197 results on '"Lucia Masarova"'
Search Results
2. Sotatercept for anemia of myelofibrosis: a phase II investigator-initiated study
- Author
-
Prithviraj Bose, Lucia Masarova, Naveen Pemmaraju, Sharon D. Bledsoe, Naval G. Daver, Elias J. Jabbour, Tapan M. Kadia, Zeev Estrov, Steven M. Kornblau, Michael Andreeff, Nitin Jain, Jorge E. Cortes, Gautam Borthakur, Yesid Alvarado, Mary Ann Richie, Mackenzie H. Dobbins, Selene A. McCrackin, Lingsha Zhou, Sherry A. Pierce, Xuemei Wang, Allison M. Pike, Guillermo Garcia-Manero, Hagop M. Kantarjian, and Srdan Verstovsek
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2024
- Full Text
- View/download PDF
3. Interferons in the treatment of myeloproliferative neoplasms
- Author
-
Pankit Vachhani, John Mascarenhas, Prithviraj Bose, Gabriela Hobbs, Abdulraheem Yacoub, Jeanne M. Palmer, Aaron T. Gerds, Lucia Masarova, Andrew T. Kuykendall, Raajit K. Rampal, Ruben Mesa, and Srdan Verstovsek
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Interferons are cytokines with immunomodulatory properties and disease-modifying effects that have been used to treat myeloproliferative neoplasms (MPNs) for more than 35 years. The initial use of interferons was limited due to difficulties with administration and a significant toxicity profile. Many of these shortcomings were addressed by covalently binding polyethylene glycol to the interferon structure, which increases the stability, prolongs activity, and reduces immunogenicity of the molecule. In the current therapeutic landscape, pegylated interferons are recommended for use in the treatment of polycythemia vera, essential thrombocythemia, and primary myelofibrosis. We review recent efficacy, molecular response, and safety data for the two available pegylated interferons, peginterferon alfa-2a (Pegasys) and ropeginterferon alfa-2b-njft (BESREMi). The practical management of interferon-based therapies is discussed, along with our opinions on whether to and how to switch from hydroxyurea to one of these therapies. Key topics and questions related to use of interferons, such as their safety and tolerability, the significance of variant allele frequency, advantages of early treatment, and what the future of interferon therapy may look like, will be examined. Pegylated interferons represent an important therapeutic option for patients with MPNs; however, more research is still required to further refine interferon therapy.
- Published
- 2024
- Full Text
- View/download PDF
4. S172: PHASE 1/2 STUDY OF ORAL DECITABINE/CEDAZURIDINE IN COMBINATION WITH VENETOCLAX IN TREATMENT-NAÏVE HIGHER-RISK MYELODYSPLASTIC SYNDROMES OR CHRONIC MYELOMONOCYTIC LEUKEMIA
- Author
-
Alex Bataller, Alexandre Bazinet, Sangeetha Venugopal, Guillermo Montalban-Bravo, Yesid Alvarado, Kelly Chien, Ghayas Issa, Nicholas Short, Danielle Hammond, Lucia Masarova, Tapan Kadia, Rashmi Kanagal-Shamanna, Stephany Hendrickson, Farhad Ravandi, Elias Jabbour, Hagop Kantarjian, and Guillermo Garcia-Manero
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
5. P364: A PHASE II STUDY OF LOW-INTENSITY CHEMOTHERAPY (MINI-HYPER-CVD) AND PONATINIB FOLLOWED BY BLINATUMOMAB AND PONATINIB IN PATIENTS WITH PHILADELPHIA CHROMOSOME-POSITIVE ACUTE LYMPHOBLASTIC LEUKEMIA
- Author
-
Fadi Haddad, Elias Jabbour, Lewis Nasr, Nicholas Short, Walid Macaron, Cedric Nasnas, Marianne Zoghbi, Ghayas Issa, Musa Yilmaz, Naval Daver, Naveen Pemmaraju, Lucia Masarova, Farhad Ravandi, Nitin Jain, Wuliamatu Deen, Christopher Loiselle, Lourdes Waller, Glenda Banks, Rebecca Garris, and Hagop Kantarjian
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
6. P672: CHARACTERISTICS AND OUTCOMES OF PATIENTS WITH CHRONIC MYELOID LEUKEMIA AND T315I MUTATION AFTER FAILURE OF PRIOR THERAPIES
- Author
-
Aram Bidikian, Fadi Haddad, Koji Sasaki, Ghayas Issa, Tapan Kadia, Nitin Jain, Yesid Alvarado, Nicholas Short, Musa Yilmaz, Lucia Masarova, Elias Jabbour, and Hagop Kantarjian
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
7. P736: ANALYSIS OF RESPONSE RATES AND OUTCOMES IN ERYTHROID-PREDOMINANT MYELODYSPLASTIC SYNDROMES (MDS) TREATED WITH VENETOCLAX-BASED REGIMENS
- Author
-
Alexandre Bazinet, Naszrin Arani, Kelly Chien, Danielle Hammond, Tapan Kadia, Gautam Borthakur, Yesid Alvarado, Koji Sasaki, Courtney Dinardo, Naveen Pemmaraju, Lucia Masarova, Nicholas Short, Musa Yilmaz, Naval Daver, Elias Jabbour, Farhad Ravandi, Rashmi Kanagal-Shamanna, Sherry Pierce, Hagop Kantarjian, Guillermo Garcia-Manero, and Guillermo Montalban-Bravo
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
8. P1025: A PILOT STUDY OF THE ANTI-SLAMF7 MONOCLONAL ANTIBODY, ELOTUZUMAB, IN PATIENTS WITH MYELOFIBROSIS.
- Author
-
Prithviraj Bose, Lucia Masarova, Naveen Pemmaraju, Mackenzie Dobbins, Nitin Jain, Hussein Abbas, Steven Kornblau, Abhishek Maiti, Ivo Veletic, Taghi Manshouri, Sharon Bledsoe, Mary Ann Richie, Nakiuda Hall-Moore, Lingsha Zhou, Xuemei Wang, Hagop Kantarjian, Zeev Estrov, and Srdan Verstovsek
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
9. P1057: INDIRECT TREATMENT COMPARISONS OF MOMELOTINIB VS PACRITINIB SAFETY AND ANEMIA OUTCOMES IN PATIENTS WITH MYELOFIBROSIS
- Author
-
Lucia Masarova, Srdan Verstovsek, Francesca Palandri, Ruben Mesa, Claire Harrison, Balazs Dobi, Boris Gorsh, Zhaohui Wang, Catherine Ellis, Dwaipayan Patnaik, and Venediktos Kapetanakis
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
10. P1062: INDIRECT TREATMENT COMPARISON OF MOMELOTINIB VS FEDRATINIB SAFETY IN PATIENTS WITH MYELOFIBROSIS
- Author
-
Francesca Palandri, Lucia Masarova, Srdan Verstovsek, Ruben Mesa, Claire Harrison, Gautam Sajeev, Boris Gorsh, Ryan Simpson, Sang Cho, Zhaohui Wang, Catherine Ellis, Sean Conlon, and James Signorovitch
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
11. PB2184: IMPACT OF TRANSFUSION BURDEN ON HEALTH-RELATED QUALITY OF LIFE AND FUNCTIONING IN PATIENTS WITH MYELOFIBROSIS: POST HOC ANALYSIS OF SIMPLIFY-1 AND -2
- Author
-
Ruben Mesa, Francesca Palandri, Srdan Verstovsek, Lucia Masarova, Claire Harrison, Flora Mazerolle, Boris Gorsh, Manal M’hari, Zhaohui Wang, Catherine Ellis, Samineh Deheshi, Jun Kawashima, Robyn Von-Maltzahn, and Antoine Regnault
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
12. PB2183: TRANSFUSION-RELATED COST OFFSETS AND TIME BURDEN IN PATIENTS WITH MYELOFIBROSIS (MF) TREATED WITH MOMELOTINIB (MMB) VS DANAZOL (DAN)
- Author
-
Lucia Masarova, Srdan Verstovsek, Tom Liu, Sumati Rao, Guatam Sajeev, Ryan Simpson, Weilong LI, Joseph Yang, Boris Gorsh, and James Signorovitch
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
13. Efficacy and safety of enasidenib and azacitidine combination in patients with IDH2 mutated acute myeloid leukemia and not eligible for intensive chemotherapy
- Author
-
Sangeetha Venugopal, Koichi Takahashi, Naval Daver, Abhishek Maiti, Gautam Borthakur, Sanam Loghavi, Nicholas. J. Short, Maro Ohanian, Lucia Masarova, Ghayas Issa, Xuemei Wang, Bueso-Ramos Carlos, Musa Yilmaz, Tapan Kadia, Michael Andreeff, Farhad Ravandi, Marina Konopleva, Hagop M. Kantarjian, and Courtney D. DiNardo
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Preclinically, enasidenib and azacitidine (ENA + AZA) synergistically enhance cell differentiation, and venetoclax (VEN), a small molecule Bcl2 inhibitor (i) is particularly effective in IDH2 mutated acute myeloid leukemia (IDH2 mut AML). This open label phase II trial enrolled patients (pts) with documented IDH2 mut AML. All patients received AZA 75 mg/m2/d x 7 d/cycle and ENA 100 mg QD continuously. Concomitant Bcl2i and FLT3i were allowed (NCT03683433).Twenty-six pts received ENA + AZA (median 68 years, range, 24–88); 7 newly diagnosed (ND) and 19 relapsed/refractory (R/R). In R/R AML patients, three had received prior ENA and none had received prior VEN. The composite complete remission rate (CRc) [complete remission (CR) or complete remission with incomplete hematologic recovery (CRi)] was 100% in ND AML, and 58% in R/R AML. Median OS was not reached in ND AML with median follow-up of 13.1 months (mo); Pts treated in first relapse had improved OS than those with ≥2 relapse (median OS not reached vs 5.2 mo; HR 0.24, 95% CI 0.07–0.79, p = 0.04). Two patients received ENA + AZA with a concomitant FLT3i, one responding ND AML patient and one nonresponding R/R AML patient. Seven R/R AML pts received ENA + AZA + VEN triplet, and with median follow up of 11.2 mo, median OS was not reached and 6-mo OS was 70%. The most frequent treatment-emergent adverse events include febrile neutropenia (23%). Adverse events of special interest included all-grade IDH differentiation syndrome (8%) and indirect hyperbilirubinemia (35%). ENA + AZA was a well-tolerated, and effective therapy for elderly pts with IDH2 mut ND AML as well as pts with R/R AML. The addition of VEN to ENA + AZA appears to improve outcomes in R/R IDH2 mut AML. Clinical trial registration information: https://clinicaltrials.gov/.NCT03683433
- Published
- 2022
- Full Text
- View/download PDF
14. Efficacy of CDK9 inhibition in therapy of post-myeloproliferative neoplasm (MPN) secondary (s) AML cells
- Author
-
Warren Fiskus, Taghi Manshouri, Christine Birdwell, Christopher P. Mill, Lucia Masarova, Prithviraj Bose, Tapan M. Kadia, Naval Daver, Courtney D. DiNardo, Gautam Borthakur, Joseph D. Khoury, Srdan Verstovsek, and Kapil N. Bhalla
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2022
- Full Text
- View/download PDF
15. Extracellular volume quantification using synthetic haematocrit assessed from native and post-contrast longitudinal relaxation T1 times of a blood pool
- Author
-
Lukas Opatril, Roman Panovsky, Jan Machal, Tomas Holecek, Lucia Masarova, Vera Feitova, Vladimir Kincl, Marek Hodejovsky, and Lenka Spinarova
- Subjects
Extracellular volume ,ECV ,Synthetic haematocrit ,Cardiovascular magnetic resonance ,CMR ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
Abstract Background In terms of cardiovascular magnetic resonance are haematocrit values required for calculation of extracellular volume fraction (ECV). Previously published studies have hypothesized that haematocrit could be calculated from T1 blood pool relaxation time, however only native T1 relaxation time values have been used and the resulting formulae had been both in reciprocal and linear proportion. The aim of the study was to generate a synthetic haematocrit formula from only native relaxation time values first, calculate whether linear or reciprocal model is more precise in haematocrit estimation and then determine whether adding post-contrast values further improve its precision. Methods One hundred thirty-nine subjects underwent CMR examination. Haematocrit was measured using standard laboratory methods. Afterwards T1 relaxation times before and after the application of a contrast agent were measured and a statistical relationship between these values was calculated. Results Different linear and reciprocal models were created to estimate the value of synthetic haematocrit and ECV. The highest coefficient of determination was observed in the combined reciprocal model “− 0.047 + (779/ blood native) − (11.36/ blood post-contrast)”. Conclusions This study provides more evidence that assessing synthetic haematocrit and synthetic ECV is feasible and statistically most accurate model to use is reciprocal. Adding post-contrast values to the calculation was proved to improve the precision of the formula statistically significantly.
- Published
- 2021
- Full Text
- View/download PDF
16. Superior efficacy of co-targeting GFI1/KDM1A and BRD4 against AML and post-MPN secondary AML cells
- Author
-
Warren Fiskus, Christopher P. Mill, Behnam Nabet, Dimuthu Perera, Christine Birdwell, Taghi Manshouri, Bernardo Lara, Tapan M. Kadia, Courtney DiNardo, Koichi Takahashi, Naval Daver, Prithviraj Bose, Lucia Masarova, Naveen Pemmaraju, Steven Kornblau, Gautam Borthakur, Guillermo Montalban-Bravo, Guillermo Garcia Manero, Sunil Sharma, Matthew Stubbs, Xiaoping Su, Michael R. Green, Cristian Coarfa, Srdan Verstovsek, Joseph D. Khoury, Christopher R. Vakoc, and Kapil N. Bhalla
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract There is an unmet need to overcome nongenetic therapy-resistance to improve outcomes in AML, especially post-myeloproliferative neoplasm (MPN) secondary (s) AML. Studies presented describe effects of genetic knockout, degradation or small molecule targeted-inhibition of GFI1/LSD1 on active enhancers, altering gene-expressions and inducing differentiation and lethality in AML and (MPN) sAML cells. A protein domain-focused CRISPR screen in LSD1 (KDM1A) inhibitor (i) treated AML cells, identified BRD4, MOZ, HDAC3 and DOT1L among the codependencies. Our findings demonstrate that co-targeting LSD1 and one of these co-dependencies exerted synergistic in vitro lethality in AML and post-MPN sAML cells. Co-treatment with LSD1i and the JAKi ruxolitinib was also synergistically lethal against post-MPN sAML cells. LSD1i pre-treatment induced GFI1, PU.1 and CEBPα but depleted c-Myc, overcoming nongenetic resistance to ruxolitinib, or to BETi in post-MPN sAML cells. Co-treatment with LSD1i and BETi or ruxolitinib exerted superior in vivo efficacy against post-MPN sAML cells. These findings highlight LSD1i-based combinations that merit testing for clinical efficacy, especially to overcome nongenetic therapy-resistance in AML and post-MPN sAML.
- Published
- 2021
- Full Text
- View/download PDF
17. Reccurent thrombus in the gigantic left atrium during effective anticoagulant therapy: case report
- Author
-
Lucia Masarova, Jan Novak, Martin Pesl, Jiri Ondrasek, Jiri Semenka, Eva Simarova, and Roman Panovsky
- Subjects
Cardiac magnetic resonance ,Echocardiography ,Atrial fibrillation ,Recurrent thrombus ,Gigantic left atrium ,Anticoagulant therapy ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
Abstract Background Gigantic left atrium is defined in the current literature as an excessive dilatation of the left atrium above 65mm. Chronic mitral valve disease is associated with the development of thrombus in the left atrium in up to 19% of all cases of mitral insufficiency and appropriate treatment must be initiated to prevent thromboembolic events. In order to diagnose thrombi in the left atrium or left atrial appendage, various imaging methods may be used, including cardiac magnetic resonance. Case presentation The case report describes a 73-year-old male who developed recurrent sessile thrombus on the posterior wall of the gigantic left atrium. A large thrombus was first detected following mitral valve surgery despite effective vitamin K antagonist anticoagulation therapy. Echocardiography and cardiac magnetic resonance were used within the diagnostic procedure and to monitor the treatment outcomes. Cardiac magnetic resonance was shown to be beneficial as it provided a more precise description of the intra-atrial masses located on the posterior left atrial wall, and in such situations, is of greater benefit than standard echocardiography. This led to the surgical removal of the intra-atrial mass; nevertheless, it was quickly followed by the recurrence of the thrombus. The anticoagulant therapy was adjusted and fortified by the introduction of acetylsalicylic acid and sequentially clopidogrel, but this also did not resolve the thrombus formation. Finally, employing a combination of rivaroxaban and clopidogrel resulted in partial thrombus regression. Therefore, various pathophysiological aspects of thrombus formation and used anticoagulation strategies are discussed. Conclusions We describe a unique case of a recurrent thrombus located on the posterior wall of the gigantic left atrium. Cardiac magnetic resonance was shown to be beneficial in providing a more precise description of the intra-atrial masses located on the posterior left atrial wall as compared to standard echocardiographic examination. Development of a thrombus after mitral valve surgery despite effective anticoagulant therapy and its final resolution by introducing a combination of rivaroxaban and clopidogrel highlights the complex etiopathogenesis of thrombus formation. This supports the potential use of this combination in tailoring an individual personalized therapy for patients with recurrent atrial thrombi.
- Published
- 2020
- Full Text
- View/download PDF
18. The effect of eltrombopag in managing thrombocytopenia associated with tyrosine kinase therapy in patients with chronic myeloid leukemia and myelofibrosis
- Author
-
Mahran Shoukier, Gautam Borthakur, Elias Jabbour, Farhad Ravandi, Guillermo Garcia-Manero, Tapan Kadia, Jairo Matthews, Lucia Masarova, Kiran Naqvi, Koji Sasaki, Srdan Verstovsek, and Jorge Cortes
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Approximately 20-50% patients with chronic phase chronic myeloid leukemia (CML-CP) treated with tyrosine kinase inhibitors (TKI) or with myelofibrosis (MF) treated with ruxolitinib develop grade ≥3 thrombocytopenia needing treatment interruptions and dose reductions. We conducted a non-randomized, phase II, single-arm study to determine the efficacy of eltrombopag for patients with CML or MF with persistent thrombocytopenia while on TKI or ruxolitinib. Eltrombopag was initiated at 50 mg/day, with dose escalation up to 300 mg daily allowed every 2 weeks. Twenty-one patients were enrolled (CML=15, MF=6); with a median age of 60 years (range, 31-97 years). The median platelet count was 44x109/L (range, 3-49x109/L) in CML and 62x109/L (range, 21-75x109/L) in MF. After a median of 18 months (range, 5-77 months), 12 of 15 patients with CML achieved complete platelet response. The median peak platelet count among responders was 154x109/L (range, 74-893x109/L). Among CML patients five could re-escalate the TKI dose and nine improved their response. None of the six patients with MF had a sustained response. Therapy was generally well tolerated. One patient discontinued therapy due to toxicity (elevated transaminases). One patient with CML developed significant thrombocytosis (>1,000x109/L). Another CML patient developed non occlusive deep venous thrombosis in the right upper extremity without thrombocytosis, and one MF patient had myocardial infarction. Eltrombopag may help improve platelet counts in CML patients receiving TKI with recurrent thrombocytopenia. Further studies are warranted (clinicaltrials gov. Identifier: NCT01428635).
- Published
- 2020
- Full Text
- View/download PDF
19. Outcomes of relapsed or refractory acute myeloid leukemia after frontline hypomethylating agent and venetoclax regimens
- Author
-
Abhishek Maiti, Caitlin R. Rausch, Jorge E. Cortes, Naveen Pemmaraju, Naval G. Daver, Farhad Ravandi, Guillermo Garcia-Manero, Gautam Borthakur, Kiran Naqvi, Maro Ohanian, Nicholas J. Short, Yesid Alvarado, Tapan M. Kadia, Koichi Takahashi, Musa Yilmaz, Nitin Jain, Steven Kornblau, Guillermo Montalban Bravo, Koji Sasaki, Michael Andreeff, Prithiviraj Bose, Alessandra Ferrajoli, Ghayas C. Issa, Elias J. Jabbour, Lucia Masarova, Philip A. Thompson, Sa Wang, Sergej Konoplev, Sherry A. Pierce, Jing Ning, Wei Qiao, John S. Welch, Hagop M. Kantarjian, Courtney D. DiNardo, and Marina Y. Konopleva
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2020
- Full Text
- View/download PDF
20. Histomorphological responses after therapy with pegylated interferon α-2a in patients with essential thrombocythemia (ET) and polycythemia vera (PV)
- Author
-
Lucia Masarova, C. Cameron Yin, Jorge E. Cortes, Marina Konopleva, Gautam Borthakur, Kate J. Newberry, Hagop M. Kantarjian, Carlos E. Bueso-Ramos, and Srdan Verstovsek
- Subjects
Essential thrombocythemia ,Polycythemia vera ,Pegylated interferon alfa-2a ,Histomorphological response ,Diseases of the blood and blood-forming organs ,RC633-647.5 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Pegylated interferon alfa-2a (PEG-IFN-α-2a) is a potent immunomodulating agent capable of inducing high rate of hematologic and even complete molecular remission in patients with essential thrombocythemia (ET) and polycythemia vera (PV). We recently reported results of a phase 2 trial of PEG-IFN-α-2a in 83 patients with ET and PV after a median follow-up of 83 months. Here we report an analysis of bone marrow (BM) responses in these patients. Methods Among 83 patients, 58 (70%, PV 25, ET 31) had evaluable BM samples. BM responses and fibrosis grading were defined according to the International Working Group for Myeloproliferative Neoplasms Research and Treatment, and the European Consensus on grading of BM fibrosis, respectively. BM was assessed prior to enrollment, and every 6–24 months while on therapy in all patients, and after therapy discontinuation in some patients. Results The median age of analyzed 58 patients was 52 years, and 29% were males. After a median follow-up of 84 months, 32 patients are still on study. Hematologic (HR) and molecular responses (MR) were seen in 93 and 69% patients, respectively. Twenty-nine patients (50%) had a BM response, including 13 (22%) with a complete BM response (BM-CR). Moreover, 13 patients (22%) have experienced complete resolution of bone marrow reticulin fibrosis. Patients with BM response had higher duration of HR and MR, and lower discontinuation rate. Furthermore, patients with BM-CR had a higher probability of complete MR. The median duration of BM-CR was 30 months, and 9 patients have maintained their BM-CR (69%), including five who have maintained their response after discontinuation of therapy. Despite this observation, the pattern of HR, MR and BM response, their durability and interrelation was heterogeneous. Conclusions Our results show the ability of PEG-IFN-α-2a to induce complete BM responses in a subset of ET and PV patients, but its correlation with durable clinically relevant treatment benefit warrants further investigation. Trial registration This study is registered with ClinicalTrials.gov (NCT00452023), and is ongoing but not enrolling new patients.
- Published
- 2017
- Full Text
- View/download PDF
21. Therapy-related myelofibrosis does not appear to exist
- Author
-
Lucia Masarova, Gabriele Todisco, Taghi Manshouri, Kate J. Newberry, Jorge E. Cortes, Hagop M. Kantarjian, Zeev Estrov, and Srdan Verstovsek
- Subjects
Specialties of internal medicine ,RC581-951 - Published
- 2017
- Full Text
- View/download PDF
22. Targeted therapy with the mutant IDH2 inhibitor enasidenib for high-risk IDH2-mutant myelodysplastic syndrome
- Author
-
Courtney D. DiNardo, Sangeetha Venugopal, Curtis Lachowiez, Koichi Takahashi, Sanam Loghavi, Guillermo Montalban-Bravo, Xuemei Wang, Hetty Carraway, Mikkael Sekeres, Ameenah Sukkur, Danielle Hammond, Kelly Chien, Abhishek Maiti, Lucia Masarova, Koji Sasaki, Yesid Alvarado, Tapan Kadia, Nicholas J. Short, Naval Daver, Gautam Borthakur, Farhad Ravandi, Hagop M. Kantarjian, Bhumika Patel, Amy Dezern, Gail Roboz, and Guillermo Garcia-Manero
- Subjects
Hematology - Abstract
The isocitrate dehydrogenase enzyme 2 (IDH2) gene is mutated in ∼5% of patients with myelodysplastic syndrome (MDS). Enasidenib is an oral, selective, mutant IDH2 inhibitor approved for IDH2-mutated (mIDH2) relapsed/refractory acute myeloid leukemia. We designed a 2-arm multicenter study to evaluate safety and efficacy of (A) the combination of enasidenib with azacitidine for newly diagnosed mIDH2 MDS, and (B) enasidenib monotherapy for mIDH2 MDS after prior hypomethylating agent (HMA) therapy. Fifty patients with mIDH2 MDS enrolled: 27 in arm A and 23 in arm B. Median age of patients was 73 years. The most common adverse events were neutropenia (40%), nausea (36%), constipation (32%), and fatigue (26%). Hyperbilirubinemia from off-target UGT1A1 inhibition occurred in 14% of patients (8%; grades 3 and 4), and IDH-inhibitor–associated differentiation syndrome (IDH-DS) in 8 patients (16%). In the combination arm, the overall response rate (ORR: complete remission [CR] + marrow CR [mCR] + partial remission) was 74%, including 70% composite CR (CRc: CR + mCR). Median time to best response was 1 month (range, 1-4), and a median of 4 cycles was received (1-32). The median overall survival (OS) was 26 months (range, 14 to not reached). In the enasidenib monotherapy cohort after HMA failure, ORR and CRc were both 35% (n = 8), with 22% CR (n = 5). Median time to first response was 27 days, and time to best response was 4.6 months (2.7-7.6 months). A median of 7 cycles was received (range, 1-29), and the median OS was 20 months (range, 11 to not reached). Enasidenib is an effective treatment option for mIDH2 MDS, both in combination with azacitidine for treatment-naïve high-risk MDS, and as a single agent after prior HMA therapy. This trial is registered at www.clinicaltrials.gov as #NCT03383575.
- Published
- 2023
23. SOHO State of the Art Updates and Next Questions: Novel Therapeutic Strategies in Development for Myelofibrosis
- Author
-
Lucia Masarova, Srdan Verstovsek, MD, PhD, Professor of Medicine, and Helen Chifotides, PhD, ELS, Senior Research Scientist and Medical/Scientific Writer
- Subjects
Cancer Research ,Oncology ,Hematology - Published
- 2023
24. Clinical outcomes associated with NPM1 mutations in patients with relapsed or refractory AML
- Author
-
Ghayas C. Issa, Aram Bidikian, Sangeetha Venugopal, Marina Konopleva, Courtney D. DiNardo, Tapan M. Kadia, Gautam Borthakur, Elias Jabbour, Naveen Pemmaraju, Musa Yilmaz, Nicholas J. Short, Abhishek Maiti, Koji Sasaki, Lucia Masarova, Sherry Pierce, Koichi Takahashi, Guilin Tang, Sanam Loghavi, Keyur Patel, Michael Andreeff, Kapil Bhalla, Guillermo Garcia-Manero, Farhad Ravandi, Hagop Kantarjian, and Naval Daver
- Subjects
Hematology - Abstract
Mutations in Nucleophosmin 1 (NPM1) are associated with a favorable prognosis in newly diagnosed acute myeloid leukemia (AML), however, their prognostic impact in relapsed/refractory (R/R) settings are unknown. In a retrospective analysis, we identified 206 patients (12%) with mutated NPM1 (NPM1c) and compared their outcomes to 1516 patients (88%) with NPM1 wild-type (NPM1wt). NPM1c was associated with higher rates of complete remission or complete remission with incomplete count recovery compared with NPM1wt following each line of salvage therapy (first salvage, 56% vs 37%; P < .0001; second salvage, 33% vs 22%; P = .02; third salvage, 24% vs 14%; P = .02). However, NPM1 mutations had no impact on relapse-free survival (RFS) and overall survival (OS) with each salvage therapy with a median OS following salvage 1, 2 or 3 therapies in NPM1c vs NPM1wt of 7.8 vs 6.0; 5.3 vs 4.1; and 3.5 vs 3.6 months, respectively. Notably, the addition of venetoclax to salvage regimens in patients with NPM1c improved RFS and OS (median RFS, 15.8 vs 4.6 months; P = .05; median OS, 14.7 vs 5.9 months; P = .02). In conclusion, NPM1 mutational status has a minimal impact on prognosis in relapsed or refractory AML; therefore, novel treatment strategies are required to improve outcomes in this entity.
- Published
- 2023
25. Long-Term Outcomes of Acute Myeloid Leukemia Following Allogeneic Stem-Cell Transplantation after Decitabine and Venetoclax: Subgroup Analysis of a Phase II Trial
- Author
-
Abhishek Maiti, Courtney D. DiNardo, Maro Ohanian, Naveen Pemmaraju, Naval Daver, Gautam Borthakur, Farhad Ravandi, Ghayas C. Issa, Guillermo Garcia-Manero, Nitin Jain, Tapan M. Kadia, Lucia Masarova, William G. Wierda, Musa Yilmaz, Yesid Alvarado, Elias Jabbour, Nicholas Short, Koichi Takahashi, Sanam Loghavi, Keyur P. Patel, Kathryn Montalbano, Sherry A. Pierce, Carol A. Bivins, Rohtesh S. Mehta, Gheath Alatrash, Uday R Popat, Partow Kebriaei, Betul Oran, Elizabeth J Shpall, Richard E Champlin, Hagop Kantarjian, and Marina Konopleva
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
26. Updated Results of CPX-351 in Combination with Gemtuzumab Ozogamicin (GO) in Relapsed Refractory (R/R) Acute Myeloid Leukemia (AML) and Post-Hypomethylating Agent (Post-HMA) Failure High-Risk Myelodysplastic Syndrome (HR-MDS)
- Author
-
Jayastu Senapati, Emmanuel Huante Almanza, Tapan M. Kadia, Guillermo Montalban-Bravo, Stefan Faderl, Koji Sasaki, Nicholas Short, Naval Daver, Courtney D. DiNardo, Lucia Masarova, Alessandra Ferrajoli, Elias Jabbour, Gautam Borthakur, Hind Al Azzawi, Marina Konopleva, Guillermo Garcia-Manero, Michael Andreeff, Hagop Kantarjian, Farhad Ravandi, and Yesid Alvarado
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
27. Impact of SF3B1 mutation in myelofibrosis
- Author
-
Jayastu Senapati, Srdan Verstovsek, Lucia Masarova, Naveen Pemmaraju, Keyur P. Patel, Guillermo Montalban-Bravo, Sherry A. Pierce, Lingsha Zhou, Guillermo Garcia-Manero, Hagop M. Kantarjian, and Prithviraj Bose
- Subjects
Cancer Research ,Oncology ,Primary Myelofibrosis ,Mutation ,Humans ,RNA Splicing Factors ,Hematology ,Phosphoproteins - Published
- 2022
28. The role of therapy in the outcome of patients with myelofibrosis
- Author
-
Lucia Masarova, Prithviraj Bose, Naveen Pemmaraju, Naval G. Daver, Koji Sasaki, Helen T. Chifotides, Lingsha Zhou, Hagop M. Kantarjian, Zeev Estrov, and Srdan Verstovsek
- Subjects
Cancer Research ,Oncology - Published
- 2023
29. Low-Dose Dasatinib (50mg Daily) Frontline Therapy in Newly Diagnosed Chronic Phase Chronic Myeloid Leukemia: 5-Year Follow-up Results
- Author
-
Georgina Gener-Ricos, Fadi G. Haddad, Koji Sasaki, Ghayas C. Issa, Jeffrey Skinner, Lucia Masarova, Gautam Borthakur, Yesid Alvarado, Guillermo Garcia-Manero, Elias Jabbour, and Hagop Kantarjian
- Subjects
Cancer Research ,Oncology ,Hematology - Published
- 2023
30. A phase Ib/II study of ivosidenib with venetoclax +/- azacitidine in IDH1-mutated myeloid malignancies
- Author
-
Curtis A. Lachowiez, Sanam Loghavi, Zhihong Zeng, Tomoyuki Tanaka, Yi June Kim, Hidetaka Uryu, Sven Turkalj, Niels Asger Jakobsen, Marlise R. Luskin, Dzifa Y. Duose, Rebecca S S. Tidwell, Nicholas J. Short, Gautam Borthakur, Tapan M. Kadia, Lucia Masarova, George D. Tippett, Prithviraj Bose, Elias J. Jabbour, Farhad Ravandi, Naval G. Daver, Guillermo Garcia-Manero, Hagop Kantarjian, Jacqueline S. Garcia, Paresh Vyas, Koichi Takahashi, Marina Konopleva, and Courtney D. DiNardo
- Subjects
General Medicine - Abstract
The safety and efficacy of combining the IDH1 inhibitor ivosidenib (IVO) with the BCL2 inhibitor venetoclax (VEN; IVO+VEN) +/- azacitidine (AZA; IVO+VEN+AZA) was evaluated in four cohorts of patients with IDH1-mutated myeloid malignancies (n=31). Most (91%) adverse events were grade 1 or 2. The maximal tolerated dose was not reached. Composite complete remission with IVO+VEN+AZA vs. IVO+VEN was 90% vs. 83%. Among MRD-evaluable patients (N=16) 63% attained MRD-negative remissions; IDH1 mutation clearance occurred in 64% of patients receiving ≥5 treatment cycles (N=14). Median EFS and OS were 36 (95% CI: 23-NR) and 42 (95% CI: 42-NR) months. Patients with signaling gene mutations appeared to particularly benefit from the triplet regimen. Longitudinal single-cell proteogenomic analyses linked co-occurring mutations, anti-apoptotic protein expression, and cell maturation to therapeutic sensitivity of IDH1-mutated clones. No IDH isoform switching or second-site IDH1 mutations were observed, indicating combination therapy may overcome established resistance pathways to single-agent IVO.
- Published
- 2023
31. Updated Phase IIb Results of Venetoclax with FLAG-IDA in Relapsed or Refractory Acute Myeloid Leukemia
- Author
-
Sai Prasad Desikan, Marina Konopleva, Koichi Takahashi, Curtis A Lachowiez, Sanam Loghavi, Lian-Chun Xiao, Tapan M. Kadia, Naval Daver, Nicholas Short, Koji Sasaki, Gautam Borthakur, Ghayas C. Issa, Abhishek Maiti, Kelly S. Chien, Yesid Alvarado, Guillermo Montalban-Bravo, Lucia Masarova, Musa Yilmaz, Michael Andreeff, Elias Jabbour, Guillermo Garcia-Manero, Steven M. Kornblau, Farhad Ravandi, Hagop Kantarjian, and Courtney D. DiNardo
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
32. A Phase II Study of the Sequential Combination of Low-Intensity Chemotherapy (mini-hyper-CVD) and Ponatinib Followed By Blinatumomab and Ponatinib in Patients with Philadelphia Chromosome-Positive (Ph+) Acute Lymphoblastic Leukemia (ALL)
- Author
-
Daniel Nguyen, Elias Jabbour, Nicholas Short, Ghayas C. Issa, Musa Yilmaz, Naval Daver, Naveen Pemmaraju, Kelly S. Chien, Lucia Masarova, Farhad Ravandi, Nitin Jain, Wuliamatu Deen, Min Zhao, Christopher Loiselle, Lourdes Waller, Glenda Banks, Rebecca Garris, and Hagop Kantarjian
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
33. Phase 2 Study of ASTX727 (cedazuridine/decitabine) Plus Venetoclax in Patients with Relapsed/Refractory Acute Myeloid Leukemia (AML) or Previously Untreated, Elderly Patients with AML Unfit for Chemotherapy
- Author
-
Tareq Abuasab, Guillermo Garcia-Manero, Nicholas Short, Yesid Alvarado, Ghayas C. Issa, Rubiul Islam, Abhishek Maiti, Musa Yilmaz, Nitin Jain, Lucia Masarova, Steven M. Kornblau, Elias Jabbour, Naveen Pemmaraju, Guillermo Montalban-Bravo, Sherry A. Pierce, Courtney D. DiNardo, Tapan M. Kadia, Naval Daver, Marina Konopleva, Hagop Kantarjian, and Farhad Ravandi
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
34. Venetoclax Combined with Cladribine, Idarubicin, Cytarabine (CLIA) As Induction Therapy in Patients with Newly Diagnosed Acute Myeloid Leukemia and High-Risk Myelodysplastic Syndrome
- Author
-
Patrick K Reville, Hagop Kantarjian, Gautam Borthakur, Musa Yilmaz, Naval Daver, Nicholas Short, Courtney D. DiNardo, Steven M. Kornblau, Naveen Pemmaraju, Nitin Jain, Yesid Alvarado, Prithviraj Bose, Elias Jabbour, Kelly S. Chien, Hussein A Abbas, Lucia Masarova, Sa A Wang, Rebecca S. S Tidwell, Michael Andreeff, Guillermo Garcia-Manero, Marina Konopleva, Farhad Ravandi, and Tapan M. Kadia
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
35. A Phase II Study of Inotuzumab Ozogamicin for the Treatment of Measurable Residual Disease-Positive B-Cell Acute Lymphoblastic Leukemia
- Author
-
Jayastu Senapati, Nicholas Short, Yesid Alvarado, Jan A. Burger, Nitin Jain, Marina Konopleva, Farhad Ravandi, Courtney D. DiNardo, Lucia Masarova, Koji Sasaki, Philip A. Thompson, Alessandra Ferrajoli, Jovitta Jacob, Ejiroghene Mayor, Anna Milton, Christopher Loiselle, Rebecca Garris, Hagop Kantarjian, and Elias Jabbour
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
36. Phase I Study of Ruxolitinib in Combination with Abemaciclib for Patients with Primary or Post-Polycythemia Vera/Essential Thrombocythemia Myelofibrosis
- Author
-
Jan Philipp Bewersdorf, Srdan Verstovsek, Andriy Derkach, Lucia Masarova, Naveen Pemmaraju, Eytan Stein, Michael Mauro, Raajit K Rampal, and Prithviraj Bose
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
37. The Frequency and Clinical Implications of ASXL2 Mutations in Myeloid Neoplasms
- Author
-
Tareq Abuasab, Srdan Verstovsek, Gautam Borthakur, Rashmi Kanagal-Shamanna, Keyur Patel, Koichi Takahashi, Lucia Masarova, Prithviraj Bose, John Villarreal, Sherry A. Pierce, Guillermo Montalban-Bravo, Guillermo Garcia-Manero, Nicholas Short, Courtney D. DiNardo, Naval Daver, Farhad Ravandi, Hagop Kantarjian, and Musa Yilmaz
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
38. A multi-arm phase Ib/II study designed for rapid, parallel evaluation of novel immunotherapy combinations in relapsed/refractory acute myeloid leukemia
- Author
-
Nicholas J. Short, Gautam Borthakur, Naveen Pemmaraju, Courtney D. Dinardo, Tapan M. Kadia, Elias Jabbour, Marina Konopleva, Walid Macaron, Jing Ning, Junsheng Ma, Sherry Pierce, Yesid Alvarado, Koji Sasaki, Koichi Takahashi, Zeev Estrov, Lucia Masarova, Ghayas C. Issa, Guillermo Montalban-Bravo, Michael Andreeff, Jan A. Burger, Darla Miller, Lynette Alexander, Aung Naing, Guillermo Garcia-Manero, Farhad Ravandi, and Naval Daver
- Subjects
Cohort Studies ,Leukemia, Myeloid, Acute ,Sulfonamides ,Cancer Research ,Oncology ,Antineoplastic Combined Chemotherapy Protocols ,Azacitidine ,Antibodies, Monoclonal ,Humans ,Immunotherapy ,Hematology ,Bridged Bicyclo Compounds, Heterocyclic ,Gemtuzumab - Abstract
We conducted a phase Ib/II multi-arm, parallel cohort study to simultaneously evaluate various immunotherapeutic agents and combinations in relapsed/refractory acute myeloid leukemia (AML). Overall, 50 patients were enrolled into one of 6 arms: (A) single agent PF-04518600 (OX40 agonist monoclonal antibody), (B) azacitidine + venetoclax + gemtuzumab ozogamicin (GO), (C) azacitidine + avelumab (anti-PD-L1 monoclonal antibody) + GO, (D) azacitidine + venetoclax + avelumab, (E) azacitidine + avelumab + PF-04518600, and (F) glasdegib + GO. Among all regimens evaluated, azacitidine + venetoclax + GO appeared most promising. In this arm, the CR/CRi rates among venetoclax-naïve and prior venetoclax-exposed patients were 50% and 22%, respectively, and the 1-year OS rate was 31%. This study shows the feasibility of a conducting a multi-arm trial to efficiently and simultaneously evaluate novel therapies in AML, a needed strategy in light of the plethora of emerging therapies. This trial was registered at www.clinicaltrials.gov as NCT03390296.
- Published
- 2022
39. SOHO State of the Art Updates and Next Questions: Novel Therapies in Development for Myelofibrosis
- Author
-
Helen T. Chifotides, Prithviraj Bose, Naveen Pemmaraju, Srdan Verstovsek, and Lucia Masarova
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Ruxolitinib ,Anemia ,Epigenesis, Genetic ,Imetelstat ,chemistry.chemical_compound ,Internal medicine ,medicine ,Humans ,Janus Kinase Inhibitors ,Myelofibrosis ,Protein Kinase Inhibitors ,Myeloproliferative Disorders ,Navitoclax ,business.industry ,Hematology ,Janus Kinase 2 ,medicine.disease ,Clinical trial ,Pacritinib ,chemistry ,Primary Myelofibrosis ,business ,Progressive disease ,medicine.drug - Abstract
Myeloproliferative neoplasms research has entered a dynamic and exciting era as we witness exponential growth of novel agents in advanced/early phase clinical trials for myelofibrosis (MF). Building on the success and pivotal role of ruxolitinib, many novel agents, spanning a wide range of mechanisms/targets (epigenetic regulation, apoptotic/intracellular signaling pathways, telomerase, bone marrow fibrosis) are in clinical development; several are studied in registrational trials and hold great potential to expand the therapeutic arsenal/shift the treatment paradigm if regulatory approval is granted. Insight into MF pathogenesis and its molecular underpinnings, preclinical studies demonstrating synergism of ruxolitinib with investigational agents, urgent unmet clinical needs (cytopenias, loss of response to JAK inhibitors); and progressive disease fueled the rapid rise of innovative therapeutics. New strategies include pairing ruxolitinib with erythroid maturation agents to manage anemia (luspatercept), designing rational combinations with ruxolitinib to boost responses in both the frontline and suboptimal response settings (pelabresib, navitoclax, parsaclisib), treatment with non-JAK inhibitor monotherapy in the second-line setting (navtemadlin, imetelstat), novel JAK inhibitors tailored to subgroups with challenging unmet needs (momelotinib and pacritinib for anemia and thrombocytopenia, respectively); and agents potentially enhancing longevity (imetelstat). Beyond typical endpoints evaluated in MF clinical trials (spleen volume reduction ≥ 35%, total symptom score reduction ≥ 50%) thus far, emerging endpoints include overall survival, progression-free survival, transfusion independence, anemia benefits, bone marrow fibrosis and driver mutation allele burden reduction. Novel biomarkers and additional clinical features are being sought to assess new agents and tailor emerging therapies to appropriate patients. New strategies are needed to optimize the design of clinical trials comparing novel combinations to standard agent monotherapy.
- Published
- 2022
40. Prediction of early (4‐week) mortality in acute myeloid leukemia with intensive chemotherapy
- Author
-
Farhad Ravandi, Nitin Jain, Naval Daver, Courtney D. DiNardo, Guillermo Garcia-Manero, Kebede H. Begna, Hagop M. Kantarjian, Guillermo Montalban Bravo, Nicholas J. Short, Ayalew Tefferi, Tapan M. Kadia, Lucia Masarova, Sherry Pierce, Gautam Borthakur, Koji Sasaki, Elias Jabbour, and Marina Konopleva
- Subjects
Adult ,Male ,medicine.medical_specialty ,Population ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Hyperuricemia ,education ,Aged ,Retrospective Studies ,education.field_of_study ,Performance status ,business.industry ,Mortality rate ,Organ dysfunction ,Myeloid leukemia ,Hematology ,Middle Aged ,Prognosis ,medicine.disease ,Leukemia, Myeloid, Acute ,Multivariate Analysis ,Cohort ,Cytarabine ,Female ,medicine.symptom ,business ,medicine.drug - Abstract
The progress with intensive chemotherapy and supportive care measures has improved survival in patients with newly diagnosed acute myeloid leukemia (AML). Given the recent development of effective low intensity therapies, an optimal decision on the therapy intensity may improve survival through the avoidance of early mortality. We reviewed the outcome of 3728 patients with newly diagnosed AML who received intensive chemotherapy between August 1980 and May 2020. Intensive chemotherapy was defined as a cumulative cytarabine dose ≥ 700 mg/m2 during induction therapy. We divided the whole cohort into a training and validation group at a 3:1 ratio. The population was divided into a training (2790 patients) and a validation cohort (938 patients). The median age was 55 years (range, 15-99). Among them, 442 patients (12%) had core-binding factor AML. Binary logistic regression identified older age, worse performance status, hyperbilirubinemia, elevated creatinine, hyperuricemia, cytogenetic abnormalities other than CBF and -Y, and pneumonia as adverse prognostic factors for an early 4-week mortality. This risk classification for early mortality was verified in the validation cohort of patients. In the validation cohort of more recently treated patients from 2000 to 2017, the 4-week mortality rates with intensive chemotherapy were 2%, 14%, and 50% in the low-, high-, and very high-risk group, respectively. The mortality rates with low intensity therapies were 3%, 9%, and 20%, respectively. The risk classification guides treatment intensity by the assessment of age, frailty, organ dysfunction, cytogenetic abnormality, and infection to avoid early mortality.
- Published
- 2021
41. Venetoclax Combined With FLAG-IDA Induction and Consolidation in Newly Diagnosed and Relapsed or Refractory Acute Myeloid Leukemia
- Author
-
Koichi Takahashi, Nitin Jain, Guillermo Garcia-Manero, Lucia Masarova, Sa Wang, Courtney D. DiNardo, Naveen Pemmaraju, Guillermo Montalban Bravo, Tapan M. Kadia, Sanam Loghavi, Musa Yilmaz, Ghayas C. Issa, Koji Sasaki, Elias Jabbour, Steven M. Kornblau, Abhishek Maiti, Marina Konopleva, Lianchun Xiao, Curtis Lachowiez, Maria Adeoti, Farhad Ravandi, Michael Andreeff, Nicholas J. Short, Gautam Borthakur, Hagop M. Kantarjian, Naval Daver, and Yesid Alvarado
- Subjects
Adult ,Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Myeloid ,Maximum Tolerated Dose ,Young Adult ,chemistry.chemical_compound ,Refractory ,hemic and lymphatic diseases ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Granulocyte Colony-Stimulating Factor ,Humans ,Medicine ,Neoplasm ,Survival rate ,Aged ,Sulfonamides ,business.industry ,Venetoclax ,Remission Induction ,Cytarabine ,Hematopoietic Stem Cell Transplantation ,Myeloid leukemia ,ORIGINAL REPORTS ,Middle Aged ,Bridged Bicyclo Compounds, Heterocyclic ,Prognosis ,medicine.disease ,Survival Rate ,Leukemia, Myeloid, Acute ,Leukemia ,medicine.anatomical_structure ,chemistry ,Drug Resistance, Neoplasm ,FLAG (chemotherapy) ,Female ,Neoplasm Recurrence, Local ,Idarubicin ,business ,Vidarabine ,Follow-Up Studies - Abstract
PURPOSE Sixty percent of newly diagnosed patients with acute myeloid leukemia (ND-AML) receiving frontline therapy attain a complete response (CR), yet 30%-40% of patients relapse. Relapsed or refractory AML (R/R-AML) remains a particularly adverse population necessitating improved therapeutic options. This phase Ib/II study evaluated the safety and efficacy of fludarabine, cytarabine, granulocyte colony-stimulating factor, and idarubicin combined with the B-cell lymphoma-2 inhibitor venetoclax in ND-AML and R/R-AML. MATERIALS AND METHODS The phase IB portion (PIB) enrolled patients with R/R-AML using a 3 + 3 dose escalation and de-escalation algorithm for identification of maximum tolerated dose and dose-limiting toxicities. The phase II portion enrolled patients into two arms to evaluate response and time-to-event end points: phase IIA (PIIA): ND-AML and phase IIB (PIIB): R/R-AML. RESULTS Sixty-eight patients have enrolled to date (PIB, 16; PIIA, 29; PIIB, 23). Median age was 46 years (range, 20-73). Grade 3 and 4 adverse events occurring in ≥ 10% of patients included febrile neutropenia (50%), bacteremia (35%), pneumonia (28%), and sepsis (12%). The overall response rate for PIB, PIIA, and PIIB was 75%, 97%, and 70% with 75%, 90%, and 61%, respectively, achieving a composite CR. Measurable residual disease–negative composite CR was attained in 96% of ND-AML and 69% of R/R-AML patients. After a median follow-up of 12 months, median overall survival (OS) for both PII cohorts was not reached. Fifty-six percent of patients proceeded to allogeneic hematopoietic stem-cell transplantation (ND-AML, 69%; R/R-AML, 46%). In R/R-AML, allogeneic hematopoietic stem-cell transplantation resulted in a significant improvement in OS (median OS, NR; 1-year OS, 87%). One-year survival post-HSCT was 94% in ND-AML and 78% in R/R-AML. CONCLUSION Fludarabine, cytarabine, granulocyte colony-stimulating factor, and idarubicin + venetoclax represents an effective intensive treatment regimen in ND-AML and R/R-AML patients, associated with deep remissions and a high rate of transition to successful transplantation.
- Published
- 2021
42. EXABS-116-MPN Extended Abstract: Novel Therapies for PV
- Author
-
Lucia Masarova
- Subjects
Cancer Research ,Oncology ,Humans ,Interferon-alpha ,Hematology ,Polycythemia Vera - Published
- 2022
43. MPN-354 Characteristics of Patients With Myelofibrosis Taking Ruxolitinib for 3 or More Years
- Author
-
Lucia Masarova, Prithviraj Bose, Naveen Pemmaraju, Hagop Kantarjian, Zeev Estrov, and Srdan Verstovsek
- Subjects
Adult ,Aged, 80 and over ,Male ,Cancer Research ,Hematology ,Middle Aged ,Pyrimidines ,Oncology ,Primary Myelofibrosis ,Nitriles ,Humans ,Pyrazoles ,Female ,Child ,Aged - Abstract
Ruxolitinib (RUX) was the first JAK1/2 inhibitor approved for patients with advanced-phase myelofibrosis (MF) with a median therapy duration of about 3 years.We aimed to characterize the clinical features of patients treated with RUX for ≥3 years.Retrospective review of medical records of 131 patients presented to our institution between January 2000 and July 2020 who received RUX for ≥3 years.Major clinical features were reported by descriptive statistics. OS was analyzed using the Kaplan-Meier model. The Cox proportional hazard model (univariate, multivariate) was used for factors associated with OS.The median age of patients was 67 years (range, 31-84) and 56% were men. Fewer than 20% of patients had hemoglobin10 g/dL or platelets150×10Forty percent of patients who remained on RUX for ≥3 years were alive at 10 years since their therapy initiation. Age remains the major predictive factor of survival on long RUX therapy.
- Published
- 2022
44. Final results of a phase 2 clinical trial of LCL161, an oral SMAC mimetic for patients with myelofibrosis
- Author
-
Marina Konopleva, Nitin Jain, Gautam Borthakur, Carlos E. Bueso-Ramos, Zeev Estrov, Elias Jabbour, Prithviraj Bose, Wei Qiao, Sherry Pierce, Uday R. Popat, Naval Daver, Courtney D. DiNardo, Bing Z. Carter, Maro Ohanian, Lingsha Zhou, Srdan Verstovsek, Sharon D. Bledsoe, Jorge E. Cortes, Po Yee Mak, Xuemei Wang, Guillermo Garcia-Manero, Tapan M. Kadia, Naveen Pemmaraju, and Lucia Masarova
- Subjects
medicine.medical_specialty ,Myeloproliferative Disorders ,Clinical Trials and Observations ,Anemia ,business.industry ,Nausea ,Smac Mimetic LCL161 ,Phases of clinical research ,Apoptosis ,Hematology ,medicine.disease ,Gastroenterology ,Thiazoles ,Primary Myelofibrosis ,Internal medicine ,Toxicity ,medicine ,Vomiting ,Humans ,medicine.symptom ,Myelofibrosis ,Adverse effect ,business - 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.
- Published
- 2021
45. Triple-Negative Primary Myelofibrosis: a Bone Marrow Pathology Group (BMPG) study
- Author
-
Yahya Al-Ghamdi, Jonathan Lake, Adam Bagg, Beenu Thakral, Sa Wang, Carlos Bueso-Ramos, Lucia Masarova, Srdan Verstovsek, Heesun Rogers, Eric Hsi, Jonathon Gralewski, Devon Chabot-Richards, Tracy George, Anton Rets, Robert Hasserjian, Olga Weinberg, Megan Parilla, Daniel A Arber, Osvaldo Padilla, Attilio Orazi, and Wayne Tam
- Abstract
Primary myelofibrosis (PMF) is a clonal myeloproliferative neoplasm driven by canonical gene mutations in JAK2, CALR or MPL in more than 80% of the cases. PMF cases that lack these canonical alterations are termed triple-negative PMF (TN-PMF). The pathologic and genetic characteristics of TN-PMF compared to conventional PMF with canonical driver mutations (DM-PMF) have not been well studied. We aim to identify clinicopathologic and molecular genetic differences between TN-PMF (n = 56) and DM-PMF (n = 89), all of which fulfilled the 2016 WHO diagnostic criteria for PMF. Compared to the control group, TN-PMF patients were more likely to have thrombocytopenia and less likely to have organomegaly. The bone marrow with TN-PMF showed fewer granulocytic elements and more frequent dyserythropoiesis. Cytogenetic analysis showed a higher incidence of trisomy 8. Targeted NGS revealed a lower frequency of ASXL1 mutations but an enrichment of ASXL1/SRSF2 co-mutations. Our findings demonstrated several clinicopathologic and molecular differences between TN-PMF and DM-PMF. These findings, particularly the observed mutation profile characterized by a higher frequency of ASXL1 and SRSF2 co-mutation, suggest that at least a subset of TN-PMF may be pathogenetically different from DM-PMF, with potential prognostic implications.
- Published
- 2022
46. Clinicopathologic correlates and natural history of atypical chronic myeloid leukemia
- Author
-
Marina Konopleva, Farhad Ravandi, Prithviraj Bose, Koichi Takahashi, Naval Daver, Kiran Naqvi, Srdan Verstovsek, Guillermo Garcia-Manero, Courtney D. DiNardo, Keyur P. Patel, Carlos E. Bueso-Ramos, Gautam Borthakur, Joseph D. Khoury, Guillermo Montalban-Bravo, Hagop M. Kantarjian, Koji Sasaki, Elias J. Jabbour, Tapan M. Kadia, Zeev Estrov, Sherry Pierce, Rashmi Kanagal-Shamanna, Kelly A. Soltysiak, Lucia Masarova, Sanam Loghavi, and Naveen Pemmaraju
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Ruxolitinib ,Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative ,03 medical and health sciences ,0302 clinical medicine ,Bone Marrow ,Internal medicine ,Humans ,Medicine ,Platelet ,Multivariable model ,030212 general & internal medicine ,Gene ,Aged ,business.industry ,GATA2 ,Hazard ratio ,Hematopoietic Stem Cell Transplantation ,Myeloid leukemia ,Prognosis ,medicine.disease ,Natural history ,Transplantation ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Mutation ,Atypical chronic myeloid leukemia ,Bone marrow ,Stem cell ,business ,medicine.drug - Abstract
Background There are limited data on the clonal mechanisms underlying leukemogenesis, prognostic factors, and optimal therapy for atypical chronic myeloid leukemia (aCML). Methods The authors evaluated the clinicopathologic features, outcomes, and responses to therapy of 65 patients with aCML. The median age was 67 years (range, 46-89 years). Results The most frequently mutated genes included ASXL1 (83%), SRSF2 (68%), and SETBP1 (58%). Mutations in SETBP1, SRSF2, TET2, and GATA2 appeared at variant allele frequencies (VAFs) greater than 40%, whereas other RAS pathway mutations were more likely to appear at low VAFs. The acquisition of new, previously undetectable mutations at transformation was observed in 63% of the evaluable patients, with the most common involving signaling pathway mutations. Hypomethylating agents (HMAs) were associated with the highest response rates but with a short duration of response (median, 2.7 months). Therapy with ruxolitinib was not associated with clinically significant responses as a single agent or in combination with an HMA. Allogeneic stem cell transplantation was the only therapy associated with improved outcomes (hazard ratio, 0.144; 95% CI, 0.035-0.593; P = .007). Age, platelet counts, bone marrow blast percentages, and serum lactate dehydrogenase (LDH) levels were independent predictors of survival and were integrated in a multivariable model that allowed the prediction of 1- and 3-year survival. Conclusions aCML is characterized by high frequencies of ASXL1, SRSF2, and SETBP1 mutations and is associated with a high risk of acute myeloid leukemia transformation. Response and survival outcomes with current therapies remain poor. The incorporation of age, platelet counts, bone marrow blast percentages, and LDH levels can allow survival prediction, and allogeneic stem cell transplantation should be considered for all eligible patients.
- Published
- 2021
47. Allogeneic stem cell transplant for patients with myeloproliferative neoplasms in blast phase: improving outcomes in the recent era
- Author
-
Rohtesh S. Mehta, Lucia Masarova, Rima M. Saliba, Amanda Olson, Naval Daver, Qaiser Bashir, Samer A. Srour, Amin M. Alousi, Stefan O. Ciurea, Ankur Varma, Prithviraj Bose, Richard E. Champlin, Uday R. Popat, Naveen Pemmaraju, Mithun Vinod Shah, Srdan Verstovsek, and Betul Oran
- Subjects
Male ,Myeloproliferative Disorders ,business.industry ,medicine.medical_treatment ,Hematopoietic Stem Cell Transplantation ,Hematology ,Hematopoietic stem cell transplantation ,Allografts ,Blast Phase ,Disease-Free Survival ,Survival Rate ,Cancer research ,Humans ,Medicine ,Female ,Stem cell ,Blast Crisis ,business ,Retrospective Studies - Published
- 2021
48. Diagnostic Performance of Erythropoietin Levels in Polycythemia Vera: Experience at a Comprehensive Cancer Center
- Author
-
Cristhiam M. Rojas-Hernandez, Lee Cheng, Lucia Masarova, Srdan Verstovsek, Alanna Barrios-Ruiz, Eric Fountain, and Daniel Davila-Gonzalez
- Subjects
Male ,Cancer Research ,medicine.medical_specialty ,Secondary Polycythemia ,Polycythemia ,Cancer Care Facilities ,03 medical and health sciences ,0302 clinical medicine ,Polycythemia vera ,Predictive Value of Tests ,hemic and lymphatic diseases ,Internal medicine ,Biomarkers, Tumor ,medicine ,Humans ,Genetic Testing ,Erythropoietin ,Polycythemia Vera ,Retrospective Studies ,Predictive marker ,business.industry ,Area under the curve ,Cancer ,Retrospective cohort study ,Hematology ,Janus Kinase 2 ,Middle Aged ,medicine.disease ,ROC Curve ,Oncology ,030220 oncology & carcinogenesis ,Mutation ,Etiology ,Female ,business ,030215 immunology ,medicine.drug - Abstract
Introduction Considering the evolving diagnostic criteria of polycythemia vera (PV), we analyzed the utility of serum erythropoietin (EPO) as a predictive marker for differentiating polycythemia vera (PV) from other etiologies of erythrocytosis. Patients and Methods We conducted a retrospective study after a review of electronical medical records from January 2005 to December 2016 with diagnosis of erythrocytosis using International Classification of Disease–specific codes. To evaluate the diagnostic performance of EPO levels and JAK2-V617F mutation, we constructed a receiver-operated characteristic curve of sensitivity versus 1-specificity for serum EPO levels and JAK2-V617F mutation as predictive markers for differentiating PV from other causes of erythrocytosis. Results We surveyed 577 patients with erythrocytosis. Median patient age was 59.2 years, 57.72% (n = 329) were male, 86.3% (n = 491) were white, and only 3.3% (n = 19) were African American. A total of 80.88% (n = 351) of those diagnosed with PV had a JAK2-V617F mutation compared to only 1.47% (n = 2) whose primary diagnosis was secondary polycythemia. When comparing JAK2-V617 mutation to the EPO level, the area under the curve of JAK2-V617 (0.8970) was statistically larger than that of EPO test (0.6765). Therefore, the PV diagnostic methodology using JAK2-V617 is better than the EPO test. An EPO level of 99% specific to predict PV but was only 12% sensitive. Conclusion In the appropriate clinical setting, cytogenetic and molecular studies such as JAK2 mutation status prevail as the most useful tools for PV case identification. The use of isolated EPO to screen patients with erythrocytosis is not a good diagnostic approach.
- Published
- 2021
49. Prognostic value of measurable residual disease after venetoclax and decitabine in acute myeloid leukemia
- Author
-
Koichi Takahashi, Prithviraj Bose, Elias Jabbour, Jan A. Burger, Musa Yilmaz, Srdan Verstovsek, Nicholas J. Short, Ghayas C. Issa, Naval Daver, Maro Ohanian, Wei Qiao, Sa A. Wang, Courtney D. DiNardo, Hagop M. Kantarjian, Caitlin R. Rausch, Kathryn S. Montalbano, Farhad Ravandi, Jing Ning, Gautam Borthakur, Tapan M. Kadia, Alessandra Ferrajoli, Philip A. Thompson, Marina Konopleva, Guillermo Garcia-Manero, Michael Andreeff, Nitin Jain, Jeffrey L. Jorgensen, Naveen Pemmaraju, Lucia Masarova, Koji Sasaki, Abhishek Maiti, Sherry Pierce, and Guillermo Montalban-Bravo
- Subjects
medicine.medical_specialty ,Myeloid ,Clinical Trials and Observations ,Decitabine ,Gastroenterology ,chemistry.chemical_compound ,Interquartile range ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Humans ,Aged ,Sulfonamides ,business.industry ,Venetoclax ,Hazard ratio ,Myeloid leukemia ,Hematology ,Bridged Bicyclo Compounds, Heterocyclic ,Prognosis ,medicine.disease ,Confidence interval ,Leukemia, Myeloid, Acute ,Leukemia ,medicine.anatomical_structure ,chemistry ,business ,medicine.drug - Abstract
Assessment of measurable residual disease (MRD) provides prognostic information in acute myeloid leukemia (AML). However, the utility of MRD with venetoclax-based lower intensity regimens is unknown. We analyzed the prognostic value of achieving a negative MRD in older/“unfit” patients with AML receiving first-line therapy with 10-day decitabine and venetoclax. MRD was evaluated in bone marrow specimens using multicolor flow cytometry (sensitivity 0.1%). Ninety-seven patients achieving either a complete remission (CR) or CR with incomplete hematologic recovery (CRi) or morphologic leukemia-free state were included. Median age was 72 years (interquartile range, 68-78 years), and 64% had adverse-risk AML. Eighty-three patients achieved CR/CRi, and 52 (54%) became MRD negative. Median time to becoming MRD negative was 2.0 months (interquartile range, 0.9-3.1 months). Patients becoming MRD negative by 2 months had longer relapse-free survival (RFS) compared with those remaining MRD positive (median RFS, not reached vs 5.2 months; hazard ratio [HR], 0.31; 95% confidence interval [CI], 0.12-0.78; P = .004), longer event-free survival (EFS) (median EFS, not reached vs 5.8 months; HR, 0.25; 95% CI, 0.12-0.55; P < .001), as well as longer overall survival (OS) (median OS, 25.1 vs 7.1 months; HR, 0.23; 95% CI, 0.11-0.51; P < .001). Patients achieving an MRD-negative CR had longer OS compared with those with an inferior response (median OS, 25.1 vs 11.6 months; HR, 0.33; 95% CI, 0.19-0.58; P < .0005). Patients becoming MRD negative within 1 month had an improved OS compared with MRD-positive patients (median OS, 25.1 vs 3.4 months; HR, 0.15; 95% CI, 0.03-0.64; P < .0001). Differential impact of MRD status on survival outcomes persisted at a later 4-month time point of evaluation. In conclusion, MRD-negative status at 1, 2, and 4 months after starting therapy confers significantly better survival in older/unfit patients with AML receiving first-line therapy with 10-day decitabine and venetoclax. This trial was registered at www.clinicaltrials.gov as #NCT03404193.
- Published
- 2021
50. Abstract CT242: A phase I study of ruxolitinib in combination with abemaciclib for patients with primary or post-polycythemia vera/essential thrombocythemia myelofibrosis
- Author
-
Jan P. Bewersdorf, Srdan Verstovsek, Andriy Derkach, Lucia Masarova, Naveen Pemmaraju, Eytan M. Stein, Michael Mauro, Raajit Rampal, and Prithviraj Bose
- Subjects
Cancer Research ,Oncology - Abstract
Introduction: Patients (pts) with primary and secondary myelofibrosis (MF) have a reduced life expectancy. While several oral Janus Kinase (JAK)-1/2 inhibitors such as ruxolitinib (RUX) have demonstrated improvements in symptom burden and spleen volume reduction compared to physician choice of best available therapy leading to their approval by the US FDA, responses to these agents are generally time-limited. Additionally, these agents are not considered to be disease-modifying and survival in pts after RUX discontinuation is poor with a median survival of 11-14 months. Thus, novel mechanism-based therapies for pts with suboptimal response or progression on RUX are needed. Preclinical Rationale: JAK2 V617F mutations promote transition from G1 to S-phase of the cell cycle via increased expression of CDC25A in preclinical models and CDC25A expression is upregulated in primary MPN patient samples. Additional cell cycle regulators such as CDK6 and Cyclin D also play a role in MF pathogenesis and drive myeloproliferation. Finally, combination of the CDK4/6 inhibitor ribociclib and RUX demonstrated reduction in spleen and liver size when compared to RUX alone in murine models, suggesting synergy (Rampal et al., CCR 2021). Methods: This multicenter, phase I dose-escalation trial evaluates the safety of RUX + the CDK4/6 inhibitor abemaciclib in pts with primary or secondary MF with intermediate-1/2 or high-risk disease by DIPSS who require treatment and had an inadequate response to RUX. An inadequate response will be defined by (I) palpable splenomegaly ≥5 cm below the left costal margin at study entry AND/OR (II) active MPN symptoms [presence of 1 symptom score ≥5 or 2 symptom scores ≥3 using the screening MPN-SAF TSS. This study follows a conventional “3+3” dose-escalation design, in which pts on a stable dose of RUX (10mg or 15mg BID) with sufficient baseline platelet and absolute neutrophil count will be treated with increasing doses of abemaciclib. Abemaciclib was chosen due to less myelosuppression compared with other CDK4/6 inhibitors. The pre-study dose of ruxolitinib will be maintained to determine the maximum tolerated dose of the combination of RUX+ abemaciclib. The primary endpoint is to determine the safety and tolerability, and to identify the recommended phase II dose of abemaciclib in MF pts on a stable dose of RUX. Secondary endpoints include overall response rate, progression-free and overall survival, and duration of response per the modified International Working Group - Myeloproliferative Neoplasms Research and Treatment (IWG-MRT) and the European Leukemia Net (ELN) 2013 consensus definitions. Correlative studies are planned to identify biomarkers predictive of response to RUX + abemaciclib, mechanisms of resistance (e.g., MAP kinase pathway activation), and any disease-modifying effects of combination therapy. Citation Format: Jan P. Bewersdorf, Srdan Verstovsek, Andriy Derkach, Lucia Masarova, Naveen Pemmaraju, Eytan M. Stein, Michael Mauro, Raajit Rampal, Prithviraj Bose. A phase I study of ruxolitinib in combination with abemaciclib for patients with primary or post-polycythemia vera/essential thrombocythemia myelofibrosis [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 2 (Clinical Trials and Late-Breaking Research); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(8_Suppl):Abstract nr CT242.
- Published
- 2023
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.