301
|
How I treat the blast phase of Philadelphia chromosome–negative myeloproliferative neoplasms. Blood 2018; 132:2339-2350. [DOI: 10.1182/blood-2018-03-785907] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 10/06/2018] [Indexed: 12/30/2022] Open
Abstract
The classic Philadelphia chromosome (Ph)–negative myeloproliferative neoplasms (MPNs) are a heterogeneous group of hematopoietic stem-cell diseases, characterized by activated JAK/STAT signaling and significant phenotypic mimicry, including a propensity for evolution to myeloid blast phase disease. Effective therapeutic options are limited for patients with Ph− MPNs in the blast phase (MPN-BP), and allogeneic stem-cell transplantation is the only known cure. Our increasing understanding of the molecular pathogenesis of this group of diseases, coupled with the increasing availability of targeted agents, has the potential to inform new subset-specific therapeutic approaches. Ultimately, progress in MPN-BP will hinge on prospective clinical and translational investigations with the goal of generating more effective treatment interventions. This case-based review highlights the molecular and clinical heterogeneities of MPN-BP and incorporates a treatment algorithm that underscores the importance of a personalized approach to this challenging group of diseases.
Collapse
|
302
|
O'Sullivan J, Mead AJ. Heterogeneity in myeloproliferative neoplasms: Causes and consequences. Adv Biol Regul 2018; 71:55-68. [PMID: 30528537 DOI: 10.1016/j.jbior.2018.11.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 11/19/2018] [Accepted: 11/20/2018] [Indexed: 01/09/2023]
Abstract
Myeloproliferative neoplasms (MPNs) are haematopoietic stem cell-derived clonal disorders characterised by proliferation of some or all myeloid lineages, depending on the subtype. MPNs are classically categorized into three disease subgroups; essential thrombocythaemia (ET), polycythaemia vera (PV) and primary myelofibrosis (PMF). The majority (>85%) of patients carry a disease-initiating or driver mutation, the most prevalent occurring in the janus kinase 2 gene (JAK2 V617F), followed by calreticulin (CALR) and myeloproliferative leukaemia virus (MPL) genes. Although these diseases are characterised by shared clinical, pathological and molecular features, one of the most challenging aspects of these disorders is the diverse clinical features which occur in each disease type, with marked variability in risks of disease complications and progression to leukaemia. A remarkable aspect of MPN biology is that the JAK2 V617F mutation, often occurring in the absence of additional mutations, generates a spectrum of phenotypes from asymptomatic ET through to aggressive MF, associated with a poor outcome. The mechanisms promoting MPN heterogeneity remain incompletely understood, but contributing factors are broad and include patient characteristics (gender, age, comorbidities and environmental exposures), additional somatic mutations, target disease-initiating cell, bone marrow microenvironment and germline genetic associations. In this review, we will address these in detail and discuss their role in heterogeneity of MPN disease phenotypes. Tailoring patient management according to the multiple different factors that influence disease phenotype may prove to be the most effective approach to modify the natural history of the disease and ultimately improve outcomes for patients.
Collapse
Affiliation(s)
- Jennifer O'Sullivan
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DS, United Kingdom.
| | - Adam J Mead
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DS, United Kingdom; NIHR Biomedical Research Centre, Churchill Hospital, Oxford, UK.
| |
Collapse
|
303
|
Pacilli A, Rotunno G, Mannarelli C, Fanelli T, Pancrazzi A, Contini E, Mannelli F, Gesullo F, Bartalucci N, Fattori GC, Paoli C, Vannucchi AM, Guglielmelli P. Mutation landscape in patients with myelofibrosis receiving ruxolitinib or hydroxyurea. Blood Cancer J 2018; 8:122. [PMID: 30467377 PMCID: PMC6250726 DOI: 10.1038/s41408-018-0152-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 09/27/2018] [Accepted: 10/15/2018] [Indexed: 12/12/2022] Open
Abstract
Refractoriness to ruxolitinib in patients with myelofibrosis (MF) was associated with clonal evolution; however, whether genetic instability is promoted by ruxolitinib remains unsettled. We evaluated the mutation landscape in 71 MF patients receiving ruxolitinib (n = 46) and hydroxyurea (n = 25) and correlated with response. A spleen volume response (SVR) was obtained in 57% and 12%, respectively. Highly heterogenous patterns of mutation acquisition/loss and/or changes of variant allele frequency (VAF) were observed in the 2 patient groups without remarkable differences. In patients receiving ruxolitinib, driver mutation type and high-molecular risk profile (HMR) at baseline did not impact on response rate, while HMR and sole ASXL1 mutations predicted for SVR loss at 3 years. In patients with SVR, a decrease of ≥ 20% of JAK2V617F VAF predicted for SVR duration. VAF increase of non-driver mutations and clonal progression at follow-up correlated with SVR loss and treatment discontinuation, and clonal progression also predicted for shorter survival. These data indicate that (i) ruxolitinib does not appreciably promote clonal evolution compared with hydroxyurea, (ii) VAF increase of pre-existing and/or (ii) acquisition of new mutations while on treatment correlated with higher rate of discontinuation and/or death, and (iv) reduction of JAK2V617F VAF associated with SVR duration.
Collapse
Affiliation(s)
- Annalisa Pacilli
- CRIMM, Centro di Ricerca e Innovazione per le Malattie Mieloproliferative, Azienda Ospedaliera Universitaria Careggi, Dipartimento di Medicina Sperimentale e Clinica, Università degli Studi, Firenze, Italy
| | - Giada Rotunno
- CRIMM, Centro di Ricerca e Innovazione per le Malattie Mieloproliferative, Azienda Ospedaliera Universitaria Careggi, Dipartimento di Medicina Sperimentale e Clinica, Università degli Studi, Firenze, Italy
| | - Carmela Mannarelli
- CRIMM, Centro di Ricerca e Innovazione per le Malattie Mieloproliferative, Azienda Ospedaliera Universitaria Careggi, Dipartimento di Medicina Sperimentale e Clinica, Università degli Studi, Firenze, Italy
| | | | - Alessandro Pancrazzi
- CRIMM, Centro di Ricerca e Innovazione per le Malattie Mieloproliferative, Azienda Ospedaliera Universitaria Careggi, Dipartimento di Medicina Sperimentale e Clinica, Università degli Studi, Firenze, Italy
| | - Elisa Contini
- CRIMM, Centro di Ricerca e Innovazione per le Malattie Mieloproliferative, Azienda Ospedaliera Universitaria Careggi, Dipartimento di Medicina Sperimentale e Clinica, Università degli Studi, Firenze, Italy
| | - Francesco Mannelli
- CRIMM, Centro di Ricerca e Innovazione per le Malattie Mieloproliferative, Azienda Ospedaliera Universitaria Careggi, Dipartimento di Medicina Sperimentale e Clinica, Università degli Studi, Firenze, Italy
| | - Francesca Gesullo
- CRIMM, Centro di Ricerca e Innovazione per le Malattie Mieloproliferative, Azienda Ospedaliera Universitaria Careggi, Dipartimento di Medicina Sperimentale e Clinica, Università degli Studi, Firenze, Italy
| | - Niccolò Bartalucci
- CRIMM, Centro di Ricerca e Innovazione per le Malattie Mieloproliferative, Azienda Ospedaliera Universitaria Careggi, Dipartimento di Medicina Sperimentale e Clinica, Università degli Studi, Firenze, Italy
| | | | - Chiara Paoli
- CRIMM, Centro di Ricerca e Innovazione per le Malattie Mieloproliferative, Azienda Ospedaliera Universitaria Careggi, Dipartimento di Medicina Sperimentale e Clinica, Università degli Studi, Firenze, Italy
| | - Alessandro M Vannucchi
- CRIMM, Centro di Ricerca e Innovazione per le Malattie Mieloproliferative, Azienda Ospedaliera Universitaria Careggi, Dipartimento di Medicina Sperimentale e Clinica, Università degli Studi, Firenze, Italy.
| | - Paola Guglielmelli
- CRIMM, Centro di Ricerca e Innovazione per le Malattie Mieloproliferative, Azienda Ospedaliera Universitaria Careggi, Dipartimento di Medicina Sperimentale e Clinica, Università degli Studi, Firenze, Italy
| |
Collapse
|
304
|
Vannucchi AM, Te Boekhorst PAW, Harrison CN, He G, Caramella M, Niederwieser D, Boyer-Perrard F, Duan M, Francillard N, Molloy B, Wroclawska M, Gisslinger H. EXPAND, a dose-finding study of ruxolitinib in patients with myelofibrosis and low platelet counts: 48-week follow-up analysis. Haematologica 2018; 104:947-954. [PMID: 30442723 PMCID: PMC6518918 DOI: 10.3324/haematol.2018.204602] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 11/13/2018] [Indexed: 12/28/2022] Open
Abstract
EEXPAND (phase Ib, dose-finding study) evaluated the starting dose of ruxolitinib in patients with myelofibrosis with baseline platelet counts of 50-99×109/L. The study consisted of dose-escalation and safety-expansion phases. Based on the baseline platelet counts, patients were assigned to stratum 1 (75-99×109/L) or stratum 2 (50-74×109/L), with the primary objective of determining the maximum safe starting dose (MSSD); key secondary objectives included safety and efficacy. At week 48 data cutoff (stratum 1, n=44; stratum 2, n=25), 24.6% (17 out of 69) of patients were still receiving treatment. The MSSD was established as ruxolitinib 10 mg twice daily in both strata. Thrombocytopenia [grade 4 (stratum 1, n=1; stratum 2, n=2)] was the only reported dose-limiting toxicity (study drug related) at 10 mg twice daily. In the MSSD cohort (stratum 1, n=20; stratum 2, n=18), adverse events (regardless of study drug relationship) led to treatment discontinuation in 15.0% and 33.3% of patients in stratum 1 and stratum 2, respectively, and dose adjustment/interruption in 45.0% and 66.7% of patients in stratum 1 and stratum 2, respectively. Three cases of on-treatment deaths were reported at the MSSD. Spleen response was achieved at week 48 in 33.3% and 30.0% of patients in stratum 1 and stratum 2, respectively. Improvements in the Total Symptom Score were also observed. In this study, ruxolitinib demonstrated acceptable tolerability in both the strata at the MSSD of 10 mg twice daily. (Registered at: clinicaltrials.gov identifier: 01317875).
Collapse
Affiliation(s)
- Alessandro M Vannucchi
- Center for Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliero Universitaria Careggi, University of Florence, Italy
| | | | - Claire N Harrison
- Guy's and St Thomas' NHS Foundation Trust, Guy's Hospital, London, UK
| | | | | | | | | | - Minghui Duan
- Peking Union Medical College Hospital, Beijing, China
| | | | | | | | - Heinz Gisslinger
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Austria
| |
Collapse
|
305
|
Treating early-stage myelofibrosis. Ann Hematol 2018; 98:241-253. [DOI: 10.1007/s00277-018-3526-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 10/12/2018] [Indexed: 01/03/2023]
|
306
|
Fowlkes S, Murray C, Fulford A, De Gelder T, Siddiq N. Myeloproliferative neoplasms (MPNs) - Part 1: An overview of the diagnosis and treatment of the "classical" MPNs. Can Oncol Nurs J 2018; 28:262-268. [PMID: 31148835 DOI: 10.5737/23688076284262268] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Myeloproliferative neoplasms (MPNs) are rare, yet potentially life-threatening, disorders caused by overproliferation of bone marrow stem cells. The symptom burden experienced by patients with the BCR-ABL1-negative MPNs (also referred to as the classical MPNs, i.e., essential thrombocythemia [ET], polycythemia vera [PV] and myelofibrosis [MF]) can be significant and can negatively impact quality of life (QOL). Since patients with these MPNs can live for several years, thereby requiring long-term treatment and follow-up, nurses play an essential role in communicating with these patients, assessing their symptoms, and educating them on treatments and self-management strategies that can reduce their symptom burden. This article, which is the first of a two-part series, was developed to provide nurses and other healthcare professionals with a review of the diagnosis and treatment of the most common classical MPNs. The second article in this series (also available in this issue) will provide nurses with practical guidance for managing the symptom burden associated with MPNs in order to help enhance the overall health and well-being of patients living with these disorders.
Collapse
Affiliation(s)
- Sabrina Fowlkes
- Chronic Myelogenous Leukemia/Myeloproliferative Neoplasms, Jewish General Hospital, Montreal, Quebec
| | - Cindy Murray
- Malignant Hematology, UHN Princess Margaret Cancer Centre, Toronto, Ontario
| | - Adrienne Fulford
- Hematology Oncology, London Health Sciences Centre, London, Ontario
| | - Tammy De Gelder
- Hamilton Health Sciences, Juravinski Hospital and Cancer Centre, Hamilton, Ontario
| | - Nancy Siddiq
- Clinical Nurse Specialist for Myeloproliferative Neoplasms (MPN), Princess Margaret Cancer Centre, Toronto, Ontario
| |
Collapse
|
307
|
Fowlkes S, Murray C, Fulford A, De Gelder T, Siddiq N. Myeloproliferative neoplasms (MPNs) - Part 2: A nursing guide to managing the symptom burden of MPNs. Can Oncol Nurs J 2018; 28:276-281. [PMID: 31148831 DOI: 10.5737/23688076284276281] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Myeloproliferative neoplasms (MPNs) are rare, yet potentially life-threatening, disorders caused by overproliferation of bone marrow stem cells. The symptom burden experienced by patients with the BCR-ABL1-negative MPNs (also referred to as the classical MPNs, i.e., essential thrombocythemia [ET], polycythemia vera [PV] and myelofibrosis [MF]) can be significant and can negatively impact quality of life (QOL). Since patients with these MPNs can live for several years, thereby requiring long-term treatment and follow-up, nurses play an essential role in communicating with these patients, assessing their symptoms, and educating them on treatments and self-management strategies that can reduce their symptom burden. This article, which is the second of a twopart series, was developed to provide nurses and other healthcare professionals with practical guidance for managing the symptom burden associated with the classical MPNs in order to help enhance their patients' overall health and QOL.
Collapse
Affiliation(s)
- Sabrina Fowlkes
- Chronic Myelogenous Leukemia/Myeloproliferative Neoplasms, Jewish General Hospital, Montreal, Quebec
| | - Cindy Murray
- Malignant Hematology, UHN Princess Margaret Cancer Centre, Toronto, Ontario
| | - Adrienne Fulford
- Hematology Oncology, London Health Sciences Centre, London, Ontario
| | - Tammy De Gelder
- Hamilton Health Sciences, Juravinski Hospital and Cancer Centre, Hamilton, Ontario
| | - Nancy Siddiq
- Clinical Nurse Specialist for Myeloproliferative Neoplasms (MPN), Princess Margaret Cancer Centre, Toronto, Ontario
| |
Collapse
|
308
|
Abikhair Burgo M, Roudiani N, Chen J, Santana AL, Doudican N, Proby C, Felsen D, Carucci JA. Ruxolitinib inhibits cyclosporine-induced proliferation of cutaneous squamous cell carcinoma. JCI Insight 2018; 3:120750. [PMID: 30185657 PMCID: PMC6171807 DOI: 10.1172/jci.insight.120750] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 07/26/2018] [Indexed: 01/05/2023] Open
Abstract
Organ transplant recipients (OTRs) on cyclosporine A (CSA) are prone to catastrophic cutaneous squamous cell carcinoma (SCC). Allograft-sparing, cancer-targeting systemic treatments are unavailable. We have shown increased risk for catastrophic SCC in OTRs via CSA-mediated induction of IL-22. Herein, we found that CSA drives SCC proliferation and tumor growth through IL-22 and JAK/STAT pathway induction. We in turn inhibited SCC growth with an FDA-approved JAK1/2 inhibitor, ruxolitinib. In human SCC cells, the greatest proliferative response to IL-22 and CSA treatment occurred in nonmetastasizing lines. IL-22 treatment upregulated JAK1 and STAT1/3 in A431 SCC cells. JAK/STAT pathway genes were highly expressed in tumors from a cohort of CSA-exposed OTRs and in SCC with high risk for metastasis. Compared with immunocompetent SCC, genes associated with innate immunity, response to DNA damage, and p53 regulation were differentially expressed in SCC from OTRs. In nude mice engrafted with human A431 cells, IL-22 and CSA treatment increased tumor growth and upregulated IL-22 receptor, JAK1, and STAT1/3 expression. Ruxolitinib treatment significantly reduced tumor volume and reversed the accelerated tumor growth. CSA and IL-22 exacerbate aggressive behavior in SCC. Targeting the IL-22 axis via selective JAK/STAT inhibition may reduce the progression of aggressive SCC in OTRs, without compromising immunosuppression.
Collapse
Affiliation(s)
- Melody Abikhair Burgo
- Ronald O. Perelman Department of Dermatology, New York University School of Medicine, New York, New York, USA
| | - Nazanin Roudiani
- Ronald O. Perelman Department of Dermatology, New York University School of Medicine, New York, New York, USA
| | - Jie Chen
- Institute for Pediatric Urology, Department of Urology, Weill Cornell Medicine, New York, New York, USA
| | - Alexis L. Santana
- Ronald O. Perelman Department of Dermatology, New York University School of Medicine, New York, New York, USA
| | - Nicole Doudican
- Ronald O. Perelman Department of Dermatology, New York University School of Medicine, New York, New York, USA
| | - Charlotte Proby
- Division of Cancer Research, Jacqui Wood Cancer Centre, University of Dundee, Dundee, Scotland, United Kingdom
| | - Diane Felsen
- Institute for Pediatric Urology, Department of Urology, Weill Cornell Medicine, New York, New York, USA
| | - John A. Carucci
- Ronald O. Perelman Department of Dermatology, New York University School of Medicine, New York, New York, USA
| |
Collapse
|
309
|
A phase 2 study of ruxolitinib in combination with azacitidine in patients with myelofibrosis. Blood 2018; 132:1664-1674. [PMID: 30185431 DOI: 10.1182/blood-2018-04-846626] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 08/27/2018] [Indexed: 12/29/2022] Open
Abstract
Ruxolitinib (RUX)-based combinations may provide benefit for patients with myelofibrosis (MF). In this open-label, nonrandomized, prospective phase 2 study, patients with MF initially received RUX twice per day continuously in 28-day cycles for the first 3 cycles. Azacitidine (AZA) 25 mg/m2 (days 1-5) was added starting with cycle 4 and could be subsequently increased to 75 mg/m2 (days 1-5). Forty-six patients were enrolled with a median follow-up of 28 months (range, 4-50+ months). An International Working Group for Myelofibrosis Research and Treatment (IWG-MRT) response was achieved in 33 patients (72%), with a median time to response of 1.8 months (range, 0.7-19.0 months). One-fourth (7 of 33) of the IWG-MRT responses occurred after the addition of AZA. A reduction of >50% in palpable spleen length at 24 weeks and at any time on the study was achieved in 62% and 71% of the evaluable patients, respectively. Among patients who achieved a >50% reduction in spleen length at 24 weeks, 95% had maintained it at 48 weeks. Notably, improvements in bone marrow reticulin fibrosis grade occurred in 57% of the patients at 24 months. Treatment discontinuations as a result of drug-related toxicities occurred in 4 patients (9%), all as a result of cytopenias. New onset grade 3 to 4 anemia and thrombocytopenia occurred in 35% and 26% of patients, respectively. RUX and AZA were safe, with encouraging spleen response rates and improvement in bone marrow fibrosis in patients with MF. This trial was registered at www.clinicaltrials.gov as #NCT01787487.
Collapse
|
310
|
Corey SJ, Jha J, McCart EA, Rittase WB, George J, Mattapallil JJ, Mehta H, Ognoon M, Bylicky MA, Summers TA, Day RM. Captopril mitigates splenomegaly and myelofibrosis in the Gata1 low murine model of myelofibrosis. J Cell Mol Med 2018; 22:4274-4282. [PMID: 29971909 PMCID: PMC6111823 DOI: 10.1111/jcmm.13710] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 05/05/2018] [Indexed: 01/06/2023] Open
Abstract
Allogeneic stem cell transplantation is currently the only curative therapy for primary myelofibrosis (MF), while the JAK2 inhibitor, ruxolitinib. Has been approved only for palliation. Other therapies are desperately needed to reverse life-threatening MF. However, the cell(s) and cytokine(s) that promote MF remain unclear. Several reports have demonstrated that captopril, an inhibitor of angiotensin-converting enzyme that blocks the production of angiotensin II (Ang II), mitigates fibrosis in heart, lung, skin and kidney. Here, we show that captopril can mitigate the development of MF in the Gata1low mouse model of primary MF. Gata1low mice were treated with 79 mg/kg/d captopril in the drinking water from 10 to 12 months of age. At 13 months of age, bone marrows were examined for fibrosis, megakaryocytosis and collagen expression; spleens were examined for megakaryocytosis, splenomegaly and collagen expression. Treatment of Gata1low mice with captopril in the drinking water was associated with normalization of the bone marrow cellularity; reduced reticulin fibres, splenomegaly and megakaryocytosis; and decreased collagen expression. Our findings suggest that treating with the ACE inhibitors captopril has a significant benefit in overcoming pathological changes associated with MF.
Collapse
Affiliation(s)
- Seth J. Corey
- Division of Pediatric Hematology, Oncology & Stem Cell TransplantationThe Massey Cancer Center at Virginia Commonwealth UniversityRichmondVAUSA
| | - Jyoti Jha
- Department of Pharmacology and Molecular TherapeuticsUniformed Services University of the Health SciencesBethesdaMDUSA
| | - Elizabeth A. McCart
- Department of Pharmacology and Molecular TherapeuticsUniformed Services University of the Health SciencesBethesdaMDUSA
| | - William B. Rittase
- Department of Pharmacology and Molecular TherapeuticsUniformed Services University of the Health SciencesBethesdaMDUSA
| | - Jeffy George
- Department of MicrobiologyUniformed Services University of the Health SciencesBethesdaMDUSA
| | - Joseph J. Mattapallil
- Department of MicrobiologyUniformed Services University of the Health SciencesBethesdaMDUSA
| | - Hrishikesh Mehta
- Division of Pediatric Hematology, Oncology & Stem Cell TransplantationThe Massey Cancer Center at Virginia Commonwealth UniversityRichmondVAUSA
| | - Mungunsukh Ognoon
- Department of AnesthesiologyUniformed Services University of the Health SciencesBethesdaMDUSA
| | - Michelle A. Bylicky
- Neuroscience Graduate ProgramUniformed Services University of the Health SciencesBethesdaMDUSA
| | - Thomas A. Summers
- Department of PathologyUniformed Services University of the Health SciencesBethesdaMDUSA
| | - Regina M. Day
- Department of Pharmacology and Molecular TherapeuticsUniformed Services University of the Health SciencesBethesdaMDUSA
| |
Collapse
|
311
|
Krauth MT, Burgstaller S, Buxhofer-Ausch V, Gastl G, Geissler K, Keil F, Krippl P, Melchardt T, Petzer A, Rumpold H, Sliwa T, Wöhrer S, Wölfler A, Gisslinger H. Ruxolitinib therapy for myelofibrosis in Austria : Consensus on therapy management. Wien Klin Wochenschr 2018; 130:495-504. [PMID: 30043249 PMCID: PMC6132876 DOI: 10.1007/s00508-018-1365-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 07/04/2018] [Indexed: 12/19/2022]
Abstract
The oral Janus associated kinase (JAK1/2) inhibitor ruxolitinib has been available for treatment of patients with intermediate or high-risk myelofibrosis in Europe since 2012. Since its introduction, the expertise of prescribing doctors with respect to ruxolitinib function, efficacy and adverse effects has consistently been augmented, resulting in therapy modalities that are better tailored to individual patients as well as in increased safety of the treatment. The present consensus on ruxolitinib therapy management has been elaborated by Austrian experts in myeloproliferative neoplasms in line with international treatment guidelines. Our recommendations aim to contribute to an improved management of patients with myelofibrosis treated with ruxolitinib.
Collapse
Affiliation(s)
- Maria-Theresa Krauth
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University Vienna, Vienna, Austria.
| | - Sonja Burgstaller
- Department of Internal Medicine IV, Klinikum Wels-Grieskirchen, Wels, Austria
| | - Veronika Buxhofer-Ausch
- Department of Internal Medicine I, Hematology with Stem Cell Transplantation, Hemostaseology and Medical Oncology, Ordensklinikum Linz-Elisabethinen, Linz, Austria
| | - Günther Gastl
- Department of Internal Medicine V, Division Hematology and Oncology, Medical University of Innsbruck, Innsbruck, Austria
| | - Klaus Geissler
- Fifth Medical Department, Hospital Hietzing, Vienna, Austria
| | - Felix Keil
- Third Medical Department, Hanusch Hospital, Vienna, Austria
| | - Peter Krippl
- Department of Internal Medicine, LKH Fürstenfeld, Krankenhausverbund Feldbach, Fürstenfeld, Austria
| | - Thomas Melchardt
- Third Medical Department, Division Hematology and Medical Oncology, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Andreas Petzer
- Department of Internal Medicine I, Hematology with Stem Cell Transplantation, Hemostaseology and Medical Oncology, Ordensklinikum Linz-Elisabethinen, Linz, Austria
| | - Holger Rumpold
- Internal Medicine II, Medical Oncology, Hematology, Gastroenterology and Rheumatology, Academic Teaching Hospital Feldkirch, Feldkirch, Austria
| | - Thamer Sliwa
- Third Medical Department, Hanusch Hospital, Vienna, Austria
| | - Stefan Wöhrer
- Permedio Center for Personalized Medicine and Sanatorium Hera Vienna, Vienna, Austria
| | - Albert Wölfler
- Division of Hematology, Medical University of Graz, Graz, Austria
| | - Heinz Gisslinger
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University Vienna, Vienna, Austria
| |
Collapse
|
312
|
Byrne M, Savani B, Savona MR. Leveraging JAK-STAT regulation in myelofibrosis to improve outcomes with allogeneic hematopoietic stem-cell transplant. Ther Adv Hematol 2018; 9:251-259. [PMID: 30210754 DOI: 10.1177/2040620718786437] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Accepted: 06/11/2018] [Indexed: 01/17/2023] Open
Abstract
Primary myelofibrosis (PMF) is a disease characterized by bone marrow fibrosis, extramedullary hematopoiesis, risk of transformation to acute myeloid leukemia, and a substantial symptom burden with diminished quality of life. Allogeneic hematopoietic cell transplantation (HCT) is the only curative option; however, disease relapse and graft versus host disease (GVHD) are significant barriers to long-term survival. The discovery of the JAK2 V617F mutation, and subsequent development of JAK inhibitors, resulted in improved survival and significant improvements in spleen volumes and symptom scores. Though the effect of JAK inhibition on transplant outcome is poorly understood, using JAK inhibition to achieve maximal response prior to HCT is standard practice at major centers. After allogeneic HCT, a significant proportion of patients with steroid-refractory GVHD have clinical responses to JAK inhibition. Targeting this pathway is a key component in the management of patients with PMF before and after allogeneic HCT.
Collapse
Affiliation(s)
- Michael Byrne
- Department of Medicine, Division of Hematology/Oncology, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, 777 Preston Research Building, 2200 Pierce Avenue, Nashville, TN 37232, USA
| | - Bipin Savani
- Vanderbilt-Ingram Cancer, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Michael R Savona
- Vanderbilt-Ingram Cancer, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
313
|
Crisà E, Cilloni D, Elli EM, Martinelli V, Palumbo GA, Pugliese N, Beggiato E, Frairia C, Cerrano M, Lanzarone G, Marchetti M, Mezzabotta M, Boccadoro M, Ferrero D. The use of erythropoiesis-stimulating agents is safe and effective in the management of anaemia in myelofibrosis patients treated with ruxolitinib. Br J Haematol 2018; 182:701-704. [DOI: 10.1111/bjh.15450] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Accepted: 05/14/2018] [Indexed: 12/20/2022]
Affiliation(s)
- Elena Crisà
- Haematology Division; A.O.U. Città della Salute e della Scienza di Torino; University of Turin; Torino Italy
| | - Daniela Cilloni
- Haematology Division; Department of Clinical and Biological Sciences; Ospedale San Luigi di Orbassano; University of Turin; Orbassano Italy
| | - Elena M. Elli
- Haematology Division; Ospedale San Gerardo; ASST Monza; Monza Italy
| | | | - Giuseppe A. Palumbo
- Haematology Division; A.O.U. Policlinico-V.Emanuele; University of Catania; Catania Italy
| | | | - Eloise Beggiato
- Haematology Division; A.O.U. Città della Salute e della Scienza di Torino; University of Turin; Torino Italy
| | - Chiara Frairia
- Haematology Division; Department of Translational Medicine; Azienda Ospedaliero-Universitaria Maggiore della Carita; Novara Italy
| | - Marco Cerrano
- Haematology Division; A.O.U. Città della Salute e della Scienza di Torino; University of Turin; Torino Italy
| | - Giuseppe Lanzarone
- Haematology Division; A.O.U. Città della Salute e della Scienza di Torino; University of Turin; Torino Italy
| | | | - Mauro Mezzabotta
- Haematology Division; Ordine Mauriziano - Ospedale Umberto I; Torino Italy
| | - Mario Boccadoro
- Haematology Division; A.O.U. Città della Salute e della Scienza di Torino; University of Turin; Torino Italy
| | - Dario Ferrero
- Haematology Division; A.O.U. Città della Salute e della Scienza di Torino; University of Turin; Torino Italy
| |
Collapse
|
314
|
Shahnaz Syed Abd Kadir S, Christopeit M, Wulf G, Wagner E, Bornhauser M, Schroeder T, Crysandt M, Mayer K, Jonas J, Stelljes M, Badbaran A, Ayuketang Ayuk F, Triviai I, Wolf D, Wolschke C, Kröger N. Impact of ruxolitinib pretreatment on outcomes after allogeneic stem cell transplantation in patients with myelofibrosis. Eur J Haematol 2018; 101:305-317. [PMID: 29791053 DOI: 10.1111/ejh.13099] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2018] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Ruxolitinib is the first approved drug for treatment of myelofibrosis, but its impact of outcome after allogeneic stem cell transplantation (ASCT) is unknown. PATIENTS AND METHODS We reported on 159 myelofibrosis patients (pts) with a median age of 59 years (r: 28-74) who received reduced intensity ASCT between 2000 and 2015 in eight German centers from related (n = 23), matched (n = 86) or mismatched (n = 50) unrelated donors. Forty-six (29%) patients received ruxolitinib at any time point prior to ASCT. The median daily dose of ruxolitinib was 30 mg (range 10-40 mg) and the median duration of treatment was 4.9 months (range 0.4-39.1 months). RESULTS Primary graft failure was seen in 2 pts (4%) in the ruxolitinib and 3 (2%) in the non-ruxolitinib group. Engraftment and incidence of acute GVHD grade II to IV and III/IV did not differ between groups (37% vs 39% and 19% vs 28%, respectively), nor did the non-relapse mortality at 2 years (23% vs 23%). A trend for lower risk of relapse was seen in the ruxolitinib group (9% vs 17%, P = .2), resulting in a similar 2 year DFS and OS (68% vs 60% and 73% vs 70%, respectively). No difference in any outcome variable could be seen between ruxolitinib responders and those who failed or lost response to ruxolitinib. CONCLUSIONS These results suggest that ruxolitinib pretreatment in myelofibrosis patient does not negatively influence outcome after allogeneic stem cell transplantation.
Collapse
Affiliation(s)
- Sharifah Shahnaz Syed Abd Kadir
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Haematology, Ampang Hospital, Selangor, Malaysia
| | - Maximilian Christopeit
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gerald Wulf
- Department of Hematology/Oncology, University Hospital Göttingen, Göttingen, Germany
| | - Eva Wagner
- Department for Hematology and Oncology, University Hospital Mainz, Mainz, Germany
| | - Martin Bornhauser
- Department for Hematology and Oncology, University Hospital Dresden, Dresden, Germany
| | - Thomas Schroeder
- Department for Hematology, Oncology and Clinical Immunology, University Hospital Duesseldorf, Düsseldorf, Germany
| | - Martina Crysandt
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Karin Mayer
- Medical Clinic 3, Oncology, Hematology, Immunoncology and Rheumatology, University Clinic Bonn (UKB), Bonn, Germany
| | - Julia Jonas
- Medical Clinic 3, Oncology, Hematology, Immunoncology and Rheumatology, University Clinic Bonn (UKB), Bonn, Germany
| | - Matthias Stelljes
- Department of Medicine A, Hematology and Oncology, University of Muenster, Muenster, Germany
| | - Anita Badbaran
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Francis Ayuketang Ayuk
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ioanna Triviai
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Dominik Wolf
- Medical Clinic 3, Oncology, Hematology, Immunoncology and Rheumatology, University Clinic Bonn (UKB), Bonn, Germany
| | - Christine Wolschke
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nicolaus Kröger
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
315
|
Takenaka K, Shimoda K, Akashi K. Recent advances in the diagnosis and management of primary myelofibrosis. Korean J Intern Med 2018; 33:679-690. [PMID: 29665657 PMCID: PMC6030412 DOI: 10.3904/kjim.2018.033] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 03/07/2018] [Indexed: 12/13/2022] Open
Abstract
Primary myelofibrosis (PMF) is a myeloproliferative neoplasm (MPN) in which dysregulation of the Janus kinase/signal transducers and activators of transcription (JAK/STAT) signaling pathways is the major pathogenic mechanism. Most patients with PMF carry a driver mutation in the JAK2, MPL (myeloproliferative leukemia), or CALR (calreticulin) genes. Mutations in epigenetic regulators and RNA splicing genes may also occur, and play critical roles in PMF disease progression. Based on revised World Health Organization diagnostic criteria for MPNs, both screening for driver mutations and bone marrow biopsy are required for a specific diagnosis. Clinical trials of JAK2 inhibitors for PMF have revealed significant efficacy for improving splenomegaly and constitutional symptoms. However, the currently available drug therapies for PMF do not improve survival. Although allogeneic stem cell transplantation is potentially curative, it is associated with substantial treatment-related morbidity and mortality. PMF is a heterogeneous disorder and decisions regarding treatments are often complicated, necessitating the use of prognostic models to determine the management of treatments for individual patients. This review focuses on the clinical aspects and outcomes of a cohort of Japanese patients with PMF, including discussion of recent advances in the management of PMF.
Collapse
Affiliation(s)
- Katsuto Takenaka
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
- Correspondence to Katsuto Takenaka, M.D. Division of Hematology, Oncology & Cardiovascular Medicine, Kyushu University Hospital, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan Tel: +81-92-642-5230 Fax: +81-92-642-5247 E-mail:
| | - Kazuya Shimoda
- Department of Gastroenterology and Hematology, University of Miyazaki Faculty of Medicine, Miyazaki, Japan
| | - Koichi Akashi
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| |
Collapse
|
316
|
LSD1 Inhibition Prolongs Survival in Mouse Models of MPN by Selectively Targeting the Disease Clone. Hemasphere 2018; 2:e54. [PMID: 31723778 PMCID: PMC6745991 DOI: 10.1097/hs9.0000000000000054] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 04/27/2018] [Indexed: 12/27/2022] Open
Abstract
Supplemental Digital Content is available in the text Despite recent advances, the myeloproliferative neoplasms (MPNs) are attended by considerable morbidity and mortality. Janus kinase (Jak) inhibitors such as ruxolitinib manage symptoms but do not substantially change the natural history of the disease. In this report, we show the effects of IMG-7289, an irreversible inhibitor of the epigenetically active lysine-specific demethylase 1 (LSD1) in mouse models of MPN. Once-daily treatment with IMG-7289 normalized or improved blood cell counts, reduced spleen volumes, restored normal splenic architecture, and reduced bone marrow fibrosis. Most importantly, LSD1 inhibition lowered mutant allele burden and improved survival. IMG-7289 selectively inhibited proliferation and induced apoptosis of JAK2V617F cells by concomitantly increasing expression and methylation of p53, and, independently, the pro-apoptotic factor PUMA and by decreasing the levels of its antiapoptotic antagonist BCLXL. These data provide a molecular understanding of the disease-modifying activity of the LSD1 inhibitor IMG-7289 that is currently undergoing clinical evaluation in patients with high-risk myelofibrosis. Moreover, low doses of IMG-7289 and ruxolitinib synergize in normalizing the MPN phenotype in mice, offering a rationale for investigating combination therapy.
Collapse
|
317
|
Palandri F, Palumbo GA, Iurlo A, Polverelli N, Benevolo G, Breccia M, Abruzzese E, Tiribelli M, Bonifacio M, Tieghi A, Isidori A, Martino B, Sgherza N, D'Adda M, Bergamaschi M, Crugnola M, Cavazzini F, Bosi C, Binotto G, Auteri G, Latagliata R, Ibatici A, Scaffidi L, Penna D, Cattaneo D, Soci F, Trawinska M, Russo D, Cuneo A, Semenzato G, Di Raimondo F, Aversa F, Lemoli RM, Heidel F, Reggiani MLB, Bartoletti D, Cavo M, Catani L, Vianelli N. Differences in presenting features, outcome and prognostic models in patients with primary myelofibrosis and post-polycythemia vera and/or post-essential thrombocythemia myelofibrosis treated with ruxolitinib. New perspective of the MYSEC-PM in a large multicenter study ⁎. Semin Hematol 2018; 55:248-255. [PMID: 30502854 DOI: 10.1053/j.seminhematol.2018.05.013] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 05/10/2018] [Accepted: 05/19/2018] [Indexed: 02/02/2023]
Abstract
Recently, the myelofibrosis secondary to PV and ET prognostic model (MYSEC-PM) was introduced to assess prognosis in myelofibrosis (MF) secondary to polycythemia vera and essential thrombocythemia (post-PV and post-ET MF), replacing the International Prognostic Scoring System (IPSS) and/or Dynamic IPSS (DIPSS) that was applied for primary MF (PMF). In a cohort of 421 ruxolitinib (RUX)-treated patients (post-PV and post-ET MF: 44.2%), we evaluated the following: (1) disease phenotype, responses, and toxicity to RUX; and (2) performance of the MYSEC-PM in post-PV or post-ET MF. While the IPSS failed to correctly stratify post-PV or post-ET MF patients at diagnosis, the MYSEC-PM identified 4 risk categories projected at significantly different survival probability (P < .001). Additionally, the MYSEC-PM maintained a prognostic value in post-PV and post-ET MF also when used over time, at RUX start. Notably, the MYSEC-PM reclassified 41.8% and 13.6% of patients into a lower and higher risk category, respectively. Finally, patients at intermediate-1 risk had significantly higher spleen responses and lower hematological toxicities compared to higher risk patients. Compared to PMF, post-PV and post-ET MF presented a more hyperproliferative disease, with higher leukocyte and/or platelet count and hemoglobin levels both at diagnosis and at RUX start. Despite comparable response rates, post-PV and post-ET MF had lower rate of RUX-induced anemia and thrombocytopenia at 3 and 6 months. The study validates MYSEC-PM in post-PV and post-ET MF prognostication. Post-PV or post-ET MF represents a separate entity compared to PMF in terms of clinical manifestations and toxicity to RUX.
Collapse
Affiliation(s)
- Francesca Palandri
- Institute of Hematology "L. and A. Seràgnoli", Sant'Orsola-Malpighi University Hospital, Bologna, Italy.
| | - Giuseppe A Palumbo
- Division of Hematology, AOU "Policlinico-V. Emanuele", University of Catania, Catania, Italy
| | - Alessandra Iurlo
- Hematology Division, IRCCS Ca' Granda - Maggiore Policlinico Hospital Foundation, University of Milan, Milan, Italy
| | - Nicola Polverelli
- Unit of Blood Diseases and Stem Cells Transplantation, Department of Clinical and Experimental Sciences, University of Brescia, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Giulia Benevolo
- Division of Hematology, Città della Salute e della Scienza Hospital, Torino, Italy
| | - Massimo Breccia
- Division of Cellular Biotechnologies and Hematology, Sapienza University, Roma, Italy
| | | | - Mario Tiribelli
- Division of Hematology and BMT, Azienda Sanitaria Universitaria Integrata di Udine, Udine, Italy
| | | | - Alessia Tieghi
- Division of Hematology, Azienda Ospedaliera-IRCCS Arcispedale Santa Maria Nuova, Reggio Emilia, Italy
| | - Alessandro Isidori
- Hematology and Stem Cell Transplant Center, AORMN Hospital, Pesaro, Italy
| | - Bruno Martino
- Division of Hematology, Azienda Ospedaliera "Bianchi Melacrino Morelli", Reggio Calabria, Italy
| | - Nicola Sgherza
- Division of Hematology, Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Mariella D'Adda
- Division of Hematology, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Micaela Bergamaschi
- Clinic of Hematology, Department of Internal Medicine (DiMI), IRCCS AOU San Martino-IST, Genova, Italy
| | - Monica Crugnola
- Division of Hematology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | | | - Costanza Bosi
- Division of Hematology, Azienda Ospedaliera of Piacenza, Piacenza, Italy
| | - Gianni Binotto
- Unit of Hematology and Clinical Immunology, University of Padova, Padova, Italy
| | - Giuseppe Auteri
- Institute of Hematology "L. and A. Seràgnoli", Sant'Orsola-Malpighi University Hospital, Bologna, Italy
| | - Roberto Latagliata
- Division of Cellular Biotechnologies and Hematology, Sapienza University, Roma, Italy
| | - Adalberto Ibatici
- Division of Hematology and Bone Marrow Transplant, IRCCS San Martino-IST, Genova, Italy
| | - Luigi Scaffidi
- Department of Hematology, University of Verona, Verona, Italy
| | - Domenico Penna
- Division of Hematology and BMT, Azienda Sanitaria Universitaria Integrata di Udine, Udine, Italy
| | - Daniele Cattaneo
- Hematology Division, IRCCS Ca' Granda - Maggiore Policlinico Hospital Foundation, University of Milan, Milan, Italy
| | - Francesco Soci
- Division of Hematology, Azienda Ospedaliera-IRCCS Arcispedale Santa Maria Nuova, Reggio Emilia, Italy
| | | | - Domenico Russo
- Unit of Blood Diseases and Stem Cells Transplantation, Department of Clinical and Experimental Sciences, University of Brescia, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Antonio Cuneo
- Division of Hematology, University of Ferrara, Ferrara, Italy
| | | | - Francesco Di Raimondo
- Division of Hematology, AOU "Policlinico-V. Emanuele", University of Catania, Catania, Italy
| | - Franco Aversa
- Division of Hematology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Roberto M Lemoli
- Clinic of Hematology, Department of Internal Medicine (DiMI), IRCCS AOU San Martino-IST, Genova, Italy
| | - Florian Heidel
- Department of Internal Medicine II, Hematology and Oncology, Friedrich-Schiller-University Medical Center, Jena, Germany
| | - Maria L B Reggiani
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Cardiology, Sant'Orsola-Malpighi University Hospital, Bologna, Italy
| | - Daniela Bartoletti
- Institute of Hematology "L. and A. Seràgnoli", Sant'Orsola-Malpighi University Hospital, Bologna, Italy
| | - Michele Cavo
- Institute of Hematology "L. and A. Seràgnoli", Sant'Orsola-Malpighi University Hospital, Bologna, Italy
| | - Lucia Catani
- Institute of Hematology "L. and A. Seràgnoli", Sant'Orsola-Malpighi University Hospital, Bologna, Italy
| | - Nicola Vianelli
- Institute of Hematology "L. and A. Seràgnoli", Sant'Orsola-Malpighi University Hospital, Bologna, Italy
| |
Collapse
|
318
|
How I treat myelofibrosis after failure of JAK inhibitors. Blood 2018; 132:492-500. [PMID: 29866811 DOI: 10.1182/blood-2018-02-785923] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 05/28/2018] [Indexed: 12/16/2022] Open
Abstract
The introduction of JAK inhibitors, leading to regulatory approval of ruxolitinib, represents a major therapeutic advance in myelofibrosis (MF). Most patients experience reduction in splenomegaly and improved quality of life from symptom improvement. It is a paradox, however, that, despite inhibition of signaling downstream of disease-related driver mutations, JAK inhibitor treatment is not associated with consistent molecular or pathologic responses in MF. Furthermore, there are important limitations to JAK inhibitor therapy including development of dose-limiting cytopenias and/or nonhematological toxicities such as neuropathy or opportunistic infections. Over half of the patients discontinue treatment within 3 years of starting treatment. Although data are sparse, clinical outcome after JAK inhibitor "failure" is likely poor; consequently, it is important to understand patterns of failure to select appropriate salvage treatment(s). An algorithmic approach, particularly one that incorporates cytogenetics/molecular data, is most helpful in selecting stem cell transplant candidates. Treatment of transplant-ineligible patients relies on a problem-based approach that includes use of investigational drugs, or consideration of splenectomy or radiotherapy. Data from early phase ruxolitinib combination studies, despite promising preclinical data, have not shown clear benefit over monotherapy thus far. Development of effective treatment strategies for MF patients failing JAK inhibitors remains a major unmet need.
Collapse
|
319
|
Al-Fayoumi S, Hashiguchi T, Shirakata Y, Mascarenhas J, Singer JW. Pilot study of the antifibrotic effects of the multikinase inhibitor pacritinib in a mouse model of liver fibrosis. J Exp Pharmacol 2018; 10:9-17. [PMID: 29785143 PMCID: PMC5953271 DOI: 10.2147/jep.s150729] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Background Fibrotic diseases result from an exuberant response to chronic inflammation. Myelofibrosis is the end result of inflammation in bone, caused by an inflammatory process triggered by production of abnormal myeloid cells driven by mutations affecting the JAK-STAT pathway. Inflammatory cytokine overproduction leads to increased mesenchymal cell proliferation, culminating in fibrosis. Although JAK2 inhibitors, such as the JAK1/2 inhibitor ruxolitinib and the JAK2/FLT3/CSF1R/IRAK1 inhibitor pacritinib suppress abnormal clone expansion in myelofibrosis, ruxolitinib does not appear to prevent or reverse bone-marrow fibrosis in most patients. In two Phase III clinical trials, pacritinib, however, demonstrated improvements in platelet counts and hemoglobin and reductions in transfusion burden in some patients with baseline cytopenias, suggesting it may improve bone-marrow function. Unlike ruxolitinib, pacritinib suppresses signaling through IRAK1, a key control point for inflammatory and fibrotic signaling. Purpose To investigate potential antifibrotic effects of pacritinib in an animal model of liver fibrosis relevant to the observed course of human disease. Methods Pacritinib, negative control (vehicle), and positive control (the angiotensin 2-receptor antagonist and PPARγ partial agonist telmisartan) were assessed in the murine Stelic animal model, which mimics the clinically observed progression from hepatic steatosis to nonalcoholic steatohepatitis, liver fibrosis, and hepatocellular carcinoma. Histopathological analysis used hematoxylin and eosin staining. Body and liver weight changes, nonalcoholic fatty-liver disease activity scores, and plasma cytokeratin 18 fragment levels (a biomarker of hepatic necrosis) were measured. Results Pacritinib-treated mice had significantly (P<0.01) reduced fibrotic areas in liver compared to vehicle control and significantly (P<0.05) lower levels of CK18. The antifibrotic effect of pacritinib was comparable to that of telmisartan, but without significant effects on fat accumulation. Conclusion These results, the first to demonstrate hepatic antifibrotic effects for pacritinib in an animal model of liver disease, provide preliminary support for potential clinical applications of pacritinib in fibrotic diseases other than myelofibrosis.
Collapse
Affiliation(s)
| | | | | | - John Mascarenhas
- Tish Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | |
Collapse
|
320
|
von Lilienfeld-Toal M, Maschmeyer G. Challenges in Infectious Diseases for Haematologists. Oncol Res Treat 2018; 41:406-410. [PMID: 29734194 DOI: 10.1159/000487439] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 02/06/2018] [Indexed: 01/11/2023]
Abstract
Infections remain a threat for patients with haematological malignancies. In accordance with the European Hematology Association roadmap we provide a concise overview regarding the most relevant current challenges in infectious diseases for haematologists. These include bacterial infections and the need for antibiotic stewardship as well as infections with community-acquired respiratory viruses, infections in patients receiving targeted therapies, re-activations of latent infections and vaccination strategies. The following review intends to summarise the most relevant information for clinicians currently caring for patients with haematological malignancies. Recommendations given are based on the guidelines published by the Infectious Diseases Working Party of the German Society of Haematology and Medical Oncology.
Collapse
|
321
|
Bose P, Swaminathan M. Janus kinase inhibition and symptom control in myeloproliferative neoplasms. Curr Med Res Opin 2018; 34:935-937. [PMID: 29415577 PMCID: PMC6329289 DOI: 10.1080/03007995.2018.1438999] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 02/07/2018] [Indexed: 10/18/2022]
Affiliation(s)
- Prithviraj Bose
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Mahesh Swaminathan
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
322
|
Aboul-Fettouh N, Nijhawan RI. Aggressive squamous cell carcinoma in a patient on the Janus kinase inhibitor ruxolitinib. JAAD Case Rep 2018; 4:455-457. [PMID: 29984281 PMCID: PMC6031591 DOI: 10.1016/j.jdcr.2018.01.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Affiliation(s)
- Nader Aboul-Fettouh
- Department of Dermatology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Rajiv I Nijhawan
- Department of Dermatology, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
323
|
Zacharaki D, Ghazanfari R, Li H, Lim HC, Scheding S. Effects of JAK1/2 inhibition on bone marrow stromal cells of myeloproliferative neoplasm (MPN) patients and healthy individuals. Eur J Haematol 2018; 101:57-67. [DOI: 10.1111/ejh.13079] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/29/2018] [Indexed: 01/03/2023]
Affiliation(s)
- Dimitra Zacharaki
- Division of Molecular Hematology & Lund Stem Cell Center; Lund University; Lund Sweden
| | - Roshanak Ghazanfari
- Division of Molecular Hematology & Lund Stem Cell Center; Lund University; Lund Sweden
| | - Hongzhe Li
- Division of Molecular Hematology & Lund Stem Cell Center; Lund University; Lund Sweden
| | - Hooi Ching Lim
- Division of Molecular Hematology & Lund Stem Cell Center; Lund University; Lund Sweden
| | - Stefan Scheding
- Division of Molecular Hematology & Lund Stem Cell Center; Lund University; Lund Sweden
- Department of Hematology; Skåne University Hospital Lund; Lund Sweden
| |
Collapse
|
324
|
Gleitz HF, Kramann R, Schneider RK. Understanding deregulated cellular and molecular dynamics in the haematopoietic stem cell niche to develop novel therapeutics for bone marrow fibrosis. J Pathol 2018; 245:138-146. [PMID: 29570794 PMCID: PMC5969225 DOI: 10.1002/path.5078] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 03/01/2018] [Accepted: 03/15/2018] [Indexed: 01/06/2023]
Abstract
Bone marrow fibrosis is the continuous replacement of blood-forming cells in the bone marrow with excessive scar tissue, leading to failure of the body to produce blood cells and ultimately to death. Myofibroblasts are fibrosis-driving cells and are well characterized in solid organ fibrosis, but their role and cellular origin in bone marrow fibrosis have remained obscure. Recent work has demonstrated that Gli1+ and leptin receptor+ mesenchymal stromal cells are progenitors of fibrosis-causing myofibroblasts in the bone marrow. Genetic ablation or pharmacological inhibition of Gli1+ mesenchymal stromal cells ameliorated fibrosis in mouse models of myelofibrosis. Conditional deletion of the platelet-derived growth factor (PDGF) receptor-α (PDGFRA) gene (Pdgfra) and inhibition of PDGFRA by imatinib in leptin receptor+ stromal cells suppressed their expansion and ameliorated bone marrow fibrosis. Understanding the cellular and molecular mechanisms in the haematopoietic stem cell niche that govern the mesenchymal stromal cell-to-myofibroblast transition and myofibroblast expansion will be critical to understand the pathogenesis of bone marrow fibrosis in both malignant and non-malignant conditions, and will guide the development of novel therapeutics. In this review, we summarize recent discoveries of mesenchymal stromal cells as part of the haematopoietic niche and as myofibroblast precursors, and discuss potential therapeutic strategies in the specific targeting of fibrotic transformation in bone marrow fibrosis. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Hélène Fe Gleitz
- Department of Haematology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Rafael Kramann
- Division of Nephrology and Clinical Immunology, RWTH Aachen University, Aachen, Germany
| | - Rebekka K Schneider
- Department of Haematology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands.,Department of Haematology, Oncology, Haemostaseology, and Stem Cell Transplantation, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
325
|
Abstract
The IL-6/JAK/STAT3 pathway is aberrantly hyperactivated in many types of cancer, and such hyperactivation is generally associated with a poor clinical prognosis. In the tumour microenvironment, IL-6/JAK/STAT3 signalling acts to drive the proliferation, survival, invasiveness, and metastasis of tumour cells, while strongly suppressing the antitumour immune response. Thus, treatments that target the IL-6/JAK/STAT3 pathway in patients with cancer are poised to provide therapeutic benefit by directly inhibiting tumour cell growth and by stimulating antitumour immunity. Agents targeting IL-6, the IL-6 receptor, or JAKs have already received FDA approval for the treatment of inflammatory conditions or myeloproliferative neoplasms and for the management of certain adverse effects of chimeric antigen receptor T cells, and are being further evaluated in patients with haematopoietic malignancies and in those with solid tumours. Novel inhibitors of the IL-6/JAK/STAT3 pathway, including STAT3-selective inhibitors, are currently in development. Herein, we review the role of IL-6/JAK/STAT3 signalling in the tumour microenvironment and the status of preclinical and clinical investigations of agents targeting this pathway. We also discuss the potential of combining IL-6/JAK/STAT3 inhibitors with currently approved therapeutic agents directed against immune-checkpoint inhibitors.
Collapse
Affiliation(s)
- Daniel E. Johnson
- Department of Otolaryngology – Head and Neck Surgery, University of California, San Francisco, CA, USA
| | - Rachel A. O’Keefe
- Department of Otolaryngology – Head and Neck Surgery, University of California, San Francisco, CA, USA
| | - Jennifer R. Grandis
- Department of Otolaryngology – Head and Neck Surgery, University of California, San Francisco, CA, USA
| |
Collapse
|
326
|
Treatment With JAK Inhibitors in Myelofibrosis Patients Nullifies the Prognostic Impact of Unfavorable Cytogenetics. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2018; 18:e201-e210. [PMID: 29574002 DOI: 10.1016/j.clml.2018.02.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 02/26/2018] [Indexed: 11/24/2022]
Abstract
INTRODUCTION In the era before Janus kinase (JAK) inhibitors, cytogenetic information was used to predict survival in myelofibrosis patients. However, the prognostic value of cytogenetics in the setting of JAK inhibitor therapy remains unknown. PATIENTS AND METHODS We performed a retrospective analysis of 180 patients with bone marrow biopsy-proven myelofibrosis from 3 US academic medical centers. We fit Cox proportional hazards models for overall survival and transformation-free survival on the bases of 3 factors: JAK inhibitor therapy as a time-dependent covariate, dichotomized cytogenetic status (favorable vs. unfavorable), and statistical interaction between the two. The median follow-up time was 37.1 months. RESULTS Among patients treated with best available therapy, unfavorable cytogenetic status was associated with decreased survival (hazard ratio = 2.31; P = .025). At initiation of JAK inhibitor therapy, unfavorable cytogenetics was (nonsignificantly) associated with increased survival compared to favorable cytogenetics (hazard ratio = 0.292; P = .172). The ratio of hazard ratios was 0.126 (P = .034). These findings were similar after adjusting for standard clinical prognostic factors as well as when measured against transformation-free survival. CONCLUSION The initiation of JAK inhibitor therapy appears to change the association between cytogenetics and overall survival. There was little difference in survival between treatment types in patients with favorable cytogenetics. However, the use of JAK inhibitor therapy among patients with unfavorable cytogenetics was not associated with worse survival compared to favorable cytogenetics. Our analyses suggest that initiation of JAK inhibitor therapy nullifies the negative prognostic implication of unfavorable cytogenetics established in the pre-JAK inhibitor therapy era.
Collapse
|
327
|
The identification of fibrosis-driving myofibroblast precursors reveals new therapeutic avenues in myelofibrosis. Blood 2018; 131:2111-2119. [PMID: 29572380 DOI: 10.1182/blood-2018-02-834820] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 03/21/2018] [Indexed: 12/14/2022] Open
Abstract
Myofibroblasts are fibrosis-driving cells and are well characterized in solid organ fibrosis, but their role and cellular origin in bone marrow fibrosis remains obscure. Recent work has demonstrated that Gli1+ and LepR+ mesenchymal stromal cells (MSCs) are progenitors of fibrosis-causing myofibroblasts in the bone marrow. Genetic ablation of Gli1+ MSCs or pharmacologic targeting of hedgehog (Hh)-Gli signaling ameliorated fibrosis in mouse models of myelofibrosis (MF). Moreover, pharmacologic or genetic intervention in platelet-derived growth factor receptor α (Pdgfrα) signaling in Lepr+ stromal cells suppressed their expansion and ameliorated MF. Improved understanding of cellular and molecular mechanisms in the hematopoietic stem cell niche that govern the transition of MSCs to myofibroblasts and myofibroblast expansion in MF has led to new paradigms in the pathogenesis and treatment of MF. Here, we highlight the central role of malignant hematopoietic clone-derived megakaryocytes in reprogramming the hematopoietic stem cell niche in MF with potential detrimental consequences for hematopoietic reconstitution after allogenic stem cell transplantation, so far the only therapeutic approach in MF considered to be curative. We and others have reported that targeting Hh-Gli signaling is a therapeutic strategy in solid organ fibrosis. Data indicate that targeting Gli proteins directly inhibits Gli1+ cell proliferation and myofibroblast differentiation, which results in reduced fibrosis severity and improved organ function. Although canonical Hh inhibition (eg, smoothened [Smo] inhibition) failed to improve pulmonary fibrosis, kidney fibrosis, or MF, the direct inhibition of Gli proteins ameliorated fibrosis. Therefore, targeting Gli proteins directly might be an interesting and novel therapeutic approach in MF.
Collapse
|
328
|
Abstract
INTRODUCTION The outlook for patients with myeloproliferative neoplasms, particularly myelofibrosis, has improved in recent years, with greater understanding of the pathogenesis and the subsequent development of a plethora of new agents. Areas covered: This article will discuss some of the advances in the field in recent years and explore in greater detail some of the most advanced emerging agents as well as those with greatest potential. An extensive literature review has been performed to identify recent clinical trials and any relevant pre-clinical work. Expert commentary: Important discoveries regarding molecular pathogenesis have led to advances in diagnostic algorithms, prognosis and ultimately also treatment strategies. However, the therapeutic armamentarium for MPN is still largely inadequate to cope with significant challenges including normalization of life span, reduction of cardiovascular complications, prevention of hematological progression and improved quality of life. Sadly, no currently available drugs have shown clear evidence of disease-modifying activity and results of early phase I and II clinical trials have been quite disappointing to date, with toxicities sometimes limiting and a lack of meaningful biological surrogate end points.
Collapse
Affiliation(s)
- Patrick M Harrington
- a Department of Haematology , Guys and St Thomas' NHS Foundation Trust , London , UK
| | - Claire N Harrison
- a Department of Haematology , Guys and St Thomas' NHS Foundation Trust , London , UK
| |
Collapse
|
329
|
Jin J, Du X, Zhou DB, Li JM, Li JY, Hou M, Liu T, Wu DP, Hu Y, Xiao ZJ. [Efficacy and safety of JAK inhibitor ruxolitinib in Chinese patients with myelofibrosis: results of a 1-year follow-up of A2202]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2018; 37:858-863. [PMID: 27801315 PMCID: PMC7364878 DOI: 10.3760/cma.j.issn.0253-2727.2016.10.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
目的 评价芦可替尼在中国骨髓纤维化患者中的疗效和安全性。 方法 63例中国骨髓纤维化患者纳入研究,男32例,女31例,中位年龄55(25~79)岁。芦可替尼起始剂量:基线PLT(100~200)×109/L者(25例)30 mg/d,基线PLT>200×109/L者(38例)40 mg/d。使用MRI/CT、欧洲癌症研究与治疗组织生活质量调查问卷核心30(EORTC QLQ-C30)和骨髓纤维化症状评估表(MFSAF)v2.0对患者进行脾脏体积、生活质量和症状评估。 结果 截至12个月随访结束,47例(74.6%)患者仍在继续治疗,25例(39.7%)患者脾脏体积较基线缩小超过35%,首次达到脾脏体积缩小≥35%的中位时间为12.71(95%CI 12.14~35.00)周。治疗期间,85.7%(54/63)的患者有不同程度的脾脏缩小,中位最佳脾脏体积缩小百分比为35.5%,48周时中位脾脏缩小体积为34.7%。治疗48周时53.1 %(26/49)的患者MFSAF症状评分降低超过50%,生活质量得到改善。最常见的血液学不良事件包括贫血和血小板计数降低,但极少造成停药。非血液学不良事件以1/2级为主。 结论 芦可替尼使中国骨髓纤维化患者的脾脏体积获得持续缩小、症状改善,不良反应可耐受。
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Z J Xiao
- Institute of Hematology and Blood Diseases Hospital, CAMS & PUMC, the State Key Laboratory of Experimental Hematology, Tianjin 300020, China
| |
Collapse
|
330
|
Kvasnicka HM, Thiele J, Bueso-Ramos CE, Sun W, Cortes J, Kantarjian HM, Verstovsek S. Long-term effects of ruxolitinib versus best available therapy on bone marrow fibrosis in patients with myelofibrosis. J Hematol Oncol 2018; 11:42. [PMID: 29544547 PMCID: PMC5856218 DOI: 10.1186/s13045-018-0585-5] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 03/01/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Myelofibrosis (MF) is a life-shortening complication of myeloproliferative neoplasms associated with ineffective hematopoiesis, splenomegaly, and progressive bone marrow (BM) fibrosis. The oral Janus kinase (JAK) 1/JAK2 inhibitor ruxolitinib has been shown to improve splenomegaly, symptom burden, and overall survival in patients with intermediate-2 or high-risk MF compared with placebo or best available therapy (BAT). METHODS The effects of ruxolitinib therapy for up to 66 months on BM morphology in 68 patients with advanced MF with variable BM fibrosis grade were compared with those in 192 matching patients treated with BAT. Available trephine biopsies underwent independent, blinded review by three hematopathologists for consensus-based adjudication of grades for reticulin fibrosis, collagen deposition, and osteosclerosis. RESULTS Ruxolitinib treatment versus BAT was associated with greater odds of BM fibrosis improvement or stabilization and decreased odds of BM fibrosis worsening based on changes from baseline in reticulin fibrosis grade. Generally, these changes were accompanied by a sustained higher level of individual spleen size reduction and regression of leukoerythroblastosis. Patients with more advanced baseline fibrosis showed lower spleen size response. CONCLUSIONS The finding that long-term ruxolitinib therapy may reverse or markedly delay BM fibrosis progression in advanced MF suggests that sustained JAK inhibition may be disease-modifying. TRIAL REGISTRATION INCB18424-251, ClinicalTrials.gov identifier NCT00509899 .
Collapse
Affiliation(s)
- Hans Michael Kvasnicka
- Senckenberg Institute of Pathology, University of Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany.
| | | | - Carlos E Bueso-Ramos
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Jorge Cortes
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hagop M Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Srdan Verstovsek
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
331
|
Leiva O, Leon C, Kah Ng S, Mangin P, Gachet C, Ravid K. The role of extracellular matrix stiffness in megakaryocyte and platelet development and function. Am J Hematol 2018; 93:430-441. [PMID: 29247535 DOI: 10.1002/ajh.25008] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 12/11/2017] [Accepted: 12/13/2017] [Indexed: 12/16/2022]
Abstract
The extracellular matrix (ECM) is a key acellular structure in constant remodeling to provide tissue cohesion and rigidity. Deregulation of the balance between matrix deposition, degradation, and crosslinking results in fibrosis. Bone marrow fibrosis (BMF) is associated with several malignant and nonmalignant pathologies severely affecting blood cell production. BMF results from abnormal deposition of collagen fibers and enhanced lysyl oxidase-mediated ECM crosslinking within the marrow, thereby increasing marrow stiffness. Bone marrow stiffness has been recently recognized as an important regulator of blood cell development, notably by modifying the fate and differentiation process of hematopoietic or mesenchymal stem cells. This review surveys the different components of the ECM and their influence on stem cell development, with a focus on the impact of the ECM composition and stiffness on the megakaryocytic lineage in health and disease. Megakaryocyte maturation and the biogenesis of their progeny, the platelets, are thought to respond to environmental mechanical forces through a number of mechanosensors, including integrins and mechanosensitive ion channels, reviewed here.
Collapse
Affiliation(s)
- Orly Leiva
- Department of Medicine; Whitaker Cardiovascular Institute, Boston University School of Medicine; Boston Massachusetts
| | - Catherine Leon
- Université de Strasbourg, INSERM, EFS Grand-Est, BPPS UMR-S 949, FMTS; Strasbourg F-67000 France
| | - Seng Kah Ng
- Department of Medicine; Whitaker Cardiovascular Institute, Boston University School of Medicine; Boston Massachusetts
| | - Pierre Mangin
- Université de Strasbourg, INSERM, EFS Grand-Est, BPPS UMR-S 949, FMTS; Strasbourg F-67000 France
| | - Christian Gachet
- Université de Strasbourg, INSERM, EFS Grand-Est, BPPS UMR-S 949, FMTS; Strasbourg F-67000 France
| | - Katya Ravid
- Department of Medicine; Whitaker Cardiovascular Institute, Boston University School of Medicine; Boston Massachusetts
| |
Collapse
|
332
|
Oritani K, Ohishi K, Okamoto S, Kirito K, Komatsu N, Tauchi T, Handa H, Saito S, Takenaka K, Shimoda K, Okada H, Amagasaki T, Wakase S, Shimozuma K, Akashi K. Effect of ruxolitinib therapy on the quality-of-life of Japanese patients with myelofibrosis. Curr Med Res Opin 2018; 34:531-537. [PMID: 29224367 DOI: 10.1080/03007995.2017.1415874] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 12/01/2017] [Accepted: 12/07/2017] [Indexed: 12/16/2022]
Abstract
OBJECTIVES Myelofibrosis (MF) is associated with a significant symptom burden that severely impacts patient quality-of-life (QoL). Ruxolitinib, a potent Janus kinase 1 (JAK1)/JAK2 inhibitor, led to substantial improvements in splenomegaly, MF-associated symptoms, and QoL in the phase 3 COMFORT studies, proving superior to placebo and best available therapy. This study evaluated the effect of ruxolitinib on symptoms and QoL in Japanese patients with MF. METHODS A pooled analysis of studies A2202 (NCT01392443) and AJP01 (NCT02087059) of ruxolitinib in Japanese patients with MF (n = 81) was conducted. Changes in total symptom score (TSS) and the European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire Core 30 were summarized. RESULTS Most patients received a starting dose of 15 or 20 mg twice daily (BID) and had a final titrated dose of ≥10 mg BID. Overall, 67.7% (44/65) achieved a ≥50% reduction from baseline in TSS at week 24. Reductions in TSS were seen in every dose group; the greatest reductions occurred in patients with a final titrated dose of 20 or 25 mg BID. Improvements in QoL were seen in patients who achieved a ≥50% reduction in TSS. Generally, improvements in TSS and individual symptoms correlated with reductions in spleen size, with those having a ≥35% reduction in spleen volume having the greatest improvements. CONCLUSIONS Consistent with COMFORT-I, ruxolitinib provided substantial improvements in symptoms and QoL in Japanese patients with MF, with higher doses of ruxolitinib associated with better responses.
Collapse
Affiliation(s)
- Kenji Oritani
- a Department of Hematology, Graduate School of Medical Sciences , International University of Health and Welfare Hospital , Tochigi , Japan
| | - Kohshi Ohishi
- b Blood Transfusion Service, Mie University Hospital , Mie , Japan
| | - Shinichiro Okamoto
- c Division of Hematology, Department of Medicine , Keio University Hospital , Tokyo , Japan
| | - Keita Kirito
- d Department of Hematology/Oncology , University of Yamanashi , Yamanashi , Japan
| | - Norio Komatsu
- e Department of Hematology , Juntendo University School of Medicine , Tokyo , Japan
| | - Tetsuzo Tauchi
- f Department of Hematology , Tokyo Medical University , Tokyo , Japan
| | - Hiroshi Handa
- g Department of Medicine , Gunma University Hospital , Gunma , Japan
| | - Shigeki Saito
- h Department of Hematology , Nagoya University Hospital , Nagoya , Japan
- i Department of Hematology and Oncology , JRC Nagoya Daini Red Cross Hospital , Aichi , Japan
| | - Katsuto Takenaka
- j Center for Cellular and Molecular Medicine, Kyushu University Hospital , Japan
| | - Kazuya Shimoda
- k Gastroenterology and Hematology , University of Miyazaki , Miyazaki , Japan
| | - Hikaru Okada
- l Department of Medical Affairs-Oncology , Novartis Pharma KK , Tokyo , Japan
| | - Taro Amagasaki
- m Clinical Development, Japan Integrated Biostatistics, Novartis Pharma KK , Tokyo , Japan
| | - Shiho Wakase
- n Department of Commercial Excellence-Oncology , Novartis Pharma KK , Tokyo , Japan
| | - Kojiro Shimozuma
- o Department of Biomedical Sciences , Ritsumeikan University , Shiga , Japan
| | - Koichi Akashi
- p Department of Medicine and Biosystemic Sciences , Kyushu University , Fukuoka , Japan
| |
Collapse
|
333
|
Lussana F, Cattaneo M, Rambaldi A, Squizzato A. Ruxolitinib-associated infections: A systematic review and meta-analysis. Am J Hematol 2018; 93:339-347. [PMID: 29150886 DOI: 10.1002/ajh.24976] [Citation(s) in RCA: 169] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 11/13/2017] [Accepted: 11/14/2017] [Indexed: 12/14/2022]
Abstract
Ruxolitinib exerts immunosuppressive activity that may increase the risk of infectious complications. We performed a systematic review of the literature with the aim of estimating the risk of infections in patients treated with ruxolitinib. Studies were identified by electronic search of MEDLINE and EMBASE database. Differences in the incidence of infectious events between ruxolitinib and comparison groups were expressed as odds ratios (ORs) and 95% confidence intervals (95% CI). Five phase III randomized clinical trials (RCTs) (3 phase IIIa with their extended phase and 2 phase IIIb), 6 phase IV studies and 28 case reports were included in this systematic review. Ruxolitinib was associated with a statistically significant increased risk of herpes zoster infection compared to control group in 3 RCTs including patients with polycythemia vera (OR 7.39 [1.33, 41.07]) and in a pooled analysis of the extended phase IIIa RCTs (OR 5.20 [95%CI 1.27, 21.18]). In the larger phase IV post-marketing study, the incidence of the most frequent infections was 8% for herpes zoster, 6.1% for bronchitis and 6% for urinary tract infections. In the published case reports, the most frequent infections were tuberculosis (N = 10), hepatitis B reactivation (N = 5) and pneumocystis jeroveci infection (N = 2). Evidence is not solid enough to accurately estimate the risk of infection in ruxolitinib-treated patients. However, published data clearly suggest that the infection risk may be clinically relevant. Well-designed studies are warranted to evaluate the risk of ruxolitinib-associated infection, in order to identify the most appropriate antimicrobial prophylactic strategy.
Collapse
Affiliation(s)
- Federico Lussana
- Hematology and Bone Marrow Transplant Unit, Azienda Ospedaliera Papa Giovanni XIII; Bergamo Italy
| | - Marco Cattaneo
- Medicina III, Ospedale San Paolo, ASST Santi Paolo e Carlo Dipartimento di Scienze della Salute; Università degli Studi di Milano; Milan Italy
| | - Alessandro Rambaldi
- Hematology and Bone Marrow Transplant Unit, Azienda Ospedaliera Papa Giovanni XIII; Bergamo Italy
- Department of Oncology and Hematology; Università degli Studi di Milano; Milan Italy
| | - Alessandro Squizzato
- Research Center on Thromboembolic Disorders and Antithrombotic Therapies, Department of Clinical and Experimental Medicine; University of Insubria; Varese Italy
| |
Collapse
|
334
|
Abstract
PURPOSE OF REVIEW Allogeneic hematopoietic stem-cell transplantation (HSCT) remains the only curative therapy for myelofibrosis. The number of HSCTs performed for this indication has been steadily increasing over the past years, even after the approval of the Janus kinase (JAK) inhibitor, ruxolitinib. This increase may be attributed to improved patient selection based on new prognostic molecular markers, more frequent use of matched unrelated donors, secondary to better (high-resolution) human leukocyte antigen typing and supportive care. Ruxolitinib approval raises new questions regarding the role of JAK inhibitors in the transplant setting. RECENT FINDINGS The current review summarizes recent updates on HSCT in myelofibrosis. Predictors for transplant outcomes, and specific considerations related to myelofibrosis patient selection for HSCT (e.g. molecular risk stratification) are reviewed. In addition, this review will consider management of myelofibrosis patients in the peritransplant period, including the role of ruxolitinib in the pretransplant period, pre and posttransplant splenomegaly, transplant protocols, posttransplant follow-up of minimal residual disease and interventions in the event of poor engraftment. SUMMARY HSCT remains a highly relevant treatment option for myelofibrosis in the era of JAK inhibitors. Recent advances may contribute to a refined definition of HSCT eligibility and identification of the optimal transplantation time, conditioning protocols and posttransplant management.
Collapse
|
335
|
Rocca S, Carrà G, Poggio P, Morotti A, Brancaccio M. Targeting few to help hundreds: JAK, MAPK and ROCK pathways as druggable targets in atypical chronic myeloid leukemia. Mol Cancer 2018; 17:40. [PMID: 29455651 PMCID: PMC5817721 DOI: 10.1186/s12943-018-0774-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 02/01/2018] [Indexed: 12/19/2022] Open
Abstract
Atypical Chronic Myeloid Leukemia (aCML) is a myeloproliferative neoplasm characterized by neutrophilic leukocytosis and dysgranulopoiesis. From a genetic point of view, aCML shows a heterogeneous mutational landscape with mutations affecting signal transduction proteins but also broad genetic modifiers and chromatin remodelers, making difficult to understand the molecular mechanisms causing the onset of the disease. The JAK-STAT, MAPK and ROCK pathways are known to be responsible for myeloproliferation in physiological conditions and to be aberrantly activated in myeloproliferative diseases. Furthermore, experimental evidences suggest the efficacy of inhibitors targeting these pathways in repressing myeloproliferation, opening the way to deep clinical investigations. However, the activation status of these pathways is rarely analyzed when genetic mutations do not occur in a component of the signaling cascade. Given that mutations in functionally unrelated genes give rise to the same pathology, it is tempting to speculate that alteration in the few signaling pathways mentioned above might be a common feature of pathological myeloproliferation. If so, targeted therapy would be an option to be considered for aCML patients.
Collapse
Affiliation(s)
- Stefania Rocca
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126, Torino, Italy
| | - Giovanna Carrà
- Department of Clinical and Biological Sciences, University of Torino, 10043, Orbassano, Italy
| | - Pietro Poggio
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126, Torino, Italy
| | - Alessandro Morotti
- Department of Clinical and Biological Sciences, University of Torino, 10043, Orbassano, Italy
| | - Mara Brancaccio
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126, Torino, Italy.
| |
Collapse
|
336
|
Reinwald M, Silva JT, Mueller NJ, Fortún J, Garzoni C, de Fijter JW, Fernández-Ruiz M, Grossi P, Aguado JM. ESCMID Study Group for Infections in Compromised Hosts (ESGICH) Consensus Document on the safety of targeted and biological therapies: an infectious diseases perspective (Intracellular signaling pathways: tyrosine kinase and mTOR inhibitors). Clin Microbiol Infect 2018; 24 Suppl 2:S53-S70. [PMID: 29454849 DOI: 10.1016/j.cmi.2018.02.009] [Citation(s) in RCA: 128] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 02/08/2018] [Accepted: 02/11/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND The present review is part of the European Society of Clinical Microbiology and Infectious Diseases (ESCMID) Study Group for Infections in Compromised Hosts (ESGICH) Consensus Document on the safety of targeted and biologic therapies. AIMS To review, from an infectious diseases perspective, the safety profile of therapies targeting different intracellular signaling pathways and to suggest preventive recommendations. SOURCES Computer-based Medline searches with MeSH terms pertaining to each agent or therapeutic family. CONTENT Although BCR-ABL tyrosine kinase inhibitors modestly increase the overall risk of infection, dasatinib has been associated with cytomegalovirus and hepatitis B virus reactivation. BRAF/MEK kinase inhibitors do not significantly affect infection susceptibility. The effect of Bruton tyrosine kinase inhibitors (ibrutinib) among patients with B-cell malignancies is difficult to distinguish from that of previous immunosuppression. However, cases of Pneumocystis jirovecii pneumonia (PCP), invasive fungal infection and progressive multifocal leukoencephalopathy have been occasionally reported. Because phosphatidylinositol-3-kinase inhibitors (idelalisib) may predispose to opportunistic infections, anti-Pneumocystis prophylaxis and prevention strategies for cytomegalovirus are recommended. No increased rates of infection have been observed with venetoclax (antiapoptotic protein Bcl-2 inhibitor). Therapy with Janus kinase inhibitors markedly increases the incidence of infection. Pretreatment screening for chronic hepatitis B virus and latent tuberculosis infection must be performed, and anti-Pneumocystis prophylaxis should be considered for patients with additional risk factors. Cancer patients receiving mTOR inhibitors face an increased incidence of overall infection, especially those with additional risk factors (prior therapies or delayed wound healing). IMPLICATIONS Specific preventive approaches are warranted in view of the increased risk of infection associated with some of the reviewed agents.
Collapse
Affiliation(s)
- M Reinwald
- Department of Hematology and Oncology, Klinikum Brandenburg, Medizinische Hochschule Brandenburg Theodor Fontane, Brandenburg an der Havel, Germany.
| | - J T Silva
- Department of Infectious Diseases, University Hospital of Badajoz, Fundación para la Formación e Investigación de los Profesionales de la Salud (FundeSalud), Badajoz, Spain
| | - N J Mueller
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - J Fortún
- Department of Infectious Diseases, Hospital Universitario 'Ramon y Cajal', Madrid, Spain; Spanish Network for Research in Infectious Diseases (REIPI RD16/0016), Instituto de Salud Carlos III, Madrid, Spain
| | - C Garzoni
- Department of Internal Medicine, Clinica Luganese, Lugano, Switzerland; Department of Infectious Disease, Clinica Luganese, Lugano, Switzerland
| | - J W de Fijter
- Department of Medicine, Division of Nephrology, Leiden University Medical Centre, Leiden, The Netherlands
| | - M Fernández-Ruiz
- Unit of Infectious Diseases, Hospital Universitario '12 de Octubre', Instituto de Investigación Hospital '12 de Octubre' (i+12), School of Medicine, Universidad Complutense, Madrid, Spain; Spanish Network for Research in Infectious Diseases (REIPI RD16/0016), Instituto de Salud Carlos III, Madrid, Spain
| | - P Grossi
- Department of Infectious and Tropical Diseases, University of Insubria, Ospedale di Circolo-Fondazioni Macchi, Varese, Italy
| | - J M Aguado
- Unit of Infectious Diseases, Hospital Universitario '12 de Octubre', Instituto de Investigación Hospital '12 de Octubre' (i+12), School of Medicine, Universidad Complutense, Madrid, Spain; Spanish Network for Research in Infectious Diseases (REIPI RD16/0016), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
337
|
Assi R, Kantarjian HM, Garcia-Manero G, Cortes JE, Pemmaraju N, Wang X, Nogueras-Gonzalez G, Jabbour E, Bose P, Kadia T, Dinardo CD, Patel K, Bueso-Ramos C, Zhou L, Pierce S, Gergis R, Tuttle C, Borthakur G, Estrov Z, Luthra R, Hidalgo-Lopez J, Verstovsek S, Daver N. A phase II trial of ruxolitinib in combination with azacytidine in myelodysplastic syndrome/myeloproliferative neoplasms. Am J Hematol 2018; 93:277-285. [PMID: 29134664 DOI: 10.1002/ajh.24972] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Revised: 11/09/2017] [Accepted: 11/10/2017] [Indexed: 01/05/2023]
Abstract
Ruxolitinib and azacytidine target distinct disease manifestations of myelodysplastic syndrome/myeloproliferative neoplasms (MDS/MPNs). Patients with MDS/MPNs initially received ruxolitinib BID (doses based on platelets count), continuously in 28-day cycles for the first 3 cycles. Azacytidine 25 mg/m2 (Day 1-5) intravenously or subcutaneously was recommended to be added to each cycle starting cycle 4 and could be increased to 75 mg/m2 (Days 1-5) for disease control. Azacytidine could be started earlier than cycle 4 and/or at higher dose in patients with rapidly proliferative disease or with elevated blasts. Thirty-five patients were treated (MDS/MPN-U, n =14; CMML, n =17; aCML, n =4), with a median follow-up of 15.2 months (range, 1.0-41.5). All patients were evaluable by the 2015 international consortium proposal of response criteria for MDS/MPNs (ICP MDS/MPN) and 20 (57%) responded. Nine patients (45%) responded after the addition of azacytidine. A greater than 50% reduction in palpable splenomegaly at 24 weeks was noted in 9/14 (64%) patients. Responders more frequently were JAK2-mutated (P = .02) and had splenomegaly (P = .03) compared to nonresponders. New onset grade 3/4 anemia and thrombocytopenia occurred in 18 (51%) and 19 (54%) patients, respectively, but required therapy discontinuation in only 1 (3%) patient. Patients with MDS/MPN-U had better median survival compared to CMML and aCML (26.5 vs 15.1 vs 8 months; P = .034). The combination of ruxolitinib and azacytidine was well-tolerated with an ICP MDS/MPN-response rate of 57% in patients with MDS/MPNs. The survival benefit was most prominent in patients with MDS/MPN-U.
Collapse
Affiliation(s)
- Rita Assi
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Hagop M. Kantarjian
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | | | - Jorge E. Cortes
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Naveen Pemmaraju
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Xuemei Wang
- Department of Biostatistics; The University of Texas MD Anderson Cancer Center; Houston Texas
| | | | - Elias Jabbour
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Prithviraj Bose
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Tapan Kadia
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Courtney D. Dinardo
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Keyur Patel
- Department of Hematopathology; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Carlos Bueso-Ramos
- Department of Hematopathology; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Lingsha Zhou
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Sherry Pierce
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Romany Gergis
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Carla Tuttle
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Gautam Borthakur
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Zeev Estrov
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Rajyalakshmi Luthra
- Department of Hematopathology; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Juliana Hidalgo-Lopez
- Department of Hematopathology; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Srdan Verstovsek
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Naval Daver
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| |
Collapse
|
338
|
Ajayi S, Becker H, Reinhardt H, Engelhardt M, Zeiser R, von Bubnoff N, Wäsch R. Ruxolitinib. Recent Results Cancer Res 2018; 212:119-132. [PMID: 30069628 DOI: 10.1007/978-3-319-91439-8_6] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Ruxolitinib, formerly known as INCB018424 or INC424, is a potent and selective oral inhibitor of Janus kinase (JAK) 1 and JAK2. Ruxolitinib has been approved for the treatment of myelofibrosis (MF) by the US Food and Drug Administration (FDA) in 2011 and by the European Medicines Agency (EMA) in 2012, followed by the approval for the treatment of hydroxyurea (HU)-resistant or -intolerant polycythemia vera (PV) in 2014. Both MF and PV are myeloproliferative neoplasms (MPNs) which are characterized by the aberrant activation of the JAK-STAT pathway. Clinically, MF features bone marrow fibrosis, splenomegaly, abnormal blood counts, and poor quality-of-life through associated symptoms. PV is characterized by the overproduction of primarily red blood cells (RBC), risk of thrombotic complications, and development of secondary MF. Ruxolitinib treatment results in a meaningful reduction in spleen size and symptom burden in the majority of MF patients and may also have a favorable effect on survival. In PV, ruxolitinib effectively controls the hematocrit and reduces splenomegaly. Since recently, ruxolitinib is also under investigation for the treatment of graft-versus-host disease (GvHD) after allogeneic hematopoietic stem cell transplantation (HSCT). Toxicities of ruxolitinib include myelosuppression, which results in dose-limiting thrombocytopenia and anemia, and viral reactivations. The metabolization of ruxolitinib through CYP3A4 needs to be considered particularly if co-administered with potent CYP3A4 inhibitors. Several further JAK inhibitors are currently under investigation for MPNs or other immuno-inflammatory diseases.
Collapse
Affiliation(s)
- Stefanie Ajayi
- Department of Hematology and Oncology, University of Freiburg Medical Center, Hugstetter Str. 55, 79106, Freiburg, Germany.,Comprehensive Cancer Center Freiburg (CCCF), Hugstetter Str. 55, 79106, Freiburg, Germany
| | - Heiko Becker
- Department of Hematology and Oncology, University of Freiburg Medical Center, Hugstetter Str. 55, 79106, Freiburg, Germany.,Comprehensive Cancer Center Freiburg (CCCF), Hugstetter Str. 55, 79106, Freiburg, Germany
| | - Heike Reinhardt
- Department of Hematology and Oncology, University of Freiburg Medical Center, Hugstetter Str. 55, 79106, Freiburg, Germany.,Comprehensive Cancer Center Freiburg (CCCF), Hugstetter Str. 55, 79106, Freiburg, Germany
| | - Monika Engelhardt
- Department of Hematology and Oncology, University of Freiburg Medical Center, Hugstetter Str. 55, 79106, Freiburg, Germany.,Comprehensive Cancer Center Freiburg (CCCF), Hugstetter Str. 55, 79106, Freiburg, Germany
| | - Robert Zeiser
- Department of Hematology and Oncology, University of Freiburg Medical Center, Hugstetter Str. 55, 79106, Freiburg, Germany.,Comprehensive Cancer Center Freiburg (CCCF), Hugstetter Str. 55, 79106, Freiburg, Germany
| | - Nikolas von Bubnoff
- Department of Hematology and Oncology, University of Freiburg Medical Center, Hugstetter Str. 55, 79106, Freiburg, Germany.,Comprehensive Cancer Center Freiburg (CCCF), Hugstetter Str. 55, 79106, Freiburg, Germany
| | - Ralph Wäsch
- Department of Hematology and Oncology, University of Freiburg Medical Center, Hugstetter Str. 55, 79106, Freiburg, Germany. .,Comprehensive Cancer Center Freiburg (CCCF), Hugstetter Str. 55, 79106, Freiburg, Germany.
| |
Collapse
|
339
|
Malhotra H, Agarwal M, Chakarborti P, Varma N, Mathews V, Bhattacharyya J, Seth T, Gyathri K, Menon H, Subramanian PG, Sharma A, Bhattacharyya M, Mehta J, Shah S, Gogoi PK, Nair R, Agarwal U, Varma S, Prasad SVVS, Mishra D. Revised myeloproliferative neoplasms working group consensus recommendations for diagnosis and management of primary myelofibrosis, polycythemia vera, and essential thrombocythemia. Indian J Med Paediatr Oncol 2018. [DOI: 10.4103/ijmpo.ijmpo_88_17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
340
|
Bose P, Gotlib J, Harrison CN, Verstovsek S. SOHO State-of-the-Art Update and Next Questions: MPN. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2018; 18:1-12. [PMID: 29277359 PMCID: PMC5915302 DOI: 10.1016/j.clml.2017.11.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 11/27/2017] [Indexed: 12/28/2022]
Abstract
The discovery of the activating Janus kinase (JAK)2V617F mutation in 2005 in most patients with the classic Philadelphia chromosome-negative myeloproliferative neoplasms (MPN) spurred intense interest in research into these disorders, culminating in the identification of activating mutations in MPL in 2006 and indels in the gene encoding calreticulin (CALR) in 2013, thus providing additional mechanistic explanations for the universal activation of JAK-signal transducer and activator of transcription (JAK-STAT) observed in these conditions, and the success of the JAK1/2 inhibitor ruxolitinib, which first received regulatory approval in 2011. The field has continued to advance rapidly since then, and the past 2 years have witnessed important changes to the classification of MPN and diagnostic criteria for polycythemia vera (PV), novel insights into the mechanisms of bone marrow fibrosis in primary myelofibrosis (PMF), increasing appreciation of the biologic differences between essential thrombocythemia (ET), prefibrotic and overt PMF, and between primary and post-PV/ET myelofibrosis (MF). Additionally, the mechanisms through which mutant CALR drives JAK-STAT pathway activation and oncogenic transformation are now better understood. Although mastocytosis is no longer included under the broad heading of MPN in the 2016 revision to the World Health Organization classification, an important milestone in mastocytosis research was reached in 2017 with the regulatory approval of midostaurin for patients with advanced systemic mastocytosis (AdvSM). In this article, we review the major recent developments in the areas of PV, ET, and MF, and also briefly summarize the literature on midostaurin and other KIT inhibitors for patients with AdvSM.
Collapse
Affiliation(s)
- Prithviraj Bose
- Department of Leukemia, University of Texas M.D. Anderson Cancer Center, Houston, TX.
| | - Jason Gotlib
- Department of Medicine - Hematology, Stanford University, Palo Alto, CA
| | | | - Srdan Verstovsek
- Department of Leukemia, University of Texas M.D. Anderson Cancer Center, Houston, TX
| |
Collapse
|
341
|
Myelofibrosis-Related Anemia: Current and Emerging Therapeutic Strategies. Hemasphere 2017; 1:e1. [PMID: 31723730 PMCID: PMC6745971 DOI: 10.1097/hs9.0000000000000001] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 10/20/2017] [Indexed: 12/15/2022] Open
Abstract
Myelofibrosis (MF) is a clonal hematopoietic stem cell disorder characterized by pathological myeloproliferation and aberrant cytokine production resulting in progressive fibrosis, inflammation, and functional compromise of the bone marrow niche. Patients with MF develop splenomegaly (due to extramedullary hematopoiesis), hypercatabolic symptoms (due to overexpression of inflammatory cytokines), and anemia (due to bone marrow failure and splenic sequestration). MF remains curable only with allogeneic hematopoietic stem cell transplantation (ASCT), a therapy that few MF patients are deemed fit to undergo. The goals of treatment are thus often palliative. The approval of the JAK inhibitor ruxolitinib has done much to address the burden of splenomegaly and constitutional symptoms of patients with MF; however, therapy-related anemia is often an anticipated downside. Anemia thus remains a challenge in the management of MF and represents a major unmet need. Intractable anemia depresses quality of life, portends poor outcomes, and can act to restrict access to palliative JAK inhibition in some patients. While therapies for MF-related anemia do exist, they are limited in their efficacy, durability, and tolerability. Therapies currently in development promise improved anemia-specific outcomes; however, are still early in the pathway to regulatory approval and regular clinical use. In this review, we will discuss established and emerging treatments for MF-related anemia. We will give particular attention to developmental therapies which herald significant progress in the understanding and management of MF-related anemia.
Collapse
|
342
|
Diaz AE, Mesa RA. Pacritinib and its use in the treatment of patients with myelofibrosis who have thrombocytopenia. Future Oncol 2017; 14:797-807. [PMID: 29235894 DOI: 10.2217/fon-2017-0494] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The treatment landscape for myelofibrosis (MF) has reached the molecular era by targeting different pathways that are implied in this myeloproliferative neoplasm. A few years ago, the first-in-class JAK1/JAK2 inhibitor ruxolitinib, demonstrated reductions in both constitutional symptoms and splenomegaly, leading to the US FDA approval. The development or worsening of cytopenias in patients receiving ruxolitinib uncovered an unmet need that has been addressed by alternative approaches. Pacritinib, a dual JAK2 and FLT3 inhibitor which also inhibits IRAK1, has demonstrated the ability to favorably impact MF-associated splenomegaly and symptom burden, while having limited myelosuppression with manageable gastrointestinal toxicity. Herein, we provide an overview of pacritinib, from early preclinical studies to the latest and ongoing PAC203 trial, as an emerging therapy for MF.
Collapse
Affiliation(s)
- Adolfo Enrique Diaz
- Hematology Oncology Division, UT Health San Antonio Cancer Center, 7979 Wurzbach Road, San Antonio, TX 78229, USA
| | - Ruben A Mesa
- Hematology Oncology Division, UT Health San Antonio Cancer Center, 7979 Wurzbach Road, San Antonio, TX 78229, USA
| |
Collapse
|
343
|
Ianotto JC, Chauveau A, Boyer-Perrard F, Gyan E, Laribi K, Cony-Makhoul P, Demory JL, de Renzis B, Dosquet C, Rey J, Roy L, Dupriez B, Knoops L, Legros L, Malou M, Hutin P, Ranta D, Benbrahim O, Ugo V, Lippert E, Kiladjian JJ. Benefits and pitfalls of pegylated interferon-α2a therapy in patients with myeloproliferative neoplasm-associated myelofibrosis: a French Intergroup of Myeloproliferative neoplasms (FIM) study. Haematologica 2017; 103:438-446. [PMID: 29217781 PMCID: PMC5830374 DOI: 10.3324/haematol.2017.181297] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 12/06/2017] [Indexed: 12/12/2022] Open
Abstract
We have previously described the safety and efficacy of pegylated interferon-α2a therapy in a cohort of 62 patients with myeloproliferative neoplasm-associated myelofibrosis followed in centers affiliated to the French Intergroup of Myeloproliferative neoplasms. In this study, we report their long-term outcomes and correlations with mutational patterns of driver and non-driver mutations analyzed by targeted next generation sequencing. The median age at diagnosis was 66 years old, the median follow-up since starting pegylated interferon was 58 months. At the time of analysis, 30 (48.4%) patients were alive including 16 still being treated with pegylated interferon. The median survival of patients with intermediate and high-risk prognostic Lille and dynamic International Prognostic Scoring System scores treated with pegylated interferon was increased in comparison to that of historical cohorts. In addition, overall survival was significantly correlated with the duration of pegylated interferon therapy (70 versus 30 months after 2 years of treatment, P<10−12). JAK2V617F allele burden was decreased by more than 50% in 58.8% of patients and two patients even achieved complete molecular response. Next-generation sequencing analyses performed in 49 patients showed that 28 (57.1%) of them carried non-driver mutations. The presence of at least one additional mutation was associated with a reduction of both overall and leukemia-free survival. These findings in a large series of patients with myelofibrosis suggest that pegylated interferon therapy may provide a survival benefit for patients with intermediate- or high-risk Lille and dynamic International Prognostic Scoring System scores. It also reduced the JAK2V617F allele burden in most patients. These results further support the use of pegylated interferon in selected patients with myelofibrosis.
Collapse
Affiliation(s)
| | - Aurélie Chauveau
- Laboratoire d'Hématologie, CHRU de Brest and INSERM U1078, Université de Bretagne Occidentale, Brest, France
| | | | - Emmanuel Gyan
- Hématologie et Thérapie Cellulaire, CRU de Cancérologie H.S. Kaplan, Tours, France
| | | | | | - Jean-Loup Demory
- Service d'Hématologie, Hôpital St Vincent de Paul, Lille, France
| | | | | | - Jerome Rey
- Département d'Hématologie, Institut Paoli-Calmette, Marseille, France
| | - Lydia Roy
- Service d'Hématologie, Hôpital de Créteil, France
| | | | - Laurent Knoops
- Cliniques Universitaires Saint-Luc and Université Catholique de Louvain, Brussels, Belgium
| | | | - Mohamed Malou
- Service d'Oncologie et D'Hématologie, Hôpital de Morlaix, France
| | - Pascal Hutin
- Service de Médecine Interne et de Maladies Infectieuses, Hôpital Laennec, Quimper, France
| | - Dana Ranta
- Département d'Hématologie, Hôpital Universitaire de Nancy, Vandoeuvre-lès-Nancy, France
| | - Omar Benbrahim
- Service d'Hématologie, Hôpital La Source, Orléans, France
| | - Valérie Ugo
- Laboratoire d'Hématologie, CHU d'Angers, France
| | - Eric Lippert
- Laboratoire d'Hématologie, CHRU de Brest and INSERM U1078, Université de Bretagne Occidentale, Brest, France
| | - Jean-Jacques Kiladjian
- Centre d'Investigation Clinique, Hôpital Saint-Louis, APHP, Université Paris Diderot, Inserm, Paris, France
| |
Collapse
|
344
|
Greenfield G, McMullin MF. A spotlight on the management of complications associated with myeloproliferative neoplasms: a clinician's perspective. Expert Rev Hematol 2017; 11:25-35. [PMID: 29183180 DOI: 10.1080/17474086.2018.1410433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
INTRODUCTION Myeloproliferative neoplasms (MPNs) are associated with a variety of symptoms and signs which cause major morbidity for the patients. The disorders are associated with increased incidence of thromboembolic and hemorrhagic events which can lead to complications and shortened life expectancy. Areas covered: Using systematic literature review and expert clinical and research experience the authors discuss strategies for the management of symptoms and signs including pruritus, fatigue, splenomegaly, and cytopenia. Cytoreduction including treatments to inhibit the JAK/STAT pathway are considered. Pathogenesis and prevention and treatment of thrombotic and hemorrhagic events and their management is addressed and the suggested management of the special situations such as surgery and pregnancy are discussed. Expert commentary: Management of disease has traditionally focused on symptom treatment and complication prevention but the discovery of driver mutations has led to treatments aiming to eliminate the clone, which should be the ultimate goal of therapy. A future challenge is to develop safe and effective MPN therapy and to personalize therapy.
Collapse
|
345
|
Pearson S, Williamson AJK, Blance R, Somervaille TCP, Taylor S, Azadbakht N, Whetton AD, Pierce A. Proteomic analysis of JAK2V617F-induced changes identifies potential new combinatorial therapeutic approaches. Leukemia 2017; 31:2717-2725. [PMID: 28533538 PMCID: PMC5729335 DOI: 10.1038/leu.2017.143] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 04/06/2017] [Accepted: 04/25/2017] [Indexed: 01/02/2023]
Abstract
In excess of 90% of patients with polycythaemia vera (PV) express a mutated form of Janus kinase 2 (JAK2), JAK2V617F. Such aberrant proteins offer great potential for the treatment of these diseases; however, inhibitors to JAK2 have had limited success in the clinic in terms of curing the disease. To understand the effects of this oncogene in haematopoietic cells with the aim of improving treatment strategies, we undertook a systematic evaluation of the effects of JAK2V617F expression using proteomics. The effects of JAK2V617F on over 5000 proteins and 2000 nuclear phosphopeptide sites were relatively quantified using either SILAC or eight-channel iTRAQ mass spectrometry. Pathway analysis of the proteins identified as changing indicated disruption to the p53 and MYC signalling pathways. These changes were confirmed using orthogonal approaches. The insight gained from this proteomic analysis led to the formation of hypothesis-driven analysis on inhibitor-mediated effects on primary cells from patients with a JAK2V617F mutation. Simultaneous inhibition of MYC and upregulation of p53 led to the preferential extinction of JAK2V617F-positive CD34+ cells, illustrating a potential therapeutic benefit from combined targeting of p53 and MYC.
Collapse
Affiliation(s)
- S Pearson
- Stem Cell and Leukaemia Proteomics Laboratory, Manchester Academic Health Science Centre, University of Manchester, Wolfson Molecular Imaging Centre, Manchester, UK
| | - A J K Williamson
- Stem Cell and Leukaemia Proteomics Laboratory, Manchester Academic Health Science Centre, University of Manchester, Wolfson Molecular Imaging Centre, Manchester, UK
| | - R Blance
- Stem Cell and Leukaemia Proteomics Laboratory, Manchester Academic Health Science Centre, University of Manchester, Wolfson Molecular Imaging Centre, Manchester, UK
| | - T C P Somervaille
- Leukaemia Biology Laboratory, Cancer Research UK Manchester Institute, University of Manchester, Manchester, UK
| | - S Taylor
- Stem Cell and Leukaemia Proteomics Laboratory, Manchester Academic Health Science Centre, University of Manchester, Wolfson Molecular Imaging Centre, Manchester, UK
| | - N Azadbakht
- Stem Cell and Leukaemia Proteomics Laboratory, Manchester Academic Health Science Centre, University of Manchester, Wolfson Molecular Imaging Centre, Manchester, UK
| | - A D Whetton
- Stem Cell and Leukaemia Proteomics Laboratory, Manchester Academic Health Science Centre, University of Manchester, Wolfson Molecular Imaging Centre, Manchester, UK
- Stoller Biomarker Discovery Centre, University of Manchester, Manchester, UK
| | - A Pierce
- Stem Cell and Leukaemia Proteomics Laboratory, Manchester Academic Health Science Centre, University of Manchester, Wolfson Molecular Imaging Centre, Manchester, UK
| |
Collapse
|
346
|
Abstract
PURPOSE OF REVIEW The purpose of the review was to provide a contemporary update of novel agents and targets under investigation in myelofibrosis in the Janus kinase (JAK) inhibitor era. RECENT FINDINGS Myelofibrosis (MF) is a clonal stem cell disease characterized by marrow fibrosis and a heterogeneous disease phenotype with a variable degree of splenomegaly, cytopenias, and constitutional symptoms that significantly impact quality of life and survival. Overactive JAK/STAT signaling is a hallmark of MF. The only approved therapy for MF, JAK1/2 inhibitor ruxolitinib, can ameliorate splenomegaly, improve symptoms, and prolong survival in some patients. Therapeutic challenges remain, however. Myelosuppression limits the use of ruxolitinib in some patients, eventual drug resistance is common, and the underlying malignant clone persists despite therapy. A deeper understanding of the pathogenesis of MF has informed the development of additional agents. Promising targets under investigation include JAK1 and JAK2 and downstream intermediates in related signaling pathways, epigenetic modifiers, pro-inflammatory cytokines, and immune regulators.
Collapse
Affiliation(s)
- Kristen Pettit
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, 5841 S. Maryland Ave, MC2115, Chicago, IL, 60637, USA
| | - Olatoyosi Odenike
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, 5841 S. Maryland Ave, MC2115, Chicago, IL, 60637, USA.
| |
Collapse
|
347
|
Kuykendall AT, Shah S, Talati C, Al Ali N, Sweet K, Padron E, Sallman DA, Lancet JE, List AF, Zuckerman KS, Komrokji RS. Between a rux and a hard place: evaluating salvage treatment and outcomes in myelofibrosis after ruxolitinib discontinuation. Ann Hematol 2017; 97:435-441. [PMID: 29189896 DOI: 10.1007/s00277-017-3194-4] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 11/20/2017] [Indexed: 11/26/2022]
Abstract
Ruxolitinib is a JAK1/2 inhibitor that is effective in managing symptoms and splenomegaly related to myelofibrosis (MF). Unfortunately, many patients must discontinue ruxolitinib, at which time treatment options are not well defined. In this study, we investigated salvage treatment options and clinical outcomes among MF patients who received and discontinued ruxolitinib outside the context of a clinical trial. Among 145 patients who received ruxolitinib, 23 died while on treatment, 58 remained on treatment at time of analysis, leaving 64 people available for analysis. Development of cytopenias was the most common reason for discontinuation (38%) after median treatment time of 3.8 months (mo). The majority of patients received some form of salvage therapy after ruxolitinib discontinuation (n = 42; 66%), with allogeneic hematopoietic stem cell transplant (alloHSCT) (n = 17), being most commonly employed. Lenalidomide, thalidomide, hydroxyurea, interferon, and danazol were used with similar frequency. The response rate to salvage treatment was 26% (8 responses) and responses were most often seen with lenalidomide or thalidomide. Improved outcomes were observed in patients who underwent alloHSCT or received salvage therapy compared to those who did not receive additional therapy. Median overall survival (OS) after ruxolitinib discontinuation was 13 months. These findings show that salvage therapy can provide clinical responses after ruxolitinib discontinuation; however, these responses are rare and outcomes in this patient population are poor. This represents an area of unmet clinical need in MF.
Collapse
Affiliation(s)
- Andrew T Kuykendall
- University of South Florida Morsani College of Medicine at H. Lee Moffitt Cancer Center, 12902 Magnolia Drive, MCC-GME, Tampa, FL, 33612, USA.
| | - Savan Shah
- Morsani College of Medicine, Department of Internal Medicine, University of South Florida, 12902 Magnolia Drive, MCC-GME, Tampa, FL, 33612, USA
| | - Chetasi Talati
- University of South Florida Morsani College of Medicine at H. Lee Moffitt Cancer Center, 12902 Magnolia Drive, MCC-GME, Tampa, FL, 33612, USA
| | - Najla Al Ali
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| | - Kendra Sweet
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| | - Eric Padron
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| | - David A Sallman
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| | - Jeffrey E Lancet
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| | - Alan F List
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| | - Kenneth S Zuckerman
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| | - Rami S Komrokji
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| |
Collapse
|
348
|
Gómez-Casares MT, Hernández-Boluda JC, Jiménez-Velasco A, Martínez-López J, Ferrario MG, Gozalbo I, Gostkorzewicz J, Subirá R. Cost-effectiveness of Ruxolitinib vs Best Available Therapy in the Treatment of Myelofibrosis in Spain. JOURNAL OF HEALTH ECONOMICS AND OUTCOMES RESEARCH 2017; 5:162-174. [PMID: 35620778 PMCID: PMC9090464 DOI: 10.36469/9808] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Introduction: Primary myelofibrosis (MF) is a rare hematologic disease belonging to the group of Philadelphia-negative chronic myeloproliferative neoplasms. Identification of the Janus Kinase (JAK) gene mutations inaugurated a new era in the targeted therapy of myeloproliferative diseases. Ruxolitinib is the first JAK1/JAK2 inhibitor specifically approved for the treatment of disease-related splenomegaly or symptoms in adult patients with primary myelofibrosis. The objective of this study was to assess the cost-effectiveness of ruxolitinib vs best available therapy (BAT) in MF patients in Spain. Methods: A decision-tree and Markov model were adapted to the Spanish setting to assess the cost-effectiveness of ruxolitinib vs. BAT on a lifetime horizon (≤15 years) from the societal perspective, while healthcare system perspective was included in the one-way sensitivity analysis. The population was assumed to be similar to that of the COMFORT-II clinical trial (CT), which was also the source of treatment efficacy data. BAT composition was derived from the same CT and validated with Spanish experts. Utilities were derived from the COMFORT-I CT. Costs included treatment, management, hospitalizations, emergency and outpatient visits, as well as adverse events and end-of-life costs. Additionally, costs associated to productivity loss were taken into account. Resource use was validated with experts and costs were extracted from Spanish sources. A probabilistic sensitivity analysis was also performed to evaluate the consistency of the results under the uncertainty or variability of the input data. Results: Patients on ruxolitinib accumulated 6.1 life years gained (LYGs), resulting in 73% extra life-years compared to patients treated with BAT (3.5LYs gained). Ruxolitinib provided 4.4 quality-adjusted life years (QALYs), with a 99% improvement compared to BAT (2.2 QALYs). This analysis gave an incremental cost of €47 199 per LYG and an incremental cost of €55 616 per QALY gained from the societal perspective. Conclusions: Ruxolitinib would be cost-effective in Spain according to the end-of-life criteria defined by the NICE and commonly referred for Spain (cost-effectiveness threshold of €61 500/QALY), in line with results published for other European countries.
Collapse
|
349
|
Griesshammer M, Sadjadian P. The BCR-ABL1-negative myeloproliferative neoplasms: a review of JAK inhibitors in the therapeutic armamentarium. Expert Opin Pharmacother 2017; 18:1929-1938. [PMID: 29134817 DOI: 10.1080/14656566.2017.1404574] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION The classical BCR-ABL1-negative myeloproliferative neoplasms (MPN) include primary myelofibrosis (PMF), polycythemia vera (PV) and essential thrombocythemia (ET). They are characterized by stem cell-derived clonal proliferation, harbor Janus kinase 2 (JAK2), or calreticulin (CALR), or myeloproliferative leukemia virus oncogene (MPL) driver mutations and exert an over activated JAK-signal transducer and activator of transcription (STAT) pathway. Therefore JAK inhibiting strategies have been successfully investigated in MPN clinical trials. Areas covered: The present review aims to provide a concise overview of the current and future role of JAK inhibitors in the therapeutic armamentarium of MPN. Expert opinion: The JAK1/JAK2 inhibitor ruxolitinib has clearly enriched the therapeutic armamentarium of MPN and is now licenced for more than five years in MF and over three years as second line in PV. Momelotinib, although of limited activity in MPN trials, demonstrated unique property of improving MF associated anemia. Less myelosuppressive or more selective JAK inhibitors like pacritinib, NS-01872 or Itacitinib are new promising agents tested in MF. JAK inhibition has become a cornerstone of MPN therapy and future efforts focus on ruxolitinib-based combinations and new JAK inhibitors.
Collapse
Affiliation(s)
- Martin Griesshammer
- a University Clinic for Haematology, Oncology, Haemostaseology and Palliative Care , Johannes Wesling Medical Center Minden, UKRUB, University of Bochum , Minden , Germany
| | - Parvis Sadjadian
- a University Clinic for Haematology, Oncology, Haemostaseology and Palliative Care , Johannes Wesling Medical Center Minden, UKRUB, University of Bochum , Minden , Germany
| |
Collapse
|
350
|
Efficacy and safety of ruxolitinib in regularly transfused patients with thalassemia: results from a phase 2a study. Blood 2017; 131:263-265. [PMID: 29097381 DOI: 10.1182/blood-2017-06-790121] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
|