1
|
Melica ME, Antonelli G, Semeraro R, Angelotti ML, Lugli G, Landini S, Ravaglia F, La Regina G, Conte C, De Chiara L, Peired AJ, Mazzinghi B, Donati M, Molli A, Steiger S, Magi A, Bartalucci N, Raglianti V, Guzzi F, Maggi L, Annunziato F, Burger A, Lazzeri E, Anders HJ, Lasagni L, Romagnani P. Differentiation of crescent-forming kidney progenitor cells into podocytes attenuates severe glomerulonephritis in mice. Sci Transl Med 2022; 14:eabg3277. [PMID: 35947676 PMCID: PMC7614034 DOI: 10.1126/scitranslmed.abg3277] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Crescentic glomerulonephritis is characterized by vascular necrosis and parietal epithelial cell hyperplasia in the space surrounding the glomerulus, resulting in the formation of crescents. Little is known about the molecular mechanisms driving this process. Inducing crescentic glomerulonephritis in two Pax2Cre reporter mouse models revealed that crescents derive from clonal expansion of single immature parietal epithelial cells. Preemptive and delayed histone deacetylase inhibition with panobinostat, a drug used to treat hematopoietic stem cell disorders, attenuated crescentic glomerulonephritis with recovery of kidney function in the two mouse models. Three-dimensional confocal microscopy and stimulated emission depletion superresolution imaging of mouse glomeruli showed that, in addition to exerting an anti-inflammatory and immunosuppressive effect, panobinostat induced differentiation of an immature hyperplastic parietal epithelial cell subset into podocytes, thereby restoring the glomerular filtration barrier. Single-cell RNA sequencing of human renal progenitor cells in vitro identified an immature stratifin-positive cell subset and revealed that expansion of this stratifin-expressing progenitor cell subset was associated with a poor outcome in human crescentic glomerulonephritis. Treatment of human parietal epithelial cells in vitro with panobinostat attenuated stratifin expression in renal progenitor cells, reduced their proliferation, and promoted their differentiation into podocytes. These results offer mechanistic insights into the formation of glomerular crescents and demonstrate that selective targeting of renal progenitor cells can attenuate crescent formation and the deterioration of kidney function in crescentic glomerulonephritis in mice.
Collapse
Affiliation(s)
- Maria Elena Melica
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence 50139, Italy,Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy
| | - Giulia Antonelli
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence 50139, Italy,Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy
| | - Roberto Semeraro
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Maria Lucia Angelotti
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence 50139, Italy,Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy
| | - Gianmarco Lugli
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy,Nephrology and Dialysis Unit, Meyer Children’s Hospital, Florence 50139, Italy
| | - Samuela Landini
- Nephrology and Dialysis Unit, Meyer Children’s Hospital, Florence 50139, Italy
| | - Fiammetta Ravaglia
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy
| | - Gilda La Regina
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy
| | - Carolina Conte
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence 50139, Italy,Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy
| | - Letizia De Chiara
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence 50139, Italy,Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy
| | - Anna Julie Peired
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence 50139, Italy,Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy
| | - Benedetta Mazzinghi
- Nephrology and Dialysis Unit, Meyer Children’s Hospital, Florence 50139, Italy
| | - Marta Donati
- Nephrology and Dialysis Unit, Meyer Children’s Hospital, Florence 50139, Italy
| | - Alice Molli
- Nephrology and Dialysis Unit, Meyer Children’s Hospital, Florence 50139, Italy
| | - Stefanie Steiger
- Division of Nephrology, Medizinische Klinik and Poliklinik IV, Klinikum der LMU München, Munich 80336, Germany
| | - Alberto Magi
- Department of Information Engineering, University of Florence, Florence, Italy
| | - Niccolò Bartalucci
- Department of Experimental and Clinical Medicine, CRIMM, Center Research and Innovation of Myeloproliferative Neoplasms, AOUC, University of Florence, Florence 50139, Italy
| | - Valentina Raglianti
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy,Nephrology and Dialysis Unit, Meyer Children’s Hospital, Florence 50139, Italy
| | - Francesco Guzzi
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy,Nephrology and Dialysis Unit, Meyer Children’s Hospital, Florence 50139, Italy
| | - Laura Maggi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Francesco Annunziato
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Alexa Burger
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Elena Lazzeri
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence 50139, Italy,Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy
| | - Hans-Joachim Anders
- Division of Nephrology, Medizinische Klinik and Poliklinik IV, Klinikum der LMU München, Munich 80336, Germany
| | - Laura Lasagni
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence 50139, Italy,Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy,Corresponding authors. and
| | - Paola Romagnani
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence 50139, Italy,Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy,Nephrology and Dialysis Unit, Meyer Children’s Hospital, Florence 50139, Italy,Corresponding authors. and
| |
Collapse
|
2
|
Pemmaraju N, Carter BZ, Bose P, Jain N, Kadia TM, Garcia-Manero G, Bueso-Ramos CE, DiNardo CD, Bledsoe S, Daver NG, Popat U, Konopleva MY, Zhou L, Pierce S, Estrov ZE, Borthakur GM, Ohanian M, Qiao W, Masarova L, Wang X, Mak PY, Cortes J, Jabbour E, Verstovsek S. Final results of a phase 2 clinical trial of LCL161, an oral SMAC mimetic for patients with myelofibrosis. Blood Adv 2021; 5:3163-3173. [PMID: 34424319 PMCID: PMC8405193 DOI: 10.1182/bloodadvances.2020003829] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 04/01/2021] [Indexed: 12/28/2022] Open
Abstract
Outcomes in patients with high-risk and treatment-resistant myelofibrosis (MF) post-JAK inhibitor therapy remain poor, with no approved drug therapies beyond the JAK inhibitor class. In certain clinical situations, such as severe thrombocytopenia, administration of most JAK inhibitors are contraindicated. Thus, there is an unmet medical need for the development of novel agents for patients with MF. SMAC mimetics [or inhibitor of apoptosis (IAP) antagonists] induce apoptosis in cancer cells. Because these agents are hypothesized to have increased activity in a tumor necrosis factor-α cytokine-rich microenvironment, as is the case with MF, we conducted a single-center, investigator-initiated phase 2 clinical trial, with a monovalent SMAC mimetic LCL161 (oral, starting dose, 1500 mg per week) in patients with intermediate to high-risk MF. In an older group, 66% with ≥2 prior therapies and a median baseline platelet count of 52 × 103/μL and 28% with ASXL1 mutations, we observed a 30% objective response by Revised International Working Group-Myeloproliferative Neoplasms Research and Treatment (IWG-MRT) 2013 criteria. Notably, 6 responding patients achieved clinical improvement of anemia: 4, hemoglobin response; 2, transfusion independence. Median OS was 34 months (range, 2.2-60.1+). Reductions of cIAPs were observed in all responders. The most common toxicity was nausea/vomiting (N/V) in 64% (mostly grade 1/2); fatigue in 46%; and dizziness/vertigo in 30%. There were 4 grade 3/4 adverse events (2, syncope; 1, N/V; 1, skin eruption/pruritis). There were 2 deaths during the study period, both unrelated to the study drug. SMAC mimetics may represent an option for older patients with thrombocytopenia or for those in whom prior JAK inhibitors has failed. This trial was registered at www.clinicaltrials.gov as #NCT02098161.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Uday Popat
- Department of Stem Cell Transplantation, and
| | | | | | | | | | | | | | - Wei Qiao
- Department of Biostatistics, MD Anderson Cancer Center, Houston, TX; and
| | | | - Xuemei Wang
- Department of Biostatistics, MD Anderson Cancer Center, Houston, TX; and
| | | | - Jorge Cortes
- Georgia Cancer Center, Augusta University, Augusta, GA
| | | | | |
Collapse
|
3
|
Palmer J, Mesa R. The role of fedratinib for the treatment of patients with primary or secondary myelofibrosis. Ther Adv Hematol 2020; 11:2040620720925201. [PMID: 32477483 PMCID: PMC7232117 DOI: 10.1177/2040620720925201] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 03/03/2020] [Indexed: 01/01/2023] Open
Abstract
Myelofibrosis (MF) is a chronic myeloid neoplasm characterized by either primary
myelofibrosis, or secondary MF following essential thrombocythemia or
polycythemia vera. Historically, therapy has been symptom directed; however, in
2011, the first janus kinase inhibitor (JAK-i) – ruxolitinib – was approved for
treatment. This medication was found to be effective in reduction of symptom
burden and spleen size; however, the median duration of response is about 3
years. In addition, many patients are intolerant or develop toxicities to
ruxolitinib, including patients with anemia, as well as thrombocytopenia.
Therefore, there is a critical need for alternate therapeutic options for
patients with MF. Additional JAK-i have been developed over the last 8 years,
including fedratinib, momelotinib, and pacritinib. Fedratinib recently received
approval for treatment of MF both in the first-line and second-line setting. It
has shown efficacy in the first-line setting, as well as in 30% of patients who
are refractory/intolerant of ruxolitinib. This review covers the trials that
have led to the approval of ruxolitinib as well as fedratinib, as well as
reviews of two JAK inhibitors that are still under clinical investigation:
momelotinib and pacritinib.
Collapse
Affiliation(s)
- Jeanne Palmer
- Division of Hematology and Oncology, Mayo Clinic, Phoenix AZ, USA
| | - Ruben Mesa
- Division of Hematology and Oncology, UT Health San Antonio MD Anderson, Mail Code 8026, 6th Floor Urschel Tower, 7979 Wurzbach Rd, San Antonio, TX 78229-3900
| |
Collapse
|
4
|
Palandri F, Breccia M, Bonifacio M, Polverelli N, Elli EM, Benevolo G, Tiribelli M, Abruzzese E, Iurlo A, Heidel FH, Bergamaschi M, Tieghi A, Crugnola M, Cavazzini F, Binotto G, Isidori A, Sgherza N, Bosi C, Martino B, Latagliata R, Auteri G, Scaffidi L, Griguolo D, Trawinska M, Cattaneo D, Catani L, Krampera M, Lemoli RM, Cuneo A, Semenzato G, Foà R, Di Raimondo F, Bartoletti D, Cavo M, Palumbo GA, Vianelli N. Life after ruxolitinib: Reasons for discontinuation, impact of disease phase, and outcomes in 218 patients with myelofibrosis. Cancer 2019; 126:1243-1252. [PMID: 31860137 DOI: 10.1002/cncr.32664] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 11/11/2019] [Accepted: 11/22/2019] [Indexed: 01/08/2023]
Abstract
BACKGROUND After discontinuing ruxolitinib, the outcome of patients with myelofibrosis reportedly has been poor. The authors investigated whether disease characteristics before the receipt of ruxolitinib may predict drug discontinuation in patients with myelofibrosis and whether reasons for drug discontinuation, disease phase at discontinuation, and salvage therapies may influence the outcome. METHODS A centralized electronic clinical database was created in 20 European hematology centers, including clinical and laboratory data for 524 patients who received ruxolitinib for myelofibrosis. RESULTS At 3 years, 40.8% of patients had stopped ruxolitinib. Baseline predictors of drug discontinuation were: intermediate-2-risk/high-risk category (Dynamic International Prognostic Score System), a platelet count <100 ×109 per liter, transfusion dependency, and unfavorable karyotype. At last contact, 268 patients (51.1%) had discontinued therapy, and the median drug exposure was 17.5 months. Fifty patients (18.7%) died while taking ruxolitinib. The reasons for discontinuation in the remaining 218 patients were the lack (22.9%) or loss (11.9%) of a spleen response, ruxolitinib-related adverse events (27.5%), progression to blast phase (23.4%), ruxolitinib-unrelated adverse events (9.2%), and allogeneic transplantation during response (5.1%). The median survival after ruxolitinib was 13.2 months and was significantly better in the 167 patients who discontinued ruxolitinib in chronic phase (27.5 vs 3.9 months for those who discontinued in blast phase; P < .001). No survival differences were observed among patients who discontinued ruxolitinib in chronic phase because of lack of response, loss of response, or ruxolitinib-related adverse events. The use of investigational agents and/or ruxolitinib rechallenge were associated with improved outcome. CONCLUSIONS The survival of patients with myelofibrosis after discontinuation of ruxolitinib is poor, particularly for those who discontinue in blast phase. Salvage therapies can improve outcome, emphasizing the need for novel therapies.
Collapse
Affiliation(s)
- Francesca Palandri
- Institute of Hematology "L. and A. Seràgnoli", St Orsola-Malpighi University Hospital, Bologna, Italy
| | - Massimo Breccia
- Division of Cellular Biotechnology and Hematology, Sapienza University, Rome, Italy
| | | | - Nicola Polverelli
- Unit of Blood Diseases and Stem Cell Transplantation, Department of Clinical and Experimental Sciences, University of Brescia, ASST-Spedali Civili di Brescia, Brescia, Italy
| | - Elena M Elli
- Hematology Division and Bone Marrow Unit, San Gerardo Hospital, Monza, Italy
| | - Giulia Benevolo
- Division of Hematology, City Hospital of Health and Science, Turin, Italy
| | - Mario Tiribelli
- Division of Hematology and Bone Marrow Transplantation, Integrated Healthcare University of Udine, Udine, Italy
| | | | - Alessandra Iurlo
- Hematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico and University of Milan, Milan, Italy
| | - Florian H Heidel
- Internal Medicine II, Hematology and Oncology, Friedrich Schiller University Medical Center, Jena, Germany
| | - Micaela Bergamaschi
- Clinic of Hematology, Department of Internal Medicine, IRCCS San Martino Hospital, University of Genoa, Genoa, Italy
| | - Alessia Tieghi
- Hematology Unit, Azienda Unità Sanitaria Locale - IRCCS, Arcispedale S.Maria Nuova, Reggio Emilia, Italy
| | - Monica Crugnola
- Division of Hematology, University Hospital of Parma, Parma, Italy
| | | | - Gianni Binotto
- Unit of Hematology and Clinical Immunology, University of Padua, Padua, Italy
| | - Alessandro Isidori
- Hematology and Stem Cell Transplantation Center, Azienda Ospedaliera Ospedali Riuniti Marche Nord (AORMN), Pesaro, Italy
| | - Nicola Sgherza
- Division of Hematology, Home for the Relief of Suffering, San Giovanni Rotondo, Italy
| | - Costanza Bosi
- Division of Hematology, Local Healthcare Unit of Piacenza, Piacenza, Italy
| | - Bruno Martino
- Division of Hematology, "Bianchi Melacrino Morelli" Hospital Corporation, Reggio Calabria, Italy
| | - Roberto Latagliata
- Division of Cellular Biotechnology and Hematology, Sapienza University, Rome, Italy
| | - Giuseppe Auteri
- Institute of Hematology "L. and A. Seràgnoli", St Orsola-Malpighi University Hospital, Bologna, Italy
| | - Luigi Scaffidi
- Department of Medicine, Section of Hematology, University of Verona, Verona, Italy
| | - Davide Griguolo
- Division of Hematology and Bone Marrow Transplantation, Integrated Healthcare University of Udine, Udine, Italy
| | | | - Daniele Cattaneo
- Hematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico and University of Milan, Milan, Italy
| | - Lucia Catani
- Institute of Hematology "L. and A. Seràgnoli", St Orsola-Malpighi University Hospital, Bologna, Italy
| | - Mauro Krampera
- Department of Medicine, Section of Hematology, University of Verona, Verona, Italy
| | - Roberto M Lemoli
- Clinic of Hematology, Department of Internal Medicine, IRCCS San Martino Hospital, University of Genoa, Genoa, Italy
| | - Antonio Cuneo
- Division of Hematology, University of Ferrara, Ferrara, Italy
| | | | - Robin Foà
- Division of Cellular Biotechnology and Hematology, Sapienza University, Rome, Italy
| | - Francesco Di Raimondo
- Division of Hematology, V. Emanuele University Polyclinic, University of Catania, Catania, Italy
| | - Daniela Bartoletti
- Institute of Hematology "L. and A. Seràgnoli", St Orsola-Malpighi University Hospital, Bologna, Italy
| | - Michele Cavo
- Institute of Hematology "L. and A. Seràgnoli", St Orsola-Malpighi University Hospital, Bologna, Italy
| | - Giuseppe A Palumbo
- Department of Medical Science, Surgery, and Advanced Technology "G. F. Ingrassia", University of Catania, Catania, Italy
| | - Nicola Vianelli
- Institute of Hematology "L. and A. Seràgnoli", St Orsola-Malpighi University Hospital, Bologna, Italy
| |
Collapse
|
5
|
Diamantopoulos P, Giannakopoulou N, Hatzidavid S, Viniou NA. Post-Myelofibrosis Acute Myeloid Leukemia Effectively Treated with a Combination of Ruxolitinib and 5-Azacytidine. Oncol Res Treat 2019; 42:532-535. [DOI: 10.1159/000502121] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 07/13/2019] [Indexed: 11/19/2022]
|
6
|
Abstract
Myelofibrosis (MF) is a myeloproliferative neoplasm that is pathologically characterized by bone marrow myeloproliferation, reticulin and collagen fibrosis, and extramedullary hematopoiesis. Constitutive activation of the Janus associated kinase (JAK)-signal transducers and activators of transcription signaling pathway with resultant elevation in pro-inflammatory cytokine levels is the pathogenic hallmark of MF. JAK inhibitors, namely ruxolitinib, have been successful in alleviating symptoms and reducing splenomegaly, but therapy-related myelosuppression has led to the further development of highly selective JAK2 inhibitors. Additionally, ruxolitinib does not appear to affect the malignant hematopoietic clone substantially, evidenced by lack of molecular remissions, bone marrow histopathologic responses, and a proportion of treated patients developing progressive disease and leukemic transformation while receiving therapy. A number of other pharmacotherapeutic strategies are currently being explored in the clinic. Non-JAK inhibitor strategies being evaluated in MF include non-JAK signaling pathway inhibitors, epigenetic-directed therapies, immune-modulating agents, anti-fibrotic agents, and telomerase inhibitors. This review highlights the current landscape of MF pharmacotherapy and explores therapeutic advances underway.
Collapse
Affiliation(s)
- Douglas Tremblay
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L Levy Place, Box 1079, New York, NY, 10029, USA
| | - Bridget Marcellino
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L Levy Place, Box 1079, New York, NY, 10029, USA
| | - John Mascarenhas
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L Levy Place, Box 1079, New York, NY, 10029, USA.
| |
Collapse
|
7
|
Reactive Oxygen Species Drive Epigenetic Changes in Radiation-Induced Fibrosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:4278658. [PMID: 30881591 PMCID: PMC6381575 DOI: 10.1155/2019/4278658] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 12/06/2018] [Accepted: 12/12/2018] [Indexed: 12/14/2022]
Abstract
Radiation-induced fibrosis (RIF) develops months to years after initial radiation exposure. RIF occurs when normal fibroblasts differentiate into myofibroblasts and lay down aberrant amounts of extracellular matrix proteins. One of the main drivers for developing RIF is reactive oxygen species (ROS) generated immediately after radiation exposure. Generation of ROS is known to induce epigenetic changes and cause differentiation of fibroblasts to myofibroblasts. Several antioxidant compounds have been shown to prevent radiation-induced epigenetic changes and the development of RIF. Therefore, reviewing the ROS-linked epigenetic changes in irradiated fibroblast cells is essential to understand the development and prevention of RIF.
Collapse
|
8
|
Gowin K, Kosiorek H, Dueck A, Mascarenhas J, Hoffman R, Reeder C, Camoriano J, Tibes R, Gano K, Palmer J, Mesa R. Multicenter phase 2 study of combination therapy with ruxolitinib and danazol in patients with myelofibrosis. Leuk Res 2017. [PMID: 28646676 DOI: 10.1016/j.leukres.2017.06.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Myelofibrosis is a myeloproliferative neoplasm that is characterized by splenomegaly, profound symptom burden, and cytopenias. JAK inhibitor therapy offers improvements in splenomegaly, symptom burden, and potentially survival; however, cytopenias remain a significant challenge. Danazol has previously demonstrated improvements in myelofibrosis-associated anemia. We conducted a phase II clinical trial evaluating the efficacy and tolerability of combination therapy with ruxolitinib, an oral JAK inhibitor, and danazol. Fourteen intermediate or high-risk MF patients were enrolled at 2 institutions. Responses per IWG-MRT criteria were stable disease in 9 patients (64.2%) clinical improvement in 3 (21.4%) all of which were spleen responses, partial response in 1 (7.1%) and progressive disease in 1 (7.1%). Despite limited IWG-MRT response, stabilization of anemia and thrombocytopenia was demonstrated. In JAK inhibitor naïve patients, 4/5 (80%) had stable or increasing hemoglobin. Of the 9 patients on prior JAK inhibitor, 5 patients (55.5%) and 8 patients (88.9%) had stable or increasing hemoglobin or platelet levels, respectively. Adverse events possibly related included grade 3 or greater hematologic toxicity in ten patients (71.4%) and non-hematologic toxicity in two patients (14.3%). Although combination therapy did not lead to increased hematologic response per IWG-MRT criteria, hematologic stabilization was observed and may be clinically useful.
Collapse
Affiliation(s)
- K Gowin
- Mayo Clinic Arizona, Department of Hematology, Phoenix, AZ, USA
| | - H Kosiorek
- Mayo Clinic Arizona, Department of Health Sciences Research, Section of Biostatistics, Phoenix, AZ, USA
| | - A Dueck
- Mayo Clinic Arizona, Department of Health Sciences Research, Section of Biostatistics, Phoenix, AZ, USA
| | - J Mascarenhas
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, USA
| | - R Hoffman
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, USA
| | - C Reeder
- Mayo Clinic Arizona, Department of Hematology, Phoenix, AZ, USA
| | - J Camoriano
- Mayo Clinic Arizona, Department of Hematology, Phoenix, AZ, USA
| | - R Tibes
- Mayo Clinic Arizona, Department of Hematology, Phoenix, AZ, USA
| | - K Gano
- Mayo Clinic Arizona, Department of Hematology, Phoenix, AZ, USA
| | - J Palmer
- Mayo Clinic Arizona, Department of Hematology, Phoenix, AZ, USA
| | - R Mesa
- Mayo Clinic Arizona, Department of Hematology, Phoenix, AZ, USA.
| |
Collapse
|
9
|
Virtgaym E, Tremblay D, Iancu-Rubin C, Hoffman R, Mascarenhas J. Imetelstat for treatment of myelofibrosis. Expert Opin Orphan Drugs 2016. [DOI: 10.1080/21678707.2017.1266934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- E. Virtgaym
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - D. Tremblay
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - C. Iancu-Rubin
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - R. Hoffman
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - J. Mascarenhas
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
10
|
Mascarenhas JO, Talpaz M, Gupta V, Foltz LM, Savona MR, Paquette R, Turner AR, Coughlin P, Winton E, Burn TC, O'Neill P, Clark J, Hunter D, Assad A, Hoffman R, Verstovsek S. Primary analysis of a phase II open-label trial of INCB039110, a selective JAK1 inhibitor, in patients with myelofibrosis. Haematologica 2016; 102:327-335. [PMID: 27789678 DOI: 10.3324/haematol.2016.151126] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 10/17/2016] [Indexed: 12/24/2022] Open
Abstract
Combined Janus kinase 1 (JAK1) and JAK2 inhibition therapy effectively reduces splenomegaly and symptom burden related to myelofibrosis but is associated with dose-dependent anemia and thrombocytopenia. In this open-label phase II study, we evaluated the efficacy and safety of three dose levels of INCB039110, a potent and selective oral JAK1 inhibitor, in patients with intermediate- or high-risk myelofibrosis and a platelet count ≥50×109/L. Of 10, 45, and 32 patients enrolled in the 100 mg twice-daily, 200 mg twice-daily, and 600 mg once-daily cohorts, respectively, 50.0%, 64.4%, and 68.8% completed week 24. A ≥50% reduction in total symptom score was achieved by 35.7% and 28.6% of patients in the 200 mg twice-daily cohort and 32.3% and 35.5% in the 600 mg once-daily cohort at week 12 (primary end point) and 24, respectively. By contrast, two patients (20%) in the 100 mg twice-daily cohort had ≥50% total symptom score reduction at weeks 12 and 24. For the 200 mg twice-daily and 600 mg once-daily cohorts, the median spleen volume reductions at week 12 were 14.2% and 17.4%, respectively. Furthermore, 21/39 (53.8%) patients who required red blood cell transfusions during the 12 weeks preceding treatment initiation achieved a ≥50% reduction in the number of red blood cell units transfused during study weeks 1-24. Only one patient discontinued for grade 3 thrombocytopenia. Non-hematologic adverse events were largely grade 1 or 2; the most common was fatigue. Treatment with INCB039110 resulted in clinically meaningful symptom relief, modest spleen volume reduction, and limited myelosuppression.
Collapse
Affiliation(s)
| | - Moshe Talpaz
- University of Michigan Cancer Center, Ann Arbor, MI, USA
| | - Vikas Gupta
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Lynda M Foltz
- St. Paul's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Michael R Savona
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | | | | | - Elliott Winton
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | | | | | | | | | | | - Ronald Hoffman
- The Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Srdan Verstovsek
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
11
|
A data-driven network model of primary myelofibrosis: transcriptional and post-transcriptional alterations in CD34+ cells. Blood Cancer J 2016; 6:e439. [PMID: 27341078 PMCID: PMC5141361 DOI: 10.1038/bcj.2016.47] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 05/19/2016] [Indexed: 12/18/2022] Open
Abstract
microRNAs (miRNAs) are relevant in the pathogenesis of primary myelofibrosis (PMF) but our understanding is limited to specific target genes and the overall systemic scenario islacking. By both knowledge-based and ab initio approaches for comparative analysis of CD34+ cells of PMF patients and healthy controls, we identified the deregulated pathways involving miRNAs and genes and new transcriptional and post-transcriptional regulatory circuits in PMF cells. These converge in a unique and integrated cellular process, in which the role of specific miRNAs is to wire, co-regulate and allow a fine crosstalk between the involved processes. The PMF pathway includes Akt signaling, linked to Rho GTPases, CDC42, PLD2, PTEN crosstalk with the hypoxia response and Calcium-linked cellular processes connected to cyclic AMP signaling. Nested on the depicted transcriptional scenario, predicted circuits are reported, opening new hypotheses. Links between miRNAs (miR-106a-5p, miR-20b-5p, miR-20a-5p, miR-17-5p, miR-19b-3p and let-7d-5p) and key transcription factors (MYCN, ATF, CEBPA, REL, IRF and FOXJ2) and their common target genes tantalizingly suggest new path to approach the disease. The study provides a global overview of transcriptional and post-transcriptional deregulations in PMF, and, unifying consolidated and predicted data, could be helpful to identify new combinatorial therapeutic strategy. Interactive PMF network model: http://compgen.bio.unipd.it/pmf-net/.
Collapse
|
12
|
Zahr AA, Salama ME, Carreau N, Tremblay D, Verstovsek S, Mesa R, Hoffman R, Mascarenhas J. Bone marrow fibrosis in myelofibrosis: pathogenesis, prognosis and targeted strategies. Haematologica 2016; 101:660-71. [PMID: 27252511 PMCID: PMC5013940 DOI: 10.3324/haematol.2015.141283] [Citation(s) in RCA: 133] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 02/08/2016] [Indexed: 12/14/2022] Open
Abstract
Bone marrow fibrosis is a central pathological feature and World Health Organization major diagnostic criterion of myelofibrosis. Although bone marrow fibrosis is seen in a variety of malignant and non-malignant disease states, the deposition of reticulin and collagen fibrosis in the bone marrow of patients with myelofibrosis is believed to be mediated by the myelofibrosis hematopoietic stem/progenitor cell, contributing to an impaired microenvironment favoring malignant over normal hematopoiesis. Increased expression of inflammatory cytokines, lysyl oxidase, transforming growth factor-β, impaired megakaryocyte function, and aberrant JAK-STAT signaling have all been implicated in the pathogenesis of bone marrow fibrosis. A number of studies indicate that bone marrow fibrosis is an adverse prognostic variable in myeloproliferative neoplasms. However, modern myelofibrosis prognostication systems utilized in risk-adapted treatment approaches do not include bone marrow fibrosis as a prognostic variable. The specific effect on bone marrow fibrosis of JAK2 inhibition, and other rationally based therapies currently being evaluated in myelofibrosis, has yet to be fully elucidated. Hematopoietic stem cell transplantation remains the only curative therapeutic approach that reliably results in resolution of bone marrow fibrosis in patients with myelofibrosis. Here we review the pathogenesis, biological consequences, and prognostic impact of bone marrow fibrosis. We discuss the rationale of various anti-fibrogenic treatment strategies targeting the clonal hematopoietic stem/progenitor cell, aberrant signaling pathways, fibrogenic cytokines, and the tumor microenvironment.
Collapse
Affiliation(s)
- Abdallah Abou Zahr
- Division of Hematology Oncology, Mount Sinai Beth Israel, New York, NY, USA
| | - Mohamed E Salama
- Associated Regional University Pathologists Laboratories, Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Nicole Carreau
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Douglas Tremblay
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Srdan Verstovsek
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | - Ruben Mesa
- Division of Hematology & Medical Oncology, Mayo Clinic Cancer Center, Scottsdale, AZ, USA
| | - Ronald Hoffman
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John Mascarenhas
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
13
|
Alimam S, Harrison C. Is there a role for pomalidomide in the treatment of myelofibrosis? Expert Opin Orphan Drugs 2016. [DOI: 10.1517/21678707.2016.1171139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
14
|
Koschmieder S, Mughal TI, Hasselbalch HC, Barosi G, Valent P, Kiladjian JJ, Jeryczynski G, Gisslinger H, Jutzi JS, Pahl HL, Hehlmann R, Maria Vannucchi A, Cervantes F, Silver RT, Barbui T. Myeloproliferative neoplasms and inflammation: whether to target the malignant clone or the inflammatory process or both. Leukemia 2016; 30:1018-24. [PMID: 26854026 DOI: 10.1038/leu.2016.12] [Citation(s) in RCA: 134] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 11/28/2015] [Accepted: 12/01/2015] [Indexed: 02/07/2023]
Abstract
The Philadelphia-negative myeloproliferative neoplasms (MPNs) are clonal disorders involving hematopoietic stem and progenitor cells and are associated with myeloproliferation, splenomegaly and constitutional symptoms. Similar signs and symptoms can also be found in patients with chronic inflammatory diseases, and inflammatory processes have been found to play an important role in the pathogenesis and progression of MPNs. Signal transduction pathways involving JAK1, JAK2, STAT3 and STAT5 are causally involved in driving both the malignant cells and the inflammatory process. Moreover, anti-inflammatory and immune-modulating drugs have been used successfully in the treatment of MPNs. However, to date, many unresoved issues remain. These include the role of somatic mutations that are present in addition to JAK2V617F, CALR and MPL W515 mutations, the interdependency of malignant and nonmalignant cells and the means to eradicate MPN-initiating and -maintaining cells. It is imperative for successful therapeutic approaches to define whether the malignant clone or the inflammatory cells or both should be targeted. The present review will cover three aspects of the role of inflammation in MPNs: inflammatory states as important differential diagnoses in cases of suspected MPN (that is, in the absence of a clonal marker), the role of inflammation in MPN pathogenesis and progression and the use of anti-inflammatory drugs for MPNs. The findings emphasize the need to separate the inflammatory processes from the malignancy in order to improve our understanding of the pathogenesis, diagnosis and treatment of patients with Philadelphia-negative MPNs.
Collapse
Affiliation(s)
- S Koschmieder
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - T I Mughal
- Division of Hematology/Oncology, Tufts University Medical Center, Boston, MA, USA
| | - H C Hasselbalch
- Department of Hematology, Roskilde Hospital, Copenhagen University Hospital, Roskilde, Denmark
| | - G Barosi
- Center for the Study and Treatment of Myelofibrosis, Biotechnology Research Laboratories, Fondazione IRCCS 'Policlinico San Matteo', Pavia, Italy
| | - P Valent
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, and Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Vienna, Austria
| | - J-J Kiladjian
- Clinical Investigations Center (INSERM CIC 1427), Hôpital Saint-Louis and Paris Diderot University, Paris, France
| | - G Jeryczynski
- Division of Hematology and Blood Coagulation, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - H Gisslinger
- Division of Hematology and Blood Coagulation, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - J S Jutzi
- Division of Molecular Hematology, University Hospital Freiburg, Center for Clinical Research, Freiburg, Germany.,Spemann Graduate School of Biology and Medicine (SGBM) and Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - H L Pahl
- Division of Molecular Hematology, University Hospital Freiburg, Center for Clinical Research, Freiburg, Germany
| | - R Hehlmann
- Universitätsmedizin Mannheim, Medizinische Fakultät Mannheim der Universität Heidelberg, Mannheim, Germany
| | - A Maria Vannucchi
- CRIMM, Centro di Ricerca e Innovazione e Laboratorio Congiunto per le Malattie Mieloproliferative, Dipartimento di Medicina Sperimentale e Clinica, Centro Denothe, Azienda Ospedaliera Universitaria Careggi, Università degli Studi, Firenze, Italy
| | - F Cervantes
- Department of Hematology, Hospital Clínic, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - R T Silver
- Myeloproliferative Neoplasm Center, Division of Hematology-Oncology, Weill Cornell Medical College, New York, NY, USA
| | - T Barbui
- Clinical Research Center and Hematology, Ospedale Papa Giovanni XXIII, Bergamo, Italy
| |
Collapse
|
15
|
Duenas-Perez AB, Mead AJ. Clinical potential of pacritinib in the treatment of myelofibrosis. Ther Adv Hematol 2015; 6:186-201. [PMID: 26288713 DOI: 10.1177/2040620715586527] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Myelofibrosis (MF) is a myeloid disorder caused by a clonal hematopoietic stem-cell proliferation associated with activation of the Janus kinase (JAK) signal transducer and activator of transcription (STAT) signaling pathways. Patients with MF often develop severe splenomegaly, marked symptom burden and significant cytopenias, with a consequent marked negative impact on quality of life and survival. The management of MF patients has dramatically improved with the development of a group of drugs that inhibit JAK signaling. The first of these agents to be approved was ruxolitinib, a JAK1/JAK2 inhibitor, which has been shown to improve both spleen size and symptoms in patients with MF. However, myelotoxicity, particularly of the platelet lineage, significantly limits the patient population who can benefit from this agent. Thus, there is an unmet need for novel agents with limited myelotoxicity to treat MF. Pacritinib, a JAK2 and FMS-like tyrosine kinase 3 (FLT3) inhibitor, has shown promising results in early phase trials with limited myelotoxicity and clinical responses that are comparable with those seen with ruxolitinib, even in patients with severe thrombocytopenia. Currently there are two large phase III clinical trials of pacritinib in MF, including patients with thrombocytopenia, and those previously treated with ruxolitinib. If the encouraging results observed in early phase clinical trials are confirmed, pacritinib will represent a new and exciting treatment option for patients with MF and particularly patients with significant cytopenias.
Collapse
Affiliation(s)
- Ana B Duenas-Perez
- Haematopoietic Stem Cell Biology, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Adam J Mead
- Haematopoietic Stem Cell Biology, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| |
Collapse
|
16
|
Daver N, Cortes J, Newberry K, Jabbour E, Zhou L, Wang X, Pierce S, Kadia T, Sasaki K, Borthakur G, Ravandi F, Pemmaraju N, Kantarjian H, Verstovsek S. Ruxolitinib in combination with lenalidomide as therapy for patients with myelofibrosis. Haematologica 2015; 100:1058-63. [PMID: 26088933 DOI: 10.3324/haematol.2015.126821] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 06/16/2015] [Indexed: 02/03/2023] Open
Abstract
Ruxolitinib and lenalidomide may target distinct clinical and pathological manifestations of myelofibrosis and prevent therapy-related worsening of blood cell counts. To determine the efficacy and safety of the combination in patients with myelofibrosis, patients were given 15 mg ruxolitinib orally twice daily in continuous 28-day cycles, plus 5 mg lenalidomide orally once daily on days 1-21. Thirty-one patients were treated, with a median followup of 28 months (range, 12 - 35+). Due to failure to meet the predetermined efficacy rules for treatment success the study was terminated early. Simultaneous administration of ruxolitinib and lenalidomide was difficult: 20 of the 23 dose interruptions occurred within the first 3 months of therapy. Lenalidomide was interrupted in all 20 cases. Fourteen patients (45%) were completely off lenalidomide within 3 months of initiation. Responses were noted in 17 patients (55%). The median time to response was 1.8 months (range, 0.4 - 31). All responses were International Working Group for Myelofibrosis Research and Treatment-defined clinical improvement in palpable spleen size. One spleen responder also met the criteria for clinical improvement in hemoglobin. The response rate was higher (73%) among patients who did not require early dose interruption than among those who required early interruption (45%). Improvements in bone marrow fibrosis and serial reductions in lactate dehydrogenase >50% were noted in 17% and 50% of evaluable responders, respectively. Alternate approaches such as sequential dosing need to be evaluated when considering novel combination strategies for myelofibrosis. This trial was registered with clinicaltrials.gov identifier: NCT01375140.
Collapse
Affiliation(s)
- Naval Daver
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jorge Cortes
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kate Newberry
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Elias Jabbour
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lingsha Zhou
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xuemei Wang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sherry Pierce
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tapan Kadia
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Koji Sasaki
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gautam Borthakur
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Farhad Ravandi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Naveen Pemmaraju
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hagop Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Srdan Verstovsek
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
17
|
|
18
|
Mascarenhas JO, Cross NCP, Mesa RA. The future of JAK inhibition in myelofibrosis and beyond. Blood Rev 2014; 28:189-96. [PMID: 25043171 DOI: 10.1016/j.blre.2014.06.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 05/19/2014] [Accepted: 06/20/2014] [Indexed: 01/06/2023]
Abstract
The identification of aberrant JAK-STAT signaling in patients with myeloproliferative neoplasms has served as the basis for the development of a new class of targeted agents. Ruxolitinib, the first-in-class oral small molecule JAK1/2 inhibitor, has demonstrated clinical efficacy and shown a potential overall survival benefit in two randomized phase III clinical trials. However, this agent has not been associated with improvements in cytopenias, molecular remissions, or resolution of bone marrow fibrosis. Therefore, further translational research is needed to improve the understanding of the pathogenetic mechanisms driving this myeloid malignancy to ultimately address remaining unmet clinical needs. A number of novel JAK inhibitors are being evaluated in ongoing clinical trials and the full clinical potential of these newer agents remains incompletely understood. The use of JAK inhibition in combination therapy approaches, as well as mono- and combination therapies in the treatment of advanced forms of polycythemia vera are also under active investigation. This review will update the reader on the current understanding of oncogenic JAK-STAT pathway activity in the pathogenesis of myeloproliferative neoplasms and the current success and limitations of anti-JAK therapy.
Collapse
Affiliation(s)
- John O Mascarenhas
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | | | | |
Collapse
|