependent patients with splenomegaly, in a 1:1 ratio, to receive ruxolitinib (110 patients) or standard therapy (112 patients). The primary end point was both hematocrit control through week 32 and at least a 35% reduction in spleen volume at week 32, as assessed by means of imaging., Results: The primary end point was achieved in 21% of the patients in the ruxolitinib group versus 1% of those in the standard-therapy group (P<0.001). Hematocrit control was achieved in 60% of patients receiving ruxolitinib and 20% of those receiving standard therapy; 38% and 1% of patients in the two groups, respectively, had at least a 35% reduction in spleen volume. A complete hematologic remission was achieved in 24% of patients in the ruxolitinib group and 9% of those in the standard-therapy group (P=0.003); 49% versus 5% had at least a 50% reduction in the total symptom score at week 32. In the ruxolitinib group, grade 3 or 4 anemia occurred in 2% of patients, and grade 3 or 4 thrombocytopenia occurred in 5%; the corresponding percentages in the standard-therapy group were 0% and 4%. Herpes zoster infection was reported in 6% of patients in the ruxolitinib group and 0% of those in the standard-therapy group (grade 1 or 2 in all cases). Thromboembolic events occurred in one patient receiving ruxolitinib and in six patients receiving standard therapy., Conclusions: In patients who had an inadequate response to or had unacceptable side effects from hydroxyurea, ruxolitinib was superior to standard therapy in controlling the hematocrit, reducing the spleen volume, and improving symptoms associated with polycythemia vera. (Funded by Incyte and others; RESPONSE ClinicalTrials.gov number, NCT01243944.).
- Published
- 2015
- Full Text
- View/download PDF
43. Use of the 46/1 haplotype to model JAK2(V617F) clonal architecture in PV patients: clonal evolution and impact of IFNα treatment.
- Author
-
Hasan S, Cassinat B, Droin N, Le Couedic JP, Favale F, Monte-Mor B, Lacout C, Fontenay M, Dosquet C, Chomienne C, Solary E, Villeval JL, Casadevall N, Kiladjian JJ, Vainchenker W, and Plo I
- Subjects
- Alleles, Clonal Evolution, Hematopoietic Stem Cells drug effects, Hematopoietic Stem Cells metabolism, Humans, Interferon-alpha therapeutic use, Models, Biological, Polycythemia Vera drug therapy, Haplotypes, Janus Kinase 2 genetics, Mutation, Polycythemia Vera genetics, Polycythemia Vera metabolism
- Published
- 2014
- Full Text
- View/download PDF
44. Revised response criteria for polycythemia vera and essential thrombocythemia: an ELN and IWG-MRT consensus project.
- Author
-
Barosi G, Mesa R, Finazzi G, Harrison C, Kiladjian JJ, Lengfelder E, McMullin MF, Passamonti F, Vannucchi AM, Besses C, Gisslinger H, Samuelsson J, Verstovsek S, Hoffman R, Pardanani A, Cervantes F, Tefferi A, and Barbui T
- Subjects
- Humans, Polycythemia Vera mortality, Societies, Medical, Thrombocythemia, Essential mortality, Consensus, International Agencies standards, Platelet Aggregation Inhibitors therapeutic use, Polycythemia Vera drug therapy, Practice Guidelines as Topic standards, Thrombocythemia, Essential drug therapy
- Abstract
Standardized response criteria to interpret and compare clinical trials are needed for approval of new therapeutic agents by regulatory agencies. The European LeukemiaNet (ELN) response criteria for essential thrombocythemia (ET) and polycythemia vera (PV) issued in 2009 have been widely adopted as end points in a number of recent clinical trials. However, evidence exists that they do not predict response or provide clinically relevant measures of benefit for the patients. This article presents revised recommendations for assessing response in ET and PV provided by a working group established by ELN and International Working Group-Myeloproliferative Neoplasms Research and Treatment. New definitions of complete and partial remission incorporate clinical, hematological, and histological response assessments that include a standardized symptom assessment form and consider absence of disease progression and vascular events. We anticipate that these criteria will be adopted widely to facilitate the development of new and more effective therapies for ET and PV.
- Published
- 2013
- Full Text
- View/download PDF
45. Treatment target in polycythemia vera.
- Author
-
McMullin MF, Harrison CN, and Kiladjian JJ
- Subjects
- Female, Humans, Male, Antineoplastic Agents therapeutic use, Cardiovascular Diseases mortality, Hematocrit, Hydroxyurea therapeutic use, Phlebotomy, Polycythemia Vera therapy, Thrombosis etiology
- Published
- 2013
- Full Text
- View/download PDF
46. Interferon and the treatment of polycythemia vera, essential thrombocythemia and myelofibrosis.
- Author
-
Silver RT, Kiladjian JJ, and Hasselbalch HC
- Subjects
- Humans, Interferons genetics, Polycythemia Vera genetics, Primary Myelofibrosis genetics, Recombinant Proteins genetics, Recombinant Proteins therapeutic use, Thrombocythemia, Essential genetics, Interferons therapeutic use, Polycythemia Vera drug therapy, Primary Myelofibrosis drug therapy, Thrombocythemia, Essential drug therapy
- Abstract
Recombinant IFN-α (rIFN-α) induces complete hematologic remissions in patients with myeloproliferative neoplasms (MPNs), but its use has been limited by side effects owing to the relatively high doses used. Now, low-dose rIFN-α is stressed, starting relatively early in the course of the MPNs. In polycythemia vera, this has resulted in a significant clinical, hematologic, morphologic and molecular response manifested by reduction in the JAK2(V617F) allele burden, sustained even after discontinuation of recombinant IFN. In essential thrombocythemia, platelet count reduction is prompt and durable without treatment for varying periods. In hypercellular primary myelofibrosis, rIFN-α has restored normal blood counts, reduced splenomegaly and induced morphologic marrow remissions. This article highlights our current use of rIFN-α in MPNs.
- Published
- 2013
- Full Text
- View/download PDF
47. Live and let (MPN cells) die!
- Author
-
Cassinat B and Kiladjian JJ
- Subjects
- Apoptosis drug effects, Drug Therapy, Combination, Hematopoietic Stem Cells pathology, Humans, Polycythemia Vera genetics, Polycythemia Vera pathology, Recombinant Proteins pharmacology, Hematopoietic Stem Cells drug effects, Imidazoles pharmacology, Interferon-alpha pharmacology, Piperazines pharmacology, Polycythemia Vera drug therapy, Polyethylene Glycols pharmacology, Proto-Oncogene Proteins c-mdm2 antagonists & inhibitors, Tumor Suppressor Protein p53 antagonists & inhibitors
- Published
- 2012
- Full Text
- View/download PDF
48. Coexistence of a myeloproliferative disorder and secondary polycythemia in the same patient.
- Author
-
Kouroupi E, Cassinat B, Schlageter MH, Dosquet C, Kiladjian JJ, and Chomienne C
- Subjects
- Aged, 80 and over, Comorbidity, Drug Resistance, Erythropoiesis drug effects, Erythropoietin blood, Humans, Hydroxyurea pharmacology, Hydroxyurea therapeutic use, Janus Kinase 2 genetics, Male, Mutation, Missense, Phlebotomy, Point Mutation, Polycystic Kidney, Autosomal Dominant complications, Polycystic Kidney, Autosomal Dominant diagnostic imaging, Polycythemia Vera drug therapy, Polycythemia Vera genetics, Ultrasonography, Polycythemia etiology, Polycythemia Vera complications
- Published
- 2012
- Full Text
- View/download PDF
49. Treatment of polycythemia vera with hydroxyurea and pipobroman: final results of a randomized trial initiated in 1980.
- Author
-
Kiladjian JJ, Chevret S, Dosquet C, Chomienne C, and Rain JD
- Subjects
- Adult, Aged, Aged, 80 and over, Cohort Studies, Female, Follow-Up Studies, Humans, Hydroxyurea adverse effects, Male, Middle Aged, Survival Analysis, Young Adult, Hydroxyurea therapeutic use, Pipobroman therapeutic use, Polycythemia Vera drug therapy
- Abstract
Purpose: The overall impact of hydroxyurea (HU) or pipobroman treatments on the long-term outcome of patients with polycythemia vera (PV) has not been assessed in randomized studies. We report final analyses from the French Polycythemia Study Group (FPSG) study, which randomly assigned HU versus pipobroman as first-line therapy in 285 patients younger than age 65 years., Patients and Methods: The full methodology has been described previously. FPSG results were updated with a median follow-up of 16.3 years. Statistical analysis was performed by using competing risks on the intention-to-treat population and according to main treatment received., Results: Median survival was 17 years for the whole cohort, 20.3 years for the HU arm, and 15.4 years for the pipobroman arm (P = .008) and differed significantly from that in the general population. At 10, 15, and 20 years, cumulative incidence of acute myeloid leukemia/myelodysplastic syndrome (AML/MDS) was 6.6%, 16.5%, and 24% in the HU arm and 13%, 34%, and 52% in the pipobroman arm (P = .004). Cumulative myelofibrosis incidence at 10, 15, and 20 years according to main treatment received was 15%, 24%, and 32% with HU versus 5%, 10%, and 21% with pipobroman (P = .02)., Conclusion: Data from this unique randomized trial comparing HU with another cytoreductive drug in PV showed that (1) survival of patients with PV treated with conventional agents differed from survival in the general population, (2) evolution to AML/MDS is the first cause of death, (3) pipobroman is leukemogenic and is unsuitable for first-line therapy, and (4) incidence of evolution to AML/MDS with HU is higher than previously reported, although consideration should be given to the natural evolution of PV.
- Published
- 2011
- Full Text
- View/download PDF
50. Molecular and clinical features of the myeloproliferative neoplasm associated with JAK2 exon 12 mutations.
- Author
-
Passamonti F, Elena C, Schnittger S, Skoda RC, Green AR, Girodon F, Kiladjian JJ, McMullin MF, Ruggeri M, Besses C, Vannucchi AM, Lippert E, Gisslinger H, Rumi E, Lehmann T, Ortmann CA, Pietra D, Pascutto C, Haferlach T, and Cazzola M
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Exons, Female, Humans, In Situ Hybridization, Leukocytosis etiology, Leukocytosis genetics, Male, Middle Aged, Polycythemia etiology, Polycythemia genetics, Polycythemia Vera complications, Polycythemia Vera mortality, Polymerase Chain Reaction, Thrombocytosis etiology, Thrombocytosis genetics, Young Adult, Janus Kinase 2 genetics, Mutation, Polycythemia Vera genetics
- Abstract
Although approximately 95% of patients with polycythemia vera (PV) harbor the V617F mutation in JAK2 exon 14, several mutations in exon 12 have been described in the remaining patients. We conducted a European collaborative study to define the molecular and clinical features of patients harboring these mutations. Overall, 106 PVs were recruited and 17 different mutations identified. Irrespective of the mutation, two-thirds of patients had isolated erythrocytosis, whereas the remaining subjects had erythrocytosis plus leukocytosis and/or thrombocytosis. Compared with JAK2 (V617F)-positive PV patients, those with exon 12 mutations had significantly higher hemoglobin level and lower platelet and leukocyte counts at diagnosis but similar incidences of thrombosis, myelofibrosis, leukemia, and death. In a multivariable analysis, age more than 60 years and prior thrombosis predicted thrombosis. These findings suggest that, despite the phenotypical difference, the outcome of JAK2 exon 12 mutations-positive PV is similar to that of JAK2 (V617F)-positive PV.
- Published
- 2011
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.