73 results on '"Konopleva MY"'
Search Results
2. The Impact of Smoking on Survival in Patients (Pts) with Newly Diagnosed Philadelphia Chromosome Positive (Ph plus ) Acute Lymphoblastic Leukemia (ALL) Treated with the Combination of Intensive Therapy with Tyrosine Kinase Inhibitor (TKI)
- Author
-
Sasaki, K, Ribera, JM, Figueroa, M, Ravandi, F, Short, NJ, Garcia-Manero, G, Daver, NG, Kadia, TM, Konopleva, MY, Jain, N, Issa, GC, Estrov, ZE, Garris, R, Khouri, R, Nasnas, P, DiNardo, CD, Naqvi, K, Kornblau, SM, Montalban-Bravo, G, Pemmaraju, N, Cortes, JE, O'Brien, SM, Chandra, J, Kantarjian, HM, and Jabbour, E
- Published
- 2019
3. Venetoclax plus low intensity chemotherapy for adults with acute lymphoblastic leukemia.
- Author
-
Luskin MR, Shimony S, Keating JH, Winer ES, Garcia JS, Stone RM, Jabbour EJ, Flamand Y, Stevenson K, Ryan JA, Zeng Z, Letai A, Konopleva MY, Jain N, and DeAngelo DJ
- Abstract
In acute lymphoblastic leukemia (ALL), the BCL2 inhibitor venetoclax may enhance the efficacy of chemotherapy allowing dose reductions that reduce toxicity. We designed a phase 1b study of venetoclax plus attenuated chemotherapy to determine the recommended phase 2 dose (RP2D) of venetoclax. The study enrolled 19 patients with ALL either newly diagnosed (≥60 years, n=11 [B-cell, n=8; T-cell, n=3]) or R/R (≥18 years, n=8 [B-cell, n=3; T-cell, n=5]). Venetoclax was given for 21 days with each cycle of mini-hyper-CVD (cyclophosphamide, vincristine, dexamethasone alternating with methotrexate and cytarabine). There were no dose limiting toxicities at dose level (DL) 1 (n=3, 400 mg/day) or DL2 (n=6, 600 mg/day); DL2 was the RP2D and explored further (n=10). The most common non-hematologic adverse events were grade 3+ infections. There were no deaths within 60 days and no patients discontinued therapy for toxicity. There was no tumor lysis syndrome, hepatotoxicity, or prolonged cytopenias. Among patients with newly diagnosed ALL, 10/11 (90.9%) achieved a measurable residual disease-negative (<0.01% sensitivity) complete remission (CR) including 6 patients with hypodiploid TP53 mutated ALL. All patients in CR bridged to hematopoietic stem cell transplant (n=9) or completed protocol (n=1). With a median follow-up of 60.0 months, median disease-free survival (DFS) for patients with newly diagnosed ALL was 54.6 months (95% CI: 35.5 months - NA) with a 2-year DFS rate of 90% (95% CI, 71% - 100%). Among patients with R/R ALL, 3/8 (37.5%) achieved CR. In summary, for patients with newly diagnosed ALL, venetoclax plus mini-HCVD is well-tolerated with promising efficacy. NCT03319901., (Copyright © 2024 American Society of Hematology.)
- Published
- 2024
- Full Text
- View/download PDF
4. Inter-racial genetic differences in myelofibrosis: a diverse inner-city center analysis.
- Author
-
Kamel J, Yan J, Rockwell B, Goldfinger M, Feldman EJ, Konopleva MY, Mantzaris I, Shastri A, Kornblum N, Gritsman K, Sica A, Shah N, Cooper D, Verma A, and Goel S
- Published
- 2024
- Full Text
- View/download PDF
5. A Weekly Low-Dose Regimen of Decitabine and Venetoclax is Efficacious and Less Myelotoxic in a Racially Diverse Cohort.
- Author
-
Goldfinger M, Mantzaris I, Shastri A, Saunthararajah Y, Gritsman K, Sica A, Kornblum N, Shah N, Levitz D, Rockwell B, Shapiro LC, Gupta R, Pradhan K, Xue X, Munoz A, Dhawan A, Fehn K, Comas M, Verceles JA, Jonas BA, Kambhampati S, Shi Y, Braunschweig I, Cooper DL, Konopleva MY, Feldman EJ, and Verma A
- Abstract
A metronomic, low-dose schedule of decitabine and Venetoclax was safe and effective in myeloid malignancies with few dose reductions or interruptions in an older diverse population. Median OS for AML and TP53 mutated patients was 16.1 and 11.3 months respectively., (Copyright © 2024 American Society of Hematology.)
- Published
- 2024
- Full Text
- View/download PDF
6. Molecular responses in decitabine- and decitabine/ venetoclax-treated patients with acute myeloid leukemia and myelodysplastic syndromes.
- Author
-
Gruszczynska A, Maiti A, Miller CA, Ramakrishnan SM, Link DC, Uy GL, Petti AA, Hayes K, DiNardo CD, Ravandi F, Ley TJ, Spencer DH, Gao F, Konopleva MY, and Welch JS
- Subjects
- Humans, Male, Aged, Female, Treatment Outcome, Middle Aged, Adult, Decitabine therapeutic use, Decitabine pharmacology, Bridged Bicyclo Compounds, Heterocyclic therapeutic use, Myelodysplastic Syndromes drug therapy, Myelodysplastic Syndromes genetics, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute mortality, Sulfonamides therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use
- Published
- 2024
- Full Text
- View/download PDF
7. Venetoclax and Cobimetinib in Relapsed/Refractory AML: A Phase 1b Trial.
- Author
-
Konopleva MY, Dail M, Daver NG, Garcia JS, Jonas BA, Yee KWL, Kelly KR, Vey N, Assouline S, Roboz GJ, Paolini S, Pollyea DA, Tafuri A, Brandwein JM, Pigneux A, Powell BL, Fenaux P, Olin RL, Visani G, Martinelli G, Onishi M, Wang J, Huang W, Dunshee DR, Hamidi H, Ott MG, Hong WJ, and Andreeff M
- Subjects
- Humans, Aged, Male, Female, Middle Aged, Aged, 80 and over, Treatment Outcome, Drug Resistance, Neoplasm drug effects, Bridged Bicyclo Compounds, Heterocyclic therapeutic use, Bridged Bicyclo Compounds, Heterocyclic pharmacology, Bridged Bicyclo Compounds, Heterocyclic administration & dosage, Sulfonamides pharmacology, Sulfonamides therapeutic use, Sulfonamides administration & dosage, Azetidines therapeutic use, Azetidines pharmacology, Azetidines administration & dosage, Piperidines therapeutic use, Piperidines pharmacology, Leukemia, Myeloid, Acute drug therapy, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Antineoplastic Combined Chemotherapy Protocols pharmacology, Antineoplastic Combined Chemotherapy Protocols adverse effects
- Abstract
Background: Therapies for relapsed/refractory acute myeloid leukemia remain limited and outcomes poor, especially amongst patients who are ineligible for cytotoxic chemotherapy or targeted therapies., Patients and Methods: This phase 1b trial evaluated venetoclax, a B-cell lymphoma-2 (BCL-2) inhibitor, plus cobimetinib, a MEK1/2 inhibitor, in patients with relapsed/refractory acute myeloid leukemia, ineligible for cytotoxic chemotherapy. Two-dimensional dose-escalation was performed for venetoclax dosed daily, and for cobimetinib dosed on days 1-21 of each 28-day cycle., Results: Thirty patients (median [range] age: 71.5 years [60-84]) received venetoclax-cobimetinib. The most common adverse events (AEs; in ≥40.0% of patients) were diarrhea (80.0%), nausea (60.0%), vomiting (40.0%), febrile neutropenia (40.0%), and fatigue (40.0%). Overall, 66.7% and 23.3% of patients experienced AEs leading to dose modification/interruption or treatment withdrawal, respectively. The composite complete remission (CRc) rate (complete remission [CR] + CR with incomplete blood count recovery + CR with incomplete platelet recovery) was 15.6%; antileukemic response rate (CRc + morphologic leukemia-free state/partial remission) was 18.8%. For the recommended phase 2 dose (venetoclax: 600 mg; cobimetinib: 40 mg), CRc and antileukemic response rates were both 12.5%. Failure to achieve an antileukemic response was associated with elevated baseline phosphorylated ERK and MCL-1 levels, but not BCL-xL. Baseline mutations in ≥1 signaling gene or TP53 were noted in nonresponders and emerged on treatment. Pharmacodynamic biomarkers revealed inconsistent, transient inhibition of the mitogen-activated protein kinase (MAPK) pathway., Conclusion: Venetoclax-cobimetinib showed limited preliminary efficacy similar to single-agent venetoclax, but with added toxicity. Our findings will inform future trials of BCL-2/MAPK pathway inhibitor combinations., Competing Interests: Disclosure MYK is a consultant for AbbVie, Inc., Genentech, Inc., and F. Hoffmann La-Roche; is an advisory board member for F. Hoffmann La-Roche; holds shares from Reata Pharmaceuticals; has received honoraria from Amgen, AbbVie, Inc., and Genentech, Inc.; and has received research funding from AbbVie, Inc., Genentech, Inc., Eli Lilly, Cellectis, Calithera, Stemline, Threshold, Flexus Biosciences, Novartis, Ablynx, and Agios. MD, MO, JW, WH, DD, and HH are employees of Genentech, Inc. and may hold Roche stock or stock options. NGD has received research funding from Daiichi Sankyo, Bristol-Myers Squibb, Pfizer, Gilead, Sevier, Genentech, Inc., Astellas, Daiichi Sankyo, AbbVie, Inc., Hanmi, Trovagene, Fate Therapeutics, Amgen, Novimmune, GlycoMimetics, Trillium, Kite Pharma, Aptose, Shattuck Labs, KAHR, ArcellX, Sanofi, Sumitomo, and ImmunoGen; and has served in a consulting or advisory role for Daiichi Sankyo, Bristol-Myers Squibb, Arog Pharmaceuticals, Pfizer, Novartis, Jazz Pharmaceuticals, Celgene, AbbVie, Inc., Astellas, Genentech, Inc., Immunogen, Servier, Syndax, Trillium, Gilead, Amgen, Shattuck Labs, ArcellX, Kite Pharma, Sumitomo, Caribou Biosciences, Sanofi, Rigel, Aptose, KURA, GlycoMimetics, and Agios. JSG has received research funding (for trials) from AbbVie, Inc., Genentech, Inc., Pfizer, Prelude, and AstraZeneca; and has served on advisory boards for AbbVie, Inc., Astellas, Bristol-Myers Squibb and Servier. BAJ is a consultant/advisor for AbbVie, Inc., Bristol-Myers Squibb, Daiichi Sankyo, Genentech, Inc., Gilead, GlycoMimetics, Jazz Pharmaceuticals, Kymera, Pfizer, Rigel, Servier, and Takeda; protocol steering committee for GlycoMimetics; data monitoring committee for Gilead; travel reimbursement/support from Rigel; and research funding to his institution from AbbVie, Inc., Amgen, Arog Pharmaceuticals, Aptose, BMS, Celgene, Daiichi Sankyo, F. Hoffmann-La Roche, Forma Therapeutics, Forty-Seven, Genentech, Inc./Roche, Gilead, GlycoMimetics, Hanmi, Immune-Onc, Incyte, Jazz Pharmaceuticals, Loxo Oncology, Pfizer, Pharmacyclics, Sigma Tau, and Treadwell. KWLY is a consultant for Astex, Bristol-Myers Squibb/Celgene, F. Hoffmann-La Roche, Novartis, Otsuka, Paladin, Pfizer, Shattuck Labs, Taiho, and Takeda; has received honorarium from AbbVie, Inc. and Novartis; and has received research funding from Astex, Forma Therapeutics, F. Hoffmann-La Roche, Genentech, Inc., Geron, Janssen, Jazz Pharmaceuticals, MedImmune, Novartis, Onconova, and Tolero. KRK, NV, SP, AT, AP, PF, and GV declare no competing financial interests. SA reports non-financial support from Roche/Genentech, Inc. during the conduct of the study; and has received personal fees from Roche Canada, Pfizer, Bristol-Myers Squibb, Palladin, and Lundbeck outside of the submitted work. GJR consulted for AbbVie, Inc., Amgen, Argenx, AstraZeneca, Bluebird Bio, Blueprint Medicines, Bristol-Myers Squibb, Caribou Biosciences, Celgene, Daiichi Sankyo, Ellipses Pharma, GlaxoSmithKline, Janssen, Jasper Pharmaceuticals, Jazz Pharmaceuticals, Molecular Partners, Novartis, Pfizer, Roche, Syndax, Takeda (IRC Chair), and Telix Pharma; and has received research funding from Janssen. DAP has received research funding from AbbVie, Inc. and has served as a consultant or advisory board member for AbbVie, Inc. and Genentech, Inc. JMB consulted for AbbVie, Inc., Bristol-Myers Squibb, Astex, Pfizer, Astellas, Taiho, and Jazz Pharmaceuticals. BLP has received research funding from Ambit Biosciences, Genentech, Inc., F. Hoffmann-La Roche, Jazz Pharmaceuticals, Novartis, Pfizer, and Cornerstone Pharmaceuticals; and is a consultant for Cornerstone Pharmaceuticals. RLO has received research support for trials from Astellas, Pfizer, Genentech, Inc., Daiichi Sankyo, and Cellectis; and consulted for AbbVie, Inc., Astellas, and Actinium. GM is a consultant for AbbVie, Inc., Celgene, Roche, Janssen, Astellas, Pfizer, and Incyte. W-JH is a former employee of Genentech, Inc. and may hold Roche stock; and is a current employee of Prelude Therapeutics. MGO is an employee of F. Hoffmann-La Roche and may hold stock or stock options. MA consulted for Amgen, AstraZeneca, Daiichi Sankyo, Syndax, GlycoMimetics, Oncoceutics, and Aptose; has received research funding from F. Hoffmann-La Roche, AstraZeneca, Amgen, Daiichi Sankyo, Jazz Pharmaceuticals, GlycoMimetics; and holds stock from Reata, Oncoceutics/Chimerix and Aptose., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
8. Pivekimab sunirine (IMGN632), a novel CD123-targeting antibody-drug conjugate, in relapsed or refractory acute myeloid leukaemia: a phase 1/2 study.
- Author
-
Daver NG, Montesinos P, DeAngelo DJ, Wang ES, Papadantonakis N, Todisco E, Sweet KL, Pemmaraju N, Lane AA, Torres-Miñana L, Thompson JE, Konopleva MY, Sloss CM, Watkins K, Bedse G, Du Y, Malcolm KE, Zweidler-McKay PA, and Kantarjian HM
- Subjects
- Humans, Female, Male, Interleukin-3 Receptor alpha Subunit, Immunoconjugates adverse effects, Leukemia, Myeloid, Acute drug therapy, Drug-Related Side Effects and Adverse Reactions, Hematologic Neoplasms, Febrile Neutropenia
- Abstract
Background: Pivekimab sunirine (IMGN632) is a first-in-class antibody-drug conjugate comprising a high-affinity CD123 antibody, cleavable linker, and novel indolinobenzodiazepine pseudodimer payload. CD123 is overexpressed in several haematological malignancies, including acute myeloid leukaemia. We present clinical data on pivekimab sunirine in relapsed or refractory acute myeloid leukaemia., Methods: This first-in-human, phase 1/2 dose-escalation and dose-expansion study enrolled participants aged 18 years or older at nine hospitals in France, Italy, Spain, and the USA with CD123+ haematological malignancies (Eastern Cooperative Oncology Group performance status of 0-1); participants reported here were in a cohort of participants with acute myeloid leukaemia who were refractory to or had relapsed on one or more previous treatments for acute myeloid leukaemia. The 3 + 3 dose-escalation phase evaluated two dosing schedules: schedule A (once every 3 weeks, on day 1 of a 3-week cycle) and fractionated schedule B (days 1, 4, and 8 of a 3-week cycle). The dose-expansion phase evaluated two cohorts: one cohort given 0·045 mg/kg of bodyweight (schedule A) and one cohort given 0·090 mg/kg of bodyweight (schedule A). The primary endpoints were the maximum tolerated dose and the recommended phase 2 dose. Antileukaemia activity (overall response and a composite complete remission assessment) was a secondary endpoint. The study is ongoing and registered with ClinicalTrials.gov, NCT03386513., Findings: Between Dec 29, 2017, and May 27, 2020, 91 participants were enrolled (schedule A, n=68; schedule B, n=23). 30 (44%) of schedule A participants were female and 38 (56%) were male; 60 (88%) were White, six (9%) were Black or African American, and two (3%) were other races. Pivekimab sunirine at doses of 0·015 mg/kg to 0·450 mg/kg in schedule A was administered in six escalating doses with no maximum tolerated dose defined; three dose-limiting toxicities were observed (reversible veno-occlusive disease; 0·180 mg/kg, n=1 and 0·450 mg/kg, n=1; and neutropenia; 0·300 mg/kg, n=1). Schedule B was not pursued further on the basis of comparative safety and antileukaemia findings with schedule A. The recommended phase 2 dose was selected as 0·045 mg/kg once every 3 weeks. At the recommended phase 2 dose (n=29), the most common grade 3 or worse treatment-related adverse events were febrile neutropenia (three [10%]), infusion-related reactions (two [7%]), and anaemia (two [7%]). Treatment-related serious adverse events occurring in 5% or more of participants treated at the recommended phase 2 dose were febrile neutropenia (two [7%]) and infusion-related reactions (two [7%]). Among 68 participants who received schedule A, one death (1%) was considered to be treatment-related (cause unknown; 0·300 mg/kg cohort). At the recommended phase 2 dose, the overall response rate was 21% (95% CI 8-40; six of 29) and the composite complete remission rate was 17% (95% CI 6-36; five of 29)., Interpretation: Pivekimab sunirine showed single-agent activity across multiple doses, with a recommended phase 2 dose of 0·045 mg/kg once every 3 weeks. These findings led to a phase 1b/2 study of pivekimab sunirine plus azacitidine and venetoclax in patients with CD123-positive acute myeloid leukaemia., Funding: ImmunoGen., Competing Interests: Declaration of interests NGD reports consultancy for Agios, Astellas, KITE, AbbVie, Gilead, Jazz, Daiichi Sankyo, BMS, ImmunoGen, Syndax, Novartis, Shattuck Labs, Trillium, Genentech, Stemline/Menarini, Celgene, Amgen, Pfizer, Servier, and Arog; and research funding from Astellas, Trovagene, FATE Therapeutics, Novimmune, KITE, Hanmi Pharm, AbbVie, Gilead, Daiichi Sankyo, BMS, ImmunoGen, Glycomimetics, Trillium, Genentech, Amgen, Pfizer, and Servier. PM reports consultancy for Daiichi Sankyo and Celgene; membership on an entity's Board of Directors or advisory committees for Daiichi Sankyo, AbbVie, Celgene, Novartis, Karyopharm, Incyte, Janssen, Pfizer, and Teva; research support from Daiichi Sankyo, Celgene, Novartis, Karyopharm, Janssen, Pfizer, and Teva; being on a speaker's bureau for Daiichi Sankyo, Celgene, Novartis, Incyte, Janssen, Pfizer and Teva; and research funding from Novartis, Janssen, Pfizer, and Teva. DJD reports research funding from AbbVie, Glycomimetics, and Novartis; and consultancy for Novartis, Amgen, Blueprint, Gilead Sciences, Incyte, Jazz Pharmaceuticals, Pfizer, Servier, and Takeda. ESW reports being on an advisory board or consulting for AbbVie, BMS, CTI Biopharma, Daiichi Sankyo, Genentech, GSK, Johnson & Johnson, Kite, Kura, Novartis, Rigel, Sellas, and Sumitomo Pharma; a speaker role for Astellas, Kura, and Pfizer; being on a data monitoring committee for AbbVie and Gilead; and being on a research committee for Gilead. NPa reports being on an advisory board for CTI Biopharma and Cambium Oncology; consultancy for CTI Biopharma; and research funding from Gilead, ONO Pharma, and AbbVie. ET reports consultancy for AbbVie, Janssen, and Jazz Pharmaceuticals; and being on an advisory board for AbbVie, Janssen, and Jazz Pharmaceuticals. KLS reports consultancy for Bristol Myers Squibb, Gilead, Jazz Arog, Pfizer, BerGenBio, Astellas, Novartis, Curis, Mablytics, Daiichi Sankyo, and Nkarta; and research funding from Jazz and Incyte. NPe reports consultancy for MustangBio, AbbVie, Celgene, Stemline Therapeutics, Novartis, and Incyte; research funding from MustangBio, Samus, AbbVie, Cellectis, Stemline Therapeutics, Novartis, Plexxikon, Daiichi-Sankyo, SagerStrong, Affymetrix, and Incyte; and honoraria from AbbVie, Celgene, and Stemline Therapeutics. AAL received research funding from AbbVie and Stemline Therapeutics; and consulting fees from Cimeio Therapeutics, IDRx, Jnana Therapeutics, N-of-One, and Qiagen. JET reports research funding from Bristol Myers Squibb and Novartis. MYK reports research funding from Allogene, Daiichi, Gilead, ImmunoGen, MEI Pharma, Precision Biosciences, Sanofi, Ablynx, Genentech, Cellectis, AbbVie, Stemline Therapeutics, Forty-Seven, and AstraZeneca; consultancy for Gilead, MEI Pharma, Sanofi, Janssen, Vincerx, Genentech, AbbVie, Stemline Therapeutics, and AstraZeneca; honoraria from BAKX Therapeutics, Genentech, AbbVie, and Stemline Therapeutics; and equity ownership, patents & royalties from Reata Pharmaceuticals. CMS, KW, GB, YD, KEM, and PAZ-M are employees of ImmunoGen. HMK reports honoraria from Pfizer, Takeda, Amphista, Amgen, Ascentage, Astellas, Agios, Biologix, Curis, Ipsen Biopharmaceuticals, KAHR, Medical LabCorp, Shenzhen Target, Stemline, Novartis, and AbbVie; and research funding from Jazz Pharma, BMS, Amgen, Ascentage, Agios, ImmunoGen, Cyclacel, Daiichi-Sankyo, Novartis, and AbbVie. LT-M declares no competing interests., (Copyright © 2024 Elsevier Ltd. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
9. Correction: Preclinical development of 1B7/CD3, a novel anti-TSLPR bispecific antibody that targets CRLF2-rearranged Ph-like B-ALL.
- Author
-
Tian Z, Shi C, Yang G, Allen JK, Shi Q, Al-Shami A, Olson JW, Smith MG, Chang Q, Kaur J, You J, Lofton TE, Gonzalez MA, Zhang Q, Zha D, Tasian SK, Jain N, Konopleva MY, Heffernan T, and Molldrem JJ
- Published
- 2024
- Full Text
- View/download PDF
10. Preclinical development of 1B7/CD3, a novel anti-TSLPR bispecific antibody that targets CRLF2-rearranged Ph-like B-ALL.
- Author
-
Tian Z, Shi C, Yang G, Allen JK, Shi Q, Al-Shami A, Olson JW, Smith MG, Chang Q, Kaur J, You J, Lofton TE, Gonzalez MA, Zhang Q, Zha D, Tasian SK, Jain N, Konopleva MY, Heffernan T, and Molldrem JJ
- Subjects
- Humans, Animals, Mice, CD3 Complex, Antigens, CD19, Cell Line, Receptors, Cytokine, Antibodies, Bispecific pharmacology, Antibodies, Bispecific therapeutic use, Lymphoma, B-Cell drug therapy, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma pathology
- Abstract
Patients harboring CRLF2-rearranged B-lineage acute lymphocytic leukemia (B-ALL) face a 5-year survival rate as low as 20%. While significant gains have been made to position targeted therapies for B-ALL treatment, continued efforts are needed to develop therapeutic options with improved duration of response. Here, first we have demonstrated that patients with CRLF2-rearranged Ph-like ALL harbor elevated thymic stromal lymphopoietin receptor (TSLPR) expression, which is comparable with CD19. Then we present and evaluate the anti-tumor characteristics of 1B7/CD3, a novel CD3-redirecting bispecific antibody (BsAb) that co-targets TSLPR. In vitro, 1B7/CD3 exhibits optimal binding to both human and cynomolgus CD3 and TSLPR. Further, 1B7/CD3 was shown to induce potent T cell activation and tumor lytic activity in both cell lines and primary B-ALL patient samples. Using humanized cell- or patient-derived xenograft models, 1B7/CD3 treatment was shown to trigger dose-dependent tumor remission or growth inhibition across donors as well as induce T cell activation and expansion. Pharmacokinetic studies in murine models revealed 1B7/CD3 to exhibit a prolonged half-life. Finally, toxicology studies using cynomolgus monkeys found that the maximum tolerated dose of 1B7/CD3 was ≤1 mg/kg. Overall, our preclinical data provide the framework for the clinical evaluation of 1B7/CD3 in patients with CRLF2-rearranged B-ALL., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
11. Response patterns and impact of MRD in patients with IDH1/2-mutated AML treated with venetoclax and hypomethylating agents.
- Author
-
Hammond D, Loghavi S, Wang SA, Konopleva MY, Kadia TM, Daver NG, Ohanian M, Issa GC, Alvarado Y, Short NJ, Sasaki K, Pemmaraju N, Montalban-Bravo G, Lachowiez CA, Maiti A, Garcia-Manero G, Jabbour EJ, Borthakur G, Ravandi F, Takahashi K, Pierce SR, Kantarjian HM, and DiNardo CD
- Subjects
- Humans, Sulfonamides therapeutic use, Isocitrate Dehydrogenase genetics, Bridged Bicyclo Compounds, Heterocyclic pharmacology, Bridged Bicyclo Compounds, Heterocyclic therapeutic use, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics
- Published
- 2023
- Full Text
- View/download PDF
12. Characteristics and clinical outcomes of patients with acute myeloid leukemia with inv(3)(q21q26.2) or t(3;3)(q21;q26.2).
- Author
-
Richard-Carpentier G, Rausch CR, Sasaki K, Hammond D, Morita K, Takahashi K, Tang G, Kanagal-Shamanna R, Bhalla K, Dinardo CD, Borthakur G, Pemmaraju N, Shpall EJ, Alousi A, Daver NG, Garcia-Manero G, Konopleva MY, Ravandi F, Kantarjian HM, and Kadia TM
- Subjects
- Humans, Middle Aged, Retrospective Studies, Translocation, Genetic, Chromosome Inversion, Transcription Factors genetics, Prognosis, Proto-Oncogene Proteins p21(ras) genetics, Leukemia, Myeloid, Acute diagnosis, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute therapy
- Abstract
Acute myeloid leukemia (AML) with inv(3)(q21q26.2)/t(3;3)(q21;q26.2) has a very poor prognosis. Determinants of clinical outcomes and optimal treatment remain uncertain. We retrospectively reviewed 108 cases of AML with inv(3)/t(3;3) and evaluated clinicopathological characteristics and clinical outcomes: 53 newly diagnosed (ND) AML and 55 relapsed/refractory (R/R) AML. Median age was 55 years. White blood cell (WBC) count ≥20x109/L and platelet count ≥140x109/L was observed in 25% and 32% of ND patients, respectively. Anomalies involving chromosome 7 were identified in 56% of patients. The most frequently mutated genes were SF3B1, PTPN11, NRAS, KRAS and ASXL1. In ND patients, the composite complete remission (CRc) rate was 46% overall; 46% with high-intensity treatments and 47% with lowintensity treatments. The 30-day mortality was 14% and 0%, with high- and low-intensity treatment, respectively. In R/R patients, the CRc rate was 14%. Venetoclax based-regimens were associated with a CRc rate of 33%. The 3-year overall survival (OS) was 8.8% and 7.1% in ND and R/R patients, respectively. The 3-year cumulative incidence of relapse was 81.7% overall. Older age, high WBC, high peripheral blast count, secondary AML and KRAS, ASXL1, DNMT3A mutations were associated with worse OS in univariable analyses. The 5-year OS rates were 44% and 6% with or without hematopoietic stem cell transplantation in CR1, respectively. AML with inv(3)/t(3;3) is associated with low CR rates, very high risk of relapse and dismal long-term survival. Intensive chemotherapy and hy pomethylating agents provide similar rates of remission and patients achieving CR benefit from hematopoietic stem cell transplantation in first CR.
- Published
- 2023
- Full Text
- View/download PDF
13. Early mortality in acute myeloid leukemia with KMT2A rearrangement is associated with high risk of bleeding and disseminated intravascular coagulation.
- Author
-
Nguyen D, Kantarjian HM, Short NJ, Qiao W, Ning J, Cuglievan B, Daver NG, DiNardo CD, Jabbour EJ, Kadia TM, Borthakur G, Garcia-Manero G, Konopleva MY, Andreeff M, Ravandi-Kashani F, Sasaki K, and Issa GC
- Subjects
- Adult, Humans, Retrospective Studies, Hemorrhage genetics, Recurrence, Gene Rearrangement, Disseminated Intravascular Coagulation genetics, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics
- Abstract
Background: Acute myeloid leukemia (AML) with rearrangement of lysine methyltransferase 2a gene (KMT2Ar) is characterized by chemotherapy resistance and high rates of relapse. However, additional causes of treatment failure or early mortality have not been well-defined in this entity., Methods: In a retrospective analysis, causes and rates of early mortality following induction treatment were compared between a cohort of adults with KMT2Ar AML (N = 172) and an age-matched cohort of patients with normal karyotype AML (N = 522)., Results: The 60-day mortality in patients with KMT2Ar AML was 15% compared with 7% with normal karyotype (p = .04). We found a significantly higher occurrence of major bleeding events (p = .005) and total bleeding events (p = .001) in KMT2Ar AML compared with diploid AML. Among evaluable patients with KMT2Ar AML, 93% exhibited overt disseminated intravascular coagulopathy compared with 54% of patients with a normal karyotype before death (p = .03). In a multivariate analysis, KMT2Ar and a monocytic phenotypic were the only independent predictors of any bleeding event in patients who died within 60 days (odds ratio, 3.5; 95% CI, 1.4-10.4; p = .03; odds ratio, 3.2; 95% CI, 1-1-9.4; p = .04, respectively)., Conclusion: In conclusion, early recognition and aggressive management of disseminated intravascular coagulopathy and coagulopathy are important considerations that could mitigate the risk of death during induction treatment in KMT2Ar AML., Plain Language Summary: Acute myeloid leukemia (AML) with rearrangement of KMT2A is characterized by chemotherapy resistance and high rates of relapse. However, additional causes of treatment failure or early mortality have not been well-defined in this entity. In this article, that KMT2A-rearranged AML is demonstrably associated with higher early mortality and an increased risk of bleeding and coagulopathy, specifically, disseminated intravascular coagulation, compared with normal karyotype AML. These findings emphasize the importance of monitoring and mitigating coagulopathy in KMT2A-rearranged leukemia similar to what is done in acute promyelocytic leukemia., (© 2023 American Cancer Society.)
- Published
- 2023
- Full Text
- View/download PDF
14. Interrogating bromodomain inhibitor resistance in KMT2A-rearranged leukemia through combinatorial CRISPR screens.
- Author
-
Wright S, Hu J, Wang H, Hyle J, Zhang Y, Du G, Konopleva MY, Kornblau SM, Djekidel MN, Rosikiewicz W, Xu B, Lu R, Yang JJ, and Li C
- Subjects
- Humans, Glycogen Synthase Kinase 3 metabolism, Cell Line, Tumor, Nuclear Proteins genetics, Nuclear Proteins metabolism, Repressor Proteins metabolism, Clustered Regularly Interspaced Short Palindromic Repeats, Leukemia drug therapy, Leukemia genetics
- Abstract
Bromo- and extra-terminal domain inhibitors (BETi) have exhibited therapeutic activities in many cancers. However, the mechanisms controlling BETi response and resistance are not well understood. We conducted genome-wide loss-of-function CRISPR screens using BETi-treated KMT2A-rearranged (KMT2A-r) cell lines. We revealed that Speckle-type POZ protein (SPOP) gene (Speckle Type BTB/POZ Protein) deficiency caused significant BETi resistance, which was further validated in cell lines and xenograft models. Proteomics analysis and a kinase-vulnerability CRISPR screen indicated that cells treated with BETi are sensitive to GSK3 perturbation. Pharmaceutical inhibition of GSK3 reversed the BETi-resistance phenotype. Based on this observation, a combination therapy regimen inhibiting both BET and GSK3 was developed to impede KMT2A-r leukemia progression in patient-derived xenografts in vivo. Our results revealed molecular mechanisms underlying BETi resistance and a promising combination treatment regimen of ABBV-744 and CHIR-98014 by utilizing unique ex vivo and in vivo KMT2A-r PDX models.
- Published
- 2023
- Full Text
- View/download PDF
15. Venetoclax and idasanutlin in relapsed/refractory AML: a nonrandomized, open-label phase 1b trial.
- Author
-
Daver NG, Dail M, Garcia JS, Jonas BA, Yee KWL, Kelly KR, Vey N, Assouline S, Roboz GJ, Paolini S, Pollyea DA, Tafuri A, Brandwein JM, Pigneux A, Powell BL, Fenaux P, Olin RL, Visani G, Martinelli G, Onishi M, Wang J, Huang W, Green C, Ott MG, Hong WJ, Konopleva MY, and Andreeff M
- Subjects
- Humans, Core Binding Factor Alpha 2 Subunit, Bridged Bicyclo Compounds, Heterocyclic adverse effects, Antineoplastic Combined Chemotherapy Protocols adverse effects, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Antineoplastic Agents therapeutic use
- Abstract
This phase 1b trial (NCT02670044) evaluated venetoclax-idasanutlin in patients with relapsed/refractory (R/R) acute myeloid leukemia (AML) ineligible for cytotoxic chemotherapy. Two-dimensional dose escalation (DE, n = 50) was performed for venetoclax daily with idasanutlin on days 1 to 5 in 28-day cycles, followed by dosing schedule optimization (n = 6) to evaluate reduced venetoclax schedules (21-/14-day dosing). Common adverse events (occurring in ≥40% of patients) included diarrhea (87.3% of patients), nausea (74.5%), vomiting (52.7%), hypokalemia (50.9%), and febrile neutropenia (45.5%). During DE, across all doses, composite complete remission (CRc; CR + CR with incomplete blood count recovery + CR with incomplete platelet count recovery) rate was 26.0% and morphologic leukemia-free state (MLFS) rate was 12%. For anticipated recommended phase 2 doses (venetoclax 600 mg + idasanutlin 150 mg; venetoclax 600 mg + idasanutlin 200 mg), the combined CRc rate was 34.3% and the MLFS rate was 14.3%. Pretreatment IDH1/2 and RUNX1 mutations were associated with higher CRc rates (50.0% and 45.0%, respectively). CRc rate in patients with TP53 mutations was 20.0%, with responses noted among those with co-occurring IDH and RUNX1 mutations. In 12 out of 36 evaluable patients, 25 emergent TP53 mutations were observed; 22 were present at baseline with low TP53 variant allele frequency (median 0.0095% [range, 0.0006-0.4]). Venetoclax-idasanutlin showed manageable safety and encouraging efficacy in unfit patients with R/R AML. IDH1/2 and RUNX1 mutations were associated with venetoclax-idasanutlin sensitivity, even in some patients with co-occurring TP53 mutations; most emergent TP53 clones were preexisting. Our findings will aid ongoing/future trials of BCL-2/MDM2 inhibitor combinations. This trial was registered at www.clinicaltrials.gov as #NCT02670044., (© 2023 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
16. A retrospective study of cladribine and low-dose cytarabine-based regimens for the treatment of chronic myelomonocytic leukemia and secondary acute myeloid leukemia.
- Author
-
Bazinet A, Darbaniyan F, Kadia TM, Venugopal S, Kanagal-Shamanna R, DiNardo CD, Borthakur G, Jabbour EJ, Daver NG, Pemmaraju N, Konopleva MY, Ravandi F, Sasaki K, Chien KS, Hammond D, Pierce SA, Kantarjian HM, Garcia-Manero G, and Montalban-Bravo G
- Subjects
- Humans, Retrospective Studies, Cladribine therapeutic use, Prospective Studies, Cytarabine adverse effects, Leukemia, Myelomonocytic, Chronic drug therapy, Leukemia, Myelomonocytic, Chronic genetics, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics
- Abstract
Background: Patients with higher risk chronic myelomonocytic leukemia (CMML) have limited therapeutic options beyond hydroxyurea and hypomethylating agents (HMAs). Regimens based on a backbone of cladribine (CLAD), low-dose cytarabine (LDAC), and an HMA are effective low-intensity therapies for acute myeloid leukemia (AML)., Methods: The authors conducted a retrospective chart review to evaluate the efficacy of CLAD/LDAC/HMA in CMML and secondary acute myeloid leukemia (sAML) arising from CMML. Responses were evaluated according to the 2006 International Working Group criteria for CMML and the 2017 European LeukemiaNet criteria for AML. The overall survival (OS), leukemia-free survival (LFS), and duration of response were evaluated with the Kaplan-Meier method. Patients were stratified on the basis of prior HMA exposure., Results: The authors identified 21 patients with CMML (eight with HMA-naive CMML and 13 with HMA-failure CMML) and 33 patients with sAML (11 with HMA-naive sAML and 22 with HMA-failure sAML) treated with CLAD/LDAC/HMA-based regimens. The CMML cohort was enriched for high-risk features (proliferative type, elevated blasts, and RAS/MAPK mutations). The overall response rate was 33% in CMML (50% in HMA-naive CMML and 23% in HMA-failure CMML) and 48% in sAML (82% in HMA-naive sAML and 32% in HMA-failure sAML). The median OS was 14.4, 8.8, 42.9, and 2.9 months for HMA-naive CMML, HMA-failure CMML, HMA-naive sAML, and HMA-failure sAML, respectively. The median LFS was 14.4 and 3.9 months for HMA-naive CMML and HMA-failure CMML, respectively., Conclusions: CLAD/LDAC/HMA-based regimens are effective in a subset of patients with higher risk CMML and sAML arising from CMML who have not previously experienced HMA failure. These findings must be confirmed in prospective studies., (© 2022 American Cancer Society.)
- Published
- 2023
- Full Text
- View/download PDF
17. Triple combination targeting methyltransferase, BCL-2, and PD-1 facilitates antileukemia responses in acute myeloid leukemia.
- Author
-
Zeng Z, Maiti A, Herbrich S, Cai T, Cavazos A, Manzella T, Ma H, Hayes K, Matthews J, DiNardo CD, Daver NG, and Konopleva MY
- Subjects
- Humans, Aged, Methyltransferases, Programmed Cell Death 1 Receptor therapeutic use, DNA Modification Methylases, Proto-Oncogene Proteins c-bcl-2 genetics, DNA therapeutic use, Antineoplastic Combined Chemotherapy Protocols adverse effects, Antineoplastic Agents therapeutic use, Leukemia, Myeloid, Acute
- Abstract
Background: A recent breakthrough therapy combining the BCL-2 inhibitor venetoclax with hypomethylating agents (HMAs) targeting DNA methyltransferase has improved outcomes for patients with acute myeloid leukemia (AML), but the responses and long-term survival in older/unfit patients and in patients with relapsed/refractory AML remain suboptimal. Recent studies showed that inhibition of BCL-2 or DNA methyltransferase modulates AML T-cell immunity., Methods: By using flow cytometry and time-of-flight mass cytometry, the authors examined the effects of the HMA decitabine combined with the BCL-2 inhibitor venetoclax (DAC/VEN therapy) on leukemia cells and T cells in patients with AML who received DAC/VEN therapy in a clinical trial. The authors investigated the response of programmed cell death protein 1 (PD-1) inhibition in the DAC/VEN-treated samples in vitro and investigated the triple combination of PD-1 inhibition with HMA/venetoclax in the trial patients who had AML., Results: DAC/VEN therapy effectively targeted leukemia cells and upregulated the expression of the immune checkpoint-inhibitory receptor PD-1 in T cells while preserving CD4-positive and CD8-positive memory T cells in a subset of patients with AML who were tested. In vitro PD-1 inhibition potentiated the antileukemia response in DAC/VEN-treated AML samples. The combined use of azacitidine, venetoclax, and nivolumab eliminated circulating blasts and leukemia stem cells/progenitor cells and expanded the percentage of CD8-positive memory T cells in an illustrative patient with relapsed AML who responded to the regimen in an ongoing clinical trial., Conclusions: Immunomodulation by targeting PD-1 enhances the therapeutic effect of combining an HMA and venetoclax in patients with AML., (© 2022 American Cancer Society.)
- Published
- 2023
- Full Text
- View/download PDF
18. Phase II Study of Venetoclax Added to Cladribine Plus Low-Dose Cytarabine Alternating With 5-Azacitidine in Older Patients With Newly Diagnosed Acute Myeloid Leukemia.
- Author
-
Kadia TM, Reville PK, Wang X, Rausch CR, Borthakur G, Pemmaraju N, Daver NG, DiNardo CD, Sasaki K, Issa GC, Ohanian M, Montalban-Bravo G, Short NJ, Jain N, Ferrajoli A, Bhalla KN, Jabbour E, Takahashi K, Malla R, Quagliato K, Kanagal-Shamanna R, Popat UR, Andreeff M, Garcia-Manero G, Konopleva MY, Ravandi F, and Kantarjian HM
- Subjects
- Aged, Humans, Azacitidine, Bridged Bicyclo Compounds, Heterocyclic, Cladribine therapeutic use, Cytarabine, Middle Aged, Aged, 80 and over, Antineoplastic Combined Chemotherapy Protocols adverse effects, Leukemia, Myeloid, Acute drug therapy
- Abstract
Purpose: The combination of venetoclax and 5-azacitidine (5-AZA) for older or unfit patients with acute myeloid leukemia (AML) improves remission rates and survival compared with 5-AZA alone. We hypothesized that the addition of venetoclax to cladribine (CLAD)/low-dose araC (low-dose cytarabine [LDAC]) alternating with 5-AZA backbone may further improve outcomes for older patients with newly diagnosed AML., Methods: This is a phase II study investigating the combination of venetoclax and CLAD/LDAC alternating with venetoclax and 5-AZA in older (≥ 60 years) or unfit patients with newly diagnosed AML. The primary objective was composite complete response (CR) rate (CR plus CR with incomplete blood count recovery); secondary end points were overall survival, disease-free survival (DFS), overall response rate, and toxicity., Results: A total of 60 patients were treated; median age was 68 years (range, 57-84 years). By European LeukemiaNet, 23%, 33%, and 43% were favorable, intermediate, and adverse risk, respectively. Fifty-six of 60 evaluable patients responded (composite CR: 93%) and 84% were negative for measurable residual disease. There was one death (2%) within 4 weeks. With a median follow-up of 22.1 months, the median overall survival and DFS have not yet been reached. The most frequent grade 3/4 nonhematologic adverse events were febrile neutropenia (n = 33) and pneumonia (n = 14). One patient developed grade 4 tumor lysis syndrome., Conclusion: Venetoclax and CLAD/LDAC alternating with venetoclax and 5-AZA is an effective regimen among older or unfit patients with newly diagnosed AML. The rates of overall survival and DFS are encouraging. Further study of this non-anthracycline-containing backbone in younger patients, unfit for intensive chemotherapy, as well as comparisons to standard frontline therapies is warranted.
- Published
- 2022
- Full Text
- View/download PDF
19. Beyond BCL-2 Inhibition in Acute Myloid Leukemia: Other Approaches to Leverage the Apoptotic Pathway.
- Author
-
Maiti A, Carter BZ, Andreeff M, and Konopleva MY
- Subjects
- Apoptosis, Cell Line, Tumor, Humans, Myeloid Cell Leukemia Sequence 1 Protein metabolism, Myeloid Cell Leukemia Sequence 1 Protein therapeutic use, Proto-Oncogene Proteins c-bcl-2 genetics, Proto-Oncogene Proteins c-bcl-2 metabolism, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute metabolism, Tumor Suppressor Protein p53 genetics
- Abstract
BCL-2 inhibition has transformed the therapeutic landscape of acute myeloid leukemia (AML) but is not curative for the majority of patients. Consequently, there has been growing interest in targeting other facets of the apoptotic machinery to improve outcomes. These approaches include targeting the intrinsic and extrinsic apoptotic pathway, inducing apoptosis via p53 activation, and others. Targeting the intrinsic apoptotic pathway includes MCL-1 antagonism and BCL-xL inhibition. MCL-1 can be targeted via direct inhibitors as well as via indirect mechanisms to downregulate MCL-1 including inhibition of cyclin dependent kinases and Nedd8 activating enzyme. The extrinsic apoptotic pathway could be harnessed via inhibition of inhibitor of apoptosis proteins, agonism of the TNF-related apoptosis-inducing ligand receptors and inhibiting FLICE-like inhibitor protein. Approaches inducing p53-mediated apoptosis are being evaluated using inhibitors of MDM2, dual inhibitor of MDM2/X in TP53 wild-type AML and p53 reactivators in TP53-mutant myeloid disorders. Several such agents are in early clinical development and rationale combinations of these agents may help improving outcomes for patients with AML., (Copyright © 2022. Published by Elsevier Inc.)
- Published
- 2022
- Full Text
- View/download PDF
20. Venetoclax combined with FLAG-IDA induction and consolidation in newly diagnosed acute myeloid leukemia.
- Author
-
DiNardo CD, Lachowiez CA, Takahashi K, Loghavi S, Kadia T, Daver N, Xiao L, Adeoti M, Short NJ, Sasaki K, Wang SA, Borthakur G, Issa G, Maiti A, Alvarado Y, Pemmaraju N, Bravo GM, Masarova L, Yilmaz M, Jain N, Andreeff M, Garcia-Manero G, Kornblau S, Ravandi F, Jabbour E, Konopleva MY, and Kantarjian HM
- Subjects
- Adult, Aged, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Cytarabine therapeutic use, Granulocyte Colony-Stimulating Factor therapeutic use, Humans, Middle Aged, Prospective Studies, Remission Induction, Vidarabine therapeutic use, Young Adult, Bridged Bicyclo Compounds, Heterocyclic adverse effects, Bridged Bicyclo Compounds, Heterocyclic therapeutic use, Idarubicin therapeutic use, Leukemia, Myeloid, Acute diagnosis, Leukemia, Myeloid, Acute drug therapy, Sulfonamides adverse effects, Sulfonamides therapeutic use
- Abstract
Multi-agent induction chemotherapy (IC) improves response rates in younger patients with acute myeloid leukemia (AML); however, relapse remains the principal cause of treatment failure. Improved induction regimens are needed. A prospective single-center phase Ib/II study evaluating fludarabine, cytarabine, G-CSF, and idarubicin combined with venetoclax (FLAG-IDA + VEN) in patients with newly diagnosed (ND) or relapsed/refractory AML. The primary efficacy endpoint was assessment of overall activity (overall response rate [ORR]: complete remission [CR] + CR with partial hematologic recovery [CRh] + CR with incomplete hematologic recovery [CRi] + morphologic leukemia free state + partial response). Secondary objectives included additional assessments of efficacy, overall survival (OS), and event-free survival (EFS). Results of the expanded ND cohort with additional follow-up are reported. Forty-five patients (median age: 44 years [range 20-65]) enrolled. ORR was 98% (N = 44/45; 95% credible interval 89.9%-99.7%). Eighty-nine percent (N = 40/45) of patients attained a composite CR (CRc + CRh + CRi) including 93% (N = 37/40) who were measurable residual disease (MRD) negative. Twenty-seven (60%) patients transitioned to allogeneic stem cell transplant (alloHSCT). Common non-hematologic adverse events included febrile neutropenia (44%; N = 20), pneumonia (22%, N = 10), bacteremia (18%, N = 8), and skin/soft tissue infections (44%, N = 20). After a median follow-up of 20 months, median EFS and OS were not reached. Estimated 24-month EFS and OS were 64% and 76%, respectively. FLAG-IDA + VEN is an active regimen in ND-AML capable of producing high MRD-negative remission rates and enabling transition to alloHSCT when appropriate in most patients. Toxicities were as expected with IC and were manageable. Estimated 24-month survival appears favorable compared to historical IC benchmarks., (© 2022 Wiley Periodicals LLC.)
- Published
- 2022
- Full Text
- View/download PDF
21. Idasanutlin plus cytarabine in relapsed or refractory acute myeloid leukemia: results of the MIRROS trial.
- Author
-
Konopleva MY, Röllig C, Cavenagh J, Deeren D, Girshova L, Krauter J, Martinelli G, Montesinos P, Schäfer JA, Ottmann O, Petrini M, Pigneux A, Rambaldi A, Recher C, Rodriguez-Veiga R, Taussig D, Vey N, Yoon SS, Ott M, Muehlbauer S, Beckermann BM, Catalani O, Genevray M, Mundt K, Jamois C, Fenaux P, and Wei AH
- Subjects
- Adult, Antineoplastic Combined Chemotherapy Protocols adverse effects, Humans, Pyrrolidines, para-Aminobenzoates therapeutic use, Cytarabine, Leukemia, Myeloid, Acute
- Abstract
The phase 3 MIRROS (MDM2 antagonist Idasanutlin in Relapsed or Refractory acute myeloid leukemia [AML] for Overall Survival) trial (NCT02545283) evaluated the efficacy and safety of the small-molecule MDM2 antagonist idasanutlin plus cytarabine in patients with relapsed/refractory (R/R) AML. Adults (n = 447) with R/R AML whose disease relapsed or was refractory after ≤2 prior induction regimens as initial treatment or following salvage chemotherapy regimen, with Eastern Cooperative Oncology Group performance status ≤2 were enrolled regardless of TP53 mutation status and randomly assigned 2:1 to idasanutlin 300 mg or placebo orally twice daily plus cytarabine 1 g/m2 IV on days 1 to 5 of 28-day cycles. At primary analysis (cutoff, November 2019), 436 patients were enrolled, including 355 in the TP53 wild-type intention-to-treat (TP53WT-ITT) population. The primary endpoint, overall survival in the TP53WT-ITT population, was not met (median, 8.3 vs 9.1 months with idasanutlin-cytarabine vs placebo-cytarabine; stratified hazard ratio [HR], 1.08; 95% confidence interval [CI], 0.81-1.45; P = .58). The complete remission (CR) rate, a key secondary endpoint, was 20.3% vs 17.1% (odds ratio [OR], 1.23; 95% CI, 0.70-2.18). The overall response rate (ORR) was 38.8% vs 22.0% (OR, 2.25; 95% CI, 1.36-3.72). Common any-grade adverse events (≥10% incidence in any arm) were diarrhea (87.0% vs 32.9%), febrile neutropenia (52.8% vs 49.3%), and nausea (52.5% vs 31.5%). In summary, despite improved ORR, adding idasanutlin to cytarabine did not improve overall survival or CR rates in patients with R/R AML., (© 2022 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
22. Pneumonitis after immune checkpoint inhibitor therapies in patients with acute myeloid leukemia: A retrospective cohort study.
- Author
-
Sheshadri A, Goizueta AA, Shannon VR, London D, Garcia-Manero G, Kantarjian HM, Ravandi-Kashani F, Kadia TM, Konopleva MY, DiNardo CD, Pierce S, Zarifa A, Albittar AA, Zhong LL, Akhmedzhanov FO, Arain MH, Alfayez M, Alotaibi A, Altan M, Naing A, Mendoza TR, Godoy MCB, Shroff G, Kim ST, Faiz SA, Kontoyiannis DP, Khawaja F, Jennings K, and Daver NG
- Subjects
- Dyspnea chemically induced, Female, Humans, Immune Checkpoint Inhibitors adverse effects, Retrospective Studies, Antineoplastic Agents, Immunological therapeutic use, Leukemia, Myeloid, Acute complications, Leukemia, Myeloid, Acute drug therapy, Lung Neoplasms drug therapy, Pneumonia chemically induced, Pneumonia diagnosis, Pneumonia epidemiology
- Abstract
Background: Immune checkpoint inhibitors (ICI), combined with hypomethylating agents, can be used to treat acute myeloid leukemia (AML), but this strategy results in a high rate of pneumonitis. The authors sought to determine risk factors for pneumonitis development and whether pneumonitis increased mortality., Methods: The authors conducted a retrospective review of 258 AML patients who received ICI-containing regimens from 2016 to 2018. A multidisciplinary adjudication committee diagnosed pneumonia and pneumonitis by reviewing symptoms, imaging, microbiology, and response to therapies. To measure risk factors for pneumonitis and mortality, multivariate Cox proportional hazards models were constructed. Pneumonia, pneumonitis, and disease progression were modeled as a time-dependent variable and incorporated a standard risk set modifying variables into the models., Results: Thirty patients developed pneumonitis (12%). Of these, 17 had partial or complete resolution, whereas 13 patients died from pneumonitis. Increasing age (hazard ratio [HR], 1.04 per year; 95% confidence interval [CI], 1.00-1.08), and baseline shortness of breath increased pneumonitis risk (HR, 2.51; 95% CI, 1.13-5.55). Female sex (HR, 0.33; 95% CI, 0.15-0.70) and increasing platelet count (HR, 0.52 per log-unit increase; 95% CI, 0.30-0.92) decreased pneumonitis risk. In adjusted models, ICI-related pneumonitis significantly increased mortality (HR, 2.84; 95% CI, 1.84-4.37)., Conclusions: ICI-related pneumonitis occurs at a high rate in AML patients and increases mortality., Lay Summary: Immune checkpoint inhibitors (ICIs) remove inhibitory signals that reduce T-cell function and allow T-cells to better attack cancer cells. In acute myeloid leukemia (AML), the effectiveness of ICIs is limited in part by inflammation of the lung, called pneumonitis. This study reviewed 258 patients with AML who received ICIs and identified 30 patients who developed pneumonitis, nearly half of whom died. Older age and baseline shortness of breath increased pneumonitis risk, whereas female sex and higher baseline platelet counts decreased pneumonitis risk. Pneumonitis increased mortality by nearly 3-fold. This work highlights the significant harm imposed by pneumonitis after ICI therapies., (© 2022 American Cancer Society.)
- Published
- 2022
- Full Text
- View/download PDF
23. Treatment-free remission after ceasing venetoclax-based therapy in patients with acute myeloid leukemia.
- Author
-
Chua CC, Hammond D, Kent A, Tiong IS, Konopleva MY, Pollyea DA, DiNardo CD, and Wei AH
- Subjects
- Aged, Bridged Bicyclo Compounds, Heterocyclic therapeutic use, Humans, Retrospective Studies, Sulfonamides, Antineoplastic Combined Chemotherapy Protocols adverse effects, Leukemia, Myeloid, Acute genetics
- Abstract
The clinical benefit of adding venetoclax (VEN) to hypomethylating agents or low-dose cytarabine in older and/or unfit patients with newly diagnosed acute myeloid leukemia (AML) has been confirmed in phase 3 studies. With the increased uptake of VEN-based therapies for patients with AML, a pertinent question is whether treatment can be safely ceased among patients who have achieved sustained remission. We hypothesized that a proportion of patients opting to cease therapy may benefit from a treatment-free remission (TFR) period without indefinite treatment. We report the retrospective outcomes of 29 patients in remission for a minimum of 12 months on VEN-based therapy, with 55% continuing therapy until disease progression and 45% electively ceasing treatment (STOP). With follow-up exceeding 5 years, we observed a median TFR lasting 45.8 months among the STOP cohort, with >50% of patients still in sustained remission at the data cutoff. The risk of relapse and duration of relapse-free and overall survival were similar between the 2 cohorts. Factors favoring sustained TFR within the cohort included NPM1 and/or IDH2 mutation at diagnosis, complete remission without measurable residual disease, and at least 12 months of VEN-based combination therapy prior to treatment cessation., (© 2022 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
24. Venetoclax resistance: mechanistic insights and future strategies.
- Author
-
Ong F, Kim K, and Konopleva MY
- Abstract
Acute myeloid leukemia (AML) is historically associated with poor prognosis, especially in older AML patients unfit for intensive chemotherapy. The development of Venetoclax, a potent oral BH3 (BCL-2 homology domain 3) mimetic, has transformed the AML treatment. However, the short duration of response and development of resistance remain major concerns. Understanding mechanisms of resistance is pivotal to devising new strategies and designing rational drug combination regimens. In this review, we will provide a comprehensive summary of the known mechanisms of resistance to Venetoclax and discuss Venetoclax-based combination therapies. Key contributing factors to Venetoclax resistance include dependencies on alternative anti-apoptotic BCL-2 family proteins and selection of the activating kinase mutations. Mutational landscape governing response to Venetoclax and strategic approaches developed considering current knowledge of mechanisms of resistance will be addressed., Competing Interests: Marina Konopleva: Consultant for AbbVie, Genentech, F. Hoffman La-Roche, Stemline Therapeutics, Amgen, Forty-Seven, KisoJi, Janssen; serves as advisory board member for Stemline Therapeutics, F. Hoffman La-Roche, Janssen; holds shares from Reata Pharmaceuticals; honoraria from Forty-Seven and F. Hoffman La-Roche; research funding from AbbVie, Genentech, F. Hoffman La-Roche, Eli Lilly, Cellectis, Calithera, Stemline Therapeutics, Ablynx, Agios, Ascentage, Astra Zeneca, Rafael Pharmaceutical, Sanofi, and Forty-Seven., (© The Author(s) 2022.)
- Published
- 2022
- Full Text
- View/download PDF
25. Improved outcomes among newly diagnosed patients with FMS-like tyrosine kinase 3 internal tandem duplication mutated acute myeloid leukemia treated with contemporary therapy: Revisiting the European LeukemiaNet adverse risk classification.
- Author
-
Reville PK, Sasaki K, Kantarjian HM, Daver NG, Yilmaz M, Dinardo CD, Short NJ, Borthakur G, Pemmaraju N, Mehta RS, Pierce S, Konoplev SN, Khoury JD, Garcia-Manero G, Konopleva MY, Jabbour E, Ravandi F, and Kadia TM
- Subjects
- Allografts, Disease-Free Survival, Female, Humans, Infant, Male, Mutation, Survival Rate, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Leukemia, Myeloid, Acute enzymology, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute mortality, Leukemia, Myeloid, Acute therapy, Risk Assessment, Stem Cell Transplantation, fms-Like Tyrosine Kinase 3 genetics
- Abstract
Mutations in fms-like tyrosine kinase 3 (FLT3) gene are common genomic alterations in acute myeloid leukemia (AML). FLT3 internal tandem duplication mutations (FLT3-ITD) have consistently been shown to be adversely prognostic, particularly those with high allelic ratio (AR). Current AML treatment strategies, including high dose cytarabine, purine analogs, FLT3 inhibitors (FLT3i), and with or without allogeneic stem cell transplant (SCT) have been shown to improve the outcomes in patients with FLT3 mutations. We analyzed a consecutive cohort of newly diagnosed patients with AML treated at a large academic medical center from January 2012 to January 2020. A total of 1576 patients with a new diagnosis of AML were reviewed. Among these, 1438 (91%) had molecular testing for FLT3 mutations and 21% (304/1438) had an FLT3 mutation, including 17% with an FLT3-ITD mutation. We show that FLT3-ITD high AR with NPM1 wild-type have significantly improved survival compared with other European LeukemiaNet (ELN) adverse risk disease. In multivariable cox proportional hazards model of patients receiving intensive or low-intensity induction regimens, FLT3 mutations did not have prognostic significance. The use of allogeneic SCT in CR1 for patients with FLT3 mutations appears to improve survival, particularly in those with ELN adverse risk disease. Overall, this data highlights the changing prognostic impact of FLT3 mutations in a contemporary era with appropriate use of induction therapy combined with targeted agents and allogenic SCT., (© 2022 Wiley Periodicals LLC.)
- Published
- 2022
- Full Text
- View/download PDF
26. How We Incorporate Venetoclax in Treatment Regimens for Acute Myeloid Leukemia.
- Author
-
Maiti A and Konopleva MY
- Subjects
- Bridged Bicyclo Compounds, Heterocyclic therapeutic use, Humans, Sulfonamides therapeutic use, Antineoplastic Combined Chemotherapy Protocols adverse effects, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics
- Abstract
Abstract: Venetoclax has transformed the therapeutic landscape of acute myeloid leukemia (AML). Hypomethylating agents with venetoclax (HMA-VEN) have significantly improved outcomes and have become the standard therapy for older/unfit patients with newly diagnosed AML and are comparable to intensive chemotherapy in salvage setting. Venetoclax with intensive chemotherapy have shown high response rates in both frontline and salvage setting in younger patients, and triplet combinations with HMA-VEN and FLT3 inhibitors have shown encouraging results in FLT3mut AML. While patients with NPM1mut, IDH1/2mut experience favorable outcomes, those with TP53mut and secondary AML may experience minimal benefit from the addition of venetoclax. Despite improved outcomes, severe cytopenias and infectious complications are common with venetoclax-based regimens. Early response evaluation, dose reductions, venetoclax interruptions, use of growth factors, and prophylactic antimicrobials may minimize such myelosuppression and risk of infections. Outcomes after failure of frontline HMA-VEN are dismal, and novel approaches are needed to abrogate primary and acquired resistance., Competing Interests: Conflicts of Interest and Source of Funding: This work was supported in part by the MD Anderson Cancer Center Support Grant CA016672 from the National Cancer Institute. A.M. was supported in part by the American Society of Clinical Oncology Young Investigator Award. M.Y.K. was supported in part by the Research Project Grant Program (R01CA235622) from the National Institutes of Health. A.M. has received research funding from Celgene Corporation, Conquer Cancer Foundation. M.Y.K. has received grants from NIH, NCI, Abbvie, Genentech, Stemline Therapeutics, Forty-Seven, Eli Lilly, Cellectis, Calithera, Ablynx, and AstraZeneca; consulting/honorarium from AbbVie, Genentech, F. Hoffman La-Roche, Stemline Therapeutics, Amgen, Forty-Seven, Kisoji; clinical trial support from Ascentage; and has stocks/royalties in Reata Pharmaceutical., (Copyright © 2022 The Author(s). Published by Wolters Kluwer Health, Inc.)
- Published
- 2022
- Full Text
- View/download PDF
27. Allogeneic hematopoietic cell transplantation for patients with blastic plasmacytoid dendritic cell neoplasm (BPDCN).
- Author
-
Bashir Q, Milton DR, Popat UR, Kebriaei P, Hosing C, Khouri IF, Rezvani K, Nieto Y, Oran B, Srour SA, Saini NY, Olson AL, Ahmed S, Al-Atrash G, Rondon G, Konopleva MY, Champlin RE, Shpall EJ, Qazilbash MH, and Pemmaraju N
- Subjects
- Acute Disease, Adult, Dendritic Cells, Humans, Remission Induction, Transplantation Conditioning adverse effects, Graft vs Host Disease etiology, Hematologic Neoplasms complications, Hematopoietic Stem Cell Transplantation adverse effects, Myeloproliferative Disorders, Skin Neoplasms pathology
- Abstract
Blastic Plasmacytoid Dendritic Cell Neoplasm (BPDCN) is an aggressive hematological malignancy; however, some patients achieve durable remission with allogeneic hematopoietic cell transplantation (allo-HCT). We report on all 17 patients with BPDCN who underwent allo-HCT at our center between 2000 and 2020. The median age was 39 (18-67) years. All (n = 16, 94%), except one patient, had systemic disease involving bone marrow and/or other organs. Ten patients (59%) were in first complete remission (CR1) at allo-HCT. The donor source was matched related or unrelated in ten (59%) and alternate donor in seven (41%) patients. Five (31%) patients developed acute graft-versus-host disease (GVHD), all grade I-II. The cumulative incidence (CI) of chronic GVHD at five-year was 34%. The CI of non-relapse mortality at one-year was 29%. Progression-free survival (PFS) rates at two-year and five-year were 49% (95% CI = 22-71%) and 39% (95% CI = 14-64%), respectively. The two-year and five-year overall survival (OS) rates were 65% (95% CI = 38-82%) and 40% (95% CI = 12-68%), respectively. The five-year rate for both PFS and OS was 80% in CR1 patients versus 0% in patients not in CR1. In conclusion, allo-HCT provides long-lasting remissions in BPDCN patients, particularly when performed in CR1., (© 2021. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2022
- Full Text
- View/download PDF
28. 2021 Update on MRD in acute myeloid leukemia: a consensus document from the European LeukemiaNet MRD Working Party.
- Author
-
Heuser M, Freeman SD, Ossenkoppele GJ, Buccisano F, Hourigan CS, Ngai LL, Tettero JM, Bachas C, Baer C, Béné MC, Bücklein V, Czyz A, Denys B, Dillon R, Feuring-Buske M, Guzman ML, Haferlach T, Han L, Herzig JK, Jorgensen JL, Kern W, Konopleva MY, Lacombe F, Libura M, Majchrzak A, Maurillo L, Ofran Y, Philippe J, Plesa A, Preudhomme C, Ravandi F, Roumier C, Subklewe M, Thol F, van de Loosdrecht AA, van der Reijden BA, Venditti A, Wierzbowska A, Valk PJM, Wood BL, Walter RB, Thiede C, Döhner K, Roboz GJ, and Cloos J
- Subjects
- Europe, Flow Cytometry methods, High-Throughput Nucleotide Sequencing methods, Humans, Leukemia, Myeloid, Acute genetics, Neoplasm, Residual genetics, Prognosis, Leukemia, Myeloid, Acute diagnosis, Neoplasm, Residual diagnosis
- Abstract
Measurable residual disease (MRD) is an important biomarker in acute myeloid leukemia (AML) that is used for prognostic, predictive, monitoring, and efficacy-response assessments. The European LeukemiaNet (ELN) MRD Working Party evaluated standardization and harmonization of MRD in an ongoing manner and has updated the 2018 ELN MRD recommendations based on significant developments in the field. New and revised recommendations were established during in-person and online meetings, and a 2-stage Delphi poll was conducted to optimize consensus. All recommendations are graded by levels of evidence and agreement. Major changes include technical specifications for next-generation sequencing-based MRD testing and integrative assessments of MRD irrespective of technology. Other topics include use of MRD as a prognostic and surrogate end point for drug testing; selection of the technique, material, and appropriate time points for MRD assessment; and clinical implications of MRD assessment. In addition to technical recommendations for flow- and molecular-MRD analysis, we provide MRD thresholds and define MRD response, and detail how MRD results should be reported and combined if several techniques are used. MRD assessment in AML is complex and clinically relevant, and standardized approaches to application, interpretation, technical conduct, and reporting are of critical importance., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
29. Outcomes of acute lymphoblastic leukemia with KMT2A (MLL) rearrangement: the MD Anderson experience.
- Author
-
Richard-Carpentier G, Kantarjian HM, Tang G, Yin CC, Khoury JD, Issa GC, Haddad F, Jain N, Ravandi F, Short NJ, DiNardo CD, Takahashi K, Konopleva MY, Daver NG, Kadia T, Garcia-Manero G, Garris R, O'Brien S, and Jabbour E
- Subjects
- Adolescent, Adult, Aged, Humans, Middle Aged, Neoplasm, Residual, Prognosis, Remission Induction, Young Adult, Hematopoietic Stem Cell Transplantation, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma therapy
- Abstract
Acute lymphoblastic leukemia (ALL) with t(4;11)(q21;q23)-KMT2A-AFF1 is associated with a poor prognosis. The impact of KMT2A rearrangements other than t(4;11) is uncertain, and the benefit of allogeneic stem cell transplantation (HSCT) is unclear. We reviewed adult patients with ALL treated at our institution from 1984 to 2019 and identified 50 out of 1102 (5%) with KMT2A rearrangement, including 42 (84%) with t(4;11)/KMT2A-AFF1 and 8 (16%) with other gene partners. The median age was 45 years (range, 18-78 years); median white blood cell count was 109.0 3 109/L (range, 0.5-1573.0). The complete remission (CR) rate was 88%, and the rate of measurable residual disease negativity by flow cytometry at CR was 41% (76% overall during follow-up). At the last follow-up, 14 patients were alive. The 5-year overall survival (OS) rate was 18% (95% confidence interval [CI], 9% to 35%), with no difference between t(4;11) and other KMT2A rearrangements (P 5 .87). In a 4-month landmark analysis, the 5-year OS rate was 32% (95% CI, 14% to 70%) in patients who underwent HSCT vs 11% (95% CI, 3-39) in others (P 5 .10). Our study confirms the poor prognosis of ALL with any KMT2A rearrangement and the role of HSCT in these patients., (© 2021 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
30. DYRK1a mediates BAFF-induced noncanonical NF-κB activation to promote autoimmunity and B-cell leukemogenesis.
- Author
-
Li Y, Xie X, Jie Z, Zhu L, Yang JY, Ko CJ, Gao T, Jain A, Jung SY, Baran N, Konopleva MY, Cheng X, and Sun SC
- Subjects
- Animals, Autoimmune Diseases immunology, Autoimmune Diseases pathology, B-Lymphocytes immunology, B-Lymphocytes pathology, Carcinogenesis immunology, Carcinogenesis pathology, Cell Line, Tumor, Humans, Leukemia, B-Cell pathology, Mice, Mice, Inbred C57BL, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma immunology, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma pathology, Dyrk Kinases, Autoimmunity, B-Cell Activating Factor immunology, Leukemia, B-Cell immunology, NF-kappa B immunology, Protein Serine-Threonine Kinases immunology, Protein-Tyrosine Kinases immunology
- Abstract
B-cell-activating factor (BAFF) mediates B-cell survival and, when deregulated, contributes to autoimmune diseases and B-cell malignancies. The mechanism connecting BAFF receptor (BAFFR) signal to downstream pathways and pathophysiological functions is not well understood. Here we identified DYRK1a as a kinase that responds to BAFF stimulation and mediates BAFF-induced B-cell survival. B-cell-specific DYRK1a deficiency causes peripheral B-cell reduction and ameliorates autoimmunity in a mouse model of lupus. An unbiased screen identified DYRK1a as a protein that interacts with TRAF3, a ubiquitin ligase component mediating degradation of the noncanonical nuclear factor (NF)-κB-inducing kinase (NIK). DYRK1a phosphorylates TRAF3 at serine-29 to interfere with its function in mediating NIK degradation, thereby facilitating BAFF-induced NIK accumulation and noncanonical NF-κB activation. Interestingly, B-cell acute lymphoblastic leukemia (B-ALL) cells express high levels of BAFFR and respond to BAFF for noncanonical NF-κB activation and survival in a DYRK1a-dependent manner. Furthermore, DYRK1a promotes a mouse model of B-ALL through activation of the noncanonical NF-κB pathway. These results establish DYRK1a as a critical BAFFR signaling mediator and provide novel insight into B-ALL pathogenesis., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
31. Prognostic impact of conventional cytogenetics in acute myeloid leukemia treated with venetoclax and decitabine.
- Author
-
Venugopal S, Maiti A, DiNardo CD, Qiao W, Ning J, Loghavi S, Daver NG, Kadia TM, Rausch CR, Alvarado Y, Ohanian M, Sasaki K, Short NJ, Takahashi K, Yilmaz M, Ravandi F, Kantarjian HM, and Konopleva MY
- Subjects
- Antineoplastic Combined Chemotherapy Protocols adverse effects, Azacitidine therapeutic use, Cytogenetic Analysis, Decitabine therapeutic use, Humans, Prognosis, Sulfonamides, Bridged Bicyclo Compounds, Heterocyclic adverse effects, Leukemia, Myeloid, Acute diagnosis, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics
- Published
- 2021
- Full Text
- View/download PDF
32. Ten-day decitabine with venetoclax versus intensive chemotherapy in relapsed or refractory acute myeloid leukemia: A propensity score-matched analysis.
- Author
-
Maiti A, DiNardo CD, Qiao W, Kadia TM, Jabbour EJ, Rausch CR, Daver NG, Short NJ, Borthakur G, Pemmaraju N, Yilmaz M, Alvarado Y, Montalbano KS, Wade A, Maduike RE, Guerrero JA, Vaughan K, Bivins CA, Pierce S, Ning J, Ravandi F, Kantarjian HM, and Konopleva MY
- Subjects
- Adult, Bridged Bicyclo Compounds, Heterocyclic, Cytarabine, Decitabine, Humans, Propensity Score, Sulfonamides, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Leukemia, Myeloid, Acute
- Abstract
Background: Relapsed/refractory (R/R) acute myeloid leukemia (AML) has poor outcomes. Although lower-intensity venetoclax-containing regimens are standard for older/unfit patients with newly diagnosed AML, it is unknown how such regimens compare with intensive chemotherapy (IC) for R/R AML., Methods: Outcomes of R/R AML treated with 10-day decitabine and venetoclax (DEC10-VEN) were compared with IC-based regimens including idarubicin with cytarabine, with or without cladribine, clofarabine, or fludarabine, with or without additional agents. Propensity scores derived from patient baseline characteristics were used to match DEC10-VEN and IC patients to minimize bias., Results: Sixty-five patients in the DEC10-VEN cohort were matched to 130 IC recipients. The median ages for the DEC10-VEN and IC groups were 64 and 58 years, respectively, and baseline characteristics were balanced between the 2 cohorts. DEC10-VEN conferred significantly higher responses compared with IC including higher overall response rate (60% vs 36%; odds ratio [OR], 3.28; P < .001), complete remission with incomplete hematologic recovery (CRi, 19% vs 6%; OR, 3.56; P = .012), minimal residual disease negativity by flow cytometry (28% vs 13%; OR, 2.48; P = .017), and lower rates of refractory disease. DEC10-VEN led to significantly longer median event-free survival compared with IC (5.7 vs 1.5 months; hazard ratio [HR], 0.46; 95% CI, 0.30-0.70; P < .001), as well as median overall survival (OS; 6.8 vs 4.7 months; HR, 0.56; 95% CI, 0.37-0.86; P = .008). DEC10-VEN was independently associated with improved OS compared with IC in multivariate analysis. Exploratory analysis for OS in 27 subgroups showed that DEC10-VEN was comparable with IC as salvage therapy for R/R AML., Conclusion: DEC10-VEN represents an appropriate salvage therapy and may offer better responses and survival compared with IC in adults with R/R AML., (© 2021 American Cancer Society.)
- Published
- 2021
- Full Text
- View/download PDF
33. Clonal dynamics and clinical implications of postremission clonal hematopoiesis in acute myeloid leukemia.
- Author
-
Tanaka T, Morita K, Loghavi S, Wang F, Furudate K, Sasaki Y, Little L, Gumbs C, Matthews J, Daver N, Pemmaraju N, DiNardo CD, Sasaki K, Yilmaz M, Kadia TM, Ravandi F, Konopleva MY, Kantarjian HM, Champlin RE, Al-Atrash G, Garcia-Manero G, Wang SA, Futreal PA, and Takahashi K
- Subjects
- Adult, Aged, Aged, 80 and over, Antineoplastic Agents therapeutic use, DNA-Binding Proteins genetics, Dioxygenases genetics, Female, Humans, Leukemia, Myeloid, Acute therapy, Male, Middle Aged, Remission Induction, Stem Cell Transplantation, Young Adult, Clonal Hematopoiesis, Leukemia, Myeloid, Acute genetics, Mutation
- Abstract
Although clonal hematopoiesis (CH) can precede the development of acute myeloid leukemia (AML), it can also persist after achieving remission. Long-term clonal dynamics and clinical implications of persistent CH are not well understood. Here, we studied the prevalence, dynamics, and clinical implications of postremission CH in 164 AML patients who attained complete remission after induction chemotherapies. Postremission CH was identified in 79 (48%) patients. Postremission CH persisted long term in 91% of the trackable patients despite treatment with various types of consolidation and maintenance therapies. Postremission CH was eradicated in 20 out of 21 (95%) patients who underwent allogeneic stem cell transplant. Although patients with postremission CH as a group had comparable hematopoiesis with those without it, patients with persistent TET2 mutations showed significant neutropenia long term. Postremission CH had little impact on relapse risk, nonrelapse mortality, and incidence of atherosclerotic cardiovascular disease, although the clinical impact of post-CR CH was heterogeneous among different mutations. These data suggest that although residual clonal hematopoietic stem cells are generally resistant to consolidation and maintenance therapies, they retain the ability to maintain normal hematopoiesis and have little impact on clinical outcomes. Larger study is needed to dissect the gene-specific heterogeneity., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
34. Outcomes of TP53-mutant acute myeloid leukemia with decitabine and venetoclax.
- Author
-
Kim K, Maiti A, Loghavi S, Pourebrahim R, Kadia TM, Rausch CR, Furudate K, Daver NG, Alvarado Y, Ohanian M, Sasaki K, Short NJ, Takahashi K, Yilmaz M, Tang G, Ravandi F, Kantarjian HM, DiNardo CD, and Konopleva MY
- Subjects
- Aged, Aged, 80 and over, Bridged Bicyclo Compounds, Heterocyclic therapeutic use, Decitabine adverse effects, Humans, Middle Aged, Sulfonamides, Tumor Suppressor Protein p53 genetics, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics
- Abstract
Background: TP53 mutation (TP53
mut ) confers an adverse prognosis in acute myeloid leukemia (AML). Venetoclax with hypomethylating agents is a current standard for older patients; however, recent reports suggest that TP53mut confers resistance to venetoclax. The authors investigated the outcomes of patients with TP53mut AML who were treated with a 10-day decitabine and venetoclax (DEC10-VEN) (ClinicalTrials.gov identifier NCT03404193)., Methods: Patients with newly diagnosed AML received decitabine 20 mg/m2 for 10 days every 4 to 6 weeks for induction, followed by decitabine for 5 days after response. The venetoclax dose was 400 mg daily. TP53mut was identified in bone marrow samples using next-generation sequencing, with sensitivity of 5%. Outcomes were analyzed according to European LeukemiaNet 2017 guidelines., Results: Among 118 patients (median age, 72 years; age range, 49-89 years), 63 (53%) had secondary AML, 39 (33%) had AML with complex karyotype, and 35 (30%) had TP53mut AML. The median TP53 variant allele frequency was 32% (interquartile range, 16%-65%), 8 patients (23%) had only a single TP53 mutation, 15 (43%) had multiple mutations, and 12 (34%) had mutation and deletion. Outcomes were significantly worse in patients who had TP53mut AML compared with those who had wild-type TP53 AML, with an overall response rate of 66% vs 89% (P = .002), a complete response/complete response with incomplete hematologic recovery rate of 57% vs 77% (P = .029), and a 60-day mortality of 26% vs 4% (P < .001), respectively. Patients with TP53mut versus wild-type TP53 had shorter overall survival at 5.2 versus 19.4 months, respectively (hazard ratio, 4.67; 95% CI, 2.44-8.93; P < .0001), and shorter relapse-free survival at 3.4 versus 18.9 months (hazard ratio, 4.80; 95% CI, 1.97-11.69; P < .0001), respectively. Outcomes with DEC10-VEN in patients with TP53mut AML were comparable to historical results with 10-day decitabine alone., Conclusions: Patients with TP53mut AML have lower response rates and shorter survival with DEC10-VEN., (© 2021 American Cancer Society.)- Published
- 2021
- Full Text
- View/download PDF
35. A phase 1b/2 study of azacitidine with PD-L1 antibody avelumab in relapsed/refractory acute myeloid leukemia.
- Author
-
Saxena K, Herbrich SM, Pemmaraju N, Kadia TM, DiNardo CD, Borthakur G, Pierce SA, Jabbour E, Wang SA, Bueso-Ramos C, Loghavi S, Tang G, Cheung CM, Alexander L, Kornblau S, Andreeff M, Garcia-Manero G, Ravandi F, Konopleva MY, and Daver N
- Subjects
- Adult, Aged, Aged, 80 and over, Antibodies, Monoclonal, Humanized therapeutic use, B7-H1 Antigen, Humans, Middle Aged, Young Adult, Azacitidine adverse effects, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute pathology
- Abstract
Background: Patients with relapsed/refractory (R/R) acute myeloid leukemia (AML) have limited treatment options. In preclinical models of AML, inhibition of the PD-1/PD-L1 axis demonstrated antileukemic activity. Avelumab is an anti-PD-L1 immune checkpoint inhibitor (ICI) approved in multiple solid tumors. The authors conducted a phase 1b/2 clinical trial to assess the safety and efficacy of azacitidine with avelumab in patients with R/R AML., Methods: Patients aged ≥18 years who had R/R AML received azacitidine 75 mg/m
2 on days 1 through 7 and avelumab on days 1 and 14 of 28-day cycles., Results: Nineteen patients were treated. The median age was 66 years (range, 22-83 years), 100% had European LeukemiaNet 2017 adverse-risk disease, and 63% had prior exposure to a hypomethylating agent. Avelumab was dosed at 3 mg/kg for the first 7 patients and at 10 mg/kg for the subsequent 12 patients. The most common grade ≥3 treatment-related adverse events were neutropenia and anemia in 2 patients each. Two patients experienced immune-related adverse events of grade 2 and grade 3 pneumonitis, respectively. The overall complete remission rate was 10.5%, and both were complete remission with residual thrombocytopenia. The median overall survival was 4.8 months. Bone marrow blasts were analyzed for immune-related markers by mass cytometry and demonstrated significantly higher expression of PD-L2 compared with PD-L1 both pretherapy and at all time points during therapy, with increasing PD-L2 expression on therapy., Conclusions: Although the combination of azacitidine and avelumab was well tolerated, clinical activity was limited. High expression of PD-L2 on bone marrow blasts may be an important mechanism of resistance to anti-PD-L1 therapy in AML., Lay Summary: This report describes the results of a phase 1b/2 study of azacitidine with the anti-PD-L1 immune checkpoint inhibitor avelumab for patients with relapsed/refractory acute myeloid leukemia (AML). The clinical activity of the combination therapy was modest, with an overall response rate of 10.5%. However, mass cytometry analysis revealed significantly higher expression of PD-L2 compared with PD-L1 on AML blasts from all patients who were analyzed at all time points. These data suggest a novel potential role for PD-L2 as a means of AML immune escape., (© 2021 American Cancer Society.)- Published
- 2021
- Full Text
- View/download PDF
36. Venetoclax Combined With FLAG-IDA Induction and Consolidation in Newly Diagnosed and Relapsed or Refractory Acute Myeloid Leukemia.
- Author
-
DiNardo CD, Lachowiez CA, Takahashi K, Loghavi S, Xiao L, Kadia T, Daver N, Adeoti M, Short NJ, Sasaki K, Wang S, Borthakur G, Issa G, Maiti A, Alvarado Y, Pemmaraju N, Montalban Bravo G, Masarova L, Yilmaz M, Jain N, Andreeff M, Jabbour E, Garcia-Manero G, Kornblau S, Ravandi F, Konopleva MY, and Kantarjian HM
- Subjects
- Adult, Aged, Bridged Bicyclo Compounds, Heterocyclic administration & dosage, Cytarabine administration & dosage, Female, Follow-Up Studies, Granulocyte Colony-Stimulating Factor administration & dosage, Hematopoietic Stem Cell Transplantation methods, Humans, Idarubicin administration & dosage, Leukemia, Myeloid, Acute pathology, Leukemia, Myeloid, Acute therapy, Male, Maximum Tolerated Dose, Middle Aged, Neoplasm Recurrence, Local pathology, Neoplasm Recurrence, Local therapy, Prognosis, Remission Induction, Sulfonamides administration & dosage, Survival Rate, Vidarabine administration & dosage, Vidarabine analogs & derivatives, Young Adult, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Drug Resistance, Neoplasm, Leukemia, Myeloid, Acute drug therapy, Neoplasm Recurrence, Local drug therapy
- 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., Competing Interests: Courtney D. DiNardoLeadership: Notable LabsStock and Other Ownership Interests: Notable LabsHonoraria: Agios, Celgene, AbbVie, Jazz Pharmaceuticals, Daiichi Sankyo, Novartis, TakedaConsulting or Advisory Role: Celgene, Agios, AbbVieResearch Funding: AbbVie, Agios, Celgene, Daiichi Sankyo Koichi TakahashiHonoraria: Celgene, Novartis, GlaxoSmithKline, SymBio Pharmaceuticals, DAVA PharmaceuticalsConsulting or Advisory Role: SymBio Pharmaceuticals, Novartis, GlaxoSmithKline, CelgeneResearch Funding: Onconova TherapeuticsTravel, Accommodations, Expenses: SymBio Pharmaceuticals, Celgene, GlaxoSmithKline, DAVA Pharmaceuticals Sanam LoghaviStock and Other Ownership Interests: AbbVie, PureTechHonoraria: Curio ScienceConsulting or Advisory Role: Seattle Genetics, Gerson Lehrman Group, Guidepoint GlobalTravel, Accommodations, Expenses: Curio Science Tapan KadiaConsulting or Advisory Role: Novartis, Jazz Pharmaceuticals, Pfizer, AbbVie/Genentech, AgiosResearch Funding: Bristol Myers Squibb, Celgene, Sanofi, Amgen, BiolineRx, Incyte, Genentech/AbbVie, Pfizer, Jazz Pharmaceuticals, AstraZeneca, Astellas Pharma, Ascentage Pharma, Genfleet, Cyclacel Naval DaverHonoraria: BMS, Jazz Pharmaceuticals, Novartis, Incyte, Otsuka, Immunogen, Pfizer, Astellas Pharma, AbbVieConsulting or Advisory Role: Celgene, Agios, Jazz Pharmaceuticals, Pfizer, AbbVie, Astellas Pharma, Daiichi Sankyo, Novartis, Bristol Myers Squibb, Otsuka, Incyte, Karyopharm Therapeutics, Sunesis Pharmaceuticals, Amgen, Immunogen, Genentech, Servier, Syndax, Trillium TherapeuticsResearch Funding: Bristol Myers Squibb, Pfizer, Immunogen, Genentech, Nohla Therapeutics, AbbVie, Astellas Pharma, Servier, Daiichi Sankyo, Novartis, Karyopharm Therapeutics, Incyte, Sunesis Pharmaceuticals, Sobi Nicholas J. ShortHonoraria: AmgenConsulting or Advisory Role: AstraZenecaResearch Funding: Takeda, Astellas Pharma Koji SasakiHonoraria: OtsukaConsulting or Advisory Role: Novartis, Pfizer, TakedaResearch Funding: NovartisTravel, Accommodations, Expenses: Otsuka Sa WangConsulting or Advisory Role: Boston Biomedical, GlaxoSmithKline, Blueprint Medicines, CapricoResearch Funding: GlaxoSmithKline, Autolus Ltd, Genmab Gautam BorthakurConsulting or Advisory Role: Argenx, PTC Therapeutics, BiolineRx, BioTheryX, Nkarta, Treadwell Therapeutics, Novartis, Catamaran Bio, TakedaResearch Funding: Incyte, GlaxoSmithKline, Cyclacel, BiolineRx, MedImmune, Lilly, Oncoceutics, Ryvu Therapeutics, Janssen Scientific Affairs, Bristol Myers Squibb, AbbVie, Novartis, AstraZeneca, Mundipharma Research, PTC Therapeutics, BioTheryX, XBiotech, Arvinas, Astex Pharmaceuticals, TCR2 Therapeutics, Nkarta, Treadwell Therapeutics, Cellestia Biotech Ghayas IssaConsulting or Advisory Role: Novartis, Kura OncologyResearch Funding: Novartis, Syndax, Kura Oncology Abhishek MaitiResearch Funding: Celgene Yesid AlvaradoResearch Funding: Jazz Pharmaceuticals, FibroGen, Sun Pharma, BerGenBio, Daiichi Sankyo/Lilly, Astex Pharmaceuticals Naveen PemmarajuEmployment: MD Anderson Cancer CenterLeadership: ASH, ASCOHonoraria: Incyte, Novartis, LFB Biotechnologies, Stemline Therapeutics, Celgene, AbbVie, MustangBio, Roche Molecular Diagnostics, Blueprint Medicines, DAVA Pharmaceuticals, SpringerConsulting or Advisory Role: Blueprint Medicines, Pacylex, Immunogen, Bristol Myers SquibbResearch Funding: Novartis, Stemline Therapeutics, Samus Therapeutics, AbbVie, Cellectis, Affymetrix/Thermo Fisher Scientific, Daiichi Sankyo, PlexxikonTravel, Accommodations, Expenses: Stemline Therapeutics, Celgene, AbbVie, DAVA Oncology, MustangBioUncompensated Relationships: Dan's House of Hope, Oncology Times Guillermo BravoResearch Funding: IFM Therapeutics Musa YilmazResearch Funding: Daiichi Sankyo, Pfizer Nitin JainHonoraria: Pharmacyclics, ADC Therapeutics, Adaptive Biotechnologies, AbbVie/Genentech, Janssen, AstraZeneca/MedImmune, Servier, Precision Biosciences, BeiGene, TG Therapeutics, Cellectis, Bristol Myers Squibb/CelgeneConsulting or Advisory Role: Pharmacyclics, ADC Therapeutics, Adaptive Biotechnologies, AbbVie/Genentech, Janssen, AstraZeneca/MedImmune, Servier, Precision Biosciences, BeiGene, TG Therapeutics, Cellectis, Bristol Myers Squibb/CelgeneResearch Funding: Pfizer, Pharmacyclics, AbbVie, Genentech/Roche, Incyte, Infinity Pharmaceuticals, Bristol Myers Squibb, Seattle Genetics, Celgene, ADC Therapeutics, Servier, AstraZeneca/MedImmune, Cellectis, Adaptive Biotechnologies, Precision Biosciences, Aprea Therapeutics, Fate Therapeutics, Kite, a Gilead company Michael AndreefStock and Other Ownership Interests: Reata Pharmaceuticals, Aptose Biosciences, Eutropics, Senti Biosciences, Oncoceutics, OncolyzeConsulting or Advisory Role: Daiichi Sankyo, Jazz Pharmaceuticals, Celgene, Amgen, AstraZenecaResearch Funding: Daiichi Sankyo, Breast Cancer Research Foundation, United Therapeutics, Ono Pharmaceutical, Karyopharm Therapeutics, CPRIT, NIH/NCI, Amgen, AstraZenecaPatents, Royalties, Other Intellectual Property: Patent with ReataTravel, Accommodations, Expenses: Aptose Biosciences, Oncoceutics, Amgen, AstraZeneca Elias JabbourConsulting or Advisory Role: Pfizer, Takeda, Amgen, AbbVie, Bristol Myers Squibb, Incyte, Adaptive Biotechnologies, Astellas Pharma, GenentechResearch Funding: Pfizer, AbbVie, Amgen, Takeda, Adaptive Biotechnologies, Ascentage Pharma Group Guillermo Garcia-ManeroHonoraria: Celgene, Astex Pharmaceuticals, Acceleron Pharma, Helssin, AbbVieConsulting or Advisory Role: Celgene, Astex Pharmaceuticals, Acceleron Pharma, Jazz Pharmaceuticals, Bristol Myers Squibb, Helsinn TherapeuticsResearch Funding: Celgene, Astex Pharmaceuticals, Amphivena, Helsinn Therapeutics, Novartis, AbbVie, Bristol Myers Squibb, Onconova Therapeutics, H3 Biomedicine, Merck Farhad RavandiHonoraria: Amgen, Pfizer, Astellas Pharma, Orsenix, Celgene, Agios, AbbVie/Genentech, AstraZeneca, Bristol Myers Squibb, Takeda, Jazz Pharmaceuticals, NovartisConsulting or Advisory Role: Amgen, Astellas Pharma, Orsenix, Celgene, Jazz Pharmaceuticals, Agios, AbbVie/Genentech, Bristol Myers Squibb, AstraZeneca, Taiho Oncology, Syros Pharmaceuticals, Certara IncResearch Funding: Bristol Myers Squibb, Amgen, Macrogenics, Xencor, Selvita, Cellerant Marina Y. KonoplevaHonoraria: AbbVie, Genentech, Roche, Amgen, Stemline Therapeutics, KisoJi BiotechnologyConsulting or Advisory Role: AbbVie, Genentech/Roche, Stemline Therapeutics, Amgen, Forty Seven, KisoJi Biotechnology, Roche, JanssenResearch Funding: AbbVie, Genentech, Roche, Lilly, Cellectis, Calithera Biosciences, Ablynx, Agios, Ascentage Pharma, AstraZeneca, SanofiPatents, Royalties, Other Intellectual Property: Novartis—Pending, Eli Lilly—Research Funding; Issued/Licensed, Reata Pharmaceutical—Research Funding (Issued, Licensed, Royalities) Hagop M. KantarjianHonoraria: AbbVie, Amgen, ARIAD, Bristol Myers Squibb, Immunogen, Orsenix, Pfizer, Agios, Takeda, Actinium PharmaceuticalsResearch Funding: Pfizer, Amgen, Bristol Myers Squibb, Novartis, ARIAD, Astex Pharmaceuticals, AbbVie, Agios, Cyclacel, Immunogen, Jazz PharmaceuticalsNo other potential conflicts of interest were reported.
- Published
- 2021
- Full Text
- View/download PDF
37. Final results of a phase 2 clinical trial of LCL161, an oral SMAC mimetic for patients with myelofibrosis.
- Author
-
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, and Verstovsek S
- Subjects
- Apoptosis, Humans, Thiazoles, Myeloproliferative Disorders, Primary Myelofibrosis drug therapy
- 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., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
38. Venetoclax enhances T cell-mediated antileukemic activity by increasing ROS production.
- Author
-
Lee JB, Khan DH, Hurren R, Xu M, Na Y, Kang H, Mirali S, Wang X, Gronda M, Jitkova Y, MacLean N, Arruda A, Alaniz Z, Konopleva MY, Andreeff M, Minden MD, Zhang L, and Schimmer AD
- Subjects
- Adult, Antineoplastic Agents therapeutic use, Bridged Bicyclo Compounds, Heterocyclic therapeutic use, Cells, Cultured, Humans, Immunity, Cellular drug effects, Leukemia, Myeloid, Acute immunology, Reactive Oxygen Species immunology, Sulfonamides therapeutic use, T-Lymphocytes immunology, Tumor Cells, Cultured, Antineoplastic Agents pharmacology, Bridged Bicyclo Compounds, Heterocyclic pharmacology, Leukemia, Myeloid, Acute drug therapy, Sulfonamides pharmacology, T-Lymphocytes drug effects
- Abstract
Venetoclax, a Bcl-2 inhibitor, in combination with the hypomethylating agent azacytidine, achieves complete remission with or without count recovery in ∼70% of treatment-naive elderly patients unfit for conventional intensive chemotherapy. However, the mechanism of action of this drug combination is not fully understood. We discovered that venetoclax directly activated T cells to increase their cytotoxicity against acute myeloid leukemia (AML) in vitro and in vivo. Venetoclax enhanced T-cell effector function by increasing reactive oxygen species generation through inhibition of respiratory chain supercomplexes formation. In addition, azacytidine induced a viral mimicry response in AML cells by activating the STING/cGAS pathway, thereby rendering the AML cells more susceptible to T cell-mediated cytotoxicity. Similar findings were seen in patients treated with venetoclax, as this treatment increased reactive oxygen species generation and activated T cells. Collectively, this study presents a new immune-mediated mechanism of action for venetoclax and azacytidine in the treatment of AML and highlights a potential combination of venetoclax and adoptive cell therapy for patients with AML., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
39. Duration of cytopenias with concomitant venetoclax and azole antifungals in acute myeloid leukemia.
- Author
-
Rausch CR, DiNardo CD, Maiti A, Jammal NJ, Kadia TM, Marx KR, Borthakur G, Savoy JM, Pemmaraju N, DiPippo AJ, Daver NG, Chew SM, Sasaki K, Issa GC, Short NJ, Takahashi K, Ohanian MN, Ning J, Xiao L, Alvarado Y, Kontoyiannis DP, Ravandi F, Kantarjian HM, and Konopleva MY
- Subjects
- Aged, Antifungal Agents adverse effects, Antineoplastic Combined Chemotherapy Protocols adverse effects, Azoles therapeutic use, Bridged Bicyclo Compounds, Heterocyclic, Humans, Sulfonamides, Leukemia, Myeloid, Acute, Thrombocytopenia chemically induced, Thrombocytopenia drug therapy
- Abstract
Background: Venetoclax (VEN) combined with the hypomethylating agent (HMA) azacitidine improves survival in patients aged ≥75 years with newly diagnosed acute myeloid leukemia (AML). VEN and HMA treatment can result in prolonged and often profound neutropenia, and this warrants antifungal prophylaxis. Azole antifungals inhibit cytochrome P450 3A4, the primary enzyme responsible for VEN metabolism; this results in VEN dose reductions for each concomitant antifungal. Limited clinical data exist on outcomes for patients treated with VEN, an HMA, and various azoles., Methods: The time to neutrophil recovery (absolute neutrophil count [ANC] > 1000 cells/mm
3 ) and platelet (PLT) recovery (PLT count > 100,000 cells/mm3 ) in 64 patients with newly diagnosed AML who achieved a response after course 1 of VEN plus an HMA were evaluated. HMA therapy included azacitidine (75 mg/m2 intravenously/subcutaneously for 7 days) or decitabine (20 mg/m2 intravenously for 5 or 10 days)., Results: Forty-seven patients (73%) received an azole: posaconazole (n = 17; 27%), voriconazole (n = 9; 14%), isavuconazole (n = 20; 31%), or fluconazole (n = 1; 2%). The median time to ANC recovery were similar for patients who did receive an azole (37 days; 95% confidence interval [CI], 34-38 days) and patients who did not receive an azole (39 days; 95% CI, 30 days to not estimable; P = .8). The median time to PLT recovery was significantly longer for patients receiving azoles (28 vs 22 days; P = .01). The median times to ANC recovery (35 vs 38 days) and PLT recovery (26 vs 32 days) were similar with posaconazole and voriconazole., Conclusions: VEN plus an HMA resulted in neutropenia and thrombocytopenia, with the latter prolonged in patients receiving concomitant azoles. Concomitant posaconazole or voriconazole and VEN (100 mg) resulted in similar ANC and PLT recovery times, suggesting the safety of these dosage combinations during course 1., (© 2021 American Cancer Society.)- Published
- 2021
- Full Text
- View/download PDF
40. Decitabine and venetoclax for IDH1/2-mutated acute myeloid leukemia.
- Author
-
Venugopal S, Maiti A, DiNardo CD, Loghavi S, Daver NG, Kadia TM, Rausch CR, Alvarado Y, Ohanian M, Sasaki K, Short NJ, Takahashi K, Yilmaz M, Ravandi F, Kantarjian HM, and Konopleva MY
- Subjects
- Adult, Aged, Aged, 80 and over, Bridged Bicyclo Compounds, Heterocyclic administration & dosage, Decitabine administration & dosage, Female, Follow-Up Studies, Humans, Kaplan-Meier Estimate, Leukemia, Myeloid, Acute genetics, Male, Middle Aged, Mutation, Neoplasms, Second Primary drug therapy, Neoplasms, Second Primary genetics, Salvage Therapy, Sulfonamides administration & dosage, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Isocitrate Dehydrogenase genetics, Leukemia, Myeloid, Acute drug therapy, Neoplasm Proteins genetics
- Published
- 2021
- Full Text
- View/download PDF
41. Impact of splicing mutations in acute myeloid leukemia treated with hypomethylating agents combined with venetoclax.
- Author
-
Lachowiez CA, Loghavi S, Furudate K, Montalban-Bravo G, Maiti A, Kadia T, Daver N, Borthakur G, Pemmaraju N, Sasaki K, Alvarado Y, Yilmaz M, Short NJ, Chien K, Ohanian M, Pierce S, Patel KP, Jabbour E, Ravandi F, Kantarjian HM, Garcia-Manero G, Takahashi K, Konopleva MY, and DiNardo CD
- Subjects
- Bridged Bicyclo Compounds, Heterocyclic, Humans, Mutation, Retrospective Studies, Serine-Arginine Splicing Factors, Splicing Factor U2AF genetics, Sulfonamides, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Nuclear Proteins genetics
- Abstract
Spliceosome mutations (SRSF2, SF3B1, U2AF1, ZRSR2), are encountered in ∼50% of secondary acute myeloid leukemia cases (sAML) and define a molecular subgroup with outcomes similar to sAML in de novo AML patients treated with intensive chemotherapy. Outcomes in patients with spliceosome mutations treated with hypomethylating agents in combination with venetoclax (HMA+VEN) remains unknown. The primary objective was to compare outcomes in patients with spliceosome mutations vs wild-type patients treated with HMA+VEN. Secondary objectives included analysis of the mutational landscape of the spliceosome cohort and assessing the impact of co-occurring mutations. We performed a retrospective cohort analysis of patients treated with HMA+VEN-based regimens at The University of Texas MD Anderson Cancer Center. A total of 119 patients (spliceosome mutated n = 39 [SRSF2, n = 24; SF3B1, n = 8; U2AF1, n = 7]; wild-type, n = 80) were included. Similar responses were observed between spliceosome and wild-type cohorts for composite complete response (CRc; 79% vs 75%, P = .65), and measurable residual disease-negative CRc (48% vs 60%, P = .34). Median overall survival for spliceosome vs wild-type patients was 35 vs 14 months (P = .58), and was not reached; 35 months and 8 months for patients with SRSF2, SF3B1, and U2AF1 mutations, respectively. IDH2 mutations were enriched in patients with SRSF2 mutations and associated with favorable outcomes (1- and 2-year overall survival [OS] of 100% and 88%). RAS mutations were enriched in patients with U2AF1 mutations and associated with inferior outcomes (median OS, 8 months). Comparable outcomes were observed between patients with vs without spliceosome mutations treated with HMA+VEN regimens, with specific co-mutation pairs demonstrating favorable outcomes., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
42. Prognostic value of measurable residual disease after venetoclax and decitabine in acute myeloid leukemia.
- Author
-
Maiti A, DiNardo CD, Wang SA, Jorgensen J, Kadia TM, Daver NG, Short NJ, Yilmaz M, Pemmaraju N, Borthakur G, Bose P, Issa GC, Ferrajoli A, Jabbour EJ, Jain N, Garcia-Manero G, Ohanian M, Takahashi K, Montalban-Bravo G, Masarova L, Burger JA, Thompson PA, Verstovsek S, Sasaki K, Andreeff M, Rausch CR, Montalbano KS, Pierce S, Qiao W, Ning J, Kantarjian HM, Konopleva MY, and Ravandi F
- Subjects
- Aged, Decitabine therapeutic use, Humans, Prognosis, Sulfonamides, Bridged Bicyclo Compounds, Heterocyclic therapeutic use, Leukemia, Myeloid, Acute diagnosis, Leukemia, Myeloid, Acute drug therapy
- 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., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
43. Venetoclax with decitabine vs intensive chemotherapy in acute myeloid leukemia: A propensity score matched analysis stratified by risk of treatment-related mortality.
- Author
-
Maiti A, Qiao W, Sasaki K, Ravandi F, Kadia TM, Jabbour EJ, Daver NG, Borthakur G, Garcia-Manero G, Pierce SA, Montalbano KS, Pemmaraju N, Naqvi K, Ohanian M, Short NJ, Alvarado Y, Takahashi K, Yilmaz M, Jain N, Kornblau SM, Andreeff M, Bose P, Ferrajoli A, Issa GC, Masarova L, Thompson PA, Rausch CR, Ning J, Kantarjian HM, DiNardo CD, and Konopleva MY
- Subjects
- Aged, Aged, 80 and over, Antineoplastic Combined Chemotherapy Protocols adverse effects, Bridged Bicyclo Compounds, Heterocyclic administration & dosage, Bridged Bicyclo Compounds, Heterocyclic adverse effects, Clinical Trials, Phase II as Topic statistics & numerical data, Combined Modality Therapy, Consolidation Chemotherapy, Cytarabine administration & dosage, Decitabine administration & dosage, Decitabine adverse effects, Drug Administration Schedule, Drug Evaluation, Female, Hematopoietic Stem Cell Transplantation, Humans, Kaplan-Meier Estimate, Leukemia, Myeloid, Acute mortality, Leukemia, Myeloid, Acute surgery, Male, Middle Aged, Progression-Free Survival, Propensity Score, Recurrence, Remission Induction, Retrospective Studies, Risk, Sulfonamides administration & dosage, Sulfonamides adverse effects, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Leukemia, Myeloid, Acute drug therapy
- Abstract
Hypomethylating agents (HMA) with venetoclax is a new standard for older/unfit patients with acute myeloid leukemia (AML). However, it is unknown how HMA with venetoclax compare to intensive chemotherapy (IC) in patients who are "fit" or "unfit" for IC. We compared outcomes of older patients with newly diagnosed AML receiving 10-day decitabine with venetoclax (DEC10-VEN) vs IC. DEC10-VEN consisted of daily venetoclax with decitabine 20 mg/m
2 for 10 days for induction and decitabine for 5 days as consolidation. The IC cohort received regimens containing cytarabine ≥1 g/m2 /d. A validated treatment-related mortality score (TRMS) was used to classify patients at high-risk or low-risk for TRM with IC. Propensity scores were used to match patients to minimize bias. Median age of the DEC10-VEN cohort (n = 85) was 72 years (range 63-89) and 28% patients were at high-risk of TRM with IC. The comparator IC group (n = 85) matched closely in terms of baseline characteristics. DEC10-VEN was associated with significantly higher CR/CRi compared to IC (81% vs 52%, P < .001), and lower rate of relapse (34% vs 56%, P = .01), 30-day mortality (1% vs 24%, P < .01), and longer overall survival (OS; 12.4 vs 4.5 months, HR = 0.48, 95%CI 0.29-0.79, P < .01). In patients at both at high-risk and low-risk of TRM, DEC10-VEN showed significantly higher CR/CRi, lower 30-day mortality, and longer OS compared to IC. Patients at both high-risk and low-risk of TRM had comparable outcomes with DEC10-VEN. In conclusion, DEC10-VEN offers better outcomes compared to intensive chemotherapy in older patients with newly diagnosed AML, particularly in those at high-risk of TRM., (© 2020 Wiley Periodicals LLC.)- Published
- 2021
- Full Text
- View/download PDF
44. Mechanisms for resistance in AML insights into molecular pathways mediating resistance to venetoclax.
- Author
-
Konopleva MY
- Subjects
- Bridged Bicyclo Compounds, Heterocyclic therapeutic use, Humans, Sulfonamides therapeutic use, Antineoplastic Combined Chemotherapy Protocols, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics
- Abstract
Resistance to therapy continues to pose hurdles in the therapeutic management of acute myeloid leukemia (AML). Although the approval and development of therapies such as venetoclax, was expected to overcome this issue, resistance remains a common occurrence in AML treatment. This review has summarized evidence that will provide insights into acquired mutations that influence response to venetoclax therapy and the utility of novel combination approaches in improving outcomes., (Copyright © 2021. Published by Elsevier Ltd.)
- Published
- 2021
- Full Text
- View/download PDF
45. Outcomes of relapsed or refractory acute myeloid leukemia after frontline hypomethylating agent and venetoclax regimens.
- Author
-
Maiti A, Rausch CR, Cortes JE, Pemmaraju N, Daver NG, Ravandi F, Garcia-Manero G, Borthakur G, Naqvi K, Ohanian M, Short NJ, Alvarado Y, Kadia TM, Takahashi K, Yilmaz M, Jain N, Kornblau S, Montalban Bravo G, Sasaki K, Andreeff M, Bose P, Ferrajoli A, Issa GC, Jabbour EJ, Masarova L, Thompson PA, Wang S, Konoplev S, Pierce SA, Ning J, Qiao W, Welch JS, Kantarjian HM, DiNardo CD, and Konopleva MY
- Subjects
- Antineoplastic Combined Chemotherapy Protocols adverse effects, Humans, Sulfonamides adverse effects, Bridged Bicyclo Compounds, Heterocyclic therapeutic use, Leukemia, Myeloid, Acute drug therapy
- Published
- 2021
- Full Text
- View/download PDF
46. Correction to: Outcomes with sequential FLT3-inhibitor-based therapies in patients with AML.
- Author
-
Yilmaz M, Alfayez M, DiNardo CD, Borthakur G, Kadia TM, Konopleva MY, Loghavi S, Kanagal-Shamanna R, Patel KP, Jabbour EJ, Garcia-Manero G, Pemmaraju N, Pierce SA, Ghayas I, Short NJ, Montalban-Bravo G, Takahashi K, Assi R, Alotaibi AS, Ohanian M, Andreeff M, Cortes JE, Kantarjian HM, Ravandi F, and Daver NG
- Published
- 2021
- Full Text
- View/download PDF
47. Triplet therapy with venetoclax, FLT3 inhibitor and decitabine for FLT3-mutated acute myeloid leukemia.
- Author
-
Maiti A, DiNardo CD, Daver NG, Rausch CR, Ravandi F, Kadia TM, Pemmaraju N, Borthakur G, Bose P, Issa GC, Short NJ, Yilmaz M, Montalban-Bravo G, Ferrajoli A, Jabbour EJ, Jain N, Ohanian M, Takahashi K, Thompson PA, Loghavi S, Montalbano KS, Pierce S, Wierda WG, Kantarjian HM, and Konopleva MY
- Subjects
- Adult, Aged, Female, Humans, Leukemia, Myeloid, Acute genetics, Male, Middle Aged, Mutation drug effects, Treatment Outcome, fms-Like Tyrosine Kinase 3 genetics, Antineoplastic Agents therapeutic use, Bridged Bicyclo Compounds, Heterocyclic therapeutic use, Decitabine therapeutic use, Leukemia, Myeloid, Acute drug therapy, Protein Kinase Inhibitors therapeutic use, Sulfonamides therapeutic use, fms-Like Tyrosine Kinase 3 antagonists & inhibitors
- Published
- 2021
- Full Text
- View/download PDF
48. A venetoclax bench-to-bedside story.
- Author
-
DiNardo CD and Konopleva MY
- Subjects
- Bridged Bicyclo Compounds, Heterocyclic therapeutic use, Sulfonamides therapeutic use
- Published
- 2021
- Full Text
- View/download PDF
49. Acute promyelocytic leukemia (APL) with an IRF2BP2-RARA fusion transcript: an aggressive APL variant.
- Author
-
Alotaibi AS, Abdulrazzaq M, Patel KP, Ravandi F, Konoplev S, Bueso-Ramos C, Yin CC, Muzzafar T, Tang G, Futreal A, Jain N, Konopleva MY, and Pemmaraju N
- Subjects
- DNA-Binding Proteins, Humans, Oncogene Proteins, Fusion genetics, Promyelocytic Leukemia Protein, Retinoic Acid Receptor alpha genetics, Transcription Factors, Leukemia, Promyelocytic, Acute genetics
- Published
- 2020
- Full Text
- View/download PDF
50. Outcomes with sequential FLT3-inhibitor-based therapies in patients with AML.
- Author
-
Yilmaz M, Alfayez M, DiNardo CD, Borthakur G, Kadia TM, Konopleva MY, Loghavi S, Kanagal-Shamanna R, Patel KP, Jabbour EJ, Garcia-Manero G, Pemmaraju N, Pierce SA, Ghayas I, Short NJ, Montalban-Bravo G, Takahashi K, Assi R, Alotaibi AS, Ohanian M, Andreeff M, Cortes JE, Kantarjian HM, Ravandi F, and Daver NG
- Subjects
- Adult, Aged, Aged, 80 and over, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Female, Humans, Leukemia, Myeloid, Acute genetics, Male, Middle Aged, Mutation drug effects, Retrospective Studies, Treatment Outcome, Young Adult, fms-Like Tyrosine Kinase 3 genetics, Leukemia, Myeloid, Acute drug therapy, Protein Kinase Inhibitors therapeutic use, fms-Like Tyrosine Kinase 3 antagonists & inhibitors
- Abstract
Background: Second-generation FLT3-inhibitors (FLT3i) demonstrated single-agent composite CR rates (CRc) of 45-55% in patients with relapsed/refractory (R/R) FLT3-mutated AML in phase II/III trials. However, > 85% of patients treated were prior FLT3i naïve. The response rates to sequential FLT3i exposure remain poorly defined., Methods: We retrospectively reviewed patients with FLT3-mutated AML between November 2006 and December 2019., Results: In frontline patients treated with a FLT3i (cohort 1), the CRc rates and median overall survival (OS) with the first (n = 56), second (n = 32), and third FLT3i-based (n = 8) therapy were 77%, 31%, and 25%, and 16.7 months, 6.0 months, and 1.4 months, respectively. In patients receiving a FLT3i-based therapy for the first time in a R/R AML setting (cohort 2), the CRc rates and median OS were 45%, 21%, and 10%, and 7.9 months, 4.0 months, and 4.1 months with the first (n = 183), second (n = 89), and third/fourth (n = 29) FLT3i-based therapy, respectively. In cohort 1, CRc rates with single-agent FLT3i (n = 21) versus FLT3i-based combinations (n = 19) in second/third sequential FLT3i exposures were 19% versus 42%, respectively. In cohort 2, the CRc rates with single-agent FLT3i (n = 82) versus FLT3i-based combinations (n = 101) in first FLT3i exposure were 34% versus 53%, respectively, and those with single-agent FLT3i (n = 63) versus FLT3i-based combinations (n = 55) in second/third/fourth sequential FLT3i exposures were 13% versus 25%, respectively., Conclusion: CRc rates drop progressively with sequential exposure to FLT3i's in FLT3-mutated AML. In all settings, CRc rates were higher with FLT3i-based combinations compared with single-agent FLT3i therapy in similar FLT3i exposure settings.
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.