216 results on '"Alice S. Mims"'
Search Results
52. Regularized Mixture Cure Models Identify a Gene Signature That Improves Risk Stratification within the Favorable-Risk Group in 2017 European Leukemianet (ELN) Classification of Acute Myeloid Leukemia (Alliance 152010)
- Author
-
Kellie J. Archer, Han Fu, Krzysztof Mrózek, Deedra Nicolet, Jessica Kohlschmidt, Alice S. Mims, Geoffrey L. Uy, Wendy Stock, John C. Byrd, and Ann-Kathrin Eisfeld
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
53. Preliminary Results from a Phase 2 Open-Label, Multicenter, Dose Optimization Clinical Study of the Safety, Tolerability, and Pharmacokinetic (PK) and Pharmacodynamic (PD) Profiles of Cfi-400945 As a Single Agent or in Combination with Azacitidine or Decitabine in Patients with Acute Myeloid Leukemia, Myelodysplastic Syndrome or Chronic Myelomonocytic Leukemia (TWT-202)
- Author
-
Brian A. Jonas, Karen Yee, Paul B. Koller, Joseph Brandwein, Alice S. Mims, Glenn C. Michelson, Linh Nguyen, Mark R Bray, Emily L Roberts-Thomson, and Gautam Borthakur
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
54. Entospletinib in Combination with Induction Chemotherapy in Previously Untreated Acute Myeloid Leukemia: Response and Predictive Significance of HOXA9 and MEIS1 Expression
- Author
-
Bhavana Bhatnagar, Arati V. Rao, James S. Blachly, Tara L. Lin, Alison Walker, Howland E. Crosswell, William Blum, Jinfeng Liu, Veerendra Munugalavadla, Lauren Long, Danjie Zhang, Mark D. Minden, Yang Pan, Hubert Serve, John C. Byrd, Alice S. Mims, Thomas Oellerich, and Shelley Orwick
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Daunorubicin ,medicine.medical_treatment ,Syk ,03 medical and health sciences ,0302 clinical medicine ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Chemotherapy ,business.industry ,Myeloid leukemia ,Induction chemotherapy ,medicine.disease ,Rash ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cytarabine ,medicine.symptom ,business ,Febrile neutropenia ,medicine.drug - Abstract
Purpose: Spleen tyrosine kinase (SYK) signaling is a proposed target in acute myeloid leukemia (AML). Sensitivity to SYK inhibition has been linked to HOXA9 and MEIS1 overexpression in preclinical studies. This trial evaluated the safety and efficacy of entospletinib, a selective inhibitor of SYK, in combination with chemotherapy in untreated AML. Patients and Methods: This was an international multicenter phase Ib/II study, entospletinib dose escalation (standard 3+3 design between 200 and 400 mg twice daily) + 7+3 (cytarabine + daunorubicin) in phase Ib and entospletinib dose expansion (400 mg twice daily) + 7+3 in phase II. Results: Fifty-three patients (n = 12, phase Ib and n = 41, phase II) with previously untreated de novo (n = 39) or secondary (n = 14) AML were enrolled (58% male; median age, 60 years) in this study. The composite complete response with entospletinib + 7+3 was 70%. Patients with baseline HOXA9 and MEIS1 expression higher than the median had improved overall survival compared with patients with below median HOXA9 and MEIS1 expression. Common adverse events were cytopenias, febrile neutropenia, and infection. There were no dose-limiting toxicities. Entospletinib-related skin rash and hyperbilirubinemia were also observed. Conclusions: Entospletinib with intensive chemotherapy was well-tolerated in patients with AML. Improved survival was observed in patients with HOXA9/MEIS1 overexpression, contrasting published data demonstrating poor survival in such patients. A randomized study will be necessary to determine whether entospletinib was a mediator this observation.
- Published
- 2020
55. Enasidenib (ENA) Monotherapy with Addition of Azacitidine in Non-Responders Is Effective in Older Patients with Newly Diagnosed IDH2 Mutated Acute Myeloid Leukemia (AML): A Completed Phase 2/1b Sub-Study of the Beat AML Master Trial
- Author
-
Jordan Chervin, Ross L. Levine, Tara L. Lin, Ying Huang, William Blum, Sonja Marcus, Tibor Kovacsovics, Ashley O. Yocum, Franchesca Druggan, Gary J. Schiller, Brian J. Druker, Mona Stefanos, Uma Borate, Matthew C. Foster, Mark R. Litzow, John C. Byrd, Nyla A. Heerema, Robert H. Collins, Abigail B. Shoben, Wendy Stock, Leonard Rosenberg, Amy Burd, Michael Boyiadzis, James M. Foran, Rebecca L. Olin, Jo-Anne Vergilio, Prapti A. Patel, Maria R. Baer, Timothy L. Chen, Eytan M. Stein, and Alice S. Mims
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Immunology ,Azacitidine ,Beat (acoustics) ,Myeloid leukemia ,Cell Biology ,Hematology ,Newly diagnosed ,Enasidenib ,Biochemistry ,IDH2 ,Non responders ,Older patients ,Internal medicine ,medicine ,business ,medicine.drug - Abstract
Background: ENA is an oral, selective inhibitor of IDH2 approved for the treatment (Tx) of patients (pts) with relapsed/refractory IDH2 mutated (IDH2m) AML. Here we report the results of a Phase 2 expansion and Phase 1b of the Beat AML Master Trial Phase 2/1b sub-study to assess the efficacy of Tx of newly diagnosed (ND) IDH2m AML pts ≥ 60 years of age with ENA monotherapy (ENAm) and subsequent response-driven addition of AZA Tx. (ClinicalTrials.gov NCT03013998). Methods: The study initiated with a 3-outcome, 2-stage Phase 2 design, which enrolled patients on ENAm for up to 5 cycles. Pts without CR/CRi after 5 cycles of ENAm, or progression/intolerance prior to this time, were transferred to Phase 1b to receive ENA + AZA (Figure 1). Key eligibility included ND IDH2m AML pts with age ≥ 60 years and ECOG performance status 0-2. Pts received ENAm 100 mg/day in continuous 28-day cycles and ENA + AZA (75 mg/m2 days 1-7 every 28 days) for Phase 1b. Response was assessed using 2017 ELN AML criteria. The primary endpoint was CR/CRi rate. The 2-stage design required 24 pts and tested the null hypothesis (H0) that CR/CRi rate equaled 20% vs 50% and then expanded to test a revised H0 of 30% vs 50% in 60 pts (conditional alpha=0.025, power=77%). Expansion also allowed further assessment of safety of this treatment regimen. Results: At data cut off (06/18/2020), 60 pts enrolled, received ENAm, and were evaluable for the primary endpoint. Median age was 75 years and 52% were female (Table 1). Median time on ENAm was 4.7 months (mos). At data cut off, 12 pts were still on ENAm Tx. Most common reasons for discontinuing ENAm were Tx failure (defined as no response to treatment) (23 or 38%), disease progression (loss of response to treatment) (7 or 12%) and adverse events (AEs; 6 or 10%). Five pts (8%) went to transplant. CR/CRi was achieved in 28 pts (47%; adjusted 95% CI 28-59, unadjusted exact 95% CI 34-60) (Table 2). Responses were higher (p=0.04) among the 44 pts with IDH2 R140 (55%) as compared to the 16 with IDH2 R172 mutation (25%) further supporting distinct biology between these subsets. After a median follow up of 14.6 mos, the median overall survival (mOS) was 24.4 mos (95% CI 10.6-not reached). The median duration of response was not reached with 12 mos estimation of 57% (95% CI 34-75). Overall, 20 ENA-related serious adverse events (SAEs) occurred in 15 pts, the most common was differentiation syndrome (12 or 20%) and 1 had ENA-related SAE of tumor lysis syndrome (1.7%). One pt had ENA-related Grade 5 AE (renal failure/death). Most common AEs of any grade (in ≥20%) were nausea, anemia, and low potassium (Table 3). The 7-day/30-day/60-day deaths observed with ENAm were 2%/5%/11%, respectively. Phase 1b: Seventeen pts had inadequate response to ENAm and transferred to Phase 1b to receive ENA + AZA. Median time on Tx (including ENAm) was 6.2 mos and median time on Tx after pts started ENA + AZA was 2.1 mos (Table 2). Most common reasons for discontinuing ENA + AZA included Tx failure (5 or 29%), disease progression (2 or 12%), transplant, death and AEs (each 2 or 12%). CR/CRi was 41% (exact 95% CI 18-67). After a median follow up of 12.7 mos, the mOS from start of ENA + AZA combination Tx was 8.9 mos. Four ENA-related SAEs occurred in 3 pts on ENA + AZA Tx and the most common was differentiation syndrome (2 or 12.5%). One dose-limiting toxicity (Grade 3 nausea) related to both Txs was observed. Most common AEs (≥20%) of any grade were anemia, low albumin and vomiting (Table 3). One death occurred at day 13 of ENA + AZA. Conclusions: In newly diagnosed pts ≥60 years old with IDH2m AML, ENA had a low early death rate, high CR/CRi rate (47%, adjusted 95% CI 28-59), and yielded durable remissions. The most common unique toxicity with ENA was differentiation syndrome that occurred in 20% of patients. In pts who did not achieve CR/CRi with ENAm, a subset of patients achieved CR/CRi with addition of AZA. This combined approach of serial therapy with ENA monotherapy followed by AZA addition in pts with sub-optimal response resulted in a mOS exceeding 2 years for pts enrolled on study. Further focus on improving response among patients with IDH2 R172 mutations, identifying subsets of pts not responding to ENA monotherapy, and integrating new targeted agents into this treatment regimen are warranted. Figure 1 Disclosures Stein: Syndax: Consultancy, Research Funding; Celgene Pharmaceuticals: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Daiichi-Sankyo: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Astellas Pharmaceuticals: Consultancy, Membership on an entity's Board of Directors or advisory committees; Seattle Genetics: Consultancy; Agios Pharmaceuticals: Consultancy, Membership on an entity's Board of Directors or advisory committees; Bayer: Research Funding; Genentech: Consultancy, Membership on an entity's Board of Directors or advisory committees; Amgen: Consultancy; Biotheryx: Consultancy; Novartis: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; PTC Therapeutics: Membership on an entity's Board of Directors or advisory committees; Syros: Membership on an entity's Board of Directors or advisory committees; Abbvie: Consultancy. Borate:Genentech: Membership on an entity's Board of Directors or advisory committees; Daiichi Sankyo: Membership on an entity's Board of Directors or advisory committees; Takeda: Membership on an entity's Board of Directors or advisory committees, Research Funding; Novartis: Membership on an entity's Board of Directors or advisory committees, Research Funding; Jazz Pharmaceuticals: Research Funding; AbbVie: Other: Investigator in AbbVie-funded clinical trials; Pfizer: Membership on an entity's Board of Directors or advisory committees, Research Funding. Baer:Takeda: Other: Institutional research funding; AbbVie: Other: Institutional research funding; Astellas: Other: Institutional research funding; Forma: Other: Institutional research funding; Kite: Other: Institutional research funding; Oscotec: Other: Institutional research funding; Incyte: Other: Institutional research funding. Kovacsovics:Agios: Honoraria; Astella: Honoraria; Pfizer: Research Funding; Novartis: Research Funding; AbbVie: Research Funding; Jazz: Honoraria. Schiller:Astellas Pharma: Honoraria, Research Funding; Celator: Research Funding; Constellation: Research Funding; Abbvie: Research Funding; Actinium: Research Funding; Ariad: Research Funding; Stemline: Speakers Bureau; Cyclacel: Research Funding; Daiichi Sankyo: Research Funding; Deciphera: Research Funding; DeltaFly: Research Funding; Bristol-Myers Squibb: Current equity holder in publicly-traded company, Research Funding; Forma: Research Funding; FujiFilm: Research Funding; Gamida: Research Funding; Genentech-Roche: Research Funding; Geron: Research Funding; Jazz Pharmaceuticals: Research Funding; Karyopharm: Research Funding; Kite Pharma: Research Funding; Mateon: Research Funding; MedImmune: Research Funding; Onconova: Research Funding; Pfizer: Current equity holder in publicly-traded company, Research Funding; Regimmune: Research Funding; Samus: Research Funding; Sangamo: Research Funding; Tolero: Research Funding; Trovagene: Research Funding; Kaiser Permanente: Consultancy; Johnson & Johnson: Current equity holder in publicly-traded company; Agios: Consultancy, Research Funding, Speakers Bureau; Amgen: Consultancy, Current equity holder in publicly-traded company, Research Funding, Speakers Bureau; AstraZeneca: Consultancy; Incyte: Consultancy, Research Funding, Speakers Bureau; Novartis: Consultancy, Research Funding; Ono Pharma: Consultancy; Celgene: Research Funding, Speakers Bureau; Sanofi: Speakers Bureau; Gilead: Speakers Bureau. Olin:Astellas: Other: Site PI; Genentech: Other: Site PI; Pfizer: Other: Site PI; Daiichi Sankyo: Other: Site PI; Genentech: Consultancy; Amgen: Consultancy. Foran:Trillium: Research Funding; Xencor: Research Funding; H3Biosciences: Research Funding; Agios: Honoraria, Research Funding; Pfizer: Membership on an entity's Board of Directors or advisory committees; BMS: Membership on an entity's Board of Directors or advisory committees; Abbvie: Research Funding; Boehringer Ingelheim: Research Funding; Actinium: Research Funding; Aprea: Research Funding; Aptose: Research Funding; Kura Oncology: Research Funding; Takeda: Research Funding; Revolution Medicine: Consultancy; Novartis: Membership on an entity's Board of Directors or advisory committees; Servier: Membership on an entity's Board of Directors or advisory committees. Lin:Trovagene: Research Funding; Tolero Pharmaceuticals: Research Funding; Seattle Genetics: Research Funding; Prescient Therapeutics: Research Funding; Abbvie: Research Funding; Bio-Path Holdings: Research Funding; Astellas Pharma: Research Funding; Aptevo: Research Funding; Incyte: Research Funding; Pfizer: Research Funding; Mateon Therapeutics: Research Funding; Ono Pharmaceutical: Research Funding; Jazz: Research Funding; Gilead Sciences: Research Funding; Genetech-Roche: Research Funding; Celyad: Research Funding; Celgene: Research Funding. Patel:DAVA Pharmaceuticals: Honoraria; Celgene: Consultancy, Speakers Bureau; Agios: Consultancy; France Foundation: Honoraria. Foster:Daiichi Sankyo: Consultancy; Bellicum Pharmaceuticals: Research Funding; Macrogenics: Consultancy, Research Funding. Druker:Leukemia & Lymphoma Society: Research Funding; Henry Stewart Talks: Patents & Royalties; Iterion Therapeutics (formerly Beta Cat Pharmaceuticals): Membership on an entity's Board of Directors or advisory committees; Vivid Biosciences: Membership on an entity's Board of Directors or advisory committees; GRAIL: Consultancy, Current equity holder in private company, Membership on an entity's Board of Directors or advisory committees; Patient True Talks: Consultancy; The RUNX1 Research Program: Membership on an entity's Board of Directors or advisory committees; Third Coast Therapeutics: Membership on an entity's Board of Directors or advisory committees; VB Therapeutics: Membership on an entity's Board of Directors or advisory committees; Gilead Sciences: Consultancy, Membership on an entity's Board of Directors or advisory committees; Millipore (formerly Upstate Biotechnology): Patents & Royalties; MolecularMD (acquired by ICON): Consultancy, Current equity holder in publicly-traded company, Membership on an entity's Board of Directors or advisory committees; Novartis Pharmaceuticals: Membership on an entity's Board of Directors or advisory committees, Patents & Royalties, Research Funding; McGraw Hill: Patents & Royalties; Merck & Co: Patents & Royalties; Cepheid: Consultancy, Membership on an entity's Board of Directors or advisory committees; Dana-Farber Cancer Institute: Patents & Royalties; EnLiven: Consultancy, Research Funding; Aptose Therapeutics Inc. (formerly Lorus): Consultancy, Current equity holder in publicly-traded company, Membership on an entity's Board of Directors or advisory committees; ARIAD: Research Funding; Blueprint Medicines: Consultancy, Current equity holder in private company, Membership on an entity's Board of Directors or advisory committees; ALLCRON: Consultancy, Membership on an entity's Board of Directors or advisory committees; Amgen: Current equity holder in publicly-traded company, Membership on an entity's Board of Directors or advisory committees; Bristol-Myers Squibb: Research Funding; Aileron Therapeutics: Membership on an entity's Board of Directors or advisory committees; Pfizer: Research Funding; Oregon Health & Science University: Patents & Royalties. Byrd:Acerta Pharma: Research Funding; Syndax: Research Funding; Vincera: Research Funding; Pharmacyclics LLC, an AbbVie Company, Janssen, Novartis, Gilead, TG Therapeutics: Other; Pharmacyclics LLC, an AbbVie Company, Gilead, TG Therapeutics, Novartis, Janssen: Speakers Bureau; Novartis: Research Funding; Kartos Therapeutics: Research Funding; Trillium: Research Funding; Leukemia and Lymphoma Society: Other; Janssen: Consultancy; Pharmacyclics LLC, an AbbVie Company, Gilead, TG Therapeutics, BeiGene: Research Funding. Levine:Loxo: Current equity holder in private company, Membership on an entity's Board of Directors or advisory committees; C4 Therapeutics: Current equity holder in private company, Membership on an entity's Board of Directors or advisory committees; Imago: Current equity holder in private company, Membership on an entity's Board of Directors or advisory committees; Celgene: Consultancy, Honoraria, Research Funding; Qiagen: Current equity holder in publicly-traded company, Membership on an entity's Board of Directors or advisory committees; Prelude Therapeutics: Research Funding; Amgen: Honoraria; Astellas: Consultancy; Morphosys: Consultancy; Novartis: Consultancy; Isoplexis: Current equity holder in private company, Membership on an entity's Board of Directors or advisory committees; Janssen: Consultancy; Lilly: Consultancy, Honoraria; Roche: Consultancy, Honoraria, Research Funding; Gilead: Honoraria. Mims:Abbvie: Membership on an entity's Board of Directors or advisory committees; Kura Oncology: Membership on an entity's Board of Directors or advisory committees; Leukemia and Lymphoma Society: Other: Senior Medical Director for Beat AML Study; Jazz Pharmaceuticals: Other: Data Safety Monitoring Board; Syndax Pharmaceuticals: Membership on an entity's Board of Directors or advisory committees; Agios: Consultancy; Novartis: Speakers Bureau. OffLabel Disclosure: Enasidenib is not approved for the treatment of newly diagnosed AML.
- Published
- 2020
56. Treating acute myeloid leukemia in the modern era: A primer
- Author
-
William Blum and Alice S. Mims
- Subjects
Cancer Research ,medicine.medical_specialty ,business.industry ,medicine.medical_treatment ,Myeloid leukemia ,Disease ,Middle Aged ,Targeted therapy ,Transplantation ,Clinical trial ,Food and drug administration ,Leukemia, Myeloid, Acute ,03 medical and health sciences ,0302 clinical medicine ,Oncology ,hemic and lymphatic diseases ,030220 oncology & carcinogenesis ,Risk stratification ,medicine ,Drug approval ,Humans ,030212 general & internal medicine ,business ,Intensive care medicine - Abstract
Recent years have seen tremendous advances in treating acute myeloid leukemia (AML), largely because of progress in understanding the genetic basis of the disease. The US Food and Drug Administration approved 7 agents for AML in the last 2 years: the first new drugs in decades. In this review, the authors discuss these new approvals in the backdrop of an overall strategy for treating AML today. Treating AML in the modern era requires: 1) access to and use of upfront genetic and cytogenetic testing, not only to describe prognosis but also to help identify the best available therapy; 2) effectively working new therapies into a conventional backbone of treatment, including transplantation; and 3) continued commitment to clinical trials designed to capitalize on advances in genetics and immunology to foster the next wave of drug approvals.
- Published
- 2020
57. Safety and efficacy of BAY1436032 in IDH1-mutant AML: phase I study results
- Author
-
Courtney D. DiNardo, Lewis R. Silverman, Charles Cai, Claudia D. Baldus, Isabelle Genvresse, Eleni Lagkadinou, Neil Palmisiano, Walter Fiedler, Alice S. Mims, Michael Jeffers, Markus Wagner, Ioannis Mantzaris, Alexander E. Perl, Gary Wilkinson, Bingyan Wu, Christine Rentzsch, Eunice S. Wang, Timothy S. Pardee, Sebastian Schwind, Stefan Kaulfuss, Michael Heuser, and Alwin Krämer
- Subjects
Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Myeloid ,IDH1 ,Mutant ,DNA Mutational Analysis ,Drug development ,Antineoplastic Agents ,Gastroenterology ,Article ,Acute myeloid leukaemia ,Pharmacokinetics ,Phase I trials ,Bone Marrow ,Internal medicine ,medicine ,Humans ,Dosing ,Molecular Targeted Therapy ,Enzyme Inhibitors ,Survival analysis ,Aged ,Aniline Compounds ,business.industry ,Hematology ,Middle Aged ,medicine.disease ,Prognosis ,Survival Analysis ,Isocitrate Dehydrogenase ,Clinical trial ,Leukemia ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,Treatment Outcome ,Oncology ,Mutation ,Benzimidazoles ,Female ,business - Abstract
The mutant IDH1 (mIDH1) inhibitor BAY1436032 demonstrated robust activity in preclinical AML models, supporting clinical evaluation. In the current dose-escalation study, BAY1436032 was orally administered to 27 mIDH1 AML subjects across 4 doses ranging from 300 to 1500 mg twice-daily. BAY1436032 exhibited a relatively short half-life and apparent non-linear pharmacokinetics after continuous dosing. Most subjects experienced only partial target inhibition as indicated by plasma R-2HG levels. BAY1436032 was safe and a maximum tolerated dose was not identified. The median treatment duration for all subjects was 3.0 months (0.49–8.5). The overall response rate was 15% (4/27; 1 CRp, 1 PR, 2 MLFS), with responding subjects experiencing a median treatment duration of 6.0 months (3.9–8.5) and robust R-2HG decreases. Thirty percent (8/27) achieved SD, with a median treatment duration of 5.5 months (3.1–7.0). Degree of R-2HG inhibition and clinical benefit did not correlate with dose. Although BAY1436032 was safe and modestly effective as monotherapy, the low overall response rate and incomplete target inhibition achieved at even the highest dose tested do not support further clinical development of this investigational agent in AML.
- Published
- 2020
58. Relapsed or primary refractory AML
- Author
-
Alice S. Mims and Kristin L. Koenig
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Myeloid ,Disease ,03 medical and health sciences ,European LeukemiaNet ,0302 clinical medicine ,Refractory ,Recurrence ,Internal medicine ,Humans ,Medicine ,MYELOID DISEASE: Edited by Martin S. Tallman ,hypomethylating agents ,business.industry ,Myeloid leukemia ,Hematology ,targeted therapy ,medicine.disease ,relapsed/refractory AML ,Leukemia, Myeloid, Acute ,Leukemia ,030104 developmental biology ,medicine.anatomical_structure ,FLAG (chemotherapy) ,Bone marrow ,business ,salvage regimens ,030215 immunology - Abstract
Acute myeloid leukemia (AML) is a biologically heterogeneous disease of the hematopoietic system characterized by clonal accumulation and expansion of immature myeloid cells in the bone marrow. Unfortunately, with current treatment strategies, only approximately 35–40% of patients at least 60 years and 5–15% of patients older than 60 years are cured of this disease [1]. Even with adaptation of cytogenetic and molecular risk-stratified therapies, 10–40% of patients do not achieve a complete remission (CR) after intensive induction therapy and are deemed to have primary refractory disease. Refractory disease is defined by the European LeukemiaNet (ELN) as the inability to attain CR or complete remission with incomplete hematologic recovery (CRi) after two courses of intensive induction treatment. Of note, this definition is not consistent throughout the literature [2]. Although some patients are able to achieve CR, greater than 50% of these patients subsequently experience disease relapse [3]. For patients who relapse, only a small fraction undergo successful salvage treatment with ability to attain a second CR [3]. Additionally, these patients are often not candidates for aggressive treatment (i.e. allogeneic stem cell transplant [alloHSCT]) given comorbid conditions and lack of suitable donors. Therefore, this leaves a large unmet clinical need for treatment of both relapsed and refractory (R/R) AML. Open in a separate window Box 1 no caption available
- Published
- 2020
59. Risk, Characteristics and Biomarkers of Cytokine Release Syndrome in Patients with Relapsed/Refractory AML or MDS Treated with CD3xCD123 Bispecific Antibody APVO436
- Author
-
Christopher R. Cogle, Alice S. Mims, Cynthia Lee, Paul J. Shami, Elizabeth Cull, Prapti A. Patel, Eunice S. Wang, Fatih M. Uckun, Tara L. Lin, and Justin M. Watts
- Subjects
Cancer Research ,medicine.medical_specialty ,Gastroenterology ,Article ,chemistry.chemical_compound ,Immunophenotyping ,Tocilizumab ,AML ,Internal medicine ,otorhinolaryngologic diseases ,Medicine ,Dexamethasone ,RC254-282 ,business.industry ,T-cells ,leukemia ,Myeloid leukemia ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,clinical study ,medicine.disease ,APVO436 ,Leukemia ,Cytokine release syndrome ,bispecific antibody ,medicine.anatomical_structure ,Oncology ,chemistry ,CD123 ,Interleukin-3 receptor ,Bone marrow ,business ,medicine.drug - Abstract
Simple Summary Cytokine release syndrome is a potentially life-threatening complication of therapy with T-cell engaging bispecific antibodies. Here we evaluated the risk, characteristics and biomarkers of treatment-emergent cytokine release syndrome in patients with relapsed/refractory acute myeloid leukemia or myelodysplastic syndrome who received weekly intravenous infusions of the CD3xCD123 bispecific antibody APVO436. Cytokine release syndrome was encountered in 10 of 46 patients (21.7%) treated with APVO436 with a cumulative Grade 3/4 cytokine release syndrome incidence of 8.7%. Cytokine profiling in patients who developed cytokine release syndrome after APVO436 infusion indicated that the predominant cytokine in this inflammatory cytokine response was IL-6. The findings from this research provide new insights regarding the biology and effective management of cytokine release syndrome in leukemia patients treated with T-cell redirecting bispecific antibodies. Abstract We evaluate the risk, characteristics and biomarkers of treatment-emergent cytokine release syndrome (CRS) in patients with relapsed/refractory acute myeloid leukemia (AML) or myelodysplastic syndrome (MDS) who received APVO436 during the dose-escalation phase of a Phase 1B study (ClinicalTrials.gov, identifier: NCT03647800). Of four patients who developed Grade ≥ 3 CRS, two received steroid prophylaxis. The dose level, gender, race, obesity, or baseline hematologic parameters in peripheral blood did not predict the risk of CRS. Patients with a higher leukemia burden as determined by a higher total WBC, higher percentage of blasts in bone marrow, or higher percentage of blasts in peripheral blood (by hematopathology or immunophenotyping) did not have a higher incidence of CRS. There was an age difference between patients who did versus patients who did not develop CRS (72.9 ± 1.6 years (Median 73.5 years) vs. 63.3 ± 2.3 years (Median: 65.0 years), which was borderline significant (p = 0.04). Premedication with steroids did not eliminate the risk of CRS. Cytokine profiling in patients who developed CRS after APVO436 infusion indicates that the predominant cytokine in this inflammatory cytokine response was IL-6. APVO436-associated CRS was generally manageable with tocilizumab with or without dexamethasone. Notably, the development of CRS after APVO436 therapy did not appear to be associated with a response. The prolonged stabilization of disease, partial remissions and complete remissions were achieved in both patients who experienced CRS, as well as patients who did not experience CRS after APVO436 infusions.
- Published
- 2021
- Full Text
- View/download PDF
60. Molecular, clinical, and prognostic implications of PTPN11 mutations in acute myeloid leukemia
- Author
-
James S. Blachly, Andrew J. Carroll, John C. Byrd, Christopher C. Oakes, Sydney Fobare, Shelley Orwick, Richard Stone, Krzysztof Mrózek, Alice S. Mims, Eunice S. Wang, Hatice Gulcin Ozer, Bayard L. Powell, Ann-Kathrin Eisfeld, Jonathan E. Kolitz, Ramiro Garzon, Jessica Kohlschmidt, Deedra Nicolet, and Erin Hertlein
- Subjects
musculoskeletal diseases ,Acute promyelocytic leukemia ,Oncology ,congenital, hereditary, and neonatal diseases and abnormalities ,medicine.medical_specialty ,NPM1 ,Protein Tyrosine Phosphatase, Non-Receptor Type 11 ,Gene mutation ,medicine.disease_cause ,Internal medicine ,Medicine ,Humans ,Mutation ,Clinical Trials as Topic ,business.industry ,Cancer ,Myeloid leukemia ,Hematology ,medicine.disease ,Prognosis ,Phosphoric Monoester Hydrolases ,PTPN11 ,Leukemia ,Leukemia, Myeloid, Acute ,business ,Nucleophosmin - Abstract
Prognostic factors associated with chemotherapy outcomes in patients with acute myeloid leukemia (AML) are extensively reported, and one gene whose mutation is recognized as conferring resistance to several newer targeted therapies is protein tyrosine phosphatase non-receptor type 11 (PTPN11). The broader clinical implications of PTPN11 mutations in AML are still not well understood. The objective of this study was to determine which cytogenetic abnormalities and gene mutations co-occur with PTPN11 mutations and how PTPN11 mutations affect outcomes of patients treated with intensive chemotherapy. We studied 1725 patients newly diagnosed with AML (excluding acute promyelocytic leukemia) enrolled onto the Cancer and Leukemia Group B/Alliance for Clinical Trials in Oncology trials. In 140 PTPN11-mutated patient samples, PTPN11 most commonly co-occurred with mutations in NPM1, DNMT3A, and TET2. PTPN11 mutations were relatively common in patients with an inv(3)(q21q26)/t(3;3)(q21;q26) and a normal karyotype but were very rare in patients with typical complex karyotype and core-binding factor AML. Mutations in the N-terminal SH2 domain of PTPN11 were associated with a higher early death rate than those in the phosphatase domain. PTPN11 mutations did not affect outcomes of NPM1-mutated patients, but these patients were less likely to have co-occurring kinase mutations (ie, FLT3-ITD), suggesting activation of overlapping signaling pathways. However, in AML patients with wild-type NPM1, PTPN11 mutations were associated with adverse patient outcomes, providing a rationale to study the biology and treatment approaches in this molecular group. This trial was registered at www.clinicaltrials.gov as #NCT00048958 (CALGB 8461), #NCT00899223 (CALGB 9665), and #NCT00900224 (CALGB 20202).
- Published
- 2021
61. Author response for 'Diagnostic utility of bronchoscopy in newly diagnosed acute leukemia patients'
- Author
-
Ashleigh Keiter, John C. Byrd, Kristin Lynn Koenig, Meixiao Long, James S. Blachly, Mark E. Lustberg, Thomas P. Curley, Gregory K. Behbehani, Alice S. Mims, Tamanna Haque, Nicole Grieselhuber, Bhavana Bhatnagar, Karilyn Larkin, Shylaja Mani, Qiuhong Zhao, Sarah A Wall, Alison R. Walker, and Sumithira Vasu
- Subjects
medicine.medical_specialty ,Acute leukemia ,Bronchoscopy ,medicine.diagnostic_test ,business.industry ,Medicine ,Newly diagnosed ,Radiology ,business - Published
- 2021
62. Off-the-Shelf Third-Party Virus-Specific T Cell Therapy to Treat JC Polyomavirus Infection in Hematopoietic Stem Cell Transplantation Recipients
- Author
-
Zeinab El Boghdadly, Xiang Zhu, Daria Heyenbruch, Stella M. Davies, Adam S. Nelson, Jamie Wilhelm, Sonata Jodele, Alice S. Mims, Carolyn Lutzko, Thomas Leemhuis, Shawn Thomas, Catherine M. Bollard, Michael D. Keller, Jeremy D. Rubinstein, Michael Grimley, Jose A. Cancelas, and Patrick J. Hanley
- Subjects
viruses ,JC virus ,Cell- and Tissue-Based Therapy ,medicine.disease_cause ,Virus ,medicine ,Immunology and Allergy ,Humans ,Child ,Retrospective Studies ,Transplantation ,Polyomavirus Infections ,business.industry ,ELISPOT ,Progressive multifocal leukoencephalopathy ,Hematopoietic Stem Cell Transplantation ,Leukoencephalopathy, Progressive Multifocal ,Hematopoietic stem cell ,Cell Biology ,Hematology ,medicine.disease ,JC Virus ,BK virus ,Graft-versus-host disease ,medicine.anatomical_structure ,Immunology ,Molecular Medicine ,business ,Viral load - Abstract
Background Progressive multifocal leukoencephalopathy (PML) is a progressive and generally fatal demyelinating neurological disease that occurs in profoundly immunocompromised patients due to infection with the human polyomavirus JC virus (JCPyV). Treatment options are limited and are largely focused on restoring T-cell immunity and outcomes are historically poor. Control of JCPyV in the setting of an immunocompromised patient by adoptive transfer of third-party virus specific T-cells (VSTs) has been described in a small number of cases. Objective To investigate treatment response and outcomes in recipients of hematopoietic stem cell transplant (HSCT) with PML treated with third-party VSTs directed against BK virus, a highly homologous polyoma virus that shares immunogenic epitopes with JCPyV. Study Design Retrospective chart review was performed on four patients who received VSTs for the treatment of PML at Cincinnati Children's Hospital Medical Center since 2019 Results VSTs were safely administered with no cases of graft-vs-host disease and no infusion reactions. One patient, who was treated almost immediately after diagnosis, was able to clear JCPyV from blood and CSF with resultant stabilization of neurologic decline. Interferon-gamma ELISpot demonstrated virus specific T-cells in the peripheral blood following infusion. Response was maintained through repeat infusions. Three other patients, all of whom had a longer delay between diagnosis and infusion, had progressive neurologic decline despite varying degree of improvement in viral load. Conclusion PML is a rare but often fatal complication following HSCT for which few treatment options are available. BK directed, JCPyV cross-reactive VSTs are a safe and viable therapeutic option and prompt administration should be considered after a diagnosis of PML is made. Key points • Virus specific T cells targeting JCPyV virus are safe with no infusional toxicity or de-novo graft versus host disease. • Virus specific T-cells have evidence of efficacy in some cases of PML, but further studies are needed to determine factors that will optimize response.
- Published
- 2021
63. A Clinical Phase 1B Study of the CD3xCD123 Bispecific Antibody APVO436 in Patients with Relapsed/Refractory Acute Myeloid Leukemia or Myelodysplastic Syndrome
- Author
-
Paul J. Shami, Prapti Patel, Fatih M. Uckun, Anoush Shahidzadeh, Justin M. Watts, Alice S. Mims, Elizabeth Cull, Christopher R. Cogle, Tara L. Lin, and Cynthia Lee
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Bispecific antibody ,T cells ,Article ,AML ,Internal medicine ,hemic and lymphatic diseases ,medicine ,MDS ,In patient ,Adverse effect ,RC254-282 ,business.industry ,leukemia ,Myeloid leukemia ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,CD123 ,bispecific antibody ,clinical study ,APVO436 ,medicine.disease ,Leukemia ,Cytokine release syndrome ,Tolerability ,Interleukin-3 receptor ,business - Abstract
Simple Summary AML is a common form of blood cancer in adults. This study was undertaken to evaluate if AML patients who have failed the available standard treatment options could tolerate and potentially benefit from a new form of therapy. This new therapy activates patients’ own immune system against AML cells. The findings from this research may provide the foundation for a potentially more effective future form of standard therapy that is less likely to fail. Abstract APVO436 is a recombinant T cell-engaging humanized bispecific antibody designed to redirect host T cell cytotoxicity in an MHC-independent manner to CD123-expressing blast cells from patients with hematologic malignancies and has exhibited single-agent anti-leukemia activity in murine xenograft models of acute myeloid leukemia (AML). In this first-in-human (FIH) multicenter phase 1B study, we sought to determine the safety and tolerability of APVO436 in R/R AML/myelodysplastic syndrome (MDS) patients and identify a clinically active recommended phase 2 dose (RP2D) level for its further clinical development. A total of 46 R/R AML/MDS patients who had failed 1–8 prior lines of therapy received APVO436 as weekly intravenous (IV) infusions at 10 different dose levels, ranging from a Minimum Anticipated Biological Effect Level (MABEL) of 0.3 mcg to 60 mcg. APVO436 exhibited a favorable safety profile with acceptable tolerability and manageable drug-related adverse events (AEs), and its maximum tolerated dose (MTD) was not reached at a weekly dose of 60 mcg. The most common APVO436-related AEs were infusion-related reactions (IRR) occurring in 13 (28.3%) patients and cytokine release syndrome (CRS) occurring in 10 (21.7%). The single dose RP2D level was identified as 0.2 mcg/kg. Preliminary efficacy signals were observed in both AML and MDS patients: Prolonged stable disease (SD), partial remissions (PR), and complete remissions (CR) were observed in R/R AML patients as best overall responses to APVO436 at the RP2D level. Three of six evaluable MDS patients had marrow CRs. The safety and preliminary evidence of efficacy of APVO436 in R/R AML and MDS patients warrant further investigation of its clinical impact potential.
- Published
- 2021
64. Outcomes of Bone Marrow Compared to Peripheral Blood for Haploidentical Transplantation
- Author
-
Karilyn Larkin, Audrey M. Sigmund, Alice S. Mims, Patrick Elder, Nicole Grieselhuber, Ayman Saad, Yvonne A. Efebera, Qiuhong Zhao, Srinivas Devarakonda, Sarah A Wall, Ashley E. Rosko, Don M. Benson, Muhammad Salman Faisal, Justin Jiang, Sumithira Vasu, Hannah Choe, Nidhi Sharma, Naresh Bumma, Abdullah Khan, Jonathan E. Brammer, Sam Penza, Samantha Jaglowski, and Maria Chaudhry
- Subjects
medicine.medical_specialty ,bone marrow ,Cyclophosphamide ,Gastroenterology ,Article ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,haploidentical transplantation ,business.industry ,Incidence (epidemiology) ,allogenic transplantation ,General Medicine ,peripheral blood ,Confidence interval ,Peripheral blood ,Transplantation ,medicine.anatomical_structure ,surgical procedures, operative ,030220 oncology & carcinogenesis ,Cohort ,Medicine ,Bone marrow ,Stem cell ,business ,030215 immunology ,medicine.drug - Abstract
Allogeneic hematopoietic cell transplantation (allo-HCT) from a haploidentical (haplo) donor has emerged as a suitable alternative in the absence of a matched donor. However, haplo-HCT patients have a higher risk of graft-versus-host disease (GVHD). Hence, bone marrow (BM) stem cell source and post-transplant cyclophosphamide (PTCy) have been routinely used to help mitigate this. Due to ease of collection, peripheral blood (PB) stem cells are increasingly being considered for haplo-HCT. We retrospectively analyzed 74 patients (42 BM and 32 PB) who underwent haplo-HCT at Ohio State University from 2009 to 2018. Median age at transplant was 60 years (yrs) for BM and 54 yrs for PB, (p = 0.45). There was no difference in OS (p = 0.13) and NRM (p = 0.75) as well as PFS (p = 0.10) or GRFS (p = 0.90) between the groups. The BM cohort showed a 3-year OS rate of 63% (95% confidence interval (CI): 46–76), and 3-year PFS of 49% (95% CI: 33–63). For the PB group, 3-year OS and PFS were 78% (95% CI: 59–89) and 68% (95% CI: 49–82), respectively. There were no differences in the incidence of acute GVHD (grade II-IV) (p = 0.31) and chronic GVHD (p = 0.18). Patients receiving BM had a significantly higher risk for relapse with relapse rates by 2 years at 36% (95% CI: 22–50) vs. 16% (95% CI: 6–31) for PB (p = 0.03). The findings from this study suggest that PB is an excellent alternative to BM for haplo-HCT.
- Published
- 2021
65. Poor Treatment Outcomes of Young (<60 Years) African American Patients (Pts) Diagnosed with Acute Myeloid Leukemia (AML) (Alliance)
- Author
-
Christopher J. Walker, Richard Stone, Ann-Kathrin Eisfeld, Bayard L. Powell, Electra D. Paskett, Qiuhong Zhao, James L. Fisher, Alice S. Mims, Jonathan E. Kolitz, Jessica Kohlschmidt, Ramiro Garzon, John C. Byrd, James S. Blachly, Krzysztof Mrózek, Deedra Nicolet, Albert de la Chapelle, Shelley Orwick, Clara D. Bloomfield, Andrew J. Carroll, and Bhavana Bhatnagar
- Subjects
African american ,medicine.medical_specialty ,education.field_of_study ,business.industry ,Immunology ,Treatment outcome ,Population ,Cell Biology ,Hematology ,Gene mutation ,Biochemistry ,Health equity ,Clinical trial ,Kite Pharma ,Alliance ,Family medicine ,medicine ,education ,business - Abstract
Background: AML is a clinically and molecularly heterogeneous disease associated with poor survival. Multiple disease-related factors including cytogenetic findings and gene mutations, as well as patient-related factors, such as demographics and African American (AA) heritage, have been identified that impact on pt outcomes. However, with recent improved survival it is unknown whether racial health disparities persist. Moreover, we are not aware of a large study that assessed possible race-associated molecular differences. Thus, the goals of our study were to 1) analyze the outcomes of adult AML pts in a nationwide population study, including possible impacts of sociodemographic, financial and racial disparities and 2) characterize molecular features of AA compared with those of Caucasian AML pts. Methods: For a nationwide population analysis, the Surveillance Epidemiology End Results (SEER) Program of the National Cancer Institute was used to identify 11,190 adults aged 18-60 years (y) diagnosed with AML (excluding acute promyelocytic leukemia) between 1986 and 2015. To characterize molecular features we performed targeted sequencing of 81 genes in 1,339 AML pts treated on frontline Cancer and Leukemia Group B/Alliance for Clinical Trials in Oncology (Alliance) protocols based on standard intensity cytarabine/anthracycline induction followed by consolidation between 1986 and 2016. No Alliance pt received an allogeneic stem cell transplant in 1st complete remission (CR). Results: The associations between demographic parameters and risk of death among SEER registry AML pts are shown in Table 1. While there was a slightly higher risk of death for men (HR 1.09) and a lower risk of death for pts with a higher median household income (>79.6k vs Conclusion: Self-reported AA race is the most important pt-associated factor associated with poor survival in AML pts < 60 y of age based on SEER. Survival analyses in Alliance pts identify AA race as independent poor survival prognosticator in AML pts besides established molecular markers. . This disparity must be urgently addressed to ensure improved outcomes for AA AML pts, and larger studies to establish molecular risk profiles are needed. Support: U10CA180821, U10CA180882 U24CA196171, https://acknowledgments.alliancefound.org; Clinicaltrials.gov Identifiers: NCT00048958, NCT00899223, NCT00900224 Disclosures Bhatnagar: KaryoPharm Therapuetics: Research Funding; Cell Therapeutics: Membership on an entity's Board of Directors or advisory committees, Research Funding; Novartis: Membership on an entity's Board of Directors or advisory committees; Astellas: Membership on an entity's Board of Directors or advisory committees; Pfizer: Membership on an entity's Board of Directors or advisory committees; KITE: Membership on an entity's Board of Directors or advisory committees. Blachly:AbbVie, AstraZeneca, KITE Pharma: Consultancy. Mims:Novartis: Speakers Bureau; Jazz Pharmaceuticals: Other: Data Safety Monitoring Board; Syndax Pharmaceuticals: Membership on an entity's Board of Directors or advisory committees; Abbvie: Membership on an entity's Board of Directors or advisory committees; Kura Oncology: Membership on an entity's Board of Directors or advisory committees; Leukemia and Lymphoma Society: Other: Senior Medical Director for Beat AML Study; Agios: Consultancy. Walker:Vigeo Therapeutics: Consultancy; Karyopharm: Current Employment, Current equity holder in publicly-traded company. Powell:Genentech: Research Funding; Jazz Pharmaceuticals: Consultancy, Other: Advisor, Research Funding; Pfizer: Research Funding; Rafael Pharmaceuticals: Consultancy, Other: Advisor, Research Funding; Novartis: Research Funding. Kolitz:Magellan: Membership on an entity's Board of Directors or advisory committees; Pfizer: Membership on an entity's Board of Directors or advisory committees. Stone:Biolinerx: Consultancy; Novartis: Consultancy, Research Funding; Celgene: Consultancy, Other; Aztra-Zeneca: Consultancy; Jazz: Consultancy; Argenix: Other; Janssen: Consultancy; Daiichi-Sankyo: Consultancy; Pfizer: Consultancy; Gemoab: Consultancy; Syndax: Consultancy, Research Funding; Takeda: Other: DSMB; Macrogenics: Consultancy; Trovagene: Consultancy; Syntrix: Other: DSMB; Abbvie: Consultancy, Research Funding; Agios: Consultancy, Research Funding; Actinium: Consultancy, Membership on an entity's Board of Directors or advisory committees; Astellas: Consultancy; Arog: Consultancy, Research Funding; Syros: Consultancy; Stemline: Consultancy. Byrd:Trillium: Research Funding; Leukemia and Lymphoma Society: Other; Acerta Pharma: Research Funding; Syndax: Research Funding; Pharmacyclics LLC, an AbbVie Company, Janssen, Novartis, Gilead, TG Therapeutics: Other; Janssen: Consultancy; Pharmacyclics LLC, an AbbVie Company, Gilead, TG Therapeutics, BeiGene: Research Funding; Pharmacyclics LLC, an AbbVie Company, Gilead, TG Therapeutics, Novartis, Janssen: Speakers Bureau; Kartos Therapeutics: Research Funding; Vincera: Research Funding; Novartis: Research Funding. Eisfeld:Karyopharm: Current Employment, Current equity holder in publicly-traded company; Vigeo Therapeutics: Consultancy.
- Published
- 2020
66. Hypomethylating agents super-responders: challenging the dogma of long-term remission for acute myeloid leukemia
- Author
-
Michael Ozga, Will Pulley, Joseph Maakaron, Alice S. Mims, Gabriel N. Mannis, Joshua F. Zeidner, and Matthew C. Foster
- Subjects
Oncology ,medicine.medical_specialty ,Myeloid ,Hematology ,business.industry ,Treatment outcome ,Myeloid leukemia ,General Medicine ,medicine.disease ,Remission induction ,Leukemia ,medicine.anatomical_structure ,Internal medicine ,medicine ,Molecular diagnostic techniques ,Long term remission ,business - Published
- 2020
67. Selinexor in combination with decitabine in patients with acute myeloid leukemia: results from a phase 1 study
- Author
-
William Blum, John C. Byrd, Sumithira Vasu, Gregory K. Behbehani, Rebecca B. Klisovic, Karilyn Larkin, Ramiro Garzon, Amy S. Ruppert, Alice S. Mims, Qiuhong Zhao, James S. Blachly, Shelley Orwick, Bhavana Bhatnagar, Christopher C. Oakes, Parvathi Ranganathan, and Alison Walker
- Subjects
Adult ,Oncology ,Cancer Research ,medicine.medical_specialty ,Decitabine ,Article ,Unmet needs ,03 medical and health sciences ,0302 clinical medicine ,Refractory ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Humans ,In patient ,Aged ,business.industry ,Treatment options ,Myeloid leukemia ,Hematology ,Triazoles ,Clinical trial ,Leukemia, Myeloid, Acute ,Hydrazines ,030220 oncology & carcinogenesis ,Azacitidine ,business ,030215 immunology ,medicine.drug - Abstract
Current treatment options for older and relapsed or refractory (R/R) acute myeloid leukemia (AML) patients are limited and represent an unmet need. Based on preclinical studies showing strong anti-leukemic effects in vivo, this phase I dose-escalation study assessed the safety and preliminary clinical activity of the oral exportin-1 inhibitor, selinexor, in combination with the hypomethylating agent, decitabine 20 mg/m(2), in adults with R/R AML and in older (age ≥60) untreated AML patients. There were no protocol-defined dose limiting toxicities. The recommended phase 2 dose of selinexor was 60mg (~35 mg/m(2)) given twice-weekly. Notable grade ≥3 toxicities included asymptomatic hyponatremia (68%), febrile neutropenia (44%), sepsis (44%), hypophosphatemia (36%), and pneumonia (28%). In 25 patients, the overall response rate was 40%. Modification of selinexor to a flat dose of 60mg, twice-weekly for two weeks after decitabine, improved tolerability of the regimen and demonstrated preliminary clinical activity in poor-risk patients with AML.
- Published
- 2019
68. Daunorubicin-cytarabine liposome (CPX-351) in the management of newly diagnosed secondary AML: A new twist on an old cocktail
- Author
-
Joseph E. Maakaron and Alice S. Mims
- Subjects
Male ,Oncology ,medicine.medical_specialty ,Anthracycline ,Daunorubicin ,Clinical Biochemistry ,Disease-Free Survival ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Humans ,Secondary Acute Myeloid Leukemia ,Aged ,Cytopenia ,business.industry ,Cytarabine ,Myeloid leukemia ,medicine.disease ,Survival Rate ,Clinical trial ,Leukemia, Myeloid, Acute ,Regimen ,030220 oncology & carcinogenesis ,Female ,business ,030215 immunology ,medicine.drug - Abstract
Initial therapy for acute myeloid leukemia (AML) remained stagnant for approximately four decades despite advances in improved understanding of pathogenesis and prognostication of the disease. Treatment has typically consisted of an anthracycline combined with continuous infusion of cytarabine for 7 days, the “7 + 3” regimen. Attempts have been made to improve on this regimen with modest improvements in response rates but no change in overalll survival, until the recent introduction of mutation-specific agents. However, the re-vamping of the delivery of both daunorubicin and cytarabine in a liposomal encapsulation, known as CPX-351, did show improvements of overall survival compared to traditional 7 + 3 in newly diagnosed secondary and therapy-related AML in patients aged 60–75. This led to the Food and Drug Administration (FDA) approval of the agent for both of these subtypes of AML in August of 2017. Herein we will review the rationale and preclinical development of CPX-351 and discuss the pivotal studies that led to its FDA approval.
- Published
- 2019
69. Acute Myeloid Leukemia, Version 3.2019, NCCN Clinical Practice Guidelines in Oncology
- Author
-
Steven Coutre, Alexander E. Perl, Richard Stone, Vijaya Raj Bhatt, Deniz Peker, Ndiya Ogba, Keith W. Pratz, Frederick R. Appelbaum, Martin S. Tallman, Michael Gary Martin, Thomas W. LeBlanc, Alice S. Mims, Dale L. Bixby, Aric C. Hall, Paul J. Shami, Jeffrey E. Lancet, Marcos de Lima, Stephen A. Strickland, Thomas Prebet, Melanie Fiorella, Meagan A. Jacoby, Lydia J. Hammond, Margaret R. O'Donnell, Guido Marcucci, Jessica K. Altman, Rebecca L. Olin, Daniel A. Pollyea, Gabriel N. Mannis, Matthew J. Wieduwilt, Farhad Ravandi, Eunice S. Wang, Kristina M. Gregory, James M. Foran, and Amir T. Fathi
- Subjects
0301 basic medicine ,Graft vs Host Disease ,Medical Oncology ,Risk Assessment ,Disease-Free Survival ,03 medical and health sciences ,0302 clinical medicine ,HLA Antigens ,hemic and lymphatic diseases ,Antineoplastic Combined Chemotherapy Protocols ,Biomarkers, Tumor ,Humans ,Transplantation, Homologous ,Medicine ,Aged ,business.industry ,Histocompatibility Testing ,Remission Induction ,Age Factors ,Hematopoietic Stem Cell Transplantation ,Myeloid leukemia ,Middle Aged ,United States ,Leukemia, Myeloid, Acute ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cytogenetic Analysis ,Cancer research ,business - Abstract
Acute myeloid leukemia (AML) is the most common form of acute leukemia among adults and accounts for the largest number of annual deaths due to leukemias in the United States. Recent advances have resulted in an expansion of treatment options for AML, especially concerning targeted therapies and low-intensity regimens. This portion of the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines) for AML focuses on the management of AML and provides recommendations on the workup, diagnostic evaluation and treatment options for younger (age
- Published
- 2019
70. Progress in the problem of relapsed or refractory acute myeloid leukemia
- Author
-
Alice S. Mims and William Blum
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Myeloid ,medicine.medical_treatment ,Antineoplastic Agents ,Drug resistance ,Disease ,Small Molecule Libraries ,03 medical and health sciences ,0302 clinical medicine ,Refractory ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Humans ,business.industry ,Myeloid leukemia ,Treatment options ,Hematology ,Immunotherapy ,medicine.disease ,Leukemia, Myeloid, Acute ,Leukemia ,030104 developmental biology ,medicine.anatomical_structure ,Drug Resistance, Neoplasm ,business ,030215 immunology - Abstract
The majority of patients with acute myeloid leukemia (AML) die from disease recurrence and historically, treatment options in both the relapsed and refractory settings of this disease have been limited. However, new insights into the molecular characterization and biology of relapsed and refractory AML have led to novel therapeutics and improvement in outcomes in these settings. The current understanding of mechanisms of disease resistance and status of treatment options both currently available and under exploration in relapsed and refractory AML are summarized in this review.The rapid approval of multiple therapeutic agents since 2017 has led to improvement in selected populations such as isocitrate dehydrogenase and fms-like tyrosine kinase 3-mutated relapsed and refractory AML with agents such as enasidenib, ivosidenib, and gilteritinib. Despite these advancements, the only current curative approach remains allogeneic transplantation and only for those minority of patients that are candidates. However, encouraging results are being seen with a multitude of novel small molecular inhibitors and immunotherapeutic approaches currently in clinical trials both as single agents and combination strategies in both upfront and relapsed/refractory AML.Continued advancements in the knowledge of various mechanisms of relapse and resistance in AML are ongoing, leading to the realization that diverse treatment strategies are needed to both prevent and manage relapsed and refractory disease.
- Published
- 2019
71. HSP90 inhibition depletes DNA repair proteins to sensitize acute myelogenous leukemia to nucleoside analog chemotherapeutics
- Author
-
Alice S. Mims, William Plunkett, Janani Ravikrishnan, Shaneice Mitchell, Jennifer A. Woyach, Vinay K. Puduvalli, Rosa Lapalombella, Deepa Sampath, Tzung Huei Lai, Shelley Orwick, John C. Byrd, Pei Jung Wu, and Chaomei Liu
- Subjects
Cancer Research ,Myeloid ,DNA Repair ,DNA repair ,medicine.medical_treatment ,Drug resistance ,Isoindoles ,Article ,Cytosine ,03 medical and health sciences ,Myelogenous ,0302 clinical medicine ,Cell Line, Tumor ,hemic and lymphatic diseases ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,DNA Breaks, Double-Stranded ,HSP90 Heat-Shock Proteins ,Chemotherapy ,business.industry ,Hematology ,medicine.disease ,Leukemia, Myeloid, Acute ,Leukemia ,medicine.anatomical_structure ,Oncology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Benzamides ,Cancer research ,Cytarabine ,Arabinonucleosides ,business ,Nucleoside ,030215 immunology ,medicine.drug - Abstract
The treatment of acute myelogenous leukemia (AML) relies heavily on cytarabine (Ara-C) containing chemotherapy. Even newly approved multi-kinase inhibitors improved survival only when combined with...
- Published
- 2019
72. Abstract 1060: Introducing a novel DHODH inhibitor with superior in vivo activity as monotherapy or in novel combination regimen with immunotherapy for hematological malignancies
- Author
-
Ola A. Elgamal, Sandip Vibhute, Sydney Fobare, Abeera Mehmood, Mariah L. Johnson, Jean Truxall, Emily Stahl, Bridget Carmichael, Shelley J. Orwick, Ramasamy Santhanam, Kasey Hill, Susheela Tridandapani, Christopher C. Coss, Alice S. Mims, Karilyn T. Larkin, Mitch A. Phelps, Sharyn D. Baker, Alex Sparreboom, Thomas E. Goodwin, Gerard Hilinski, Chad E. Bennett, Erin Hertlein, and John C. Byrd
- Subjects
Cancer Research ,Oncology - Abstract
Acute myeloid leukemia (AML) is characterized by the uncontrolled expansion of un-differentiated hematopoietic progenitor myeloblasts. AML treatment is very challenging owing to its complex heterogeneity resulting in a dismal 5-year overall survival rate particularly in elderly patients unfit for standard induction chemotherapy. The expansion of AML requires the availability of sufficient nucleotides supporting the anabolic processes required for AML growth thus, targeting nucleotide biosynthesis can halt AML progression. Indeed, targeting dihydroorotate dehydrogenase (DHODH), a critical rate-limiting step in the de novo pyrimidine synthesis pathway not only induced cytotoxicity but has been shown to promote blast differentiation in a HOXA9/MEIS1 over-expressing model. We sought to develop a DHODH inhibitor that had superior properties to those reported for AML therapy. Compound 41 (cmpd 41) demonstrates sub-nanomolar 50% inhibitory concentration for DHODH biochemical activity and potent in vitro activity across several AML cell lines and primary AML cells independent of mutational subtype, including mutated TP53. Cmpd 41 also demonstrated superior in vivo anti-leukemic activity in multiple AML xenograft models as monotherapy and demonstrated synergy with a hypomethylating agent, decitabine in TP53 mutated AML. Given the heterogeneity of AML and frequent emergence of resistant clones, we aimed to investigate ways to enhance response to DHODH inhibitors through combination. After in vitro treatment of AML cell lines and primary patient samples with DHODH inhibitors, we observed an increase in CD38 surface expression suggesting synergy with CD38 targeting monoclonal antibody (mAb) immunotherapies. Indeed, we are the first to report synergy between DHODH inhibitors and anti-CD38 mAb in AML which emphasizes the synergy between this promising novel class of agents with immunotherapies via recruiting innate immunity. Consequently, given the relevance of CD38 mAb therapy to multiple myeloma (MM), we extended these studies to MM and remarkably found that cmpd 41 was highly efficacious as a monotherapy and in combination with CD38 mAb, resulted in complete tumor regression in a subcutaneous MM xenograft model. In summary, we introduce a best in class DHODH inhibitor with a data-driven combination strategy for both AML and MM. Our studies suggest a highly synergistic combination strategy involving immunotherapy for AML and other hematologic malignancies. Citation Format: Ola A. Elgamal, Sandip Vibhute, Sydney Fobare, Abeera Mehmood, Mariah L. Johnson, Jean Truxall, Emily Stahl, Bridget Carmichael, Shelley J. Orwick, Ramasamy Santhanam, Kasey Hill, Susheela Tridandapani, Christopher C. Coss, Alice S. Mims, Karilyn T. Larkin, Mitch A. Phelps, Sharyn D. Baker, Alex Sparreboom, Thomas E. Goodwin, Gerard Hilinski, Chad E. Bennett, Erin Hertlein, John C. Byrd. Introducing a novel DHODH inhibitor with superior in vivo activity as monotherapy or in novel combination regimen with immunotherapy for hematological malignancies [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 1060.
- Published
- 2022
73. A phase 1b/2 study of TP-0903 and decitabine targeting mutant TP53 and/or complex karyotype in patients with untreated acute myeloid leukemia ≥ age 60 years: Phase 1b interim results
- Author
-
Alice S. Mims, Ying Huang, Eric Eisenmann, Daelynn Buelow, Ronan T. Swords, Matthew Charles Foster, Tara L. Lin, Maria R. Baer, Tibor Kovacsovics, Zeina Al-Mansour, Mona Stefanos, Franchesca Druggan, Timothy Chen, Ashley Yocum, Uma Borate, Brian J. Druker, Amy Burd, Ross L. Levine, Sharyn D. Baker, and John C. Byrd
- Subjects
Cancer Research ,Oncology - Abstract
7027 Background: TP-0903 is a multi-kinase inhibitor designed to target AXL, a receptor tyrosine kinase, and also inhibits cell cycle regulators such as Chk1/2 and other AML associated kinases. TP-0903 has shown prior anti-tumor activity at a safe dose in solid tumors. In pre-clinical AML studies, TP-0903 shows potent cytotoxicity in TP53 mutant ( TP53m) AML cell lines, an adverse prognostic genomic sub-group of AML. TP-0903 also had synergistic activity with decitabine (dec) in TP53m AML and prolonged survival in xenograft and genetically engineered mouse models. We report here on the initial safety and clinical results from the Leukemia and Lymphoma Society’s ongoing Beat AML phase 1b/2 (Ph1b/2) trial of TP-0903 in combination with dec (ClinicalTrials.gov NCT03013998). Methods: Newly diagnosed AML pts ≥60 years with TP53m and/or complex karyotype (≥3 abnormalities) were selected for a Ph1b/2 dose escalation study of TP-0903 combined with dec. Seven Ph1b pts were given TP-0903 every 28-day cycle from days 1-21 (Dose level (DL) 1 = 37 mg/day) and dec IV days 1-10 (20 mg/m2). A standard 3+3 design was used to evaluate the safety and tolerability. Nine additional patients enrolled onto Ph2 at DL1, but further assessments of safety, pharmacokinetics (PK) and correlative data was used to update the final recommended Ph2 dose (RP2D) of TP-0903 to DL-1 (25 mg/day) with dec. Results: At data cutoff (10Jan2022), 16 total pts were accrued. Ph1b treated 7 pts at DL1, 6 were DLT evaluable, and no DLTs were observed. Ph2 enrolled and treated 9 pts at DL1 before concerns of delayed count recovery led to the reduction of the Ph2 dose of TP-0903 to DL-1 (25 mg/day). For all 16 pts treated at DL1, 1 pt achieved CR, 4 pts CRh, and 1 pt CRi, for a composite CR (CR/CRh/CRi) rate of 37.5% (95% CI, 15.2-64.6), with 4 pts achieving MRD negativity by central flow cytometry. For the remaining 10 pts, 1 pt achieved MLFS (6%), 6 pts had stable disease (37.5%), 1 pt had treatment failure (6%), and 2 pts were not evaluable (12.5%) due to withdrawal of consent and death from early disease progression. Two pts (1 CR and 1 CRh) proceeded to stem cell transplantation. The most common grade 3 and above treatment-related AEs include decreased neutrophil counts (37.5%), platelet counts (31.3%), and anemia (18.8%). Finally, PK and correlative data analysis looking at soluble Axl and Gas6 also supported reduction to DL-1. Conclusions: Initial results with DL1 suggest that TP-0903/dec shows preliminary clinical activity in the prognostically poor TP53m/complex karyotype AML sub-group, with 4 pts achieving MRD negative status out of 6 patients who achieved a CR/CRh/CRi (66%). After further patients were treated on DL1, the toxicity profile and correlative data supported the de-escalation to DL-1 as the RP2D. The Ph2 study is ongoing to determine the clinical activity of this new RP2D (DL-1). Clinical trial information: NCT03013998.
- Published
- 2022
74. Type of prior genotoxic insult determines the genomic characteristics of therapy‐related myeloid neoplasms
- Author
-
Ann-Kathrin Eisfeld, Dan Jones, Alice S. Mims, Nicole Grieselhuber, Weiqiang Zhao, Tamanna Haque, John C. Byrd, Sumithira Vasu, Bhavana Bhatnagar, James S. Blachly, Meixiao Long, Caner Saygin, Gregory K. Behbehani, Karilyn Larkin, Michael Ozga, and Alison Walker
- Subjects
Myeloid ,Therapy related ,business.industry ,media_common.quotation_subject ,Smoking ,Antineoplastic Agents ,Neoplasms, Second Primary ,Hematology ,Insult ,medicine.anatomical_structure ,Mutation Rate ,Leukemia, Myeloid ,Risk Factors ,Mutation ,Cancer research ,Humans ,Medicine ,business ,DNA Damage ,media_common - Published
- 2021
75. Ivosidenib or enasidenib combined with intensive chemotherapy in patients with newly diagnosed AML: a phase 1 study
- Author
-
Daniel A. Pollyea, Anthony S. Stein, Mikhail Roshal, Michael R. Savona, Olatoyosi Odenike, Courtney D. DiNardo, Hongfang Wang, Eric S. Winer, Richard Stone, Aleksandra Franovic, Alice S. Mims, Gert J. Ossenkoppele, Lei Hua, Hartmut Döhner, Martin S. Tallman, Bin Wu, Amir T. Fathi, Prapti A. Patel, Sung Choe, Frederik Lersch, Salah Nabhan, Caroline Almon, Bob Löwenberg, Keith W. Pratz, Mark G. Frattini, Bin Fan, Eytan M. Stein, Michael Cooper, Christopher S. Seet, Hagop M. Kantarjian, James K. McCloskey, Hematology laboratory, CCA - Cancer Treatment and quality of life, and Hematology
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Myeloid ,business.industry ,Immunology ,Myeloid leukemia ,Consolidation Chemotherapy ,Cell Biology ,Hematology ,Enasidenib ,medicine.disease ,Biochemistry ,IDH2 ,QT interval ,Gastroenterology ,03 medical and health sciences ,Leukemia ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Refractory ,030220 oncology & carcinogenesis ,Internal medicine ,Medicine ,business - Abstract
Ivosidenib (AG-120) and enasidenib (AG-221) are targeted oral inhibitors of the mutant isocitrate dehydrogenase (mIDH) 1 and 2 enzymes, respectively. Given their effectiveness as single agents in mIDH1/2 relapsed or refractory acute myeloid leukemia (AML), this phase 1 study evaluated the safety and efficacy of ivosidenib or enasidenib combined with intensive chemotherapy in patients with newly diagnosed mIDH1/2 AML. Ivosidenib 500 mg once daily and enasidenib 100 mg once daily were well tolerated in this setting, with safety profiles generally consistent with those of induction and consolidation chemotherapy alone. The frequency of IDH differentiation syndrome was low, as expected given the concurrent administration of cytotoxic chemotherapy. In patients receiving ivosidenib, the frequency and grades of QT interval prolongation were similar to those observed with ivosidenib monotherapy. Increases in total bilirubin were more frequently observed in patients treated with enasidenib, consistent with this inhibitor’s known potential to inhibit UGT1A1, but did not appear to have significant clinical consequences. In patients receiving ivosidenib (n = 60) or enasidenib (n = 91), end-of-induction complete remission (CR) rates were 55% and 47%, respectively, and CR/CR with incomplete neutrophil or platelet recovery (CR/CRi/CRp) rates were 72% and 63%, respectively. In patients with a best overall response of CR/CRi/CRp, 16/41 (39%) receiving ivosidenib had IDH1 mutation clearance and 15/64 (23%) receiving enasidenib had IDH2 mutation clearance by digital polymerase chain reaction; furthermore, 16/20 (80%) and 10/16 (63%), respectively, became negative for measurable residual disease by multiparameter flow cytometry. This trial was registered at www.clinicaltrials.gov as #NCT02632708.
- Published
- 2021
76. Novel Targeted Therapeutics in Acute Myeloid Leukemia: an Embarrassment of Riches
- Author
-
Alice S. Mims and Nicole Grieselhuber
- Subjects
Cancer Research ,medicine.medical_specialty ,IDH1 ,medicine.medical_treatment ,Article ,Targeted therapy ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,hemic and lymphatic diseases ,Biomarkers, Tumor ,Medicine ,Humans ,Molecular Targeted Therapy ,neoplasms ,Protein Kinase Inhibitors ,Clinical Trials as Topic ,Hematology ,business.industry ,Drug discovery ,Myeloid leukemia ,Disease Management ,DOT1L ,Clinical trial ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,Treatment Outcome ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Bone marrow ,Disease Susceptibility ,business ,Nucleophosmin ,030215 immunology - Abstract
PURPOSE OF REVIEW: Acute myeloid leukemia (AML) is an aggressive malignancy of the bone marrow that has a poor prognosis with traditional cytotoxic chemotherapy, especially in elderly patients. In recent years, small molecule inhibitors targeting AML associated IDH1, IDH2 and FLT3 mutations have been FDA approved. However, the majority of AML cases do not have a targetable mutation. A variety of novel agents targeting both previously untargetable mutations and general pathways in AML are currently being investigated. Herein, we review selected new targeted therapies currently in early phase clinical investigation in AML. RECENT FINDINGS: The DOT1L inhibitor pinometostat in KMT2A rearranged AML, the menin inhibitors KO-539 and SYNDX-5613 in KMT2Ar and NPM1 mutated AML and the mutant TP53 inhibitor APR-246 are examples of novel agents targeting specific mutations in AML. In addition, BET inhibitors, polo-like kinase inhibitors and MDM2 inhibitors are promising new drug classes for AML which do not depend on the presence of a particular mutation. SUMMARY: AML remains in incurable disease for many patients but advances in genomics, epigenetics and drug discovery have led to the development of many potential novel therapeutic agents, many of which are being investigated in on-going clinical trials. Additional studies will be necessary to determine how best to incorporate these novel agents into routine clinical treatment of AML.
- Published
- 2021
77. Targeting DNA damage repair functions of two histone deacetylases, HDAC8 and SIRT6, sensitizes acute myeloid leukemia to NAMPT inhibition
- Author
-
Samuel K. Kulp, Nicole Grieselhuber, James S. Blachly, Tierney Kauffman, Deepa Sampath, Hannah Phillips, Matthew Cannon, Lapo Alinari, Youssef Youssef, Pearlly S. Yan, Katie Williams, Larry Beaver, Lai Tzung-Huei, Steven Sher, Allison M. Mustonen, Christopher C. Coss, Alice S. Mims, John C. Byrd, Shelley Orwick, Lindsey T. Brinton, Daniel Canfield, Pu Zhang, Shaneice Mitchell, Amy Lehman, Wing Keung Chan, and Rosa Lapalombella
- Subjects
0301 basic medicine ,Male ,Cancer Research ,DNA End-Joining Repair ,Indoles ,Combination therapy ,DNA repair ,Nicotinamide phosphoribosyltransferase ,Aminopyridines ,Synthetic lethality ,Hydroxamic Acids ,Article ,Histone Deacetylases ,03 medical and health sciences ,chemistry.chemical_compound ,Gene Knockout Techniques ,Mice ,0302 clinical medicine ,Cell Line, Tumor ,Antineoplastic Combined Chemotherapy Protocols ,Medicine ,Animals ,Humans ,Sirtuins ,Nicotinamide Phosphoribosyltransferase ,Acrylamides ,business.industry ,Myeloid leukemia ,Recombinational DNA Repair ,HDAC8 ,Phenylbutyrates ,Xenograft Model Antitumor Assays ,Non-homologous end joining ,Repressor Proteins ,Haematopoiesis ,Leukemia, Myeloid, Acute ,030104 developmental biology ,Oncology ,chemistry ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Cancer research ,Cytokines ,business ,DNA Damage - Abstract
Purpose: Nicotinamide phosphoribosyltransferase (NAMPT) inhibitors (NAMPTi) are currently in development, but may be limited as single-agent therapy due to compound-specific toxicity and cancer metabolic plasticity allowing resistance development. To potentially lower the doses of NAMPTis required for therapeutic benefit against acute myeloid leukemia (AML), we performed a genome-wide CRISPRi screen to identify rational disease-specific partners for a novel NAMPTi, KPT-9274. Experimental Design: Cell lines and primary cells were analyzed for cell viability, self-renewal, and responses at RNA and protein levels with loss-of-function approaches and pharmacologic treatments. In vivo efficacy of combination therapy was evaluated with a xenograft model. Results: We identified two histone deacetylases (HDAC), HDAC8 and SIRT6, whose knockout conferred synthetic lethality with KPT-9274 in AML. Furthermore, HDAC8-specific inhibitor, PCI-34051, or clinical class I HDAC inhibitor, AR-42, in combination with KPT-9274, synergistically decreased the survival of AML cells in a dose-dependent manner. AR-42/KPT-9274 cotreatment attenuated colony-forming potentials of patient cells while sparing healthy hematopoietic cells. Importantly, combined therapy demonstrated promising in vivo efficacy compared with KPT-9274 or AR-42 monotherapy. Mechanistically, genetic inhibition of SIRT6 potentiated the effect of KPT-9274 on PARP-1 suppression by abolishing mono-ADP ribosylation. AR-42/KPT-9274 cotreatment resulted in synergistic attenuation of homologous recombination and nonhomologous end joining pathways in cell lines and leukemia-initiating cells. Conclusions: Our findings provide evidence that HDAC8 inhibition- or shSIRT6-induced DNA repair deficiencies are potently synergistic with NAMPT targeting, with minimal toxicity toward normal cells, providing a rationale for a novel–novel combination-based treatment for AML.
- Published
- 2021
78. Effect of Early Post-Transplantation Tacrolimus Concentration on the Risk of Acute Graft-Versus-Host Disease in Allogenic Stem Cell Transplantation
- Author
-
Don M. Benson, Jonathan E. Brammer, Nicole Grieselhuber, Karilyn Larkin, Nidhi Sharma, Naresh Bumma, Qiuhong Zhao, Abdullah Khan, Patrick Elder, Samantha Jaglowski, Alice S. Mims, Bin Ni, Hannah Choe, Yvonne A. Efebera, Srinivas Devarakonda, Sam Penza, Maria Chaudhry, Basem M. William, Ayman Saad, Marcin Puto, Ashley E. Rosko, Sumithira Vasu, and Sarah A Wall
- Subjects
Cancer Research ,medicine.medical_specialty ,chemical and pharmacologic phenomena ,Lower risk ,Gastroenterology ,lcsh:RC254-282 ,Article ,03 medical and health sciences ,0302 clinical medicine ,immune system diseases ,allogeneic stem cell transplantation ,Internal medicine ,hemic and lymphatic diseases ,graft versus host disease ,medicine ,Cumulative incidence ,tacrolimus ,relapse ,business.industry ,Myeloid leukemia ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Tacrolimus ,Calcineurin ,Transplantation ,stomatognathic diseases ,Graft-versus-host disease ,surgical procedures, operative ,Oncology ,Quartile ,030220 oncology & carcinogenesis ,business ,030215 immunology - Abstract
Acute graft versus host disease (aGVHD) remains a leading cause of morbidity and mortality in allogeneic hematopoietic stem cell transplant (allo-HSCT). Tacrolimus (TAC), a calcineurin inhibitor that prevents T-cell activation, is commonly used as a GVHD prophylaxis. However, there is variability in the serum concentrations of TAC, and little is known on the impact of early TAC levels on aGVHD. We retrospectively analyzed 673 consecutive patients undergoing allo-HSCT at the Ohio State University between 2002 and 2016. Week 1 TAC was associated with a lower risk of aGVHD II–IV at TAC level ≥ 10.15 ng/mL (p = 0.03) compared to the lowest quartile. The cumulative incidence of relapse at 1, 3 and 5 years was 33%, 38% and 41%, respectively. TAC levels at week 2, ≥ 11.55 ng/mL, were associated with an increased risk of relapse (p = 0.01) compared to the lowest quartile. Subset analysis with acute myeloid leukemia and myelodysplastic syndrome patients showed significantly reduced aGVHD with TAC level ≥ 10.15 ng/mL at week 1 and a higher risk of relapse associated with week 2 TAC level ≥11.55 ng/mL (p = 0.02). Hence, achieving ≥10 ng/mL during the first week of HCT may mitigate the risk of aGVHD. However, levels (>, 11 ng/mL) beyond the first week may be associated with suppressed graft versus tumor effect and higher relapse.
- Published
- 2021
79. PP2A is a therapeutically targetable driver of cell fate decisions via a c-Myc/p21 axis in human and murine acute myeloid leukemia
- Author
-
Alice S. Mims, Rosa Lapalombella, Nicole Grieselhuber, Min Chen, Erin Hertlein, Swagata Goswami, Rajeswaran Mani, Yo-Ting Tsai, Frank Frissora, Raymond D. Devine, Ralf Bundschuh, Logan A. Walker, Larry Beaver, Gregory K. Behbehani, Kevan Zapolnik, Pearlly S. Yan, Eileen Y. Hu, Jessica Nunes, Alison Walker, Zhiliang Xie, Chad Bennett, Chi-Ling Chiang, John C. Byrd, Sumithira Vasu, X. Mo, Karilyn Larkin, Natarajan Muthusamy, Mitch A. Phelps, and Ann Marie Ventura
- Subjects
Cyclin-Dependent Kinase Inhibitor p21 ,Myeloid ,Cellular differentiation ,Immunology ,Cell fate determination ,Biology ,Biochemistry ,Proto-Oncogene Proteins c-myc ,Mice ,hemic and lymphatic diseases ,Cell Line, Tumor ,medicine ,Gene silencing ,Animals ,Humans ,Protein Phosphatase 2 ,Mice, Knockout ,Myeloid leukemia ,Cell Biology ,Hematology ,Cell cycle ,medicine.disease ,Leukemia ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,Cancer research ,Stem cell - Abstract
Dysregulated cellular differentiation is a hallmark of acute leukemogenesis. Phosphatases are widely suppressed in cancers but have not been traditionally associated with differentiation. In this study, we found that the silencing of protein phosphatase 2A (PP2A) directly blocks differentiation in acute myeloid leukemia (AML). Gene expression and mass cytometric profiling revealed that PP2A activation modulates cell cycle and transcriptional regulators that program terminal myeloid differentiation. Using a novel pharmacological agent, OSU-2S, in parallel with genetic approaches, we discovered that PP2A enforced c-Myc and p21 dependent terminal differentiation, proliferation arrest, and apoptosis in AML. Finally, we demonstrated that PP2A activation decreased leukemia-initiating stem cells, increased leukemic blast maturation, and improved overall survival in murine Tet2−/−Flt3ITD/WT and human cell-line derived xenograft AML models in vivo. Our findings identify the PP2A/c-Myc/p21 axis as a critical regulator of the differentiation/proliferation switch in AML that can be therapeutically targeted in malignancies with dysregulated maturation fate.
- Published
- 2021
80. High Early Death Rates, Treatment Resistance and Short Survival of Black Adolescent and Young Adults with Acute Myeloid Leukemia
- Author
-
Karilyn T. Larkin, Deedra Nicolet, Benjamin J. Kelly, Krzysztof Mrózek, Stephanie LaHaye, Katherine E. Miller, Saranga Wijeratne, Gregory Wheeler, Jessica Kohlschmidt, James S. Blachly, Alice S. Mims, Christopher J. Walker, Christopher C. Oakes, Shelley Orwick, Isaiah Boateng, Jill Buss, Adrienne Heyrosa, Helee Desai, Andrew J. Carroll, William Blum, Bayard L. Powell, Jonathan E. Kolitz, Joseph O. Moore, Robert J. Mayer, Richard A. Larson, Richard M. Stone, Electra D. Paskett, John C. Byrd, Elaine R. Mardis, and Ann-Kathrin Eisfeld
- Subjects
History ,Polymers and Plastics ,Business and International Management ,Industrial and Manufacturing Engineering - Published
- 2021
81. Outcomes for Patients with IDH-Mutated Acute Myeloid Leukemia Undergoing Allogeneic Hematopoietic Cell Transplantation
- Author
-
Steven L. McAfee, Joseph H. Antin, Amir T. Fathi, Corey Cutler, Rizwan Romee, Marlise R. Luskin, Shuli Li, Areej El-Jawahri, Yi Bin Chen, Zachariah DeFilipp, Ann-Kathrin Eisfeld, Dan Jones, Evan C. Chen, Mahasweta Gooptu, John Koreth, Alice S. Mims, Robert J. Soiffer, and Vincent T. Ho
- Subjects
Clinical trial ,Transplantation ,European LeukemiaNet ,medicine.medical_specialty ,Hematopoietic cell ,Maintenance therapy ,business.industry ,Patient age ,Family medicine ,Medicine ,Patient characteristics ,General hospital ,business - Abstract
Background: Disease relapse after hematopoietic cell transplantation (HCT) is a major cause of treatment failure for patients with acute myeloid leukemia (AML). Maintenance therapy following HCT for patients with targetable mutations such as mutated IDH1 or IDH2 may improve outcomes, and clinical trials evaluating this strategy are ongoing. However, current clinical outcomes of IDH1 - and IDH2 -mutated AML patients following HCT have not been well-described. Methods: In this multicenter retrospective analysis, 112 adult patients with either IDH1 - or IDH2 -mutated AML who underwent HCT were identified at Massachusetts General Hospital, Dana Farber Cancer Institute, and Ohio State University. Mutation testing was performed using next-generation sequencing panels. Patient characteristics were collected retrospectively, and their outcomes — including progression-free survival (PFS), overall survival (OS), relapse, and non-relapse mortality — were analyzed. Findings: The median patient age was 64·1 years. 78·5% of patients had intermediate- or adverse-risk disease by European LeukemiaNET criteria. Commonly detected co-mutations were DNMT3A (35·7%), NPM1 (33·1%), and FLT3-ITD (13·4%). The median follow-up was 27·5 months. For IDH1- mutated patients, the 1- and 2-year PFS was 75% and 58%, respectively, and the 1- and 2-year OS was 78% and 74%, respectively. For IDH2- mutated patients, the 1- and 2-year PFS was 64% and 58%, respectively, and the 1- and 2-year OS was 75% and 68%, respectively. Interpretation: Our analysis provides important benchmarks for analysis and interpretation of results emerging from ongoing clinical trials evaluating maintenance IDH1 and IDH2 inhibitor therapy for AML patients following HCT. Funding: This project has no funding source to report. Declaration of Interests: Dr. Eisfeld reports other from Karyopharm, personal fees from Vigeo, outside the submitted work. Dr. E Chen has nothing to disclose. Dr. Li has nothing to disclose. Dr. Luskin has nothing to disclose. Dr. Mims reports other from Jazz, other from Syndax, other from Abbvie, other from Kura oncology, personal fees from Agios, other from Novartis, outside the submitted work. Dr. Jones reports personal fees from Pharmacyclics LLC, outside the submitted work. Dr. Antin has nothing to disclose. Dr. Cutler reports other from Incyte, other from Kadmon, other from Jazz, other from Medsenic, other from Generon, other from Mesoblast, outside the submitted work. Dr. Koreth reports personal fees from Equilium, personal fees from Amgen, personal fees from Moderna Therapeutics, personal fees from Biolojic Design, personal fees from EMD Serono, other from Therakos, other from Cugene, grants from Miltenyi, grants from BMS, grants from Reneron, other from Clinigen, outside the submitted work. Dr. Gooptu has nothing to disclose. Dr. Romee has nothing to disclose. Dr. El-Jawahri has nothing to disclose. Dr. McAfee has nothing to disclose. Dr. Defilipp reports grants from Incyte, grants from Regimmune, personal fees from Syndax, outside the submitted work. Dr. Soiffer reports other from Kiadis, other from Gilead, other from Rheos, other from Cugene, other from Precision Bioscience, other from Mana Therapeutics, other from VOR Biopharma, other from Novartis, other from Juno, other from Celgene, other from Alexion, other from National Marrow Donor Program, outside the submitted work. Dr. YB Chen reports personal fees from Incyte, personal fees from Takeda, personal fees from Magenta, personal fees from Kiadis, other from Actinium, other from Equilium, other from Abbvie, outside the submitted work. Dr. Fathi reports grants from Takeda, personal fees from Boston Biomedical, personal fees from PTC Therapeutics, personal fees from Amphivena, personal fees from Astellas, personal fees from Daiichi Sankyo, personal fees from Novartis, grants and personal fees from Celgene/BMS, personal fees from Trovagene, personal fees from Forty Seven , personal fees from NewLink Genetics , personal fees from Pfizer, personal fees from Abbvie, grants and personal fees from Seattle Genetics , grants and personal fees from Agios, personal fees from Amgen, personal fees from Trillium, personal fees from Kura Oncology, personal fees from Blueprint, personal fees from Genentech, outside the submitted work. Ethics Approval Statement: This study was approved by the Institutional Review Boards at the Dana-Farber Harvard Cancer Center and Ohio State University Comprehensive Cancer Center
- Published
- 2021
82. Poor Survival and Differential Impact of Genetic Features of Black Patients with Acute Myeloid Leukemia
- Author
-
Bayard L. Powell, Bhavana Bhatnagar, Alice S. Mims, Isaiah Boateng, Deedra Nicolet, Richard Stone, James S. Blachly, Shelley Orwick, Jessica Kohlschmidt, Ann-Kathrin Eisfeld, Albert de la Chapelle, Sophia E. Maharry, Jonathan E. Kolitz, Ramiro Garzon, Andrew J. Carroll, John C. Byrd, James L. Fisher, Krzysztof Mrózek, Electra D. Paskett, Brian Giacopelli, Qiuhong Zhao, Christopher C. Oakes, and Christopher J. Walker
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Patient demographics ,MEDLINE ,Disease ,Article ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Surveillance, Epidemiology, and End Results ,Ethnicity ,Medicine ,Humans ,Socioeconomic status ,Differential impact ,Retrospective Studies ,business.industry ,Myeloid leukemia ,United States ,Clinical trial ,Black or African American ,Leukemia, Myeloid, Acute ,030104 developmental biology ,Treatment Outcome ,030220 oncology & carcinogenesis ,business - Abstract
Clinical outcome of patients with acute myeloid leukemia (AML) is associated with cytogenetic and molecular factors and patient demographics (e.g., age and race). We compared survival of 25,523 non-Hispanic Black and White adults with AML using Surveillance Epidemiology and End Results (SEER) Program data and performed mutational profiling of 1,339 patients with AML treated on frontline Alliance for Clinical Trials in Oncology (Alliance) protocols. Black patients had shorter survival than White patients, both in SEER and in the setting of Alliance clinical trials. The disparity was especially pronounced in Black patients Significance: We show that young Black patients have not benefited as much as White patients from recent progress in AML treatment in the United States. Our data suggest that both socioeconomic factors and differences in disease biology contribute to the survival disparity and need to be urgently addressed. See related commentary by Vyas, p. 540. This article is highlighted in the In This Issue feature, p. 521
- Published
- 2020
83. Precision medicine treatment in acute myeloid leukemia using prospective genomic profiling: feasibility and preliminary efficacy of the Beat AML Master Trial
- Author
-
Nyla A. Heerema, Ashley Owen Yocum, Timothy L. Chen, Eric Allan Severson, Leonard Rosenberg, Michael Boyiadzis, Martha Arellano, Brian J. Druker, Rebecca L. Olin, Tibor Kovacsovics, Robert H. Collins, Amy Burd, Abigail B. Shoben, Maria R. Baer, Olatoyosi Odenike, Sonja Marcus, Mark R. Litzow, Elie Traer, Michael W. Deininger, Uma Borate, Tara L. Lin, Alice S. Mims, Molly Rae Miller, William Blum, John C. Byrd, Gary J. Schiller, Vu H. Duong, Jo Anne Vergilio, Mona Stefanos, Prapti A. Patel, Christine Vietz, James M. Foran, Matthew C. Foster, Tim Brennan, Amy S. Ruppert, Wendy Stock, Brian Ball, Ross L. Levine, Alison Walker, and Eytan M. Stein
- Subjects
0301 basic medicine ,Oncology ,Male ,Myeloid ,Palliative care ,Medical and Health Sciences ,0302 clinical medicine ,hemic and lymphatic diseases ,Antineoplastic Combined Chemotherapy Protocols ,Monoclonal ,80 and over ,Precision Medicine ,Humanized ,Cancer ,Aged, 80 and over ,Pediatric ,Leukemia ,Tumor ,Cytarabine ,Myeloid leukemia ,General Medicine ,Genomics ,Hematology ,Middle Aged ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,Treatment Outcome ,030220 oncology & carcinogenesis ,6.1 Pharmaceuticals ,Female ,medicine.medical_specialty ,Childhood Leukemia ,Pediatric Cancer ,Clinical Trials and Supportive Activities ,Immunology ,Acute ,Antibodies, Monoclonal, Humanized ,Article ,General Biochemistry, Genetics and Molecular Biology ,Antibodies ,03 medical and health sciences ,Rare Diseases ,Clinical Research ,Internal medicine ,Biomarkers, Tumor ,medicine ,Genetics ,Humans ,neoplasms ,Survival analysis ,Aged ,business.industry ,Daunorubicin ,Evaluation of treatments and therapeutic interventions ,Precision medicine ,medicine.disease ,Survival Analysis ,Clinical trial ,030104 developmental biology ,Good Health and Well Being ,Mutation ,business ,Biomarkers - Abstract
Acute myeloid leukemia (AML) is the most common diagnosed leukemia. In older adults, AML confers an adverse outcome1,2. AML originates from a dominant mutation, then acquires collaborative transformative mutations leading to myeloid transformation and clinical/biological heterogeneity. Currently, AML treatment is initiated rapidly, precluding the ability to consider the mutational profile of a patient’s leukemia for treatment decisions. Untreated patients with AML ≥ 60 years were prospectively enrolled on the ongoing Beat AML trial (ClinicalTrials.gov NCT03013998 ), which aims to provide cytogenetic and mutational data within 7 days (d) from sample receipt and before treatment selection, followed by treatment assignment to a sub-study based on the dominant clone. A total of 487 patients with suspected AML were enrolled; 395 were eligible. Median age was 72 years (range 60–92 years; 38% ≥75 years); 374 patients (94.7%) had genetic and cytogenetic analysis completed within 7 d and were centrally assigned to a Beat AML sub-study; 224 (56.7%) were enrolled on a Beat AML sub-study. The remaining 171 patients elected standard of care (SOC) (103), investigational therapy (28) or palliative care (40); 9 died before treatment assignment. Demographic, laboratory and molecular characteristics were not significantly different between patients on the Beat AML sub-studies and those receiving SOC (induction with cytarabine + daunorubicin (7 + 3 or equivalent) or hypomethylation agent). Thirty-day mortality was less frequent and overall survival was significantly longer for patients enrolled on the Beat AML sub-studies versus those who elected SOC. A precision medicine therapy strategy in AML is feasible within 7 d, allowing patients and physicians to rapidly incorporate genomic data into treatment decisions without increasing early death or adversely impacting overall survival. Preliminary results from the Beat AML umbrella trial demonstrates the feasibility and efficacy of applying prospective genomic profiling for matching newly diagnosed patients with AML with targeted therapies.
- Published
- 2020
84. Comparison of fixed dose reduced-intensity conditioning with fludarabine and busulfan to PK-guided busulfan AUC (FluBu4K) in hematopoietic stem cell transplant for AML/MDS
- Author
-
Tyler Dickerson, Qiuhong Zhao, Patrick Elder, Jonathan E. Brammer, Alice S. Mims, Julianna Roddy, Hannah Choe, Ayman Saad, Brendan Rasor, Karilyn Larkin, Marcin Puto, Basem M. William, Sam Penza, Sarah A Wall, Samantha Jaglowski, and Sumithira Vasu
- Subjects
Oncology ,Adult ,Cancer Research ,medicine.medical_specialty ,Transplantation Conditioning ,Graft vs Host Disease ,Fixed dose ,Article ,03 medical and health sciences ,0302 clinical medicine ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Humans ,Transplantation, Homologous ,Busulfan ,Retrospective Studies ,business.industry ,Hematopoietic Stem Cell Transplantation ,Hematopoietic stem cell ,Retrospective cohort study ,Hematology ,humanities ,Fludarabine ,Regimen ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,Tolerability ,030220 oncology & carcinogenesis ,Reduced Intensity Conditioning ,Area Under Curve ,Neoplasm Recurrence, Local ,business ,Vidarabine ,030215 immunology ,medicine.drug - Abstract
A retrospective cohort study was conducted to assess differences in efficacy and tolerability between a busulfan AUC target of 16.4 mgxHr/L per day (FluBu4K) and a conventional RIC regimen (FluBu2). Adult patients with a diagnosis of AML or MDS who received fludarabine + busulfan conditioning with or without antithymocyte globulin between 2015–2018 were included. The primary outcome was relapse free survival. Overall, 74 patients received conditioning with either FluBu4K or FluBu2. At 18 months, relapse-free survival was not significantly different, at 63.9% with FluBu4k compared to 57.5% with FluBu2 (p=0.49). There was a statistically significant difference in the cumulative incidence of relapse at 18 months in favor of the FluBu4K regimen, at 12.0% vs 32.5% (p=0.047). The results of this study indicate that for select patients, there may be benefit in choosing targeted FluBu4K over FluBu2. Adverse effects other than mucositis were not significantly different.
- Published
- 2020
85. Incidence of venous thrombosis after peg-asparaginase in adolescent and young adults with acute lymphoblastic leukemia
- Author
-
Brynne, Underwood, Qiuhong, Zhao, Alison R, Walker, Alice S, Mims, Sumithira, Vasu, Meixiao, Long, Tamanna, Z Haque, Bradley W, Blaser, Nicole R, Grieselhuber, Sarah A, Wall, Gregory K, Behbehani, James S, Blachly, Karilyn, Larkin, John C, Byrd, Ramiro, Garzon, Tzu-Fei, Wang, and Bhavana, Bhatnagar
- Abstract
There are limited data describing incidence of symptomatic venous thromboembolism (VTE) in adolescent and young adult (AYA) acute lymphoblastic leukemia (ALL) patients receiving peg-asparaginase.Single-institution retrospective analysis of 44 AYA ALL patients treated with peg-asparaginase. Rates of VTE and proposed risk factors were assessed.18 patients (41%) had a symptomatic VTE following peg-asparaginase. The cumulative incidence rate was 25% (95% CI: 13-38%) within 30 days of the initial dose. Personal history of thrombosis was statistically significantly associated with an increased risk of VTE with HR of 2.73 (95% CI: 1.40-5.33, p = 0.003) after adjusting for gender.These data indicate a high rate of VTE in the AYA ALL population following treatment with peg-asparaginase.
- Published
- 2020
86. Clinical activity of axicabtagene ciloleucel in adult patients with Richter syndrome
- Author
-
Jennifer A. Woyach, Ayman Saad, Sarah A Wall, Polina Shindiapina, Alice S. Mims, Bhavana Bhatnagar, Jonathan E. Brammer, Samantha Jaglowski, Hannah K. Choe, Lynn O'Donnell, Karilyn Larkin, Sam Penza, Basem M. William, Seema A. Bhat, Yvonne A. Efebera, Adam Kittai, Kerry A. Rogers, John C. Byrd, Sumithira Vasu, Meixiao Long, and David A. Bond
- Subjects
Adult ,Pediatrics ,medicine.medical_specialty ,Richter syndrome ,Biological Products ,Adult patients ,business.industry ,Antigens, CD19 ,MEDLINE ,Hematology ,Immunotherapy, Adoptive ,Leukemia, Lymphocytic, Chronic, B-Cell ,Text mining ,medicine ,Commentary ,Humans ,Lymphoma, Large B-Cell, Diffuse ,business - Published
- 2020
87. Genomic analysis of cellular hierarchy in acute myeloid leukemia using ultrasensitive LC-FACSeq
- Author
-
Tzyy-Jye Doong, Nicole Grieselhuber, Nyla A. Heerema, Gerard Lozanski, Cecelia R. Miller, Karilyn Larkin, Tierney Kauffman, Rosa Lapalombella, Arletta Lozanski, John C. Byrd, Clara D. Bloomfield, Casey Cempre, Shelley Orwick, Bhavana Bhatnagar, Alice S. Mims, Lynne V. Abruzzo, Gregory K. Behbehani, Eileen Hu, Virginia M. Goettl, Alison Walker, Steven Sher, Pu Zhang, Caner Saygin, Jordan N. Skinner, James S. Blachly, Jadwiga Labanowska, and Deedra Nicolet
- Subjects
0301 basic medicine ,Adult ,Male ,Cancer Research ,Biology ,Somatic evolution in cancer ,Article ,Acute myeloid leukaemia ,Immunophenotyping ,Clonal Evolution ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,medicine ,Biomarkers, Tumor ,Humans ,Cancer genetics ,Aged ,Aged, 80 and over ,Myeloid leukemia ,Hematopoietic stem cell ,Correction ,Hematology ,Genomics ,Amplicon ,Middle Aged ,medicine.disease ,Flow Cytometry ,Hematopoietic Stem Cells ,Prognosis ,Haematopoiesis ,Leukemia ,Leukemia, Myeloid, Acute ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,DNA methylation ,Mutation ,Cancer research ,Neoplastic Stem Cells ,Female ,Stem cell ,Follow-Up Studies - Abstract
Hematopoiesis is hierarchical, and it has been postulated that acute myeloid leukemia (AML) is organized similarly with leukemia stem cells (LSCs) residing at the apex. Limited cells acquired by fluorescence activated cell sorting in tandem with targeted amplicon-based sequencing (LC-FACSeq) enables identification of mutations in small subpopulations of cells, such as LSCs. Leveraging this, we studied clonal compositions of immunophenotypically-defined compartments in AML through genomic and functional analyses at diagnosis, remission and relapse in 88 AML patients. Mutations involving DNA methylation pathways, transcription factors and spliceosomal machinery did not differ across compartments, while signaling pathway mutations were less frequent in putative LSCs. We also provide insights into TP53-mutated AML by demonstrating stepwise acquisition of mutations beginning from the preleukemic hematopoietic stem cell stage. In 10 analyzed cases, acquisition of additional mutations and del(17p) led to genetic and functional heterogeneity within the LSC pool with subclones harboring varying degrees of clonogenic potential. Finally, we use LC-FACSeq to track clonal evolution in serial samples, which can also be a powerful tool to direct targeted therapy against measurable residual disease. Therefore, studying clinically significant small subpopulations of cells can improve our understanding of AML biology and offers advantages over bulk sequencing to monitor the evolution of disease.
- Published
- 2020
88. A phase I study of lenalidomide plus chemotherapy with idarubicin and cytarabine in patients with relapsed or refractory acute myeloid leukemia and high-risk myelodysplastic syndrome
- Author
-
Bhavana Bhatnagar, Karilyn Larkin, Shelley Orwick, Katherine Walsh, Apollinaire Ngankeu, Shylaja Mani, John C. Byrd, Sumithira Vasu, William Blum, Alice S. Mims, Ramiro Garzon, James S. Blachly, Charles Thomas Gregory, Mitch A. Phelps, Caner Saygin, Rebecca B. Klisovic, Michael R. Grever, Guido Marcucci, and Alison Walker
- Subjects
Oncology ,Adult ,Male ,medicine.medical_specialty ,Adolescent ,medicine.medical_treatment ,Disease-Free Survival ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,hemic and lymphatic diseases ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Idarubicin ,Humans ,Survival rate ,Lenalidomide ,Research Articles ,Aged ,Chemotherapy ,business.industry ,Myelodysplastic syndromes ,Cytarabine ,Hematology ,Middle Aged ,medicine.disease ,Transplantation ,Survival Rate ,Leukemia, Myeloid, Acute ,030220 oncology & carcinogenesis ,Myelodysplastic Syndromes ,Female ,business ,Febrile neutropenia ,030215 immunology ,medicine.drug ,Research Article - Abstract
Patients with relapsed/refractory (R/R) acute myeloid leukemia (AML) have poor outcomes and hematopoietic cell transplantation (HCT) is the only curative treatment. New targeted therapies improved survival in select patients with specific mutations, however management of patients without these molecular alterations is an unmet need. We conducted a phase one study of lenalidomide in combination with cytarabine/idarubicin salvage chemotherapy in patients with R/R AML and high‐risk myelodysplastic syndromes. A total of 33 patients were enrolled in the study (30 AML, 3 MDS), and treated at three dose levels with 3 + 3 design. Dose‐limiting toxicity (DLT) was seen in eight patients, including four hematologic DLTs. The most commonly observed non‐hematologic serious adverse events were febrile neutropenia, rash, sepsis and renal injury. Dose level −1, consisting of 25 mg/d lenalidomide D1‐21, 1 g/m2 cytarabine D5‐8, and 8 mg/m2 idarubicin D5‐7 was determined to be the maximum tolerated dose. Note, 15/33 (45%) of patients were able to receive pre‐planned 21 days of lenalidomide. Overall, 18 patients achieved complete remission (CR) (n = 14) or CR with incomplete count recovery (CRi) (n = 4) with total CR/CRi rate of 56%. The 1‐year and 2‐year overall survival (OS) were 24% and 10%, respectively. Among responders, 10/18 underwent allogeneic HCT and had a 1‐year OS of 40%. There was no molecular pattern associated with response. These data demonstrate that the combination had clinical activity in R/R AML. This regimen should be further investigated for patients who relapsed after HCT, and as a bridge therapy to HCT. (ClinicalTrials.gov identifier: NCT01132586).
- Published
- 2020
89. Additional gene mutations may refine the 2017 European LeukemiaNet classification in adult patients with de novo acute myeloid leukemia aged <60 years
- Author
-
James S. Blachly, Ann-Kathrin Eisfeld, Richard Stone, Christopher J. Walker, Krzysztof Mrózek, Albert de la Chapelle, Bayard E. Powell, Andrew J. Carroll, Alice S. Mims, Deedra Nicolet, Jessica Kohlschmidt, Jonathan E. Kolitz, Dimitrios Papaioannou, Clara D. Bloomfield, and John C. Byrd
- Subjects
0301 basic medicine ,Oncology ,Adult ,Male ,Cancer Research ,NPM1 ,medicine.medical_specialty ,Adolescent ,Gene mutation ,Group B ,Article ,03 medical and health sciences ,European LeukemiaNet ,Young Adult ,0302 clinical medicine ,Risk Factors ,Internal medicine ,medicine ,Humans ,business.industry ,Myeloid leukemia ,Cancer ,Hematology ,Middle Aged ,medicine.disease ,Prognosis ,Clinical trial ,Leukemia ,Leukemia, Myeloid, Acute ,030104 developmental biology ,030220 oncology & carcinogenesis ,Mutation ,Female ,business ,Nucleophosmin - Abstract
The European LeukemiaNet (ELN) recommendations for diagnosis and management of acute myeloid leukemia (AML) have become an important tool to assess patients’ prognosis and guide treatment. We tested the prognostic impact of the 2017 ELN classification in a large cohort of 863 AML patients aged
- Published
- 2020
90. CD33-Targeted Therapies: Beating the Disease or Beaten to Death?
- Author
-
Meixiao Long, Veronika Bachanova, Alice S. Mims, Joseph Maakaron, and John Rogosheske
- Subjects
Myeloid ,Gemtuzumab ozogamicin ,CD33 ,Sialic Acid Binding Ig-like Lectin 3 ,030226 pharmacology & pharmacy ,Lintuzumab ,Immunotherapy, Adoptive ,03 medical and health sciences ,0302 clinical medicine ,hemic and lymphatic diseases ,medicine ,Pharmacology (medical) ,Progenitor cell ,Pharmacology ,Clinical Trials as Topic ,Receptors, Chimeric Antigen ,Dose-Response Relationship, Drug ,business.industry ,Vadastuximab Talirine ,Stem Cells ,Myeloid leukemia ,Gemtuzumab ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cancer research ,Stem cell ,business ,medicine.drug - Abstract
CD33 is a transmembrane protein that is found on cells of myeloid lineage. It is also intensely expressed on acute myeloid leukemia (AML) progenitor cells but not on normal stem cells. It internalizes on binding and dimerization, making it a specific and ideal target for AML therapeutics and drug delivery. Several targeted therapies have been tested and many are still currently in development. Gemtuzumab ozogamicin was the first and only CD33-directed antibody-drug conjugate to be US Food and Drug Administration approved for AML. Other targeted agents have not achieved such success. Promising new strategies include cellular therapy mechanisms and linker molecules. This is an exciting target that requires a considerable amount of precision to yield clinical benefit.
- Published
- 2020
91. Entospletinib in Combination with Induction Chemotherapy in Previously Untreated Acute Myeloid Leukemia: Response and Predictive Significance of
- Author
-
Alison R, Walker, John C, Byrd, James S, Blachly, Bhavana, Bhatnagar, Alice S, Mims, Shelley, Orwick, Tara L, Lin, Howland E, Crosswell, Danjie, Zhang, Mark D, Minden, Veerendra, Munugalavadla, Lauren, Long, Jinfeng, Liu, Yang, Pan, Thomas, Oellerich, Hubert, Serve, Arati V, Rao, and William G, Blum
- Subjects
Adult ,Homeodomain Proteins ,Male ,Indazoles ,Daunorubicin ,Cytarabine ,Induction Chemotherapy ,Middle Aged ,Gene Expression Regulation, Neoplastic ,Leukemia, Myeloid, Acute ,Pyrazines ,Humans ,Syk Kinase ,Female ,Myeloid Ecotropic Viral Integration Site 1 Protein - Abstract
Spleen tyrosine kinase (SYK) signaling is a proposed target in acute myeloid leukemia (AML). Sensitivity to SYK inhibition has been linked toThis was an international multicenter phase Ib/II study, entospletinib dose escalation (standard 3+3 design between 200 and 400 mg twice daily) + 7+3 (cytarabine + daunorubicin) in phase Ib and entospletinib dose expansion (400 mg twice daily) + 7+3 in phase II.Fifty-three patients (Entospletinib with intensive chemotherapy was well-tolerated in patients with AML. Improved survival was observed in patients with
- Published
- 2020
92. Comparison of clinical and molecular characteristics of patients with acute myeloid leukemia and either TP73 or TP53 mutations
- Author
-
Shelley Orwick, Jessica Kohlschmidt, Richard Stone, Dimitrios Papaioannou, James S. Blachly, Clara D. Bloomfield, Bayard L. Powell, Deepa Sampath, Alice S. Mims, Krzysztof Mrόzek, Deedra Nicolet, Jonathan E. Kolitz, John C. Byrd, and Ann-Kathrin Eisfeld
- Subjects
Male ,Cancer Research ,Genotype ,MEDLINE ,Kaplan-Meier Estimate ,Tp53 mutation ,Article ,Text mining ,Medicine ,Humans ,Genetic Predisposition to Disease ,Alleles ,Genetic Association Studies ,business.industry ,Myeloid leukemia ,Tumor Protein p73 ,Hematology ,Prognosis ,Leukemia, Myeloid, Acute ,Phenotype ,Oncology ,Mutation ,Cancer research ,Female ,Tumor Suppressor Protein p53 ,business - Published
- 2020
93. Molecular mechanisms mediating relapse following ivosidenib monotherapy in IDH1-mutant relapsed or refractory AML
- Author
-
Sung Choe, Zenon D. Konteatis, Chris Bowden, Brandon Nicolay, Courtney D. DiNardo, Bin Wu, Jessica K. Altman, Alice S. Mims, Lenny Dang, Scott A. Biller, Guowen Liu, Daniel A. Pollyea, Parham Nejad, Hongfang Wang, Eyal C. Attar, Richard Stone, Stéphane de Botton, Wei Liu, Vickie Zhang, Eytan M. Stein, Kevin Marks, Justin M. Watts, Gail J. Roboz, Meredith Goldwasser, Lynn Quek, Amir T. Fathi, Hua Liu, Hagop M. Kantarjian, and Martin S. Tallman
- Subjects
0301 basic medicine ,Myeloid ,IDH1 ,Combination therapy ,Pyridines ,Glycine ,IDH2 ,Receptor tyrosine kinase ,03 medical and health sciences ,0302 clinical medicine ,Recurrence ,medicine ,Humans ,Myeloid Neoplasia ,biology ,business.industry ,Myeloid leukemia ,Hematology ,medicine.disease ,Isocitrate Dehydrogenase ,Leukemia ,Leukemia, Myeloid, Acute ,030104 developmental biology ,medicine.anatomical_structure ,Isocitrate dehydrogenase ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,business - Abstract
Isocitrate dehydrogenase (IDH) 1 and 2 mutations result in overproduction of D-2-hydroxyglutarate (2-HG) and impaired cellular differentiation. Ivosidenib, a targeted mutant IDH1 (mIDH1) enzyme inhibitor, can restore normal differentiation and results in clinical responses in a subset of patients with mIDH1 relapsed/refractory (R/R) acute myeloid leukemia (AML). We explored mechanisms of ivosidenib resistance in 174 patients with confirmed mIDH1 R/R AML from a phase 1 trial. Receptor tyrosine kinase (RTK) pathway mutations were associated with primary resistance to ivosidenib. Multiple mechanisms contributed to acquired resistance, particularly outgrowth of RTK pathway mutations and 2-HG–restoring mutations (second-site IDH1 mutations, IDH2 mutations). Observation of multiple concurrent mechanisms in individual patients underscores the complex biology of resistance and has important implications for rational combination therapy design. This trial was registered at www.clinicaltrials.gov as #NCT02074839
- Published
- 2020
94. Complement-mediated thrombotic microangiopathy as a link between endothelial damage and steroid-refractory GVHD
- Author
-
Alice S. Mims, Basem M. William, Martha Yearsley, Stella M. Davies, Jonathan E. Brammer, Steven M. Devine, Hannah Choe, Parvathi Ranganathan, Samantha Jaglowski, Yvonne A. Efebera, Spero R. Cataland, Haiwa Wu, Luke Blower, Sam Penza, Akwasi Agyeman, Qiuhong Zhao, Shangbin Yang, Matthew Bostic, Sumithira Vasu, and Sarah A Wall
- Subjects
Adult ,Male ,medicine.medical_specialty ,Thrombotic microangiopathy ,Graft vs Host Disease ,Gastroenterology ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,immune system diseases ,Internal medicine ,parasitic diseases ,medicine ,Humans ,Endothelium ,Young adult ,Risk factor ,Aged ,Transplantation ,Thrombotic Microangiopathies ,business.industry ,Hazard ratio ,Histology ,Hematology ,Middle Aged ,medicine.disease ,Pathophysiology ,Complement system ,surgical procedures, operative ,030220 oncology & carcinogenesis ,population characteristics ,Female ,Complication ,business ,human activities ,030215 immunology - Abstract
Transplant-associated thrombotic microangiopathy (TA-TMA), a complication of hematopoietic cell transplant (HCT), is associated with significant morbidity and mortality. The pathophysiology and overlap of TA-TMA with other posttransplant complications such as graft-versus-host disease (GVHD) is poorly understood. We retrospectively identified cases of TA-TMA among patients with grade 3/4 gastrointestinal (GI) GVHD, reviewed intestinal biopsy specimens, and performed correlative testing of biomarkers associated with TA-TMA. TA-TMA was more common in patients with steroid-refractory GVHD compared with steroid-responsive GVHD (79.3% vs 42.1%; P = .001). Among patients surviving 100 days post-HCT, 1-year survival from day 100 was significantly better for patients who had not developed TA-TMA in the first 100 days (69.5% vs 36.7%; P < .001). Only 1 of 7 proposed TA-TMA histology criteria (mucosal hemorrhage) differed significantly based on GVHD steroid response. In multivariable modeling, steroid-refractory GVHD was a risk factor for development of TA-TMA (hazard ratio, 3.09; 95% confidence interval, 1.68-5.67; P < .001). There were no differences in complement activation at GVHD onset; however, 2 to 6 weeks later, patients with TA-TMA had higher levels of BBPlus and C5b-9, markers of alternative and terminal pathway activation (BBPlus: median, 600 vs 209.3 ng/mL; P = .0045) (C5b-9: median, 425.9 vs 258.4 ng/mL; P = .029). TA-TMA is associated with poor overall survival (OS) following HCT and may be detected early by histologic findings and may be differentiated from GVHD by measurement of alternative and terminal complement pathway activation. It is unknown whether treatment of TA-TMA will improve survival in steroid-refractory GVHD.
- Published
- 2018
95. Durable Remissions with Ivosidenib inIDH1-Mutated Relapsed or Refractory AML
- Author
-
Courtney D. DiNardo, Hua Liu, Gail J. Roboz, Hongfang Wang, Martha Arellano, Samuel V. Agresta, Vickie Zhang, Ronan T. Swords, Martin S. Tallman, Geoffrey L. Uy, William B. Donnellan, David Dai, Stephanie M. Kapsalis, Christophe Willekens, Jessica K. Altman, James M. Foran, A. Pigneux, Meredith Goldwasser, Sung Choe, Gabriel N. Mannis, Eyal C. Attar, James L. Slack, Amir T. Fathi, Harry P. Erba, Bin Wu, Bin Fan, Robert K. Stuart, S. de Botton, Hagop M. Kantarjian, Richard Stone, Mikkael A. Sekeres, Katharine E. Yen, Hua Yang, Elie Traer, Eytan M. Stein, Robert H. Collins, Alice S. Mims, Daniel A. Pollyea, Anthony S. Stein, and Gabrielle T. Prince
- Subjects
Male ,0301 basic medicine ,Myeloid ,Pyridines ,Administration, Oral ,Cell Count ,Gastroenterology ,Hemoglobins ,0302 clinical medicine ,Recurrence ,hemic and lymphatic diseases ,Leukocytosis ,Enzyme Inhibitors ,Aged, 80 and over ,education.field_of_study ,Remission Induction ,Myeloid leukemia ,General Medicine ,Middle Aged ,Isocitrate Dehydrogenase ,Survival Rate ,Leukemia, Myeloid, Acute ,Leukemia ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Female ,medicine.symptom ,Adult ,medicine.medical_specialty ,Adolescent ,Population ,Glycine ,Enasidenib ,Young Adult ,03 medical and health sciences ,Refractory ,Internal medicine ,medicine ,Humans ,education ,Survival rate ,Aged ,Dose-Response Relationship, Drug ,business.industry ,medicine.disease ,030104 developmental biology ,Drug Resistance, Neoplasm ,Mutation ,business ,Follow-Up Studies - Abstract
Mutations in the gene encoding isocitrate dehydrogenase 1 ( IDH1) occur in 6 to 10% of patients with acute myeloid leukemia (AML). Ivosidenib (AG-120) is an oral, targeted, small-molecule inhibitor of mutant IDH1.We conducted a phase 1 dose-escalation and dose-expansion study of ivosidenib monotherapy in IDH1-mutated AML. Safety and efficacy were assessed in all treated patients. The primary efficacy population included patients with relapsed or refractory AML receiving 500 mg of ivosidenib daily with at least 6 months of follow-up.Overall, 258 patients received ivosidenib and had safety outcomes assessed. Among patients with relapsed or refractory AML (179 patients), treatment-related adverse events of grade 3 or higher that occurred in at least 3 patients were prolongation of the QT interval (in 7.8% of the patients), the IDH differentiation syndrome (in 3.9%), anemia (in 2.2%), thrombocytopenia or a decrease in the platelet count (in 3.4%), and leukocytosis (in 1.7%). In the primary efficacy population (125 patients), the rate of complete remission or complete remission with partial hematologic recovery was 30.4% (95% confidence interval [CI], 22.5 to 39.3), the rate of complete remission was 21.6% (95% CI, 14.7 to 29.8), and the overall response rate was 41.6% (95% CI, 32.9 to 50.8). The median durations of these responses were 8.2 months (95% CI, 5.5 to 12.0), 9.3 months (95% CI, 5.6 to 18.3), and 6.5 months (95% CI, 4.6 to 9.3), respectively. Transfusion independence was attained in 29 of 84 patients (35%), and patients who had a response had fewer infections and febrile neutropenia episodes than those who did not have a response. Among 34 patients who had a complete remission or complete remission with partial hematologic recovery, 7 (21%) had no residual detectable IDH1 mutations on digital polymerase-chain-reaction assay. No preexisting co-occurring single gene mutation predicted clinical response or resistance to treatment.In patients with advanced IDH1-mutated relapsed or refractory AML, ivosidenib at a dose of 500 mg daily was associated with a low frequency of grade 3 or higher treatment-related adverse events and with transfusion independence, durable remissions, and molecular remissions in some patients with complete remission. (Funded by Agios Pharmaceuticals; ClinicalTrials.gov number, NCT02074839 .).
- Published
- 2018
96. NF1 mutations are recurrent in adult acute myeloid leukemia and confer poor outcome
- Author
-
Alice S. Mims, Jessica Kohlschmidt, Sophia E. Maharry, Jonathan E. Kolitz, Christopher J. Walker, Shelley Orwick, Eunice S. Wang, Bayard L. Powell, James S. Blachly, Ann-Kathrin Eisfeld, Richard Stone, John C. Byrd, Deedra Nicolet, Albert de la Chapelle, Krzysztof Mrózek, Andrew J. Carroll, and Clara D. Bloomfield
- Subjects
Male ,Adult ,0301 basic medicine ,Oncology ,congenital, hereditary, and neonatal diseases and abnormalities ,Cancer Research ,medicine.medical_specialty ,Myeloid ,Adolescent ,Nonsense mutation ,Article ,Frameshift mutation ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Risk Factors ,Internal medicine ,medicine ,Humans ,Missense mutation ,neoplasms ,Aged ,Aged, 80 and over ,Neurofibromin 1 ,business.industry ,Remission Induction ,Myeloid leukemia ,Adult Acute Myeloid Leukemia ,Hematology ,Middle Aged ,Prognosis ,medicine.disease ,nervous system diseases ,3. Good health ,Leukemia, Myeloid, Acute ,Leukemia ,030104 developmental biology ,medicine.anatomical_structure ,Targeted Mutation ,030220 oncology & carcinogenesis ,Mutation ,Female ,business - Abstract
Targeted mutation assessment of 81 genes in 1021 adults with de novo acute myeloid leukemia (AML) identified recurrent mutations in the neurofibromin 1 (NF1) gene in 52 (5.1%) patients, including 36 (5.2%) younger and 16 (4.8%) older patients, which suggests NF1 belongs to the 20 most frequently mutated genes in adult AML. NF1 mutations were found throughout the gene, and comprised missense, frameshift, and nonsense mutations. One mutation hotspot, at amino acid threonine 676 (Thr676), was found in 27% of AML patients with NF1 mutations. NF1-mutated patients belonged more often to the adverse European LeukemiaNet (ELN) risk category than NF1 wild-type patients. Among patients aged
- Published
- 2018
97. The interleukin-3 receptor CD123 targeted SL-401 mediates potent cytotoxic activity against CD34+CD123+ cells from acute myeloid leukemia/myelodysplastic syndrome patients and healthy donors
- Author
-
Alice S. Mims, Amber Gordon, Gerard Lozanski, Bhavani Gopalakrishnan, David M. Lucas, Rajeswaran Mani, Natarajan Muthusamy, William Blum, Alison Walker, Rahul Ramaswamy, Xiaokui Mo, Minh Tran, John C. Byrd, Sumithira Vasu, Adrienne M. Dorrance, Swagata Goswami, Christopher L. Brooks, Donna Bucci, and Ronni Wasmuth
- Subjects
0301 basic medicine ,Myeloid ,business.industry ,CD34 ,Myeloid leukemia ,Hematology ,medicine.disease ,3. Good health ,03 medical and health sciences ,Leukemia ,Haematopoiesis ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,hemic and lymphatic diseases ,030220 oncology & carcinogenesis ,Cord blood ,Cancer research ,Medicine ,Progenitor cell ,Stem cell ,business - Abstract
Diseases with clonal hematopoiesis such as myelodysplastic syndrome and acute myeloid leukemia have high rates of relapse. Only a small subset of acute myeloid leukemia patients are cured with chemotherapy alone. Relapse in these diseases occurs at least in part due to the failure to eradicate leukemic stem cells or hematopoietic stem cells in myelodysplastic syndrome. CD123, the alpha chain of the interleukin-3 receptor heterodimer, is expressed on the majority of leukemic stem cells and myelodysplastic syndrome hematopoietic stem cells and in 80% of acute myeloid leukemia. Here, we report indiscriminate killing of CD123+ normal and acute myeloid leukemia / myelodysplastic syndrome cells by SL-401, a diphtheria toxin interleukin-3 fusion protein. SL-401 induced cytotoxicity of CD123+ primary cells/blasts from acute myeloid leukemia and myelodysplastic syndrome patients but not CD123- lymphoid cells. Importantly, SL-401 was highly active even in cells expressing low levels of CD123, with minimal effect on modulation of the CD123 target in acute myeloid leukemia. SL-401 significantly prolonged survival of leukemic mice in acute myeloid leukemia patient-derived xenograft mouse models. In addition to primary samples, studies on normal cord blood and healthy marrow show that SL-401 has activity against normal hematopoietic progenitors. These findings indicate potential use of SL-401 as a "bridge-to-transplant" before allogeneic hematopoietic cell transplantation in acute myeloid leukemia / myelodysplastic syndrome patients.
- Published
- 2018
98. Incidence and survival of hematological cancers among adults ages ≥75 years
- Author
-
Julie A. Stephens, Alice S. Mims, Jessica L. Krok-Schoen, James L. Fisher, Sabarish Ayyappan, Jennifer A. Woyach, and Ashley E. Rosko
- Subjects
Cancer Research ,medicine.medical_specialty ,Chronic lymphocytic leukemia ,Population ,elderly ,03 medical and health sciences ,0302 clinical medicine ,hemic and lymphatic diseases ,Internal medicine ,Acute lymphocytic leukemia ,Epidemiology ,medicine ,Radiology, Nuclear Medicine and imaging ,education ,RC254-282 ,older adults ,Original Research ,Cancer ,education.field_of_study ,Acute leukemia ,Relative survival ,business.industry ,Incidence (epidemiology) ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Myeloid leukemia ,hematologic malignancies ,medicine.disease ,Oncology ,030220 oncology & carcinogenesis ,business ,Cancer Prevention ,SEER Program ,030215 immunology - Abstract
Evaluating population‐based data of hematologic malignancies (HMs) in older adults provides prognostic information for this growing demographic. Incidence rates and one‐ and five‐year relative survival rates were examined for specific HMs among adults ages ≥75 years using data from the Surveillance, Epidemiology and End Results (SEER) Program. Hematologic malignancy cases (Hodgkin lymphoma (HL), non‐Hodgkin lymphoma (NHL), multiple myeloma (MM), acute lymphocytic leukemia (ALL), chronic lymphocytic leukemia (CLL), acute myeloid leukemia (AML), and chronic myeloid leukemia (CML)) were reported to one of 18 SEER registries. Recent average annual (2010–2014) incidence rates and incidence trends from 1973 to 2014 were examined for cases ages ≥75 years. One‐ and five‐year relative cancer survival rates were examined for adults ages ≥75 years diagnosed 2007–2013, with follow‐up into 2014. From 1973 to 2014, incidence rates increased for NHL, MM, and AML, decreased for HL, and remained relatively stable for ALL, CLL, and CML among adults ages ≥75 years. The highest one‐ and five‐year relative survival rates were observed among adults with CLL ages 75–84 years (1 year: 91.8% (95% CI = 91.8–90.8)) and 5 years: 76.5% (95% CI = 74.2–78.6)). The lowest one‐ and five‐year survival rates were observed among adults with AML ages 75–84 (1 year: 18.2% (95% CI = 74.2–78.6) and 5 years: 2.7% (95% CI = 2.0–3.6)). Survival for older adults ages ≥75 years with HMs is poor, particularly for acute leukemia. Understanding the heterogeneity in HM outcomes among older patients may help clinicians better address the hematological cancer burden and mortality in the aging population.
- Published
- 2018
99. A novel regimen for relapsed/refractory adult acute myeloid leukemia using a KMT2A partial tandem duplication targeted therapy: results of phase 1 study NCI 8485
- Author
-
Alison Walker, Katherine Walsh, Clara D. Bloomfield, William Blum, James S. Blachly, Susan P. Whitman, Anjali Mishra, Gerard Lozanski, John C. Byrd, Sumithira Vasu, Rebecca B. Klisovic, Michael A. Caligiuri, Guido Marcucci, Richard Piekarz, Steven M. Devine, Shelley Orwick, Dan Jones, Michael R. Grever, Alice S. Mims, Ramiro Garzon, and Nyla A. Heerema
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Myeloid ,Decitabine ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Medicine ,Vorinostat ,biology ,business.industry ,Myeloid leukemia ,Adult Acute Myeloid Leukemia ,Hematology ,medicine.disease ,3. Good health ,Leukemia ,030104 developmental biology ,medicine.anatomical_structure ,KMT2A ,030220 oncology & carcinogenesis ,Cytarabine ,biology.protein ,business ,medicine.drug - Abstract
KMT2A partial tandem duplication occurs in approximately 5-10% of patients with acute myeloid leukemia and is associated with adverse prognosis. KMT2A wild type is epigenetically silenced in KMT2A partial tandem duplication; re-expression can be induced with DNA methyltransferase and/or histone deacetylase inhibitors in vitro, sensitizing myeloid blasts to chemotherapy. We hypothesized that epigenetic silencing of KMT2A wildtype contributes to KMT2A partial tandem duplication-associated leukemogenesis and pharmacologic re-expression activates apoptotic mechanisms important for chemoresponse. We developed a regimen for this unique molecular subset, but due to relatively low frequency of KMT2A partial tandem duplication, this dose finding study was conducted in relapsed/refractory disease regardless of molecular subtype. Seventeen adults (< age 60) with relapsed/refractory acute myeloid leukemia were treated on study. Patients received decitabine 20 milligrams/meter2 daily on days 1-10 and vorinostat 400 milligrams daily on days 5-10. Cytarabine was dose-escalated from 1.5 grams/meter2 every 12 hours to 3 grams/meter2 every 12 hours on days 12, 14 and 16. Two patients experienced dose limiting toxicities at dose level 1 due to prolonged myelosuppression. However, as both patients achieved complete remission after Day 42, the protocol was amended to adjust the definition of hematologic dose limiting toxicity. No further dose limiting toxicities were found. Six of 17 patients achieved complete remission including 2 of 4 patients with KMT2A partial tandem duplication. Combination therapy with decitabine, vorinostat and cytarabine was tolerated in younger relapsed/refractory acute myeloid leukemia and should be explored further focusing on the KMT2A partial tandem duplication subset. (clinicaltrials.gov identifier 01130506).
- Published
- 2018
100. Combination of the low anticoagulant heparin CX-01 with chemotherapy for the treatment of acute myeloid leukemia
- Author
-
Peter Ahorukomeye, Martha Glenn, Paul J. Shami, Tibor Kovacsovics, Stephen Marcus, Linda M Bavisotto, Jeremy Pantin, Thomas P. Kennedy, Gerardo Gutierrez-Sanchez, Alice S. Mims, Michael W. Deininger, Kenneth M. Boucher, Narayanam V. Rao, Mohamed E. Salama, and Ken M. Kosak
- Subjects
Adult ,Male ,0301 basic medicine ,medicine.medical_specialty ,Myeloid ,Clinical Trials and Observations ,medicine.medical_treatment ,Antineoplastic Agents ,Pilot Projects ,Gastroenterology ,Young Adult ,03 medical and health sciences ,Myelogenous ,Internal medicine ,medicine ,Humans ,Idarubicin ,Aged ,Chemotherapy ,Heparin ,business.industry ,Anticoagulants ,Myeloid leukemia ,Hematology ,Middle Aged ,medicine.disease ,Survival Analysis ,Chemotherapy regimen ,Leukemia, Myeloid, Acute ,Leukemia ,Treatment Outcome ,030104 developmental biology ,medicine.anatomical_structure ,Cytarabine ,Drug Therapy, Combination ,Female ,business ,medicine.drug - Abstract
Relapses in acute myelogenous leukemia (AML) are a result of quiescent leukemic stem cells (LSCs) in marrow stromal niches, where they resist chemotherapy. LSCs employ CXCL12/CXCR4 to home toward protective marrow niches. Heparin disrupts CXCL12-mediated sequestration of cells in the marrow. CX-01 is a low-anticoagulant heparin derivative. In this pilot study, we combined CX-01 with chemotherapy for the treatment of AML. Induction consisted of cytarabine and idarubicin (7 + 3) with CX-01. Twelve patients were enrolled (median age, 56 years; 3 women). Three, 5, and 4 patients had good-, intermediate-, and poor-risk disease, respectively. Day 14 bone marrows were available on 11 patients and were aplastic in all without detectable leukemia. Eleven patients (92%) had morphologic complete remission after 1 induction (CR1). Eight patients were alive at a median follow-up of 24 months (4 patients in CR1). Three patients received an allogeneic stem cell transplant in CR1. Median disease-free survival was 14.8 months. Median overall survival was not attained at the maximum follow-up time of 29.4 months. No CX-01-associated serious adverse events occurred. Median day to an untransfused platelet count of at least 20 × 109/L was 21. CX-01 is well tolerated when combined with intensive therapy for AML and appears associated with enhanced count recovery and treatment efficacy.
- Published
- 2018
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.