101
|
McKinnell Z, Karel D, Tuerff D, SH Abrahim M, Nassereddine S. Acute Myeloid Leukemia Following Myeloproliferative Neoplasms: A Review of What We Know, What We Do Not Know, and Emerging Treatment Strategies. J Hematol 2022; 11:197-209. [PMID: 36632576 PMCID: PMC9822656 DOI: 10.14740/jh1042] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 10/15/2022] [Indexed: 01/04/2023] Open
Abstract
Acute myeloid leukemia (AML) arising from myeloproliferative neoplasms (MPNs) represents a small subtype of secondary AML (sAML). This entity is well known to be associated with poor responses to available treatment options and dismal outcomes. To date, there are no standardized treatment options and there has been very little therapeutic advancement in recent years. This is a stark contrast to other subsets of AML for which there have been significant advances in therapeutic approaches, especially for patients with targetable mutations. We aim to focus our review on the incidence, risk factors for leukemogenesis, pathogenesis, molecular landscape, and emerging therapeutic options in post-myeloproliferative neoplasm acute myeloid leukemia (post-MPN AML).
Collapse
Affiliation(s)
- Zoe McKinnell
- Department of Hematology and Oncology, George Washington University Hospital, Washington, DC, USA
| | - Daniel Karel
- Department of Hematology and Oncology, George Washington University Hospital, Washington, DC, USA
| | - Daniel Tuerff
- Department of Hematology and Oncology, George Washington University Hospital, Washington, DC, USA
| | - Marwa SH Abrahim
- Department of Hematology and Oncology, George Washington University Hospital, Washington, DC, USA
| | - Samah Nassereddine
- Department of Hematology and Oncology, George Washington University Hospital, Washington, DC, USA,Corresponding Author: Samah Nassereddine, Department of Hematology and Oncology, George Washington University and George Washington Cancer Center, Washington, DC, USA.
| |
Collapse
|
102
|
Vinogradova OY, Pankrashkina MM, Shikhbabaeva DI, Chernikov MV, Neverova AL, Ivanova VL, Nikitin EA, Usikova EV, Ptushkin VV. Possibilities of targeted therapy for myelofibrosis: Moscow experience. ONCOHEMATOLOGY 2022. [DOI: 10.17650/1818-8346-2022-17-4-94-105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Background. For many years the primary aim of treatment strategy for ph-negative myeloproliferative neoplasms has been to restrain disease progression, with lasting relief and management of symptoms to improve patients’ quality of life. Generally, this did not lead to a significant increase in life expectancy with primary myelofibrosis and didn’t decrease the risk of fibrosis in patients with polycythemia vera and essential thrombocythemia. To date a new class of targeted drugs has been developed, it is JAK2 inhibitors with pathogenetic effects. The results of clinical trials showed the high efficacy of the first registered drug of this its kind – ruxolitinib – that includes a faster reduction in the symptoms of tumor intoxication and in symptoms associated with the development of splenomegaly and increase in the overall survival rates. It is known that the data obtained during clinical trials of medicines may differ from the results obtained in routine clinical practice. In actual practice drugs are used in a much wider heterogeneous population of patients, less limited first of all by age and comorbid characteristics. It is possible to analyze cohorts of patients including a larger number of clinical cases with a longer follow-up period. In this regard of great interest is the actual clinical experience of long-term use of ruxolitinib in patients whose set is limited only by clinical contraindications for prescribing the drug.Aim. To present our own actual experience of targeted therapy of myelofibrosis and compare the results obtained with the data of clinical trials.Materials and methods. Our analysis includes data from 141 patients (67 (47.5 %) men and 74 (52.5 %) women) in a chronic phase myelofibrosis. All patients received ruxolitinib. Of these, 109 (69 %) patients had primary myelofibrosis, 26 (16 %) – postpolycythemia myelofibrosis, 6 (4 %) – postessential thrombocythemia myelofibrosis. The median age at the start of therapy was 62 (18–84) years. The median disease duration before ruxolitinib was prescribed – 79 (1–401) months. According to the dIpSS (dynamic International prognostic Scoring System) criteria, 13 % of patients were assigned to the low risk group, 38 % – to the intermediate-1, 36 % – to the intermediate-2, 13 % – to the high risk group. Most patients (52 %) had grade 3 bone marrow fibrosis.Results. The median duration of treatment was 18 (range from 1 to 115) months. Symptoms of intoxication were relieved 74 (81 %) of 91 patients, the spleen size decreased in 81 % of patients (the spleen size returned to normal in 25 % of patients). The increase in the median hemoglobin level was 15 %. The proportion of patients requiring blood transfusion decreased by 4 times (from 39 to 9 %). Mean platelet levels normalized in most patients with baseline high and low platelet levels. A complete clinical and hematological response was achieved in 16 % (n = 23) of cases, a partial response – in 26 % (n = 37) of cases, clinical improvement – in 21 % (n = 30), disease stabilization – in 33 % (n = 46) of cases. No response was received in 1 (1 %) patient and in 3 (3 %) cases there was progression of the disease. At the time of analysis, 81 (57 %) of 141 patients were continuing the ruxolitinib treatment. The fatal outcome in 33 (22 %) patients was associated with concomitant diseases, among which 20 (14 %) died from proven COvId-19 infection. Overall survival: 1-year 81 %, 2-year 73 %, 5-year 50 %. Overall survival excluding deaths due to COvId-19: 1-year 92 %, 2-year 85 %, 5-year 70 %. Massive splenomegaly and a high degree of fibrosis were unfavorable predictors of prognosis of overall survival.Conclusion. Target therapy with Janus kinase inhibitor ruxolitinib has demonstrated high efficacy in patients with myelofibrosis in routine clinical practice. The most rapid effect ruxolitinib had on the spleen size and the symptoms of intoxication. Tolerability of ruxolitinib therapy was generally satisfactory. The overall and progression-free survival rates in patients with myelofibrosis, receiving ruxolitinib in the clinical setting was consistent with the results of international multicenter clinical trials.
Collapse
Affiliation(s)
- O. Yu. Vinogradova
- Moscow City Hematology Center, S.P. Botkin City Clinical Hospital, Moscow Healthcare Department; Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Ministry of Health of Russia; N.I. Pirogov Russian National Research Medical University, Ministry of Health of Russia
| | - M. M. Pankrashkina
- Moscow City Hematology Center, S.P. Botkin City Clinical Hospital, Moscow Healthcare Department
| | - D. I. Shikhbabaeva
- Moscow City Hematology Center, S.P. Botkin City Clinical Hospital, Moscow Healthcare Department
| | - M. V. Chernikov
- Moscow City Hematology Center, S.P. Botkin City Clinical Hospital, Moscow Healthcare Department
| | - A. L. Neverova
- Moscow City Hematology Center, S.P. Botkin City Clinical Hospital, Moscow Healthcare Department
| | - V. L. Ivanova
- Moscow City Hematology Center, S.P. Botkin City Clinical Hospital, Moscow Healthcare Department
| | - E. A. Nikitin
- Moscow City Hematology Center, S.P. Botkin City Clinical Hospital, Moscow Healthcare Department
| | - E. V. Usikova
- Moscow City Hematology Center, S.P. Botkin City Clinical Hospital, Moscow Healthcare Department
| | - V. V. Ptushkin
- Moscow City Hematology Center, S.P. Botkin City Clinical Hospital, Moscow Healthcare Department; Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Ministry of Health of Russia; N.I. Pirogov Russian National Research Medical University, Ministry of Health of Russia
| |
Collapse
|
103
|
Jalles C, Lepelley M, Mouret S, Charles J, Leccia MT, Trabelsi S. Skin cancers under Janus kinase inhibitors: A World Health Organization drug safety database analysis. Therapie 2022; 77:649-656. [PMID: 35710462 DOI: 10.1016/j.therap.2022.04.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 03/27/2022] [Accepted: 04/29/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Janus kinase (JAK) inhibitors are targeted therapies with a potential imunomodulatory and anti-inflammatory effect, indicated in various dysimmune pathologies. Skin cancers have been reported to occur in patients treated with JAK inhibitors. However, drug safety in clinical trials did not confirm that risk, but these studies are performed on controlled population and in a limited time of follow up. OBJECTIVES The aim of this study is to evaluate in real life condition if a disproportionality signal exists between JAK inhibitors treatment and skin cancers. METHODS We performed cases/non cases analysis in VigiBase® (the World Health Organization international database of suspected adverse drug reaction) using information component to search for a disproportionality signal of skin cancers from JAK inhibitor. We extracted all reports of skin cancers from the French Pharmacovigilance database occurring since 1978 up to 31st December 2019 for the three existing JAK inhibitors on market: ruxolitinib, tofacitinib and baricitinib. Only melanoma, squamous cell carcinoma and Merkel cell carcinoma were analyzed, according to the pathophysiology of these cancers and their link with immunosuppression. RESULTS A disproportionality signal was found positive for squamous cell carcinoma with ruxolitinib (IC025=3.92) and tofacitinib (IC025=0.82), for melanoma with ruxolitinib (IC025=0.81) and tofacitinib (IC025=0.74), and Merkel cell carcinoma with ruxolitinib (IC025=4) and tofactinib (IC025=1.01) and only for Merkel cell carcinoma with baricitinib (IC025=0.53). Moreover, Merkel cell carcinoma, a very rare skin cancer more prevalent in immunodepressed patients was particularly represented in our sample and was associated with a significant disproportionality signal with all the studied JAK inhibitors. CONCLUSION Our study shows that JAK inhibitors could be associated with an extra risk to develop skin cancers. Could an anti-viral or immunovigilance disruption mechanism brought by JAK inhibitors explain an over-risk with Merkel cell carcinoma, which were notably represented in our sample? Considering pharmacovigilance method limitations, further pharmacoepidemiological studies are required to assess a causal link between JAK inhibitors treatment and skin cancers development.
Collapse
Affiliation(s)
- Cédric Jalles
- Dermatology Department, Grenoble Alpes University Hospital, Hôpital Michallon Site Nord BD de la Chantourne, 38700 La Tronche, France.
| | - Marion Lepelley
- Pharmacology Department, Grenoble Alpes University Hospital, 38700 La Tronche, France
| | - Stéphane Mouret
- Dermatology Department, Grenoble Alpes University Hospital, Hôpital Michallon Site Nord BD de la Chantourne, 38700 La Tronche, France
| | - Julie Charles
- Dermatology Department, Grenoble Alpes University Hospital, Hôpital Michallon Site Nord BD de la Chantourne, 38700 La Tronche, France; University Grenoble Alpes, 38400 Saint-Martin-d'Hères, France
| | - Marie-Thérèse Leccia
- Dermatology Department, Grenoble Alpes University Hospital, Hôpital Michallon Site Nord BD de la Chantourne, 38700 La Tronche, France; University Grenoble Alpes, 38400 Saint-Martin-d'Hères, France
| | - Sabiha Trabelsi
- Dermatology Department, Grenoble Alpes University Hospital, Hôpital Michallon Site Nord BD de la Chantourne, 38700 La Tronche, France
| |
Collapse
|
104
|
Breccia M, Assanto GM, Laganà A, Scalzulli E, Martelli M. Novel therapeutic agents for myelofibrosis after failure or suboptimal response to JAK2 inhbitors. Curr Opin Oncol 2022; 34:729-737. [PMID: 36017560 DOI: 10.1097/cco.0000000000000898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW JAK2 inhibitors have changed the therapeutic strategies for the management of primary and secondary myelofibrosis. Ruxolitinib, the first available agent, improved disease-related symptoms, spleen volume, and overall survival compared to conventional chemotherapy. It has been revealed that after 3 years of treatment, about 50% of patients discontinued ruxolitinib for resistance and/or intolerance and should be candidate to a second line of treatment. RECENT FINDINGS Second-generation tyrosine kinase inhibitors have been tested in this setting, but all these new drugs do not significantly impact on disease progression. Novel agents are in developments that target on different pathways, alone or in combination with JAK2 inhibitors. SUMMARY In this review, we summarize all the clinical efficacy and safety data of these drugs providing a vision of the possible future.
Collapse
Affiliation(s)
- Massimo Breccia
- Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | | | | | | | | |
Collapse
|
105
|
Severe ARDS due to Ruxolitinib discontinuation syndrome: case presentation and literature review. Heliyon 2022; 8:e11782. [DOI: 10.1016/j.heliyon.2022.e11782] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 06/06/2022] [Accepted: 11/14/2022] [Indexed: 11/30/2022] Open
|
106
|
Perram J, Ross DM, McLornan D, Gowin K, Kröger N, Gupta V, Lewis C, Gagelmann N, Hamad N. Innovative strategies to improve hematopoietic stem cell transplant outcomes in myelofibrosis. Am J Hematol 2022; 97:1464-1477. [PMID: 35802782 PMCID: PMC9796730 DOI: 10.1002/ajh.26654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/28/2022] [Accepted: 07/05/2022] [Indexed: 01/28/2023]
Abstract
Myelofibrosis (MF) is a clonal myeloproliferative neoplasm characterized by inflammation, marrow fibrosis, and an inherent risk of blastic transformation. Hematopoietic allogeneic stem cell transplant is the only potentially curative therapy for this disease, however, survival gains observed for other transplant indications over the past two decades have not been realized for MF. The role of transplantation may also evolve with the use of novel targeted agents. The chronic inflammatory state associated with MF necessitates pretransplantation assessment of end-organ function. Applying the transplant methodology employed for other myeloid disorders to patients with MF fails to acknowledge differences in the underlying disease pathophysiology. Limited understanding of the causes of poor transplant outcomes in this cohort has prevented refinement of transplant eligibility criteria in MF. There is increasing evidence of heterogeneity in molecular disease grade, beyond the clinical manifestations which have traditionally guided transplant timing. Exploring the physiological consequences of disease chronicity unique to MF, acknowledging the heterogeneity in disease grade, and using advanced prognostic models, molecular diagnostics and other organ function diagnostic tools, we present an innovative review of strategies with the potential to improve transplant outcomes in this disease. Larger, prospective studies which consider the impact of molecular-based disease grade are needed for MF transplantation.
Collapse
Affiliation(s)
- Jacinta Perram
- Department of Bone Marrow Transplantation and HaematologySt Vincent's HospitalDarlinghurstNew South WalesAustralia,School of Clinical Medicine, UNSW Medicine & HealthKensingtonNew South WalesAustralia
| | - David M. Ross
- Department of Haematology and Bone Marrow TransplantationRoyal Adelaide HospitalAdelaideSouth AustraliaAustralia,Centre for Cancer BiologySA Pathology and University of South AustraliaAdelaideSouth AustraliaAustralia
| | - Donal McLornan
- Department of Haematology and Stem Cell TransplantationUniversity College London Hospitals NHSLondonUK
| | - Krisstina Gowin
- Department of Hematology and OncologyBone Marrow Transplant and Cellular Therapy, University of ArizonaTucsonArizonaUSA
| | - Nicolas Kröger
- Department of Stem Cell TransplantationUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Vikas Gupta
- Medical Oncology and HaematologyPrincess Margaret Cancer CentreTorontoOntarioCanada
| | - Clinton Lewis
- Department of HaematologyAuckland City HospitalAucklandNew Zealand
| | - Nico Gagelmann
- Department of Stem Cell TransplantationUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Nada Hamad
- Department of Bone Marrow Transplantation and HaematologySt Vincent's HospitalDarlinghurstNew South WalesAustralia,School of Clinical Medicine, UNSW Medicine & HealthKensingtonNew South WalesAustralia,School of MedicineUniversity of Notre Dame AustraliaFremantleWestern AustraliaAustralia
| |
Collapse
|
107
|
Jones E, Dillon B, Swan D, Thachil J. Practical management of the haemorrhagic complications of myeloproliferative neoplasms. Br J Haematol 2022; 199:313-321. [PMID: 35724983 PMCID: PMC9796684 DOI: 10.1111/bjh.18322] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/06/2022] [Accepted: 06/09/2022] [Indexed: 01/07/2023]
Abstract
Myeloproliferative neoplasms can be associated with bleeding manifestations which can cause significant morbidities. Although haematologists are aware of the likelihood of this complication in the setting of myeloproliferative neoplasms, it may often be overlooked especially in patients with no extreme elevation of blood counts and those with myelofibrosis. Acquired von Willebrand syndrome and platelet dysfunction are the two common diagnoses to be considered in this regard. In this review article, we discuss the mechanisms for the development of these rare bleeding disorders, their diagnosis and practical management.
Collapse
Affiliation(s)
- Edward Jones
- Department of HaematologySt James' HospitalDublinIreland
| | - Bryan Dillon
- Department of HaematologySt James' HospitalDublinIreland
| | - Dawn Swan
- National University IrelandGalwayIreland
| | - Jecko Thachil
- Department of HaematologyManchester University Hospitals NHS Foundation TrustManchesterUK
| |
Collapse
|
108
|
High Incidence of Herpes Zoster in Patients Using Ruxolitinib for Myeloproliferative Neoplasms: Need for Prophylaxis. Hemasphere 2022; 6:e793. [PMID: 36325270 PMCID: PMC9619234 DOI: 10.1097/hs9.0000000000000793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 09/22/2022] [Indexed: 11/23/2022] Open
|
109
|
England JT, Gupta V. Fedratinib: a pharmacotherapeutic option for JAK-inhibitor naïve and exposed patients with myelofibrosis. Expert Opin Pharmacother 2022; 23:1677-1686. [PMID: 36252265 DOI: 10.1080/14656566.2022.2135989] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Ruxolitinib is the most commonly used JAK-inhibitor (JAKi) for the management of symptoms related to splenomegaly and cytokine-mediated inflammation in patients with myelofibrosis (MF), but is limited by variable durability of response with most patients experiencing failure after 2-3 years. Long-term data on other approved JAKi, fedratinib and pacritinib, are not available due to the clinical hold put on pivotal trials for toxicity concerns. AREAS COVERED Following the initial hold for concern of Wernicke's encephalopathy, fedratinib was approved by the Food and Drug Administration (FDA) in 2019 for MF. We review the data available from early, and late phase critical trials, outline a role for fedratinib in the current treatment landscape of MF, and highlight the knowledge gaps in optimizing use of fedratinib. EXPERT OPINION The JAKARTA and JAKARTA2 trials established efficacy in spleen volume response (SVR) and symptom reduction in JAKi-naïve and ruxolitinib-exposed MF patients, respectively. Further trials, FREEDOM and FREEDOM2, are in progress to understand long-term effects of fedratinib; and include strategies to mitigate gastrointestinal toxicity, monitor thiamine levels and surveil for encephalopathy. We use fedratinib for symptomatic MF following ruxolitinib failure in patients without significant cytopenias; with practical strategies for monitoring and managing potential toxicity.
Collapse
Affiliation(s)
- James T England
- Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Vikas Gupta
- Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
110
|
Jung EH, Hong J, Kim SY, Park Y, Yuh YJ, Mun YC, Lee WS, Park SK, Bang SM. Real-World Outcomes of Ruxolitinib in Patients With Myelofibrosis Focusing on Red Blood Cell Transfusion: A Multicenter Study From the MPN Working Party of the Korean Society of Hematology. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2022; 22:e931-e937. [PMID: 35858905 DOI: 10.1016/j.clml.2022.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 06/21/2022] [Indexed: 06/15/2023]
Abstract
INTRODUCTION/BACKGROUND Ruxolitinib is an established treatment for myelofibrosis (MF) that has demonstrated clinical benefit by reducing spleen size and debilitating MF-related symptoms. However, despite the efficacy of ruxolitinib, anemia remains a major adverse event that causes dose modification or discontinuation in real-world practice. Additionally, dependence on red blood cell (RBC) transfusion (TF) is common during treatment; therefore, we explored the outcome of ruxolitinib therapy with a primary focus on RBC TF. PATIENTS/METHODS We retrospectively reviewed the medical records of 123 MF patients treated with ruxolitinib between January 2012 and April 2020 at eight academic centers in Korea. RESULTS At ruxolitinib initiation, 38 patients (30.9%) underwent ≥ 2 units of RBC TF over 8 weeks. The most common reason for permanent discontinuation was intolerant anemia (10/63, 15.9%). The most common reasons for temporary interruption were nonhematologic toxicity (26/55, 21.1%), anemia (23/55, 18.7%) and thrombocytopenia (13/55, 10.6%). Among the 123 patients in the study, 57 (46.3%), 42 (34.1%), and 40 patients (32.5%) who were receiving or stopped ruxolitinib therapy had a status of RBC TF dependence, long-term RBC TF dependence, or severe RBC TF dependence, respectively. The presence of ≥ 2 units of RBC transfusion over 8 weeks at ruxolitinib initiation was an independent risk factor for persistent RBC TF dependence. CONCLUSION The requirement for RBC TF is commonly encountered during treatment of MF with ruxolitinib, particularly among those with pre-existing ≥ 2 units of RBC TF over 8 weeks. For those patients, overcoming the barrier of maintenance TF is demanding.
Collapse
Affiliation(s)
- Eun Hee Jung
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Junshik Hong
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea.
| | - Sung-Yong Kim
- Division of Hematology-Oncology, Department of Internal Medicine, Konkuk University Medical Center, Seoul, Republic of Korea
| | - Young Park
- Division of Hematology-Oncology, Department of Internal Medicine, Korea University Anam Hospital, Seoul, Republic of Korea
| | - Young Jin Yuh
- Division of Hematology-Oncology, Department of Internal Medicine, Sanggye Paik Hospital, Inje University College of Medicine, Seoul, Republic of Korea
| | - Yeung-Chul Mun
- Department of Hematology-Oncology, Ewha Womans University Mokdong Hospital, Seoul, Republic of Korea
| | - Won-Sik Lee
- Department of Internal Medicine, Busan Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | - Sung-Kyu Park
- Division of Hematology & Oncology, Department of Internal Medicine, Soonchunhyang University College of Medicine, Seoul, Republic of Korea
| | - Soo Mee Bang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| |
Collapse
|
111
|
Prakash S, Alhariri S, Hassan M, Patel PK, Corral J. Improvement in Primary Autoimmune Myelofibrosis Following a Short Course of Steroids and Intravenous Immunoglobulins. Cureus 2022; 14:e29735. [PMID: 36340546 PMCID: PMC9621728 DOI: 10.7759/cureus.29735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/28/2022] [Indexed: 11/24/2022] Open
Abstract
Bone marrow fibrosis (BMF) is a histopathological finding appreciated in a multitude of conditions such as myeloproliferative diseases and malignant neoplasms, along with autoimmune disorders. Autoimmune myelofibrosis (AIMF) is a particularly uncommon etiology of benign BMF. AIMF may be primary with serologic evidence of autoantibodies or secondary to an underlying autoimmune disease. The authors aim to emphasize the importance of distinguishing between primary versus secondary causes owing to significant prognostic and therapeutic discrepancies and in hopes of expediting the diagnostic journey. Research has recommended a treatment strategy of high-dose steroids followed by a steroid taper. However, our patient responded positively to a short course of high-dose steroids and intravenous immunoglobulins (IVIG) as evidenced by an improvement in cytopenias and bone marrow fibrosis grading. This outcome warrants further research on the necessity of steroid tapers in AIMF.
Collapse
|
112
|
Genomic Mutations of the STAT5 Transcription Factor Are Associated with Human Cancer and Immune Diseases. Int J Mol Sci 2022; 23:ijms231911297. [PMID: 36232600 PMCID: PMC9569778 DOI: 10.3390/ijms231911297] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 09/16/2022] [Accepted: 09/21/2022] [Indexed: 11/17/2022] Open
Abstract
Signal transducer and activation of transcription 5 (STAT5) is a key transcription factor that regulates various biological processes in mammalian development. Aberrant regulation of STAT5 has also been causally linked to many diseases, including cancers and immune-related diseases. Although persistent activation of STAT5 due to dysregulation of the signaling cascade has been reported to be associated with the progression of solid tumors and leukemia, various genomic mutations of STAT5 have also been found to cause a wide range of diseases. The present review comprehensively summarizes results of recent studies evaluating the intrinsic function of STAT5 and the link between STAT5 mutations and human diseases. This review also describes the types of disease models useful for investigating the mechanism underlying STAT5-driven disease progression. These findings provide basic knowledge for understanding the regulatory mechanisms of STAT5 and the progression of various diseases resulting from aberrant regulation of STAT5. Moreover, this review may provide insights needed to create optimal disease models that reflect human disease associated STAT5 mutations and to design gene therapies to correct STAT5 mutations.
Collapse
|
113
|
Guglielmelli P, Kiladjian JJ, Vannucchi AM, Duan M, Meng H, Pan L, He G, Verstovsek S, Boyer F, Barraco F, Niederwieser D, Pungolino E, Liberati AM, Harrison C, Roussou P, Wroclawska M, Karumanchi D, Sinclair K, Te Boekhorst PAW, Gisslinger H. Efficacy and safety of ruxolitinib in patients with myelofibrosis and low platelet count (50 × 10 9/L to <100 × 10 9/L) at baseline: the final analysis of EXPAND. Ther Adv Hematol 2022; 13:20406207221118429. [PMID: 36105914 PMCID: PMC9465569 DOI: 10.1177/20406207221118429] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 07/20/2022] [Indexed: 12/05/2022] Open
Abstract
Background: Thrombocytopenia is a common feature of myelofibrosis (MF), a
myeloproliferative neoplasm driven by dysregulated JAK/STAT signaling;
however, pivotal trials assessing the efficacy of ruxolitinib (a JAK1/2
inhibitor) excluded MF patients with low platelet counts
(<100 × 109/L). Objectives: Determination of the maximum safe starting dose (MSSD) of ruxolitinib was the
primary endpoint, with long-term safety and efficacy as secondary and
exploratory endpoints, respectively. Design: EXPAND (NCT01317875) was a phase 1b, open-label, ruxolitinib dose-finding
study in patients with MF and low platelet counts (50 to
<100 × 109/L). Methods: Patients were stratified according to baseline platelet count into stratum 1
(S1, 75 to <100 × 109/L) or stratum 2 (S2, 50 to
<75 × 109/L). Previous analyses established the MSSD at 10
mg twice daily (bid); long-term results are reported here. Results: Of 69 enrolled patients, 38 received ruxolitinib at the MSSD (S1,
n = 20; S2, n = 18) and are the focus
of this analysis. The incidence of adverse events was consistent with the
known safety profile of ruxolitinib, with thrombocytopenia (S1, 50%; S2,
78%) and anemia (S1, 55%; S2, 44%) the most frequently reported adverse
events and no new or unexpected safety signals. Substantial clinical
benefits were observed for patients in both strata: 50% (10/20) and 67%
(12/18) of patients in S1 and S2, respectively, achieved a spleen response
(defined as ⩾50% reduction in spleen length from baseline) at any time
during the study. Conclusion: The final safety and efficacy results from EXPAND support the use of a 10 mg
bid starting dose of ruxolitinib in patients with MF and platelet counts 50
to <100 × 109/L. Registration: ClinicalTrials.gov NCT01317875.
Collapse
Affiliation(s)
- Paola Guglielmelli
- CRIMM, Centro di Ricerca e Innovazione per le Malattie Mieloproliferative, AOU Careggi, Dipartimento di Medicina Sperimentale e Clinica, University of Florence, Viale Pieraccini 6, 50134 Firenze, Italy
| | - Jean-Jacques Kiladjian
- APHP, Hôpital Saint-Louis, Centre d'Investigations Cliniques, INSERM CIC 1427, Université de Paris, Paris, France
| | - Alessandro M Vannucchi
- CRIMM, Centro di Ricerca e Innovazione per le Malattie Mieloproliferative, AOU Careggi, Dipartimento di Medicina Sperimentale e Clinica, University of Florence, Florence, Italy
| | - Minghui Duan
- Peking Union Medical College Hospital, Beijing, China
| | - Haitao Meng
- Department of Hematology, The First Affiliated Hospital, College of Medicine, Institute of Hematology, Zhejiang University, Hangzhou, China
| | - Ling Pan
- West China Hospital, Sichuan University, Chengdu, China
| | - Guangsheng He
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Srdan Verstovsek
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | - Dietger Niederwieser
- Department of Hematology and Medical Oncology, University of Leipzig, Leipzig, Germany
| | - Ester Pungolino
- Division of Hematology, ASST Grande Ospedale Metropolitano Niguarda, Milano, Italy
| | - Anna Marina Liberati
- Azienda Ospedaliera Santa Maria di Terni, Università degli Studi di Perugia, Terni, Italy
| | | | | | | | | | | | | | | |
Collapse
|
114
|
Gerds AT, Gotlib J, Ali H, Bose P, Dunbar A, Elshoury A, George TI, Gundabolu K, Hexner E, Hobbs GS, Jain T, Jamieson C, Kaesberg PR, Kuykendall AT, Madanat Y, McMahon B, Mohan SR, Nadiminti KV, Oh S, Pardanani A, Podoltsev N, Rein L, Salit R, Stein BL, Talpaz M, Vachhani P, Wadleigh M, Wall S, Ward DC, Bergman MA, Hochstetler C. Myeloproliferative Neoplasms, Version 3.2022, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2022; 20:1033-1062. [PMID: 36075392 DOI: 10.6004/jnccn.2022.0046] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The classic Philadelphia chromosome-negative myeloproliferative neoplasms (MPN) consist of myelofibrosis, polycythemia vera, and essential thrombocythemia and are a heterogeneous group of clonal blood disorders characterized by an overproduction of blood cells. The NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines) for MPN were developed as a result of meetings convened by a multidisciplinary panel with expertise in MPN, with the goal of providing recommendations for the management of MPN in adults. The Guidelines include recommendations for the diagnostic workup, risk stratification, treatment, and supportive care strategies for the management of myelofibrosis, polycythemia vera, and essential thrombocythemia. Assessment of symptoms at baseline and monitoring of symptom status during the course of treatment is recommended for all patients. This article focuses on the recommendations as outlined in the NCCN Guidelines for the diagnosis of MPN and the risk stratification, management, and supportive care relevant to MF.
Collapse
Affiliation(s)
- Aaron T Gerds
- Case Comprehensive Cancer Center/University Hospitals Seidman Cancer Center and Cleveland Clinic Taussig Cancer Institute
| | | | - Haris Ali
- City of Hope National Medical Center
| | | | | | | | | | | | | | | | - Tania Jain
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins
| | | | | | | | | | | | | | | | - Stephen Oh
- Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine
| | | | | | | | - Rachel Salit
- Fred Hutchinson Cancer Research Center/Seattle Cancer Care Alliance
| | - Brady L Stein
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University
| | | | | | | | - Sarah Wall
- The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute
| | - Dawn C Ward
- UCLA Jonsson Comprehensive Cancer Center; and
| | | | | |
Collapse
|
115
|
Zhao Y, Wang D, Liang Y, Xu C, Shi L, Tong J. Expression profiles analysis identifies specific interferon-stimulated signatures as potential diagnostic and predictive indicators of JAK2V617F+ myelofibrosis. Front Genet 2022; 13:927018. [PMID: 36061178 PMCID: PMC9434717 DOI: 10.3389/fgene.2022.927018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 07/05/2022] [Indexed: 11/26/2022] Open
Abstract
Objective: This study aimed to identify specific dysregulated genes with potential diagnostic and predictive values for JAK2V617F+ myelofibrosis. Methods: Two gene expression datasets of CD34+ hematopoietic stem and progenitor cells (HSPCs) from patients with JAK2V617F+ myeloproliferative neoplasm (MPN) [n = 66, including polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF)] and healthy controls (HC) (n = 30) were acquired from the GEO (Gene Expression Omnibus) database. The differentially expressed genes (DEGs) were screened between each JAK2V617F+ MPN entity and HC. Subsequently, functional enrichment analyses, including Kyoto Encyclopedia of Genes and Genomes (KEGG), Reactome, and Gene Set Enrichment Analysis (GSEA), were conducted to decipher the important biological effects of DEGs. Protein–protein interaction (PPI) networks of the DEGs were constructed to identify hub genes and significant modules. Another two gene expression profiles of patients with JAK2V617F+ MPN [n = 23, including PV, ET, secondary myelofibrosis (SMF), and PMF] and HC (n = 6) from GEO were used as external validation datasets to prove the reliability of the identified signatures. Results: KEGG analysis revealed the upregulated genes in three JAK2V617F+ MPN entities compared with HC were essentially enriched in inflammatory pathways and immune response signaling pathways, and the number of these pathways enriched in PMF was obviously more than that in PV and ET. Following the PPI analysis, 10 genes primarily related to inflammation and immune response were found upregulated in different JAK2V617F+ MPN entities. In addition, Reactome enrichment analysis indicated that interferon signaling pathways were enriched specifically in PMF but not in PV or ET. Furthermore, several interferon (IFN)-stimulated genes were identified to be uniquely upregulated in JAK2V617F+ PMF. The external datasets validated the upregulation of four interferon-related genes (OAS1, IFITM3, GBP1, and GBP2) in JAK2V617F+ myelofibrosis. The receiver operating characteristic (ROC) curves indicate that the four genes have high area under the ROC curve (AUC) values when distinguishing JAK2V617F+ myelofibrosis from PV or ET. Conclusion: Four interferon-stimulated genes (OAS1, IFITM3, GBP1, and GBP2) exclusively upregulated in JAK2V617F+ myelofibrosis might have the potential to be the auxiliary molecular diagnostic and predictive indicators of myelofibrosis.
Collapse
|
116
|
Breccia M, Palandri F, Guglielmelli P, Palumbo GA, Malato A, Mendicino F, Ricco A, Sant’Antonio E, Tiribelli M, Iurlo A. Management of Myelofibrosis during Treatment with Ruxolitinib: A Real-World Perspective in Case of Resistance and/or Intolerance. Curr Oncol 2022; 29:4970-4980. [PMID: 35877255 PMCID: PMC9325304 DOI: 10.3390/curroncol29070395] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 07/04/2022] [Accepted: 07/12/2022] [Indexed: 11/16/2022] Open
Abstract
The development and approval of ruxolitinib, the first JAK1/2 inhibitor indicated to treat myelofibrosis, has improved patient outcomes, with higher spleen and symptoms responses, improved quality of life, and overall survival. Despite this, several unmet needs remain, including the absence of resistance criteria, suboptimal response, the timing of allogeneic transplant, and the management of patients in case of intolerance. Here, we report the results of the second survey led by the "MPN Lab" collaboration, which aimed to report physicians' perspectives on these topics. As in our first survey, physicians were selected throughout Italy, and we included those with extensive experience in treating myeloproliferative neoplasms and those with less experience representing clinical practice in the real world. The results presented here, summarized using descriptive analyses, highlight the need for a clear definition of response to ruxolitinib as well as recommendations to guide the management of ruxolitinib under specific conditions including anemia, thrombocytopenia, infections, and non-melanoma skin cancers.
Collapse
Affiliation(s)
- Massimo Breccia
- Hematology, Department of Precision and Translational Medicine, Policlinico Umberto 1, Sapienza University, 00161 Rome, Italy
| | - Francesca Palandri
- Istituto di Ematologia “Seràgnoli”, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Paola Guglielmelli
- Center of Research and Innovation of Myeloproliferative Neoplasms, AOU Careggi, University of Florence, 50134 Florence, Italy;
| | - Giuseppe Alberto Palumbo
- Dipartimento di Scienze Mediche, Chirurgiche e Tecnologie Avanzate “G.F. Ingrassia”, University of Catania, 95124 Catania, Italy;
| | - Alessandra Malato
- UOC di Oncoematologia Ospedali Riuniti Villa Sofia-Cervello Palermo, 90146 Palermo, Italy;
| | - Francesco Mendicino
- Hematology Unit, Department of Hemato-Oncology, Ospedale Annunziata, 87100 Cosenza, Italy;
| | - Alessandra Ricco
- Department of Emergency and Organ Transplantation (DETO), Hematology Section, University of Bari, 70121 Bari, Italy;
| | - Emanuela Sant’Antonio
- Department of Oncology, Division of Hematology, Azienda USL Toscana Nord Ovest, 55100 Lucca, Italy;
- Medical Genetics, University of Siena, 53100 Siena, Italy
| | - Mario Tiribelli
- Division of Hematology and Bone Marrow Transplantation, Department of Medical Area, University of Udine, 33100 Udine, Italy;
| | - Alessandra Iurlo
- Hematology Division, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| |
Collapse
|
117
|
Mascarenhas JO, Verstovsek S. The clinical dilemma of JAK inhibitor failure in myelofibrosis: Predictive characteristics and outcomes. Cancer 2022; 128:2717-2727. [PMID: 35385124 PMCID: PMC9324085 DOI: 10.1002/cncr.34222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/11/2022] [Accepted: 03/15/2022] [Indexed: 11/10/2022]
Abstract
Two Janus-associated kinase inhibitors (JAKi) (initially ruxolitinib and, more recently, fedratinib) have been approved as treatment options for patients who have intermediate-risk and high-risk myelofibrosis (MF), with pivotal trials demonstrating improvements in spleen volume, disease symptoms, and quality of life. At the same time, however, clinical trial experiences with JAKi agents in MF have demonstrated a high frequency of discontinuations because of adverse events or progressive disease. In addition, overall survival benefits and clinical and molecular predictors of response have not been established in this population, for which the disease burden is high and treatment options are limited. Consistently poor outcomes have been documented after JAKi discontinuation, with survival durations after ruxolitinib ranging from 11 to 16 months across several studies. To address such a high unmet therapeutic need, various non-JAKi agents are being actively explored (in combination with ruxolitinib in first-line or salvage settings and/or as monotherapy in JAKi-pretreated patients) in phase 3 clinical trials, including pelabresib (a bromodomain and extraterminal domain inhibitor), navitoclax (a B-cell lymphoma 2/B-cell lymphoma 2-xL inhibitor), parsaclisib (a phosphoinositide 3-kinase inhibitor), navtemadlin (formerly KRT-232; a murine double-minute chromosome 2 inhibitor), and imetelstat (a telomerase inhibitor). The breadth of data expected from these trials will provide insight into the ability of non-JAKi treatments to modify the natural history of MF.
Collapse
Affiliation(s)
- John O. Mascarenhas
- Tisch Cancer InstituteIcahn School of Medicine at Mount SinaiNew YorkNew York
| | - Srdan Verstovsek
- Leukemia DepartmentThe University of TexasMD Anderson Cancer CenterHoustonTexas
| |
Collapse
|
118
|
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
|
119
|
Pastor-Galán I, Martín I, Ferrer B, Hernández-Boluda JC. Impact of molecular profiling on the management of patients with myelofibrosis. Cancer Treat Rev 2022; 109:102435. [PMID: 35839532 DOI: 10.1016/j.ctrv.2022.102435] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/30/2022] [Accepted: 07/04/2022] [Indexed: 11/02/2022]
Abstract
Myelofibrosis (MF) is a chronic myeloproliferative neoplasm (MPN) characterized by a highly heterogeneous clinical course, which can be complicated by severe constitutional symptoms, massive splenomegaly, progressive bone marrow failure, cardiovascular events, and development of acute leukemia. Constitutive signaling through the JAK-STAT pathway plays a fundamental role in its pathogenesis, generally due to activating mutations of JAK2, CALR and MPL genes (i.e., the MPN driver mutations), present in most MF patients. Next Generation Sequencing (NGS) panel testing has shown that additional somatic mutations can already be detected at the time of diagnosis in more than half of patients, and that they accumulate along the disease course. These mutations, mostly affecting epigenetic modifiers or spliceosome components, may cooperate with MPN drivers to favor clonal dominance or influence the clinical phenotype, and some, such as high molecular risk mutations, correlate with a more aggressive clinical course with poor treatment response. The current main role of molecular profiling in clinical practice is prognostication, principally for selecting high-risk patients who may be candidates for transplantation, the only curative treatment for MF to date. To this end, contemporary prognostic models incorporating molecular data are useful tools to discriminate different risk categories. Aside from certain clinical situations, decisions regarding medical treatment are not based on patient molecular profiling, yet this approach may become more relevant in novel treatment strategies, such as the use of vaccines against the mutant forms of JAK2 or CALR, or drugs directed against actionable molecular targets.
Collapse
Affiliation(s)
| | - Iván Martín
- Hospital Clínico Universitario-INCLIVA, Valencia, Spain
| | - Blanca Ferrer
- Hospital Clínico Universitario-INCLIVA, Valencia, Spain
| | | |
Collapse
|
120
|
Ibarra J, Elbanna YA, Kurylowicz K, Ciboddo M, Greenbaum HS, Arellano NS, Rodriguez D, Evers M, Bock-Hughes A, Liu C, Smith Q, Lutze J, Baumeister J, Kalmer M, Olschok K, Nicholson B, Silva D, Maxwell L, Dowgielewicz J, Rumi E, Pietra D, Casetti IC, Catricala S, Koschmieder S, Gurbuxani S, Schneider RK, Oakes SA, Elf SE. Type I but Not Type II Calreticulin Mutations Activate the IRE1α/XBP1 Pathway of the Unfolded Protein Response to Drive Myeloproliferative Neoplasms. Blood Cancer Discov 2022; 3:298-315. [PMID: 35405004 PMCID: PMC9338758 DOI: 10.1158/2643-3230.bcd-21-0144] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 01/21/2022] [Accepted: 04/08/2022] [Indexed: 01/09/2023] Open
Abstract
Approximately 20% of patients with myeloproliferative neoplasms (MPN) harbor mutations in the gene calreticulin (CALR), with 80% of those mutations classified as either type I or type II. While type II CALR-mutant proteins retain many of the Ca2+ binding sites present in the wild-type protein, type I CALR-mutant proteins lose these residues. The functional consequences of this differential loss of Ca2+ binding sites remain unexplored. Here, we show that the loss of Ca2+ binding residues in the type I mutant CALR protein directly impairs its Ca2+ binding ability, which in turn leads to depleted endoplasmic reticulum (ER) Ca2+ and subsequent activation of the IRE1α/XBP1 pathway of the unfolded protein response. Genetic or pharmacologic inhibition of IRE1α/XBP1 signaling induces cell death in type I mutant but not type II mutant or wild-type CALR-expressing cells, and abrogates type I mutant CALR-driven MPN disease progression in vivo. SIGNIFICANCE Current targeted therapies for CALR-mutated MPNs are not curative and fail to differentiate between type I- versus type II-driven disease. To improve treatment strategies, it is critical to identify CALR mutation type-specific vulnerabilities. Here we show that IRE1α/XBP1 represents a unique, targetable dependency specific to type I CALR-mutated MPNs. This article is highlighted in the In This Issue feature, p. 265.
Collapse
Affiliation(s)
- Juan Ibarra
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
- Committee on Cancer Biology, University of Chicago, Chicago, Illinois
| | - Yassmin A. Elbanna
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
| | - Katarzyna Kurylowicz
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
| | - Michele Ciboddo
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
| | - Harrison S. Greenbaum
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
| | - Nicole S. Arellano
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
| | - Deborah Rodriguez
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
| | - Maria Evers
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
- Committee on Cancer Biology, University of Chicago, Chicago, Illinois
| | - Althea Bock-Hughes
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
- Committee on Molecular Metabolism and Nutrition, University of Chicago, Chicago, Illinois
| | - Chenyu Liu
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
| | - Quinn Smith
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
| | - Julian Lutze
- Committee on Cancer Biology, University of Chicago, Chicago, Illinois
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, Illinois
| | - Julian Baumeister
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University and Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Milena Kalmer
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University and Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Kathrin Olschok
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University and Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Benjamin Nicholson
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
- Committee on Cancer Biology, University of Chicago, Chicago, Illinois
| | - Diane Silva
- Department of Pathology, University of Chicago, Chicago, Illinois
| | - Luke Maxwell
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
| | - Jonathan Dowgielewicz
- Committee on Molecular Metabolism and Nutrition, University of Chicago, Chicago, Illinois
| | - Elisa Rumi
- Department of Hematology Oncology, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Daniela Pietra
- Department of Hematology Oncology, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
| | | | - Silvia Catricala
- Department of Hematology Oncology, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
| | - Steffen Koschmieder
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University and Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | | | - Rebekka K. Schneider
- Department of Cell Biology, Institute for Biomedical Technologies, RWTH Aachen University, Aachen, Germany
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Scott A. Oakes
- Committee on Cancer Biology, University of Chicago, Chicago, Illinois
- Department of Pathology, University of Chicago, Chicago, Illinois
| | - Shannon E. Elf
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
- Committee on Cancer Biology, University of Chicago, Chicago, Illinois
| |
Collapse
|
121
|
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
|
122
|
Lysine-Specific Demethylase 1 (LSD1/KDM1A) Inhibition as a Target for Disease Modification in Myelofibrosis. Cells 2022; 11:cells11132107. [PMID: 35805191 PMCID: PMC9265913 DOI: 10.3390/cells11132107] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 06/28/2022] [Accepted: 07/02/2022] [Indexed: 02/04/2023] Open
Abstract
Myelofibrosis (MF) is the most symptomatic form of myeloproliferative neoplasm and carries the worst outcome. Allogeneic hematopoietic stem cell transplantation is the only therapy with potential for cure at present, but is limited by significant mortality and morbidity. JAK inhibition is the mainstay of treatment for intermediate- and high-risk MF. Ruxolitinib is the most widely used JAK1/2 inhibitor and provides durable effects in controlling symptom burden and spleen volumes. Nevertheless, ruxolitinib may not adequately address the underlying disease biology. Its effects on mutant allele burden, bone marrow fibrosis, and the prevention of leukemic transformation are minimal. Multiple small molecules are being tested in multiple phase 2 and 3 studies as either monotherapy or in combination with JAK2 inhibitors. In this review, the role of LSD1/KDM1A inhibition as a potential disease-modification strategy in patients with myelofibrosis is described and discussed.
Collapse
|
123
|
Duek A, Berla M, Ellis MH. Recent advances in the treatment of polycythemia vera. Leuk Lymphoma 2022; 63:1801-1809. [DOI: 10.1080/10428194.2022.2057491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Adrian Duek
- Hematology Institute Assuta Medical Center, Ashdod, Israel
| | - Maya Berla
- Hematology Institute Meir Medical Center, Kfar Saba, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Martin H. Ellis
- Hematology Institute Meir Medical Center, Kfar Saba, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
124
|
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: 0.7] [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
|
125
|
Zeidan AM, Cook RJ, Bordoni R, Berenson JR, Edenfield WJ, Mohan S, Zhou G, Asatiani E, Srinivas N, Savona MR. A Phase 1/2 Study of the Oral Janus Kinase 1 Inhibitors INCB052793 and Itacitinib Alone or in Combination With Standard Therapies for Advanced Hematologic Malignancies. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2022; 22:523-534. [PMID: 35260349 DOI: 10.1016/j.clml.2022.01.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 01/07/2022] [Accepted: 01/16/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND The Janus kinase (JAK)/signal transducers and activators of transcription pathway has been implicated in the pathogenesis and progression of various hematologic malignancies. JAK1-regulated cytokines stimulate proliferation and growth of malignant cells and resistance to certain therapies. PATIENTS AND METHODS This phase 1/2 study evaluated 2 oral, novel JAK1 inhibitors (INCB052793 and itacitinib) in advanced hematologic malignancies. Phase 1a assessed dose escalation and expansion of INCB052793 monotherapy. Phase 1b evaluated INCB052793 plus standard therapy in relapsed/refractory multiple myeloma, acute myeloid leukemia (AML), or myelodysplastic syndrome (MDS). Phase 2 evaluated INCB052793 or itacitinib plus azacitidine in DNA methyltransferase inhibitor (DNMTi)-refractory AML or MDS. Primary endpoints included safety and tolerability for phase 1, and objective response rate for phase 2. RESULTS Fifty-eight patients were enrolled, all received study treatment and discontinued either treatment or participation in the study. The most common reasons for treatment discontinuation were progressive disease (35.4% and 50.0%) and adverse events (22.9% and 20.0%) for INCB052793 and itacitinib plus azacitidine, respectively. In phase 1, 12 of 39 patients (31%) achieved an objective response; 35 mg once daily was selected as the phase 2 dose. Two patients with DNMTi-refractory disease had an objective response in phase 2. The study was terminated for lack of efficacy. CONCLUSION Inhibition of JAK1 with INCB052793 (monotherapy or combination therapy) or itacitinib plus azacitidine did not demonstrate clinically meaningful responses in these patients with hematopoietic malignancies.
Collapse
Affiliation(s)
- Amer M Zeidan
- Section of Hematology, Department of Internal Medicine, Yale University and Yale Cancer Center, New Haven, CT
| | | | | | | | | | - Sanjay Mohan
- Department of Internal Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN
| | | | | | | | - Michael R Savona
- Department of Internal Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN.
| |
Collapse
|
126
|
Baek DW, Cho HJ, Lee JM, Kim J, Moon JH, Sohn SK. Light and shade of ruxolitinib: positive role of early treatment with ruxolitinib and ruxolitinib withdrawal syndrome in patients with myelofibrosis. Expert Rev Hematol 2022; 15:573-581. [PMID: 35679520 DOI: 10.1080/17474086.2022.2088499] [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 Myelofibrosis (MF) is characterized by ineffective and hepatosplenic extramedullary hematopoiesis due to fibrotic changes in the bone marrow and systemic manifestations due to aberrant cytokine release. Ruxolitinib (RUX) is the first JAK1/JAK2 inhibitor that is clinically approved to treat splenomegaly by ameliorating inflammatory cytokines and myeloproliferation in MF. AREAS COVERED Patients with less advanced MF may also achieve better outcome and successful treatment with RUX. However, approximately 40% of the patients failed to achieve a stable response or have shown to be intolerant to RUX, and most of them discontinued RUX. In patients who need to discontinue or reduce the dose of RUX for any reason, RUX is known to induce a paradoxical accumulation of JAK activation loop phosphorylation that is causing RUX discontinuation syndrome (RDS). To review the topic of MF and RUX, we searched relevant literatures using PubMed. EXPERT OPINION RUX treatment in lower IPSS risk patients who present with splenomegaly and disease-associated symptoms can be helpful. A careful discontinuation strategy with steroids may reduce the probability of RDS, and the recognition of RDS with early re-introduction of RUX is important in the treatment of severe cases of RDS.
Collapse
Affiliation(s)
- Dong Won Baek
- Department of Hematology/Oncology, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Hee Jeong Cho
- Department of Hematology/Oncology, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Jung Min Lee
- Department of Hematology/Oncology, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Juhyung Kim
- Department of Hematology/Oncology, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Joon Ho Moon
- Department of Hematology/Oncology, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Sang Kyun Sohn
- Department of Hematology/Oncology, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, Daegu, South Korea
| |
Collapse
|
127
|
Gangat N, McCullough K, Al-Kali A, Begna KH, Patnaik MM, Litzow MR, Hogan W, Shah M, Alkhateeb H, Mangaonkar A, Foran JM, Palmer JM, Pardanani A, Tefferi A. Limited activity of fedratinib in myelofibrosis patients relapsed/refractory to ruxolitinib 20 mg twice daily or higher: A real-world experience. Br J Haematol 2022; 198:e54-e58. [PMID: 35614565 DOI: 10.1111/bjh.18284] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 05/09/2022] [Accepted: 05/15/2022] [Indexed: 11/30/2022]
Affiliation(s)
- Naseema Gangat
- Division of Hematology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Aref Al-Kali
- Division of Hematology, Mayo Clinic, Rochester, Minnesota, USA
| | - Kebede H Begna
- Division of Hematology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Mark R Litzow
- Division of Hematology, Mayo Clinic, Rochester, Minnesota, USA
| | - William Hogan
- Division of Hematology, Mayo Clinic, Rochester, Minnesota, USA
| | - Mithun Shah
- Division of Hematology, Mayo Clinic, Rochester, Minnesota, USA
| | | | | | - James M Foran
- Division of Hematology, Mayo Clinic, Jacksonville, Florida, USA
| | - Jeanne M Palmer
- Division of Hematology, Mayo Clinic, Scottsdale, Arizona, USA
| | | | - Ayalew Tefferi
- Division of Hematology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
128
|
Harrison CN, Garcia JS, Somervaille TC, Foran JM, Verstovsek S, Jamieson C, Mesa R, Ritchie EK, Tantravahi SK, Vachhani P, O'Connell CL, Komrokji RS, Harb J, Hutti JE, Holes L, Masud AA, Nuthalapati S, Potluri J, Pemmaraju N. Addition of Navitoclax to Ongoing Ruxolitinib Therapy for Patients With Myelofibrosis With Progression or Suboptimal Response: Phase II Safety and Efficacy. J Clin Oncol 2022; 40:1671-1680. [PMID: 35180010 PMCID: PMC9113204 DOI: 10.1200/jco.21.02188] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 12/21/2021] [Accepted: 01/13/2022] [Indexed: 12/17/2022] Open
Abstract
PURPOSE Targeting the BCL-XL pathway has demonstrated the ability to overcome Janus kinase inhibitor resistance in preclinical models. This phase II trial investigated the efficacy and safety of adding BCL-XL/BCL-2 inhibitor navitoclax to ruxolitinib therapy in patients with myelofibrosis with progression or suboptimal response to ruxolitinib monotherapy (ClinicalTrials.gov identifier: NCT03222609). METHODS Thirty-four adult patients with intermediate-/high-risk myelofibrosis who had progression or suboptimal response on stable ruxolitinib dose (≥ 10 mg twice daily) were administered navitoclax at 50 mg once daily starting dose, followed by escalation to a maximum of 300 mg once daily in once in weekly increments (if platelets were ≥ 75 × 109/L). The primary end point was ≥ 35% spleen volume reduction (SVR35) from baseline at week 24. Secondary end points included ≥ 50% reduction in total symptom score (TSS50) from baseline at week 24, hemoglobin improvement, change in bone marrow fibrosis (BMF) grade, and safety. RESULTS High molecular risk mutations were identified in 58% of patients, and 52% harbored ≥ 3 mutations. SVR35 was achieved by 26.5% of patients at week 24, and by 41%, at any time on study, with an estimated median duration of SVR35 of 13.8 months. TSS50 was achieved by 30% (6 of 20) of patients at week 24, and BMF improved by 1-2 grades in 33% (11 of 33) of evaluable patients. Anemia response was achieved by 64% (7 of 11), including one patient with baseline transfusion dependence. Median overall survival was not reached with a median follow-up of 21.6 months. The most common adverse event was reversible thrombocytopenia without clinically significant bleeding (88%). CONCLUSION The addition of navitoclax to ruxolitinib in patients with persistent or progressive myelofibrosis resulted in durable SVR35, improved TSS, hemoglobin response, and BMF. Further investigation is underway to qualify the potential for disease modification.
Collapse
Affiliation(s)
| | | | - Tim C.P. Somervaille
- The Christie NHS Foundation Trust, Manchester, United Kingdom
- Cancer Research UK Manchester Institute, The University of Manchester, Manchester, United Kingdom
| | | | | | - Catriona Jamieson
- University of California San Diego Moores Cancer Center, La Jolla, CA
| | - Ruben Mesa
- University of Texas Health San Antonio, San Antonio, TX
| | - Ellen K. Ritchie
- Division of Hematology and Oncology, Weill Cornell Medical College, New York, NY
| | | | | | - Casey L. O'Connell
- University of Southern California Keck School of Medicine, Los Angeles, CA
| | | | | | | | | | | | | | | | | |
Collapse
|
129
|
Maffioli M. Making a case for disease-modifying agents in myelofibrosis. Lancet Haematol 2022; 9:e391-e392. [DOI: 10.1016/s2352-3026(22)00140-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/04/2022] [Accepted: 05/06/2022] [Indexed: 11/28/2022]
|
130
|
Cheung V, England J, Maze D, Sibai H. Osteolytic lesion in polycythemia vera: First report and review of literature. EJHAEM 2022; 3:526-532. [PMID: 35846060 PMCID: PMC9176128 DOI: 10.1002/jha2.420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 03/06/2022] [Accepted: 03/09/2022] [Indexed: 11/10/2022]
Affiliation(s)
- Verna Cheung
- Princess Margaret Cancer CentreUniversity of Health NetworkTorontoCanada
- Faculty of NursingUniversity of TorontoTorontoOntarioCanada
| | - James England
- Princess Margaret Cancer CentreUniversity of Health NetworkTorontoCanada
- Department of MedicineUniversity of TorontoTorontoOntarioCanada
| | - Dawn Maze
- Princess Margaret Cancer CentreUniversity of Health NetworkTorontoCanada
- Department of MedicineUniversity of TorontoTorontoOntarioCanada
| | - Hassan Sibai
- Princess Margaret Cancer CentreUniversity of Health NetworkTorontoCanada
- Department of MedicineUniversity of TorontoTorontoOntarioCanada
| |
Collapse
|
131
|
Wang F, Qiu T, Wang H, Yang Q. State-of-the-Art Review on Myelofibrosis Therapies. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2022; 22:e350-e362. [PMID: 34903489 DOI: 10.1016/j.clml.2021.11.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/08/2021] [Accepted: 11/11/2021] [Indexed: 06/14/2023]
Abstract
Myelofibrosis (MF) is a BCR-ABL1-negative myeloproliferative neoplasm characterized by anemia, extramedullary hematopoiesis, bone marrow fibrosis, splenomegaly, constitutional symptoms and acute myeloid leukemia progression. Currently, allogeneic haematopoietic stem cell transplantation (AHSCT) therapy is the only curative option for MF patients. However, AHSCT is strictly limited due to the high rates of morbidity and mortality. Janus kinase 2 (JAK2) inhibitor Ruxolitinib is the first-line treatment for intermediate-II or high-risk MF patients with splenomegaly and constitutional symptoms, but most MF patients develop resistance or intolerance to Ruxolitinib. Therefore, MF treatment is a challenge for the medical community. This review summarizes 3 investigated directions for MF therapy: monotherapies of JAK inhibitors, monotherapies of non-JAK targeted agents, combination therapies of Ruxolitinib and other agents. We emphasize combination of Ruxolitinib and other agents is a promising strategy.
Collapse
Affiliation(s)
- Fuping Wang
- Beijing Key Laboratory of Resistant Gene Resources and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Tian Qiu
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haitao Wang
- Department of Hematology, Fourth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Qiong Yang
- Beijing Key Laboratory of Resistant Gene Resources and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, China.
| |
Collapse
|
132
|
Pemmaraju N, Garcia JS, Potluri J, Harb JG, Sun Y, Jung P, Qin QQ, Tantravahi SK, Verstovsek S, Harrison C. Addition of navitoclax to ongoing ruxolitinib treatment in patients with myelofibrosis (REFINE): a post-hoc analysis of molecular biomarkers in a phase 2 study. THE LANCET HAEMATOLOGY 2022; 9:e434-e444. [DOI: 10.1016/s2352-3026(22)00116-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 04/01/2022] [Accepted: 04/06/2022] [Indexed: 12/20/2022]
|
133
|
Ogai A, Yagi K, Ito F, Domoto H, Shiomi T, Chin K. Fatal Disseminated Tuberculosis and Concurrent Disseminated Cryptococcosis in a Ruxolitinib-treated Patient with Primary Myelofibrosis: A Case Report and Literature Review. Intern Med 2022; 61:1271-1278. [PMID: 34565769 PMCID: PMC9107979 DOI: 10.2169/internalmedicine.6436-20] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Ruxolitinib, a Janus kinase inhibitor, improves symptoms in patients with myelofibrosis. However, its association with the development of opportunistic infections has been a concern. We herein report a 71-year-old man with primary myelofibrosis who developed disseminated tuberculosis and concurrent disseminated cryptococcosis during ruxolitinib treatment. We also reviewed the literature on disseminated tuberculosis and/or cryptococcosis associated with ruxolitinib treatment. This is the first case of disseminated tuberculosis and concurrent disseminated cryptococcosis during treatment with ruxolitinib. We therefore suggest considering not only disseminated tuberculosis but also cryptococcosis in the differential diagnosis of patients with abnormal pulmonary shadows during ruxolitinib treatment.
Collapse
Affiliation(s)
- Asuka Ogai
- Department of Hematology, Department of Medicine, Keiyu Hospital, Japan
| | - Kazuma Yagi
- Department of Pulmonary Medicine, Department of Medicine, Keiyu Hospital, Japan
| | - Fumimaro Ito
- Department of Pulmonary Medicine, Department of Medicine, Keiyu Hospital, Japan
| | | | - Tetsuya Shiomi
- Department of Pulmonary Medicine, Department of Medicine, Keiyu Hospital, Japan
| | - Kenko Chin
- Department of Hematology, Department of Medicine, Keiyu Hospital, Japan
| |
Collapse
|
134
|
Tan YX, Luo J, Huang JX, Luo DM, Liang HY, Zhou X, Liu XL, Xu N. [Analysis of the effect of gene mutations on the efficacy of ruxolitinib in patients with myelofibrosis based on second-generation sequencing technology]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2022; 43:323-329. [PMID: 35680632 PMCID: PMC9189480 DOI: 10.3760/cma.j.issn.0253-2727.2022.04.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Indexed: 12/01/2022]
Abstract
Objective: To assess the effect of gene mutations on the efficacy of ruxolitinib for treating myelofibrosis (MF) . Methods: We retrospectively analyzed the clinical data of 56 patients with MF treated with ruxolitinib from July 2017 to December 2020 and applied second-generation sequencing (NGS) technology to detect 127 hematologic tumor-related gene mutations. Additionally, we analyzed the relationship between mutated genes and the efficacy of ruxolitinib. Results: ①Among the 56 patients, there were 36 cases of primary bone marrow fibrosis (PMF) , 9 cases of bone marrow fibrosis (ppv-mf) after polycythemia vera, and 11 cases of bone marrow fibrosis (PET-MF) after primary thrombocytosis (ET) . ②Fifty-six patients with MF taking ruxolitinib underwent NGS, among whom, 50 (89.29%) carried driver mutations, 22 (39.29%) carried ≥3 mutations, and 29 (51.79%) carried high-risk mutations (HMR) . ③ For patients with MF carrying ≥ 3 mutations, ruxolitinib still had a better effect of improving somatic symptoms and shrinking the spleen (P=0.001, P<0.001) , but TTF and PFS were significantly shorter in patients carrying ≥ 3 mutations (P=0.007, P=0.042) . ④For patients carrying ≥ 2 HMR mutations, ruxolitinib was less effective in shrinking the spleen than in those who did not carry HMR (t= 10.471, P=0.034) , and the TTF and PFS were significantly shorter in patients carrying ≥2 HMR mutations (P<0.001, P=0.001) . ⑤Ruxolitinib had poorer effects on spleen reduction, symptom improvement, and stabilization of myelofibrosis in patients carrying additional mutations in ASXL1, EZH2, and SRSF2. Moreover, patients carrying ASXL1 and EZH2 mutations had significantly shorter TTF [ASXL1: 360 (55-1270) d vs 440 (55-1268) d, z=-3.115, P=0.002; EZH2: 327 (55-975) d vs 404 (50-1270) d, z=-3.219, P=0.001], and significantly shorter PFS compared to non-carriers [ASXL1: 457 (50-1331) d vs 574 (55-1437) d, z=-3.219, P=0.001) ; 428 (55-1331) d vs 505 (55-1437) d, z=-2.576, P=0.008]. Conclusion: The type and number of mutations carried by patients with myelofibrosis and HMR impact the efficacy of ruxolitinib.
Collapse
Affiliation(s)
- Y X Tan
- Department of Hematology, Nan fang Hospital, Southern Medical University, Guangzhou 510515, China
| | - J Luo
- Department of Hematology, Nan fang Hospital, Southern Medical University, Guangzhou 510515, China
| | - J X Huang
- Department of Hematology, Yuebei People's Hospital, Shaoguan 512025, China
| | - D M Luo
- Department of Hematology, Nan fang Hospital, Southern Medical University, Guangzhou 510515, China
| | - H Y Liang
- Department of Hematology, Nan fang Hospital, Southern Medical University, Guangzhou 510515, China
| | - X Zhou
- Department of Hematology, Nan fang Hospital, Southern Medical University, Guangzhou 510515, China
| | - X L Liu
- Department of Hematology, Nan fang Hospital, Southern Medical University, Guangzhou 510515, China
| | - N Xu
- Department of Hematology, Nan fang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
135
|
Philadelphia chromosome-negative myeloproliferative neoplasms: clinical aspects and treatment options. Int J Hematol 2022; 115:616-618. [DOI: 10.1007/s12185-022-03344-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 03/30/2022] [Accepted: 03/30/2022] [Indexed: 10/18/2022]
|
136
|
Tvorogov D, Thompson‐Peach CAL, Foßelteder J, Dottore M, Stomski F, Onnesha SA, Lim K, Moretti PAB, Pitson SM, Ross DM, Reinisch A, Thomas D, Lopez AF. Targeting human CALR-mutated MPN progenitors with a neoepitope-directed monoclonal antibody. EMBO Rep 2022; 23:e52904. [PMID: 35156745 PMCID: PMC8982588 DOI: 10.15252/embr.202152904] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 01/23/2022] [Accepted: 01/26/2022] [Indexed: 01/02/2023] Open
Abstract
Calreticulin (CALR) is recurrently mutated in myelofibrosis via a frameshift that removes an endoplasmic reticulum retention signal, creating a neoepitope potentially targetable by immunotherapeutic approaches. We developed a specific rat monoclonal IgG2α antibody, 4D7, directed against the common sequence encoded by both insertion and deletion mutations. 4D7 selectively bound to cells co-expressing mutant CALR and thrombopoietin receptor (TpoR) and blocked JAK-STAT signalling, TPO-independent proliferation and megakaryocyte differentiation of mutant CALR myelofibrosis progenitors by disrupting the binding of CALR dimers to TpoR. Importantly, 4D7 inhibited proliferation of patient samples with both insertion and deletion CALR mutations but not JAK2 V617F and prolonged survival in xenografted bone marrow models of mutant CALR-dependent myeloproliferation. Together, our data demonstrate a novel therapeutic approach to target a problematic disease driven by a recurrent somatic mutation that would normally be considered undruggable.
Collapse
Affiliation(s)
- Denis Tvorogov
- Centre for Cancer BiologySA Pathology and University of South AustraliaAdelaideSAAustralia
| | - Chloe A L Thompson‐Peach
- Cancer ProgramPrecision Medicine ThemeSouth Australian Health and Medical Research Institute (SAHMRI)University of AdelaideAdelaideSAAustralia
- Discipline of MedicineAdelaide Medical SchoolThe University of AdelaideAdelaideSAAustralia
| | - Johannes Foßelteder
- Department of Internal MedicineDivision of HaematologyMedical University of GrazGrazAustria
| | - Mara Dottore
- Centre for Cancer BiologySA Pathology and University of South AustraliaAdelaideSAAustralia
| | - Frank Stomski
- Centre for Cancer BiologySA Pathology and University of South AustraliaAdelaideSAAustralia
| | - Suraiya A Onnesha
- Cancer ProgramPrecision Medicine ThemeSouth Australian Health and Medical Research Institute (SAHMRI)University of AdelaideAdelaideSAAustralia
- Discipline of MedicineAdelaide Medical SchoolThe University of AdelaideAdelaideSAAustralia
| | - Kelly Lim
- Cancer ProgramPrecision Medicine ThemeSouth Australian Health and Medical Research Institute (SAHMRI)University of AdelaideAdelaideSAAustralia
- Discipline of MedicineAdelaide Medical SchoolThe University of AdelaideAdelaideSAAustralia
| | - Paul A B Moretti
- Centre for Cancer BiologySA Pathology and University of South AustraliaAdelaideSAAustralia
| | - Stuart M Pitson
- Centre for Cancer BiologySA Pathology and University of South AustraliaAdelaideSAAustralia
- Discipline of MedicineAdelaide Medical SchoolThe University of AdelaideAdelaideSAAustralia
| | - David M Ross
- Centre for Cancer BiologySA Pathology and University of South AustraliaAdelaideSAAustralia
- Cancer ProgramPrecision Medicine ThemeSouth Australian Health and Medical Research Institute (SAHMRI)University of AdelaideAdelaideSAAustralia
- Department of HaematologyFlinders University and Medical CentreAdelaideSAAustralia
| | - Andreas Reinisch
- Department of Internal MedicineDivision of HaematologyMedical University of GrazGrazAustria
- Department of Blood Group Serology and Transfusion MedicineMedical University of GrazGrazAustria
| | - Daniel Thomas
- Cancer ProgramPrecision Medicine ThemeSouth Australian Health and Medical Research Institute (SAHMRI)University of AdelaideAdelaideSAAustralia
- Discipline of MedicineAdelaide Medical SchoolThe University of AdelaideAdelaideSAAustralia
| | - Angel F Lopez
- Centre for Cancer BiologySA Pathology and University of South AustraliaAdelaideSAAustralia
- Discipline of MedicineAdelaide Medical SchoolThe University of AdelaideAdelaideSAAustralia
| |
Collapse
|
137
|
The Use of Allogeneic Hematopoietic Stem Cell Transplantation in Primary Myelofibrosis. J Pers Med 2022; 12:jpm12040571. [PMID: 35455686 PMCID: PMC9025208 DOI: 10.3390/jpm12040571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/04/2022] [Accepted: 03/14/2022] [Indexed: 01/27/2023] Open
Abstract
Primary myelofibrosis (PMF) is a BCR-ABL1 negative myeloproliferative neoplasm characterized by clonal proliferation of myeloid cells. This leads to reactive bone marrow fibrosis, ultimately resulting in progressive marrow failure, hepatosplenomegaly, and extramedullary hematopoiesis. PMF is considered the most aggressive of the BCR-ABL1 negative myeloproliferative neoplasms with the least favorable prognosis. Constitutional symptoms are common, which can impact an individual’s quality of life and leukemic transformation remains an important cause of death in PMF patients. The development of the Janus kinase 2 (JAK2) inhibitors have provided a good option for management of PMF-related symptoms. Unfortunately, these agents have not been shown to improve overall survival or significantly alter the course of disease. Allogenic hematopoietic stem cell transplantation (allo-HSCT) remains the only curative treatment option in PMF. However, allo-HSCT is associated with significant treatment-related morbidity and mortality and has historically been reserved for younger, high-risk patients. This review examines patient, disease, and transplant-specific factors which may impact transplant-related outcomes in PMF. Through the vast improvements in donor selection, conditioning regimens, and post-transplant care, allo-HSCT may provide a safe and effective curative option for a broader range of PMF patients in the future.
Collapse
|
138
|
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
|
139
|
Loscocco GG, Vannucchi AM. Role of JAK inhibitors in myeloproliferative neoplasms: current point of view and perspectives. Int J Hematol 2022; 115:626-644. [PMID: 35352288 DOI: 10.1007/s12185-022-03335-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 03/06/2022] [Accepted: 03/15/2022] [Indexed: 12/29/2022]
Abstract
Classic Philadelphia-negative myeloproliferative neoplasms (MPN) include polycythemia vera (PV), essential thrombocythemia (ET), and myelofibrosis (MF), classified as primary (PMF), or secondary to PV or ET. All MPN, regardless of the underlying driver mutation in JAK2/CALR/MPL, are invariably associated with dysregulation of JAK/STAT pathway. The discovery of JAK2V617F point mutation prompted the development of small molecules inhibitors of JAK tyrosine kinases (JAK inhibitors-JAKi). To date, among JAKi, ruxolitinib (RUX) and fedratinib (FEDR) are approved for intermediate and high-risk MF, and RUX is also an option for high-risk PV patients inadequately controlled by or intolerant to hydroxyurea. While not yet registered, pacritinib (PAC) and momelotinib (MMB), proved to be effective particularly in thrombocytopenic and anemic MF patients, respectively. In most cases, JAKi are effective in reducing splenomegaly and alleviating disease-related symptoms. However, almost 50% lose response by three years and dose-dependent toxicities may lead to suboptimal dosing or treatment discontinuation. To date, although not being disease-modifying agents, JAKi represent the therapeutic backbone particularly in MF patient. To optimize therapeutic strategies, many trials with drug combinations of JAKi with novel molecules are ongoing. This review critically discusses the role of JAKi in the modern management of patients with MPN.
Collapse
Affiliation(s)
- Giuseppe G Loscocco
- Department of Experimental and Clinical Medicine, University of Florence, CRIMM, Center of Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliero-Universitaria Careggi, Largo Brambilla, 3 pad 27B, 50134, Florence, Italy
- Doctorate School GenOMec, University of Siena, Siena, Italy
| | - Alessandro M Vannucchi
- Department of Experimental and Clinical Medicine, University of Florence, CRIMM, Center of Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliero-Universitaria Careggi, Largo Brambilla, 3 pad 27B, 50134, Florence, Italy.
| |
Collapse
|
140
|
Analysis of factors associated with the development of myelofibrosis in polycythemia vera and essential thrombocythemia patients: a single-center experience. J Hematop 2022. [DOI: 10.1007/s12308-022-00488-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
141
|
Verachi P, Gobbo F, Martelli F, Martinelli A, Sarli G, Dunbar A, Levine RL, Hoffman R, Massucci MT, Brandolini L, Giorgio C, Allegretti M, Migliaccio AR. The CXCR1/CXCR2 Inhibitor Reparixin Alters the Development of Myelofibrosis in the Gata1 low Mice. Front Oncol 2022; 12:853484. [PMID: 35392239 PMCID: PMC8982152 DOI: 10.3389/fonc.2022.853484] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 02/16/2022] [Indexed: 11/13/2022] Open
Abstract
A major role for human (h)CXCL8 (interleukin-8) in the pathobiology of myelofibrosis (MF) has been suggested by observations indicating that MF megakaryocytes express increased levels of hCXCL8 and that plasma levels of this cytokine in MF patients are predictive of poor patient outcomes. Here, we demonstrate that, in addition to high levels of TGF-β, the megakaryocytes from the bone marrow of the Gata1 low mouse model of myelofibrosis express high levels of murine (m)CXCL1, the murine equivalent of hCXCL8, and its receptors CXCR1 and CXCR2. Treatment with the CXCR1/R2 inhibitor, Reparixin in aged-matched Gata1 low mice demonstrated reductions in bone marrow and splenic fibrosis. Of note, the levels of fibrosis detected using two independent methods (Gomori and reticulin staining) were inversely correlated with plasma levels of Reparixin. Immunostaining of marrow sections indicated that the bone marrow from the Reparixin-treated group expressed lower levels of TGF-β1 than those expressed by the bone marrow from vehicle-treated mice while the levels of mCXCL1, and expression of CXCR1 and CXCR2, were similar to that of vehicle-treated mice. Moreover, immunofluorescence analyses performed on bone marrow sections from Gata1 low mice indicated that treatment with Reparixin induced expression of GATA1 while reducing expression of collagen III in megakaryocytes. These data suggest that in Gata1low mice, Reparixin reduces fibrosis by reducing TGF-β1 and collagen III expression while increasing GATA1 in megakaryocytes. Our results provide a preclinical rationale for further evaluation of this drug alone and in combination with current JAK inhibitor therapy for the treatment of patients with myelofibrosis.
Collapse
Affiliation(s)
- Paola Verachi
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum University, Bologna, Italy
| | - Francesca Gobbo
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum University, Bologna, Italy
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Fabrizio Martelli
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Andrea Martinelli
- Center for Animal Experimentation and Well-Being, Istituto Superiore di Santà, Rome, Italy
| | - Giuseppe Sarli
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Andrew Dunbar
- Human Oncology & Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Leukemia Service, Department of Medicine and Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Ross L. Levine
- Human Oncology & Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Leukemia Service, Department of Medicine and Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Ronald Hoffman
- Division of Hematology/Oncology, Tisch Cancer Institute and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | | | | | | | | | - Anna Rita Migliaccio
- Center for Integrated Biomedical Research, Campus Bio-medico, Rome, Italy
- Altius Institute for Biomedical Sciences, Seattle, WA, United States
| |
Collapse
|
142
|
Verstovsek S, Yu J, Scherber RM, Verma S, Dieyi C, Chen CC, Parasuraman S. Changes in the incidence and overall survival of patients with myeloproliferative neoplasms between 2002 and 2016 in the United States. Leuk Lymphoma 2022; 63:694-702. [PMID: 34689695 DOI: 10.1080/10428194.2021.1992756] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 10/05/2021] [Accepted: 10/09/2021] [Indexed: 01/08/2023]
Abstract
This analysis examined trends in incidence and survival among US adults with myeloproliferative neoplasms, including essential thrombocythemia (ET; n = 14,676), polycythemia vera (PV; n = 15,141), and primary myelofibrosis (PMF; n = 4214), using Surveillance, Epidemiology, and End User Results (SEER) data (SEER 18; 2002-2016). Incidence and survival rates over the study period and by diagnosis year (per 5-year time frames: 2002-2006; 2007-2011; 2012-2016) were assessed. The overall incidence rates (95% CI) were 1.55 (1.52-1.57) for ET, 1.57 (1.55-1.60) for PV, and 0.44 (0.43-0.45) per 100,000 person-years for PMF, with rising ET incidence. Five-year mortality over the study period was 19.2%, 19.0%, and 51.0% for ET, PV, and PMF, respectively. Improved survival over time was observed for PV and PMF, but not for ET. These findings highlight the need for effective ET therapies, as ET incidence has risen without concurrent improvements in survival over the past 2 decades.
Collapse
Affiliation(s)
- Srdan Verstovsek
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jingbo Yu
- Incyte Corporation, Wilmington, DE, USA
| | | | | | | | | | | |
Collapse
|
143
|
Dutta A, Nath D, Yang Y, Le BT, Rahman MFU, Faughnan P, Wang Z, Stuver M, He R, Tan W, Hutchison RE, Foulks JM, Warner SL, Zang C, Mohi G. Genetic ablation of Pim1 or pharmacologic inhibition with TP-3654 ameliorates myelofibrosis in murine models. Leukemia 2022; 36:746-759. [PMID: 34741118 PMCID: PMC8891046 DOI: 10.1038/s41375-021-01464-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 10/19/2021] [Accepted: 10/21/2021] [Indexed: 12/14/2022]
Abstract
Myelofibrosis (MF) is the deadliest form of myeloproliferative neoplasm (MPN). The JAK inhibitor Ruxolitinib can reduce constitutional symptoms but it does not substantially improve bone marrow fibrosis. Pim1 expression is significantly elevated in MPN/MF hematopoietic progenitors. Here, we show that genetic ablation of Pim1 blocked the development of myelofibrosis induced by Jak2V617F and MPLW515L. Pharmacologic inhibition of Pim1 with a second-generation Pim kinase inhibitor TP-3654 significantly reduced leukocytosis and splenomegaly, and attenuated bone marrow fibrosis in Jak2V617F and MPLW515L mouse models of MF. Combined treatment of TP-3654 and Ruxolitinib resulted in greater reduction of spleen size, normalization of blood leukocyte counts and abrogation of bone marrow fibrosis in murine models of MF. TP-3654 treatment also preferentially inhibited Jak2V617F mutant hematopoietic progenitors in mice. Mechanistically, we show that TP-3654 treatment significantly inhibits mTORC1, MYC and TGF-β signaling in Jak2V617F mutant hematopoietic cells and diminishes the expression of fibrotic markers in the bone marrow. Collectively, our results suggest that Pim1 plays an important role in the pathogenesis of MF, and inhibition of Pim1 with TP-3654 might be useful for treatment of MF.
Collapse
Affiliation(s)
- Avik Dutta
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Dipmoy Nath
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Yue Yang
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Bao T Le
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Mohammad Ferdous-Ur Rahman
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Patrick Faughnan
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Zhenjia Wang
- Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Matthew Stuver
- Department of Pharmacology, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, NY, USA
| | - Rongquan He
- Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Wuwei Tan
- Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Robert E Hutchison
- Department of Pathology, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, NY, USA
| | - Jason M Foulks
- Sumitomo Dainippon Pharma Oncology, Inc (formerly Tolero Pharmaceuticals, Inc), Lehi, UT, USA
| | - Steven L Warner
- Sumitomo Dainippon Pharma Oncology, Inc (formerly Tolero Pharmaceuticals, Inc), Lehi, UT, USA
| | - Chongzhi Zang
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, USA
- Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, VA, USA
- Department of Public Health Sciences, University of Virginia School of Medicine, Charlottesville, VA, USA
- University of Virginia Cancer Center, Charlottesville, VA, USA
| | - Golam Mohi
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, USA.
- University of Virginia Cancer Center, Charlottesville, VA, USA.
| |
Collapse
|
144
|
Novel treatments for myelofibrosis: beyond JAK inhibitors. Int J Hematol 2022; 115:645-658. [PMID: 35182376 DOI: 10.1007/s12185-022-03299-8] [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: 01/11/2022] [Revised: 01/24/2022] [Accepted: 01/24/2022] [Indexed: 10/19/2022]
Abstract
Myelofibrosis is a chronic hematologic malignancy characterized by constitutional symptoms, bone marrow fibrosis, extramedullary hematopoiesis resulting in splenomegaly and a propensity toward leukemic progression. Given the central role of the JAK-STAT pathway in the pathobiology of myelofibrosis, JAK inhibitors are the mainstay of current pharmacologic management. Although these therapies have produced meaningful improvements in splenomegaly and symptom burden, JAK inhibitors do not significantly impact disease progression. In addition, many patients are ineligible because of disease-related cytopenias, which are exacerbated by JAK inhibitors. Therefore, there is a continued effort to identify targets outside the JAK-STAT pathway. In this review, we discuss novel therapies in development for myelofibrosis. We focus on the preclinical rationale, efficacy and safety data for non-JAK inhibitor therapies that have published or presented clinical data. Specifically, we discuss agents that target epigenetic modification (pelabresib, bomedemstat), apoptosis (navitoclax, navtemdalin), signaling pathways (parsaclisib), bone marrow fibrosis (AVID200, PRM-151), in addition to other targets including telomerase (imetelstat), selective inhibitor of nuclear transport (selinexor), CD123 (tagraxofusp) and erythroid maturation (luspatercept). We end by providing commentary on the ongoing and future therapeutic development in myelofibrosis.
Collapse
|
145
|
Bochicchio MT, Di Battista V, Poggio P, Carrà G, Morotti A, Brancaccio M, Lucchesi A. Understanding Aberrant Signaling to Elude Therapy Escape Mechanisms in Myeloproliferative Neoplasms. Cancers (Basel) 2022; 14:cancers14040972. [PMID: 35205715 PMCID: PMC8870427 DOI: 10.3390/cancers14040972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/08/2022] [Accepted: 02/10/2022] [Indexed: 02/01/2023] Open
Abstract
Aberrant signaling in myeloproliferative neoplasms may arise from alterations in genes coding for signal transduction proteins or epigenetic regulators. Both mutated and normal cells cooperate, altering fragile balances in bone marrow niches and fueling persistent inflammation through paracrine or systemic signals. Despite the hopes placed in targeted therapies, myeloid proliferative neoplasms remain incurable diseases in patients not eligible for stem cell transplantation. Due to the emergence of drug resistance, patient management is often very difficult in the long term. Unexpected connections among signal transduction pathways highlighted in neoplastic cells suggest new strategies to overcome neoplastic cell adaptation.
Collapse
Affiliation(s)
- Maria Teresa Bochicchio
- Biosciences Laboratory, IRCCS Istituto Scientifico Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy;
| | - Valeria Di Battista
- Hematology Unit, IRCCS Istituto Scientifico Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy;
| | - Pietro Poggio
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy;
| | - Giovanna Carrà
- Department of Clinical and Biological Sciences, University of Torino, 10043 Orbassano, Italy;
| | - Alessandro Morotti
- Department of Clinical and Biological Sciences, University of Torino, 10043 Orbassano, Italy;
- Correspondence: (A.M.); (M.B.); (A.L.)
| | - Mara Brancaccio
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy;
- Correspondence: (A.M.); (M.B.); (A.L.)
| | - Alessandro Lucchesi
- Hematology Unit, IRCCS Istituto Scientifico Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy;
- Correspondence: (A.M.); (M.B.); (A.L.)
| |
Collapse
|
146
|
Chojecki AL, DiSogra KY, Arnall J, Cowgill N, Soni A, Packman CH, Sanikommu SR, Shah NA, Ragon BK, Knight TG, Ai J, Avalos BR, Copelan EA, Grunwald MR. Optimization of physician and specialty pharmacy clinical workflow in assessment of risk category and symptom burden in patients with myelofibrosis (MF). Leuk Lymphoma 2022; 63:1515-1517. [PMID: 35105270 DOI: 10.1080/10428194.2022.2032037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Aleksander L Chojecki
- Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| | - Kristyn Y DiSogra
- Atrium Health Specialty Pharmacy Service, Atrium Health, Charlotte, NC, USA
| | - Justin Arnall
- Atrium Health Specialty Pharmacy Service, Atrium Health, Charlotte, NC, USA
| | - Nicole Cowgill
- Atrium Health Specialty Pharmacy Service, Atrium Health, Charlotte, NC, USA
| | - Amy Soni
- Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| | - Charles H Packman
- Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| | - Srinivasa Reddy Sanikommu
- Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| | - Nilay A Shah
- Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| | - Brittany K Ragon
- Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| | - Thomas G Knight
- Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| | - Jing Ai
- Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| | - Belinda R Avalos
- Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| | - Edward A Copelan
- Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| | - Michael R Grunwald
- Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| |
Collapse
|
147
|
Lin JQ, Li SQ, Li S, Kiamanesh EF, Aasi SZ, Kwong BY, Su Chang AL. A 10-year retrospective cohort study of ruxolitinib and association with nonmelanoma skin cancer in patients with polycythemia vera and myelofibrosis. J Am Acad Dermatol 2022; 86:339-344. [PMID: 34648874 DOI: 10.1016/j.jaad.2021.10.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 09/27/2021] [Accepted: 10/04/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND Clinical trials report occurrence of nonmelanoma skin cancers (NMSCs) with ruxolitinib in patients with polycythemia vera (PV) or myelofibrosis (MF); however, the level of risk and effect of covariates are not known in the real-world setting. OBJECTIVE To systematically assess the risk of developing NMSC after ruxolitinib exposure in patients with PV or MF. METHODS A 10-year retrospective cohort of patients with PV or MF at Stanford Medical Center was identified and matched according to age, gender, race, Charlson Comorbidity Index, disease diagnosis, and follow-up time. The main outcome measure was hazard ratio (HR) for NMSC (comprised of basal cell carcinoma and squamous cell carcinoma [SCC]) after ruxolitinib exposure, adjusted for covariates. RESULTS The study cohort consisted of 564 patients (188 exposed to ruxolitinib for at least 4 weeks, 376 unexposed). Ruxolitinib-exposed patients with PV or MF had an adjusted NMSC HR of 2.69 (95% CI, 1.03-7.02). In particular, ruxolitinib exposure was associated with SCC (HR, 3.24; 95% CI, 1.45-7.22), with non-Janus kinase 2-mutated patients showing even higher SCC risk (HR, 7.40; 95% CI, 2.54-21.63). LIMITATIONS Retrospective design. CONCLUSIONS Our real-world results indicate that SCC risk is increased in patients with PV or MF taking ruxolitinib and support consideration of skin cancer monitoring.
Collapse
Affiliation(s)
- John Q Lin
- Department of Dermatology, Stanford University School of Medicine, Redwood City, California
| | | | - Shufeng Li
- Department of Dermatology, Stanford University School of Medicine, Redwood City, California
| | - Eileen F Kiamanesh
- Research Informatics Center, Stanford University School of Medicine, Stanford, California
| | - Sumaira Z Aasi
- Department of Dermatology, Stanford University School of Medicine, Redwood City, California
| | - Bernice Y Kwong
- Department of Dermatology, Stanford University School of Medicine, Redwood City, California
| | - Anne Lynn Su Chang
- Department of Dermatology, Stanford University School of Medicine, Redwood City, California.
| |
Collapse
|
148
|
Ciochetto Z, Wainaina N, Graham MB, Corey A, Abid MB. Cryptococcal infection with ruxolitinib in primary myelofibrosis: A case report and literature review. Clin Case Rep 2022; 10:e05461. [PMID: 35369391 PMCID: PMC8858788 DOI: 10.1002/ccr3.5461] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 01/24/2022] [Accepted: 02/06/2022] [Indexed: 12/16/2022] Open
Abstract
Cryptococcus neoformans (CN) is an encapsulated yeast that is found worldwide. It causes self-limiting infections in immunocompetent hosts; however, infections due to CN could be disseminated and potentially life-threatening in immunocompromised hosts. Herein, we present a patient with primary myelofibrosis who received ruxolitinib and developed disseminated cryptococcosis due to CN. We further discuss immune compromising factors indigenous to myeloproliferative neoplasms, ruxolitinib, and immunological pathways associated with janus kinase inhibition. We further review other cases of cryptococcal infections in patients receiving ruxolitinib reported in the literature. The report underscores the importance of suspecting infections with intracellular pathogens early in the course of illness in patients with higher rates of cumulative immunosuppression. A high clinical suspicion should be maintained when caring for such immunosuppressed patients receiving immunomodulatory agents as severe, disseminated infections can present atypically and lead to worse outcomes.
Collapse
Affiliation(s)
- Zachary Ciochetto
- Division of Infectious DiseasesMedical College of WisconsinMilwaukeeWisconsinUSA
| | - Njeri Wainaina
- Division of Infectious DiseasesMedical College of WisconsinMilwaukeeWisconsinUSA
| | - Mary Beth Graham
- Division of Infectious DiseasesMedical College of WisconsinMilwaukeeWisconsinUSA
| | - Anna Corey
- Division of Infectious DiseasesMedical College of WisconsinMilwaukeeWisconsinUSA
| | - Muhammad Bilal Abid
- Division of Infectious DiseasesMedical College of WisconsinMilwaukeeWisconsinUSA
| |
Collapse
|
149
|
Bader MS, Meyer SC. JAK2 in Myeloproliferative Neoplasms: Still a Protagonist. Pharmaceuticals (Basel) 2022; 15:ph15020160. [PMID: 35215273 PMCID: PMC8874480 DOI: 10.3390/ph15020160] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/25/2022] [Accepted: 01/26/2022] [Indexed: 02/01/2023] Open
Abstract
The discovery of the activating V617F mutation in Janus kinase 2 (JAK2) has been decisive for the understanding of myeloproliferative neoplasms (MPN). Activated JAK2 signaling by JAK2, CALR, and MPL mutations has become a focus for the development of targeted therapies for patients with MPN. JAK2 inhibitors now represent a standard of clinical care for certain forms of MPN and offer important benefits for MPN patients. However, several key aspects remain unsolved regarding the targeted therapy of MPN with JAK2 inhibitors, such as reducing the MPN clone and how to avoid or overcome a loss of response. Here, we summarize the current knowledge on the structure and signaling of JAK2 as central elements of MPN pathogenesis and feature benefits and limitations of therapeutic JAK2 targeting in MPN.
Collapse
Affiliation(s)
| | - Sara Christina Meyer
- Division of Hematology, University Hospital Basel, CH-4031 Basel, Switzerland;
- Department of Biomedicine, University Hospital Basel and University of Basel, CH-4031 Basel, Switzerland
- Correspondence:
| |
Collapse
|
150
|
Vachhani P, Verstovsek S, Bose P. Disease Modification in Myelofibrosis: An Elusive Goal? J Clin Oncol 2022; 40:1147-1154. [PMID: 35084934 PMCID: PMC8987221 DOI: 10.1200/jco.21.02246] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Affiliation(s)
- Pankit Vachhani
- Department of Medicine, Division of Hematology and Oncology, The University of Alabama at Birmingham, Birmingham, AL
| | - Srdan Verstovsek
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Prithviraj Bose
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|