202 results on '"Roboz GJ"'
Search Results
2. Oral azacitidine prolongs survival of patients with AML in remission independently of measurable residual disease status
- Author
-
Roboz, GJ, Ravandi, F, Wei, AH, Dombret, H, Thol, F, Voso, MT, Schuh, AC, Porkka, K, La Torre, I, Skikne, B, Zhong, J, Beach, CL, Risueno, A, Menezes, DL, Ossenkoppele, G, Dohner, H, Roboz, GJ, Ravandi, F, Wei, AH, Dombret, H, Thol, F, Voso, MT, Schuh, AC, Porkka, K, La Torre, I, Skikne, B, Zhong, J, Beach, CL, Risueno, A, Menezes, DL, Ossenkoppele, G, and Dohner, H
- Abstract
Measurable residual disease (MRD) in patients with acute myeloid leukemia (AML) in remission after intensive chemotherapy is predictive of early relapse and poor survival. Postremission maintenance therapy that prolongs MRD negativity or converts MRD+ patients to MRD- status may delay or prevent relapse and improve overall survival (OS). In the phase 3 QUAZAR AML-001 trial, oral azacitidine (oral-AZA; formerly CC-486), a hypomethylating agent, significantly prolonged OS and relapse-free survival (RFS) compared with placebo in patients aged ≥55 years with AML in first remission after intensive chemotherapy who were not candidates for hematopoietic stem cell transplantation. In this trial, MRD (≥0.1% leukemic cells in bone marrow) was assessed by multiparameter flow cytometry in serial samples collected at baseline and on day 1 of every 3 cycles. As expected, baseline MRD status was significantly associated with both OS and RFS. Multivariate analyses showed oral-AZA significantly improved OS and RFS vs placebo independent of baseline MRD status. Oral-AZA treatment also extended the duration of MRD negativity by 6 months vs placebo and resulted in a higher rate of conversion from MRD+ at baseline to MRD- during treatment: 37% vs 19%, respectively. In the oral-AZA arm, 24% of MRD responders achieved MRD negativity >6 months after treatment initiation. Although presence or absence of MRD was a strong prognostic indicator of OS and RFS, there were added survival benefits with oral-AZA maintenance therapy compared with placebo, independent of patients' MRD status at baseline. Registered at clinicaltrials.gov as #NCT01757535.
- Published
- 2022
3. Phase I trials of the lysine-specific demethylase 1 inhibitor, GSK2879552, as mono- and combination-therapy in relapsed/refractory acute myeloid leukemia or high-risk myelodysplastic syndromes
- Author
-
Roboz, GJ, Yee, K, Verma, A, Borthakur, G, de la Fuente Burguera, A, Sanz, G, Mohammad, HP, Kruger, RG, Karpinich, NO, Ferron-Brady, G, Acusta, A, Del Buono, H, Collingwood, T, Ballas, M, Dhar, A, Wei, AH, Roboz, GJ, Yee, K, Verma, A, Borthakur, G, de la Fuente Burguera, A, Sanz, G, Mohammad, HP, Kruger, RG, Karpinich, NO, Ferron-Brady, G, Acusta, A, Del Buono, H, Collingwood, T, Ballas, M, Dhar, A, and Wei, AH
- Published
- 2022
4. Phase I trials of the lysine-specific demethylase 1 inhibitor, GSK2879552, as mono- and combination-therapy in relapsed/refractory acute myeloid leukemia or high-risk myelodysplastic syndromes
- Author
-
Roboz GJ, Yee K, Verma A, Borthakur G, de la Fuente Burguera A, Sanz G, Mohammad HP, Kruger RG, Karpinich NO, Ferron-Brady G, Acusta A, Del Buono H, Collingwood T, Ballas M, Dhar A, and Wei AH
- Published
- 2022
5. 2021 update measurable residual disease in acute myeloid leukemia: European leukemia net working party consensus document
- Author
-
Heuser, M, Freeman, Sd, Ossenkoppele, Gj, Buccisano, F, Hourigan, Cs, Ngai, Ll, Tettero, Jm, Bachas, C, Baer, C, Béné, Mc, Buecklein, V, Czyz, A, Denys, B, Dillon, R, Feuring-Buske, M, Guzman, Ml, Haferlach, T, Han, L, Herzig, Jk, Jorgensen, Jl, Kern, W, Konopleva, My, Lacombe, F, Libura, M, Majchrzak, A, Maurillo, L, Ofran, Y, Philippé, J, Plesa, A, Preudhomme, C, Ravandi, F, Roumier, C, Subklewe, M, Thol, F, van de Loosdrecht, Aa, van der Reijden, Ba, Venditti, A, Wierzbowska, A, Valk, Pjm, Wood, Bl, Walter, Rb, Thiede, C, Döhner, K, Roboz, Gj, and Cloos, J
- Subjects
Settore MED/15 - Published
- 2021
6. Oral azacitidine preserves favorable level of fatigue and health-related quality of life for patients with acute myeloid leukemia in remission: results from the phase III, placebo-controlled QUAZAR AML-001 trial
- Author
-
Roboz, GJ, Doehner, H, Pocock, C, Dombret, H, Ravandi, F, Jang, JH, Selleslag, D, Mayer, JR, Martens, UM, Liesveld, J, Bernal, T, Wang, MC, Yu, P, Shi, L, Guo, S, La Torre, I, Skikne, B, Dong, Q, Braverman, J, Nehme, SA, Beach, CL, Wei, AH, Roboz, GJ, Doehner, H, Pocock, C, Dombret, H, Ravandi, F, Jang, JH, Selleslag, D, Mayer, JR, Martens, UM, Liesveld, J, Bernal, T, Wang, MC, Yu, P, Shi, L, Guo, S, La Torre, I, Skikne, B, Dong, Q, Braverman, J, Nehme, SA, Beach, CL, and Wei, AH
- Abstract
Not available.
- Published
- 2021
7. Management of adverse events in patients with acute myeloid leukemia in remission receiving oral azacitidine: experience from the phase 3 randomized QUAZAR AML-001 trial
- Author
-
Ravandi, F, Roboz, GJ, Wei, AH, Dohner, H, Pocock, C, Selleslag, D, Montesinos, P, Sayar, H, Musso, M, Figuera-Alvarez, A, Safah, H, Tse, W, Sohn, SK, Hiwase, D, Chevassut, T, Pierdomenico, F, La Torre, I, Skikne, B, Bailey, R, Zhong, J, Beach, CL, Dombret, H, Ravandi, F, Roboz, GJ, Wei, AH, Dohner, H, Pocock, C, Selleslag, D, Montesinos, P, Sayar, H, Musso, M, Figuera-Alvarez, A, Safah, H, Tse, W, Sohn, SK, Hiwase, D, Chevassut, T, Pierdomenico, F, La Torre, I, Skikne, B, Bailey, R, Zhong, J, Beach, CL, and Dombret, H
- Abstract
BACKGROUND: Most older patients with acute myeloid leukemia (AML) who attain morphologic remission with intensive chemotherapy (IC) will eventually relapse and post-relapse prognosis is dismal. In the pivotal QUAZAR AML-001 trial, oral azacitidine maintenance therapy significantly prolonged overall survival by 9.9 months (P < 0.001) and relapse-free survival by 5.3 months (P < 0.001) compared with placebo in patients with AML in first remission after IC who were not candidates for transplant. Currently, the QUAZAR AML-001 trial provides the most comprehensive safety information associated with oral azacitidine maintenance therapy. Reviewed here are common adverse events (AEs) during oral azacitidine treatment in QUAZAR AML-001, and practical recommendations for AE management based on guidance from international cancer consortiums, regulatory authorities, and the authors' clinical experience treating patients in the trial. METHODS: QUAZAR AML-001 is an international, placebo-controlled randomized phase 3 study. Patients aged ≥ 55 years with AML and intermediate- or poor-risk cytogenetics at diagnosis, who had attained first complete remission (CR) or CR with incomplete blood count recovery (CRi) within 4 months before study entry, were randomized 1:1 to receive oral azacitidine 300 mg or placebo once-daily for 14 days in repeated 28-day cycles. Safety was assessed in all patients who received ≥ 1 dose of study drug. RESULTS: A total of 469 patients received oral azacitidine (n = 236) or placebo (n = 233). Median age was 68 years. Patients received a median of 12 (range 1-80) oral azacitidine treatment cycles or 6 (1-73) placebo cycles. Gastrointestinal AEs were common and typically low-grade. The most frequent grade 3-4 AEs during oral azacitidine therapy were hematologic events. AEs infrequently required permanent discontinuation of oral azacitidine (13%), suggesting they were effectively managed with use of concomitant medications and oral azacitidine dosing modifi
- Published
- 2021
8. Special considerations in the management of adult patients with acute leukaemias and myeloid neoplasms in the COVID-19 era: recommendations from a panel of international experts
- Author
-
Zeidan, AM, Boddu, PC, Patnaik, MM, Bewersdorf, JP, Stahl, M, Rampal, RK, Shallis, RM, Steensma, DP, Savona, MR, Sekeres, MA, Roboz, GJ, DeAngelo, DJ, Schuh, AC, Padron, E, Zeidner, JF, Walter, RB, Onida, F, Fathi, AT, DeZern, A, Hobbs, G, Stein, EM, Vyas, P, Wei, AH, Bowen, DT, Montesinos, P, Griffiths, EA, Verma, AK, Keyzner, A, Bar-Natan, M, Navada, SC, Kremyanskaya, M, Goldberg, AD, Al-Kali, A, Heaney, ML, Nazha, A, Salman, H, Luger, S, Pratz, KW, Konig, H, Komrokji, R, Deininger, M, Cirici, BX, Bhatt, VR, Silverman, LR, Erba, HP, Fenaux, P, Platzbecker, U, Santini, V, Wang, ES, Tallman, MS, Stone, RM, and Mascarenhas, J
- Abstract
The ongoing COVID-19 pandemic caused by severe acute respiratory syndrome coronavirus 2 is a global public health crisis. Multiple observations indicate poorer post-infection outcomes for patients with cancer than for the general population. Herein, we highlight the challenges in caring for patients with acute leukaemias and myeloid neoplasms amid the COVID-19 pandemic. We summarise key changes related to service allocation, clinical and supportive care, clinical trial participation, and ethical considerations regarding the use of lifesaving measures for these patients. We recognise that these recommendations might be more applicable to high-income countries and might not be generalisable because of regional differences in health-care infrastructure, individual circumstances, and a complex and highly fluid health-care environment. Despite these limitations, we aim to provide a general framework for the care of patients with acute leukaemias and myeloid neoplasms during the COVID-19 pandemic on the basis of recommendations from international experts.
- Published
- 2020
9. Applicability and reproducibility of acute myeloid leukaemia stem cell assessment in a multi-centre setting
- Author
-
Hanekamp, D, Snel, A, Kelder, A, Scholten, WJ, Khan, NM, Metzner, M, Irno-Consalvo, M, Sugita, M, Jong, A, Oude Alink, Sjoerd, Eidhof, H, Wilhelm, MJ, Feuring-Buske, M, Slomp, J, van der Velden, Vincent, Sonneveld, EDJA, Guzman, MG, Roboz, GJ, Buccisano, F, Vyas, P, Freeman, SH, Bachas, C, Ossenkoppele, GL, Schuurhuis, GJ, Cloos, J, Hanekamp, D, Snel, A, Kelder, A, Scholten, WJ, Khan, NM, Metzner, M, Irno-Consalvo, M, Sugita, M, Jong, A, Oude Alink, Sjoerd, Eidhof, H, Wilhelm, MJ, Feuring-Buske, M, Slomp, J, van der Velden, Vincent, Sonneveld, EDJA, Guzman, MG, Roboz, GJ, Buccisano, F, Vyas, P, Freeman, SH, Bachas, C, Ossenkoppele, GL, Schuurhuis, GJ, and Cloos, J
- Published
- 2020
10. A-type proanthocyanidins selectively target acute myeloid leukemia cells in vitro and in vivo
- Author
-
Bystrom, LM, additional, Zong, H, additional, Martinez, LAL, additional, Neto, C, additional, Roboz, GJ, additional, Rivella, S, additional, and Guzman, ML, additional
- Published
- 2016
- Full Text
- View/download PDF
11. Pregnancy in a woman with chronic neutrophilic leukemia.
- Author
-
Taylor J, Roboz GJ, Baergen RN, Genc MR, Taylor, Jolyn, Roboz, Gail J, Baergen, Rebecca N, and Genc, Mehmet R
- Abstract
Background: Chronic neutrophilic leukemia is a rare myeloproliferative disorder in women of reproductive age.Case: A pregnant woman with an established diagnosis of chronic neutrophilic leukemia presented at 26 weeks of gestation with splenomegaly, thrombocytopenia, leukocytosis, and anemia. Thrombocytopenia was refractory to medical treatment and, in part, was attributed to splenic sequestration. She delivered a healthy neonate at 35 weeks of gestation by repeat cesarean delivery under general anesthesia. Her preoperative platelet count was 30,000/mL and she was transfused platelets throughout the perioperative period. Her postpartum course was complicated by intraabdominal hemorrhage and severe preeclampsia. She recovered with intensive medical and surgical management.Conclusion: Chronic neutrophilic leukemia poses difficult challenges during pregnancy and requires a multidisciplinary approach. [ABSTRACT FROM AUTHOR]- Published
- 2013
- Full Text
- View/download PDF
12. Pomalidomide is active in the treatment of anemia associated with myelofibrosis.
- Author
-
Tefferi A, Verstovsek S, Barosi G, Passamonti F, Roboz GJ, Gisslinger H, Paquette RL, Cervantes F, Rivera CE, Deeg HJ, Thiele J, Kvasnicka HM, Vardiman JW, Zhang Y, Bekele BN, Mesa RA, Gale RP, Kantarjian HM, Tefferi, Ayalew, and Verstovsek, Srdan
- Published
- 2009
- Full Text
- View/download PDF
13. Dasatinib in the treatment of chronic myeloid leukemia in accelerated phase after imatinib failure: the START a trial.
- Author
-
Apperley JF, Cortes JE, Kim DW, Roy L, Roboz GJ, Rosti G, Bullorsky EO, Abruzzese E, Hochhaus A, Heim D, de Souza CA, Larson RA, Lipton JH, Khoury HJ, Kim HJ, Sillaber C, Hughes TP, Erben P, Van Tornout J, and Stone RM
- Published
- 2009
- Full Text
- View/download PDF
14. On the use of lonafarnib in myelodysplastic syndrome and chronic myelomonocytic leukemia
- Author
-
E. Frank, Paul Kirschmeier, Jeffrey H. Lipton, Josy Reiffers, Stephen G. O'Brien, Johan Lanng Nielsen, Alan F. List, Giovanni Martinelli, Eric J. Feldman, François Guilhot, Yali Zhu, Stephen H. Petersdorf, Tessa L. Holyoake, Gail J. Roboz, Paul Statkevich, S. Loechner, A R Turner, Philippe Colombat, Jorge E. Cortes, Daniel J. DeAngelo, Bengt Simonsson, Juliet N. Barker, Frédéric Maloisel, Feldman EJ, Cortes J, DeAngelo DJ, Holyoake T, Simonsson B, O'Brien SG, Reiffers J, Turner AR, Roboz GJ, Lipton JH, Maloisel F, Colombat P, Martinelli G, Nielsen JL, Petersdorf S, Guilhot F, Barker J, Kirschmeier P, Frank E, Statkevich P, Zhu Y, Loechner S, and List A.
- Subjects
Adult ,Cancer Research ,medicine.medical_specialty ,Maximum Tolerated Dose ,Gastrointestinal Diseases ,Pyridines ,Nausea ,medicine.medical_treatment ,MYELODISPLASTIC SYNDROME ,Chronic myelomonocytic leukemia ,Anorexia ,Gastroenterology ,chemistry.chemical_compound ,Piperidines ,Internal medicine ,hemic and lymphatic diseases ,medicine ,Farnesyltranstransferase ,Humans ,Lonafarnib ,Enzyme Inhibitors ,Aged ,Aged, 80 and over ,Chemotherapy ,Hematology ,business.industry ,Remission Induction ,Farnesyltransferase inhibitor ,CHRONIC MYELOMONOCYTIC LEUKEMIA ,Leukemia, Myelomonocytic, Chronic ,LONAFARNIB ,Middle Aged ,medicine.disease ,Leukemia ,Treatment Outcome ,Oncology ,chemistry ,Myelodysplastic Syndromes ,Immunology ,Drug Monitoring ,medicine.symptom ,business - Abstract
Udgivelsesdato: 2008-Sep Lonafarnib is an orally bio-available farnesyltransferase inhibitor that prevents farnesylation of specific target proteins including Ras. In a multicenter study, 67 patients with advanced myelodysplastic syndrome (MDS) and chronic myelomonocytic leukemia (CMML) were treated with a continuous oral dose of 200-300 mg of lonafarnib and were evaluated for hematologic, pathologic and pharmacodynamic response. The median age of patients was 70 years (range 44-86). There were 32 patients with MDS (RAEB-20 and RAEB-t-12) and 35 with CMML. Overall 16 (24%) of the patients responded with two patients achieving a complete remission and one a partial response. Responses were seen in 6/32 and 10/35 patients with MDS and CMML, respectively. Of the 19 patients who were platelet transfusion-dependent prior to treatment, 5 (26%) became transfusion-free for a median duration of 185 days. A decrease in the farnesylation of the HDJ-2 protein measured in patient-derived cells was observed in the majority of patients during treatment with lonafarnib, but no clear correlation between changes in farnesylation and clinical effect could be made. Gastrointestinal toxicity was significant with 19% of patients discontinuing therapy due to diarrhea, nausea and/or anorexia. Lonafarnib has demonstrable activity in patients with advanced MDS and CMML.
- Published
- 2008
- Full Text
- View/download PDF
15. Dasatinib induces significant hematologic and cytogenetic responses in patients with imatinib-resistant or -intolerant chronic myeloid leukemia in accelerated phase
- Author
-
Gail J. Roboz, Martin C. Müller, Ashwin Gollerkeri, François Guilhot, Jeffrey H. Lipton, Michele Baccarani, Dong-Wook Kim, Richard A. Larson, Eduardo Bullorsky, Sergio Amadori, Andreas Hochhaus, Carmino Antonio De Souza, Dominik Heim, Susan Branford, Moshe Talpaz, Prasheen Agarwal, Jane F. Apperley, Guilhot F, Apperley J, Kim DW, Bullorsky EO, Baccarani M, Roboz GJ, Amadori S, de Souza CA, Lipton JH, Hochhaus A, Heim D, Larson RA, Branford S, Muller MC, Agarwal P, Gollerkeri A, and Talpaz M.
- Subjects
Adult ,Male ,medicine.medical_specialty ,Immunology ,Dasatinib ,Fusion Proteins, bcr-abl ,Antineoplastic Agents ,Biochemistry ,Imatinib resistant ,Gastroenterology ,Piperazines ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,hemic and lymphatic diseases ,Internal medicine ,Humans ,Point Mutation ,Medicine ,In patient ,Adverse effect ,Accelerated phase ,Aged ,Dose Modification ,Aged, 80 and over ,business.industry ,Myeloid leukemia ,Cell Biology ,Hematology ,Middle Aged ,Protein-Tyrosine Kinases ,Blood Cell Count ,Thiazoles ,Pyrimidines ,Treatment Outcome ,Imatinib mesylate ,Drug Resistance, Neoplasm ,Benzamides ,Cytogenetic Analysis ,Disease Progression ,Imatinib Mesylate ,Female ,business ,Settore MED/15 - Malattie del Sangue ,medicine.drug - Abstract
Treatment options are limited for patients with imatinib-resistant or -intolerant accelerated phase chronic myeloid leukemia (CML-AP). Dasatinib is a novel, potent, oral, multitargeted kinase inhibitor of BCR-ABL and SRC-family kinases that showed marked efficacy in a phase 1 trial of patients with imatinib-resistant CML. Results are presented for 107 patients with CML-AP with imatinib-resistance or -intolerance from a phase 2, open-label study further evaluating dasatinib efficacy and safety. At 8 months' minimum follow-up, 81%, 64%, and 39% of patients achieved overall, major (MaHR), and complete hematologic responses, respectively, whereas 33% and 24% attained major and complete cytogenetic remission. Of 69 patients who achieved MaHR, 7 progressed. Seventy-six percent of patients are estimated to be alive and progression-free at 10 months. Response rates for the 60% of patients with baseline BCR-ABL mutations did not differ from the total population. Dasatinib was well tolerated: most nonhematologic adverse events (AEs) were mild to moderate; no imatinib-intolerant patients discontinued dasatinib because of AEs. Although common (76% of patients with severe neutropenia), cytopenias were manageable through dose modification. In summary, dasatinib induced significant hematologic and cytogenetic responses in patients with imatinib resistance or intolerance, was well tolerated, and may represent a potent new therapeutic option for CML-AP. Further follow-up is warranted. This trial was registered at www.clinicaltrials.gov as #CA180005.
- Published
- 2007
16. Data-driven, harmonised classification system for myelodysplastic syndromes: a consensus paper from the International Consortium for Myelodysplastic Syndromes.
- Author
-
Komrokji RS, Lanino L, Ball S, Bewersdorf JP, Marchetti M, Maggioni G, Travaglino E, Al Ali NH, Fenaux P, Platzbecker U, Santini V, Diez-Campelo M, Singh A, Jain AG, Aguirre LE, Tinsley-Vance SM, Schwabkey ZI, Chan O, Xie Z, Brunner AM, Kuykendall AT, Bennett JM, Buckstein R, Bejar R, Carraway HE, DeZern AE, Griffiths EA, Halene S, Hasserjian RP, Lancet J, List AF, Loghavi S, Odenike O, Padron E, Patnaik MM, Roboz GJ, Stahl M, Sekeres MA, Steensma DP, Savona MR, Taylor J, Xu ML, Sweet K, Sallman DA, Nimer SD, Hourigan CS, Wei AH, Sauta E, D'Amico S, Asti G, Castellani G, Delleani M, Campagna A, Borate UM, Sanz G, Efficace F, Gore SD, Kim TK, Daver N, Garcia-Manero G, Rozman M, Orfao A, Wang SA, Foucar MK, Germing U, Haferlach T, Scheinberg P, Miyazaki Y, Iastrebner M, Kulasekararaj A, Cluzeau T, Kordasti S, van de Loosdrecht AA, Ades L, Zeidan AM, and Della Porta MG
- Subjects
- Humans, Consensus, Myelodysplastic Syndromes classification, Myelodysplastic Syndromes diagnosis, Myelodysplastic Syndromes genetics
- Abstract
The WHO and International Consensus Classification 2022 classifications of myelodysplastic syndromes enhance diagnostic precision and refine decision-making processes in these diseases. However, some discrepancies still exist and potentially cause inconsistency in their adoption in a clinical setting. We adopted a data-driven approach to provide a harmonisation between these two classification systems. We investigated the importance of genomic features and their effect on the cluster assignment process to define harmonised entity labels. A panel of expert haematologists, haematopathologists, and data scientists who are members of the International Consortium for Myelodysplastic Syndromes was formed and a modified Delphi consensus process was adopted to harmonise morphologically defined categories without a distinct genomic profile. The panel held regular online meetings and participated in a two-round survey using an online voting tool. We identified nine clusters with distinct genomic features. The cluster of highest hierarchical importance was characterised by biallelic TP53 inactivation. Cluster assignment was irrespective of blast count. Individuals with monoallelic TP53 inactivation were assigned to other clusters. Hierarchically, the second most important group included myelodysplastic syndromes with del(5q). Isolated del(5q) and less than 5% of blast cells in the bone marrow were the most relevant label-defining features. The third most important cluster included myelodysplastic syndromes with mutated SF3B1. The absence of isolated del(5q), del(7q)/-7, abn3q26.2, complex karyotype, RUNX1 mutations, or biallelic TP53 were the basis for a harmonised label of this category. Morphologically defined myelodysplastic syndrome entities showed large genomic heterogeneity that was not efficiently captured by single-lineage versus multilineage dysplasia, marrow blasts, hypocellularity, or fibrosis. We investigated the biological continuum between myelodysplastic syndromes with more than 10% bone marrow blasts and acute myeloid leukaemia, and found only a partial overlap in genetic features. After the survey, myelodysplastic syndromes with low blasts (ie, less than 5%) and myelodysplastic syndromes with increased blasts (ie, 5% or more) were recognised as disease entities. Our data-driven approach can efficiently harmonise current classifications of myelodysplastic syndromes and provide a reference for patient management in a real-world setting., Competing Interests: Declaration of interests UG reports speaker honoraria from Novartis, AbbVie, and BMS; and institutional research support from BMS, AbbVie, and Jazz Pharmaceuticals. FE reports consultancy or advisory roles for AbbVie, Incyte, Syros, Novartis, and Jazz Pharmaceuticals. SH reports research support from STORM Therapeutics and AstraZeneca. EAG reports honoraria from AAMDS, MedscapeLIVE!, MediCom Worldwide, MJH Life Sciences, ASH, MDS International Foundation, and Physicians’ Education Resource; consulting fees from AbbVie, Alexion, Apellis, Takeda Oncology, Astex/Taiho Oncology, Alexion/AstraZeneca Rare Disease, Celgene/BMS, CTI BioPharma, Novartis, Partner Therapeutics, Picnic Health, and Servier; and research funding from Alexion, Apellis, Astex /Otsuka/Taiho Oncology, Blueprint Medicines, Celldex Therapeutics, Genentech, and NextCure. MAS reports participation on advisory boards for BMS, Kurome, Schrödinger, and Karyopharm. MD-C reports participation on a data safety monitoring board or advisory board for BMS, Novartis, Blueprint Medicines, GSK, Agios, Hemavan, Syros, Keros, Curis, and Astex/Otsuka; and payment or honoraria for lectures, presentations, speakers’ bureaus, manuscript writing, or educational events for BMS, Novartis, and Keros. UP reports research support and honoraria from BMS, Geron, Curis, AbbVie, and Janssen. RBe reports employment or equity from Aptose Biosciences; participation on advisory boards for BMS, Servier, NeoGenomics, and Geron; being Data Monitoring Committee Chair for Gilead, Ipsen, and Keros; and consultancy for TenSixteen. YM reports honoraria from Nippon Shinyaku, BMS, Novartis, Sumitomo Pharma, Kyowa Kirin, AbbVie, Daiichi Sankyo, Takeda, Janssen, Astellas, Pfizer, Eisai, and Otsuka; and research funding from Chugai. AED reports participation on advisory boards, consultancy, or honoraria from Celgene/BMS, Agios, Novartis, Astellas, and Gilead; and participation on clinical trial committees or data safety monitoring boards for Novartis, AbbVie, Kura, Geron, Servier, Keros, and Celgene/BMS. DPS reports employment by Ajax Therapeutics; former employment by Novartis; and minor equity in Arrowhead and Bluebird. TKK reports consultancy for Agenus and ImmunoBiome. AK reports research support from Novartis and BMS; consulting fees from Alexion, Novartis, Amgen, Agios, Pfizer, Samsung, Celgene, F Hoffmann-La Roche, and Sobi; honoraria from Alexion, Novartis, Pfizer, Amgen, Samsung, Celgene, F Hoffmann-La Roche, BMS, Sobi, and Silence Therapeutics; and speakers fees from Alexion, Novartis, Amgen, Pfizer, Celgene, F Hoffmann-La Roche, and Sobi. All other authors declare no competing interests., (Copyright © 2024 Elsevier Ltd. All rights reserved, including those for text and data mining, AI training, and similar technologies.)
- Published
- 2024
- Full Text
- View/download PDF
17. Transplant Outcomes in Myelofibrosis: Impact of Donor Type (Cord Blood Grafts Supported by CD34+ selected Cells [Haplo-Cord] Versus Matched Donors).
- Author
-
Ghalehsari N, Castillo Tokumori F, Chen Z, Liu M, Mayer SA, Zeinah GA, Shore TB, Ritchie EK, Silver RT, Scandura JM, Roboz GJ, van Besien K, and Gomez-Arteaga A
- Subjects
- Humans, Male, Middle Aged, Female, Adult, Aged, Treatment Outcome, Tissue Donors statistics & numerical data, Hematopoietic Stem Cell Transplantation methods, Unrelated Donors, Transplantation, Homologous, Primary Myelofibrosis therapy, Primary Myelofibrosis mortality, Antigens, CD34, Graft vs Host Disease, Cord Blood Stem Cell Transplantation methods
- Abstract
Despite the established potentially curative role of allogeneic hematopoietic cell transplantation (allo-HCT) in managing myelofibrosis (MF), the choice of alternative donors for patients lacking matched donors remains a challenge, and the optimal graft source in this disease entity continues to be an ongoing debate. We aimed to evaluate the impact of donor type: umbilical cord blood transplant supported with CD34+ selected haploidentical donor (haplo-cord) versus adult matched related donor (MRD) and matched unrelated donor (MUD) in 40 adult patients with primary or secondary MF, including those progressing to accelerated phase (AP) or blast phase (BP), who underwent their first allo-HCT. The primary objective of this study was to analyze the impact of stem cell source on primary endpoints of overall survival (OS), graft-versus-host disease, and non-relapse mortality (NRM). Median follow-up for all alive patients was 53 months (range 0.3-63 months). Nine patients (22.5%) underwent an MRD allo-HCT, 15 patients (37.5%) underwent a MUD allo-HCT, and 16 patients (40%) underwent a haplo-cord allo-HCT. Four patients died without neutrophil engraftment: 3 (19%) in haplo-cord group and one (4%) in MRD/MUD group. The cumulative incidence of absolute neutrophil engraftment by day 60 was 80% (95% CI 45-94) in the haplo-cord group and 92% (95% CI 65-98) in the MRD/MUD group (P = .09). The cumulative incidence of platelet engraftment by day 60 was 59% (95% CI 27-81) in haplo-cord group and 75% (95% CI 51-88) in MRD/MUD group (P = .4). OS was 62% at 1 year (95% CI 49-79) and 34% at 3 years (95% CI 21-55). The 3-year OS was similar between the haplo-cord group and the MRD/MUD (37% versus 32%, P = .9). The 1-year OS for AP/BP patients was 50% (95% CI 27-93) in the haplo-cord group, compared to 40% (95% CI 19-86) in the MRD/MUD. The 1-year OS for chronic phase CP patients was 83% (95% CI 58-100) in the haplo-cord group, compared to 79% (95% CI 60-100) in the MRD/MUD group. The cumulative incidence of relapse at 3 years in the haplo-cord group was 13% (95% CI 1.8-34), and in the MRD/MUD group was 28% (95% CI 10-49) (P = .36). One-year NRM was 38% (95% CI 15-61) in the haplo-cord group and 33% (95% CI 15-52) in the MRD/MUD group. Three-year NRM was 48% (95% CI 19-72) in the haplo-cord group and 54% (95% CI 29-73) in MRD/MUD group (P = .95). We showed no significant difference in OS, relapse, and NRM outcomes after haplo-cord transplant compared to adult matched donors' grafts (MRD or MUD) in MF patients. However, there were more graft failures in patients transplanted with a haplo-cord transplants and delayed engraftments with inadequate haplo myeloid bridges. Despite the small sample size in our study, considering our findings and the availability of other alternative donors, using haplo-cord platforms may no longer be justified for MF unless the UCB engraftment dynamics can be optimized., Competing Interests: Conflict of Interest Statement The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this article., (Copyright © 2024 The American Society for Transplantation and Cellular Therapy. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
18. How I use Maintenance Therapy in Acute Myeloid Leukemia.
- Author
-
Roboz GJ and Canaani J
- Abstract
Outcomes for acute myeloid leukemia have improved significantly in the last decade with the approval of novel therapeutics targeting diverse vulnerabilities of leukemic cells, expanded access to stem cell transplantation, and improved safety of transplantation. While attainment of initial remission is now an expected outcome in most AML patients receiving intensive or non-intensive induction regimens, maintaining long-term remission and decreasing the risk of relapse remain critical challenges. Maintenance approaches employing assorted agents have yielded variable success and only recently have been integrated to the standard of care. We present four commonly encountered clinical scenarios that highlight challenges facing physicians as they care for AML patients in remission and contemplate using post-remission maintenance. Using published studies and emerging clinical data, we discuss our approach to maintenance treatment in AML, emphasizing that selection of a specific strategy is an individualized decision based on leukemia biology and risk stratification, presence of targetable mutations, initial treatment approach, performance status, and feasibility of allogeneic stem cell transplantation., (Copyright © 2024 American Society of Hematology.)
- Published
- 2024
- Full Text
- View/download PDF
19. Contemporary Approach to the Diagnosis and Classification of Myelodysplastic Neoplasms/Syndromes-Recommendations From the International Consortium for Myelodysplastic Neoplasms/Syndromes (MDS [icMDS]).
- Author
-
Aakash F, Gisriel SD, Zeidan AM, Bennett JM, Bejar R, Bewersdorf JP, Borate UM, Boultwood J, Brunner AM, Buckstein R, Carraway HE, Churpek JE, Daver NG, DeZern AE, Efficace F, Fenaux P, Figueroa ME, Garcia-Manero G, Gore SD, Greenberg PL, Griffiths EA, Halene S, Hourigan CS, Kim TK, Kim N, Komrokji RS, Kutchroo VK, List AF, Little RF, Majeti R, Nazha A, Nimer SD, Odenike O, Padron E, Patnaik MM, Platzbecker U, Della Porta MG, Roboz GJ, Sallman DA, Santini V, Sanz G, Savona MR, Sekeres MA, Stahl M, Starczynowski DT, Steensma DP, Taylor J, Abdel-Wahab O, Wei AH, Xie Z, Xu ML, Hasserjian RP, and Loghavi S
- Abstract
Myelodysplastic neoplasms/syndromes (MDS) are a heterogeneous group of biologically distinct entities characterized by variable degrees of ineffective hematopoiesis. Recently, 2 classification systems (the 5th edition of the World Health Organization Classification of Haematolymphoid tTumours and the International Consensus Classification) further subcharacterized MDS into morphologically and genetically defined groups. Accurate diagnosis and subclassification of MDS require a multistep systemic approach. The International Consortium for MDS (icMDS) summarizes a contemporary, practical, and multimodal approach to MDS diagnosis and classification., (Copyright © 2024 United States & Canadian Academy of Pathology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
20. Final phase 1 substudy results of ivosidenib for patients with mutant IDH1 relapsed/refractory myelodysplastic syndrome.
- Author
-
DiNardo CD, Roboz GJ, Watts JM, Madanat YF, Prince GT, Baratam P, de Botton S, Stein A, Foran JM, Arellano ML, Sallman DA, Hossain M, Marchione DM, Bai X, Patel PA, Kapsalis SM, Garcia-Manero G, and Fathi AT
- Subjects
- Humans, Female, Male, Middle Aged, Aged, Treatment Outcome, Adult, Recurrence, Antineoplastic Agents therapeutic use, Antineoplastic Agents adverse effects, Aged, 80 and over, Isocitrate Dehydrogenase genetics, Isocitrate Dehydrogenase antagonists & inhibitors, Pyridines therapeutic use, Pyridines adverse effects, Pyridines administration & dosage, Myelodysplastic Syndromes drug therapy, Myelodysplastic Syndromes genetics, Myelodysplastic Syndromes mortality, Glycine analogs & derivatives, Glycine therapeutic use, Glycine adverse effects, Glycine administration & dosage, Mutation
- Abstract
Abstract: Ivosidenib is a first-in-class mutant isocitrate dehydrogenase 1 (mIDH1) inhibitor with efficacy and tolerability in patients with advanced mIDH1 hematologic malignancies, leading to approval in frontline and relapsed/refractory (R/R) mIDH1 acute myeloid leukemia. We report final data from a phase 1 single-arm substudy of once-daily ivosidenib in patients with R/R mIDH1 myelodysplastic syndrome (MDS) after failure of standard-of-care therapies. Primary objectives were to determine safety, tolerability, and clinical activity. The primary efficacy end point was the complete remission (CR) + partial remission (PR) rate. Nineteen patients were enrolled; 18 were included in the efficacy analysis. Treatment-related adverse events occurred in 8 (42.1%) patients, including a grade 1 QT interval prolongation in 1 (5.3%) patient and grade 2 differentiation syndrome in 2 (10.5%) patients. Rates of CR + PR and objective response (CR + PR + marrow CR) were 38.9% (95% confidence interval [CI], 17.3-64.3) and 83.3% (95% CI, 58.6-96.4), respectively. Kaplan-Meier estimates showed a 68.6% probability of patients in CR achieving a remission duration of ≥5 years, and a median overall survival of 35.7 months. Of note, 71.4% and 75.0% baseline red blood cell (RBC)- and platelet-transfusion-dependent patients, respectively, became transfusion independent (TI; no transfusion for ≥56 days); 81.8% and 100% of baseline RBC and platelet TI patients, respectively, remained TI. One (5.3%) patient proceeded to a hematopoietic stem cell transplant. In conclusion, ivosidenib is clinically active, with durable remissions and a manageable safety profile observed in these patients. This trial was registered at www.ClinicalTrials.gov as #NCT02074839., (© 2024 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
21. Olutasidenib in post-venetoclax patients with mutant isocitrate dehydrogenase 1 (m IDH1 ) acute myeloid leukemia (AML).
- Author
-
Cortes J, Jonas BA, Schiller G, Mims A, Roboz GJ, Wei AH, Montesinos P, Ferrell PB, Yee KW, Fenaux P, Schwarer A, and Watts JM
- Subjects
- Humans, Middle Aged, Female, Male, Aged, Adult, Pyridines therapeutic use, Pyridines adverse effects, Treatment Outcome, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Antineoplastic Combined Chemotherapy Protocols adverse effects, Remission Induction, Antineoplastic Agents therapeutic use, Antineoplastic Agents adverse effects, Drug Resistance, Neoplasm genetics, Aged, 80 and over, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Isocitrate Dehydrogenase genetics, Isocitrate Dehydrogenase antagonists & inhibitors, Bridged Bicyclo Compounds, Heterocyclic therapeutic use, Bridged Bicyclo Compounds, Heterocyclic adverse effects, Sulfonamides therapeutic use, Sulfonamides adverse effects, Mutation
- Abstract
Olutasidenib, a potent, selective, oral, mutant isocitrate dehydrogenase 1 (mIDH1) inhibitor, is FDA-approved for relapsed/refractory (R/R) acute myeloid leukemia (AML). Here we report efficacy and safety of olutasidenib in 18 patients with m IDH1 AML who were relapsed (10), refractory (6) or had complete remission with incomplete hematologic recovery (CRi; 2) to a venetoclax combination. Of the 16 patients who were R/R, 4 (25%) achieved complete remission (CR), one (6.3%) achieved CR with partial hematologic recovery (CRh), and 7 (43.8%) achieved a composite complete remission (CRc). Median time to CRc was 1.9 months (range 1-2.8). As of data cutoff (18 June 2021), median duration of CRc was not reached (range, 1.2-NR, ongoing at 30.4+ months). Both patients with CRi at study entry achieved a CR. Safety was consistent with the overall profile of olutasidenib. Olutasidenib offers a valuable treatment option for patients with m IDH1 AML previously treated with venetoclax.
- Published
- 2024
- Full Text
- View/download PDF
22. Venetoclax and Cobimetinib in Relapsed/Refractory AML: A Phase 1b Trial.
- Author
-
Konopleva MY, Dail M, Daver NG, Garcia JS, Jonas BA, Yee KWL, Kelly KR, Vey N, Assouline S, Roboz GJ, Paolini S, Pollyea DA, Tafuri A, Brandwein JM, Pigneux A, Powell BL, Fenaux P, Olin RL, Visani G, Martinelli G, Onishi M, Wang J, Huang W, Dunshee DR, Hamidi H, Ott MG, Hong WJ, and Andreeff M
- Subjects
- Humans, Aged, Male, Female, Middle Aged, Aged, 80 and over, Treatment Outcome, Drug Resistance, Neoplasm drug effects, Bridged Bicyclo Compounds, Heterocyclic therapeutic use, Bridged Bicyclo Compounds, Heterocyclic pharmacology, Bridged Bicyclo Compounds, Heterocyclic administration & dosage, Sulfonamides pharmacology, Sulfonamides therapeutic use, Sulfonamides administration & dosage, Azetidines therapeutic use, Azetidines pharmacology, Azetidines administration & dosage, Piperidines therapeutic use, Piperidines pharmacology, Leukemia, Myeloid, Acute drug therapy, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Antineoplastic Combined Chemotherapy Protocols pharmacology, Antineoplastic Combined Chemotherapy Protocols adverse effects
- Abstract
Background: Therapies for relapsed/refractory acute myeloid leukemia remain limited and outcomes poor, especially amongst patients who are ineligible for cytotoxic chemotherapy or targeted therapies., Patients and Methods: This phase 1b trial evaluated venetoclax, a B-cell lymphoma-2 (BCL-2) inhibitor, plus cobimetinib, a MEK1/2 inhibitor, in patients with relapsed/refractory acute myeloid leukemia, ineligible for cytotoxic chemotherapy. Two-dimensional dose-escalation was performed for venetoclax dosed daily, and for cobimetinib dosed on days 1-21 of each 28-day cycle., Results: Thirty patients (median [range] age: 71.5 years [60-84]) received venetoclax-cobimetinib. The most common adverse events (AEs; in ≥40.0% of patients) were diarrhea (80.0%), nausea (60.0%), vomiting (40.0%), febrile neutropenia (40.0%), and fatigue (40.0%). Overall, 66.7% and 23.3% of patients experienced AEs leading to dose modification/interruption or treatment withdrawal, respectively. The composite complete remission (CRc) rate (complete remission [CR] + CR with incomplete blood count recovery + CR with incomplete platelet recovery) was 15.6%; antileukemic response rate (CRc + morphologic leukemia-free state/partial remission) was 18.8%. For the recommended phase 2 dose (venetoclax: 600 mg; cobimetinib: 40 mg), CRc and antileukemic response rates were both 12.5%. Failure to achieve an antileukemic response was associated with elevated baseline phosphorylated ERK and MCL-1 levels, but not BCL-xL. Baseline mutations in ≥1 signaling gene or TP53 were noted in nonresponders and emerged on treatment. Pharmacodynamic biomarkers revealed inconsistent, transient inhibition of the mitogen-activated protein kinase (MAPK) pathway., Conclusion: Venetoclax-cobimetinib showed limited preliminary efficacy similar to single-agent venetoclax, but with added toxicity. Our findings will inform future trials of BCL-2/MAPK pathway inhibitor combinations., Competing Interests: Disclosure MYK is a consultant for AbbVie, Inc., Genentech, Inc., and F. Hoffmann La-Roche; is an advisory board member for F. Hoffmann La-Roche; holds shares from Reata Pharmaceuticals; has received honoraria from Amgen, AbbVie, Inc., and Genentech, Inc.; and has received research funding from AbbVie, Inc., Genentech, Inc., Eli Lilly, Cellectis, Calithera, Stemline, Threshold, Flexus Biosciences, Novartis, Ablynx, and Agios. MD, MO, JW, WH, DD, and HH are employees of Genentech, Inc. and may hold Roche stock or stock options. NGD has received research funding from Daiichi Sankyo, Bristol-Myers Squibb, Pfizer, Gilead, Sevier, Genentech, Inc., Astellas, Daiichi Sankyo, AbbVie, Inc., Hanmi, Trovagene, Fate Therapeutics, Amgen, Novimmune, GlycoMimetics, Trillium, Kite Pharma, Aptose, Shattuck Labs, KAHR, ArcellX, Sanofi, Sumitomo, and ImmunoGen; and has served in a consulting or advisory role for Daiichi Sankyo, Bristol-Myers Squibb, Arog Pharmaceuticals, Pfizer, Novartis, Jazz Pharmaceuticals, Celgene, AbbVie, Inc., Astellas, Genentech, Inc., Immunogen, Servier, Syndax, Trillium, Gilead, Amgen, Shattuck Labs, ArcellX, Kite Pharma, Sumitomo, Caribou Biosciences, Sanofi, Rigel, Aptose, KURA, GlycoMimetics, and Agios. JSG has received research funding (for trials) from AbbVie, Inc., Genentech, Inc., Pfizer, Prelude, and AstraZeneca; and has served on advisory boards for AbbVie, Inc., Astellas, Bristol-Myers Squibb and Servier. BAJ is a consultant/advisor for AbbVie, Inc., Bristol-Myers Squibb, Daiichi Sankyo, Genentech, Inc., Gilead, GlycoMimetics, Jazz Pharmaceuticals, Kymera, Pfizer, Rigel, Servier, and Takeda; protocol steering committee for GlycoMimetics; data monitoring committee for Gilead; travel reimbursement/support from Rigel; and research funding to his institution from AbbVie, Inc., Amgen, Arog Pharmaceuticals, Aptose, BMS, Celgene, Daiichi Sankyo, F. Hoffmann-La Roche, Forma Therapeutics, Forty-Seven, Genentech, Inc./Roche, Gilead, GlycoMimetics, Hanmi, Immune-Onc, Incyte, Jazz Pharmaceuticals, Loxo Oncology, Pfizer, Pharmacyclics, Sigma Tau, and Treadwell. KWLY is a consultant for Astex, Bristol-Myers Squibb/Celgene, F. Hoffmann-La Roche, Novartis, Otsuka, Paladin, Pfizer, Shattuck Labs, Taiho, and Takeda; has received honorarium from AbbVie, Inc. and Novartis; and has received research funding from Astex, Forma Therapeutics, F. Hoffmann-La Roche, Genentech, Inc., Geron, Janssen, Jazz Pharmaceuticals, MedImmune, Novartis, Onconova, and Tolero. KRK, NV, SP, AT, AP, PF, and GV declare no competing financial interests. SA reports non-financial support from Roche/Genentech, Inc. during the conduct of the study; and has received personal fees from Roche Canada, Pfizer, Bristol-Myers Squibb, Palladin, and Lundbeck outside of the submitted work. GJR consulted for AbbVie, Inc., Amgen, Argenx, AstraZeneca, Bluebird Bio, Blueprint Medicines, Bristol-Myers Squibb, Caribou Biosciences, Celgene, Daiichi Sankyo, Ellipses Pharma, GlaxoSmithKline, Janssen, Jasper Pharmaceuticals, Jazz Pharmaceuticals, Molecular Partners, Novartis, Pfizer, Roche, Syndax, Takeda (IRC Chair), and Telix Pharma; and has received research funding from Janssen. DAP has received research funding from AbbVie, Inc. and has served as a consultant or advisory board member for AbbVie, Inc. and Genentech, Inc. JMB consulted for AbbVie, Inc., Bristol-Myers Squibb, Astex, Pfizer, Astellas, Taiho, and Jazz Pharmaceuticals. BLP has received research funding from Ambit Biosciences, Genentech, Inc., F. Hoffmann-La Roche, Jazz Pharmaceuticals, Novartis, Pfizer, and Cornerstone Pharmaceuticals; and is a consultant for Cornerstone Pharmaceuticals. RLO has received research support for trials from Astellas, Pfizer, Genentech, Inc., Daiichi Sankyo, and Cellectis; and consulted for AbbVie, Inc., Astellas, and Actinium. GM is a consultant for AbbVie, Inc., Celgene, Roche, Janssen, Astellas, Pfizer, and Incyte. W-JH is a former employee of Genentech, Inc. and may hold Roche stock; and is a current employee of Prelude Therapeutics. MGO is an employee of F. Hoffmann-La Roche and may hold stock or stock options. MA consulted for Amgen, AstraZeneca, Daiichi Sankyo, Syndax, GlycoMimetics, Oncoceutics, and Aptose; has received research funding from F. Hoffmann-La Roche, AstraZeneca, Amgen, Daiichi Sankyo, Jazz Pharmaceuticals, GlycoMimetics; and holds stock from Reata, Oncoceutics/Chimerix and Aptose., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
23. Toward a more patient-centered drug development process in clinical trials for patients with myelodysplastic syndromes/neoplasms (MDS): Practical considerations from the International Consortium for MDS (icMDS).
- Author
-
Efficace F, Buckstein R, Abel GA, Giesinger JM, Fenaux P, Bewersdorf JP, Brunner AM, Bejar R, Borate U, DeZern AE, Greenberg P, Roboz GJ, Savona MR, Sparano F, Boultwood J, Komrokji R, Sallman DA, Xie Z, Sanz G, Carraway HE, Taylor J, Nimer SD, Della Porta MG, Santini V, Stahl M, Platzbecker U, Sekeres MA, and Zeidan AM
- Abstract
Notable treatment advances have been made in recent years for patients with myelodysplastic syndromes/neoplasms (MDS), and several new drugs are under development. For example, the emerging availability of oral MDS therapies holds the promise of improving patients' health-related quality of life (HRQoL). Within this rapidly evolving landscape, the inclusion of HRQoL and other patient-reported outcomes (PROs) is critical to inform the benefit/risk assessment of new therapies or to assess whether patients live longer and better, for what will likely remain a largely incurable disease. We provide practical considerations to support investigators in generating high-quality PRO data in future MDS trials. We first describe several challenges that are to be thoughtfully considered when designing an MDS-focused clinical trial with a PRO endpoint. We then discuss aspects related to the design of the study, including PRO assessment strategies. We also discuss statistical approaches illustrating the potential value of time-to-event analyses and their implications within the estimand framework. Finally, based on a literature review of MDS randomized controlled trials with a PRO endpoint, we note the PRO items that deserve special attention when reporting future MDS trial results. We hope these practical considerations will facilitate the generation of rigorous PRO data that can robustly inform MDS patient care and support treatment decision-making for this patient population., Competing Interests: Fabio Efficace had consultancy or advisory role for AbbVie, Incyte, Syros, Novartis, and JAZZ Pharmaceuticals outside the submitted work. Rena Buckstein: Research funding and honoraria for advisory boards and speaking engagements from BMS, TAIHO, and Abbvie. Gregory A. Abel has consulted for Novartis and Geron outside the submitted work. Pierre Fenaux received research funding from BMS, Abbvie, Jazz Pharmaceuticals, Novartis, and Janssen; and had a consultancy with and received honoraria from BMS, Abbvie, Jazz Pharmaceuticals, and Novartis. Andrew M. Brunner received consulting or advisory board honoraria from Novartis, Acceleron, Agios, Abbvie, Takeda, Celgene/BMS, Keros Therapeutics, Taiho, Gilead; and has research support from the NIH SPORE in Myeloid Malignancies, and from the Edward P. Evans Foundation. Rafael Bejar owns equity in and is employed by Aptose Biosciences; he has served as an advisor to BMS, Servier, Gilead, and Ipsen; he is on the SAB for NeoGenomics. Amy E DeZern participated in advisory boards, and/or had a consultancy with and received honoraria from Celgene/BMS, Agios, Regenergon, Sobi, Novartis, Astellas, and Gilead. Amy E. DeZern served on clinical trial committees for Novartis, Abbvie, Kura, Geron, and Celgene/BMS. Gail J. Roboz: Consultancy: Abbvie, Amgen, Astra Zeneca, Bristol‐Myers Squibb, Caribou Biosciences, Celgene, Daiichi Sankyo, Ellipses Pharma, Genoptix, Geron, GlaxoSmithKline, Janssen, Jasper Pharmaceuticals, Jazz Pharmaceuticals, Molecular Partners, Novartis, Pfizer, Oncoverity, OncoPrecision, Rigel, Roche, Syndax, Takeda (IRC Chair), Telix Pharma; and research support: Janssen. Michael R. Savona: Membership on a board or advisory committee: Bristol Myers Squibb, CTI, Forma, Geron, GSK, Karyopharm, Rigel Ryvu, Taiho, Takeda, Treadwell; patents and royalties: Boehringer Ingelheim, Empath Biosciences; research funding: ALX Oncology, Astex, Incyte, Takeda, TG Therapeutics; equity ownership: Empath Biosciences, Karyopharm, Ryvu; consultancy: Forma, Geron, Karyopharm, Ryvu. Rami Komrokji: Abbvie: Speaker Bureau, Advisory board; BMS: Research grant, Advisory board; DSI: Advisory board; Geron: Consultancy; Janssen: Consultancy; Jazz: Speaker Bureau, Advisory board; Pharma Essentia: Speaker Bureau, Advisory board; Rigel: Speaker Bureau, Advisory board; Servio: Speaker Bureau, Advisory board; Sobi: Speaker Bureau, Advisory board; Sumitomo Pharma: consultancy, Advisory board. David A. Sallman served on the advisory board or panel for Agios, Avencell, BlueBird Bio, BMS, Dark Blue, Jasper Therapeutics, Kite, Magenta Therapeutics, NKARTA, Novartis, Rigel Shattuck Labs, Servier, Syndax, Syros; and had a consultancy with AbbBie, Gilead, Molecular Partners AG, Takeda. Guillermo Sanz received honoraria, advisory board membership, or consultation fees from AbbVie, BMS, ExCellThera, Novartis, Roche, and Takeda and participated in sponsored speaker's bureau for BMS, Novartis, and Takeda. Hetty E. Carraway: Consultancy: Abbvie, Amgen, Bristol‐Myers Squibb, Celgene, Daiichi Sankyo, Jazz Pharmaceuticals, Novartis, Rigel, Syndax, Servier; and research support: Celgene. Maximilian Stahl consulted for Curis Oncology and Boston Consulting; served on the advisory board for Novartis and Kymera, GSK, Rigel, and Sierra Oncology; and participated in GME activity for Novartis, Curis Oncology, Haymarket Media, and Clinical care options (CCO). Mikkael A. Sekeres has served on advisory boards for BMS, Novartis, Kurome, and Gilead. Amer M. Zeidan received research funding (institutional) from Celgene/BMS, Abbvie, Astex, Pfizer, Medimmune/Astra Zeneca, Boehringer‐Ingelheim, Cardiff oncology, Incyte, Takeda, Novartis, Aprea, and ADC Therapeutics. He participated in advisory boards, and/or had a consultancy with and received honoraria from AbbVie, Otsuka, Pfizer, Celgene/BMS, Jazz, Incyte, Agios, Boehringer‐Ingelheim, Novartis, Acceleron, Astellas, Daiichi Sankyo, Cardinal Health, Taiho, Seattle Genetics, BeyondSpring, Cardiff Oncology, Takeda, Ionis, Amgen, Janssen, Epizyme, Syndax, Gilead, Kura, Chiesi, ALX Oncology, BioCryst, Notable, Orum, and Tyme. He served on clinical trial committees for Novartis, Abbvie, Gilead, BioCryst, Abbvie, ALX Oncology, Geron, and Celgene/BMS. Fabio Efficace, Rena Buckstein, and Gregory A. Abel report being involved in the development and validation of the QUALMS, one of the PROs discussed in this paper. The QUALMS is copyrighted by Dana‐Farber Cancer Institute (DFCI) and the Children's Hospital of Eastern Ontario (CHEO). The remaining authors declare no conflict of interest., (© 2024 The Authors. HemaSphere published by John Wiley & Sons Ltd on behalf of European Hematology Association.)
- Published
- 2024
- Full Text
- View/download PDF
24. Guadecitabine vs TC in relapsed/refractory AML after intensive chemotherapy: a randomized phase 3 ASTRAL-2 trial.
- Author
-
Roboz GJ, Sanz G, Griffiths EA, Yee K, Kantarjian H, Récher C, Byrne MT, Patkowska E, Kim HJ, Thomas X, Moors I, Stock W, Illés Á, Fenaux P, Miyazaki Y, Yamauchi T, O'Connell CL, Hao Y, Keer HN, Azab M, and Döhner H
- Subjects
- Humans, Female, Middle Aged, Male, Aged, Adult, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Antineoplastic Combined Chemotherapy Protocols adverse effects, Recurrence, Treatment Outcome, Cytarabine therapeutic use, Aged, 80 and over, Young Adult, Drug Resistance, Neoplasm, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute mortality, Azacitidine therapeutic use, Azacitidine analogs & derivatives
- Abstract
Abstract: Guadecitabine is a novel hypomethylating agent (HMA) resistant to deamination by cytidine deaminase. Patients with relapsed/refractory acute myeloid leukemia (AML) were randomly assigned to guadecitabine or a preselected treatment choice (TC) of high-intensity chemotherapy, low-intensity treatment with HMAs or low-dose cytarabine, or best supportive care (BSC). The primary end point was overall survival (OS). A total of 302 patients were randomly assigned to guadecitabine (n = 148) or TC (n = 154). Preselected TCs were low-intensity treatment (n = 233 [77%; mainly HMAs]), high-intensity chemotherapy (n = 63 [21%]), and BSC (n = 6 [2%]). The median OS were 6.4 and 5.4 months for guadecitabine and TC, respectively (hazard ratio 0.88 [95% confidence interval, 0.67-1.14]; log-rank P = .33). Survival benefit for guadecitabine was suggested in several prospective subgroups, including age <65 years, Eastern Cooperative Oncology Group performance status 0 to 1, refractory AML, and lower peripheral blood blasts ≤30%. Complete response (CR) + CR with partial hematologic recovery rates were 17% for guadecitabine vs 8% for TC (P < .01); CR+CR with incomplete count recovery rates were 27% for guadecitabine vs 14% for TC (P < .01). Safety was comparable for the 2 arms, but guadecitabine had a higher rate of grade ≥3 neutropenia (32% vs 17%; P < .01). This study did not demonstrate an OS benefit for guadecitabine. Clinical response rates were higher for guadecitabine, with comparable safety to TC. There was an OS benefit for guadecitabine in several prespecified subgroups. This study was registered at www.clinicaltrials.gov as #NCT02920008., (© 2024 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
25. Efficacy and safety of bosutinib in patients treated with prior imatinib and/or dasatinib and/or nilotinib: Subgroup analyses from the phase 4 BYOND study.
- Author
-
Smith BD, Brümmendorf TH, Roboz GJ, Gambacorti-Passerini C, Charbonnier A, Viqueira A, Leip E, Purcell S, Goldman EH, Giles F, Ernst T, Hochhaus A, and Rosti G
- Subjects
- Humans, Imatinib Mesylate adverse effects, Dasatinib adverse effects, Philadelphia Chromosome, Protein Kinase Inhibitors adverse effects, Pyrimidines, Pathologic Complete Response, Antineoplastic Agents adverse effects, Leukemia, Myeloid, Chronic-Phase drug therapy, Aniline Compounds, Nitriles, Quinolines
- Abstract
The BYOND study evaluated the efficacy and safety of bosutinib 500 mg once daily in patients with chronic myeloid leukemia (CML) resistant/intolerant to prior tyrosine kinase inhibitors (TKIs). These post-hoc analyses assessed the efficacy and safety of bosutinib by resistance or intolerance to prior TKIs (imatinib-resistant vs dasatinib/nilotinib-resistant vs TKI-intolerant), and cross-intolerance between bosutinib and prior TKIs (imatinib, dasatinib, nilotinib), in patients with Philadelphia chromosome-positive chronic phase CML. Data are reported after ≥3 years' follow-up. Of 156 patients with Philadelphia chromosome-positive chronic phase CML, 53 were imatinib-resistant, 29 dasatinib/nilotinib-resistant, and 74 intolerant to all prior TKIs; cumulative complete cytogenetic response rates at any time were 83.7%, 61.5%, and 86.8%, and cumulative major molecular response rates at any time were 72.9%, 40.7%, and 82.4%, respectively. Of 141, 95, and 79 patients who received prior imatinib, dasatinib, and nilotinib, 64 (45.4%), 71 (74.7%), and 60 (75.9%) discontinued the respective TKI due to intolerance; of these, 2 (3.1%), 5 (7.0%), and 0 had cross-intolerance with bosutinib. The response rates observed in TKI-resistant and TKI-intolerant patients, and low cross-intolerance between bosutinib and prior TKIs, further support bosutinib use for patients with Philadelphia chromosome-positive chronic phase CML resistant/intolerant to prior TKIs. TRIAL REGISTRATION: ClinicalTrials.gov: NCT02228382., Competing Interests: Declaration of Competing Interest B. Douglas Smith: honoraria for consulting to Agios, Celgene, Jazz Pharmaceuticals, Novartis and Pfizer, and received research support from Pfizer. Tim H. Brümmendorf: consultant for Janssen, Merck, Novartis, Pfizer, and Takeda, and received research support from Novartis and Pfizer. Gail J. Roboz: consultancy, advisory board or data and safety monitoring committee for AbbVie, Actinium, Agios, Amphivena, Argenx, Array Biopharma, Astex, Astellas, AstraZeneca, Bayer, Celgene, Celltrion, Daiichi Sankyo, Eisai, Epizyme, Helsinn, Janssen, Jasper Therapeutics, Jazz, MEI Pharma – IDMC Chair, Novartis, Orsenix, Otsuka, Pfizer, Roche/Genentech, Sandoz, Takeda – IRC Chair, Trovagene. Received research support from Cellectis and Pfizer. Carlo Gambacorti-Passerini: provides consultancy to Bristol-Myers Squibb and received honoraria and research support from Pfizer. Aude Charbonnier: provides consultancy to Novartis and Pfizer; speaker’s bureau for Incyte; received research support from Pfizer. Andrea Viqueira, Eric Leip, Simon Purcell, and Erinn Goldman: employees of and have stock/stock options in Pfizer. Francis Giles: consultant to Actuate Therapeutics Inc, provides expert testimony to Novartis, and received research support from Pfizer. Thomas Ernst and Andreas Hochhaus: received research support from Bristol-Myers Squibb, Incyte, Novartis, and Pfizer. Gianantonio Rosti: received research support from Pfizer and served on the speaker bureau for Bristol-Myers Squibb, Incyte, Novartis, and Pfizer., (Copyright © 2024. Published by Elsevier Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
26. Survival outcomes in patients with acute myeloid leukaemia who received subsequent therapy for relapse in QUAZAR AML-001.
- Author
-
Ravandi F, Döhner H, Wei AH, Montesinos P, Pfeilstöcker M, Papayannidis C, Lai Y, Wang K, See WL, de Menezes DL, Petrlik E, Prebet T, and Roboz GJ
- Subjects
- Humans, Antimetabolites, Antineoplastic therapeutic use, Neoplasm Recurrence, Local drug therapy, Remission Induction, Chronic Disease, Antimetabolites therapeutic use, Azacitidine therapeutic use, Leukemia, Myeloid, Acute drug therapy
- Abstract
In the phase 3 QUAZAR AML-001 trial (NCT01757535) of patients with acute myeloid leukaemia (AML) in remission following intensive chemotherapy (IC) and ineligible for haematopoietic stem cell transplant (HSCT), oral azacitidine (Oral-AZA) maintenance significantly prolonged overall survival (OS) versus placebo. The impact of subsequent treatment following maintenance has not been evaluated. In this post hoc analysis, OS was estimated for patients who received subsequent AML therapy, and by regimen received (IC or lower-intensity therapy). First subsequent therapy (FST) was administered after treatment discontinuation in 134/238 Oral-AZA and 173/234 placebo patients. OS from randomization in patients who received FST after Oral-AZA versus placebo was 17.8 versus 12.9 months (HR: 0.82 [95% CI: 0.64-1.04], median follow-up: 56.7 months); OS from FST was similar between arms. Among patients who received injectable hypomethylating agents as FST, median OS was 8.2 versus 4.9 months in the Oral-AZA versus placebo groups (HR: 0.66 [95% CI: 0.41-1.06]). Forty-eight patients (16/238 Oral-AZA, 32/234 placebo) received HSCT following treatment discontinuation, including six Oral-AZA patients still in first remission; Oral-AZA OS benefit persisted when censoring these patients. Oral-AZA maintenance can prolong AML remission duration without negatively impacting survival outcomes after salvage therapies., (© 2023 Bristol Myers Squibb and The Authors. British Journal of Haematology published by British Society for Haematology and John Wiley & Sons Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
27. Oral decitabine-cedazuridine versus intravenous decitabine for myelodysplastic syndromes and chronic myelomonocytic leukaemia (ASCERTAIN): a registrational, randomised, crossover, pharmacokinetics, phase 3 study.
- Author
-
Garcia-Manero G, McCloskey J, Griffiths EA, Yee KWL, Zeidan AM, Al-Kali A, Deeg HJ, Patel PA, Sabloff M, Keating MM, Zhu N, Gabrail NY, Fazal S, Maly J, Odenike O, Kantarjian H, DeZern AE, O'Connell CL, Roboz GJ, Busque L, Buckstein R, Amin H, Randhawa J, Leber B, Shastri A, Dao KH, Oganesian A, Hao Y, Keer HN, Azab M, and Savona MR
- Subjects
- Male, Humans, Female, Decitabine adverse effects, Treatment Outcome, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Leukemia, Myelomonocytic, Chronic drug therapy, Myelodysplastic Syndromes drug therapy, Myelodysplastic Syndromes etiology, Pneumonia etiology
- Abstract
Background: The DNA methyltransferase inhibitors azacitidine and decitabine for individuals with myelodysplastic syndromes or chronic myelomonocytic leukaemia are available in parenteral form. Oral therapy with similar exposure for these diseases would offer potential treatment benefits. We aimed to compare the safety and pharmacokinetics of oral decitabine plus the cytidine deaminase inhibitor cedazuridine versus intravenous decitabine., Methods: We did a registrational, multicentre, open-label, crossover, phase 3 trial of individuals with myelodysplastic syndromes or chronic myelomonocytic leukaemia and individuals with acute myeloid leukaemia, enrolled as separate cohorts; results for only participants with myelodysplastic syndromes or chronic myelomonocytic leukaemia are reported here. In 37 academic and community-based clinics in Canada and the USA, we enrolled individuals aged 18 years or older who were candidates to receive intravenous decitabine, with Eastern Cooperative Oncology Group performance status 0 or 1 and a life expectancy of at least 3 months. Participants were randomly assigned (1:1) to receive 5 days of oral decitabine-cedazuridine (one tablet once daily containing 35 mg decitabine and 100 mg cedazuridine as a fixed-dose combination) or intravenous decitabine (20 mg/m
2 per day by continuous 1-h intravenous infusion) in a 28-day treatment cycle, followed by 5 days of the other formulation in the next treatment cycle. Thereafter, all participants received oral decitabine-cedazuridine from the third cycle on until treatment discontinuation. The primary endpoint was total decitabine exposure over 5 days with oral decitabine-cedazuridine versus intravenous decitabine for cycles 1 and 2, measured as area under the curve in participants who received the full treatment dose in cycles 1 and 2 and had decitabine daily AUC0-24 for both oral decitabine-cedazuridine and intravenous decitabine (ie, paired cycles). On completion of the study, all patients were rolled over to a maintenance study. This study is registered with ClinicalTrials.gov, NCT03306264., Findings: Between Feb 8, 2018, and June 7, 2021, 173 individuals were screened, 138 (80%) participants were randomly assigned to a treatment sequence, and 133 (96%) participants (87 [65%] men and 46 [35%] women; 121 [91%] White, four [3%] Black or African-American, three [2%] Asian, and five [4%] not reported) received treatment. Median follow-up was 966 days (IQR 917-1050). Primary endpoint of total exposure of oral decitabine-cedazuridine versus intravenous decitabine was 98·93% (90% CI 92·66-105·60), indicating equivalent pharmacokinetic exposure on the basis of area under the curve. The safety profiles of oral decitabine-cedazuridine and intravenous decitabine were similar. The most frequent adverse events of grade 3 or worse were thrombocytopenia (81 [61%] of 133 participants), neutropenia (76 [57%] participants), and anaemia (67 [50%] participants). The incidence of serious adverse events in cycles 1-2 was 31% (40 of 130 participants) with oral decitabine-cedazuridine and 18% (24 of 132 participants) with intravenous decitabine. There were five treatment-related deaths; two deemed related to oral therapy (sepsis and pneumonia) and three to intravenous treatment (septic shock [n=2] and pneumonia [n=1])., Interpretation: Oral decitabine-cedazuridine was pharmacologically and pharmacodynamically equivalent to intravenous decitabine. The results support use of oral decitabine-cedazuridine as a safe and effective alternative to intravenous decitabine for treatment of individuals with myelodysplastic syndromes or chronic myelomonocytic leukaemia., Funding: Astex Pharmaceuticals., Competing Interests: Declaration of interests GG-M has consulted for Acceleron; received honoraria and research funding from AbbVie, Astex, Bristol Myers Squibb, Curis, Genentech, and Novartis; received honoraria from Aprea; and received research funding from Gilead. JMc has received research funding from Astex; consulted for AbbVie, Apellis, BMS, CTI BioPharma, Novartis, Pfizer, and Takeda; received honoraria from BMS, Blueprint, Incyte, Jazz, Novartis, Stemline, and Takeda; and is an equity holder in COTA Healthcare. EAG has received research funding from Astex, Blueprint, and Celldex; received research funding from and consulted and served on advisory committees for Alexion and BMS/Celgene; consulted for and served on advisory committees for AbbVie, Apellis, AstraZeneca, CTI BioPharma, Genentech, Novartis, Taiho, and Takeda; and received honoraria from Aplastic Anemia and MDS International Foundation, Medicom, Physician Educational Resource, and Picnic Health. KWLY has consulted for BMS/Celgene, GSK, Jazz, Novartis, Pfizer, Shattuck, Taiho, and Takeda; received research funding from Astex, Forma, Genentech, Geron, Gilead, Janssen, Jazz, Novartis, Roche, Pfizer, and Treadwell; and received honoraria from AbbVie and Novartis. AMZ has received research funding from AbbVie, ADC Therapeutics, Amgen, Aprea, Astex, AstraZeneca, BMS, Boehringer-Ingelheim, Cardiff Oncology, Celgene, Incyte, Medimmune, Novartis, Otsuka, Pfizer, Takeda, and Trovagene; consulted for AbbVie, Acceleron, Agios, Amgen, Aprea, Astellas, BeyondSpring, BMS, Boehringer-Ingelheim, Cardiff, Cardinal Health, Celgene, Daiichi Sankyo, Epizyme, Geron, Gilead, Incyte, Ionis, Janssen, Jazz, Kura, Novartis, Pfizer, Seattle Genetics, Syndax, Taiho, Takeda, Trovagene, and Tyme; received honoraria from AbbVie, Acceleron, Agios, Amgen, Aprea, Astellas, BeyondSpring, BMS, Boehringer-Ingelheim, Cardiff, Cardinal, Celgene, Daiichi Sankyo, Epizyme, Geron, Gilead, Incyte, Ionis, Janssen, Jazz, Kura, Novartis, Otsuka, Pfizer, Seattle Genetics, Syndax, Taiho, Takeda, Trovagene, and Tyme; received travel expenses from Cardiff, Novartis, and Pfizer; and had leadership roles in boards or committees for AbbVie, BMS, Celgene, Geron, Gilead, Kura, and Novartis. AA-K's institution has received research support from Astex. HJD, NZ, NYG, JMa, HA, and JR declare no competing interests. PAP has served on an advisory committee for Agios. MS has served on advisory committees for AbbVie, BMS, Celgene, Jazz, Novartis, Pfizer, Roche, and Taiho and served on advisory committees for and received research funding from Actinium and Astellas. M-MK has served on advisory boards for AstraZeneca, BeiGene, BMS, Seattle Genetics, and Taiho. SF has consulted for and received honoraria from Blueprint, CTI, Gilead, GSK, Incyte, Janssen, Jazz, Karyopharm, Novartis, PharmaEssentia, Sanofi, Servier, Stemline, Taiho, and Takeda. OO has consulted for AbbVie, BMS/Celgene, CTI, Impact, Novartis, and Taiho and his institution has received research funding from AbbVie, Agios, Aprea, Astex, AstraZeneca, BMS/Celgene, CTI, Incyte, Janssen, Kartos, NS Pharma, and OncoTherapy. HK has received grants from AbbVie, Amgen, Ascentage, BMS, Daiichi-Sankyo, Immunogen, Jazz, and Novartis and received honoraria from AbbVie, Amgen, Amphista, Ascentage, Astellas, Biologix, Curis, Ipsen, KAHR, Labcorp, Novartis, Pfizer, Shenzhen Target Rx, Stemline, and Takeda. AED has consulted for Taiho; received honoraria from BMS, Novartis, and Gilead; served as chair of a data safety monitoring board for Geron; and is the 2023 American Society of Hematology Education Chair. CLO has received research funding from Astex and Genentech. GJR has consulted or served on advisory boards or data and safety monitoring committees for AbbVie, Actiunuim, Agios, Amgen, Astellas, AstraZeneca, Bluebird, Blueprint, Bluebird Bio, BMS, Catamaran, Celgene, Daiichi, GSK, Helsinn, Janssen, Jasper, Jazz, Mesoblast, Novartis, Pfizer, Roche, Syndax, Takeda, and Trovagene and received research support from Janssen. LB has consulted for Novartis. RB has received honoraria and research funding from BMS and Taiho; received research funding from Takeda; and received honoraria from AbbVie. BL has received honoraria and served on advisory committees and speakers bureaus for AbbVie, Amgen, and BMS and consulted for, received honoraria, and served on advisory committees and speakers bureaus for Novartis and Pfizer. AS has consulted for and received research funding from Kymera, consulted for Janssen and Rigel, and received honoraria from National Association for Continuing Education. K-HD, AO, and HNK are employed by Astex. YH and MA were employed in the past 24 months by and consult for Astex. MRS has consulted and served on advisory boards for and received travel expenses from BMS, CTI, Forma, Geron, GSK, and Taiho; received travel expenses from Astex; is an equity holder in and served on advisory committees for Karyopharm and Ryvu; and received research funding from ALX Oncology, Astex, Incyte, and Takeda., (Copyright © 2024 Elsevier Ltd. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
28. Use of tamibarotene, a potent and selective RARα agonist, in combination with azacitidine in patients with relapsed and refractory AML with RARA gene overexpression.
- Author
-
Stein EM, de Botton S, Cluzeau T, Pigneux A, Liesveld JL, Cook RJ, Rousselot P, Rizzieri DA, Braun T, Roboz GJ, Lebon D, Heiblig M, Baker K, Volkert A, Paul S, Rajagopal N, Roth DA, Kelly M, and Peterlin P
- Subjects
- Humans, Azacitidine therapeutic use, Retinoic Acid Receptor alpha, Antineoplastic Combined Chemotherapy Protocols adverse effects, Myelodysplastic Syndromes genetics, Leukemia, Myeloid, Acute diagnosis, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics
- Abstract
Tamibarotene-based therapy is a novel targeted approach for the treatment of relapsed/refractory (R/R) acute myeloid leukemia (AML) with retinoic acid receptor alpha ( RARA ) gene overexpression. Approximately, 50% of higher-risk myelodysplastic syndrome (MDS) patients and approximately 30% of AML patients are positive for RARA overexpression using a blood-based biomarker test that measures RARA expression in peripheral blasts. A phase 2 study investigating the activity of tamibarotene in patients with RARA overexpression was conducted in patients with AML and MDS (NCT02807558). In 28 patients with R/R AML and RARA overexpression treated with tamibarotene in combination with azacitidine, the median overall survival was 5.9 months. In 21 response-evaluable patients, the complete remission/complete remission with incomplete hematologic recovery (CR/CRi) rate was 19%, and median time to initial CR/CRi was 1.2 months. The favorable safety profile and preliminary clinical activity support the development of combination therapies with tamibarotene in myeloid malignancies with RARA overexpression.
- Published
- 2023
- Full Text
- View/download PDF
29. Measurable residual disease monitoring in patients with acute myeloid leukemia treated with lower-intensity therapy: Roadmap from an ELN-DAVID expert panel.
- Author
-
Ravandi F, Cloos J, Buccisano F, Dillon R, Döhner K, Freeman SD, Hourigan CS, Ossenkoppele GJ, Roboz GJ, Subklewe M, Thiede C, Arnhardt I, Valk PJM, Venditti A, Wei AH, Walter RB, and Heuser M
- Subjects
- Humans, Prognosis, Treatment Outcome, Neoplasm, Residual diagnosis, Leukemia, Myeloid, Acute diagnosis, Leukemia, Myeloid, Acute drug therapy
- Abstract
With the availability of effective targeted agents, significant changes have occurred in the management of patients with acute myeloid leukemia (AML) over the past several years, particularly for those considered unfit for intensive chemotherapy. While testing for measurable residual disease (MRD) is now routinely performed in patients treated with intensive chemotherapy to refine prognosis and, possibly, inform treatment decision-making, its value in the context of lower-intensity regimens is unclear. As such regimens have gained in popularity and can be associated with higher response rates, the need to better define the role of MRD assessment and the appropriate time points and assays used for this purpose has increased. This report outlines a roadmap for MRD testing in patients with AML treated with lower-intensity regimens. Experts from the European LeukemiaNet (ELN)-DAVID AML MRD working group reviewed all available data to propose a framework for MRD testing in future trials and clinical practice. A Delphi poll served to optimize consensus. Establishment of uniform standards for MRD assessments in lower-intensity regimens used in treating patients with AML is clinically relevant and important for optimizing testing and, ultimately, improving treatment outcomes of these patients., (© 2023 Wiley Periodicals LLC.)
- Published
- 2023
- Full Text
- View/download PDF
30. Classification, risk stratification and response assessment in myelodysplastic syndromes/neoplasms (MDS): A state-of-the-art report on behalf of the International Consortium for MDS (icMDS).
- Author
-
Stahl M, Bewersdorf JP, Xie Z, Porta MGD, Komrokji R, Xu ML, Abdel-Wahab O, Taylor J, Steensma DP, Starczynowski DT, Sekeres MA, Sanz G, Sallman DA, Roboz GJ, Platzbecker U, Patnaik MM, Padron E, Odenike O, Nimer SD, Nazha A, Majeti R, Loghavi S, Little RF, List AF, Kim TK, Hourigan CS, Hasserjian RP, Halene S, Griffiths EA, Gore SD, Greenberg P, Figueroa ME, Fenaux P, Efficace F, DeZern AE, Daver NG, Churpek JE, Carraway HE, Buckstein R, Brunner AM, Boultwood J, Borate U, Bejar R, Bennett JM, Wei AH, Santini V, Savona MR, and Zeidan AM
- Subjects
- Humans, Risk Assessment, Quality of Life, Prognosis, Neoplasms, Myelodysplastic Syndromes diagnosis, Myelodysplastic Syndromes therapy
- Abstract
The guidelines for classification, prognostication, and response assessment of myelodysplastic syndromes/neoplasms (MDS) have all recently been updated. In this report on behalf of the International Consortium for MDS (icMDS) we summarize these developments. We first critically examine the updated World Health Organization (WHO) classification and the International Consensus Classification (ICC) of MDS. We then compare traditional and molecularly based risk MDS risk assessment tools. Lastly, we discuss limitations of criteria in measuring therapeutic benefit and highlight how the International Working Group (IWG) 2018 and 2023 response criteria addressed these deficiencies and are endorsed by the icMDS. We also address the importance of patient centered care by discussing the value of quality-of-life assessment. We hope that the reader of this review will have a better understanding of how to classify MDS, predict clinical outcomes and evaluate therapeutic outcomes., Competing Interests: Declaration of Competing Interest Maximilian Stahl consulted for Curis Oncology and Boston Consulting; served on the advisory board for Novartis and Kymera, GSK, Rigel, Sierra Oncology; and participated in GME activity for Novartis, Curis Oncology, Haymarket Media and Clinical care options (CCO). Elizabeth A. Griffiths has received honoraria for advisory board membership from AbbVie, Alexion Pharmaceuticals, Apellis, Celgene/BMS, CTI Biopharma, Genentech, Novartis, Picnic Health, Takeda Oncology, Taiho Oncology. EAG has received research funding from Astex Pharmaceuticals, AstraZeneca Rare Disease, Alexion Pharmaceuticals, Apellis Pharmaceuticals, Blueprint Medicines, Genentech Inc., and honoraria for CME activities from Physicians Educational Resource, MediComWorldwide, American Society of Hematology, AAMDS International Foundation. Ravindra Majeti is on the Advisory Boards of Kodikaz Therapeutic Solutions, Syros Pharmaceuticals, TenSixteen Bio, Roche, and Cullgen Inc. and is an inventor on several patents related to CD47 cancer immunotherapy licensed to Gilead Sciences. R.M. receives research support from Gilead Sciences. Ravindra Majeti. is a co-founder and equity holder of Pheast Therapeutics, MyeloGene, and Orbital Therapeutics. Stephanie Halene consulted for Forma Therapeutics. Daniel T. Starczynowski is a consultant and received research funding from Kymera Therapeutics, Kurome Therapeutics, Captor Therapeutics, and Tolero Therapeutics. Daniel T. Starczynowski has equity in Kurome Therapeutics. Gail Roboz: Consultancy: Abbvie, Amgen, Argenx, Astra Zeneca, Bluebird Bio, Blueprint Medicines, Bristol-Myers Squibb, Caribou Biosciences, Celgene, Daiichi Sankyo, Ellipses Pharma, GlaxoSmithKline, Janssen, Jasper Pharmaceuticals, Jazz Pharmaceuticals, Molecular Partners, Novartis, Pfizer, Rigel, Roche, Syndax, Takeda (IRC Chair), Telix Pharma Research support: Janssen. David A. Sallman served on the advisory board of Aprea, AvenCell, BlueBird Bio, BMS, Intellia, Kite, Novartis, Shattuck Labs, Servier, Syndax. David A. Sallman served as a consultant for AbbVie, Magenta, Molecular Partners AG, Takeda and on the speakers' bureau for BMS, Incyte, Servier; David A. Sallman received research funding from Aprea, Jazz. Mrinal Patnaik received research funding from Kura Oncology and StemLine Pharmaceuticals. Andrew Brunner received consulting or advisory board honoraria from Novartis, Acceleron, Agios, Abbvie, Takeda, Celgene/BMS, Keros Therapeutics, Taiho, Gilead. Andrew Brunner has research support from the NIH SPORE in Myeloid Malignancies, and from the Edward P. Evans Foundation. Tae Kon Kim received research funding from Nextcure and is a consultant for Agenus. Alan List is employed by and has equity in Precision BioSciences, and has served as a consultant for Halia Therapeutics, CTI Biopharma, Aileron. Naval Daver has received research funding from Daiichi-Sankyo, Bristol-Myers Squibb, Pfizer, Gilead, Sevier, Genentech, Astellas, Daiichi-Sankyo, Abbvie, Hanmi, Trovagene, FATE therapeutics, Amgen, Novimmune, Glycomimetics, Trillium, and ImmunoGen and has served in a consulting or advisory role for Daiichi-Sankyo, Bristol-Myers Squibb, Arog, Pfizer, Novartis, Jazz, Celgene, AbbVie, Astellas, Genentech, Immunogen, Servier, Syndax, Trillium, Gilead, Amgen, Shattuck labs, and Agios. Guillermo Sanz received honoraria, advisory board membership or consultation fees from AbbVie, BMS, ExCellThera, Novartis, Roche, and Takeda and participated in sponsored speaker's bureau for BMS, Novartis, and Takeda. Mikkael A. Sekeres has served on advisory boards for BMS, Novartis, Kurome, and Gilead. Pierre Fenaux received research funding from BMS, Abbvie, Jazz Pharmaceuticals, Novartis, and Janssen. Pierre Fenaux had a consultancy with and received honoraria from BMS, Abbvie, Jazz Pharmaceuticals, and Novartis. Fabio Efficace had a consultancy or advisory role for AbbVie, Incyte, Janssen, and Syros, outside the submitted work. Omar Abdel-Wahab has served as a consultant for H3B Biomedicine, Foundation Medicine Inc., Merck, Prelude Therapeutics, and Janssen, and is on the Scientific Advisory Board of Envisagenics Inc., AIChemy, Harmonic Discovery Inc., and Pfizer Boulder; Omar Abdel-Wahab has received prior research funding from H3B Biomedicine and LOXO Oncology unrelated to the current manuscript. Andrew H.Wei has served on advisory boards for Novartis, Astra Zeneca, Astellas, Janssen, Jazz, Amgen, Roche, Pfizer, Abbvie, Servier, Gilead, BMS, Shoreline, Macrogenics, Novartis and Agios; receives research funding to the Institution from Novartis, Abbvie, Servier, Janssen, BMS, Syndax, Astex, Astra Zeneca, Amgen; serves on speaker's bureaus for Abbvie, Novartis, BMS, Servier, Astellas; Andrew H.Wei is an employee of the Walter and Eliza Hall Institute (WEHI). WEHI receives milestone and royalty payments related to the development of Venetoclax. Current and past employees of Walter and Eliza Hall Institute may be eligible for financial benefits related to these payments. Andrew H. Wei receives such a financial benefit. Amer M. Zeidan received research funding (institutional) from Celgene/BMS, Abbvie, Astex, Pfizer, Medimmune/AstraZeneca, Boehringer-Ingelheim, Cardiff oncology, Incyte, Takeda, Novartis, Aprea, and ADC Therapeutics. AMZ participated in advisory boards, and/or had a consultancy with and received honoraria from AbbVie, Otsuka, Pfizer, Celgene/BMS, Jazz, Incyte, Agios, Boehringer-Ingelheim, Novartis, Acceleron, Astellas, Daiichi Sankyo, Cardinal Health, Taiho, Seattle Genetics, BeyondSpring, Cardiff Oncology, Takeda, Ionis, Amgen, Janssen, Epizyme, Syndax, Gilead, Kura, Chiesi, ALX Oncology, BioCryst, Notable, Orum, and Tyme. AMZ served on clinical trial committees for Novartis, Abbvie, Gilead, BioCryst, Abbvie, ALX Oncology, Geron and Celgene/BMS. The National Heart, Lung, and Blood Institute receives research funding for the laboratory of Dr. Hourigan from Sellas and from the Foundation of the NIH AML MRD Biomarkers Consortium. All other authors have no conflicts of interest to disclose., (Copyright © 2023 Elsevier Ltd. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
31. Correction: Clinical impact of the genomic landscape and leukemogenic trajectories in non-intensively treated elderly acute myeloid leukemia patients.
- Author
-
Jahn E, Saadati M, Fenaux P, Gobbi M, Roboz GJ, Bullinger L, Lutsik P, Riedel A, Plass C, Jahn N, Walter C, Holzmann K, Hao Y, Naim S, Schreck N, Krzykalla J, Benner A, Keer HN, Azab M, Döhner K, and Döhner H
- Published
- 2023
- Full Text
- View/download PDF
32. Clinical impact of the genomic landscape and leukemogenic trajectories in non-intensively treated elderly acute myeloid leukemia patients.
- Author
-
Jahn E, Saadati M, Fenaux P, Gobbi M, Roboz GJ, Bullinger L, Lutsik P, Riedel A, Plass C, Jahn N, Walter C, Holzmann K, Hao Y, Naim S, Schreck N, Krzykalla J, Benner A, Keer HN, Azab M, Döhner K, and Döhner H
- Subjects
- Humans, Aged, Prospective Studies, Mutation, Prognosis, Genomics, Transcription Factors genetics, fms-Like Tyrosine Kinase 3 genetics, fms-Like Tyrosine Kinase 3 therapeutic use, Nucleophosmin, Leukemia, Myeloid, Acute therapy, Leukemia, Myeloid, Acute drug therapy
- Abstract
To characterize the genomic landscape and leukemogenic pathways of older, newly diagnosed, non-intensively treated patients with AML and to study the clinical implications, comprehensive genetics analyses were performed including targeted DNA sequencing of 263 genes in 604 patients treated in a prospective Phase III clinical trial. Leukemic trajectories were delineated using oncogenetic tree modeling and hierarchical clustering, and prognostic groups were derived from multivariable Cox regression models. Clonal hematopoiesis-related genes (ASXL1, TET2, SRSF2, DNMT3A) were most frequently mutated. The oncogenetic modeling algorithm produced a tree with five branches with ASXL1, DDX41, DNMT3A, TET2, and TP53 emanating from the root suggesting leukemia-initiating events which gave rise to further subbranches with distinct subclones. Unsupervised clustering mirrored the genetic groups identified by the tree model. Multivariable analysis identified FLT3 internal tandem duplications (ITD), SRSF2, and TP53 mutations as poor prognostic factors, while DDX41 mutations exerted an exceptionally favorable effect. Subsequent backwards elimination based on the Akaike information criterion delineated three genetic risk groups: DDX41 mutations (favorable-risk), DDX41
wildtype /FLT3-ITDneg /TP53wildtype (intermediate-risk), and FLT3-ITD or TP53 mutations (high-risk). Our data identified distinct trajectories of leukemia development in older AML patients and provide a basis for a clinically meaningful genetic outcome stratification for patients receiving less intensive therapies., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
33. Survival outcomes with oral azacitidine maintenance in patients with acute myeloid leukemia in remission by receipt of initial chemotherapy: subgroup analyses from the phase III QUAZAR AML-001 trial.
- Author
-
Wei AH, Roboz GJ, Dombret H, Dohner H, Schuh AC, Montesinos P, Selleslag D, Bondarenko SN, Prebet T, Lai Y, Skikne B, Beach CL, and Ravandi F
- Subjects
- Humans, Antimetabolites, Antineoplastic therapeutic use, Remission Induction, Azacitidine therapeutic use, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute mortality
- Published
- 2023
- Full Text
- View/download PDF
34. A phase 1b study of venetoclax and alvocidib in patients with relapsed/refractory acute myeloid leukemia.
- Author
-
Jonas BA, Hou JZ, Roboz GJ, Alvares CL, Jeyakumar D, Edwards JR, Erba HP, Kelly RJ, Röllig C, Fiedler W, Brackman D, Siddani SR, Chyla B, Hilger-Rolfe J, and Watts JM
- Subjects
- Humans, Bridged Bicyclo Compounds, Heterocyclic therapeutic use, Antineoplastic Combined Chemotherapy Protocols adverse effects, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute etiology
- Abstract
Relapsed/refractory (R/R) Acute Myeloid Leukemia (AML) is a genetically complex and heterogeneous disease with a poor prognosis and limited treatment options. Thus, there is an urgent need to develop therapeutic combinations to overcome drug resistance in AML. This open-label, multicenter, international, phase 1b study evaluated the safety, efficacy, and pharmacokinetics of venetoclax in combination with alvocidib in patients with R/R AML. Patients were treated with escalating doses of venetoclax (400, 600, and 800 mg QD, orally, days 1-28) and alvocidib (45 and 60 mg/m
2 , intravenously, days 1-3) in 28-day cycles. The combination was found to be safe and tolerable, with no maximum tolerated dose reached. Drug-related Grade ≥3 adverse events were reported in 23 (65.7%) for venetoclax and 24 (68.6%) for alvocidib. No drug-related AEs were fatal. Gastrointestinal toxicities, including diarrhea, nausea, and vomiting were notable and frequent; otherwise, the toxicities reported were consistent with the safety profile of both agents. The response rate was modest (complete remission [CR] + incomplete CR [CRi], 11.4%; CR + CRi + partial response rate + morphologic leukemia-free state, 20%). There was no change in alvocidib pharmacokinetics with increasing doses of venetoclax. However, when venetoclax was administered with alvocidib, AUC24 and Cmax decreased by 18% and 19%, respectively. A recommended phase 2 dose was not established due to lack of meaningful increase in efficacy across all cohorts compared to what was previously observed with each agent alone. Future studies could consider the role of the sequence, dosing, and the use of a more selective MCL1 inhibitor for the R/R AML population., (© 2023 AbbVie Inc. Hematological Oncology published by John Wiley & Sons Ltd.)- Published
- 2023
- Full Text
- View/download PDF
35. Guadecitabine vs treatment choice in newly diagnosed acute myeloid leukemia: a global phase 3 randomized study.
- Author
-
Fenaux P, Gobbi M, Kropf PL, Issa JJ, Roboz GJ, Mayer J, Krauter J, Robak T, Kantarjian H, Novak J, Jedrzejczak WW, Thomas X, Ojeda-Uribe M, Miyazaki Y, Min YH, Yeh SP, Brandwein J, Gercheva-Kyuchukova L, Demeter J, Griffiths E, Yee K, Döhner K, Hao Y, Keer H, Azab M, and Döhner H
- Subjects
- Humans, Treatment Outcome, Cytarabine adverse effects, Azacitidine adverse effects, Leukemia, Myeloid, Acute diagnosis, Leukemia, Myeloid, Acute drug therapy
- Abstract
This phase 3 study evaluated the efficacy and safety of the new hypomethylating agent guadecitabine (n = 408) vs a preselected treatment choice (TC; n = 407) of azacitidine, decitabine, or low-dose cytarabine in patients with acute myeloid leukemia unfit to receive intensive induction chemotherapy. Half of the patients (50%) had poor Eastern Cooperative Oncology Group Performance Status (2-3). The coprimary end points were complete remission (19% and 17% of patients for guadecitabine and TC, respectively [stratified P = .48]) and overall survival (median survival 7.1 and 8.5 months for guadecitabine and TC, respectively [hazard ratio, 0.97; 95% confidence interval, 0.83-1.14; stratified log-rank P = .73]). One- and 2-year survival estimates were 37% and 18% for guadecitabine and 36% and 14% for TC, respectively. A large proportion of patients (42%) received <4 cycles of treatment in both the arms. In a post hoc analysis of patients who received ≥4 treatment cycles, guadecitabine was associated with longer median survival vs TC (15.6 vs 13.0 months [hazard ratio, 0.78; 95% confidence interval, 0.64-0.96; log-rank P = .02]). There was no significant difference in the proportion of patients with grade ≥3 adverse events (AEs) between guadecitabine (92%) and TC (88%); however, grade ≥3 AEs of febrile neutropenia, neutropenia, and pneumonia were higher with guadecitabine. In conclusion, no significant difference was observed in the efficacy of guadecitabine and TC in the overall population. This trial was registered at www.clinicaltrials.gov as #NCT02348489., (© 2023 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
36. Correction: Integrative analysis identifies an older female-linked AML patient group with better risk in ECOG-ACRIN Cancer Research Group's clinical trial E3999.
- Author
-
Rapaport F, Seier K, Neelamraju Y, Hassane D, Baslan T, Gildea DT, Haddox S, Lee T, Murdock HM, Sheridan C, Thurmond A, Wang L, Carroll M, Cripe LD, Fernandez H, Mason CE, Paietta E, Roboz GJ, Sun Z, Tallman MS, Zhang Y, Gönen M, Levine R, Melnick AM, Kleppe M, and Garrett-Bakelman FE
- Published
- 2023
- Full Text
- View/download PDF
37. Current landscape of translational and clinical research in myelodysplastic syndromes/neoplasms (MDS): Proceedings from the 1 st International Workshop on MDS (iwMDS) Of the International Consortium for MDS (icMDS).
- Author
-
Bewersdorf JP, Xie Z, Bejar R, Borate U, Boultwood J, Brunner AM, Buckstein R, Carraway HE, Churpek JE, Daver NG, Porta MGD, DeZern AE, Fenaux P, Figueroa ME, Gore SD, Griffiths EA, Halene S, Hasserjian RP, Hourigan CS, Kim TK, Komrokji R, Kuchroo VK, List AF, Loghavi S, Majeti R, Odenike O, Patnaik MM, Platzbecker U, Roboz GJ, Sallman DA, Santini V, Sanz G, Sekeres MA, Stahl M, Starczynowski DT, Steensma DP, Taylor J, Abdel-Wahab O, Xu ML, Savona MR, Wei AH, and Zeidan AM
- Subjects
- Animals, Humans, Epigenomics, Cell- and Tissue-Based Therapy, Protein Processing, Post-Translational, Neoplasms, Myelodysplastic Syndromes etiology, Myelodysplastic Syndromes therapy
- Abstract
Biological events that contribute to the pathogenesis of myelodysplastic syndromes/neoplasms (MDS) are becoming increasingly characterized and are being translated into rationally designed therapeutic strategies. Herein, we provide updates from the first International Workshop on MDS (iwMDS) of the International Consortium for MDS (icMDS) detailing recent advances in understanding the genetic landscape of MDS, including germline predisposition, epigenetic and immune dysregulation, the complexities of clonal hematopoiesis progression to MDS, as well as novel animal models of the disease. Connected to this progress is the development of novel therapies targeting specific molecular alterations, the innate immune system, and immune checkpoint inhibitors. While some of these agents have entered clinical trials (e.g., splicing modulators, IRAK1/4 inhibitors, anti-CD47 and anti-TIM3 antibodies, and cellular therapies), none have been approved for MDS. Additional preclinical and clinical work is needed to develop a truly individualized approach to the care of MDS patients., Competing Interests: Declaration of Competing Interest Maximilian Stahl consulted for Curis Oncology and Boston Consulting; served on the advisory board for Novartis and Kymera; and participated in GME activity for Novartis, Curis Oncology, Haymarket Media and Clinical care options (CCO). Elizabeth A. Griffiths has received honoraria for advisory board membership from AbbVie, Alexion Pharmaceuticals, Apellis, Celgene/BMS, CTI Biopharma, Genentech, Novartis, Picnic Health, Takeda Oncology, Taiho Oncology. EAG has received research funding from Astex Pharmaceuticals, AstraZeneca Rare Disease, Alexion Pharmaceuticals, Apellis Pharmaceuticals, Blueprint Medicines, Genentech Inc., and honoraria for CME activities from Physicians Educational Resource, MediComWorldwide, American Society of Hematology, AAMDS International Foundation. Ravindra Majeti is on the Advisory Boards of Kodikaz Therapeutic Solutions, Syros Pharmaceuticals, TenSixteen Bio, Roche, and Cullgen Inc. and is an inventor on a number of patents related to CD47 cancer immunotherapy licensed to Gilead Sciences. R.M. receives research support from Gilead Sciences. Ravindra Majeti. is a co-founder and equity holder of Pheast Therapeutics, MyeloGene, and Orbital Therapeutics. Stephanie Halene consulted for Forma Therapeutics. Daniel T. Starczynowski is a consultant and received research funding from Kymera Therapeutics, Kurome Therapeutics, Captor Therapeutics, and Tolero Therapeutics. Daniel T. Starczynowski has equity in Kurome Therapeutics. David A. Sallman served on the advisory board of Aprea, AvenCell, BlueBird Bio, BMS, Intellia, Kite, Novartis, Shattuck Labs, Servier, Syndax. David A. Sallman served as a consultant for AbbVie, Magenta, Molecular Partners AG, Takeda and on the speakers' bureau for BMS, Incyte, Servier; David A. Sallman received research funding from Aprea, Jazz. Mrinal Patnaik received research funding from Kura Oncology and StemLine Pharmaceuticals. Andrew Brunner received consulting or advisory board honoraria from Novartis, Acceleron, Agios, Abbvie, Takeda, Celgene/BMS, Keros Therapeutics, Taiho, Gilead. Andrew Brunner has research support from the NIH SPORE in Myeloid Malignancies, and from the Edward P. Evans Foundation. Tae Kon Kim received research funding from Nextcure and is a consultant for Agenus. Alan List is employed by and has equity in Precision BioSciences, and has served as a consultant for Halia Therapeutics, CTI Biopharma, Aileron. Naval Daver has received research funding from Daiichi-Sankyo, Bristol-Myers Squibb, Pfizer, Gilead, Sevier, Genentech, Astellas, Daiichi-Sankyo, Abbvie, Hanmi, Trovagene, FATE therapeutics, Amgen, Novimmune, Glycomimetics, Trillium, and ImmunoGen and has served in a consulting or advisory role for Daiichi-Sankyo, Bristol-Myers Squibb, Arog, Pfizer, Novartis, Jazz, Celgene, AbbVie, Astellas, Genentech, Immunogen, Servier, Syndax, Trillium, Gilead, Amgen, Shattuck labs, and Agios. Guillermo Sanz received honoraria, advisory board membership or consultation fees from AbbVie, BMS, ExCellThera, Novartis, Roche, and Takeda and participated in sponsored speaker's bureau for BMS, Novartis, and Takeda. Mikkael A. Sekeres has served on advisory boards for BMS, Novartis, Kurome, and Gilead. Pierre Fenaux received research funding from BMS, Abbvie, Jazz Pharmaceuticals, Novartis, and Janssen. Pierre Fenaux had a consultancy with and received honoraria from BMS, Abbvie, Jazz Pharmaceuticals, and Novartis. Omar Abdel-Wahab has served as a consultant for H3B Biomedicine, Foundation Medicine Inc., Merck, Prelude Therapeutics, and Janssen, and is on the Scientific Advisory Board of Envisagenics Inc., AIChemy, Harmonic Discovery Inc., and Pfizer Boulder; Omar Abdel-Wahab has received prior research funding from H3B Biomedicine and LOXO Oncology unrelated to the current manuscript. Andrew H.Wei has served on advisory boards for Novartis, Astra Zeneca, Astellas, Janssen, Jazz, Amgen, Roche, Pfizer, Abbvie, Servier, Gilead, BMS, Shoreline, Macrogenics, Novartis and Agios; receives research funding to the Institution from Novartis, Abbvie, Servier, Janssen, BMS, Syndax, Astex, Astra Zeneca, Amgen; serves on speaker's bureaus for Abbvie, Novartis, BMS, Servier, Astellas; Andrew H.Wei is an employee of the Walter and Eliza Hall Institute (WEHI). WEHI receives milestone and royalty payments related to the development of Venetoclax. Current and past employees of Walter and Eliza Hall Institute may be eligible for financial benefits related to these payments. Andrew H.Wei receives such a financial benefit. Valeria Santini served in advisory boards from Abbvie, BMS, Geron, Gilead,Menarini, Novartis, Servier, Syros, ad received research support from BMS. Amer M. Zeidan received research funding (institutional) from Celgene/BMS, Abbvie, Astex, Pfizer, Medimmune/AstraZeneca, Boehringer-Ingelheim, Cardiff oncology, Incyte, Takeda, Novartis, Aprea, and ADC Therapeutics. AMZ participated in advisory boards, and/or had a consultancy with and received honoraria from AbbVie, Otsuka, Pfizer, Celgene/BMS, Jazz, Incyte, Agios, Boehringer-Ingelheim, Novartis, Acceleron, Astellas, Daiichi Sankyo, Cardinal Health, Taiho, Seattle Genetics, BeyondSpring, Cardiff Oncology, Takeda, Ionis, Amgen, Janssen, Epizyme, Syndax, Gilead, Kura, Chiesi, ALX Oncology, BioCryst, Notable, Orum, and Tyme. AMZ served on clinical trial committees for Novartis, Abbvie, Gilead, BioCryst, Abbvie, ALX Oncology, Geron and Celgene/BMS. The National Heart, Lung, and Blood Institute receives research funding for the laboratory of Dr. Hourigan from Sellas and from the Foundation of the NIH AML MRD Biomarkers Consortium. All other authors have no conflicts of interest to disclose., (Copyright © 2023 Elsevier Ltd. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
38. Transplantation Referral Patterns for Patients with Newly Diagnosed Higher-Risk Myelodysplastic Syndromes and Acute Myeloid Leukemia at Academic and Community Sites in the Connect® Myeloid Disease Registry: Potential Barriers to Care.
- Author
-
Tomlinson B, de Lima M, Cogle CR, Thompson MA, Grinblatt DL, Pollyea DA, Komrokji RS, Roboz GJ, Savona MR, Sekeres MA, Abedi M, Garcia-Manero G, Kurtin SE, Maciejewski JP, Patel JL, Revicki DA, George TI, Flick ED, Kiselev P, Louis CU, DeGutis IS, Nifenecker M, Erba HP, Steensma DP, and Scott BL
- Subjects
- Humans, Aged, Registries, Health Services Accessibility, Myelodysplastic Syndromes therapy, Leukemia, Myeloid, Acute therapy, Hematopoietic Stem Cell Transplantation
- Abstract
Hematopoietic stem cell transplantation (HCT) is indicated for patients with higher-risk (HR) myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML). Age, performance status, patient frailty, comorbidities, and nonclinical factors (eg, cost, distance to site) are all recognized as important clinical factors that can influence HCT referral patterns and patient outcomes; however, the proportion of eligible patients referred for HCT in routine clinical practice is largely unknown. This study aimed to assess patterns of consideration for HCT among patients with HR-MDS and AML enrolled in the Connect® Myeloid Disease Registry at community/government (CO/GOV)- or academic (AC)-based sites, as well as to identify factors associated with rates of transplantation referral. We assessed patterns of consideration for and completion of HCT in patients with HR-MDS and AML enrolled between December 12, 2013, and March 6, 2020, in the Connect Myeloid Disease Registry at 164 CO/GOV and AC sites. Registry sites recorded whether patients were considered for transplantation at baseline and at each follow-up visit. The following answers were possible: "considered potentially eligible," "not considered potentially eligible," or "not assessed." Sites also recorded whether patients subsequently underwent HCT at each follow-up visit. Rates of consideration for HCT between CO/GOV and AC sites were compared using multivariable logistic regression analysis with covariates for age and comorbidity. Among the 778 patients with HR-MDS or AML enrolled in the Connect Myeloid Disease Registry, patients at CO/GOV sites were less likely to be considered potentially eligible for HCT than patients at AC sites (27.9% versus 43.9%; P < .0001). Multivariable logistic regression analysis with factors for age (<65 versus ≥65 years) and ACE-27 comorbidity grade (<2 versus ≥2) showed that patients at CO/GOV sites were significantly less likely than those at AC sites to be considered potentially eligible for HCT (odds ratio, 1.6, 95% confidence interval, 1.1 to 2.4; P = .0155). Among patients considered eligible for HCT, 45.1% (65 of 144) of those at CO/GOV sites and 35.7% (41 of 115) of those at AC sites underwent transplantation (P = .12). Approximately one-half of all patients at CO/GOV (50.1%) and AC (45.4%) sites were not considered potentially eligible for HCT; the most common reasons were age at CO/GOV sites (71.5%) and comorbidities at AC sites (52.1%). Across all sites, 17.4% of patients were reported as not assessed (and thus not considered) for HCT by their treating physician (20.7% at CO/GOV sites and 10.7% at AC sites; P = .0005). These findings suggest that many patients with HR-MDS and AML who may be candidates for HCT are not receiving assessment or consideration for transplantation in clinical practice. In addition, treatment at CO/GOV sites and age remain significant barriers to ensuring that all potentially eligible patients are assessed for HCT., (Copyright © 2023 The American Society for Transplantation and Cellular Therapy. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
39. An agenda to advance research in myelodysplastic syndromes: a TOP 10 priority list from the first international workshop in MDS.
- Author
-
Stahl M, Abdel-Wahab O, Wei AH, Savona MR, Xu ML, Xie Z, Taylor J, Starczynowski D, Sanz GF, Sallman DA, Santini V, Roboz GJ, Patnaik MM, Padron E, Odenike O, Nazha A, Nimer SD, Majeti R, Little RF, Gore S, List AF, Kutchroo V, Komrokji RS, Kim TK, Kim N, Hourigan CS, Hasserjian RP, Halene S, Griffiths EA, Greenberg PL, Figueroa M, Fenaux P, Efficace F, DeZern AE, Della Porta MG, Daver NG, Churpek JE, Carraway HE, Brunner AM, Borate U, Bennett JM, Bejar R, Boultwood J, Loghavi S, Bewersdorf JP, Platzbecker U, Steensma DP, Sekeres MA, Buckstein RJ, and Zeidan AM
- Subjects
- Humans, Chromosome Aberrations, Myelodysplastic Syndromes diagnosis, Myelodysplastic Syndromes therapy
- Published
- 2023
- Full Text
- View/download PDF
40. Targeting RARA overexpression with tamibarotene, a potent and selective RARα agonist, is a novel approach in AML.
- Author
-
de Botton S, Cluzeau T, Vigil C, Cook RJ, Rousselot P, Rizzieri DA, Liesveld JL, Fenaux P, Braun T, Banos A, Jurcic JG, Sekeres MA, Savona MR, Roboz GJ, Bixby D, Madigan K, Volkert A, Stephens K, Kang-Fortner Q, Baker K, Paul S, McKeown M, Carulli J, Eaton M, Hodgson G, Fiore C, Kelly MJ, Roth DA, and Stein EM
- Subjects
- Humans, Retinoic Acid Receptor alpha, Azacitidine adverse effects, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute chemically induced, Myelodysplastic Syndromes drug therapy, Leukemia, Promyelocytic, Acute drug therapy
- Abstract
A superenhancer at the retinoic acid receptor alpha (RARA) gene is associated with RARA mRNA overexpression in ∼30% of non-acute promyelocytic leukemia acute myeloid leukemia (AML) and in ∼50% of myelodysplastic syndromes (MDS). RARA overexpression is an actionable target for treatment with tamibarotene, an oral potent and selective RARα agonist. Sensitivity to the RARα agonist tamibarotene was demonstrated in RARA-high but not RARA-low preclinical AML models. The combination of oral tamibarotene plus azacitidine was evaluated in a phase 2 clinical study in 51 newly diagnosed unfit patients with AML identified as RARA-positive (n = 22) or RARA-negative (n = 29) for RARA mRNA overexpression in peripheral blasts using a blood-based biomarker test. In 18 response-evaluable RARA-positive patients, complete remission (CR)/CR with incomplete hematologic recovery rate was 61%, CR rate was 50%, and time to initial composite CR was rapid at 1.2 months. Transfusion independence was attained by 72% of RARA-positive patients. In contrast, 28 response-evaluable RARA-negative patients had response rates that were consistent with azacitidine monotherapy. Tamibarotene in combination with azacitidine was well tolerated. The majority of nonhematologic adverse events were low grade and hematologic adverse events were comparable to single-agent azacitidine, demonstrating that there was no additional myelosuppression when tamibarotene was combined with azacitidine. These results support further evaluation of tamibarotene-based treatment strategies in patients with AML or MDS with RARA overexpression to provide a targeted approach with the goal of improving patient outcomes. This trial was registered at www.clinicaltrials.gov as #NCT02807558., (© 2023 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
41. Long-term survival with oral azacitidine for patients with acute myeloid leukemia in first remission after chemotherapy: Updated results from the randomized, placebo-controlled, phase 3 QUAZAR AML-001 trial.
- Author
-
Wei AH, Döhner H, Sayar H, Ravandi F, Montesinos P, Dombret H, Selleslag D, Porkka K, Jang JH, Skikne B, Beach CL, Prebet T, Zhang G, Risueño A, Ugidos M, See WL, Menezes D, and Roboz GJ
- Subjects
- Humans, Antimetabolites, Antineoplastic therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Azacitidine therapeutic use, Leukemia, Myeloid, Acute drug therapy
- Published
- 2023
- Full Text
- View/download PDF
42. Eprenetapopt combined with venetoclax and azacitidine in TP53-mutated acute myeloid leukaemia: a phase 1, dose-finding and expansion study.
- Author
-
Garcia-Manero G, Goldberg AD, Winer ES, Altman JK, Fathi AT, Odenike O, Roboz GJ, Sweet K, Miller C, Wennborg A, Hickman DK, Kanagal-Shamanna R, Kantarjian H, Lancet J, Komrokji R, Attar EC, and Sallman DA
- Subjects
- Aged, Female, Humans, Male, Antineoplastic Combined Chemotherapy Protocols adverse effects, Azacitidine adverse effects, Treatment Outcome, Tumor Suppressor Protein p53 genetics, Middle Aged, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Thrombocytopenia drug therapy
- Abstract
Background: TP53-mutated acute myeloid leukaemia is associated with poor outcomes. Eprenetapopt (APR-246) is a first-in-class, small-molecule p53 reactivator. We aimed to evaluate the combination of eprenetapopt and venetoclax with or without azacitidine in patients with TP53-mutated acute myeloid leukaemia., Methods: This phase 1, multicentre, open-label, dose-finding and cohort expansion study was done at eight academic research hospitals in the USA. Inclusion criteria were age of at least 18 years; at least one pathogenic TP53 mutation; treatment-naive acute myeloid leukaemia according to the 2016 WHO classification; an ECOG performance status of 0-2; and a life expectancy of at least 12 weeks. In dose-finding cohort 1 patients received previous therapy with hypomethylating agents for myelodysplastic syndromes. In dose-finding cohort 2, previous use of hypomethylating agents was not permitted. Treatment cycles were 28 days. Patients in cohort 1 received intravenous eprenetapopt 4·5 g/day on days 1-4 and oral venetoclax 400 mg/day on days 1-28; those in cohort 2 also received subcutaneous or intravenous azacitidine 75 mg/m
2 on days 1-7. The expansion part of the study proceeded with patients enrolled as in cohort 2. Primary endpoints were safety in all cohorts (assessed in patients receiving at least one dose of assigned treatment) and complete response in the expansion cohort (assessed in patients who completed at least one treatment cycle and had at least one post-treatment clinical response assessment). The trial is registered with ClinicalTrials.gov, NCT04214860, and is complete., Findings: Between Jan 3, 2020, and July 22, 2021, 49 patients were enrolled across all cohorts. Six patients were initially enrolled into each of dose-finding cohorts 1 and 2; after no dose-limiting toxicities were observed, cohort 2 was expanded to enrol an additional 37 patients. The median age was 67 years (IQR 59-73). 24 (49%) of 49 patients were female and 25 (51%) male, and 40 (82%) were White. At data cutoff (Oct 1, 2021), the median length of follow-up was 9·5 months (IQR 6·1-11·5). No dose-limiting toxicities were recorded and the recommended phase 2 dose for eprenetapopt combinations was 4·5 g/day on days 1-4. Across all patients, adverse events of grade 3 or worse occurring in at least 20% of patients were febrile neutropenia (23 [47%] of 49 patients), thrombocytopenia (18 [37%] patients), leukopenia (12 [25%] patients), and anaemia (11 [22%] patients). Treatment-related serious adverse events occurred in 13 (27%) of 49 patients and there was one (2%) treatment-related death (sepsis). 25 (64%, 95% CI 47-79) of 39 patients had an overall response with eprenetapopt and venetoclax with azacytidine; 15 (38%, 23-55) had a complete response., Interpretation: Eprenetapopt and venetoclax with azacitidine had an acceptable safety profile and encouraging activity, supporting further frontline evaluation of this combination in the treatment of TP53-mutated acute myeloid leukaemia., Funding: Aprea Therapeutics., Competing Interests: Declaration of interests GG-M reports research funding from Aprea Therapeutics. ADG reports research funding from Aprea Therapeutics; consulting fees from AbbVie, Aptose Biosciences, Astellas Pharma, Daiichi Sankyo, and Genentech; honoraria from AbbVie and Dava Oncology; and advisory board membership for AbbVie. ESW reports consulting fees from Pfizer, Jazz Pharmaceuticals, and Takeda. JKA reports research funding from Astellas Pharma and OPEN Health Group. ATF reports grants from Agios Pharmaceuticals/Servier, Celgene/Bristol-Myers Squibb (BMS), and AbbVie; and consulting fees from Pfizer, Trillium Health Partners, AbbVie, Kura Oncology, Blueprint Medicines, Genentech, Novartis, Trovagene, Agios Pharmaceuticals/Servier, Celgene/BMS, MorphoSys, Kite Pharma, Foghorn Therapeutics, Takeda, Amgen, Seattle Genetics, NewLink Genetics, Forty Seven, Ipsen, ImmunoGen, Mablytics, PureTech Health, Forma Therapeutics, Daiichi Sankyo, EnClear Therapies, and Astellas Pharma. OO reports research funding from Aprea Therapeutics; grants from AbbVie, Agios Pharmaceuticals, Astex, AstraZeneca, BMS, Celgene, CTI BioPharma, Daiichi Sankyo, Incyte, Janssen Pharmaceuticals, Kartos Therapeutics, Loxo Oncology, Novartis, NS Pharma, and Oncotherapy Sciences; advisory board membership for BMS, Celgene, Novartis, Taiho Pharmaceutical, and Kymera Therapeutics; membership of a data safety monitoring board for Threadwell Therapeutics; and leadership as Vice Chair of the medical advisory board of the Aplastic Anemia and MDS International Foundation. Kxs consulting fees from Astellas Pharma, BMS, Gilead Sciences, and Novartis and advisory board membership for BerGenBio, Curis, and Mablytics. GJR reports research funding from Aprea Therapeutics; grants from Janssen Pharmaceuticals; and consulting fees from Actinium Pharmaceuticals, Agios Pharmaceuticals, Amgen, Astellas Pharma, AstraZeneca, BMS, Blueprint Medicines, bluebird bio, Catamaran Bio, Celgene, Daiichi Sankyo, GlaxoSmithKline, Helsinn, Janssen Pharmaceuticals, Jasper Therapeutics, Jazz Pharmaceuticals, Mesoblast, Novartis, Pfizer, Roche, Syndax Pharmaceuticals, Takeda, and Trovagene. JL reports consulting fees from AbbVie, Agios Pharmaceuticals, Astellas Pharma, BerGenBio, Boxer Capital, Celgene, Daiichi Sankyo, The Dedham Group, ElevateBio, Jasper Therapeutics, Jazz Pharmaceuticals, Millenium Pharmaceuticals, Novartis, and Servier. DKH, AW, and ECA report employment with Aprea Therapeutics. HK reports grants from AbbVie, Amgen, Ascentage Pharma, BMS, Daiichi Sankyo, ImmunoGen, Jazz Pharmaceuticals, and Novartis and honoraria from AbbVie, Amgen, Amphista Therapeutics, Ascentage Pharma, Astellas Pharma, Biologix Pharma, Curis, Ipsen Biopharmaceuticals, KAHR Medical, Labcorp, Novartis, Pfizer, Shenzhen TargetRx, Stemline Therapeutics, and Takeda. DAS reports research funding from Aprea Therapeutics and Jazz Pharmaceuticals; consulting fees from AbbVie, Takeda, Zentalis Pharmaceuticals, and Molecular Partners; speakers bureaus from BMS and Incyte; and advisory board membership for AvenCell, Jasper Therapeutics, bluebird bio, BMS, Shattuck Labs, Intellia Therapeutics, Novartis, Gilead Sciences, Janssen Pharmaceuticals, Curis, Syndax Pharmaceuticals, Agios Pharmaceuticals, and Servier. All other authors declare no competing interests., (Copyright © 2023 The Author(s). Published by Elsevier Ltd. Published by Elsevier Ltd.. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
43. Venetoclax and idasanutlin in relapsed/refractory AML: a nonrandomized, open-label phase 1b trial.
- Author
-
Daver NG, Dail M, Garcia JS, Jonas BA, Yee KWL, Kelly KR, Vey N, Assouline S, Roboz GJ, Paolini S, Pollyea DA, Tafuri A, Brandwein JM, Pigneux A, Powell BL, Fenaux P, Olin RL, Visani G, Martinelli G, Onishi M, Wang J, Huang W, Green C, Ott MG, Hong WJ, Konopleva MY, and Andreeff M
- Subjects
- Humans, Core Binding Factor Alpha 2 Subunit, Bridged Bicyclo Compounds, Heterocyclic adverse effects, Antineoplastic Combined Chemotherapy Protocols adverse effects, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Antineoplastic Agents therapeutic use
- Abstract
This phase 1b trial (NCT02670044) evaluated venetoclax-idasanutlin in patients with relapsed/refractory (R/R) acute myeloid leukemia (AML) ineligible for cytotoxic chemotherapy. Two-dimensional dose escalation (DE, n = 50) was performed for venetoclax daily with idasanutlin on days 1 to 5 in 28-day cycles, followed by dosing schedule optimization (n = 6) to evaluate reduced venetoclax schedules (21-/14-day dosing). Common adverse events (occurring in ≥40% of patients) included diarrhea (87.3% of patients), nausea (74.5%), vomiting (52.7%), hypokalemia (50.9%), and febrile neutropenia (45.5%). During DE, across all doses, composite complete remission (CRc; CR + CR with incomplete blood count recovery + CR with incomplete platelet count recovery) rate was 26.0% and morphologic leukemia-free state (MLFS) rate was 12%. For anticipated recommended phase 2 doses (venetoclax 600 mg + idasanutlin 150 mg; venetoclax 600 mg + idasanutlin 200 mg), the combined CRc rate was 34.3% and the MLFS rate was 14.3%. Pretreatment IDH1/2 and RUNX1 mutations were associated with higher CRc rates (50.0% and 45.0%, respectively). CRc rate in patients with TP53 mutations was 20.0%, with responses noted among those with co-occurring IDH and RUNX1 mutations. In 12 out of 36 evaluable patients, 25 emergent TP53 mutations were observed; 22 were present at baseline with low TP53 variant allele frequency (median 0.0095% [range, 0.0006-0.4]). Venetoclax-idasanutlin showed manageable safety and encouraging efficacy in unfit patients with R/R AML. IDH1/2 and RUNX1 mutations were associated with venetoclax-idasanutlin sensitivity, even in some patients with co-occurring TP53 mutations; most emergent TP53 clones were preexisting. Our findings will aid ongoing/future trials of BCL-2/MDM2 inhibitor combinations. This trial was registered at www.clinicaltrials.gov as #NCT02670044., (© 2023 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
44. A phase Ib trial of mivavotinib (TAK-659), a dual SYK/FLT3 inhibitor, in patients with relapsed/refractory acute myeloid leukemia.
- Author
-
Pratz KW, Kaplan J, Levy M, Bixby D, Burke PW, Erba H, Wise-Draper TM, Roboz GJ, Papadantonakis N, Rajkhowa T, Hernandez D, Dobler I, Gregory RC, Li C, Wang S, Stumpo K, Kannan K, Miao H, and Levis M
- Subjects
- Humans, Protein Kinase Inhibitors adverse effects, Pyrimidines adverse effects, Syk Kinase, fms-Like Tyrosine Kinase 3, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics
- Abstract
Mivavotinib (TAK-659) is an investigational type 1 tyrosine kinase inhibitor with dual activity against spleen tyrosine kinase (SYK) and FMS-like tyrosine kinase 3 (FLT3). We conducted a phase Ib study to investigate the safety, tolerability, and efficacy of mivavotinib in patients with refractory and/or relapsed (R/R) acute myeloid leukemia (AML). Both daily (QD) and twice daily (BID) dosing regimens were evaluated. A total of 43 patients were enrolled, and there were 5 complete responses (4 with incomplete count recovery). In the QD dosing regimen, the maximum tolerated dose (MTD) was not reached up to 160 mg QD per protocol; 140 mg QD was identified as the recommended phase II dose. In the BID dosing regimen, the MTD was 60 mg BID. Thirty patients (70%) experienced a bleeding event on study; the majority were grades 1 or 2, were resolved without mivavotinib modification, and were not considered related to study treatment. Eleven patients (26%) experienced grade ≥3 bleeding events, which were observed most frequently with the 80 mg BID dose. We conducted platelet aggregation studies to investigate the potential role of mivavotinib-mediated SYK inhibition on platelet function. The bleeding events observed may have been the result of several confounding factors, including AML disease status, associated thrombocytopenia, and high doses of mivavotinib. Overall, these findings indicate that the activity of mivavotinib in R/R AML is modest. Furthermore, any future clinical investigation of this agent should be undertaken with caution, particularly in thrombocytopenic patients, due to the potential bleeding risk of SYK inhibition. ClinicalTrials.gov: NCT02323113.
- Published
- 2023
- Full Text
- View/download PDF
45. Immunogenicity of COVID-19 mRNA vaccines in patients with acute myeloid leukemia and myelodysplastic syndrome.
- Author
-
Helfgott DC, Racine-Brzostek S, Short KJ, Zhao Z, Christos P, Nino I, Niu T, Contreras J, Ritchie EK, Desai P, Samuel M, and Roboz GJ
- Subjects
- Adult, Humans, 2019-nCoV Vaccine mRNA-1273, Antibodies, Neutralizing, Antibodies, Viral, BNT162 Vaccine, mRNA Vaccines, SARS-CoV-2, Vaccination, COVID-19 prevention & control, COVID-19 Vaccines adverse effects, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute therapy, Myelodysplastic Syndromes therapy
- Abstract
Immunocompromised patients are susceptible to complications from severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). The mRNA vaccines BNT162b2 and mRNA-1273 are effective in immunocompetent adults, but have diminished activity in immunocompromised patients. We measured anti-spike SARS-CoV-2 antibody (anti-S) response, avidity, and surrogate neutralizing antibody activity in COVID-19 vaccinated patients with acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS). Anti-S was induced in 89% of AML and 88% of MDS patients, but median levels were significantly lower than in healthy controls. SARS-CoV-2 antibody avidity and neutralizing activity from AML patients were significantly lower than controls. Antibody avidity was significantly greater in patients after mRNA-1273 versus BNT162b2; there were trends toward higher anti-S levels and greater neutralizing antibody activity after mRNA-1273 vaccination. Patients with AML and MDS are likely to respond to COVID-19 mRNA vaccination, but differences in anti-S levels, avidity, and neutralizing antibody activity may affect clinical outcomes and require further study.
- Published
- 2023
- Full Text
- View/download PDF
46. North American Blastic Plasmacytoid Dendritic Cell Neoplasm Consortium: position on standards of care and areas of need.
- Author
-
Pemmaraju N, Kantarjian H, Sweet K, Wang E, Senapati J, Wilson NR, Konopleva M, Frankel AE, Gupta V, Mesa R, Ulrickson M, Gorak E, Bhatia S, Budak-Alpdogan T, Mason J, Garcia-Romero MT, Lopez-Santiago N, Cesarman-Maus G, Vachhani P, Lee S, Bhatt VR, Blum W, Walter RB, Bixby D, Gojo I, Duvic M, Rampal RK, de Lima M, Foran J, Fathi AT, Hall AC, Jacoby MA, Lancet J, Mannis G, Stein AS, Mims A, Rizzieri D, Olin R, Perl A, Schiller G, Shami P, Stone RM, Strickland S, Wieduwilt MJ, Daver N, Ravandi F, Vasu S, Guzman M, Roboz GJ, Khoury J, Qazilbash M, Aung PP, Cuglievan B, Madanat Y, Kharfan-Dabaja MA, Pawlowska A, Taylor J, Tallman M, Dhakal P, and Lane AA
- Subjects
- Child, Humans, Aged, Standard of Care, Interleukin-3 Receptor alpha Subunit, Dendritic Cells pathology, Neoplasm Recurrence, Local pathology, Acute Disease, North America, Myeloproliferative Disorders pathology, Hematologic Neoplasms pathology, Skin Neoplasms pathology
- Abstract
Blastic plasmacytoid dendritic cell neoplasm (BPDCN) is a rare hematologic malignancy with historically poor outcomes and no worldwide consensus treatment approach. Unique among most hematologic malignancies for its frequent cutaneous involvement, BPDCN can also invade other extramedullary compartments, including the central nervous system. Generally affecting older adults, many patients are unfit to receive intensive chemotherapy, and although hematopoietic stem cell transplantation is preferred for younger, fit individuals, not all are eligible. One recent therapeutic breakthrough is that all BPDCNs express CD123 (IL3Rα) and that this accessible surface marker can be pharmacologically targeted. The first-in-class agent for BPDCN, tagraxofusp, which targets CD123, was approved in December 2018 in the United States for patients with BPDCN aged ≥2 years. Despite favorable response rates in the frontline setting, many patients still relapse in the setting of monotherapy, and outcomes in patients with relapsed/refractory BPDCN remain dismal. Therefore, novel approaches targeting both CD123 and other targets are actively being investigated. To begin to formally address the state of the field, we formed a new collaborative initiative, the North American BPDCN Consortium (NABC). This group of experts, which includes a multidisciplinary panel of hematologists/oncologists, hematopoietic stem cell transplant physicians, pathologists, dermatologists, and pediatric oncologists, was tasked with defining the current standard of care in the field and identifying the most important research questions and future directions in BPDCN. The position findings of the NABC's inaugural meetings are presented herein., (© 2023 by The American Society of Hematology.)
- Published
- 2023
- Full Text
- View/download PDF
47. Finding consistency in classifications of myeloid neoplasms: a perspective on behalf of the International Workshop for Myelodysplastic Syndromes.
- Author
-
Zeidan AM, Bewersdorf JP, Buckstein R, Sekeres MA, Steensma DP, Platzbecker U, Loghavi S, Boultwood J, Bejar R, Bennett JM, Borate U, Brunner AM, Carraway H, Churpek JE, Daver NG, Della Porta M, DeZern AE, Efficace F, Fenaux P, Figueroa ME, Greenberg P, Griffiths EA, Halene S, Hasserjian RP, Hourigan CS, Kim N, Kim TK, Komrokji RS, Kutchroo V, List AF, Little RF, Majeti R, Nazha A, Nimer SD, Odenike O, Padron E, Patnaik MM, Roboz GJ, Sallman DA, Sanz G, Stahl M, Starczynowski DT, Taylor J, Xie Z, Xu M, Savona MR, Wei AH, Abdel-Wahab O, and Santini V
- Subjects
- Humans, Neoplasms, Myelodysplastic Syndromes, Myeloproliferative Disorders, Leukemia, Myeloid, Acute
- Published
- 2022
- Full Text
- View/download PDF
48. Prognostic impact of NPM1 and FLT3 mutations in patients with AML in first remission treated with oral azacitidine.
- Author
-
Döhner H, Wei AH, Roboz GJ, Montesinos P, Thol FR, Ravandi F, Dombret H, Porkka K, Sandhu I, Skikne B, See WL, Ugidos M, Risueño A, Chan ET, Thakurta A, Beach CL, and Lopes de Menezes D
- Subjects
- Azacitidine therapeutic use, Humans, Mutation, Neoplasm, Residual, Nucleophosmin, Prognosis, Protein-Tyrosine Kinases genetics, Recurrence, Remission Induction, fms-Like Tyrosine Kinase 3 genetics, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Nuclear Proteins genetics
- Abstract
The randomized, placebo-controlled, phase 3 QUAZAR AML-001 trial (ClinicalTrials.gov identifier: NCT01757535) evaluated oral azacitidine (Oral-AZA) in patients with acute myeloid leukemia (AML) in first remission after intensive chemotherapy (IC) who were not candidates for hematopoietic stem cell transplantation. Eligible patients were randomized 1:1 to Oral-AZA 300 mg or placebo for 14 days per 28-day cycle. We evaluated relapse-free survival (RFS) and overall survival (OS) in patient subgroups defined by NPM1 and FLT3 mutational status at AML diagnosis and whether survival outcomes in these subgroups were influenced by presence of post-IC measurable residual disease (MRD). Gene mutations at diagnosis were collected from patient case report forms; MRD was determined centrally by multiparameter flow cytometry. Overall, 469 of 472 randomized patients (99.4%) had available mutational data; 137 patients (29.2%) had NPM1 mutations (NPM1mut), 66 patients (14.1%) had FLT3 mutations (FLT3mut; with internal tandem duplications [ITD], tyrosine kinase domain mutations [TKDmut], or both), and 30 patients (6.4%) had NPM1mut and FLT3-ITD at diagnosis. Among patients with NPM1mut, OS and RFS were improved with Oral-AZA by 37% (hazard ratio [HR], 0.63; 95% confidence interval [CI], 0.41-0.98) and 45% (HR, 0.55; 95% CI, 0.35-0.84), respectively, vs placebo. Median OS was improved numerically with Oral-AZA among patients with NPM1mut whether without MRD (48.6 months vs 31.4 months with placebo) or with MRD (46.1 months vs 10.0 months with placebo) post-IC. Among patients with FLT3mut, Oral-AZA improved OS and RFS by 37% (HR, 0.63; 95% CI, 0.35-1.12) and 49% (HR, 0.51; 95% CI, 0.27-0.95), respectively, vs placebo. Median OS with Oral-AZA vs placebo was 28.2 months vs 16.2 months, respectively, for patients with FLT3mut and without MRD and 24.0 months vs 8.0 months for patients with FLT3mut and MRD. In multivariate analyses, Oral-AZA significantly improved survival independent of NPM1 or FLT3 mutational status, cytogenetic risk, or post-IC MRD status., (© 2022 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
49. Integrative analysis identifies an older female-linked AML patient group with better risk in ECOG-ACRIN Cancer Research Group's clinical trial E3999.
- Author
-
Rapaport F, Seier K, Neelamraju Y, Hassane D, Baslan T, Gildea DT, Haddox S, Lee T, Murdock HM, Sheridan C, Thurmond A, Wang L, Carroll M, Cripe LD, Fernandez H, Mason CE, Paietta E, Roboz GJ, Sun Z, Tallman MS, Zhang Y, Gönen M, Levine R, Melnick AM, Kleppe M, and Garrett-Bakelman FE
- Subjects
- Female, Humans, Leukemia, Myeloid, Acute diagnosis, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics
- Published
- 2022
- Full Text
- View/download PDF
50. Prospective comparison of outcomes with azacitidine and decitabine in patients with AML ineligible for intensive chemotherapy.
- Author
-
Zeidan AM, Fenaux P, Gobbi M, Mayer J, Roboz GJ, Krauter J, Robak T, Kantarjian HM, Novák J, Jedrzejczak WW, Thomas X, Ojeda-Uribe M, Miyazaki Y, Min YH, Yeh SP, Brandwein JM, Gercheva L, Demeter J, Griffiths EA, Yee KWL, Issa JJ, Bewersdorf JP, Keer H, Hao Y, Azab M, and Döhner H
- Subjects
- Antimetabolites, Antineoplastic therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Decitabine adverse effects, Humans, Treatment Outcome, Azacitidine therapeutic use, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute etiology
- Published
- 2022
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.