1
|
Al-Ali HK, Gerds AT, Grunwald MR, Yu J. A Review of Real-World Experience With Ruxolitinib for Myelofibrosis. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2024:S2152-2650(24)02465-0. [PMID: 39837682 DOI: 10.1016/j.clml.2024.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/20/2024] [Accepted: 12/23/2024] [Indexed: 01/23/2025]
Abstract
Myelofibrosis (MF) is a rare myeloproliferative neoplasm characterized by progressive bone marrow fibrosis and splenomegaly. Ruxolitinib is the standard-of-care first-line treatment option for MF. This review summarizes real-world effectiveness and safety of ruxolitinib in more than 4500 patients with MF from real-world settings, including expanded-access and phase 4 trials, as well as registry, postmarketing, and retrospective studies in the 10 years since regulatory approval. Consistent with results from the phase 3 COMFORT trials, real-world evidence supports the effectiveness of ruxolitinib in improving splenomegaly and MF symptoms while significantly increasing overall survival. Real-world safety data have also been aligned with results from the COMFORT trials. Transient anemia, thrombocytopenia, and infections are the most frequently observed adverse events (AEs) but rarely required ruxolitinib discontinuation. Other nonhematologic AEs are generally mild, and grade ≥ 3 events rarely occur. Importantly, real-world evidence also supports the effectiveness of ruxolitinib in patient groups that were poorly represented in clinical trials, including those with lower-risk MF, those presenting with thrombocytopenia or anemia, and those who have previously discontinued ruxolitinib treatment. Finally, cost-effectiveness analyses show ruxolitinib to be cost-effective in Europe and North America. Taken together, real-world studies reinforce the efficacy, safety, and cost-effectiveness of ruxolitinib for the treatment of patients with MF, supporting results from prospective clinical trials. Furthermore, they demonstrate the clinical benefit of ruxolitinib in patient subgroups poorly represented in clinical trials and the value of dose modifications or re-treatment after interruptions to optimize outcomes.
Collapse
Affiliation(s)
| | - Aaron T Gerds
- Cleveland Clinic Taussig Cancer Institute, Cleveland, OH
| | | | | |
Collapse
|
2
|
Petit C, de Lavallade H, Harrison C. What are the therapeutic options for previously treated myelofibrosis? Expert Rev Hematol 2024:1-12. [PMID: 39494849 DOI: 10.1080/17474086.2024.2423367] [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: 09/04/2024] [Accepted: 10/27/2024] [Indexed: 11/05/2024]
Abstract
INTRODUCTION The disruption of the JAK/STAT signaling pathway is a defining feature of myelofibrosis (MF). The introduction of JAK inhibitors (JAKi) has transformed the therapeutic approach to MF, becoming essential to treatment and reshaping the management landscape. While JAKi are now the preferred first-line treatment for most patients, various management options are available for those who do not respond to initial therapy. AREAS COVERED This review focuses on management options for patients with MF, with particular emphasis on therapeutic strategies following the failure of first-line JAKi. It provides a comprehensive overview of the current treatment landscape, including alternative JAKi and other approaches. The review is based on an extensive literature search using available databases (PubMed, Cochrane …) and relevant web resources (clinicaltrials.gov). EXPERT OPINION Ruxolitinib benefits in MF often diminish after 3-4 years, with complications like thrombocytopenia and anemia. Three newer JAKi offer alternatives with similar efficacy and varied side effects. Stem cell transplantation is a curative option for a minority, ideally timed at peak response to JAKi. Research aims to enhance first-line treatments and restore responses in resistant patients. Future therapies may include novel combinations or immunotherapies targeting specific mutations, requiring collaboration between patient, clinical, and pharmaceutical communities.
Collapse
Affiliation(s)
- Cassandre Petit
- Department of Haematology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Hugues de Lavallade
- Department of Haematology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Claire Harrison
- Department of Haematology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| |
Collapse
|
3
|
Pinczés LI, Jóna Á, Mezei G, Kenyeres A, Vekszler PP, Illés Á, Simon Z. Successful ruxolitinib rechallenge after fedratinib failure in a patient with overt myelofibrosis. Ann Hematol 2024; 103:4817-4819. [PMID: 39090340 DOI: 10.1007/s00277-024-05825-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 05/30/2024] [Indexed: 08/04/2024]
Affiliation(s)
- László Imre Pinczés
- Division of Hematology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
- Doctoral School of Clinical Medicine, University of Debrecen, Debrecen, Hungary.
| | - Ádám Jóna
- Division of Hematology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Doctoral School of Clinical Medicine, University of Debrecen, Debrecen, Hungary
| | - Gabriella Mezei
- Division of Hematology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Doctoral School of Clinical Medicine, University of Debrecen, Debrecen, Hungary
| | - Anna Kenyeres
- Division of Hematology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Doctoral School of Clinical Medicine, University of Debrecen, Debrecen, Hungary
| | - Péter Pambó Vekszler
- Division of Hematology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Doctoral School of Clinical Medicine, University of Debrecen, Debrecen, Hungary
| | - Árpád Illés
- Division of Hematology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Doctoral School of Clinical Medicine, University of Debrecen, Debrecen, Hungary
| | - Zsófia Simon
- Division of Hematology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Doctoral School of Clinical Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
4
|
Bose P. Management of Patients with Early Myelofibrosis: A Discussion of Best Practices. Curr Hematol Malig Rep 2024; 19:111-119. [PMID: 38441783 PMCID: PMC11127825 DOI: 10.1007/s11899-024-00729-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2024] [Indexed: 05/26/2024]
Abstract
PURPOSE OF REVIEW Summarize best practices for management of patients with early myelofibrosis (MF). RECENT FINDINGS Myelofibrosis is a progressive myeloproliferative neoplasm (MPN) that generally produces burdensome symptoms and ultimately leads to worse overall survival than that observed in healthy controls or patients with other MPNs. Several Janus kinase inhibitors and various interferon formulations are now available for treatment of MF, with ruxolitinib notable for extending overall survival in addition to improving MF signs and symptoms. The chronic nature of the disease can lead some patients to avoid immediate treatment in favor of a watch-and-wait approach. This review summarizes the patient management approach taken in my practice, providing guidance and a discussion of best practices with an emphasis on the importance and clinical benefits of active treatment in early MF. In particular, a case is made to consider treatment with ruxolitinib for patients with intermediate-1 risk disease and to minimize delay between diagnosis and treatment initiation for patients with intermediate or high-risk disease.
Collapse
Affiliation(s)
- Prithviraj Bose
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
5
|
Bose P, Kuykendall AT, Miller C, Kurtin S, Farina K, Harting DM, Mascarenhas JO, Mesa RA. Moving Beyond Ruxolitinib Failure in Myelofibrosis: Evolving Strategies for Second Line Therapy. Expert Opin Pharmacother 2023; 24:1091-1100. [PMID: 37163478 DOI: 10.1080/14656566.2023.2213435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Ruxolitinib has been the cornerstone of pharmacologic therapy for myelofibrosis for over a decade. However, the last several years have witnessed the regulatory approval of other Janus kinase (JAK) inhibitors for myelofibrosis, i.e. fedratinib, pacritinib, and US approval of momelotinib is widely anticipated in 2023. Due to the multifaceted clinical presentation of myelofibrosis, a watertight definition of ruxolitinib failure has remained elusive, as "progression" on ruxolitinib can take many forms and management is highly nuanced. Yet, the availability of other JAK inhibitors and potential future availability of non-JAK inhibitor agents for myelofibrosis make a consensus on management of ruxolitinib failure critically important. This consensus paper summarizes a discussion between multiple academic and community physician experts, a pharmacist and an advanced practice provider around the issues to be considered for the optimal care of patients with myelofibrosis whose disease is refractory to or does not respond adequately to ruxolitinib, or who exhibit intolerance to ruxolitinib. The panel identified several areas of consensus, as well as some areas where more data to inform evidence-based practice are needed. In some situations, maintaining ruxolitinib while adding another agent, e.g. to address anemia, is appropriate, whereas in others, switching to a different drug has merit.
Collapse
Affiliation(s)
- Prithviraj Bose
- University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | | | - Sandra Kurtin
- University of Arizona Cancer Center, Tucson, Arizona
| | - Kyle Farina
- The Mount Sinai Hospital, Department of Pharmacy, New York, New York
| | | | | | - Ruben A Mesa
- Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston Salem and Charlotte, North Carolina
| |
Collapse
|
6
|
Mascarenhas J, Nguyen H, Saunders A, Oliver L, Tomkinson H, Perry R, McBride A. Defining ruxolitinib failure and transition to next-line therapy for patients with myelofibrosis: a modified Delphi panel consensus study. Future Oncol 2023. [PMID: 37161798 DOI: 10.2217/fon-2022-1298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023] Open
Abstract
Aim: To define ruxolitinib failure and develop parameters to guide transition to next-line therapy for patients with myelofibrosis. Methods: A modified Delphi panel with 14 hematologists-oncologists. Survey concepts included defining primary refractory status, loss of response, disease progression, intolerance and transition to next-line therapy. Results: Ruxolitinib failure may be defined as no improvement in symptoms or spleen size, progressive disease or ruxolitinib intolerance, following a maximally tolerated dose for ≥3 months. Loss of spleen response 1 month after initial response may prompt discontinuation. Lack of evidence to inform transition to next-line therapy was noted; tapering ruxolitinib should be considered according to ruxolitinib dose and patient characteristics. Conclusion: Expert consensus was provided on defining ruxolitinib failure and transition to next-line therapy as summarized in this position paper, which may support considerations in the development of future clinical practice guidelines.
Collapse
Affiliation(s)
- John Mascarenhas
- Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Hiep Nguyen
- Bristol Myers Squibb, Princeton, NJ 08540, USA
| | | | | | | | | | - Ali McBride
- Bristol Myers Squibb, Princeton, NJ 08540, USA
| |
Collapse
|
7
|
Ayala R, Fernández RA, García‐Gutiérrez V, Alvarez‐Larrán A, Osorio S, Sánchez‐Pina JM, Carreño‐Tarragona G, Álvarez N, Gómez‐Casares MT, Duran A, Gorrochategi J, Hernández‐Boluda JC, Martínez‐López J. Janus kinase inhibitor ruxolitinib in combination with nilotinib and prednisone in patients with myelofibrosis (RuNiC study): A phase Ib, multicenter study. EJHAEM 2023; 4:401-409. [PMID: 37206258 PMCID: PMC10188506 DOI: 10.1002/jha2.685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/16/2023] [Accepted: 03/22/2023] [Indexed: 05/21/2023]
Abstract
This phase Ib, non-randomized, open-label study evaluates the safety and tolerability of ruxolitinib in combination with nilotinib and prednisone in patients with naïve or ruxolitinib-resistant myelofibrosis (MF). A total of 15 patients with primary or secondary MF received the study treatment; 13 patients had received prior ruxolitinib treatment (86.7%). Eight patients completed seven cycles (53.3%) and six patients completed twelve cycles of treatment (40%). All the patients experienced at least one adverse event (AE) during the study (the most common AEs were hyperglycemia, asthenia, and thrombocytopenia), and 14 patients registered at least one treatment-related AE (the most common treatment-related AEs were hyperglycemia (22.2%; three grade 3 cases). Five treatment-related serious AEs (SAEs) were reported in two patients (13.3%). No deaths were registered throughout the study. No dose-limiting toxicity was observed. Four out of fifteen (27%) patients experienced a 100% spleen size reduction at Cycle 7, and two additional patients achieved a >50% spleen size reduction, representing an overall response rate of 40% at Cycle 7. In conclusion, the tolerability of this combination was acceptable, and hyperglycemia was the most frequent treatment-related AE. Ruxolitinib in combination with nilotinib and prednisone showed relevant clinical activity in patients with MF. This trial was registered with EudraCT Number 2016-005214-21.
Collapse
Affiliation(s)
- Rosa Ayala
- Haematological Malignancies Clinical Research UnitHospital Universitario 12 de Octubre, Universidad Complutense, CNIO, CIBERONCMadridSpain
| | | | | | | | - Santiago Osorio
- Hematology Department Hospital General UGregorio MarañónMadridSpain
| | | | | | - Noemi Álvarez
- Department of Translational HematologyResearch Institute Hospital 12 de Octubre (i+12)MadridSpain
| | | | - Antonia Duran
- Hematology Department Hospital Universitario Son EspasesPalma de MallorcaSpain
| | | | | | - Joaquín Martínez‐López
- Haematological Malignancies Clinical Research UnitHospital Universitario 12 de Octubre, Universidad Complutense, CNIO, CIBERONCMadridSpain
| |
Collapse
|
8
|
Pemmaraju N, Bose P, Rampal R, Gerds AT, Fleischman A, Verstovsek S. Ten years after ruxolitinib approval for myelofibrosis: a review of clinical efficacy. Leuk Lymphoma 2023:1-19. [PMID: 37081809 DOI: 10.1080/10428194.2023.2196593] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2023]
Abstract
Myelofibrosis (MF) is a chronic myeloproliferative neoplasm characterized by splenomegaly, abnormal cytokine expression, cytopenias, and progressive bone marrow fibrosis. The disease often manifests with burdensome symptoms and is associated with reduced survival. Ruxolitinib, an oral Janus kinase (JAK) 1 and JAK2 inhibitor, was the first agent approved for MF. As a first-in-class targeted treatment, ruxolitinib approval transformed the MF treatment approach and remains standard of care. In addition, targeted inhibition of JAK1/JAK2 signaling, a key molecular pathway underlying MF pathogenesis, and the large volume of literature evaluating ruxolitinib, have led to a better understanding of the disease and improved management in general. Here we review ruxolitinib efficacy in patients with MF in the 10 years following approval, including demonstration of clinical benefit in the phase 3 COMFORT-I/II trials, real-world evidence, translational studies, and expanded access data. Lastly, future directions for MF treatment are discussed, including ruxolitinib-based combination therapies.
Collapse
Affiliation(s)
- Naveen Pemmaraju
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Prithviraj Bose
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Raajit Rampal
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Aaron T Gerds
- Cleveland Clinic Taussig Cancer Institute, Cleveland, OH, USA
| | - Angela Fleischman
- Division of Hematology/Oncology, Medicine, University of California, Irvine, CA, USA
| | - Srdan Verstovsek
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
9
|
Downes CEJ, McClure BJ, McDougal DP, Heatley SL, Bruning JB, Thomas D, Yeung DT, White DL. JAK2 Alterations in Acute Lymphoblastic Leukemia: Molecular Insights for Superior Precision Medicine Strategies. Front Cell Dev Biol 2022; 10:942053. [PMID: 35903543 PMCID: PMC9315936 DOI: 10.3389/fcell.2022.942053] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 06/16/2022] [Indexed: 11/13/2022] Open
Abstract
Acute lymphoblastic leukemia (ALL) is the most common pediatric cancer, arising from immature lymphocytes that show uncontrolled proliferation and arrested differentiation. Genomic alterations affecting Janus kinase 2 (JAK2) correlate with some of the poorest outcomes within the Philadelphia-like subtype of ALL. Given the success of kinase inhibitors in the treatment of chronic myeloid leukemia, the discovery of activating JAK2 point mutations and JAK2 fusion genes in ALL, was a breakthrough for potential targeted therapies. However, the molecular mechanisms by which these alterations activate JAK2 and promote downstream signaling is poorly understood. Furthermore, as clinical data regarding the limitations of approved JAK inhibitors in myeloproliferative disorders matures, there is a growing awareness of the need for alternative precision medicine approaches for specific JAK2 lesions. This review focuses on the molecular mechanisms behind ALL-associated JAK2 mutations and JAK2 fusion genes, known and potential causes of JAK-inhibitor resistance, and how JAK2 alterations could be targeted using alternative and novel rationally designed therapies to guide precision medicine approaches for these high-risk subtypes of ALL.
Collapse
Affiliation(s)
- Charlotte EJ. Downes
- Blood Cancer Program, Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
- School of Biological Sciences, Faculty of Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Barbara J. McClure
- Blood Cancer Program, Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Daniel P. McDougal
- School of Biological Sciences, Faculty of Sciences, University of Adelaide, Adelaide, SA, Australia
- Institute for Photonics and Advanced Sensing (IPAS), University of Adelaide, Adelaide, SA, Australia
| | - Susan L. Heatley
- Blood Cancer Program, Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
- Australian and New Zealand Children’s Oncology Group (ANZCHOG), Clayton, VIC, Australia
| | - John B. Bruning
- School of Biological Sciences, Faculty of Sciences, University of Adelaide, Adelaide, SA, Australia
- Institute for Photonics and Advanced Sensing (IPAS), University of Adelaide, Adelaide, SA, Australia
| | - Daniel Thomas
- Blood Cancer Program, Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - David T. Yeung
- Blood Cancer Program, Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
- Department of Haematology, Royal Adelaide Hospital and SA Pathology, Adelaide, SA, Australia
| | - Deborah L. White
- Blood Cancer Program, Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
- School of Biological Sciences, Faculty of Sciences, University of Adelaide, Adelaide, SA, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
- Australian and New Zealand Children’s Oncology Group (ANZCHOG), Clayton, VIC, Australia
| |
Collapse
|
10
|
Saha C, Harrison C. Fedratinib, the first selective JAK2 inhibitor approved for treatment of myelofibrosis - an option beyond ruxolitinib. Expert Rev Hematol 2022; 15:583-595. [PMID: 35787092 DOI: 10.1080/17474086.2022.2098105] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Introduction: Myelofibrosis, a life shortening clonal disorder, presents with a constellation of features: bone marrow fibrosis, abnormal blood counts, extramedullary hematopoiesis, splenomegaly, thrombohemorrhagic complications and constitutional symptoms. Until recently Ruxolitinib, a JAK1 and 2 inhibitor, has been the only targeted therapy available for transplant-ineligible patients requiring treatment for splenomegaly and disease related symptoms. However, most patients discontinue Ruxolitinib after 3-5 years, mostly due to loss of response. There has been an unmet need for this patient group. In August 2019 Fedratinib (INREBIC® capsules, Impact Biomedicines, Inc., a wholly owned subsidiary of Bristol Meyer Squibb), a JAK2 inhibitor, was approved by US FDA for treatment of myelofibrosis in both JAK inhibitor naïve and pre-treated patients for the management of symptoms and splenomegaly.Areas covered: Here, we discuss the development, evidence base to date for Fedratinib. Including early and late phase, and ongoing trials, safety issues, potential role and current position of Fedratinib in the treatment of myelofibrosis, as well as future direction of targeted therapy in myelofibrosis.Expert opinion: Fedratinib presents a much needed option of treatment, particularly, for patients failing Ruxolitinib, with response rates that are quite similar. Nonetheless, there remain important questions including sequencing and options for combining therapy.
Collapse
Affiliation(s)
- Chandan Saha
- Department of Hematology, Guy's and St Thomas' NHS Foundation Trust, London
| | - Claire Harrison
- Department of Hematology, Guy's and St Thomas' NHS Foundation Trust, London
| |
Collapse
|
11
|
Bose P, Mesa RA. Novel strategies for challenging scenarios encountered in managing myelofibrosis. Leuk Lymphoma 2022; 63:774-788. [PMID: 34775887 PMCID: PMC11666286 DOI: 10.1080/10428194.2021.1999443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 10/16/2021] [Indexed: 10/19/2022]
Abstract
Given its rarity, multi-faceted clinical presentation and the relative paucity of approved therapies, the management of myeloproliferative neoplasm (MPN)-associated myelofibrosis (MF) can be challenging. Janus kinase (JAK) inhibitors, the only approved agents at present, have brought many clinical benefits to patients, with prolongation of survival also demonstrated for ruxolitinib. However, these agents have clear limitations. Optimal management of anemia in MF remains a major unmet need. Neither ruxolitinib nor fedratinib is recommended for use in patients with severe thrombocytopenia, i.e. platelets <50 × 109/L, who have a particularly poor prognosis. The search for the optimal partner for JAK inhibitors to address some of the shortcomings of these agents (e.g. limited ability to improve bone marrow fibrosis, cytopenias and induce molecular responses) and achieve meaningful 'disease modification' continues. This has led to the development of a number of rational, preclinically synergistic combinations for use either upfront or in the setting of sub-optimal response to JAK inhibition. Finally, the outlook for patients whose disease progresses on JAK inhibitor therapy continues to be grim, and agents with alternative mechanisms of action may be needed in this setting. In this article, we use a case-based approach to illustrate challenges commonly encountered in clinical practice and our management of the same. Fortunately, there has been enormous growth in drug development efforts in the MF space in the last few years, some of which appear poised to bear fruit in the very near future.
Collapse
Affiliation(s)
- Prithviraj Bose
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ruben A Mesa
- Mays Cancer Center, UT Health San Antonio MD Anderson, San Antonio, TX, USA
| |
Collapse
|
12
|
Waksal JA, Tremblay D, Mascarenhas J. Clinical Utility of Fedratinib in Myelofibrosis. Onco Targets Ther 2021; 14:4509-4521. [PMID: 34456572 PMCID: PMC8387309 DOI: 10.2147/ott.s267001] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 08/10/2021] [Indexed: 12/17/2022] Open
Abstract
Myelofibrosis (MF) is a clonal hematologic malignancy characterized by bone marrow fibrosis, extramedullary hematopoiesis, splenomegaly, and constitutional symptoms with a propensity towards leukemic transformation. Constitutive activation of the JAK/STAT pathway is a well-described pathogenic feature of MF. Allogeneic stem cell transplant is the only curative therapy, but due to high morbidity and mortality this option is not available for most patients. There are two approved targeted therapy options for MF, ruxolitinib and fedratinib. In this review, we discuss the clinical utility of fedratinib in the myelofibrosis treatment paradigm. Fedratinib has shown impressive pre-clinical and clinical efficacy in patients with untreated MF as well as in those with ruxolitinib intolerance and those with relapsed/refractory MF. Here, we review the pre-clinical and clinical trials that led to the approval of fedratinib, and the ongoing late-phase trials. We highlight several areas regarding the clinical utility of fedratinib that remain unanswered. We discuss the limitations of fedratinib and address areas that are understudied and require further clinical evaluation and research. The approval of fedratinib has provided a significant expansion to the very limited treatment armamentarium available to patients with MF.
Collapse
Affiliation(s)
- Julian A Waksal
- Department of Hematology and Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Douglas Tremblay
- Department of Hematology and Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John Mascarenhas
- Department of Hematology and Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
13
|
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
|
14
|
Abstract
Following the discovery of the JAK2V617F mutation in myeloproliferative neoplasms in 2005, fedratinib was developed as a small molecular inhibitor of JAK2. It was optimized to yield low-nanomolar activity against JAK2 (50% inhibitory concentration = 3 nM) and was identified to be selective for JAK2 relative to other JAK family members (eg, JAK1, JAK3, and TYK2). It quickly moved into clinical development with a phase 1 clinical trial opening in 2008, where a favorable impact on spleen and myelofibrosis (MF) symptom responses was reported. A phase 3 trial in JAK2 inhibitor treatment-naive MF patients followed in 2011 (JAKARTA); a phase 2 trial in MF patients resistant or intolerant to ruxolitinib followed in 2012 (JAKARTA-2). Clinical development suffered a major setback between 2013 and 2017 when the US Food and Drug Administration (FDA) placed fedratinib on clinical hold due to the development of symptoms concerning for Wernicke encephalopathy (WE) in 8 of 608 subjects (1.3%) who had received the drug. It was ultimately concluded that there was no evidence that fedratinib directly induces WE, but clear risk factors (eg, poor nutrition, uncontrolled gastrointestinal toxicity) were identified. In August 2019, the FDA approved fedratinib for the treatment of adults with intermediate-2 or high-risk MF. Notably, approval includes a "black box warning" on the risk of serious and fatal encephalopathy, including WE. FDA approval was granted on the basis of the JAKARTA studies in which the primary end points (ie, spleen and MF symptom responses) were met in ∼35% to 40% of patients (JAKARTA) and 25% to 30% of patients (JAKARTA-2), respectively.
Collapse
|
15
|
Ross DM, Babon JJ, Tvorogov D, Thomas D. Persistence of myelofibrosis treated with ruxolitinib: biology and clinical implications. Haematologica 2021; 106:1244-1253. [PMID: 33472356 PMCID: PMC8094080 DOI: 10.3324/haematol.2020.262691] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Indexed: 12/18/2022] Open
Abstract
Activation of JAK-STAT signaling is one of the hallmarks of myelofibrosis, a myeloproliferative neoplasm that leads to inflammation, progressive bone marrow failure, and a risk of leukemic transformation. Around 90% of patients with myelofibrosis have a mutation in JAK2, MPL, or CALR: so-called 'driver' mutations that lead to activation of JAK2. Ruxolitinib, and other JAK2 inhibitors in clinical use, provide clinical benefit but do not have a major impact on the abnormal hematopoietic clone. This phenomenon is termed 'persistence', in contrast to usual patterns of resistance. Multiple groups have shown that type 1 inhibitors of JAK2, which bind the active conformation of the enzyme, lead to JAK2 becoming resistant to degradation with consequent accumulation of phospho-JAK2. In turn, this can lead to exacerbation of inflammatory manifestations when the JAK inhibitor is discontinued, and it may also contribute to disease persistence. The ways in which JAK2 V617F and CALR mutations lead to activation of JAK-STAT signaling are incompletely understood. We summarize what is known about pathological JAK-STAT activation in myelofibrosis and how this might lead to future novel therapies for myelofibrosis with greater disease-modifying potential.
Collapse
Affiliation(s)
- David M Ross
- Department of Hematology and Bone Marrow Transplantation, Royal Adelaide Hospital, Adelaide; Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide; Precision Medicine Theme, South Australian Health and Medical Research Institute, and Adelaide Medical School, University of Adelaide.
| | - Jeffrey J Babon
- The Walter and Eliza Hall Institute of Medical Research and Department of Medical Biology, University of Melbourne, Parkville
| | - Denis Tvorogov
- Centre for Cancer Biology, University of South Australia and SA Pathology
| | - Daniel Thomas
- Precision Medicine Theme, South Australian Health and Medical Research Institute, and Adelaide Medical School, University of Adelaide
| |
Collapse
|
16
|
Palandri F, Tiribelli M, Breccia M, Bartoletti D, Elli EM, Benevolo G, Martino B, Cavazzini F, Tieghi A, Iurlo A, Abruzzese E, Pugliese N, Binotto G, Caocci G, Auteri G, Cattaneo D, Trawinska MM, Stella R, Scaffidi L, Polverelli N, Micucci G, Masselli E, Crugnola M, Bosi C, Heidel FH, Latagliata R, Pane F, Cuneo A, Krampera M, Semenzato G, Lemoli RM, Cavo M, Vianelli N, Bonifacio M, Palumbo GA. Ruxolitinib rechallenge in resistant or intolerant patients with myelofibrosis: Frequency, therapeutic effects, and impact on outcome. Cancer 2021; 127:2657-2665. [PMID: 33794557 DOI: 10.1002/cncr.33541] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/24/2021] [Accepted: 03/01/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND After ruxolitinib discontinuation, the outcome of patients with myelofibrosis (MF) is poor with scarce therapeutic possibilities. METHODS The authors performed a subanalysis of an observational, retrospective study (RUX-MF) that included 703 MF patients treated with ruxolitinib to investigate 1) the frequency and reasons for ruxolitinib rechallenge, 2) its therapeutic effects, and 3) its impact on overall survival. RESULTS A total of 219 patients (31.2%) discontinued ruxolitinib for ≥14 days and survived for ≥30 days. In 60 patients (27.4%), ruxolitinib was rechallenged for ≥14 days (RUX-again patients), whereas 159 patients (72.6%) discontinued it permanently (RUX-stop patients). The baseline characteristics of the 2 cohorts were comparable, but discontinuation due to a lack/loss of spleen response was lower in RUX-again patients (P = .004). In comparison with the disease status at the first ruxolitinib stop, at its restart, there was a significant increase in patients with large splenomegaly (P < .001) and a high Total Symptom Score (TSS; P < .001). During the rechallenge, 44.6% and 48.3% of the patients had spleen and symptom improvements, respectively, with a significant increase in the number of patients with a TSS reduction (P = .01). Although the use of a ruxolitinib dose > 10 mg twice daily predicted better spleen (P = .05) and symptom improvements (P = .02), the reasons for/duration of ruxolitinib discontinuation and the use of other therapies before rechallenge were not associated with rechallenge efficacy. At 1 and 2 years, 33.3% and 48.3% of RUX-again patients, respectively, had permanently discontinued ruxolitinib. The median overall survival was 27.9 months, and it was significantly longer for RUX-again patients (P = .004). CONCLUSIONS Ruxolitinib rechallenge was mainly used in intolerant patients; there were clinical improvements and a possible survival advantage in many cases, but there was a substantial rate of permanent discontinuation. Ruxolitinib rechallenge should be balanced against newer therapeutic possibilities.
Collapse
Affiliation(s)
- Francesca Palandri
- Istituto di Ematologia "Seràgnoli," IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Mario Tiribelli
- Division of Hematology and Bone Marrow Transplantation, Azienda Sanitaria Universitaria Integrata di Udine, Udine, Italy
| | - Massimo Breccia
- Division of Cellular Biotechnologies and Hematology, University Sapienza, Rome, Italy
| | - Daniela Bartoletti
- Istituto di Ematologia "Seràgnoli," IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università di Bologna, Bologna, Italy
| | - Elena M Elli
- Hematology Division and Bone Marrow Unit, San Gerardo Hospital, Azienda Socio Sanitaria Territoriale Monza, Monza, Italy
| | - Giulia Benevolo
- Division of Hematology, Città della Salute e della Scienza Hospital, Turin, Italy
| | - Bruno Martino
- Division of Hematology, Azienda Ospedaliera "Bianchi Melacrino Morelli", Reggio Calabria, Italy
| | | | - Alessia Tieghi
- Department of Hematology, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Alessandra Iurlo
- Hematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Novella Pugliese
- Department of Clinical Medicine and Surgery, Federico II University Medical School, Naples, Italy
| | - Gianni Binotto
- Unit of Hematology and Clinical Immunology, University of Padua, Padua, Italy
| | - Giovanni Caocci
- Ematologia, Ospedale Businco, Università degli Studi di Cagliari, Cagliari, Italy
| | - Giuseppe Auteri
- Istituto di Ematologia "Seràgnoli," IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università di Bologna, Bologna, Italy
| | - Daniele Cattaneo
- Hematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Rossella Stella
- Division of Hematology and Bone Marrow Transplantation, Azienda Sanitaria Universitaria Integrata di Udine, Udine, Italy
| | - Luigi Scaffidi
- Section of Hematology, University of Verona, Verona, Italy
| | - Nicola Polverelli
- Unit of Blood Diseases and Stem Cell Transplantation, Azienda Socio Sanitaria Territoriale degli Spedali Civili di Brescia, Brescia, Italy
| | - Giorgia Micucci
- Hematology and Stem Cell Transplant Center, Azienda Ospedaliera Ospedali Riuniti Marche Nord, Pesaro, Italy
| | - Elena Masselli
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Monica Crugnola
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Costanza Bosi
- Division of Hematology, AUSL di Piacenza, Piacenza, Italy
| | - Florian H Heidel
- Hematology and Oncology, Friedrich Schiller University Medical Center, Jena, Germany
| | | | - Fabrizio Pane
- Department of Clinical Medicine and Surgery, Federico II University Medical School, Naples, Italy
| | - Antonio Cuneo
- Division of Hematology, University of Ferrara, Ferrara, Italy
| | - Mauro Krampera
- Section of Hematology, University of Verona, Verona, Italy
| | | | - Roberto M Lemoli
- Clinic of Hematology, Department of Internal Medicine, University of Genoa, Genoa, Italy.,IRCCS Policlinico San Martino, Genova, Italy
| | - Michele Cavo
- Istituto di Ematologia "Seràgnoli," IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università di Bologna, Bologna, Italy
| | - Nicola Vianelli
- Istituto di Ematologia "Seràgnoli," IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | | | - Giuseppe A Palumbo
- Department of Scienze Mediche, Chirurgiche e Tecnologie Avanzate "G. F. Ingrassia," University of Catania, Italy
| |
Collapse
|
17
|
Ruxolitinib discontinuation syndrome: incidence, risk factors, and management in 251 patients with myelofibrosis. Blood Cancer J 2021; 11:4. [PMID: 33414394 PMCID: PMC7791065 DOI: 10.1038/s41408-020-00392-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/02/2020] [Accepted: 11/10/2020] [Indexed: 12/25/2022] Open
|
18
|
Garcia J, Compte A, Bassa P, Mourello S, Ortiz S, Riera E. A polycythemia vera case demonstrated on 18F-Choline PET/CT. Rev Esp Med Nucl Imagen Mol 2021. [DOI: 10.1016/j.remnie.2020.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
19
|
Garcia JR, Compte A, Bassa P, Mourello S, Ortiz S, Riera E. A polycythemia vera case demonstrated on 18F-Choline PET/CT. Rev Esp Med Nucl Imagen Mol 2020; 40:50-51. [PMID: 33011102 DOI: 10.1016/j.remn.2020.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 04/14/2020] [Accepted: 04/16/2020] [Indexed: 11/25/2022]
Affiliation(s)
- J R Garcia
- CETIR ASCIRES Grupo biomédico, Barcelona, España.
| | - A Compte
- CETIR ASCIRES Grupo biomédico, Barcelona, España
| | - P Bassa
- CETIR ASCIRES Grupo biomédico, Barcelona, España
| | - S Mourello
- CETIR ASCIRES Grupo biomédico, Barcelona, España
| | - S Ortiz
- CETIR ASCIRES Grupo biomédico, Barcelona, España
| | - E Riera
- CETIR ASCIRES Grupo biomédico, Barcelona, España
| |
Collapse
|
20
|
Bose P, Verstovsek S. JAK Inhibition for the Treatment of Myelofibrosis: Limitations and Future Perspectives. Hemasphere 2020; 4:e424. [PMID: 32903304 PMCID: PMC7375176 DOI: 10.1097/hs9.0000000000000424] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 05/26/2020] [Indexed: 12/24/2022] Open
Abstract
The 2011 approval of ruxolitinib ushered in the Janus kinase (JAK) inhibitor era in the treatment of myelofibrosis (MF), and 2019 saw the US approval of fedratinib. The first therapeutic agents approved by regulatory authorities for MF, these drugs attenuate the overactive JAK-signal transducer and activator of transcription (STAT) signaling universally present in these patients, translating into major clinical benefits in terms of spleen shrinkage and symptom improvement. These, in turn, confer a survival advantage on patients with advanced disease, demonstrated in the case of ruxolitinib, for which long-term follow-up data are available. However, JAK inhibitors do not improve cytopenias in most patients, have relatively modest effects on bone marrow fibrosis and driver mutation allele burden, and clinical resistance eventually develops. Furthermore, they do not modify the risk of transformation to blast phase; indeed, their mechanism of action may be more anti-inflammatory than truly disease-modifying. This has spurred interest in rational combinations of JAK inhibitors with other agents that may improve cytopenias and drugs that could potentially modify the natural history of MF. Newer JAK inhibitors that are distinguished from ruxolitinib and fedratinib by their ability to improve anemia (eg, momelotinib) or safety and efficacy in severely thrombocytopenic patients (eg, pacritinib) are in phase 3 clinical trials. There is also interest in developing inhibitors that are highly selective for mutant JAK2, as well as "type II" JAK2 inhibitors. Overall, although current JAK inhibitors have limitations, they will likely continue to form the backbone of MF therapy for the foreseeable future.
Collapse
Affiliation(s)
- Prithviraj Bose
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Srdan Verstovsek
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
21
|
Bose P, Verstovsek S. Management of myelofibrosis after ruxolitinib failure. Leuk Lymphoma 2020; 61:1797-1809. [PMID: 32297800 PMCID: PMC8565616 DOI: 10.1080/10428194.2020.1749606] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Revised: 03/16/2020] [Accepted: 03/25/2020] [Indexed: 12/12/2022]
Abstract
Over the last decade, the Janus kinase1/2 (JAK1/2) inhibitor ruxolitinib has emerged as a cornerstone of myelofibrosis (MF) management. Ruxolitinib improves splenomegaly and symptoms regardless of driver mutation status, and confers a survival advantage in patients with intermediate-2/high risk MF. However, cytopenias remain problematic, and evidence for a robust anti-clonal effect is lacking. Furthermore, the median duration of spleen response to ruxolitinib in clinical trials is approximately 3 years, and ruxolitinib does not appear to affect the risk of leukemic transformation. There is no therapy approved specifically for patients whose disease 'progresses' on ruxolitinib, defining which remains challenging. The recent regulatory approval of the JAK2 inihibitor fedratinib partially fulfills this unmet need, but much remains to be done. Other JAK inhibitors and a plethora of novel agents are being studied in the ruxolitinib 'failure' setting, as well as 'add-on' therapies to ruxolitinib in patients having a 'sub-optimal' response.
Collapse
Affiliation(s)
- Prithviraj Bose
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Srdan Verstovsek
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
22
|
Harrison CN, Schaap N, Mesa RA. Management of myelofibrosis after ruxolitinib failure. Ann Hematol 2020; 99:1177-1191. [PMID: 32198525 PMCID: PMC7237516 DOI: 10.1007/s00277-020-04002-9] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 03/12/2020] [Indexed: 12/20/2022]
Abstract
Myelofibrosis is a BCR-ABL1-negative myeloproliferative neoplasm characterized by anemia, progressive splenomegaly, extramedullary hematopoiesis, bone marrow fibrosis, constitutional symptoms, leukemic progression, and shortened survival. Constitutive activation of the Janus kinase/signal transducers and activators of transcription (JAK-STAT) pathway, and other cellular pathways downstream, leads to myeloproliferation, proinflammatory cytokine expression, and bone marrow remodeling. Transplant is the only curative option for myelofibrosis, but high rates of morbidity and mortality limit eligibility. Several prognostic models have been developed to facilitate treatment decisions. Until the recent approval of fedratinib, a JAK2 inhibitor, ruxolitinib was the only available JAK inhibitor for treatment of intermediate- or high-risk myelofibrosis. Ruxolitinib reduces splenomegaly to some degree in almost all treated patients; however, many patients cannot tolerate ruxolitinib due to dose-dependent drug-related cytopenias, and even patients with a good initial response often develop resistance to ruxolitinib after 2-3 years of therapy. Currently, there is no consensus definition of ruxolitinib failure. Until fedratinib approval, strategies to overcome ruxolitinib resistance or intolerance were mainly different approaches to continued ruxolitinib therapy, including dosing modifications and ruxolitinib rechallenge. Fedratinib and two other JAK2 inhibitors in later stages of clinical development, pacritinib and momelotinib, have been shown to induce clinical responses and improve symptoms in patients previously treated with ruxolitinib. Fedratinib induces robust spleen responses, and pacritinib and momelotinib may have preferential activity in patients with severe cytopenias. Reviewed here are strategies to ameliorate ruxolitinib resistance or intolerance, and outcomes of clinical trials in patients with myelofibrosis receiving second-line JAK inhibitors after ruxolitinib treatment.
Collapse
Affiliation(s)
- Claire N Harrison
- Guy's and St Thomas' Hospital Foundation Trust, Westminster Bridge Rd, London, SE1 7EH, UK.
| | | | - Ruben A Mesa
- University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| |
Collapse
|
23
|
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: 3.2] [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
|
24
|
Bose P. Advances in potential treatment options for myeloproliferative neoplasm associated myelofibrosis. Expert Opin Orphan Drugs 2019; 7:415-425. [PMID: 33094033 PMCID: PMC7577425 DOI: 10.1080/21678707.2019.1664900] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 09/04/2019] [Indexed: 12/11/2022]
Abstract
INTRODUCTION The Janus kinase (JAK)1/2 inhibitor ruxolitinib provides rapid, sustained and often dramatic benefits to patients with myelofibrosis, inducing spleen shrinkage and ameliorating symptoms, and improves survival. However, the drug has little effect on the underlying bone marrow fibrosis or on mutant allele burden, and clinical resistance eventually develops. Furthermore, ruxolitinib-induced cytopenias can be challenging in everyday practice. AREAS COVERED The developmental therapeutics landscape in MF is discussed. This includes potential partners for ruxolitinib being developed with an aim to improve cytopenias, or to enhance its disease-modifying effects. The development of other JAK inhibitors with efficacy post-ruxolitinib or other unique attributes is being pursued in earnest. Agents with novel mechanisms of action are being studied in patients whose disease responds sub-optimally to, is refractory to or progresses after ruxolitinib. EXPERT OPINION The JAK inhibitors fedratinib, pacritinib and momelotinib are clearly active, and it is expected that one or more of these will become licensed in the future. The activin receptor ligand traps are promising as treatments for anemia. Imetelstat has shown interesting activity post-ruxolitinib, and azactidine may be a useful partner for ruxolitinib in some patients. Appropriately, multiple pre-clinical and clinical leads are being pursued in this difficult therapeutic area.
Collapse
Affiliation(s)
- Prithviraj Bose
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
25
|
MF management. Hemasphere 2019; 3:HemaSphere-2019-0024. [PMID: 35309812 PMCID: PMC8925717 DOI: 10.1097/hs9.0000000000000210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 03/12/2019] [Accepted: 03/13/2019] [Indexed: 11/25/2022] Open
|
26
|
Agbaria S, Haim A, Fares F, Zubidat AE. Epigenetic modification in 4T1 mouse breast cancer model by artificial light at night and melatonin - the role of DNA-methyltransferase. Chronobiol Int 2019; 36:629-643. [PMID: 30746962 DOI: 10.1080/07420528.2019.1574265] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Currently, one of the most disputed hypotheses regarding breast cancer (BC) development is exposure to short wavelength artificial light at night (ALAN) as multiple studies suggest a possible link between them. This link is suggested to be mediated by nocturnal melatonin suppression that plays an integral role in circadian regulations including cell division. The objective of the research was to evaluate effects of 1 × 30 min/midnight ALAN (134 µ Wcm-2, 460 nm) with or without nocturnal melatonin supplement on tumor development and epigenetic responses in 4T1 tumor-bearing BALB/c mice. Mice were monitored for body mass (Wb) and tumor volume for 3 weeks and thereafter urine samples were collected at regular intervals for determining daily rhythms of 6-sulfatoxymelatonin (6-SMT). Finally, mice were sacrificed and the tumor, lungs, liver, and spleen were excised for analyzing the total activity of DNA methyltransferases (DNMT) and global DNA methylation (GDM) levels. Mice exposed to ALAN significantly reduced 6-SMT levels and increased Wb, tumor volume, and lung metastasis compared with controls. These effects were diminished by melatonin. The DNMT activity and GDM levels showed tissue-specific response. The enzymatic activity and GDM levels were lower in tumor and liver and higher in spleen and lungs under ALAN compared with controls. Our results suggest that ALAN disrupts the melatonin rhythm and potentially leading to increased BC burden by affecting DNMT activity and GDM levels. These data may also be applicable to early detection and management of BC by monitoring melatonin and GDM levels as early biomarker of ALAN circadian disruption.
Collapse
Affiliation(s)
- Sahar Agbaria
- a Department of Human Biology , University of Haifa , Haifa , Israel
| | - Abraham Haim
- b The Israeli Center for Interdisciplinary Research in Chronobiology , University of Haifa , Haifa , Israel
| | - Fuad Fares
- a Department of Human Biology , University of Haifa , Haifa , Israel.,c Department of Molecular Genetics , Carmel Medical Center , Haifa , Israel
| | - Abed E Zubidat
- b The Israeli Center for Interdisciplinary Research in Chronobiology , University of Haifa , Haifa , Israel
| |
Collapse
|