1
|
Wachowiak J, Galimard JE, Dalissier A, Rihani R, AlSaedi H, Wynn RF, Dalle JH, Peffault de Latour R, Sedlacek P, Balduzzi A, Schroeder T, Bodova I, Gonzalez Vicent M, Gruhn B, Hamladji RM, Krivan G, Patrick K, Sobkowiak-Sobierajska A, Stepensky P, Unal A, Amrolia P, Perez Martinez A, Rialland F, Aljurf M, Isgro A, Toren A, Bierings M, Corbacioglu S, Kałwak K. Outcomes of allogeneic haematopoietic cell transplantation for myelofibrosis in children and adolescents: the retrospective study of the EBMT Paediatric Diseases WP. Bone Marrow Transplant 2024; 59:1057-1069. [PMID: 38627449 PMCID: PMC11296951 DOI: 10.1038/s41409-024-02286-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 03/28/2024] [Accepted: 04/09/2024] [Indexed: 08/04/2024]
Abstract
This retrospective study evaluated 35 children (median age 5.2 years; range 0.4-18) with myelofibrosis (MF), including 33 with primary myelofibrosis and 2 with secondary myelofibrosis transplanted from matched sibling donor (MSD) (n = 17) or non-MSD (n = 18) between 2000 and 2022. Conditioning was usually chemotherapy-based (n = 33) and myeloablative (n = 32). Fifteen patients received bone marrow (BM), 14 haematopoietic cells (HC) from peripheral blood (PB), and 6 from cord blood (CB). Day +100 acute GvHD II-IV incidence was significantly lower after MSD-haematopoietic cell transplantation (MSD-HCT) than after non-MSD-HCT [18.8% (4.3-41.1) vs 58.8% (31-78.6); p = 0.01]. Six-year non-relapse mortality (NRM) was 18% (7.1-32.8), relapse incidence was 15.9% (5.6-30.9), progression-free survival (PFS) was 66.1% (47-79.7), GvHD-free relapse-free survival was 50% (30.6-66.7), and overall survival (OS) was 71.1% (51.4-84). Six-year PFS and OS were significantly higher after BM transplantation compared to HCT from other sources [85.1% (52.3-96.1) vs 50.8% (26.3-71), p = 0.03, and 90.9% (50.8-98.7) vs 54% (28.1-74.2), p = 0.01, respectively], whereas NRM was significantly lower [0% vs 32% (12.3-53.9); p = 0.02]. This first multicentre study on outcomes of allogeneic HCT in children with myelofibrosis proves feasibility and curative effect of transplantation in these children, suggests that bone marrow transplantation is associated with better outcomes, and indicates the need for further studies.
Collapse
Affiliation(s)
- Jacek Wachowiak
- Department of Pediatric Oncology, Hematology and Transplantology, University of Medical Sciences, Poznan, Poland.
| | - Jacques-Emmanuel Galimard
- EBMT Paris Study Office, Paris, France
- Department of Haematology, Saint Antoine Hospital, INSERM UMR 938, Sorbonne University, Paris, France
| | - Arnaud Dalissier
- EBMT Paris Study Office, Paris, France
- Department of Haematology, Saint Antoine Hospital, INSERM UMR 938, Sorbonne University, Paris, France
| | - Rawad Rihani
- Department of Pediatrics, Pediatric Blood and Marrow Transplantation and Cellular Therapy Program, King Hussein Cancer Center, Amman, Jordan
| | - Hawazen AlSaedi
- Department of Pediatric Hematology and Oncology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Robert F Wynn
- Department of Blood and Marrow Transplant, Royal Manchester Children's Hospital, Manchester, UK
| | | | - Régis Peffault de Latour
- Department of Hematology and Stem Cell Transplantation, Saint Louis Hospital, Paris Cité Université, Paris, France
| | - Petr Sedlacek
- Department of Pediatric Hematology and Oncology, University Hospital Motol, Prague, Czechia
| | - Adriana Balduzzi
- Hematopoietic Stem Cell Transplant Unit, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- Department of Medicine and Surgery, Milano-Bicocca University, Milano, Italy
| | - Thomas Schroeder
- Department of Bone Marrow Transplantation, University of Essen, Essen, Germany
| | - Ivana Bodova
- University Children's Hospital, Bratislava, Slovakia
| | | | - Bernd Gruhn
- Department of Pediatrics, Jena University Hospital, Jena, Germany
| | - Rose-Marie Hamladji
- Centre Pierre et Marie Curie, Service Hématologie Greffe de Moëlle, Alger, Algeria
| | - Gergely Krivan
- Department of Paediatric Haematology and Stem Cell Transplantation, Central Hospital for Southern Pest, National Institute for Hematology and Infectology, Budapest, Hungary
| | - Katharine Patrick
- Sheffield Children's Hospital, Western Bank, Sheffield, United Kingdom
| | | | - Polina Stepensky
- BMT and Cancer Immunotherapy Department, Hadassah Hebrew University Medical Centre, Jerusalem, Israel
| | - Ali Unal
- Department Hematology and Bone Marrow Transplant Center Erciyes University Medical School, Kayseri, Turkey
| | - Persis Amrolia
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | | | - Fanny Rialland
- Hotel Dieu, CHU Nantes Dept. D'Hematologie, Nantes, France
| | - Mahmoud Aljurf
- King Faisal Specialist Hospital & Research Centre (Adult), Riyadh, Saudi Arabia
| | | | - Amos Toren
- Edmond & Lily Safra Children's Hospital, Division of Pediatric Hematology, Oncology & BMT, Sheba Medical Center, Tel_Hashomer, Israel
| | - Marc Bierings
- Princess Maxima Center, University Hospital for Children (WKZ), Stem cell transplantation, Utrecht, The Netherlands
| | - Selim Corbacioglu
- Department of Pediatrics, Division of Pediatric Oncology, Stem Cell Transplant, University Hospital of Regensburg, Regensburg, Germany
| | - Krzysztof Kałwak
- Department and Clinic of Pediatric Oncology, Hematology and Bone Marrow Transplantation, Wroclaw Medical University, Wroclaw, Poland
| |
Collapse
|
2
|
Tefferi A, Pardanani A. Momelotinib for myelofibrosis: our 14 years of experience with 100 clinical trial patients and recent FDA approval. Blood Cancer J 2024; 14:47. [PMID: 38499521 PMCID: PMC10948874 DOI: 10.1038/s41408-024-01029-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 02/20/2024] [Accepted: 03/05/2024] [Indexed: 03/20/2024] Open
Affiliation(s)
- Ayalew Tefferi
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN, USA.
| | - Animesh Pardanani
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
3
|
Tefferi A, Pardanani A, Gangat N. Momelotinib expands the therapeutic armamentarium for myelofibrosis: Impact on hierarchy of treatment choices. Am J Hematol 2024; 99:300-308. [PMID: 38164985 DOI: 10.1002/ajh.27163] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 11/05/2023] [Indexed: 01/03/2024]
Abstract
The primary objective of treatment in myelofibrosis (MF) is prolongation of life, which is currently accomplished only by allogeneic hematopoietic stem cell transplantation (AHSCT). Determination of optimal timing for AHSCT is facilitated by molecular risk stratification. Non-transplant treatment options in MF are palliative in scope and include Janus kinase 2 (JAK2) inhibitors (JAKi): momelotinib (FDA approved on September 15, 2023), ruxolitinib (November 16, 2011), fedratinib (August 16, 2019), and pacritinib (February 28, 2022); all four JAKi are effective in reducing spleen size and alleviating symptoms, considered a drug class effect and attributed to their canonical JAK-STAT inhibitory mechanism of action. In addition, momelotinib exhibits erythropoietic effect, attributed to alleviation of ineffective erythropoiesis through inhibition of activin A receptor type-I (ACVR1). In transplant-ineligible or deferred patients, the order of treatment preference is based on specific symptoms and individual assessment of risk tolerance. Because of drug-induced immunosuppression and other toxicities attributed to JAKi, we prefer non-JAKi drugs as initial treatment for MF-associated anemia that is not accompanied by treatment-requiring splenomegaly or constitutional symptoms. Otherwise, it is reasonable to consider momelotinib as the first-line JAKi treatment of choice, in order to target the triad of quality-of-life offenders in MF: anemia, splenomegaly, and constitutional symptoms/cachexia. For second-line therapy, we favor ruxolitinib, over fedratinib, based on toxicity profile. Pacritinib and fedratinib provide alternative options in the presence of severe thrombocytopenia or ruxolitinib-resistance/intolerance, respectively. Splenectomy remains a viable option for drug-resistant symptomatic splenomegaly and cytopenia.
Collapse
Affiliation(s)
- Ayalew Tefferi
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Animesh Pardanani
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Naseema Gangat
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
4
|
McLornan DP, Psaila B, Ewing J, Innes A, Arami S, Brady J, Butt NM, Cargo C, Cross NCP, Francis S, Frewin R, Garg M, Godfrey AL, Green A, Khan A, Knapper S, Lambert J, McGregor A, McMullin MF, Nangalia J, Neelakantan P, Woodley C, Mead A, Somervaille TCP, Harrison CN. The management of myelofibrosis: A British Society for Haematology Guideline. Br J Haematol 2024; 204:136-150. [PMID: 38037886 DOI: 10.1111/bjh.19186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 10/03/2023] [Accepted: 10/20/2023] [Indexed: 12/02/2023]
Affiliation(s)
- Donal P McLornan
- Department of Haematology, University College London Hospitals, London, UK
| | - Bethan Psaila
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Haematology, Churchill Hospital, Oxford University NHS Trust, Oxford, UK
| | - Joanne Ewing
- Department of Haematology, University Hospitals Birmingham Trust, London, UK
| | - Andrew Innes
- Department of Haematology, Imperial College, London, UK
| | - Siamak Arami
- Department of Haematology, London Northwest Healthcare University NHS Trust, London, UK
| | - Jessica Brady
- Department of Clinical Oncology, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Nauman M Butt
- Department of Haematology, The Clatterbridge Cancer Centre NHS Foundation Trust, Liverpool, UK
| | - Catherine Cargo
- Department of Haematology, Leeds Teaching Hospitals NHS Foundation Trust, Leeds, UK
| | | | - Sebastian Francis
- Department of Haematology, Sheffield Teaching Hospital NHS Foundation Trust, Sheffield, UK
| | - Rebecca Frewin
- Department of Haematology, Gloucestershire Hospitals NHS Foundation Trust, Gloucester, UK
| | - Mamta Garg
- Department of Haematology, University Hospitals Leicester NHS Trust, Leicester, UK
| | - Anna L Godfrey
- Haematopathology & Oncology Diagnostics Service, Department of Haematology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Anna Green
- Department of Histopathology, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Alesia Khan
- Department of Haematology, Leeds Teaching Hospitals NHS Foundation Trust, Leeds, UK
| | - Steve Knapper
- Department of Haematology, Cardiff University, Cardiff, UK
| | - Jonathan Lambert
- Department of Haematology, University College London Hospitals, London, UK
| | | | | | - Jyoti Nangalia
- Wellcome Sanger Institute, University of Cambridge, Cambridge, UK
| | - Pratap Neelakantan
- Department of Haematology, Royal Berkshire NHS Foundation Trust, London, UK
| | - Claire Woodley
- Department of Haematology, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Adam Mead
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Haematology, Churchill Hospital, Oxford University NHS Trust, Oxford, UK
| | - Tim C P Somervaille
- Cancer Research UK Manchester Institute & The Christie NHS Foundation Trust, Manchester, UK
| | - Claire N Harrison
- Department of Haematology, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| |
Collapse
|
5
|
Palmer J. Are transplant indications changing for myelofibrosis? HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2023; 2023:676-681. [PMID: 38066916 PMCID: PMC10727025 DOI: 10.1182/hematology.2023000453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
Myelofibrosis is a devastating myeloid malignancy characterized by dysregulation of the JAK-STAT pathway, resulting in splenomegaly, constitutional symptoms, anemia, thrombocytopenia, leukocytosis, and an increased likelihood of progression to acute leukemia. The only curative option is allogeneic stem cell transplantation. The numbers of transplants have been increasing every year, and although there have been improvements in survival, there remain many unanswered questions. In this review, we will evaluate patient selection and appropriate timing for transplantation. We will cover the current prognostic scoring systems, which can aid in the decision of when to move forward with transplant. We will also review the different donor options, as well as the conditioning regimens. The peritransplant management of splenomegaly will be reviewed. We will discuss management of posttransplant complications such as loss of donor chimerism or disease relapse. Finally, we will review what is known about the outlook of patients who have undergone allogeneic stem cell transplant with regards to quality of life and long-term survival.
Collapse
Affiliation(s)
- Jeanne Palmer
- Division of Hematology/Medical Oncology, Mayo Clinic, Phoenix, AZ
| |
Collapse
|
6
|
Amé S, Barraco F, Ianotto J, Jourdan E, Rey J, Viallard J, Wémeau M, Kiladjian J. Advances in management of primary myelofibrosis and polycythaemia vera: Implications in clinical practice. EJHAEM 2023; 4:779-791. [PMID: 37601853 PMCID: PMC10435696 DOI: 10.1002/jha2.734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 05/31/2023] [Indexed: 08/22/2023]
Abstract
Primary myelofibrosis (PMF) and polycythaemia vera (PV) are rare BCR-ABL1-negative myeloproliferative neoplasms, associated with an increased risk of thrombosis, haemorrhagic complications and progression to fibrosis or leukaemia or fibrosis for PV. Both diseases are characterised by biological and clinical heterogeneity, leading to great variability in their management in routine clinical practice. In this review, we present an updated overview of the diagnosis, prognosis and treatment of PMF and PV, and we discuss how our multidisciplinary expert group based across France translates this evidence-based knowledge into routine clinical practice.
Collapse
Affiliation(s)
- Shanti Amé
- Department of HaematologyInstitut de Cancérologie Strasbourg Europe (ICANS)StrasbourgFrance
| | - Fiorenza Barraco
- Department of HaematologyLyon Sud Hospital CentrePierre‐BéniteFrance
| | | | - Eric Jourdan
- Department of Clinical HaematologyUniversity Hospital of NimesNimesFrance
| | - Jérôme Rey
- Department of HaematologyInstitute Paoli‐CalmettesMarseilleFrance
| | | | - Mathieu Wémeau
- Department of HaematologyHospital Centre of RoubaixRoubaixFrance
| | | |
Collapse
|
7
|
Passamonti F, Harrison CN, Mesa RA, Kiladjian JJ, Vannucchi AM, Verstovsek S. Anemia in myelofibrosis: current and emerging treatment options. Crit Rev Oncol Hematol 2022; 180:103862. [DOI: 10.1016/j.critrevonc.2022.103862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 10/17/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022] Open
|
8
|
Sastow D, Mascarenhas J, Tremblay D. Thrombocytopenia in Patients With Myelofibrosis: Pathogenesis, Prevalence, Prognostic Impact, and Treatment. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2022; 22:e507-e520. [PMID: 35221248 DOI: 10.1016/j.clml.2022.01.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 01/26/2022] [Accepted: 01/28/2022] [Indexed: 06/14/2023]
Abstract
Myelofibrosis (MF) is a clonal hematopoietic stem cell neoplasm, characterized by pathologic myeloproliferation associated with inflammatory and pro-angiogenic cytokine release, that results in functional compromise of the bone marrow. Thrombocytopenia is a disease-related feature of MF, which portends a poor prognosis impacting overall survival (OS) and leukemia free survival. Thrombocytopenia in MF has multiple causes including ineffective hematopoiesis, splenic sequestration, and treatment-related effects. Presently, allogeneic hematopoietic stem cell transplantation (HSCT) remains the only curable treatment for MF, which, unfortunately, is only a viable option for a minority of patients. All other currently available therapies are either focused on improving cytopenias or the alleviating systemic symptoms and burdensome splenomegaly. While JAK2 inhibitors have moved to the forefront of MF therapy, available JAK inhibitors are advised against in patients with severe thrombocytopenia (platelets < 50 × 109/L). In this review, we describe the pathogenesis, prevalence, and prognostic significance of thrombocytopenia in MF. We also explore the value and limitations of treatments directed at addressing cytopenias, splenomegaly and symptom burden, and those with potential disease modification. We conclude by proposing a treatment algorithm for patients with MF and severe thrombocytopenia.
Collapse
Affiliation(s)
- Dahniel Sastow
- Department of Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - John Mascarenhas
- Division of Hematology and Medical Oncology, Tisch Cancer Institute Icahn School of Medicine at Mount Sinai, New York, NY
| | - Douglas Tremblay
- Division of Hematology and Medical Oncology, Tisch Cancer Institute Icahn School of Medicine at Mount Sinai, New York, NY.
| |
Collapse
|
9
|
Salit RB. The role of JAK inhibitors in hematopoietic cell transplantation. Bone Marrow Transplant 2022; 57:857-865. [PMID: 35388118 DOI: 10.1038/s41409-022-01649-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 02/07/2022] [Accepted: 03/16/2022] [Indexed: 01/03/2023]
Abstract
The Janus Kinase (JAK)/Signal Transducers and Activators of Transcription (STAT) pathway is essential for both the regulation of hematopoiesis and the control of inflammation. Disruption of this pathway can lead to inflammatory and malignant disease processes. JAK inhibitors, designed to control the downstream effects of pro-inflammatory and pro-angiogenic cytokines, have been successfully used in pre-clinical models and clinical studies of patients with autoimmune diseases, hematologic malignancies, and the hematopoietic cell transplantation (HCT) complication graft versus host disease (GVHD). In the last decade, JAK inhibitors Ruxolitinib, Fedratinib, and most recently Pacritinib have been United States Federal Drug Administration (FDA) approved for the treatment of myelofibrosis (MF). Ruxolitinib was also recently approved for the treatment of steroid refractory acute as well as chronic GVHD; JAK inhibitors are currently under evaluation in the pre-HCT setting in MF and for the prevention of GVHD. This review will focus on the role of JAK inhibitors in the treatment of hematologic malignancies, the potential function of pre-HCT JAK inhibitors in patients with MF, and the role of JAK inhibitors in the prevention and treatment of acute and chronic GVHD.
Collapse
Affiliation(s)
- Rachel B Salit
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.
- Department of Medicine, University of Washington Medical Center, Seattle, WA, USA.
| |
Collapse
|
10
|
Yilmaz M, Verstovsek S. Managing patients with myelofibrosis and thrombocytopenia. Expert Rev Hematol 2022; 15:233-241. [PMID: 35316110 DOI: 10.1080/17474086.2022.2057296] [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: 11/04/2022]
Abstract
INTRODUCTION : Given the progressive nature of myelofibrosis, the incidence of thrombocytopenia increases over time. Furthermore, approved drugs ruxolitinib and fedratinib, induce thrombocytopenia. Hence, treatment of myelofibrosis patients with low platelet counts is an unmet need. AREAS COVERED : This review summarizes the current and emerging treatment options available for patients with myelofibrosis and thrombocytopenia. In the first section of this review, we summarized the use of JAK inhibitors in patients with thrombocytopenia, and in the second part, we focused on use of therapies other than JAK Inhibitors such as steroids, immunomodulatory agents, androgens and other novel agents. EXPERT OPINION : Up to 25% of patients with myelofibrosis have platelet counts below 100,000 at presentation. Patients with thrombocytopenia are more likely to be anemic and PRBC transfusion-dependent, as well as have high-risk disease characteristics and a poor overall survival rate.. Among all JAK inhibitors studied in phase 3 clinical trials, pacritinib seems not to induce significant thrombocytopenia while maintaining a good spleen response. Severe thrombocytopenia is a major impediment to myelofibrosis therapy, and more research, particularly on novel therapeutic agents aimed at cytopenic patient populations, is needed.
Collapse
Affiliation(s)
- Musa Yilmaz
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Srdan Verstovsek
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
11
|
Yoon J, Pettit K. Improving symptom burden and quality of life in patients with myelofibrosis: current strategies and future directions. Expert Rev Hematol 2021; 14:607-619. [PMID: 34148506 DOI: 10.1080/17474086.2021.1944096] [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: 10/21/2022]
Abstract
INTRODUCTION Myelofibrosis (MF) is a complex and aggressive hematologic malignancy resulting from JAK/STAT-driven myeloproliferation and abnormal fibrogenesis. The clinical manifestations are heterogeneous and negatively impact quality of life and survival. JAK inhibitors improve symptoms and splenomegaly to a variable degree in a proportion of patients, but the effects for many patients are insufficient or short-lived. AREAS COVERED This review examines the constellation of symptoms that befall patients with MF, describes methods to quantify and serially monitor these symptoms, and evaluates pharmacologic and non-pharmacologic interventions for disease-related symptoms. The review also includes a discussion of areas of unmet medical need, and proposes future methods for meeting this need. EXPERT OPINION The treatment landscape for MF is evolving rapidly. The most effective therapies or combinations of therapies will likely simultaneously impact both the malignant hematopoietic stem cell and mechanisms of aberrant fibrogenesis that drive this disease. The goals of treatment for patients with myelofibrosis should be to improve length and quality of life. Clinical trials must be designed with these goals in mind, with endpoints focused on overall survival and symptom reduction, as opposed to surrogate endpoints such as spleen volume reduction.
Collapse
Affiliation(s)
- James Yoon
- Department of Medicine, Division of Hematology/Oncology, Michigan Medicine and University of Michigan Medical School, Ann Arbor, Michigan, United States
| | - Kristen Pettit
- Department of Medicine, Division of Hematology/Oncology, Michigan Medicine and University of Michigan Medical School, Ann Arbor, Michigan, United States
| |
Collapse
|
12
|
Placidi L, Nardini M, Cusumano D, Boldrini L, Catucci F, Chiloiro G, Votta C, Valentini V, Indovina L. Dosimetric accuracy of dual isocenter irradiation in low magnetic field resonance guided radiotherapy system for extended abdominal tumours. Phys Med 2021; 84:149-158. [PMID: 33895666 DOI: 10.1016/j.ejmp.2021.03.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 03/23/2021] [Accepted: 03/30/2021] [Indexed: 10/21/2022] Open
Abstract
PURPOSE Due to limited field size of Magnetic Resonance Linear Accelerators (MR-Linac), some treatments could require a dual-isocenter planning approach to achieve a complete target coverage and thus exploit the benefits of the online adaptation. This study evaluates the dosimetric accuracy of the dual-isocenter intensity modulated radiation therapy (IMRT) delivery technique for MR-Linac. MATERIAL AND METHODS Dual-isocenter multi leaf collimator (MLC) and couch accuracy tests have been performed to evaluate the delivery accuracy of the system. A mono-isocenter plan delivered in clinical practice has then been retrospectively re-planned with dual-isocenter technique. The dual-isocenter plan has been re-calculated and delivered on a 3-dimensional (3D) ArcCHECK phantom and 2-dimensional (2D) films to assess its dosimetric accuracy in terms of gamma analysis. Clinical and planning target volume (CTV and PTV respectively) coverage robustness was then investigated after the introduction of ± 2 mm and ± 5 mm positioning errors by shifting the couch. RESULTS MLC and couch accuracy tests confirmed the system accuracy in delivering a dual-isocenter irradiation. 2D/3D gamma analysis results occurred always to be above 95% if considered a gamma criteria 1%/2 mm and 1%/1 mm respectively for the 2D and 3D analysis. The mean variations for CTV D98% and PTV V95% were 0.2% and 1.1% respectively when positioning error was introduced separately in each direction, while the maximum observed variations were 0.9% (CTV) and 3.7% (PTV). CONCLUSION The dosimetric accuracy of dual-isocenter irradiation has been verified for MR-Linac, achieving accurate and robust treatment strategy and improving dose conformality also in presence of targets whose extension exceeds the nominal maximum field size.
Collapse
Affiliation(s)
- L Placidi
- Fondazione Policlinico Universitario ''A. Gemelli'' IRCCS, Italy; Università Cattolica del Sacro Cuore, Rome, Italy
| | - M Nardini
- Fondazione Policlinico Universitario ''A. Gemelli'' IRCCS, Italy; Università Cattolica del Sacro Cuore, Rome, Italy.
| | - D Cusumano
- Fondazione Policlinico Universitario ''A. Gemelli'' IRCCS, Italy
| | - L Boldrini
- Fondazione Policlinico Universitario ''A. Gemelli'' IRCCS, Italy
| | - F Catucci
- Fondazione Policlinico Universitario ''A. Gemelli'' IRCCS, Italy
| | - G Chiloiro
- Fondazione Policlinico Universitario ''A. Gemelli'' IRCCS, Italy
| | - C Votta
- Fondazione Policlinico Universitario ''A. Gemelli'' IRCCS, Italy
| | - V Valentini
- Fondazione Policlinico Universitario ''A. Gemelli'' IRCCS, Italy; Università Cattolica del Sacro Cuore, Rome, Italy
| | - L Indovina
- Fondazione Policlinico Universitario ''A. Gemelli'' IRCCS, Italy
| |
Collapse
|
13
|
Polverelli N, Mauff K, Kröger N, Robin M, Beelen D, Beauvais D, Chevallier P, Mohty M, Passweg J, Rubio MT, Maertens J, Finke J, Bornhäuser M, Vrhovac R, Helbig G, Mear J, Castagna L, Reményi P, Angelucci E, Karakasis D, Rifòn J, Sirait T, Russo D, Wreede L, Czerw T, Hernández‐Boluda JC, Hayden P, McLornan D, Yakoub‐Agha I. Impact of spleen size and splenectomy on outcomes of allogeneic hematopoietic cell transplantation for myelofibrosis: A retrospective analysis by the chronic malignancies working party on behalf of European society for blood and marrow transplantation (EBMT). Am J Hematol 2021; 96:69-79. [PMID: 33064301 DOI: 10.1002/ajh.26020] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 10/07/2020] [Accepted: 10/12/2020] [Indexed: 12/12/2022]
Abstract
The role of spleen size and splenectomy for the prediction of post-allogeneic hematopoietic stem cell transplant (allo-HCT) outcome in myelofibrosis remains under debate. In EBMT registry, we identified a cohort of 1195 myelofibrosis patients transplanted between 2000-2017 after either fludarabine-busulfan or fludarabine-melphalan regimens. Overall, splenectomy was performed in 202 (16.9%) patients and its use decreased over time (28.3% in 2000-2009 vs 14.1% in 2010-2017 period). By multivariate analysis, splenectomy was associated with less NRM (HR 0.64, 95% CI 0.44-0.93, P = .018) but increased risk of relapse (HR 1.43, 95% CI 1.01-2.02, P = .042), with no significant impact on OS (HR 0.86, 95% CI 0.67-1.12, P = .274). However, in subset analysis comparing the impact of splenectomy vs specific spleen sizes, for patients with progressive disease, an improved survival was seen in splenectomised subjects compared to those patients with a palpable spleen length ≥ 15 cm (HR 0.44, 95% CI 0.28-0.69, P < .001), caused by a significant reduction in NRM (HR 0.26, 95% CI 0.14-0.49, P < .001), without significantly increased relapse risk (HR 1.47, 95% CI 0.87-2.49, P = .147). Overall, despite the possible biases typical of retrospective cohorts, this study highlights the potential detrimental effect of massive splenomegaly in transplant outcome and supports the role of splenectomy for myelofibrosis patients with progressive disease and large splenomegaly.
Collapse
Affiliation(s)
- Nicola Polverelli
- Unit of Blood Diseases and Stem Cells Transplantation, Department of Clinical and Experimental Sciences University of Brescia ASST Spedali Civili of Brescia
| | | | - Nicolaus Kröger
- Department of Hematology University Hospital Eppendorf Hamburg Germany
| | - Marie Robin
- Department of Hematology Hôpital Saint‐Louis, APHP Paris France
| | - Dietrich Beelen
- Department of Hematology University Hospital of Essen Essen Germany
| | | | | | - Mohamad Mohty
- Sorbonne University Paris France
- Service d'Hématologie Clinique et Thérapie cellulaire Hopital Saint‐Antoine Paris France
- INSERM Paris France
| | - Jakob Passweg
- Department of Hematology University Hospital Basel Basel Switzerland
| | | | - Johan Maertens
- Department of Hematology University Hospital Gasthuisberg Leuven Belgium
| | - Jürgen Finke
- Department of Hematology University of Freiburg Freiburg Germany
| | - Martin Bornhäuser
- Department of Hematology University Hospital Dresded Dresden Germany
| | - Radovan Vrhovac
- Department of Hematology University Hospital Center Rebro Zagreb Croatia
| | - Grzegorz Helbig
- Department of Hematology Silesian Medical Academy Katowice Poland
| | - Jean‐Baptiste Mear
- Department of Hematology Centre Hospitalier Universitaire de Rennes Rennes France
| | - Luca Castagna
- Department of Hematology Centre de Recherche en Cancérologie de Marseille Marseille France
| | - Péter Reményi
- Department of Hematology Dél‐pesti Centrumkórház Budapest Hungary
| | - Emanuele Angelucci
- Hematology and Transplant Center IRCCS Ospedale Policlinico San Martino Genoa Italy
| | | | - Jose Rifòn
- Department of Hematology Clínica Universitaria de Navarra Pamplona Spain
| | | | - Domenico Russo
- Unit of Blood Diseases and Stem Cells Transplantation, Department of Clinical and Experimental Sciences University of Brescia ASST Spedali Civili of Brescia
| | - Liesbeth Wreede
- Department of Biomedical Data Sciences LUMC Leiden Netherlands
| | - Tomasz Czerw
- Department of Bone Marrow Transplantation and Onco‐Hematology Maria Sklodowska‐Curie National Research Institute of Oncology Gliwice Poland
| | | | - Patrick Hayden
- Department of Hematology St. James's Hospital Dublin Ireland
| | - Donal McLornan
- Department of Hematology Guy's and St Thomas' NHS Foundation Trust and University College London Hospitals London UK
| | | |
Collapse
|
14
|
Tefferi A. Primary myelofibrosis: 2021 update on diagnosis, risk-stratification and management. Am J Hematol 2021; 96:145-162. [PMID: 33197049 DOI: 10.1002/ajh.26050] [Citation(s) in RCA: 178] [Impact Index Per Article: 59.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 11/11/2020] [Indexed: 12/16/2022]
Abstract
DISEASE OVERVIEW Primary myelofibrosis (PMF) is a myeloproliferative neoplasm (MPN) characterized by stem cell-derived clonal myeloproliferation that is often but not always accompanied by JAK2, CALR, or MPL mutations. Additional disease features include bone marrow reticulin/collagen fibrosis, aberrant inflammatory cytokine expression, anemia, hepatosplenomegaly, extramedullary hematopoiesis (EMH), constitutional symptoms, cachexia, leukemic progression, and shortened survival. DIAGNOSIS Bone marrow morphology is the primary basis for diagnosis. Presence of JAK2, CALR, or MPL mutation, expected in around 90% of the patients, is supportive but not essential for diagnosis; these mutations are also prevalent in the closely related MPNs, namely polycythemia vera (PV) and essential thrombocythemia (ET). The 2016 World Health Organization classification system distinguishes "prefibrotic" from "overtly fibrotic" PMF; the former might mimic ET in its presentation. Furthermore, approximately 15% of patients with ET or PV might progress into a PMF-like phenotype (post-ET/PV MF) during their clinical course. ADVERSE MUTATIONS SRSF2, ASXL1, and U2AF1-Q157 mutations predict inferior survival in PMF, independent of each other and other risk factors. RAS/CBL mutations predicted resistance to ruxolitinib therapy. ADVERSE KARYOTYPE Very high risk abnormalities include -7, inv (3), i(17q), +21, +19, 12p-, and 11q-. RISK STRATIFICATION Two new prognostic systems for PMF have recently been introduced: GIPSS (genetically-inspired prognostic scoring system) and MIPSS70+ version 2.0 (MIPSSv2; mutation- and karyotype-enhanced international prognostic scoring system). GIPSS is based exclusively on mutations and karyotype. MIPSSv2 includes, in addition, clinical risk factors. GIPSS features four and MIPSSv2 five risk categories. RISK-ADAPTED THERAPY Observation alone is advised for MIPSSv2 "low" and "very low" risk disease (estimated 10-year survival 56%-92%); allogeneic hematopoietic stem cell transplant (AHSCT) is the preferred treatment for "very high" and "high" risk disease (estimated 10-year survival 0%-13%); treatment-requiring patients with intermediate-risk disease (estimated 10-year survival 30%) are best served by participating in clinical trials. In non-transplant candidates, conventional treatment for anemia includes androgens, prednisone, thalidomide, and danazol; for symptomatic splenomegaly, hydroxyurea and ruxolitinib; and for constitutional symptoms, ruxolitinib. Fedratinib, another JAK2 inhibitor, has now been FDA-approved for use in ruxolitinib failures. Splenectomy is considered for drug-refractory splenomegaly and involved field radiotherapy for non-hepatosplenic EMH and extremity bone pain. NEW DIRECTIONS A number of new agents, alone or in combination with ruxolitinib, are currently under investigation for MF treatment (ClinicalTrials.gov); preliminary results from some of these clinical trials were presented at the 2020 ASH annual meeting and highlighted in the current document.
Collapse
Affiliation(s)
- Ayalew Tefferi
- Division of Hematology, Department of Medicine Mayo Clinic Rochester Minnesota USA
| |
Collapse
|
15
|
Genthon A, Killian M, Mertz P, Cathebras P, Gimenez De Mestral S, Guyotat D, Chalayer E. [Myelofibrosis: A review]. Rev Med Interne 2020; 42:101-109. [PMID: 33243417 DOI: 10.1016/j.revmed.2020.08.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 08/28/2020] [Accepted: 08/30/2020] [Indexed: 11/28/2022]
Abstract
Myelofibrosis is a BCR-ABL1-negative chronic myeloproliferative neoplasm that includes primary myelofibrosis, post-polycythemia vera myelofibrosis, and post-essential thrombocythemia myelofibrosis. It is characterized by stem cell-derived clonal proliferation that is often, but not always, accompanied by somatic mutations, which are classified into driver mutations (JAK2, CALR, or MPL), subclonal mutations and fibrosis on bone marrow biopsy. Myelofibrosis commonly demonstrates splenomegaly, constitutional symptoms, anemia, thrombocytosis, or thrombocytopenia. Patients may also be asymptomatic. Complications as thromboembolic or hemorrhagic events can reveal the disease. Primary myelofibrosis is the least common myeloproliferative neoplasm but is associated with poor survival and acute leukemic transformation. In contrast to the significant progress made in understanding the disease's pathogenesis, treatment for myelofibrosis remains largely palliative. The JAK2 inhibitor, ruxolitinib is not sufficient in eliminating the underlying myeloid progenitor clone, as disease inevitably returns with therapy discontinuation. Allogeneic hematopoietic stem cell transplantation is the only therapeutic option that offers potential cure. The development of novel treatment strategies aimed at slowing or even reversing disease progression, prolonging patient survival and preventing evolution to blast-phase are still lacking.
Collapse
Affiliation(s)
- A Genthon
- Service d'hématologie clinique et de thérapie cellulaire, hôpital Saint-Antoine, AP-HP, Paris, France; Médecine Sorbonne université, Paris, France
| | - M Killian
- Service de médecine interne, hôpital Nord, CHU de Saint-Étienne, 42055 Saint-Étienne cedex 02, France
| | - P Mertz
- Service de rhumatologie, hôpitaux universitaires de Strasbourg, Strasbourg, France; Inserm UMR_S1109, laboratoire d'immunorhumatologie moléculaire, Centre national de référence des maladies systémiques et autoimmunes rares Est Sud-Ouest (RESO), université de Strasbourg, 67000 Strasbourg, France
| | - P Cathebras
- Service de médecine interne, hôpital Nord, CHU de Saint-Étienne, 42055 Saint-Étienne cedex 02, France
| | - S Gimenez De Mestral
- Pathology department, Sorbonne université, hôpital Saint-Antoine, AP-HP, 75012 Paris, France
| | - D Guyotat
- Département d'hématologie et thérapie cellulaire, institut de cancérologie Lucien-Neuwirth, Saint-Étienne, France
| | - E Chalayer
- Département d'hématologie et thérapie cellulaire, institut de cancérologie Lucien-Neuwirth, Saint-Étienne, France; Inserm, SAINBIOSE, U1059, dysfonction vasculaire et hémostase, université Jean-Monnet, Saint-Étienne, France.
| |
Collapse
|
16
|
Dunbar AJ, Rampal RK, Levine R. Leukemia secondary to myeloproliferative neoplasms. Blood 2020; 136:61-70. [PMID: 32430500 PMCID: PMC7332899 DOI: 10.1182/blood.2019000943] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 01/23/2020] [Indexed: 12/11/2022] Open
Abstract
Secondary acute myeloid leukemias (AMLs) evolving from an antecedent myeloproliferative neoplasm (MPN) are characterized by a unique set of cytogenetic and molecular features distinct from de novo AML. Given the high frequency of poor-risk cytogenetic and molecular features, malignant clones are frequently insensitive to traditional AML chemotherapeutic agents. Allogeneic stem cell transplant, the only treatment modality shown to have any beneficial long-term outcome, is often not possible given the advanced age of patients at time of diagnosis and frequent presence of competing comorbidities. Even in this setting, relapse rates remain high. As a result, outcomes are generally poor and there remains a significant unmet need for novel therapeutic strategies. Although advances in cancer genomics have dramatically enhanced our understanding of the molecular events governing clonal evolution in MPNs, the cell-intrinsic and -extrinsic mechanisms driving leukemic transformation at this level remain poorly understood. Here, we review known risk factors for the development of leukemic transformation in MPNs, recent progress made in our understanding of the molecular features associated with leukemic transformation, current treatment strategies, and emerging therapeutic options for this high-risk myeloid malignancy.
Collapse
MESH Headings
- Abnormal Karyotype
- Allografts
- Antineoplastic Agents/therapeutic use
- Cell Transformation, Neoplastic
- Chromosome Aberrations
- Clonal Evolution
- Combined Modality Therapy
- Comorbidity
- Disease Progression
- Drug Resistance, Neoplasm
- Drugs, Investigational/therapeutic use
- Genes, Neoplasm
- Hematopoietic Stem Cell Transplantation
- Humans
- Leukemia, Myeloid, Acute/etiology
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/therapy
- Models, Biological
- Mutation
- Myeloproliferative Disorders/genetics
- Myeloproliferative Disorders/pathology
- Neoplasm Proteins/genetics
- Recurrence
- Risk Factors
- Single-Cell Analysis
- Therapies, Investigational
Collapse
Affiliation(s)
- Andrew J Dunbar
- Leukemia Service, Department of Medicine
- Center for Hematologic Malignancies
| | - Raajit K Rampal
- Leukemia Service, Department of Medicine
- Center for Hematologic Malignancies
| | - Ross Levine
- Leukemia Service, Department of Medicine
- Center for Hematologic Malignancies
- Human Oncology and Pathogenesis Program, and
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
17
|
Asher S, McLornan DP, Harrison CN. Current and future therapies for myelofibrosis. Blood Rev 2020; 42:100715. [PMID: 32536371 DOI: 10.1016/j.blre.2020.100715] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 07/21/2019] [Accepted: 05/05/2020] [Indexed: 12/16/2022]
Abstract
Myelofibrosis is classified as a 'Philadelphia-chromosome negative' clonal myeloproliferative disorder. The heterogeneity of this condition and patient population and array of often challenging clinical manifestations can frequently make therapeutic decisions challenging. Despite many advances in therapy with targeted and combination approaches, following an enhanced understanding of underlying disease pathogenesis, cure only remains achievable with allogeneic stem cell transplant. This option is often limited to a small group of younger transplant-eligible patients with more advanced disease who have both a suitable donor and no or few co-morbidities. In this article, we will discuss up-to-date disease prognostication, common clinical challenges associated with myelofibrosis and both standard and novel therapeutic approaches. Increasingly complex prognostic modelling utilises patient-specific, haematological and genomic parameters to improve the accuracy of risk assessment and predict disease progression. We will also focus on difficult clinical scenarios such as disease-associated anaemia, thrombocytopenia and extremes of age. Future and evolving therapies within this field are highly anticipated and novel JAK inhibitor and non-JAK inhibitor-based therapy will also be discussed, including the new challenge of how to switch from one JAK inhibitor therapy to another.
Collapse
Affiliation(s)
- Samir Asher
- Department of Haematology, Guy's and St Thomas' NHS Foundation Trust, London SE1 9RT, UK
| | - Donal P McLornan
- Department of Haematology, Guy's and St Thomas' NHS Foundation Trust, London SE1 9RT, UK
| | - Claire N Harrison
- Department of Haematology, Guy's and St Thomas' NHS Foundation Trust, London SE1 9RT, UK.
| |
Collapse
|
18
|
Tremblay D, Schwartz M, Bakst R, Patel R, Schiano T, Kremyanskaya M, Hoffman R, Mascarenhas J. Modern management of splenomegaly in patients with myelofibrosis. Ann Hematol 2020; 99:1441-1451. [PMID: 32417942 DOI: 10.1007/s00277-020-04069-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 04/28/2020] [Indexed: 12/17/2022]
Abstract
Myelofibrosis (MF) is a chronic myeloproliferative neoplasm which can lead to massive splenomegaly secondary to extramedullary hematopoiesis. Patients frequently exhibit debilitating symptoms including pain and early satiety, in addition to cellular sequestration causing severe cytopenias. JAK 1/2 inhibitors, such as ruxolitinib and fedratinib, are the mainstay of therapy and produce significant and durable reductions in spleen volume. However, many patients are not eligible for JAK 2 inhibitor therapy or become refractory to treatment over time. Novel therapies are in development that can reduce the degree of splenomegaly for some of these patients. However, splenectomy, splenic irradiation, and partial splenic artery embolization remain valuable therapeutic options in select patients. In this review, we will discuss currently available pharmacologic therapies and describe promising drugs currently in development. We will also delve into the efficacy and safety concerns of splenectomy, splenic irradiation, and partial splenic artery embolization. Finally, we will propose a treatment algorithm to help guide clinicians in the management of symptomatic splenomegaly in patients with MF.
Collapse
Affiliation(s)
- Douglas Tremblay
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY, USA
| | - Myron Schwartz
- Recanati/Miller Transplantation Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Richard Bakst
- Department of Radiation of Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rahul Patel
- Division of Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Thomas Schiano
- Recanati/Miller Transplantation Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Marina Kremyanskaya
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY, USA
| | - Ronald Hoffman
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY, USA
| | - John Mascarenhas
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY, USA.
| |
Collapse
|
19
|
Malato A, Rossi E, Tiribelli M, Mendicino F, Pugliese N. Splenectomy in Myelofibrosis: Indications, Efficacy, and Complications. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2020; 20:588-595. [PMID: 32482540 DOI: 10.1016/j.clml.2020.04.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 04/20/2020] [Accepted: 04/22/2020] [Indexed: 10/24/2022]
Abstract
Splenomegaly, which may range from a few centimeters below the left costal border to massive dimensions, is one of the most characteristic features in patients with advanced myelofibrosis (MF). Splenectomy may offer an effective therapeutic option for treating massive splenomegaly in patients with MF, and especially in cases of disease refractory to conventional drugs, but it is associated with a number of complications as well as substantial morbidity and mortality. Whether splenectomy should be performed before allogeneic hematopoietic stem-cell transplantation is also controversial, and there is a lack of prospective randomized clinical trials that assess the role of splenectomy before hematopoietic stem-cell transplantation in patients with MF. Although splenectomy is not routinely performed before transplantation, it may be appropriate in patients with massive splenomegaly and related symptoms, so long as the higher risk of graft failure in such cases is taken into account. This review aims to describe the efficacy, indications, and complications of splenectomy in patients with MF; and to evaluate the long-term impact of splenectomy on patient survival and risk of disease transformation.
Collapse
Affiliation(s)
- Alessandra Malato
- UOC di Ematologia I ad Indirizzo Oncologico, Ospedali Riuniti Villa Sofia-Cervello, Palermo, Italy.
| | - Elena Rossi
- Istituto di Ematologia, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Image, radiation therapy, oncology and hematology Diagnosis, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Mario Tiribelli
- Division of Hematology and Bone Marrow Transplantation, Department of Medical Area, University of Udine, Udine, Italy
| | - Francesco Mendicino
- Hematology Unit, Department of Hemato-oncology, Ospedale Annunziata, Cosenza, Italy
| | - Novella Pugliese
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| |
Collapse
|
20
|
Gangat N, Tefferi A. Myelofibrosis biology and contemporary management. Br J Haematol 2020; 191:152-170. [PMID: 32196650 DOI: 10.1111/bjh.16576] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 02/20/2020] [Accepted: 02/21/2020] [Indexed: 12/25/2022]
Abstract
Myelofibrosis is an enigmatic myeloproliferative neoplasm, despite noteworthy strides in understanding its genetic underpinnings. Driver mutations involving JAK2, CALR or MPL in 90% of patients mediate constitutive JAK-STAT signaling which, in concert with epigenetic alterations (ASXL1, DNMT3A, SRSF2, EZH2, IDH1/2 mutations), play a fundamental role in disease pathogenesis. Aberrant immature megakaryocytes are a quintessential feature, exhibiting reduced GATA1 protein expression and secreting a plethora of pro-inflammatory cytokines (IL-1 ß, TGF-ß), growth factors (b-FGF, PDGF, VEGF) in addition to extra cellular matrix components (fibronectin, laminin, collagens). The ensuing disrupted interactions amongst the megakaryocytes, osteoblasts, endothelium, stromal cells and myofibroblasts within the bone marrow culminate in the development of fibrosis and osteosclerosis. Presently, prognostic assessment tools for primary myelofibrosis (PMF) are centered on genetics, with incorporation of cytogenetic and molecular information into the mutation-enhanced (MIPSS 70-plus version 2.0) and genetically-inspired (GIPSS) prognostic scoring systems. Both models illustrate substantial clinical heterogeneity in PMF and serve as the crux for risk-adapted therapeutic decisions. A major challenge remains the dearth of disease-modifying drugs, whereas allogeneic transplant offers the chance of long-term remission for some patients. Our review serves to synopsise current appreciation of the pathogenesis of myelofibrosis together with emerging management strategies.
Collapse
|
21
|
Gupta V, Cerquozzi S, Foltz L, Hillis C, Devlin R, Elsawy M, Grewal K, Hamm C, McNamara C, Sirhan S, Leber B. Patterns of Ruxolitinib Therapy Failure and Its Management in Myelofibrosis: Perspectives of the Canadian Myeloproliferative Neoplasm Group. JCO Oncol Pract 2020; 16:351-359. [PMID: 32134707 PMCID: PMC7359776 DOI: 10.1200/jop.19.00506] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Ruxolitinib improves splenomegaly and other disease-related symptoms in patients with myelofibrosis, but over time, many patients lose this benefit. It is difficult to determine whether this is due to resistance or intolerance to the drug; thus, we have used the more inclusive term of ruxolitinib failure. The survival of patients with myelofibrosis after ruxolitinib failure is poor but varies significantly by the pattern of the failure, underlining the need for a clinically appropriate classification. In this review, we propose diagnostic guidance for early recognition of the pattern of ruxolitinib failure and we recommend treatment options. The most frequent patterns of ruxolitinib failure are loss or failure to obtain a significant reduction in splenomegaly or symptom response, and the development or persistence of clinically significant cytopenias. Ruxolitinib dose modification and other ancillary therapies are sometimes helpful, and splenectomy is a palliative option in selected cases. Stem-cell transplantation is the only curative option for these patterns of failure, but its restricted applicability due to toxicity highlights the importance of ongoing clinical trials in this area. Recent approval of fedratinib by the US Food and Drug Administration provides an alternative option for patients with suboptimal or loss of spleen response. The transformation of myelofibrosis to accelerated or blast phase is an infrequent form of failure with an extremely poor prognosis, whereby patients who are ineligible for transplantation have limited treatment options.
Collapse
Affiliation(s)
- Vikas Gupta
- Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Sonia Cerquozzi
- Tom Baker Cancer Centre, University of Calgary, Calgary, Alberta, Canada
| | - Lynda Foltz
- St Paul's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Christopher Hillis
- Juravinski Cancer Centre, McMaster University, Hamilton, Ontario, Canada
| | - Rebecca Devlin
- Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | | | - Kuljit Grewal
- Memorial University of Newfoundland, St John's, Newfoundland and Labrador, Canada
| | - Caroline Hamm
- Windsor Regional Cancer Program, Windsor, Ontario, Canada
| | - Caroline McNamara
- Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | | | - Brian Leber
- Juravinski Cancer Centre, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
22
|
Mannelli L, Guglielmelli P, Vannucchi AM. Stem cell transplant for the treatment of myelofibrosis. Expert Rev Hematol 2020; 13:363-374. [DOI: 10.1080/17474086.2020.1733406] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Lara 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
- Department of Medical Biotechnologies, University of Siena, Siena, 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
| | - 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
| |
Collapse
|
23
|
Coltro G, Mannelli F, Vergoni F, Santi R, Massi D, Siliani LM, Marzullo A, Bonifacio S, Pelo E, Pacilli A, Paoli C, Franci A, Calabresi L, Bosi A, Vannucchi AM, Guglielmelli P. Extramedullary blastic transformation of primary myelofibrosis in the form of disseminated myeloid sarcoma: a case report and review of the literature. Clin Exp Med 2020; 20:313-320. [PMID: 32065308 DOI: 10.1007/s10238-020-00616-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 02/10/2020] [Indexed: 12/11/2022]
Abstract
Splenomegaly is a key clinical manifestation of myelofibrosis, and splenectomy is currently indicated in patients with drug refractory, symptomatic splenomegaly or with the aim of improving refractory cytopenias. Transformation to acute myeloid leukemia occurs in up to 20% of patients with myelofibrosis, while cases of myeloid sarcoma have been reported very unfrequently. In this manuscript, we report the case of a 60-year-old man with a history of primary myelofibrosis who underwent splenectomy because of drug-refractory massive splenomegaly, systemic symptoms and anemia. At the opening of the peritoneal cavity, the spleen resulted massively enlarged and tenaciously entrapped by a pervasive neoplastic-like tissue. The extensive involvement of the abdomen fatally complicated the surgical procedure. At postmortem examination, the spleen showed a diffuse infiltration of immature cells that were also found in the peritoneum, bowel, liver, lungs and myocardium. After immunohistochemical, cytogenetic, flow cytometric and molecular characterization of neoplastic population, a diagnosis of disseminated myeloid sarcoma of the spleen was made. This case report highlights a very unusual case of myeloid sarcoma originating from the spleen in a patient with myelofibrosis who had no evidence of bone marrow or peripheral blood involvement by leukemic cells. Molecular characterization showed that leukemic cells originated from the founding clone of the chronic phase. The sarcoma could not be suspected based on clinical findings and was diagnosed only at the time of surgical procedure and autopsy. This case suggests that leukemic transformation of myelofibrosis can originate outside the bone marrow and, presumably rarely, present as a granulocytic sarcoma.
Collapse
Affiliation(s)
- Giacomo Coltro
- CRIMM, Center of Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliero-Universitaria Careggi, 50134, Florence, Italy.,Department of Experimental and Clinical Medicine, DenoThe Excellence Center, University of Florence, Florence, Italy.,Hematology Unit, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Francesco Mannelli
- CRIMM, Center of Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliero-Universitaria Careggi, 50134, Florence, Italy
| | - Federica Vergoni
- Istologia Patologica e Diagnostica Molecolare, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Raffaella Santi
- Istologia Patologica e Diagnostica Molecolare, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Daniela Massi
- Istologia Patologica e Diagnostica Molecolare, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy.,Department of Surgery and Translational Medicine, Division of Pathology, University of Florence, Florence, Italy
| | - Luisa Margherita Siliani
- Division of Oncological and Robotic Surgery, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Antonella Marzullo
- Division of Genetic Diagnosis, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Stefania Bonifacio
- Division of Genetic Diagnosis, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Elisabetta Pelo
- Division of Genetic Diagnosis, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Annalisa Pacilli
- CRIMM, Center of Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliero-Universitaria Careggi, 50134, Florence, Italy.,Department of Experimental and Clinical Medicine, DenoThe Excellence Center, University of Florence, Florence, Italy
| | - Chiara Paoli
- CRIMM, Center of Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliero-Universitaria Careggi, 50134, Florence, Italy.,Department of Experimental and Clinical Medicine, DenoThe Excellence Center, University of Florence, Florence, Italy
| | - Annalisa Franci
- CRIMM, Center of Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliero-Universitaria Careggi, 50134, Florence, Italy.,Department of Experimental and Clinical Medicine, DenoThe Excellence Center, University of Florence, Florence, Italy
| | - Laura Calabresi
- CRIMM, Center of Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliero-Universitaria Careggi, 50134, Florence, Italy.,Department of Experimental and Clinical Medicine, DenoThe Excellence Center, University of Florence, Florence, Italy
| | - Alberto Bosi
- Hematology Unit, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Alessandro Maria Vannucchi
- CRIMM, Center of Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliero-Universitaria Careggi, 50134, Florence, Italy.,Department of Experimental and Clinical Medicine, DenoThe Excellence Center, University of Florence, Florence, Italy
| | - Paola Guglielmelli
- CRIMM, Center of Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliero-Universitaria Careggi, 50134, Florence, Italy. .,Department of Experimental and Clinical Medicine, DenoThe Excellence Center, University of Florence, Florence, Italy.
| |
Collapse
|
24
|
The role of allogeneic stem-cell transplant in myelofibrosis in the era of JAK inhibitors: a case-based review. Bone Marrow Transplant 2019; 55:708-716. [PMID: 31534197 PMCID: PMC7113188 DOI: 10.1038/s41409-019-0683-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 07/29/2019] [Accepted: 08/02/2019] [Indexed: 01/11/2023]
Abstract
Allogeneic hematopoietic stem-cell transplantation (HSCT) is, at present, the only potentially curative therapy for myelofibrosis (MF). Despite many improvements, outcomes of HSCT are still burdened by substantial morbidity and high transplant-related mortality. Allogeneic transplant is generally considered in intermediate-2 and high-risk patients aged <70 years, but the optimal selection of patients and timing of the procedure remains under debate, as does as the role of JAK inhibitors in candidates for HSCT. Starting from a real-life clinical case scenario, herein we examine some of the crucial issues of HSCT for MF in light of recent refinements on MF risk stratification, data on the use of ruxolitinib before and after transplant and findings on the impact of different conditioning regimens and donor selection.
Collapse
|
25
|
Barabanshikova MV, Zubarovsky IN, Savrasov VM, Korolkov AJ, Baykov VV, Botina AV, Vlasova JJ, Moiseev IS, Darskaya EI, Morozova EV, Afanasyev BV. Splenectomy following JAK1/JAK2 inhibitor therapy in patients with myelofibrosis undergoing allogeneic stem cell transplantation. Hematol Oncol Stem Cell Ther 2019; 12:140-145. [DOI: 10.1016/j.hemonc.2019.03.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 02/10/2019] [Accepted: 03/07/2019] [Indexed: 01/21/2023] Open
|
26
|
Leukemic Transformation of Myeloproliferative Neoplasms: Therapeutic and Genomic Considerations. Curr Hematol Malig Rep 2019; 13:588-595. [PMID: 30353413 DOI: 10.1007/s11899-018-0491-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE OF REVIEW Although BCR-ABL1-negative myeloproliferative neoplasms (MPN) are chronic, clonal hematopoietic stem cell (HSC) disorders marked by proliferation of one or more myeloid lineages, a substantial proportion of patients transform to acute myeloid leukemia. Leukemic transformation (LT) from a pre-existing MPN carries a dismal prognosis. Here, we review recent genetic, biological, and clinical data regarding LT. RECENT FINDINGS In the last decade, DNA sequencing has revolutionized our understanding of the genomic landscape of LT. Mutations in TP53, ASXL1, EZH2, IDH1/2, and SRSF2 are significantly associated with increased risk of LT of MPNs. Preclinical modeling of these mutations is underway and has yielded important biological insights, some of which have therapeutic implications. Recent progress has led to the identification of recurrent genomic alterations in patients with LT. This has allowed mechanistic and therapeutic insight into the process of LT. In turn, this may lead to more mechanism-based therapeutic strategies that may improve patient outcomes.
Collapse
|
27
|
Tavares RS, Nonino A, Pagnano KBB, Nascimento ACKVD, Conchon M, Fogliatto LM, Funke VAM, Bendit I, Clementino NCD, Chauffaille MDLLF, Bernardo WM, Santos FPDS. Guideline on myeloproliferative neoplasms: Associacão Brasileira de Hematologia, Hemoterapia e Terapia Cellular: Project guidelines: Associação Médica Brasileira - 2019. Hematol Transfus Cell Ther 2019; 41 Suppl 1:1-73. [PMID: 31248788 PMCID: PMC6630088 DOI: 10.1016/j.htct.2019.03.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 03/20/2019] [Indexed: 12/22/2022] Open
Affiliation(s)
| | - Alexandre Nonino
- Instituto Hospital de Base do Distrito Federal (IHBDF), Brasília, DF, Brazil
| | | | | | | | | | | | - Israel Bendit
- Hospital Das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil
| | | | | | - Wanderley Marques Bernardo
- Hospital Das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil; Associação Médica Brasileira (AMB), São Paulo, SP, Brazil
| | | |
Collapse
|
28
|
Mondello P, Di Mirto C, Cuzzocrea S, Arrigo C, Mian M, Pitini V. Interferon Alpha Has a Strong Anti-tumor Effect in Philadelphia-negative Myeloproliferative Neoplasms. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2019; 19:e489-e495. [PMID: 31231012 DOI: 10.1016/j.clml.2019.03.027] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 03/12/2019] [Accepted: 03/25/2019] [Indexed: 01/29/2023]
Abstract
BACKGROUND Despite the important progress in the research of myeloproliferative neoplasms (MPN), treatment options are still limited. Currently, a cytoreductive approach is the backbone treatment, with hydroxyurea (HU) being the most important agent. However, this drug is not always well-tolerated and has been questionably linked to a potential leukemogenic effect. A valid alternative is interferon alfa (IFN-α), but it is reserved for selected patients owing to the more frequent side effects and the lack of final results from the studies directly comparing IFN-α with HU, which is why we provided the results of the so far largest real-life analysis. PATIENTS AND METHODS From 2000 to 2016, 63 patients with Philadelphia-negative MPN prospectively received either HU or IFN-α. RESULTS During a median follow-up period of 121 months (range, 88-168 months), 97% of the patients treated with IFN-α achieved a hematologic response (60% complete, 37% partial) compared with 78% in the HU group (56% complete, 20% partial; P < .01). Molecular responses were limited to patients treated with IFN-α. IFN-α was well-tolerated with no secondary malignancy, whereas HU was associated with more toxic events and cases of leukemic transformation. A significantly longer progression-free survival (5.0 vs. 3.1 years; P < .001) and overall survival (7.8 vs. 5.8 years; P = .006) were observed in the IFN-α group compared with the HU cohort. CONCLUSION Our data support IFN-α as a more valid therapeutic option owing to its more profound hematologic responses, durable molecular remissions, long-term disease control, and reduced risk of leukemic transformation with a favorable toxicity profile.
Collapse
Affiliation(s)
- Patrizia Mondello
- Department of Medicine, Division of Hematology and Medical Oncology, Weill Cornell Medical College, New York, NY; Department of Human Pathology, University of Messina, Messina, Italy; Department of Biological and Environmental Sciences, University of Messina, Messina, Italy.
| | - Cristian Di Mirto
- Department of Human Pathology, University of Messina, Messina, Italy
| | - Salvatore Cuzzocrea
- Department of Biological and Environmental Sciences, University of Messina, Messina, Italy
| | - Carmela Arrigo
- Department of Human Pathology, University of Messina, Messina, Italy
| | - Michael Mian
- Internal Medicine V, Hematology and Oncology, Medical University of Innsbruck, Innsbruck, Austria; Department of Hematology and CBMT, Ospedale di Bolzano, Bolzano, Italy
| | - Vincenzo Pitini
- Department of Human Pathology, University of Messina, Messina, Italy
| |
Collapse
|
29
|
Tefferi A. Primary myelofibrosis: 2019 update on diagnosis, risk-stratification and management. Am J Hematol 2018; 93:1551-1560. [PMID: 30039550 DOI: 10.1002/ajh.25230] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 07/18/2018] [Indexed: 12/12/2022]
Abstract
DISEASE OVERVIEW Primary myelofibrosis (PMF) is a myeloproliferative neoplasm (MPN) characterized by stem cell-derived clonal myeloproliferation that is often but not always accompanied by JAK2, CALR, or MPL mutations; additional disease features include bone marrow stromal reaction including reticulin fibrosis, abnormal cytokine expression, anemia, hepatosplenomegaly, extramedullary hematopoiesis (EMH), constitutional symptoms, cachexia, leukemic progression, and shortened survival. DIAGNOSIS Diagnosis of PMF is based on bone marrow morphology. Presence of JAK2, CALR, or MPL mutation, expected in ∼ 90% of the patients, is supportive but not essential for diagnosis. The revised 2016 World Health Organization (WHO) classification system distinguishes "prefibrotic" from "overtly fibrotic" PMF; the former might mimic ET in its presentation and it is prognostically relevant to distinguish the two. RISK STRATIFICATION Two new prognostic systems for PMF have recently been introduced: GIPSS (genetically inspired prognostic scoring system) and MIPSS70+ version 2.0 (mutation- and karyotype-enhanced international prognostic scoring system). GIPSS is based exclusively on mutations and karyotype. MIPSS70+ version 2.0 utilizes both genetic and clinical risk factors. GIPSS features four and MIPSS70+ version 2.0 five risk categories. MIPSS70+ version 2.0 requires an online score calculator (http://www.mipss70score.it) while GIPPS offers a lower complexity prognostic tool. RISK-ADAPTED THERAPY Observation alone is advised for MIPSS70+ version 2.0 "low" and "very low" risk disease (estimated 10-year survival 56%-92%); allogeneic stem cell transplant is the preferred treatment of choice for "very high" and "high" risk disease (estimated 10-year survival 0-13%); treatment-requiring patients with intermediate-risk disease (estimated 10-year survival 30%) are best served by participating in clinical trials. All other treatment approaches, including the use of JAK2 inhibitors, are mostly palliative and should not be used in the absence of clear treatment indications. Conventional treatment for anemia includes androgens, prednisone, thalidomide and danazol, for symptomatic splenomegaly hydroxyurea and ruxolitinib and for constitutional symptoms ruxolitinib. Splenectomy is considered for drug-refractory splenomegaly and involved field radiotherapy for nonhepatosplenic EMH and extremity bone pain.
Collapse
Affiliation(s)
- Ayalew Tefferi
- Division of Hematology, Department of Medicine; Mayo Clinic; Rochester Minnesota
| |
Collapse
|
30
|
Pophali P, Horna P, Lasho TL, Finke CM, Ketterling RP, Gangat N, Nagorney D, Tefferi A, Patnaik MM. Splenectomy in patients with chronic myelomonocytic leukemia: Indications, histopathological findings and clinical outcomes in a single institutional series of thirty-nine patients. Am J Hematol 2018; 93:1347-1357. [PMID: 30105755 PMCID: PMC6196105 DOI: 10.1002/ajh.25246] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 08/03/2018] [Accepted: 08/06/2018] [Indexed: 12/13/2022]
Abstract
In a 28-year period, 39 (7%) patients with chronic myelomonocytic leukemia (CMML) (median age 66 years, 64% male) underwent a splenectomy at our institution. Primary indications for splenectomy were refractory thrombocytopenia (36%), progressive spleen related symptoms (33%), emergent splenectomy for splenic rupture (21%), refractory anemia (8%), and prior to allogeneic stem cell transplant (3%). Eleven (28%) patients had anemia at the time of splenectomy, of which 3 (27%) were autoimmune. The median time to splenectomy from CMML diagnosis was 6 months (0-40); perioperative morbidity and mortality rates were 43% and 13%, while the median postsplenectomy survival was 25 months (11-38). Durable remission in spleen related symptoms, thrombocytopenia, complications from splenic rupture, and anemia were achieved in 85%, 50%, 62%, and 21% of patients, respectively. Perioperative morbidity (n = 30) included infections/sepsis in 6 (20%), intraabdominal bleeding in 4 (13%), venous thromboembolism (VTE) in 3 (10%), and acute lung injury in 2 (7%) patients. The median duration of hospital stay was 6 days (1-25), with 5 deaths occurring secondary to respiratory failure (n = 2), multiorgan dysfunction (n = 2) and hemorrhagic shock (n = 1). There was no difference in overall survival between CMML patients that underwent splenectomy, in comparison to those that did not. Unlike in myelofibrosis, portal hypertension was not an indication for splenectomy and no patients developed post-splenectomy thrombocytosis. In conclusion, apart from being a lifesaving emergent modality in the event of splenic rupture, splenectomy has an important palliative role in patients with CMML, with significant and durable improvements in spleen related symptoms and refractory cytopenias.
Collapse
Affiliation(s)
- Prateek Pophali
- Division of Hematology, Department of Internal Medicine,
Mayo Clinic, Rochester, MN
| | - Pedro Horna
- Division of Hematopathology, Department of Laboratory
Medicine, Mayo Clinic, Rochester, MN
| | - Terra L. Lasho
- Division of Hematology, Department of Internal Medicine,
Mayo Clinic, Rochester, MN
| | - Christy M. Finke
- Division of Hematology, Department of Internal Medicine,
Mayo Clinic, Rochester, MN
| | - Rhett P. Ketterling
- Division of Hematopathology, Department of Laboratory
Medicine, Mayo Clinic, Rochester, MN
| | - Naseema Gangat
- Division of Hematology, Department of Internal Medicine,
Mayo Clinic, Rochester, MN
| | - David Nagorney
- Department of General Surgery, Mayo Clinic, Rochester,
MN
| | - Ayalew Tefferi
- Division of Hematology, Department of Internal Medicine,
Mayo Clinic, Rochester, MN
| | - Mrinal M. Patnaik
- Division of Hematology, Department of Internal Medicine,
Mayo Clinic, Rochester, MN
| |
Collapse
|
31
|
Mudireddy M, Gangat N, Lasho TL, Finke C, Hanson CA, Ketterling RP, Ashrani AA, Pardanani A, Nagorney DM, Tefferi A. Early thrombotic events and preemptive systemic anticoagulation following splenectomy for myelofibrosis. Am J Hematol 2018; 93:E235-E238. [PMID: 29981287 DOI: 10.1002/ajh.25203] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 06/18/2018] [Indexed: 11/10/2022]
Affiliation(s)
- Mythri Mudireddy
- Division of Hematology, Department of Internal Medicine; Mayo Clinic; Rochester Minnesota
| | - Naseema Gangat
- Division of Hematology, Department of Internal Medicine; Mayo Clinic; Rochester Minnesota
| | - Terra L. Lasho
- Division of Hematology, Department of Internal Medicine; Mayo Clinic; Rochester Minnesota
| | - Christy Finke
- Division of Hematology, Department of Internal Medicine; Mayo Clinic; Rochester Minnesota
| | - Curtis A. Hanson
- Department of Laboratory Medicine; Mayo Clinic; Rochester Minnesota
| | | | - Aneel A. Ashrani
- Division of Hematology, Department of Internal Medicine; Mayo Clinic; Rochester Minnesota
| | - Animesh Pardanani
- Division of Hematology, Department of Internal Medicine; Mayo Clinic; Rochester Minnesota
| | | | - Ayalew Tefferi
- Division of Hematology, Department of Internal Medicine; Mayo Clinic; Rochester Minnesota
| |
Collapse
|
32
|
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: 5.5] [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
|
33
|
Transplant Decisions in Patients with Myelofibrosis: Should Mutations Be the Judge? Biol Blood Marrow Transplant 2018; 24:649-658. [DOI: 10.1016/j.bbmt.2017.10.037] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 10/26/2017] [Indexed: 02/06/2023]
|
34
|
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: 36] [Impact Index Per Article: 5.1] [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
|
35
|
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
|
36
|
Tefferi A, Mudireddy M, Gangat N, Hanson CA, Ketterling RP, Pardanani A, Nagorney DM. Risk factors and a prognostic model for postsplenectomy survival in myelofibrosis. Am J Hematol 2017; 92:1187-1192. [PMID: 28782256 DOI: 10.1002/ajh.24881] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 08/02/2017] [Indexed: 01/04/2023]
Abstract
Palliative treatment in myelofibrosis (MF) includes transfusion support, JAK2 inhibitors, involved field radiotherapy and splenectomy. To assist in selecting patients who are likely to benefit from splenectomy, we looked into risk factors for postsplenectomy survival, in 120 consecutive cases (median age 66 years); at the time of splenectomy, 61% displayed red cell transfusion need, 49% platelet count <100 × 10(9)/L, 25% leukocyte count >25 × 10(9)/L, 60% constitutional symptoms and 13% circulating blasts ≥5%; dynamic international prognostic scoring system risk categories were 21% high, 55% intermediate-2, 21% intermediate-1 and 3% low. Among informative cases, karyotype was abnormal in 60% and driver mutational status was JAK2 75%, CALR 15%, MPL 4% and triple-negative 6%. At median follow-up of 1.3 years, from time of splenectomy, 95 (79%) deaths and 30 (25%) leukemic transformations were recorded. Median postsplenectomy survival was 1.5 years; in multivariable analysis, survival was adversely affected by age >65 years, transfusion need, leukocyte count >25 × 10(9)/L and circulating blasts ≥5%; these variables were subsequently used to devise an HR-weighted scoring system with high (3-4 risk factors), intermediate (2 risk factors) and low (0-1 risk factors) risk categories; the corresponding postsplenectomy median survivals were 0.3 (HR 5.9, 95% CI 3.2-11.0), 1.3 (HR 2.9, 95% CI 1.8-4.6) and 2.9 years. Postsplenectomy survival was not affected by driver mutational status or occurrence of leukemic transformation. Leukemia-free survival was predicted by very high risk karyotype. The observations from the current study might help identify appropriate candidates for splenectomy in MF.
Collapse
|
37
|
de la Pinta C, Fernández Lizarbe E, Montero Luis Á, Domínguez Rullán JA, Sancho García S. Treatment of symptomatic splenomegaly with low doses of radiotherapy: Retrospective analysis and review of the literature. Tech Innov Patient Support Radiat Oncol 2017; 3-4:23-29. [PMID: 32095563 PMCID: PMC7033798 DOI: 10.1016/j.tipsro.2017.08.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 07/09/2017] [Accepted: 08/09/2017] [Indexed: 01/28/2023] Open
Abstract
OBJECTIVES To evaluate the effectiveness of low doses of radiation therapy for symptomatic splenomegaly in malignant and benign diseases. PATIENTS AND METHODS 5 patients with symptomatic splenomegaly were treated with low doses of radiation in our centre (January 2008-December 2016). 4/5 patients had malignant neoplasia (acute myeloid leukemia, non Hogdkin lymphoma and prolymphocytic B cell leukemia) and splenomegaly was caused by extramedullary hematopoiesis. 1/5 patient had benign disease (HBV liver cirrhosis) and splenomegaly was caused by vascular ectasia. Median age was 73 years (range 61-86 years). There were 4 females and 1 male. These patients had exclusively splenic pain or abdominal discomfort in 20%, exclusively cytopenias 40% and both 40%. Patients needed radiation therapy for symptomatic control. Dose per fraction was 0.5 Gy every two days; total dose initially prescribed 10 Gy. IGRT were performed in all patients to ensure an appropriate position and to adapt the treatment volume to the changes in the spleen volume along the treatment. Median craneocaudal length size of the spleen was more than 26 cm (range 15.2-34.9 cm). RESULTS Median radiation doses were 4.85 Gy (range 2.5-10). Median craneocaudal spleen size reduction was 4.6 cm (0-8 cm). Splenic pain and abdominal disturbances improved in all patients. Median increase of haemoglobin and platelets levels was 1.6 mg/dl and 27.950 cells respectively in the first week after the end of radiotherapy.One patient had to interrupt her treatment due to grade II neutropenia. No other toxicities were described. With a median follow-up of 39 months (16-89 months), only one recurrence was described at 24 months and consisted of thrombocytopenia. The patient received a second course of radiotherapy with excellent response. CONCLUSION Low doses of radiation therapy for treatment of symptomatic splenomegaly were effective, with a low rate of side effects. Splenic pain and abdominal discomfort completely improved and cytopenias rised to secure levels.
Collapse
|
38
|
Devlin R, Gupta V. Myelofibrosis: to transplant or not to transplant? HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2016; 2016:543-551. [PMID: 27913527 PMCID: PMC6142493 DOI: 10.1182/asheducation-2016.1.543] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Hematopoietic cell transplantation (HCT) is the only curative therapeutic modality for myelofibrosis (MF) at present. The optimal timing of HCT is not known in the presence of wider availability of less risky nontransplant therapies such as JAK 1/2 inhibitors. Careful review of patient, disease, and transplant-related factors is required in the appropriate selection of HCT vs the best available nontransplant therapies. We highlight some of the relevant issues and positioning of HCT in light of evolving data on JAK 1/2 inhibitors. The goal of this study is to provide the reader with updated evidence of HCT for MF, recognizing that knowledge in this area is limited by the absence of comparative studies between HCT and nontransplant therapies. Prospective studies are needed for better information on: the determination of optimal timing and conditioning regimens, the best way to integrate JAK inhibitors in the HCT protocols, and the impact of JAK inhibitors on graft-versus-host disease.
Collapse
Affiliation(s)
- Rebecca Devlin
- The Elizabeth and Tony Comper Myeloproliferative Neoplasm Program, Princess Margaret Cancer Center, Toronto, Ontario, Canada
| | - Vikas Gupta
- The Elizabeth and Tony Comper Myeloproliferative Neoplasm Program, Princess Margaret Cancer Center, Toronto, Ontario, Canada
| |
Collapse
|
39
|
Tefferi A. Primary myelofibrosis: 2017 update on diagnosis, risk-stratification, and management. Am J Hematol 2016; 91:1262-1271. [PMID: 27870387 DOI: 10.1002/ajh.24592] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 10/20/2016] [Indexed: 12/13/2022]
Abstract
Disease overview: Primary myelofibrosis (PMF) is a myeloproliferative neoplasm (MPN) characterized by stem cell-derived clonal myeloproliferation that is often but not always accompanied by JAK2, CALR or MPL mutation, abnormal cytokine expression, bone marrow fibrosis, anemia, splenomegaly, extramedullary hematopoiesis (EMH), constitutional symptoms, cachexia, leukemic progression and shortened survival. DIAGNOSIS Diagnosis is based on bone marrow morphology. The presence of JAK2, CALR or MPL mutation is supportive but not essential for diagnosis; approximately 90% of patients carry one of these mutations and 10% are "triple-negative." None of these mutations are specific to PMF and are also seen in essential thrombocythemia (ET). According to the revised 2016 World Health Organization (WHO) classification and diagnostic criteria, "prefibrotic" PMF (pre-PMF) is distinguished from "overtly fibrotic" PMF; the former might mimic ET in its presentation and it is prognostically relevant to distinguish the two. Risk stratification: The Dynamic International Prognostic Scoring System-plus (DIPSS-plus) uses eight predictors of inferior survival: age >65 years, hemoglobin <10 g/dL, leukocytes >25 × 109 /L, circulating blasts ≥1%, constitutional symptoms, red cell transfusion dependency, platelet count <100 × 109 /L and unfavorable karyotype (i.e., complex karyotype or sole or two abnormalities that include +8, -7/7q-, i(17q), inv(3), 5/5q-, 12p-, or 11q23 rearrangement). The presence of 0, 1, "2 or 3" and ≥4 adverse factors defines low, intermediate-1, intermediate-2 and high-risk disease with median survivals of approximately 15.4, 6.5, 2.9 and 1.3 years, respectively. Most recently, DIPSS-plus-independent adverse prognostic relevance has been demonstrated for certain mutations including ASXL1 and SRSF2 whereas patients with type 1/like CALR mutations, compared to their counterparts with other driver mutations, displayed significantly better survival. Risk-adapted therapy: Observation alone is a reasonable treatment strategy for asymptomatic low or intermediate-1 DIPSS-plus risk disease, especially in the absence of high-risk mutations. All other patients with high or intermediate-2 risk disease, or those harboring high-risk mutations such as ASXL1 or SRSF2, should be considered for stem cell transplant, which is currently the only treatment modality with the potential to favorably modify the natural history of the disease. Non-transplant candidates should be encouraged to participate in clinical trials, since the value of conventional drug therapy, including the use of JAK2 inhibitors, is limited to symptoms palliation and reduction in spleen size. Specifically, JAK2 inhibitors have not been shown to induce complete clinical or cytogenetic remissions or significantly affect JAK2/CALR/MPL mutant allele burden. Splenectomy is considered for drug-refractory splenomegaly. Involved field radiotherapy is most useful for post-splenectomy hepatomegaly, non-hepatosplenic EMH, PMF-associated pulmonary hypertension and extremity bone pain. Am. J. Hematol. 91:1262-1271, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Ayalew Tefferi
- Division of Hematology, Department of MedicineMayo ClinicRochester Minnesota
| |
Collapse
|
40
|
Abstract
Myelofibrosis (MF) is a myeloproliferative neoplasm that presents either as a primary disease or evolves secondarily from polycythemia vera or essential thrombocythemia to post-polycythemia vera MF or post-essential thrombocythemia MF, respectively. Myelofibrosis is characterized by stem cell-derived clonal myeloproliferation, abnormal cytokine expression, bone marrow fibrosis, anemia, splenomegaly, extramedullary hematopoiesis, constitutional symptoms, cachexia, leukemic progression, and shortened survival. Therapeutic options for patients with MF have been limited to the use of cytoreductive agents, predominantly hydroxyurea; splenectomy and splenic irradiation for treatment of splenomegaly; and management of anemia with transfusions, erythropoiesis-stimulating agents, androgens, and immunomodulatory agents along with steroids. The only curative option is allogeneic stem cell transplantation (ASCT), which is associated with high morbidity and mortality risks. Recently, JAK (Janus kinase) inhibitor therapies have become available and proven to be palliative in primary MF patients with hydroxyurea-refractory splenomegaly and severe constitutional symptoms. The purpose of this article is to review the clinical features of MF; discuss different treatment strategies, including ASCT; and discuss the potential danger and benefit of using JAK inhibitors prior to ASCT.
Collapse
|
41
|
Aruch D, Schwartz M, Mascarenhas J, Kremyanskaya M, Newsom C, Hoffman R. Continued Role of Splenectomy in the Management of Patients With Myelofibrosis. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2016; 16:e133-e137. [PMID: 27373368 DOI: 10.1016/j.clml.2016.06.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 05/13/2016] [Accepted: 06/01/2016] [Indexed: 11/30/2022]
Affiliation(s)
- Daniel Aruch
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Myron Schwartz
- Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY
| | - John Mascarenhas
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Marina Kremyanskaya
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Carrie Newsom
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Ronald Hoffman
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY.
| |
Collapse
|
42
|
Farhadfar N, Cerquozzi S, Patnaik M, Tefferi A. Allogeneic Hematopoietic Stem-Cell Transplantation for Myelofibrosis: A Practical Review. J Oncol Pract 2016; 12:611-21. [DOI: 10.1200/jop.2016.013268] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Myelofibrosis is a myeloproliferative neoplasm with cardinal features of extramedullary hematopoiesis, hepatosplenomegaly, cytopenias, and constitutional symptoms that result in shortened survival and leukemic transformation. It is a disease predominantly of the elderly, and currently available therapies only offer symptom control without curative benefit or ability to alter disease progression. Allogeneic hematopoietic stem-cell transplant (HSCT) is the only potentially curative intervention; however, this is only feasible in younger and medically fit patients and selectively offered to those with high-risk disease. Despite ongoing advancements, HSCT is associated with substantial morbidity and mortality, and the determination of which patients with myelofibrosis are ideal candidates and the selection of the opportune moment to proceed with transplantation remains challenging. This review summarizes our current recommendations for the role of and indications for HSCT in myelofibrosis.
Collapse
|
43
|
Song MK, Chung JS, Lim SN, Lee GW, Lee SM, Lee NK, Choi JC, Oh SY. Usefulness of spleen volume measured by computed tomography for predicting clinical outcome in primary myelofibrosis. Int J Hematol 2016; 104:476-84. [PMID: 27349913 DOI: 10.1007/s12185-016-2050-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 06/20/2016] [Accepted: 06/21/2016] [Indexed: 11/28/2022]
Abstract
Although splenomegaly is major characteristic of primary myelofibrosis (PMF), splenomegaly has been devalued due to a less reliable method based on physical examination (PEx). We evaluated whether spleen volume (SV) on CT would accurately predict clinical outcomes in PMF. A total of 188 patients were enrolled. SV was quantitated by the automatic volume software. In ROC curve, the SV predicted prognosis more accurately than spleen length by PEx (p < 0.001). The ideal cut-off value was 378.1 cm(3) for SV, which was divided into high- and low-volume status. Patients with low SV status had superior leukemia-free survival and overall survival compared to high SV status (p < 0.001, p < 0.001) In the Cox analysis, old age ≥65 years (p = 0.004, p = 0.001), low Hemoglobin <10.0 g/dL (p = 0.023, p = 0.021), high WBC counts ≥25 × 10(9)/L (p = 0.003, p = 0.006), peripheral blasts ≥1 % (p = 0.029, p = 0.020), unfavorable cytogenetic abnormalities (p = 0.025, p = 0.028), and high SV status (p = 0.004, p = 0.003) were independently associated with survivals. SV measured by CT was important for predicting survival in patients with PMF.
Collapse
Affiliation(s)
- Moo-Kon Song
- Department of Hematology-Oncology, Hanyang University Hanmaeum Changwon Hospital, WoniDae-Ro 682 road, 21 Changwon, 51497, Republic of Korea.
| | - Joo-Seop Chung
- Department of Hematology-Oncology, Pusan National University Hospital, Busan, Korea
| | - Sung-Nam Lim
- Department of Hematology, Haeundae Paik Hospital, Busan, Korea
| | - Gyeong-Won Lee
- Department of Hematology, Gyeong-Sang National University Hospital, School of Medicine, Gyeong-Sang National University, Jinju, Korea
| | - Sang-Min Lee
- Department of Hematology, Busan Paik Hospital, Busan, Korea
| | - Nam-Kyung Lee
- Department of Radiology, Pusan National University Hospital, Busan, Korea
| | - Jae-Cheol Choi
- Department of Laboratory Medicine, Hanyang University Hanmaeum Changwon Hospital, Changwon, Korea
| | - So-Yeon Oh
- Department of Hematology-Oncology, Pusan National University Yangsan Hospital, Yangsan, Korea
| |
Collapse
|
44
|
Al-Ali HK, Vannucchi AM. Managing patients with myelofibrosis and low platelet counts. Ann Hematol 2016; 96:537-548. [PMID: 27209535 DOI: 10.1007/s00277-016-2697-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 05/10/2016] [Indexed: 12/18/2022]
Abstract
Myelofibrosis (MF) is a chronic myeloproliferative neoplasm characterized by bone marrow fibrosis, ineffective hematopoiesis, splenomegaly, constitutional symptoms, and shortened survival. Patients often experience multiple disease-associated, as well as treatment-emergent, cytopenias, including thrombocytopenia. However, patients with MF with thrombocytopenia have few therapeutic options, and there is little information on the management of these patients. Several Janus kinase (JAK) inhibitors have been developed for the treatment of MF, with one (ruxolitinib) having been approved. However, given their mechanism of action, JAK inhibitors are associated with high rates of thrombocytopenia. Patients can be successfully managed with dose modifications, but little is known about the safety and efficacy of these agents in patients with thrombocytopenia. Recent studies of JAK inhibitors in patients with MF who have low platelet counts have had mixed results. This review discusses the prevalence, prognostic implications, and management of thrombocytopenia in MF and the different therapeutic options available for this patient population, with an emphasis on current clinical experience with targeted therapies, as well as recent findings from several clinical studies currently underway.
Collapse
Affiliation(s)
| | - Alessandro M Vannucchi
- CRIMM, Center for Research and Innovation of Myeloproliferative Neoplasms, AOU Careggi, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| |
Collapse
|
45
|
Primary myelofibrosis: current therapeutic options. Rev Bras Hematol Hemoter 2016; 38:257-63. [PMID: 27521865 PMCID: PMC4997889 DOI: 10.1016/j.bjhh.2016.04.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 04/05/2016] [Indexed: 12/24/2022] Open
Abstract
Primary myelofibrosis is a Philadelphia-negative myeloproliferative neoplasm characterized by clonal myeloid expansion, followed by progressive fibrous connective tissue deposition in the bone marrow, resulting in bone marrow failure. Clonal evolution can also occur, with an increased risk of transformation to acute myeloid leukemia. In addition, disabling constitutional symptoms secondary to the high circulating levels of proinflammatory cytokines and hepatosplenomegaly frequently impair quality of life. Herein the main current treatment options for primary myelofibrosis patients are discussed, contemplating disease-modifying therapeutics in addition to palliative measures, in an individualized patient-based approach.
Collapse
|
46
|
Cervantes F, Correa JG, Hernandez-Boluda JC. Alleviating anemia and thrombocytopenia in myelofibrosis patients. Expert Rev Hematol 2016; 9:489-96. [DOI: 10.1586/17474086.2016.1154452] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
47
|
Patel AB, Vellore NA, Deininger MW. New Strategies in Myeloproliferative Neoplasms: The Evolving Genetic and Therapeutic Landscape. Clin Cancer Res 2016; 22:1037-47. [PMID: 26933174 PMCID: PMC4826348 DOI: 10.1158/1078-0432.ccr-15-0905] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The classical BCR-ABL1-negative myeloproliferative neoplasms (MPN) include essential thrombocythemia (ET), polycythemia vera (PV), and myelofibrosis (MF). Although these clonal disorders share certain clinical and genetic features, MF in particular is distinct for its complex mutational landscape, severe disease phenotype, and poor prognosis. The genetic complexity inherent to MF has made this disease extremely challenging to treat. Pharmacologic JAK inhibition has proven to be a transformative therapy in MPNs, alleviating symptom burden and improving survival, but has been hampered by off-target toxicities and, as monotherapy, has shown limited effects on mutant allele burden. In this review, we discuss the genetic heterogeneity contributing to the pathogenesis of MPNs, focusing on novel driver and epigenetic mutations and how they relate to combination therapeutic strategies. We discuss results from ongoing studies of new JAK inhibitors and report on new drugs and drug combinations that have demonstrated success in early preclinical and clinical trials, including type II JAK inhibitors, antifibrotic agents, and telomerase inhibitors.
Collapse
Affiliation(s)
- Ami B. Patel
- University of Utah Huntsman Cancer Institute, 2000 Circle of Hope Drive, Salt Lake City, UT 84112-5550
| | - Nadeem A. Vellore
- University of Utah Huntsman Cancer Institute, 2000 Circle of Hope Drive, Salt Lake City, UT, 84112-5550
| | - Michael W. Deininger
- Chief of Hematology, University of Utah Huntsman Cancer Institute, 2000 Circle of Hope Drive, Salt Lake City, UT, 84112-5550
| |
Collapse
|
48
|
Beauverd Y, McLornan DP, Harrison CN. Pacritinib: a new agent for the management of myelofibrosis? Expert Opin Pharmacother 2016; 16:2381-90. [PMID: 26389774 DOI: 10.1517/14656566.2015.1088831] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Myelofibrosis (MF) is a clonal haematological disease associated with recurrent somatic gene mutations (JAK2V617F, MPL, CALR) and constitutive activation of the Janus kinase (JAK)/Signal Transducer and Activator of Transcription pathway. MF is often characterised by debilitating symptoms and JAK inhibitors (JAKIs) have revolutionised available therapeutic options. Ruxolitinib, a JAK1 and 2 inhibitor, is the only currently approved agent. Several other JAKIs are undergoing evaluation in the clinical trial setting and Pacritinib , a novel JAK2 and FLT3 inhibitor, is at an advanced stage of investigation with recent completion of a Phase III trial and another ongoing. AREAS COVERED Within this article we focus on pacritinib, summarising the development, preclinical and up-to-date results from the Phase I - III trials. We present the most recent data on efficacy and safety and indirectly compare this novel JAKI with ruxolitinib. EXPERT OPINION The kinome array data for pacritinib suggests that it has a range of targets differing to those for ruxolitinib. Pacritinib appears to be an effective agent for the control of MF-related symptoms and splenomegaly with potentially fewer haematological side-effects when compared with ruxolitinib and seems a particularly promising agent for anaemic and thrombocytopenic patients. It is also an attractive drug for potential combination studies due to its good tolerability.
Collapse
Affiliation(s)
- Yan Beauverd
- a 1 Guy's and St. Thomas' NHS Foundation Trust, Department of Haematology , London, UK
| | - Donal P McLornan
- a 1 Guy's and St. Thomas' NHS Foundation Trust, Department of Haematology , London, UK.,b 2 King's College Hospital NHS Foundation Trust, Department of Haematological Medicine , London, UK
| | - Claire N Harrison
- c 3 Guy's and St. Thomas' NHS Foundation Trust, Department of Haematology , London, UK
| |
Collapse
|
49
|
Real-World Assessment of Clinical Outcomes in Patients with Lower-Risk Myelofibrosis Receiving Treatment with Ruxolitinib. Adv Hematol 2015; 2015:848473. [PMID: 26635878 PMCID: PMC4655278 DOI: 10.1155/2015/848473] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 09/28/2015] [Accepted: 10/01/2015] [Indexed: 11/17/2022] Open
Abstract
Few trial-based assessments of ruxolitinib in patients with lower-risk myelofibrosis (MF) have been conducted, and no studies have made such assessments in a real-world population. We assessed changes in spleen size and constitutional symptoms during ruxolitinib treatment using a retrospective, observational review of anonymized US medical record data of patients diagnosed with IPSS low-risk (n = 25) or intermediate-1-risk (n = 83) MF. The majority of patients were male (low risk, 60%; intermediate-1 risk, 69%). Most patients (92% and 77%) were still receiving ruxolitinib at the medical record abstraction date (median observation/exposure time, 8 months). The proportion of patients with moderate or severe palpable splenomegaly (≥10 cm) decreased from diagnosis (56%) to best response (12%). Fatigue was reported in 47% of patients and was the most common constitutional symptom. For most symptoms in both risk groups, shifts in the distribution of severity from more to less severe from diagnosis to best response were observed. Both patients with low-risk and intermediate-1-risk MF experienced a substantial decrease in spleen size with ruxolitinib treatment in real-world settings. For most symptoms examined, there were distinct improvements in the distribution of severity during ruxolitinib treatment. These findings suggest that patients with lower-risk MF may benefit clinically from ruxolitinib treatment.
Collapse
|
50
|
Vannucchi AM, Barbui T, Cervantes F, Harrison C, Kiladjian JJ, Kröger N, Thiele J, Buske C. Philadelphia chromosome-negative chronic myeloproliferative neoplasms: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 2015; 26 Suppl 5:v85-99. [PMID: 26242182 DOI: 10.1093/annonc/mdv203] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
MESH Headings
- Humans
- Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/diagnosis
- Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/pathology
- Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/therapy
- Philadelphia Chromosome
Collapse
Affiliation(s)
- A M Vannucchi
- Department of Experimental and Clinical Medicine, University of Florence, Florence
| | - T Barbui
- Hematology and Foundation for Research, Ospedale Papa Giovanni XXIII, Bergamo, Italy
| | - F Cervantes
- Department of Hematology, Hospital Clínic, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - C Harrison
- Department of Haematology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - J-J Kiladjian
- Centre d'Investigations Cliniques (INSERM CIC1427), Hôpital Saint-Louis and Paris Diderot University, Paris, France
| | - N Kröger
- Department of Stem Cell Transplantation, University Hospital Hamburg, Hamburg
| | | | - C Buske
- Comprehensive Cancer Center Ulm, Institute of Experimental Cancer Research, University Hospital Ulm, Ulm, Germany
| |
Collapse
|