217 results on '"Robert K. Stuart"'
Search Results
2. Phase 3 results for vosaroxin/cytarabine in the subset of patients ≥60 years old with refractory/early relapsed acute myeloid leukemia
- Author
-
Farhad Ravandi, Ellen K. Ritchie, Hamid Sayar, Jeffrey E. Lancet, Michael D. Craig, Norbert Vey, Stephen A. Strickland, Gary J. Schiller, Elias Jabbour, Arnaud Pigneux, Heinz-August Horst, Christian Récher, Virginia M. Klimek, Jorge E. Cortes, Angelo-Michele Carella, Miklos Egyed, Utz Krug, Judith A. Fox, Adam R. Craig, Renee Ward, Jennifer A. Smith, Gary Acton, Hagop M. Kantarjian, and Robert K. Stuart
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2018
- Full Text
- View/download PDF
3. A phase 1b/2 study of vosaroxin in combination with cytarabine in patients with relapsed or refractory acute myeloid leukemia
- Author
-
Jeffrey E. Lancet, Gail J. Roboz, Larry D. Cripe, Glenn C. Michelson, Judith A. Fox, Richard D. Leavitt, Tianling Chen, Rachael Hawtin, Adam R. Craig, Farhad Ravandi, Michael B. Maris, Robert K. Stuart, and Judith E. Karp
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Vosaroxin is a first-in-class anticancer quinolone derivative that intercalates DNA and inhibits topoisomerase II. This study assessed the safety and tolerability of vosaroxin plus cytarabine in patients with relapsed/refractory acute myeloid leukemia. Escalating vosaroxin doses (10-minute infusion; 10–90 mg/m2; days 1, 4) were given in combination with cytarabine on one of two schedules: schedule A (24-hour continuous intravenous infusion, 400 mg/m2/day, days 1–5) or schedule B (2-hour intravenous infusion, 1 g/m2/day, days 1–5). Following dose escalation, enrollment was expanded at the maximum tolerated dose. Of 110 patients enrolled, 108 received treatment. The maximum tolerated dose of vosaroxin was 80 mg/m2 for schedule A (dose-limiting toxicities: grade 3 bowel obstruction and stomatitis) and was not reached for schedule B (recommended phase 2 dose: 90 mg/m2). In the efficacy population (all patients in first relapse or with primary refractory disease treated with vosaroxin 80–90 mg/m2; n=69), the complete remission rate was 25% and the complete remission/complete remission with incomplete blood count recovery rate was 28%. The 30-day all-cause mortality rate was 2.5% among all patients treated at a dose of 80–90 mg/m2. Based upon these results, a phase 3 trial of vosaroxin plus cytarabine was initiated in patients with relapsed/refractory acute myeloid leukemia.
- Published
- 2015
- Full Text
- View/download PDF
4. Data from Phase I Trial of Bortezomib (PS-341; NSC 681239) and 'Nonhybrid' (Bolus) Infusion Schedule of Alvocidib (Flavopiridol; NSC 649890) in Patients with Recurrent or Refractory Indolent B-cell Neoplasms
- Author
-
Steven Grant, Daniel Sullivan, John D. Roberts, Domenico Coppola, William D. Figg, L. Austin Doyle, A. Dimitrios Colevas, Kevin T. Hogan, Heidi Sankala, Martha Wellons, Caryn Weir-Wiggins, Ellen Shrader, Mary Beth Tombes, Connie Honeycutt, Loveleen Kang, Jana Dawson, Cody J. Peer, Wen Wan, Viswanathan Ramakrishnan, Robert K. Stuart, G. David Roodman, Rachid C. Baz, Prithviraj Bose, E. Brent Perkins, Maciej Kmieciak, and Beata Holkova
- Abstract
Purpose: This phase I study was conducted to determine the dose-limiting toxicities (DLT) and maximum tolerated dose (MTD) for the combination of bortezomib and alvocidib in patients with B-cell malignancies (multiple myeloma, indolent lymphoma, Waldenstrom macroglobulinemia, and mantle cell lymphoma).Experimental Design: Patients received bortezomib (intravenous push), followed by alvocidib (1-hour infusion), on days 1, 4, 8, and 11 of a 21-day treatment cycle. Patients experiencing responses or stable disease continued on treatment at the investigator's discretion. A standard 3+3 dose-escalation design was used to identify the MTD based on DLTs, and pharmacokinetic and pharmacodynamic studies were conducted.Results: A total of 44 patients were enrolled, with 39 patients assessed for response. The MTD was established as 1.3 mg/m2 for bortezomib and 40 mg/m2 for alvocidib. The most common hematologic toxicities included leukopenia, lymphopenia, neutropenia, and thrombocytopenia. The most common nonhematologic toxicities included diarrhea, fatigue, and sensory neuropathy. Three complete remissions (8%) and 10 partial remissions (26%) were observed for a total response rate of 33%. Pharmacokinetic findings with the current dosing regimen were consistent with the comparable literature and the hybrid dosing regimen. Pharmacodynamic study results did not correlate with clinical responses.Conclusions: The combination of bortezomib and alvocidib is tolerable, and an MTD has been established for this schedule. The regimen appears to be efficacious in patients with relapsed/refractory multiple myeloma or indolent non-Hodgkin lymphoma. As the nonhybrid regimen is less cumbersome than the previous hybrid dosing schedule regimen, the current schedule is recommended for successor studies. Clin Cancer Res; 20(22); 5652–62. ©2014 AACR.
- Published
- 2023
- Full Text
- View/download PDF
5. Supplementary Materials from Phase I Trial of Bortezomib (PS-341; NSC 681239) and 'Nonhybrid' (Bolus) Infusion Schedule of Alvocidib (Flavopiridol; NSC 649890) in Patients with Recurrent or Refractory Indolent B-cell Neoplasms
- Author
-
Steven Grant, Daniel Sullivan, John D. Roberts, Domenico Coppola, William D. Figg, L. Austin Doyle, A. Dimitrios Colevas, Kevin T. Hogan, Heidi Sankala, Martha Wellons, Caryn Weir-Wiggins, Ellen Shrader, Mary Beth Tombes, Connie Honeycutt, Loveleen Kang, Jana Dawson, Cody J. Peer, Wen Wan, Viswanathan Ramakrishnan, Robert K. Stuart, G. David Roodman, Rachid C. Baz, Prithviraj Bose, E. Brent Perkins, Maciej Kmieciak, and Beata Holkova
- Abstract
Supplementary Materials. Supplemental Table 1. C1D1 alvocidib pharmacokinetics by dose Supplemental Figure 1. Sample alvocidib concentration-time profile. Samples were obtained pre-infusion, immediately following infusion, and at 1, 2, 4, 8, 12, and 24 hours post-infusion.
- Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Data from Phase I Trial of Bortezomib (PS-341; NSC 681239) and Alvocidib (Flavopiridol; NSC 649890) in Patients with Recurrent or Refractory B-Cell Neoplasms
- Author
-
Steven Grant, John D. Roberts, Daniel M. Sullivan, John Wright, Austin Doyle, Sarah Kolla, William D. Figg, Cody Peer, Robert K. Stuart, Jana Dawson, Loveleen Kang, Domenico Coppola, G. David Roodman, Kevin T. Hogan, Martha D. Wellons, Neha Talreja, Ellen Shrader, Mary Beth Tombes, Viswanathan Ramakrishnan, E. Brent Perkins, and Beata Holkova
- Abstract
Supplementary Figures S1-S2; Supplementary Tables S1-S2.
- Published
- 2023
- Full Text
- View/download PDF
7. Phase 3 randomized trial of chemotherapy with or without oblimersen in older AML patients: CALGB 10201 (Alliance)
- Author
-
Robert K. Stuart, Grerk Sutamtewagul, Ann-Kathrin Eisfeld, Ravi Vij, Krzysztof Mrózek, Jessica Kohlschmidt, Mohan Thakuri, Richard Stone, William Blum, Wendy Stock, Andrew J. Carroll, Jonathan E. Kolitz, Richard A. Larson, Guido Marcucci, Eunice S. Wang, Clara D. Bloomfield, Jun Yin, John C. Byrd, Alison Walker, and Sawyer B. Jacobson
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Myeloid ,Clinical Trials and Observations ,Daunorubicin ,Chronic lymphocytic leukemia ,medicine.medical_treatment ,03 medical and health sciences ,0302 clinical medicine ,hemic and lymphatic diseases ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,neoplasms ,Aged ,Chemotherapy ,business.industry ,Oblimersen ,Cytarabine ,Hematology ,Thionucleotides ,medicine.disease ,Chemotherapy regimen ,Leukemia, Myeloid, Acute ,Leukemia ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,business ,medicine.drug - Abstract
Overexpression of B-cell leukemia/lymphoma 2 (BCL2) renders acute myeloid leukemia (AML) cells resistant to chemotherapy and has been associated with unfavorable outcomes. Oblimersen (G3139) is a phosphorothioate 18-mer antisense oligonucleotide directed against the first 6 BCL2 codons. In a phase 1 study of AML patients treated with G3139, cytarabine, and daunorubicin induction with cytarabine consolidation, no antisense-related toxicity was reported, and BCL2 downregulation occurred in patients achieving complete remission. In this phase 3 trial, untreated older AML patients were randomized to cytarabine (100 mg/m2 per day on days 4-10) and daunorubicin (60 mg/m2 per day on days 4-6) followed by cytarabine consolidation (2000 mg/m2 per day on days 4-8) with (arm A) or without (arm B) G3139 (7 mg/m2 per day on days 1-10 [induction] or days 1-8 [consolidation]). A total of 506 patients were enrolled. No differences in toxicity were observed between arms. Estimated overall survival (OS) at 1 year was 43% for arm A and 40% for arm B (1-sided log rank P = .13), with no differences in disease-free (DFS; P = .26) or event-free survival (P = .80). Subgroup analyses showed patients age
- Published
- 2021
- Full Text
- View/download PDF
8. CPX-351 versus 7+3 cytarabine and daunorubicin chemotherapy in older adults with newly diagnosed high-risk or secondary acute myeloid leukaemia: 5-year results of a randomised, open-label, multicentre, phase 3 trial
- Author
-
Dale L. Bixby, Jeffrey E. Lancet, Robert K. Stuart, Scott R. Solomon, Laura F. Newell, Ellen K. Ritchie, Donna E. Hogge, Geoffrey L. Uy, Daniel H. Ryan, Stephen A. Strickland, Gary J. Schiller, Stefan Faderl, Tara L. Lin, Jorge E. Cortes, Jonathan E. Kolitz, and Matthew J Wieduwilt
- Subjects
Male ,medicine.medical_specialty ,Myeloid ,Daunorubicin ,medicine.medical_treatment ,Kaplan-Meier Estimate ,Hematopoietic stem cell transplantation ,Drug Administration Schedule ,law.invention ,Randomized controlled trial ,law ,Internal medicine ,Humans ,Medicine ,Infusions, Intravenous ,Adverse effect ,Aged ,Proportional Hazards Models ,Chemotherapy ,business.industry ,Cytarabine ,Hematopoietic Stem Cell Transplantation ,Induction chemotherapy ,Neoplasms, Second Primary ,Hematology ,Middle Aged ,Leukemia, Myeloid, Acute ,Treatment Outcome ,medicine.anatomical_structure ,Female ,business ,Follow-Up Studies ,medicine.drug - Abstract
Summary Background Daunorubicin and cytarabine are used as standard induction chemotherapy for patients with acute myeloid leukaemia. CPX-351 is a dual-drug liposomal encapsulation of daunorubicin and cytarabine in a synergistic 1:5 molar ratio. Primary analysis of the phase 3 trial in adults aged 60–75 years with newly diagnosed high-risk or secondary acute myeloid leukaemia provided support for approval of CPX-351 by the US Food and Drug Administration and European Medicines Agency. We describe the prospectively planned final 5-year follow-up results. Methods This randomised, open-label, multicentre, phase 3 trial was done across 39 academic and regional cancer centres in the USA and Canada. Eligible patients were aged 60–75 years and had a pathological diagnosis of acute myeloid leukaemia according to WHO 2008 criteria, no previous induction therapy for acute myeloid leukaemia, and an Eastern Cooperative Oncology Group performance status of 0–2. Patients were randomly assigned 1:1 (stratified by age and acute myeloid leukaemia subtype) to receive up to two induction cycles of CPX-351 (100 units/m2 administered as a 90-min intravenous infusion on days 1, 3, and 5; on days 1 and 3 for the second induction) or standard chemotherapy (cytarabine 100 mg/m2 per day continuous intravenous infusion for 7 days plus intravenous daunorubicin 60 mg/m2 on days 1, 2, and 3 [7+3]; cytarabine for 5 days and daunorubicin on days 1 and 2 for the second induction [5+2]). Patients with complete remission or complete remission with incomplete neutrophil or platelet recovery could receive up to tw cycles of consolidation therapy with CPX-351 (65 units/m2 90-min infusion on days 1 and 3) or chemotherapy (5+2, same dosage as in the second induction cycle). The primary outcome was overall survival analysed in all randomly assigned patients. No additional adverse events were collected with long-term follow-up, except data for deaths. This trial is registered with ClinicalTrials.gov , NCT01696084 , and is complete. Findings Between Dec 20, 2012, and Nov 11, 2014, 309 patients with newly diagnosed high-risk or secondary acute myeloid leukaemia were enrolled and randomly assigned to receive CPX-351 (153 patients) or 7+3 (156 patients). At a median follow-up of 60·91 months (IQR 60·06–62·98) in the CPX-351 group and 59·93 months (59·73–60·50) in the 7+3 group, median overall survival was 9·33 months (95% CI 6·37–11·86) with CPX-351 and 5·95 months (4·99–7·75) with 7+3 (HR 0·70, 95% CI 0·55–0·91). 5-year overall survival was 18% (95% CI 12–25%) in the CPX-351 group and 8% (4–13%) in the 7+3 group. The most common cause of death in both groups was progressive leukaemia (70 [56%] of 124 deaths in the CPX-351 group and 74 [53%] of 140 deaths in the 7+3 group). Six (5%) of 124 deaths in the CPX-351 group and seven (5%) of 140 deaths in the 7+3 group were considered related to study treatment. Interpretation After 5 years of follow-up, the improved overall survival with CPX-351 versus 7+3 was maintained, which supports the previous evidence that CPX-351 can contribute to long-term remission and improved overall survival in patients aged 60–75 years with newly diagnosed high-risk or secondary acute myeloid leukaemia. Funding Jazz Pharmaceuticals.
- Published
- 2021
- Full Text
- View/download PDF
9. Final analysis of a phase 1/2b study of ibrutinib combined with carfilzomib/dexamethasone in patients with relapsed/refractory multiple myeloma
- Author
-
Ruben Niesvizky, Saurabh Chhabra, Chatchada Karanes, Robert F. Cornell, Yihua Lee, Saad Z. Usmani, Jeffrey Matous, Robert K. Stuart, Larry D. Anderson, Jason Valent, Cristina Gasparetto, Zeena Salman, Emily Liu, Ajai Chari, Matthew A. Lunning, Chaim Shustik, and Saulius Girnius
- Subjects
Male ,Oncology ,Cancer Research ,Bruton's tyrosine kinase inhibitor ,Dexamethasone ,chemistry.chemical_compound ,0302 clinical medicine ,Piperidines ,Original Research Articles ,Antineoplastic Combined Chemotherapy Protocols ,Original Research Article ,Multiple myeloma ,Aged, 80 and over ,education.field_of_study ,carfilzomib ,Bortezomib ,Hematology ,General Medicine ,Middle Aged ,Prognosis ,Survival Rate ,multiple myeloma ,030220 oncology & carcinogenesis ,Ibrutinib ,Female ,Oligopeptides ,medicine.drug ,Adult ,medicine.medical_specialty ,Population ,03 medical and health sciences ,Refractory ,ibrutinib ,Internal medicine ,medicine ,Humans ,education ,Adverse effect ,Aged ,Salvage Therapy ,business.industry ,Adenine ,medicine.disease ,Carfilzomib ,Pyrimidines ,chemistry ,Drug Resistance, Neoplasm ,Pyrazoles ,Neoplasm Recurrence, Local ,business ,Follow-Up Studies ,030215 immunology - Abstract
Patients with multiple myeloma (MM) inevitably relapse on initial treatment regimens, and novel combination therapies are needed. Ibrutinib is a first‐in‐class, once‐daily inhibitor of Bruton's tyrosine kinase, an enzyme implicated in growth and survival of MM cells. Preclinical data suggest supra‐additivity or synergy between ibrutinib and proteasome inhibitors (PIs) against MM. This phase 1/2b study evaluated the efficacy and safety of ibrutinib plus the PI carfilzomib and dexamethasone in patients with relapsed/refractory MM (RRMM). In this final analysis, we report results in patients who received the recommended phase 2 dose (RP2D; ibrutinib 840 mg and carfilzomib 36 mg/m2 with dexamethasone), which was determined in phase 1. The primary efficacy endpoint was overall response rate (ORR). Fifty‐nine patients with RRMM received the RP2D (18 in phase 1 and 41 in phase 2b). These patients had received a median of three prior lines of therapy; 69% were refractory to bortezomib, and 90% were refractory to their last treatment. ORR in the RP2D population was 71% (stringent complete response and complete response: 3% each). Median duration of clinical benefit and median duration of response were both 6.5 months. Median progression‐free survival (PFS) was 7.4 months, and median overall survival (OS) was 35.9 months. High‐risk patients had comparable ORR and median PFS (67% and 7.7 months, respectively) to non–high‐risk patients, whose ORR was 73% and median PFS was 6.9 months, whereas median OS in high‐risk patients was 13.9 months and not reached in non–high‐risk patients. The most common grade ≥3 hematologic treatment‐emergent adverse events (TEAEs) were anemia and thrombocytopenia (17% each); the most common grade ≥3 non‐hematologic TEAE was hypertension (19%). In patients with RRMM treated with multiple previous lines of therapy, ibrutinib plus carfilzomib demonstrated anticancer activity within the expected efficacy range. No new safety signals were identified and the combination was well‐tolerated.
- Published
- 2020
- Full Text
- View/download PDF
10. Gilteritinib or Chemotherapy for Relapsed or Refractory FLT3-Mutated AML
- Author
-
Robert K. Stuart, Je-Hwan Lee, Andreas Neubauer, Jorge E. Cortes, Richard A. Larson, Timothy S. Pardee, Mark J. Levis, Chaofeng Liu, Harry P. Erba, Sung Soo Yoon, Celalettin Ustun, Ellin Berman, Maria R. Baer, Nikolai A. Podoltsev, Wen-Chien Chou, Giovanni Martinelli, Nahla Hasabou, Pau Montesinos, Antonio Di Stasi, Stefania Paolini, Alexander E. Perl, Francesco Fabbiano, Hisayuki Yokoyama, Xuan Liu, Margaret Kasner, Rebecca L. Olin, Naoko Hosono, Erkut Bahceci, Amir T. Fathi, Fabio Ciceri, Christian Recher, Perl, A. E., Martinelli, G., Cortes, J. E., Neubauer, A., Berman, E., Paolini, S., Montesinos, P., Baer, M. R., Larson, R. A., Ustun, C., Fabbiano, F., Erba, H. P., Di Stasi, A., Stuart, R., Olin, R., Kasner, M., Ciceri, F., Chou, W. -C., Podoltsev, N., Recher, C., Yokoyama, H., Hosono, N., Yoon, S. -S., Lee, J. -H., Pardee, T., Fathi, A. T., Liu, C., Hasabou, N., Liu, X., Bahceci, E., and Levis, M. J.
- Subjects
Myeloid ,medicine.medical_treatment ,Salvage therapy ,030204 cardiovascular system & hematology ,03 medical and health sciences ,chemistry.chemical_compound ,fluids and secretions ,0302 clinical medicine ,hemic and lymphatic diseases ,Medicine ,030212 general & internal medicine ,Quizartinib ,Chemotherapy ,business.industry ,Myeloid leukemia ,hemic and immune systems ,General Medicine ,medicine.disease ,Leukemia ,medicine.anatomical_structure ,chemistry ,embryonic structures ,Fms-Like Tyrosine Kinase 3 ,Cancer research ,business ,Crenolanib - Abstract
BACKGROUND Patients with relapsed or refractory acute myeloid leukemia (AML) with mutations in the FMS-like tyrosine kinase 3 gene (FLT3) infrequently have a response to salvage chemotherapy. Gilteritinib is an oral, potent, selective FLT3 inhibitor with single-agent activity in relapsed or refractory FLT3-mutated AML. METHODS In a phase 3 trial, we randomly assigned adults with relapsed or refractory FLT3mutated AML in a 2:1 ratio to receive either gilteritinib (at a dose of 120 mg per day) or salvage chemotherapy. The two primary end points were overall survival and the percentage of patients who had complete remission with full or partial hematologic recovery. Secondary end points included event-free survival (freedom from treatment failure [i.e., relapse or lack of remission] or death) and the percentage of patients who had complete remission. RESULTS Of 371 eligible patients, 247 were randomly assigned to the gilteritinib group and 124 to the salvage chemotherapy group. The median overall survival in the gilteritinib group was significantly longer than that in the chemotherapy group (9.3 months vs. 5.6 months; hazard ratio for death, 0.64; 95% confidence interval [CI], 0.49 to 0.83; P
- Published
- 2019
- Full Text
- View/download PDF
11. A phase 2 study to assess the pharmacokinetics and pharmacodynamics of CPX-351 and its effects on cardiac repolarization in patients with acute leukemias
- Author
-
Robert K. Stuart, Arthur C. Louie, Timothy Callahan, Lawrence D. Mayer, Kamalika Banerjee, Eric Rubenstein, Barry D. Liboiron, Donna Alvarez, Laura C. Michaelis, Laura F. Newell, Qi Wang, Tara L. Lin, and Helen S. Pentikis
- Subjects
Adult ,Male ,0301 basic medicine ,Cancer Research ,medicine.medical_specialty ,Daunorubicin ,Acute lymphoblastic leukemia ,Toxicology ,QT interval ,03 medical and health sciences ,0302 clinical medicine ,Pharmacokinetics ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Pharmacology (medical) ,Aged ,Aged, 80 and over ,Pharmacology ,Cardiotoxicity ,Acute leukemia ,Acute myeloid leukemia ,Cardiac repolarization ,business.industry ,Cytarabine ,Middle Aged ,medicine.disease ,Drug Combinations ,Leukemia, Myeloid, Acute ,Treatment Outcome ,030104 developmental biology ,Pharmacodynamics ,Oncology ,030220 oncology & carcinogenesis ,Liposomes ,Cardiology ,Original Article ,Female ,business ,Febrile neutropenia ,Half-Life ,medicine.drug - Abstract
Purpose Daunorubicin can induce left ventricular dysfunction and QT interval prolongation. This study assessed the effects of CPX-351, a liposomal encapsulation of cytarabine and daunorubicin, on cardiac repolarization. Methods Twenty-six adults with acute leukemia were treated with CPX-351 for 1–2 induction cycles and ≤ 4 consolidation cycles. The primary endpoint was mean change in QTcF from baseline. Results Mean QTcF changes were 30 h. Thirteen (50%) patients achieved remission. The most common adverse events were febrile neutropenia, fatigue, and nausea. Conclusions The cytarabine and daunorubicin in CPX-351 liposomes were metabolized and excreted similarly to conventional formulation; however, plasma pharmacokinetics were altered. CPX-351 did not prolong the QT interval, suggesting that CPX-351 may induce less cardiotoxicity than previously reported for conventional daunorubicin. Trial registration Clinicaltrials.gov identifier: NCT02238925. Electronic supplementary material The online version of this article (10.1007/s00280-019-03856-9) contains supplementary material, which is available to authorized users.
- Published
- 2019
- Full Text
- View/download PDF
12. Pracinostat plus azacitidine in older patients with newly diagnosed acute myeloid leukemia: results of a phase 2 study
- Author
-
Guillermo Garcia-Manero, Mohan Tummala, Samer K. Khaled, Bruno C. Medeiros, Martha Arellano, Lori J Maness-Harris, Yasmin Abaza, Olatoyosi Odenike, Richard G. Ghalie, Hamid Sayar, Robert K. Stuart, Abdulraheem Yacoub, Elie Traer, Ruben Giorgino, Prapti A. Patel, Koichi Takahashi, Ehab Atallah, Kasra Karamlou, and Mrinal M. Patnaik
- Subjects
Male ,medicine.medical_specialty ,Clinical Trials and Observations ,Pracinostat ,Azacitidine ,Phases of clinical research ,Antineoplastic Agents ,Gastroenterology ,chemistry.chemical_compound ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Clinical endpoint ,Humans ,Survival analysis ,Aged ,Aged, 80 and over ,business.industry ,Induction chemotherapy ,Hematology ,medicine.disease ,Survival Analysis ,Leukemia, Myeloid, Acute ,Regimen ,Treatment Outcome ,chemistry ,Benzimidazoles ,Female ,business ,Febrile neutropenia ,medicine.drug - Abstract
Pracinostat, a potent oral pan-histone deacetylase inhibitor with modest single-agent activity in acute myeloid leukemia (AML), has shown synergistic antitumor activity when combined with azacitidine. This single-group, multicenter phase 2 study assessed the safety and efficacy of pracinostat combined with azacitidine in patients who were at least 65 years old with newly diagnosed AML and who were ineligible for standard induction chemotherapy. Patients received pracinostat 60 mg/d, 3 d/wk, for 3 consecutive weeks, plus azacitidine 75 mg/m2 daily for 7 days in a 28-day cycle. Primary endpoints were complete remission (CR), CR with incomplete count recovery (CRi), and morphologic leukemia-free state (MLFS) rates of the combination. Secondary endpoints included safety, progression-free survival (PFS), and overall survival (OS) of the regimen. Fifty patients (33 de novo, 12 secondary, and 5 therapy-related AML) were enrolled. Twenty-six patients (52%) achieved the primary endpoint of CR (42%), CRi (4%), and MLFS (6%). Median OS and PFS were 19.1 months (95% confidence interval [CI], 10-26.5 months) and 12.6 months (95% CI, 10-17.7 months), respectively, with a 1-year OS rate of 62%. Forty-three patients (86%) experienced at least 1 grade 3 or worse treatment-emergent adverse event with the combination, with infections (52%), thrombocytopenia (46%), and febrile neutropenia (44%) reported as the most common toxicities. The 30- and 60-day all-cause mortality rates were 2% and 10%, respectively. DNA sequencing revealed somatic mutations at baseline, and clearance rates correlated with response to treatment. Pracinostat plus azacitidine is a well-tolerated and active regimen in the frontline treatment of older patients with AML unfit for intensive therapy. A larger controlled trial is ongoing. This trial was registered at www.clinicaltrials.gov as #NCT01912274.
- Published
- 2019
- Full Text
- View/download PDF
13. Comparative Analysis of Calcineurin Inhibitor-Based Methotrexate and Mycophenolate Mofetil-Containing Regimens for Prevention of Graft-versus-Host Disease after Reduced-Intensity Conditioning Allogeneic Transplantation
- Author
-
Yoshihiro Inamoto, Harry C. Schouten, Amin M. Alousi, Ravi Vij, David I. Marks, John E. Wagner, Joseph Pidala, Joseph H. Antin, Margaret L. MacMillan, Gérard Socié, Ayman Saad, Shahrukh K. Hashmi, Luciano J. Costa, Mukta Arora, Hisham Abdel-Azim, Hélène Schoemans, Michael T. Hemmer, Robert Peter Gale, Stephen R. Spellman, Daniel R. Couriel, Saurabh Chhabra, Taiga Nishihori, Sachiko Seo, Alvaro Urbano-Ispizua, Bipin N. Savani, Brenda W. Cooper, Muna Qayed, Robert J. Hayashi, Leo F. Verdonck, Mark R. Litzow, Jean A. Yared, Usama Gergis, Navneet S. Majhail, Betty K. Hamilton, Mahmoud Aljurf, Robert K. Stuart, Rammurti T. Kamble, Leslie Lehmann, Takanori Teshima, Ying Liu, Rodrigo Martino, Jean-Yves Cahn, Olle Ringdén, Peiman Hematti, Melhem Solh, Dennis Dong Hwan Kim, RS: GROW - R3 - Innovative Cancer Diagnostics & Therapy, Interne Geneeskunde, and MUMC+: MA Hematologie (9)
- Subjects
Male ,Transplantation Conditioning ,BLOOD ,Tissue transplantation ,GVHD PROPHYLAXIS ,TACROLIMUS ,Graft vs Host Disease ,Stem cells ,Gastroenterology ,Empelts de teixits ,immune system diseases ,hemic and lymphatic diseases ,Leukemia ,Mycophenolate mofetil ,Hematopoietic Stem Cell Transplantation ,Drugs ,Hematology ,Middle Aged ,Allogeneic hematopoietic cell transplantation ,Allografts ,Survival Rate ,COMPARING METHOTREXATE ,Myeloid leukemia ,surgical procedures, operative ,Calcineurin inhibitor Tacrolimus ,Cyclosporine ,Female ,TRIAL ,Cèl·lules mare ,Life Sciences & Biomedicine ,Medicaments ,Adult ,medicine.medical_specialty ,Allogeneic transplantation ,Leucèmia mieloide ,BONE-MARROW ,Immunology ,Calcineurin Inhibitors ,chemical and pharmacologic phenomena ,Disease-Free Survival ,Article ,Myelogenous ,Internal medicine ,medicine ,Humans ,Leucèmia limfocítica crònica ,Aged ,Retrospective Studies ,Transplantation ,Science & Technology ,business.industry ,Siblings ,STEM-CELL TRANSPLANTATION ,Mycophenolic Acid ,Graft-versus-host disease prophylaxis ,medicine.disease ,Tacrolimus ,Calcineurin ,Reduced-intensity conditioning ,Graft-versus-host disease ,Methotrexate ,Myelodysplastic Syndromes ,Chronic lymphocytic leukemia ,business ,Chronic myelogenous leukemia - Abstract
The combination of a calcineurin inhibitor (CNI) such as tacrolimus (TAC) or cyclosporine (CYSP) with methotrexate (MTX) or with mycophenolate mofetil (MMF) has been commonly used for graft-versus-host disease (GVHD) prophylaxis after reduced-intensity conditioning (RIC) allogeneic hematopoietic cell transplantation (alloHCT), but there are limited data comparing efficacy of the 2 regimens. We evaluated 1564 adult patients who underwent RIC alloHCT for acute myelogenous leukemia (AML) and acute lymphoblastic leukemia (ALL), chronic myelogenous leukemia (CML), and myelodysplastic syndrome (MDS) from 2000 to 2013 using HLA-identical sibling (matched related donor [MRD]) or unrelated donor (URD) peripheral blood graft and received CYSP or TAC with MTX or MMF for GVHD prophylaxis. Primary outcomes of the study were acute and chronic GVHD and overall survival (OS). The study divided the patient population into 4 cohorts based on regimen: MMF-TAC, MMF-CYSP, MTX-TAC, and MTX-CYSP. In the URD group, MMF-CYSP was associated with increased risk of grade II to IV acute GVHD (relative risk [RR], 1.78; P < .001) and grade III to IV acute GVHD (RR, 1.93; P = .006) compared with MTX-TAC. In the URD group, use of MMF-TAC (versus MTX-TAC) lead to higher nonrelapse mortality. (hazard ratio, 1.48; P = .008). In either group, no there was no difference in chronic GVHD, disease-free survival, and OS among the GVHD prophylaxis regimens. For RIC alloHCT using MRD, there are no differences in outcomes based on GVHD prophylaxis. However, with URD RIC alloHCT, MMF-CYSP was inferior to MTX-based regimens for acute GVHD prevention, but all the regimens were equivalent in terms of chronic GVHD and OS. Prospective studies, targeting URD recipients are needed to confirm these results. ispartof: BIOLOGY OF BLOOD AND MARROW TRANSPLANTATION vol:25 issue:1 pages:73-85 ispartof: location:United States status: published
- Published
- 2019
- Full Text
- View/download PDF
14. Lower-intensity CPX-351 + venetoclax for patients with newly diagnosed AML who are unfit for intensive chemotherapy
- Author
-
Geoffrey L. Uy, Vinod A. Pullarkat, Praneeth Baratam, Robert K. Stuart, Roland B. Walter, Eric S. Winer, Stefan Faderl, Vijayalakshmi Chandrasekaran, Qi Wang, Divya Chakravarthy, Ronald Cheung, and Tara L. Lin
- Subjects
Cancer Research ,Oncology - Abstract
7031 Background: CPX-351 (US: Vyxeos; Europe: Vyxeos liposomal) is a dual-drug liposomal encapsulation of daunorubicin and cytarabine in a synergistic 1:5 molar ratio. CPX-351 is approved for newly diagnosed, therapy-related AML or AML with myelodysplasia-related changes in patients (pts) who are candidates for intensive chemotherapy (IC) and aged ≥1 year in the US and adults in Europe. However, the appropriate dosage of CPX-351 in pts unfit for IC may be different from the label dosage. Venetoclax (VEN; BCL-2 inhibitor) + low-dose cytarabine has demonstrated efficacy in unfit pts with AML, and drug synergism/additivity in preclinical studies provided a rationale for combining CPX-351 + VEN clinically. Our study evaluates the safety and efficacy of lower-intensity CPX-351 + VEN in adults with newly diagnosed AML who are unfit for IC. Methods: This is an ongoing, open-label, phase 1b study (NCT04038437). Pts who achieve at least partial remission after 1 or 2 cycles may receive up to 4 similar cycles in the dose-exploration phase (DEP) or up to 8 similar cycles in the expansion phase (EP). Pts are assessed for response (morphology, measurable residual disease [MRD]) and monitored for safety and survival. Results: The data include 31 pts enrolled by 9/15/2021, with a data cutoff of 12/2/2021: 4 pts in DEP at dose level 1 (CPX-351 20 units/m2 on Days 1 and 3 + VEN 400 mg on Days 2 to 21 of each cycle), 7 pts in DEP at dose level 2 (CPX-351 40 units/m2 + VEN 400 mg), and a total of 20 pts in DEP and EP at dose level 1b (CPX-351 30 units/m2 + VEN 400 mg; established as the recommended phase 2 dose). Pts were unfit for IC based on age ≥75 y (n = 15) or health (ECOG PS of 2 to 3 and/or comorbidities [n = 16]). Median age was 74 y (range: 60, 90); 65% were male; 77% had de novo AML; 58% had poor-risk disease; and 23% had a TP53 mutation. Nonhematologic treatment-emergent adverse events (TEAEs) in ≥20% of pts were diarrhea (26%), cough (23%), dyspnea (23%), and nausea (23%). Hematologic grade ≥3 TEAEs were reported in 17 (55%) pts; no nonhematologic grade ≥3 TEAE was reported in > 10% of pts. There were no deaths by Day 30; mortality at Day 60 was 13%, with deaths due to myocardial infarction unrelated to therapy (n = 1), worsening lung infection (n = 1), and disease progression (n = 2). Median (IQR) recovery times were 30 d (22, 34.5) to neutrophils ≥500/μL and 21 d (21, 27) to platelets ≥50,000/μL. Complete remission (CR) or CR with incomplete neutrophil or platelet recovery (CRi) was achieved by 16/28 (57%) evaluable pts. All 16 of these pts achieved remission (CR or CRi) after the first cycle. MRD negativity was achieved by 12/16 (75%) pts with CR or CRi, primarily after Cycle 1 (Cycle 1: n = 8; Cycle 2: n = 2; Cycle 3: n = 1; Cycle 4: n = 1). Survival data are not yet mature. Conclusions: Lower-intensity CPX-351 + VEN was generally well tolerated in adults with newly diagnosed AML who are unfit for IC and showed promising initial efficacy, with CR or CRi in the majority of pts. Clinical trial information: NCT04038437.
- Published
- 2022
- Full Text
- View/download PDF
15. Autologous and allogeneic hematopoietic cell transplantation for diffuse large B-cell lymphoma-type Richter syndrome
- Author
-
Yue Chen, Amelia Langston, Silvy Lachance, Joseph P. McGuirk, Amer Assal, Matthew Greenwood, Alex F. Herrera, Samantha Jaglowski, Mehdi Hamadani, Henrik Sengeloev, Adrienne A. Phillips, Mitchell E. Horwitz, Carlos Litovich, Richard T. Maziarz, Mohammad A. H. Mian, Robert K. Stuart, Kevin Rakszawski, Nosha Farhadfar, Theresa Hahn, Ruthee-Lu Bayer, Bassam Mattar, Mohamed A. Kharfan-Dabaja, Shalini Shenoy, Qaiser Bashir, Zachariah DeFilipp, Caron A. Jacobson, Melanie Coleman, Craig S. Sauter, Sunita Nathan, and Kwang Woo Ahn
- Subjects
Oncology ,medicine.medical_specialty ,medicine.medical_treatment ,Chronic lymphocytic leukemia ,Aggressive lymphoma ,Hematopoietic stem cell transplantation ,Transplantation, Autologous ,immune system diseases ,Internal medicine ,hemic and lymphatic diseases ,Medicine ,Humans ,Not evaluated ,Transplantation ,business.industry ,Incidence (epidemiology) ,Hematopoietic Stem Cell Transplantation ,Hematology ,medicine.disease ,Prognosis ,Leukemia, Lymphocytic, Chronic, B-Cell ,Lymphoma ,surgical procedures, operative ,Lymphoma, Large B-Cell, Diffuse ,business ,Diffuse large B-cell lymphoma - Abstract
Richter syndrome (RS) represents a transformation from chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL) to aggressive lymphoma, most commonly diffuse large B-cell lymphoma (DLBCL), which is associated with a dismal prognosis. Patients with DLBCL-RS have poor outcomes with DLBCL-directed therapy; thus, consolidation with hematopoietic cell transplantation (HCT) has been used, with durable remissions observed. Studies reporting HCT outcomes in patients with DLBCL-RS have been small, have not evaluated the prognostic impact of cytogenetic risk factors, and were conducted prior to the era of novel targeted therapy of CLL/SLL. We performed a Center for International Blood and Transplant Research registry study evaluating outcomes after autologous HCT (auto-HCT; n = 53) and allogeneic HCT (allo-HCT; n = 118) in patients with DLBCL-RS treated in the modern era. More auto-HCT recipients were in complete response (CR) at HCT relative to allo-HCT recipients (66% vs 34%), whereas a higher proportion of allo-HCT recipients had 17p deletion (33% vs 7%) and had previously received novel agents (39% vs 10%). In the auto-HCT cohort, the 3-year relapse incidence, progression-free survival (PFS), and overall survival (OS) were 37%, 48%, and 57%, respectively. Among allo-HCT recipients, the 3-year relapse incidence, PFS, and OS were 30%, 43%, and 52%, respectively. In the allo-HCT cohort, deeper response at HCT was associated with outcomes (3-year PFS/OS, 66%/77% CR vs 43%/57% partial response vs 5%/15% resistant; P < .0001 for both), whereas cytogenetic abnormalities and prior novel therapy did not impact outcomes. In our study, HCT resulted in durable remissions in therapy-sensitive patients with DLBCL-RS treated in the era of targeted CLL/SLL therapy, including patients with high-risk features.
- Published
- 2021
- Full Text
- View/download PDF
16. MDS-335: Ivosidenib (IVO) in Patients with IDH1-Mutant Relapsed/Refractory Myelodysplastic Syndrome (R/R MDS): Updated Enrollment for the MDS Sub-Study
- Author
-
Vickie Zhang, Xavier Thomas, James M. Foran, Stéphane de Botton, Martha Arellano, Amir T. Fathi, Justin M. Watts, Richard Stone, Courtney D. DiNardo, Harry P. Erba, Guillermo Garcia-Manero, Ian R Lemieux, Geoffrey L. Uy, Gail J. Roboz, Eytan M. Stein, Robert K. Stuart, Anthony S. Stein, Arnaud Pigneux, Gabrielle T. Prince, David A. Sallman, Stephanie M. Kapsalis, and Prapti A. Patel
- Subjects
Cancer Research ,Cytopenia ,medicine.medical_specialty ,business.industry ,Induction chemotherapy ,Context (language use) ,Hematology ,medicine.disease ,Discontinuation ,Clinical trial ,Oncology ,Tolerability ,hemic and lymphatic diseases ,Pharmacodynamics ,Internal medicine ,medicine ,Adverse effect ,business - Abstract
Context: Mutations in isocitrate dehydrogenase 1 (IDH1) occur in ~3% of patients with MDS and are associated with increased transformation to acute myeloid leukemia (AML). IVO is an oral, potent, targeted inhibitor of the mutant IDH1 (mIDH1) enzyme and is FDA-approved for mIDH1 R/R AML and mIDH1 newly diagnosed AML in patients ≥75 years old or with comorbidities precluding the use of intensive induction chemotherapy. In the first-in-human, phase 1 study of IVO in patients with mIDH1 advanced hematologic malignancies (NCT02074839), 12 patients with R/R MDS, with median age 72.5 years (range 52–78), received IVO 500 mg once daily (QD). All patients received prior MDS treatment. Investigator-assessed ORR (CR + PR + marrow CR, per IWG 2006) was 75% (95% CI 43–95), with median duration of response of 21.4 months (95% CI 2.3–NE). Nine (75%) patients were transfusion-independent for ≥56 days during treatment. No dose-limiting toxicities or adverse events leading to treatment discontinuation were reported among patients with MDS. Based on these encouraging data, the FDA granted Breakthrough Therapy Designation to IVO in mIDH1 MDS; the study was amended to enroll additional patients with mIDH1 R/R MDS. Objective: To evaluate safety, tolerability, clinical activity, and pharmacokinetics/pharmacodynamics of IVO in patients with mIDH1 R/R MDS. Design: A sub-study of the single-arm, open-label, phase 1 dose escalation and expansion study of IVO in mIDH1 advanced hematologic malignancies, evaluating patients with R/R MDS. Patients must have R/R disease after prior standard therapy; high disease burden based on cytopenia and/or transfusion dependence at baseline; an Eastern Cooperative Oncology Group performance status score of 0–2; and be amenable to bone marrow aspirate and/or core biopsy at specified study timepoints. Patients with documented AML are not eligible. IVO 500 mg QD orally on days 1–28 of 28-day cycles. Results: Study is open; enrollment of ~23 patients from the US and France planned. Results not yet available. Conclusions: This sub-study will provide additional insights into safety, tolerability, clinical activity, and pharmacokinetics/pharmacodynamics of treatment with IVO in patients with mIDH1 R/R MDS. Funding: Agios; Servier.
- Published
- 2021
- Full Text
- View/download PDF
17. Comanagement Strategy Between Academic Institutions and Community Practices to Reduce Induction Mortality in Acute Promyelocytic Leukemia
- Author
-
Leonard T. Heffner, Sheldon Bolds, Asim Pati, Amy A. Langston, Morgan L. McLemore, Stephanie Debragga, Prachi Karkhanis, Robert K. Stuart, Anand Jillella, Michael R. Grunwald, Manila Gaddh, Vamsi Kota, Martha Arellano, Jorge E. Cortes, Chao Zhang, Catherine R. Caprara, Kathryn Sarah Simon, Mohamed M. El Geneidy, Shruthi H. Krishnamurthy, Jonathan M. Gerber, Elliott F. Winton, Jose Tongol, and Asad Bashey
- Subjects
Oncology ,Acute promyelocytic leukemia ,medicine.medical_specialty ,Universities ,Population ,MEDLINE ,Hemorrhage ,Seer data ,03 medical and health sciences ,0302 clinical medicine ,Leukemia, Promyelocytic, Acute ,Internal medicine ,medicine ,Humans ,Prospective Studies ,education ,Sweden ,education.field_of_study ,Oncology (nursing) ,business.industry ,Health Policy ,Middle Aged ,medicine.disease ,Clinical trial ,Leukemia ,030220 oncology & carcinogenesis ,business ,030215 immunology - Abstract
PURPOSE: Acute promyelocytic leukemia (APL) is a curable leukemia with > 90% survival in clinical trials. Population-based studies from Sweden and US SEER data have shown long-term survival rates of 62% and 65.7%, with the lower rate being from a higher percentage of early deaths. METHODS: In this prospective, multicenter trial, we developed a simplified algorithm that focused on prevention and early treatment of the three main causes of death: bleeding, differentiation syndrome, and infection. All patients with a diagnosis of APL were included. The initial 6 months were spent educating oncologists about early deaths in APL. At the time of suspicion of an APL, an expert was contacted. The algorithm was made available followed by discussion of the treatment plan. Communication between expert and treating physician was frequent in the first 2 weeks, during which time most deaths take place. RESULTS: Between September 2013 and April 2016, 120 patients enrolled in the study from 32 hospitals. The median age was 52.5 years, with 39% > 60 years and 25% with an age-adjusted Charlson comorbidity index > 4. Sixty-three percent of patients were managed at community centers. Two patients did not meet the criteria for analysis, and of 118 evaluable patients, 10 died, with an early mortality rate of 8.5%. With a median follow-up of 27.3 months, the overall survival was 84.5%. CONCLUSION: Induction mortality can be decreased and population-wide survival improved in APL with the use of standardized treatment guidelines. Support from experts who have more experience with induction therapy is crucial and helps to improve the outcomes.
- Published
- 2020
18. A Phase 1 Trial of CNDO-109–Activated Natural Killer Cells in Patients with High-Risk Acute Myeloid Leukemia
- Author
-
Michael Szarek, Leonid Gorelik, Sarah Cooley, John F. DiPersio, Robert K. Stuart, Julie M. Curtsinger, Amandeep Salhotra, Eric K. Rowinsky, Nova Silver, Mark W. Lowdell, Peter Westervelt, Todd A. Fehniger, Jeffrey S. Miller, and Timothy Mark Hillman
- Subjects
Adult ,Male ,0301 basic medicine ,Oncology ,medicine.medical_specialty ,Adoptive cell transfer ,Transplantation Conditioning ,Myeloid ,Cyclophosphamide ,medicine.medical_treatment ,Cell Count ,Immunotherapy, Adoptive ,Disease-Free Survival ,Article ,03 medical and health sciences ,Cell Line, Tumor ,Internal medicine ,Secondary Prevention ,medicine ,Humans ,Aged ,Transplantation ,business.industry ,Myeloid leukemia ,Microchimerism ,Hematology ,Immunotherapy ,Middle Aged ,Tissue Donors ,Fludarabine ,Killer Cells, Natural ,Leukemia, Myeloid, Acute ,Treatment Outcome ,030104 developmental biology ,medicine.anatomical_structure ,Cytokine ,Transplantation, Haploidentical ,Female ,business ,medicine.drug - Abstract
Natural killer (NK) cells are an emerging immunotherapy approach to acute myeloid leukemia (AML); however, the optimal approach to activate NK cells before adoptive transfer remains unclear. Human NK cells that are primed with the CTV-1 leukemia cell line lysate CNDO-109 exhibit enhanced cytotoxicity against NK-cell-resistant cell lines. To translate this finding to the clinic, CNDO-109-activated NK cells (CNDO-109-NK cells) isolated from related human leukocyte antigen complex (HLA)-haploidentical donors were evaluated in a phase 1 dose-escalation trial at doses of 3×10^5 (n=3), 1×10^6 (n=3), and 3×10^6 (n=6) cells/kg in patients with AML in first complete remission (CR1) at high risk for recurrence. Before CNDO-109-NK cell administration, patients were treated with lymphodepleting fludarabine/cyclophosphamide. CNDO-109-NK cells were well tolerated, and no dose-limiting toxicities were observed at the highest tested dose. The median relapse free (RFS) survival by dose level was 105 (3×10^5), 156 (1×10^6), and 337 (3×10^6) days. Two patients remain relapse-free in post-trial follow-up, with RFS durations exceeding 42.5 months. Donor NK cell micro-chimerism was detected on Day 7 in 10 of 12 patients, with 3 patients having evidence of donor cells on day 14 or later. This trial establishes that CNDO-109-NK cells generated from related HLA-haploidentical donors, cryopreserved, and then safely administered to AML patients with transient persistence without exogenous cytokine support. Three durable complete remissions of 32.6 to 47.6+ months were observed, suggesting additional clinical investigation of CNDO-109-NK cells for patients with myeloid malignancies, alone or in combination with additional immunotherapy strategies, is warranted.
- Published
- 2018
- Full Text
- View/download PDF
19. Durable Remissions with Ivosidenib inIDH1-Mutated Relapsed or Refractory AML
- Author
-
Courtney D. DiNardo, Hua Liu, Gail J. Roboz, Hongfang Wang, Martha Arellano, Samuel V. Agresta, Vickie Zhang, Ronan T. Swords, Martin S. Tallman, Geoffrey L. Uy, William B. Donnellan, David Dai, Stephanie M. Kapsalis, Christophe Willekens, Jessica K. Altman, James M. Foran, A. Pigneux, Meredith Goldwasser, Sung Choe, Gabriel N. Mannis, Eyal C. Attar, James L. Slack, Amir T. Fathi, Harry P. Erba, Bin Wu, Bin Fan, Robert K. Stuart, S. de Botton, Hagop M. Kantarjian, Richard Stone, Mikkael A. Sekeres, Katharine E. Yen, Hua Yang, Elie Traer, Eytan M. Stein, Robert H. Collins, Alice S. Mims, Daniel A. Pollyea, Anthony S. Stein, and Gabrielle T. Prince
- Subjects
Male ,0301 basic medicine ,Myeloid ,Pyridines ,Administration, Oral ,Cell Count ,Gastroenterology ,Hemoglobins ,0302 clinical medicine ,Recurrence ,hemic and lymphatic diseases ,Leukocytosis ,Enzyme Inhibitors ,Aged, 80 and over ,education.field_of_study ,Remission Induction ,Myeloid leukemia ,General Medicine ,Middle Aged ,Isocitrate Dehydrogenase ,Survival Rate ,Leukemia, Myeloid, Acute ,Leukemia ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Female ,medicine.symptom ,Adult ,medicine.medical_specialty ,Adolescent ,Population ,Glycine ,Enasidenib ,Young Adult ,03 medical and health sciences ,Refractory ,Internal medicine ,medicine ,Humans ,education ,Survival rate ,Aged ,Dose-Response Relationship, Drug ,business.industry ,medicine.disease ,030104 developmental biology ,Drug Resistance, Neoplasm ,Mutation ,business ,Follow-Up Studies - Abstract
Mutations in the gene encoding isocitrate dehydrogenase 1 ( IDH1) occur in 6 to 10% of patients with acute myeloid leukemia (AML). Ivosidenib (AG-120) is an oral, targeted, small-molecule inhibitor of mutant IDH1.We conducted a phase 1 dose-escalation and dose-expansion study of ivosidenib monotherapy in IDH1-mutated AML. Safety and efficacy were assessed in all treated patients. The primary efficacy population included patients with relapsed or refractory AML receiving 500 mg of ivosidenib daily with at least 6 months of follow-up.Overall, 258 patients received ivosidenib and had safety outcomes assessed. Among patients with relapsed or refractory AML (179 patients), treatment-related adverse events of grade 3 or higher that occurred in at least 3 patients were prolongation of the QT interval (in 7.8% of the patients), the IDH differentiation syndrome (in 3.9%), anemia (in 2.2%), thrombocytopenia or a decrease in the platelet count (in 3.4%), and leukocytosis (in 1.7%). In the primary efficacy population (125 patients), the rate of complete remission or complete remission with partial hematologic recovery was 30.4% (95% confidence interval [CI], 22.5 to 39.3), the rate of complete remission was 21.6% (95% CI, 14.7 to 29.8), and the overall response rate was 41.6% (95% CI, 32.9 to 50.8). The median durations of these responses were 8.2 months (95% CI, 5.5 to 12.0), 9.3 months (95% CI, 5.6 to 18.3), and 6.5 months (95% CI, 4.6 to 9.3), respectively. Transfusion independence was attained in 29 of 84 patients (35%), and patients who had a response had fewer infections and febrile neutropenia episodes than those who did not have a response. Among 34 patients who had a complete remission or complete remission with partial hematologic recovery, 7 (21%) had no residual detectable IDH1 mutations on digital polymerase-chain-reaction assay. No preexisting co-occurring single gene mutation predicted clinical response or resistance to treatment.In patients with advanced IDH1-mutated relapsed or refractory AML, ivosidenib at a dose of 500 mg daily was associated with a low frequency of grade 3 or higher treatment-related adverse events and with transfusion independence, durable remissions, and molecular remissions in some patients with complete remission. (Funded by Agios Pharmaceuticals; ClinicalTrials.gov number, NCT02074839 .).
- Published
- 2018
- Full Text
- View/download PDF
20. Phase 3 results for vosaroxin/cytarabine in the subset of patients ≥60 years old with refractory/early relapsed acute myeloid leukemia
- Author
-
Hamid Sayar, Heinz A. Horst, Stephen A. Strickland, Arnaud Pigneux, Gary J. Schiller, Michael Craig, Robert K. Stuart, Miklos Egyed, Renee Ward, Elias Jabbour, Jennifer A. Smith, Hagop M. Kantarjian, Jorge E. Cortes, Gary Acton, Utz Krug, Angelo Michele Carella, Farhad Ravandi, Ellen K. Ritchie, Judith A. Fox, Adam R. Craig, Norbert Vey, Jeffrey E. Lancet, Virginia M. Klimek, and Christian Recher
- Subjects
Oncology ,medicine.medical_specialty ,Standard of care ,business.industry ,Myeloid leukemia ,Hematology ,Vosaroxin ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,chemistry ,Refractory ,hemic and lymphatic diseases ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,Cytarabine ,In patient ,Online Only Articles ,business ,030215 immunology ,medicine.drug - Abstract
Refractory/early relapsed (Ref/eRel) acute myeloid leukemia (AML) in patients ≥60 years old is the most important unmet medical need in the salvage setting, where outcomes are exceptionally poor and no standard of care exists.[1][1] Vosaroxin is a first-in-class anticancer quinolone derivative
- Published
- 2018
- Full Text
- View/download PDF
21. Web-Based Clinical Trials Information for Patients: An Accrual Aid?
- Author
-
Lawrence B. Afrin, John Gentry, and Robert K. Stuart
- Published
- 1997
22. Phase 1b Study of Lower-Dose CPX-351 Plus Venetoclax As First-Line Treatment for Patients with AML Who Are Unfit for Intensive Chemotherapy: Preliminary Safety and Efficacy Results
- Author
-
Robert K. Stuart, Ronald S. Cheung, Vinod Pullarkat, Vijayalakshmi Chandrasekaran, Qi Wang, Stefan Faderl, Divya Chakravarthy, Tara L. Lin, Praneeth Baratam, and Geoffrey L. Uy
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Venetoclax ,Immunology ,Cell Biology ,Hematology ,Intensive chemotherapy ,Biochemistry ,First line treatment ,chemistry.chemical_compound ,chemistry ,Internal medicine ,Medicine ,business - Abstract
Background: CPX-351 (United States: Vyxeos ®; Europe: Vyxeos ® Liposomal), a dual-drug liposomal encapsulation of daunorubicin and cytarabine in a synergistic 1:5 molar ratio, is approved for the treatment of newly diagnosed therapy-related acute myeloid leukemia (AML) or AML with myelodysplasia-related changes in adults and pediatric patients aged ≥1 year in the United States and in adults in the European Union. In a phase 3 study in adults aged 60 to 75 years with newly diagnosed high-risk/secondary AML who were eligible for intensive chemotherapy (IC), after 5 years of follow-up CPX-351 significantly improved median overall survival versus conventional 7+3 cytarabine/daunorubicin, with a comparable safety profile. Venetoclax (VEN; BCL-2 inhibitor) + low-dose cytarabine has demonstrated efficacy in unfit patients with AML, and preclinical data support a rationale for combining CPX-351 + VEN. This study evaluates the safety and efficacy of lower-dose CPX-351 + VEN in adults with newly diagnosed AML who are considered unfit to receive IC. Methods: This is an ongoing, open-label, multicenter, phase 1b study (NCT04038437) to determine the recommended phase 2 dose (RP2D) and evaluate the safety and efficacy of lower-dose CPX-351 + VEN in adults with newly diagnosed AML who are considered unfit to receive IC. The dose-exploration phase (3+3 design; n ≤24) evaluated multiple dose levels of CPX-351 on Days 1 and 3 + VEN 400 mg on Days 2 to 21 of each cycle to determine the RP2D. Patients who achieve at least partial remission after 1 or 2 cycles may receive up to 4 similar cycles of CPX-351 + VEN. During the expansion phase, 20 additional patients will receive CPX-351 + VEN at the RP2D. Patients are assessed for response by morphology and measurable residual disease (MRD) testing and are monitored for safety and survival. Results: This preliminary analysis includes data from 14 enrolled patients. Patients were considered unfit for IC based on age ≥75 years (n = 7 [50%]) or health (aged 18 to 74 years with an EGOC performance status of 2 or 3 [n = 3 (21%)] and/or comorbidities [n = 5 (36%]). Overall, 50% of the patients had poor-risk cytogenetics, 64% were male, 71% had a diagnosis of de novo AML, and 29% had mutated TP53 (Table 1). Four patients received dose level 1 (CPX-351 20 units/m 2 [daunorubicin 8.8 mg/m 2 + cytarabine 20 mg/m 2] + VEN), with no dose-limiting toxicities (DLTs) observed in the 3 evaluable patients. Seven patients were subsequently enrolled in dose level 2 (CPX-351 40 units/m 2 [daunorubicin 17.6 mg/m 2 + cytarabine 40 mg/m 2] + VEN), with 6 patients evaluable for DLTs. At dose level 2, 1 patient experienced 2 DLTs (grade 3 tumor lysis syndrome and liver injury); review of the overall safety profile led to a protocol amendment that permitted de-escalation to dose level 1b (CPX-351 30 units/m 2 [daunorubicin 13.2 mg/m 2 + cytarabine 30 mg/m 2] + VEN). Three patients received dose level 1b with no DLTs and a safety profile comparable to dose level 1. Together, these data established dose level 1b as the RP2D. The most common nonhematologic treatment-emergent adverse events (TEAEs) were gastrointestinal events and peripheral edema (Table 2). The majority (86%) of patients experienced a grade ≥3 TEAE, primarily myelosuppression; the only nonhematologic grade ≥3 TEAE in >10% of patients was tumor lysis syndrome (14%). No patient experienced early mortality by Day 30; the mortality rate at Day 60 was 7% due to 1 death in the dose level 1 cohort (myocardial infarction considered unrelated to treatment). Among the 12 patients evaluable for efficacy across dose levels, 8 (67%) achieved a best response of complete remission (CR): 3/4 (75%) in dose level 1, 3/5 (60%) in dose level 2, and 2/3 (67%) in dose level 1b. All patients who achieved a best response of CR entered into either CR or CR with incomplete neutrophil or platelet recovery after the first treatment cycle. Confirmation of MRD status is currently ongoing. Conclusions: Preliminary results from this ongoing phase 1b study established a RP2D of CPX-351 30 units/m 2 on Days 1 and 3 + VEN 400 mg on Days 2 to 21 in adults with newly diagnosed AML who were considered unfit to receive IC. The combination of lower-dose CPX-351 + VEN was generally well tolerated and demonstrated promising initial efficacy, with achievement of CR in the majority of patients. This study is ongoing and enrolling 20 additional patients to further evaluate the RP2D. Figure 1 Figure 1. Disclosures Uy: Novartis: Consultancy; GlaxoSmithKline: Consultancy; Agios: Consultancy; AbbVie: Consultancy; Macrogenics: Research Funding; Astellas: Honoraria, Speakers Bureau; Genentech: Consultancy; Jazz: Consultancy. Pullarkat: AbbVie, Amgen, Genentech, Jazz Pharmaceuticals, Novartis, Pfizer, and Servier: Membership on an entity's Board of Directors or advisory committees; Amgen, Dova, and Novartis: Consultancy, Honoraria. Baratam: Celgene/Bristol Myers Squibb: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau. Stuart: Sunesis Pharmaceuticals: Consultancy; Sunesis Pharmaceuticals: Honoraria; Sunesis Pharmaceuticals: Other: Travel Support; Agios, Astellas Pharma, Bayer AG, Incyte, Jazz Pharmaceuticals, Ono Pharmaceuticals, and Sunesis Pharmaceuticals: Research Funding. Faderl: Jazz Pharmaceuticals: Current Employment, Current equity holder in publicly-traded company. Chandrasekaran: Jazz Pharmaceuticals: Current Employment, Current equity holder in publicly-traded company. Wang: Jazz Pharmaceuticals: Current Employment, Current equity holder in publicly-traded company. Chakravarthy: Jazz Pharmaceuticals: Current Employment, Current equity holder in publicly-traded company. Cheung: Jazz Pharmaceuticals: Current Employment, Current equity holder in publicly-traded company. Lin: AbbVie, Aptevo Therapeutics, Astellas Pharma, Bio-Path Holdings, Celgene, Celyad, Genentech-Roche, Gilead Sciences, Incyte, Jazz Pharmaceuticals, Novartis, Ono Pharmaceutical, Pfizer, Prescient Therapeutics, Seattle Genetics, Tolero, Trovagene: Research Funding. OffLabel Disclosure: combination of CPX-351 [Vyxeos] and venetoclax in adults with previously untreated AML
- Published
- 2021
- Full Text
- View/download PDF
23. Consolidation outcomes in CPX-351 versus cytarabine/daunorubicin-treated older patients with high-risk/secondary acute myeloid leukemia
- Author
-
Ellen K. Ritchie, Donna E. Hogge, Kathleen F. Villa, Stephen A. Strickland, Jeffrey E. Lancet, Robert J. Ryan, Stuart L. Goldberg, Robert K. Stuart, Jonathan E. Kolitz, Michael Chiarella, Jorge E. Cortes, and Arthur C. Louie
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Daunorubicin ,Phases of clinical research ,Newly diagnosed ,03 medical and health sciences ,0302 clinical medicine ,Older patients ,Internal medicine ,Cytarabine/daunorubicin ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Secondary Acute Myeloid Leukemia ,Humans ,Aged ,business.industry ,Cytarabine ,Hematology ,Middle Aged ,Leukemia, Myeloid, Acute ,030220 oncology & carcinogenesis ,business ,030215 immunology ,medicine.drug - Abstract
CPX-351 is a dual-drug liposomal encapsulation of cytarabine/daunorubicin. In a phase 3 study (ClinicalTrials.gov Identifier: NCT01696084), patients aged 60-75 years with newly diagnosed, high-risk/secondary AML received 1-2 induction cycles with CPX-351 or 7 + 3 chemotherapy; those achieving complete remission (including with incomplete platelet or neutrophil recovery) could receive up to 2 consolidation cycles with CPX-351 or 5 + 2 chemotherapy, respectively. In this exploratory analysis of the subgroup of patients who received consolidation, median overall survival was prolonged among patients receiving CPX-351 induction/consolidation versus 7 + 3/5 + 2 (25.43 vs. 8.53 months; HR = 0.44 [95% CI: 0.25-0.77]). The safety profile of CPX-351 consolidation was consistent with that of the overall study. Outpatient administration of CPX-351 consolidation occurred in 51%-61% of patients and did not diminish overall survival. These findings suggest consolidation with CPX-351 in this patient population contributed to the prolonged overall survival versus 7 + 3/5 + 2, building upon findings from the overall study population, and provide evidence that, with careful monitoring, some patients can successfully receive CPX-351 as outpatients.
- Published
- 2019
24. Inferior Outcomes with Cyclosporine and Mycophenolate Mofetil after Myeloablative Allogeneic Hematopoietic Cell Transplantation
- Author
-
Dennis Dong Hwan Kim, Jean-Yves Cahn, Olle Ringdén, Joseph Pidala, Margaret L. MacMillan, J. Schriber, Leo F. Verdonck, Mahmoud Aljurf, Joseph H. Antin, Robert K. Stuart, Ayman Saad, Mitchell S. Cairo, Eneida R. Nemecek, Ying Liu, David I. Marks, Daniel R. Couriel, Saurabh Chhabra, Stephen R. Spellman, Usama Gergis, Mukta Arora, Betty K. Hamilton, Christopher E. Dandoy, Taiga Nishihori, Yoshihiro Inamoto, Sung Won Choi, Bipin N. Savani, Amin M. Alousi, Christopher Bredeson, Edmund K. Waller, Gérard Socié, Robert Peter Gale, Navneet S. Majhail, Luciano J. Costa, Sachiko Seo, Mona Wirk, Rammurti T. Kamble, Takanori Teshima, Michael T. Hemmer, and Peiman Hematti
- Subjects
Adult ,Male ,medicine.medical_specialty ,Combination therapy ,Adolescent ,Urology ,Graft vs Host Disease ,chemical and pharmacologic phenomena ,Mycophenolate ,Disease-Free Survival ,Article ,immune system diseases ,Medicine ,Humans ,Aged ,Transplantation ,business.industry ,Hazard ratio ,Hematopoietic Stem Cell Transplantation ,Hematology ,Middle Aged ,Mycophenolic Acid ,medicine.disease ,Allografts ,Tacrolimus ,Calcineurin ,Survival Rate ,surgical procedures, operative ,Graft-versus-host disease ,Cyclosporine ,Methotrexate ,Female ,business ,medicine.drug - Abstract
Combination therapy with a calcineurin inhibitor (CNI), such as cyclosporine (CSA) or tacrolimus (Tac), and methotrexate (MTX) or mycophenolate mofetil (MMF) is a widely used approach to graft-versus-host disease (GVHD) prevention. Data on the comparative effectiveness of MMF compared with MTX are limited and conflicting, however. We analyzed data from the Center for International Blood and Marrow Transplant Research for adult patients undergoing first myeloablative hematopoietic cell transplantation (HCT) from an HLA-identical matched related donor (MRD; n = 3979) or matched unrelated donor (URD; n = 4163) using CSA+MMF, CSA+MTX, Tac+MMF, or Tac+MTX for GVHD prevention between 2000 and 2013. Within the MRD cohort, 2252 patients received CSA+MTX, 1391 received Tac+MTX, 114 received CSA+MMF, and 222 received Tac+MMF. Recipients of CSA+MMF had a higher incidence of acute GVHD grade II-IV (hazard ratio [HR], 1.65; 95% confidence interval [CI], 1.24 to 2.20; P.001) and grade III-IV (HR, 1.92; 95% CI, 1.31 to 2.83; P.001) compared with Tac+MTX. The use of CSA+MMF was also associated with inferior overall survival (OS) (HR, 2.31; 95% CI, 1.73 to 3.09; P.001) due to higher transplantation-related mortality (TRM) (HR, 4.03; 95% CI, 2.61 to 6.23; P.001) compared with Tac+MTX. Within the URD cohort, 974 patients received CSA+MTX, 2697 received Tac+MTX, 68 received CSA+MMF, and 424 received Tac+MMF. CSA+MMF was again significantly associated with a higher incidence of grade III-IV acute GVHD (HR, 2.31; 95% CI, 1.57 to 3.42; P0001), worse OS (HR, 2.36; 95% CI, 1.67 to 3.35; P.001), and higher TRM (HR, 3.09; 95% CI, 2.00 to 4.77; P.001), compared with Tac+MTX and other regimens. Thus, this large retrospective comparison of MMF versus MTX in combination with CSA or Tac demonstrates significantly worse GVHD and survival outcomes with CSA+MMF compared with Tac+MTX.
- Published
- 2019
25. MDS-265: Ivosidenib (IVO) in Patients with IDH1-Mutant Relapsed/Refractory Myelodysplastic Syndrome (R/R MDS): Updated Enrollment of a Phase 1 Dose Escalation and Expansion Study
- Author
-
Martha Arellano, Anthony S. Stein, David A. Sallman, Arnaud Pigneux, Gabrielle T. Prince, Guillermo Garcia-Manero, Prapti A. Patel, Thomas Winkler, Ian R Lemieux, Hua Liu, Richard Stone, Stéphane de Botton, Justin M. Watts, James M. Foran, Amir T. Fathi, Abdulafeez Oluyadi, Courtney D. DiNardo, Xavier Thomas, Bin Wu, Robert K. Stuart, Geoffrey L. Uy, Harry P. Erba, Gail J. Roboz, and Eytan M. Stein
- Subjects
Cancer Research ,medicine.medical_specialty ,Cytopenia ,education.field_of_study ,business.industry ,Myelodysplastic syndromes ,Population ,Induction chemotherapy ,Context (language use) ,Hematology ,medicine.disease ,Clinical trial ,Oncology ,Tolerability ,hemic and lymphatic diseases ,Internal medicine ,Pharmacodynamics ,Medicine ,business ,education - Abstract
Context: IVO is an oral, potent, targeted inhibitor of the mutant IDH1 (mIDH1) enzyme, approved in the US for the treatment of AML with a susceptible IDH1 mutation in adults with newly diagnosed AML ≥75 years of age or having comorbidities precluding intensive induction chemotherapy, and in adults with R/R AML. In a phase 1 dose escalation and expansion study of IVO in mIDH1 advanced hematologic malignancies ( NCT02074839 ), 12 patients with R/R MDS received IVO 500 mg once daily (QD). Median age was 72.5 years (range 52–78). All patients had received prior treatment for MDS, with 3 (25.0%) and 1 (8.3%) having received 2 or ≥3 prior therapies, respectively. Investigator-assessed ORR (CR + PR + marrow CR) per International Working Group 2006 criteria was 75.0% (95% CI 42.8%, 94.5%) with a median duration of 21.4 months (95% CI 2.3, not estimable). Nine patients (75.0%) were transfusion independent for ≥56 days during study treatment. Based on these data, the FDA recently granted a Breakthrough Therapy Designation status for IVO monotherapy in this indication, and the study has been amended to enroll additional mIDH1 R/R MDS patients. Objective: To assess safety, tolerability, clinical activity, pharmacokinetics, and pharmacodynamics of IVO in patients with mIDH1 R/R MDS. Design: This is a sub-study of the phase 1 study of IVO in mIDH1 advanced hematologic malignancies to evaluate patients with mIDH1 R/R MDS. Patients must have R/R disease after treatment with standard agents indicated for MDS and high disease burden based on cytopenia and/or transfusion dependence at baseline. IVO will be administered at a dose of 500 mg QD orally on Days 1–28 of 28-day cycles. Results: The study is open and will enroll ∼23 patients from the US and France; results not yet available. Conclusions: The favorable efficacy and safety of IVO in the small population of patients with mIDH1 R/R MDS in the phase 1 clinical study supports further evaluation in this MDS sub-study. This sub-study will provide additional insights into safety, tolerability, clinical activity, and pharmacokinetics/pharmacodynamics of treatment with IVO in patients with mIDH1 R/R MDS. Funding: Agios.
- Published
- 2020
- Full Text
- View/download PDF
26. Five-year final results of a phase III study of CPX-351 versus 7+3 in older adults with newly diagnosed high-risk/secondary AML
- Author
-
Stephen A. Strickland, Jonathan E. Kolitz, Gary J. Schiller, Donna E. Hogge, Robert K. Stuart, Stefan Faderl, Laura F. Newell, Jorge E. Cortes, Tara L. Lin, Daniel H. Ryan, Jeffrey E. Lancet, Matthew J. Wieduwilt, Scott R. Solomon, Ellen K. Ritchie, Geoffrey L. Uy, Dale L. Bixby, and Yu-Lin Chang
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Liposome ,business.industry ,Daunorubicin ,Newly diagnosed ,Secondary AML ,03 medical and health sciences ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Internal medicine ,CYTARABINE LIPOSOME ,Cytarabine ,Medicine ,business ,030215 immunology ,medicine.drug - Abstract
7510 Background: CPX-351 (Vyxeos; daunorubicin and cytarabine liposome for injection), a dual-drug liposomal encapsulation of cytarabine [C] and daunorubicin [D], is approved by the FDA and EMA for the treatment of adults with newly diagnosed therapy-related AML or AML with myelodysplasia-related changes. Primary analysis of the pivotal phase 3 study (NCT01696084) that formed the basis for these approvals evaluated patients (pts) aged 60-75 y with newly diagnosed high-risk/secondary AML and found that CPX-351 significantly improved median overall survival (OS) vs conventional 7+3, with a comparable safety profile. Here, we report the prospectively planned final 5-y follow-up results from this phase 3 study. Methods: Pts were randomized 1:1 to receive ≤2 induction cycles of CPX-351 (100 units/m2 [C 100 mg/m2 + D 44 mg/m2] as a 90-min infusion on Days 1, 3, 5 [2nd induction: Days 1, 3]) or 7+3 (C 100 mg/m2/d continuously for 7 d + D 60 mg/m2 on Days 1-3 [2nd induction: 5+2]). Pts achieving complete remission (CR) or CR with incomplete platelet or neutrophil recovery could receive up to 2 consolidation cycles. Pts could receive a hematopoietic cell transplant (HCT) at the physician’s discretion. Pts were followed until death or up to 5 y following randomization. Results: In total, 309 pts were randomized to CPX-351 (n = 153) or 7+3 (n = 156). The survival rate at 5 y was higher for CPX-351 vs 7+3 (18% vs 8%; Table). Among pts who died, the most common primary cause of death was progressive leukemia in both arms (CPX-351: 56%; 7+3: 53%). After a median follow-up of 60.65 mo, improved median OS with CPX-351 vs 7+3 was maintained: 9.33 vs 5.95 mo; Kaplan-Meier (KM) OS curves plateaued at ~30 mo. HCT was received by 53 (35%) vs 39 (25%) pts after CPX-351 vs 7+3; among these pts, the survival rate at 5 y was higher for CPX-351 vs 7+3 (52% vs 23%), and median OS landmarked from the HCT date was not reached for CPX-351 vs 10.25 mo for 7+3 (Table). Conclusions: After 5 y of follow-up, improved OS was maintained in this phase 3 study, supporting that CPX-351 has the ability to produce or contribute to long-term remission and survival in older pts with newly diagnosed high-risk/secondary AML. Clinical trial information: NCT01696084 . [Table: see text]
- Published
- 2020
- Full Text
- View/download PDF
27. Myeloablative vs reduced-intensity conditioning allogeneic hematopoietic cell transplantation for chronic myeloid leukemia
- Author
-
Ronald Sobecks, Wael Saber, Richard F. Olsson, Jack W. Hsu, Matt Kalaycio, Melhem Solh, Zachariah DeFilipp, Sandeep Jain, Nirav N. Shah, Ann E. Woolfrey, Attaphol Pawarode, Andrew Daly, Jacob M. Rowe, Betty K. Hamilton, Amer Assal, Zhen-Huan Hu, Hillard M. Lazarus, Siddhartha Ganguly, Yoshihiro Inamoto, Harry C. Schouten, Olle Ringdén, Celalettin Ustun, Sunita Nathan, Kwang Woo Ahn, Edward A. Copelan, Mark R. Litzow, Ayman Saad, Jean A. Yared, Abraham S. Kanate, Robert K. Stuart, Uday R. Popat, Saurabh Chhabra, Jan Cerny, Bipin N. Savani, H. Jean Khoury, Shahinaz M. Gadalla, Robert Peter Gale, Sachiko Seo, Edwin P. Alyea, and Gerhard C. Hildebrandt
- Subjects
Oncology ,Adult ,Male ,medicine.medical_specialty ,Transplantation Conditioning ,Adolescent ,medicine.medical_treatment ,Graft vs Host Disease ,Hematopoietic stem cell transplantation ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Recurrence ,Internal medicine ,hemic and lymphatic diseases ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,medicine ,Humans ,Transplantation, Homologous ,Cumulative incidence ,Prospective Studies ,Survival rate ,neoplasms ,Alemtuzumab ,Proportional Hazards Models ,Transplantation ,Hematology ,business.industry ,Hazard ratio ,Hematopoietic Stem Cell Transplantation ,Myeloid leukemia ,Middle Aged ,medicine.disease ,respiratory tract diseases ,Survival Rate ,Leukemia ,surgical procedures, operative ,030220 oncology & carcinogenesis ,Multivariate Analysis ,Female ,business ,030215 immunology - Abstract
Allogeneic hematopoietic cell transplantation (allo-HCT) is a potentially curative treatment of chronic myeloid leukemia (CML). Optimal conditioning intensity for allo-HCT for CML in the era of tyrosine kinase inhibitors (TKIs) is unknown. Using the Center for International Blood and Marrow Transplant Research database, we sought to determine whether reduced-intensity/nonmyeloablative conditioning (RIC) allo-HCT and myeloablative conditioning (MAC) result in similar outcomes in CML patients. We evaluated 1395 CML allo-HCT recipients between the ages of 18 and 60 years. The disease status at transplant was divided into the following categories: chronic phase 1, chronic phase 2 or greater, and accelerated phase. Patients in blast phase at transplant and alternative donor transplants were excluded. The primary outcome was overall survival (OS) after allo-HCT. MAC (n = 1204) and RIC allo-HCT recipients (n = 191) from 2007 to 2014 were included. Patient, disease, and transplantation characteristics were similar, with a few exceptions. Multivariable analysis showed no significant difference in OS between MAC and RIC groups. In addition, leukemia-free survival and nonrelapse mortality did not differ significantly between the 2 groups. Compared with MAC, the RIC group had a higher risk of early relapse after allo-HCT (hazard ratio [HR], 1.85; P = .001). The cumulative incidence of chronic graft-versus-host disease (cGVHD) was lower with RIC than with MAC (HR, 0.77; P = .02). RIC provides similar survival and lower cGVHD compared with MAC and therefore may be a reasonable alternative to MAC for CML patients in the TKI era.
- Published
- 2018
28. CPX-351 (cytarabine and daunorubicin) Liposome for Injection Versus Conventional Cytarabine Plus Daunorubicin in Older Patients With Newly Diagnosed Secondary Acute Myeloid Leukemia
- Author
-
Jeffrey E. Lancet, Daniel H. Ryan, Michael Chiarella, Robert K. Stuart, Geoffrey L. Uy, Jorge E. Cortes, Dale L. Bixby, Kamalika Banerjee, Richard Stone, Antje Hoering, Matthew J. Wieduwilt, Arthur C. Louie, Bruno C. Medeiros, Tara L. Lin, Donna E. Hogge, Laura F. Newell, Jonathan E. Kolitz, Scott R. Solomon, Ellen K. Ritchie, Stephen A. Strickland, and Gary J. Schiller
- Subjects
0301 basic medicine ,Male ,Myeloid ,Cancer Research ,medicine.medical_specialty ,Daunorubicin ,medicine.medical_treatment ,Clinical Sciences ,Oncology and Carcinogenesis ,Enasidenib ,Acute ,Gastroenterology ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Neoplasms ,Antineoplastic Combined Chemotherapy Protocols ,Hematologic Malignancy ,Medicine ,Secondary Acute Myeloid Leukemia ,Humans ,Oncology & Carcinogenesis ,Aged ,Chemotherapy ,Leukemia ,business.industry ,Hazard ratio ,Cytarabine ,Neoplasms, Second Primary ,Middle Aged ,medicine.disease ,Regimen ,Leukemia, Myeloid, Acute ,030104 developmental biology ,Nanomedicine ,Second Primary ,Oncology ,030220 oncology & carcinogenesis ,Liposomes ,Female ,business ,RAPID COMMUNICATION ,medicine.drug - Abstract
Purpose CPX-351 is a dual-drug liposomal encapsulation of cytarabine and daunorubicin that delivers a synergistic 5:1 drug ratio into leukemia cells to a greater extent than normal bone marrow cells. Prior clinical studies demonstrated a sustained drug ratio and exposure in vivo and prolonged survival versus standard-of-care cytarabine plus daunorubicin chemotherapy (7+3 regimen) in older patients with newly diagnosed secondary acute myeloid leukemia (sAML). Patients and Methods In this open-label, randomized, phase III trial, 309 patients age 60 to 75 years with newly diagnosed high-risk/sAML received one to two induction cycles of CPX-351 or 7+3 followed by consolidation therapy with a similar regimen. The primary end point was overall survival. Results CPX-351 significantly improved median overall survival versus 7+3 (9.56 v 5.95 months; hazard ratio, 0.69; 95% CI, 0.52 to 0.90; one-sided P = .003). Overall remission rate was also significantly higher with CPX-351 versus 7+3 (47.7% v 33.3%; two-sided P = .016). Improved outcomes were observed across age-groups and AML subtypes. The incidences of nonhematologic adverse events were comparable between arms, despite a longer treatment phase and prolonged time to neutrophil and platelet count recovery with CPX-351. Early mortality rates with CPX-351 and 7+3 were 5.9% and 10.6% (two-sided P = .149) through day 30 and 13.7% and 21.2% (two-sided P = .097) through day 60. Conclusion CPX-351 treatment is associated with significantly longer survival compared with conventional 7+3 in older adults with newly diagnosed sAML. The safety profile of CPX-351 was similar to that of conventional 7+3 therapy.
- Published
- 2018
29. S876 GILTERITINIB SIGNIFICANTLY PROLONGS OVERALL SURVIVAL IN PATIENTS WITH FLT3-MUTATED (FLT3MUT+) RELAPSED/REFRACTORY (R/R) ACUTE MYELOID LEUKEMIA (AML): RESULTS FROM THE PHASE 3 ADMIRAL TRIAL
- Author
-
Rebecca L. Olin, Celalettin Ustun, Erkut Bahceci, A. Di Stasi, Wen-Chien Chou, Pau Montesinos, Naoko Hosono, Fabio Ciceri, Amir T. Fathi, Francesco Fabbiano, Ellin Berman, Nikolai A. Podoltsev, X. Liu, Christian Recher, Je-Hwan Lee, Alexander E. Perl, Maria R. Baer, Hisayuki Yokoyama, Chaofeng Liu, Andreas Neubauer, Richard A. Larson, Stefania Paolini, Margaret Kasner, Robert K. Stuart, Jorge E. Cortes, Giovanni Martinelli, Timothy S. Pardee, Sung-Soo Yoon, and Mark J. Levis
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Internal medicine ,Relapsed refractory ,medicine ,Gilteritinib ,Overall survival ,Myeloid leukemia ,In patient ,Hematology ,business - Published
- 2019
- Full Text
- View/download PDF
30. Vosaroxin plus cytarabine versus placebo plus cytarabine in patients with first relapsed or refractory acute myeloid leukaemia (VALOR): a randomised, controlled, double-blind, multinational, phase 3 study
- Author
-
Ellen K. Ritchie, Judith A. Fox, Cyrus R. Mehta, Olatoyosi Odenike, Andrew H. Wei, Renee Ward, Gail J. Roboz, Michael Heuser, Norbert Vey, Gary Acton, Stephen A. Strickland, Elias Jabbour, Jeffrey E. Lancet, Arnaud Pigneux, Adam R. Craig, Donna E. Hogge, Gary J. Schiller, Gianluca Gaidano, Michael Craig, Violaine Havelange, Bayard L. Powell, Lloyd E. Damon, Xavier Thomas, Heinz A. Horst, Jennifer A. Smith, Christian Recher, Harry P. Erba, Robert K. Stuart, Angelo Michele Carella, Farhad Ravandi, Hagop M. Kantarjian, Jorge E. Cortes, Johan Maertens, Hans Günter Derigs, Virginia M. Klimek, Hamid Sayar, UCL - SSS/DDUV - Institut de Duve, and UCL - (SLuc) Service d'hématologie
- Subjects
Myeloid ,Male ,Phases of clinical research ,Kaplan-Meier Estimate ,Vosaroxin ,Gastroenterology ,chemistry.chemical_compound ,Antineoplastic Combined Chemotherapy Protocols ,80 and over ,Clinical endpoint ,Cancer ,Aged, 80 and over ,Leukemia ,Remission Induction ,Cytarabine ,Hematology ,Middle Aged ,Leukemia, Myeloid, Acute ,Treatment Outcome ,Local ,Oncology ,6.1 Pharmaceuticals ,Female ,medicine.drug ,Adult ,medicine.medical_specialty ,Clinical Trials and Supportive Activities ,Oncology and Carcinogenesis ,Acute ,Neutropenia ,Placebo ,Article ,Disease-Free Survival ,Double-Blind Method ,Clinical Research ,Internal medicine ,medicine ,Humans ,Oncology & Carcinogenesis ,Naphthyridines ,Aged ,Intention-to-treat analysis ,business.industry ,Evaluation of treatments and therapeutic interventions ,medicine.disease ,Surgery ,Thiazoles ,Neoplasm Recurrence ,chemistry ,Neoplasm Recurrence, Local ,business ,Febrile neutropenia - Abstract
BACKGROUND: Safe and effective treatments are urgently needed for patients with relapsed or refractory acute myeloid leukaemia. We investigated the efficacy and safety of vosaroxin, a first-in-class anticancer quinolone derivative, plus cytarabine in patients with relapsed or refractory acute myeloid leukaemia. METHODS: This phase 3, double-blind, placebo-controlled trial was undertaken at 101 international sites. Eligible patients with acute myeloid leukaemia were aged 18 years of age or older and had refractory disease or were in first relapse after one or two cycles of previous induction chemotherapy, including at least one cycle of anthracycline (or anthracenedione) plus cytarabine. Patients were randomly assigned 1:1 to vosaroxin (90 mg/m(2) intravenously on days 1 and 4 in a first cycle; 70 mg/m(2) in subsequent cycles) plus cytarabine (1 g/m(2) intravenously on days 1-5) or placebo plus cytarabine through a central interactive voice system with a permuted block procedure stratified by disease status, age, and geographical location. All participants were masked to treatment assignment. The primary efficacy endpoint was overall survival and the primary safety endpoint was 30-day and 60-day all-cause mortality. Efficacy analyses were done by intention to treat; safety analyses included all treated patients. This study is registered with ClinicalTrials.gov, number NCT01191801. FINDINGS: Between Dec 17, 2010, and Sept 25, 2013, 711 patients were randomly assigned to vosaroxin plus cytarabine (n=356) or placebo plus cytarabine (n=355). At the final analysis, median overall survival was 7·5 months (95% CI 6·4-8·5) in the vosaroxin plus cytarabine group and 6·1 months (5·2-7·1) in the placebo plus cytarabine group (hazard ratio 0·87, 95% CI 0·73-1·02; unstratified log-rank p=0·061; stratified p=0·024). A higher proportion of patients achieved complete remission in the vosaroxin plus cytarabine group than in the placebo plus cytarabine group (107 [30%] of 356 patients vs 58 [16%] of 355 patients, p
- Published
- 2015
- Full Text
- View/download PDF
31. Dacetuzumab plus rituximab, ifosfamide, carboplatin and etoposide as salvage therapy for patients with diffuse large B-cell lymphoma relapsing after rituximab, cyclophosphamide, doxorubicin, vincristine and prednisolone: a randomized, double-blind, placebo-controlled phase 2b trial
- Author
-
Ranjana H. Advani, Andres Forero-Torres, Stephen M. Ansell, Luis Fayad, Bertrand Coiffier, Robert K. Stuart, David Belada, Kazimierz Kuliczkowski, Edmund Ng, Jonathan G. Drachman, and Nancy L. Bartlett
- Subjects
Adult ,Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Prednisolone ,Salvage therapy ,Kaplan-Meier Estimate ,Pharmacology ,Antibodies, Monoclonal, Humanized ,Carboplatin ,chemistry.chemical_compound ,Double-Blind Method ,immune system diseases ,hemic and lymphatic diseases ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Ifosfamide ,Cyclophosphamide ,Etoposide ,Aged ,Salvage Therapy ,business.industry ,Remission Induction ,Dacetuzumab ,Hematology ,Middle Aged ,medicine.disease ,Thrombocytopenia ,Treatment Outcome ,Cough ,chemistry ,Doxorubicin ,Vincristine ,Female ,Rituximab ,Lymphoma, Large B-Cell, Diffuse ,Neoplasm Recurrence, Local ,business ,Diffuse large B-cell lymphoma ,medicine.drug - Abstract
Single-agent dacetuzumab has demonstrated antitumor activity in relapsed/refractory diffuse large B-cell lymphoma (DLBCL). Preclinical data demonstrated improved dacetuzumab antitumor activity in combination with rituximab, ± chemotherapy. We designed a phase 2b, double-blind, placebo-controlled trial to compare rituximab, ifosfamide, carboplatin and etoposide (R-ICE) + dacetuzumab with R-ICE + placebo in patients with DLBCL who relapsed after rituximab, cyclophosphamide, doxorubicin, vincristine and prednisolone (R-CHOP) (ClinicalTrials.gov #NCT00529503). The primary endpoint was complete response (CR); additional endpoints included failure-free survival and overall survival (OS). Overall, 151 patients were randomized (75 dacetuzumab, 76 placebo). No notable differences between arms in demographics or subsequent treatment parameters were observed. Cytopenias, cough and infection were more frequent with dacetuzumab. Futility analysis failed to demonstrate higher CR rates with dacetuzumab (36% dacetuzumab, 42% placebo); consequently, enrollment was stopped. Unplanned post hoc analysis showed that patients who underwent subsequent autologous stem cell transplant experienced improvement in OS (hazard ratio = 0.195, p = 0.004), which may be explained by potential immunomodulatory effects of dacetuzumab on antigen-presenting cells.
- Published
- 2015
- Full Text
- View/download PDF
32. Elevation of c-MYC Disrupts HLA Class II–Mediated Immune Recognition of Human B Cell Tumors
- Author
-
Bettina Kempkes, Janice S. Blum, Shereen Amria, Jason M. God, Christine Cameron, Georg W. Bornkamm, Robert K. Stuart, Azizul Haque, Azim Hossain, and Janette Figueroa
- Subjects
Lymphoma, B-Cell ,T cell ,Blotting, Western ,Immunology ,Human leukocyte antigen ,Biology ,Mass Spectrometry ,Article ,Flow cytometry ,Proto-Oncogene Proteins c-myc ,Tumor Cells, Cultured ,medicine ,Humans ,Immunology and Allergy ,Transcription factor ,B cell ,Antigen Presentation ,Oncogene ,medicine.diagnostic_test ,Lymphoblast ,Histocompatibility Antigens Class II ,Flow Cytometry ,medicine.disease ,Lymphoma ,medicine.anatomical_structure ,Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization ,Cancer research ,Tumor Escape - Abstract
Elevated levels of the transcription factor c-myc are strongly associated with various cancers, and in particular B cell lymphomas. Although many of c-MYC’s functions have been elucidated, its effect on the presentation of Ag through the HLA class II pathway has not been reported previously. This is an issue of considerable importance, given the low immunogenicity of many c-MYC–positive tumors. We report in this paper that increased c-MYC expression has a negative effect on the ability of B cell lymphomas to functionally present Ags/peptides to CD4+ T cells. This defect was associated with alterations in the expression of distinct cofactors as well as interactions of antigenic peptides with class II molecules required for the presentation of class II–peptide complexes and T cell engagement. Using early passage Burkitt’s lymphoma (BL) tumors and transformed cells, we show that compared with B lymphoblasts, BL cells express decreased levels of the class II editor HLA-DM, lysosomal thiol-reductase GILT, and a 47-kDa enolase-like protein. Functional Ag presentation was partially restored in BL cells treated with a c-MYC inhibitor, demonstrating the impact of this oncogene on Ag recognition. This restoration of HLA class II–mediated Ag presentation in early passage BL tumors/cells was linked to enhanced HLA-DM expression and a concurrent decrease in HLA-DO in BL cells. Taken together, these results reveal c-MYC exerts suppressive effects at several critical checkpoints in Ag presentation, which contribute to the immunoevasive properties of BL tumors.
- Published
- 2015
- Full Text
- View/download PDF
33. Vosaroxin in relapsed/refractory acute myeloid leukemia: efficacy and safety in the context of the current treatment landscape
- Author
-
Robert K. Stuart and Valeriy Sedov
- Subjects
0301 basic medicine ,Oncology ,Chemotherapy ,medicine.medical_specialty ,Anthracycline ,business.industry ,medicine.medical_treatment ,Myeloid leukemia ,Reviews ,Context (language use) ,Hematology ,Vosaroxin ,03 medical and health sciences ,chemistry.chemical_compound ,Regimen ,030104 developmental biology ,0302 clinical medicine ,chemistry ,Refractory ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,Cytarabine ,business ,medicine.drug - Abstract
Treatment for acute myeloid leukemia (AML) generally consists of a combination of cytarabine and an anthracycline. Although induction therapy leads to complete remission (CR) for most patients, refractoriness to chemotherapy or relapse after initial response is associated with poor outcomes. The 1-year survival rates after first relapse have been reported at 29%, declining to 11% at 5 years. Prognosis is particularly poor among older patients whose higher prevalence of unfavorable cytogenetics and high frequency of comorbidities diminish their ability to tolerate intensive chemotherapy. There is no standard of care for relapsed/refractory (R/R) AML, and no new therapies have shown consistently superior outcomes in this setting in over two decades. Vosaroxin is an anticancer quinolone derivative (AQD) that was evaluated in combination with cytarabine for the treatment of R/R AML in the randomized, double-blind, placebo-controlled, phase III VALOR study ( n = 711). Compared with placebo/cytarabine, the vosaroxin/cytarabine regimen demonstrated favorable CR rates and survival in patients ⩾60 years of age, with toxicities similar to other AML regimens. Here we review outcomes of recent studies of commonly used chemotherapy regimens for the treatment of R/R AML and evaluate the results of the VALOR trial in the context of the current treatment landscape.
- Published
- 2017
34. Phase I Trial of Bortezomib (PS-341; NSC 681239) and 'Nonhybrid' (Bolus) Infusion Schedule of Alvocidib (Flavopiridol; NSC 649890) in Patients with Recurrent or Refractory Indolent B-cell Neoplasms
- Author
-
Caryn Weir-Wiggins, Wen Wan, John D. Roberts, Maciej Kmieciak, Viswanathan Ramakrishnan, Loveleen Kang, Rachid Baz, Cody J. Peer, Steven Grant, G. David Roodman, E. Brent Perkins, L. Austin Doyle, Prithviraj Bose, Martha D. Wellons, Robert K. Stuart, A. Dimitrios Colevas, Daniel C. Sullivan, Heidi Sankala, Beata Holkova, Ellen Shrader, William D. Figg, Kevin T. Hogan, Connie Honeycutt, Domenico Coppola, Mary Beth Tombes, and Jana L. Dawson
- Subjects
Adult ,Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Neutropenia ,Pharmacology ,Article ,Drug Administration Schedule ,Bortezomib ,chemistry.chemical_compound ,Piperidines ,Recurrence ,hemic and lymphatic diseases ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Multiple myeloma ,Aged ,Flavonoids ,B-Lymphocytes ,Leukopenia ,business.industry ,Waldenstrom macroglobulinemia ,Middle Aged ,Alvocidib ,medicine.disease ,Boronic Acids ,Combined Modality Therapy ,Lymphoproliferative Disorders ,Regimen ,Treatment Outcome ,chemistry ,Pyrazines ,Retreatment ,Female ,Mantle cell lymphoma ,Drug Monitoring ,medicine.symptom ,business ,medicine.drug - Abstract
Purpose: This phase I study was conducted to determine the dose-limiting toxicities (DLT) and maximum tolerated dose (MTD) for the combination of bortezomib and alvocidib in patients with B-cell malignancies (multiple myeloma, indolent lymphoma, Waldenstrom macroglobulinemia, and mantle cell lymphoma). Experimental Design: Patients received bortezomib (intravenous push), followed by alvocidib (1-hour infusion), on days 1, 4, 8, and 11 of a 21-day treatment cycle. Patients experiencing responses or stable disease continued on treatment at the investigator's discretion. A standard 3+3 dose-escalation design was used to identify the MTD based on DLTs, and pharmacokinetic and pharmacodynamic studies were conducted. Results: A total of 44 patients were enrolled, with 39 patients assessed for response. The MTD was established as 1.3 mg/m2 for bortezomib and 40 mg/m2 for alvocidib. The most common hematologic toxicities included leukopenia, lymphopenia, neutropenia, and thrombocytopenia. The most common nonhematologic toxicities included diarrhea, fatigue, and sensory neuropathy. Three complete remissions (8%) and 10 partial remissions (26%) were observed for a total response rate of 33%. Pharmacokinetic findings with the current dosing regimen were consistent with the comparable literature and the hybrid dosing regimen. Pharmacodynamic study results did not correlate with clinical responses. Conclusions: The combination of bortezomib and alvocidib is tolerable, and an MTD has been established for this schedule. The regimen appears to be efficacious in patients with relapsed/refractory multiple myeloma or indolent non-Hodgkin lymphoma. As the nonhybrid regimen is less cumbersome than the previous hybrid dosing schedule regimen, the current schedule is recommended for successor studies. Clin Cancer Res; 20(22); 5652–62. ©2014 AACR.
- Published
- 2014
- Full Text
- View/download PDF
35. A phase 1b/2 study of vosaroxin in combination with cytarabine in patients with relapsed or refractory acute myeloid leukemia
- Author
-
Glenn Michelson, Larry D. Cripe, Gail J. Roboz, Judith E. Karp, Farhad Ravandi, Jeffrey E. Lancet, Richard D. Leavitt, Rachael E. Hawtin, Tianling Chen, Adam R. Craig, Robert K. Stuart, Michael B. Maris, and Judith A. Fox
- Subjects
Adult ,Male ,medicine.medical_specialty ,Adolescent ,Maximum Tolerated Dose ,Population ,Salvage therapy ,Pharmacology ,Vosaroxin ,Gastroenterology ,Cohort Studies ,Young Adult ,chemistry.chemical_compound ,Refractory ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Naphthyridines ,education ,Aged ,Neoplasm Staging ,Salvage Therapy ,education.field_of_study ,business.industry ,Remission Induction ,Cytarabine ,Myeloid leukemia ,Articles ,Hematology ,Middle Aged ,Prognosis ,medicine.disease ,Survival Rate ,Leukemia, Myeloid, Acute ,Thiazoles ,Leukemia ,chemistry ,Tolerability ,Drug Resistance, Neoplasm ,Female ,Neoplasm Recurrence, Local ,business ,Follow-Up Studies ,medicine.drug - Abstract
Vosaroxin is a first-in-class anticancer quinolone derivative that intercalates DNA and inhibits topoisomerase II. This study assessed the safety and tolerability of vosaroxin plus cytarabine in patients with relapsed/refractory acute myeloid leukemia. Escalating vosaroxin doses (10-minute infusion; 10-90 mg/m(2); days 1, 4) were given in combination with cytarabine on one of two schedules: schedule A (24-hour continuous intravenous infusion, 400 mg/m(2)/day, days 1-5) or schedule B (2-hour intravenous infusion, 1 g/m(2)/day, days 1-5). Following dose escalation, enrollment was expanded at the maximum tolerated dose. Of 110 patients enrolled, 108 received treatment. The maximum tolerated dose of vosaroxin was 80 mg/m(2) for schedule A (dose-limiting toxicities: grade 3 bowel obstruction and stomatitis) and was not reached for schedule B (recommended phase 2 dose: 90 mg/m(2)). In the efficacy population (all patients in first relapse or with primary refractory disease treated with vosaroxin 80-90 mg/m(2); n=69), the complete remission rate was 25% and the complete remission/complete remission with incomplete blood count recovery rate was 28%. The 30-day all-cause mortality rate was 2.5% among all patients treated at a dose of 80-90 mg/m(2). Based upon these results, a phase 3 trial of vosaroxin plus cytarabine was initiated in patients with relapsed/refractory acute myeloid leukemia. (Clinicaltrials.gov identifier: NCT00541866).
- Published
- 2014
- Full Text
- View/download PDF
36. Similar dynamics of intraapheresis autologous CD34+ recruitment and collection efficiency in patients undergoing mobilization with or without plerixafor
- Author
-
Kathy Hogan Edwards, Amanda Littleton, Molly Schneider, Henning Schade, Cindy Kramer, Milos N. Budisavljevic, Yubin Kang, Saurabh Chhabra, Coleen Butcher, Robert K. Stuart, and Luciano J. Costa
- Subjects
medicine.medical_specialty ,Mobilization ,business.industry ,Plerixafor ,Immunology ,Urology ,CD34 ,Blood volume ,Hematology ,Leukapheresis ,medicine.disease ,Immunology and Allergy ,Medicine ,business ,Prospective cohort study ,Multiple myeloma ,Hematopoietic Stem Cell Mobilization ,medicine.drug - Abstract
Background Compared with growth factor (G) alone, the combination of G with plerixafor (G + P) increases peripheral blood CD34+ count (PB-CD34+) and improves CD34+ collection yield (yCD34+) in multiple myeloma and lymphoma patients undergoing autologous hematopoietic progenitor cell (AHPC) mobilization. It is unknown whether the improved yCD34+ with G + P results entirely from expansion of PB-CD34+ or also from increased intraapheresis CD34+ recruitment and collection efficiency. Study Design and Methods We retrospectively studied 192 patients who underwent AHPC mobilization and collection with G (n = 73) or G + P (n = 119) to compare the adjusted relative efficiency (aRE), the proportion of the circulating CD34+ pool that is captured for each blood volume processed. Additionally, in a prospective cohort of nine patients mobilizing with G and 11 with G + P, PB-CD34+ after leukapheresis allowed calculation of the recruitment coefficient (RC), proportion of the initial CD34+ pool recruited from the marrow into peripheral blood for each blood volume processed. Results There was no difference in aRE between G and G + P (0.50 vs. 0.46; p = 0.37) and no substantial decline in aRE with higher blood volumes processed in either group. RC was also not different between G and G + P (median, 0.39 and 0.38, respectively; p = 0.7). Prediction of yCD34+ was determined essentially by PB-CD34+ and not affected independently by plerixafor. Conclusion Kinetics of intraapheresis CD34+ recruitment and collection is proportional to PB-CD34+ but not influenced further by plerixafor.
- Published
- 2014
- Full Text
- View/download PDF
37. Prediction of Poor Mobilization of Autologous CD34+ Cells with Growth Factor in Multiple Myeloma Patients: Implications for Risk-Stratification
- Author
-
Morie A. Gertz, Shaji Kumar, Elizabeth J. Nista, Angela Dispenzieri, Martha Q. Lacy, Yubin Kang, Kathy Hogan Edwards, Luciano J. Costa, Cindy Kramer, Robert K. Stuart, and Francis K. Buadi
- Subjects
Adult ,Male ,Oncology ,medicine.medical_specialty ,Filgrastim ,medicine.medical_treatment ,CD34 ,Antigens, CD34 ,Cohort Studies ,Internal medicine ,medicine ,Humans ,Platelet ,Multiple myeloma ,Aged ,Retrospective Studies ,Lenalidomide ,Transplantation ,Mobilization ,business.industry ,Growth factor ,Plerixafor ,Hematology ,Middle Aged ,medicine.disease ,Hematopoietic Stem Cell Mobilization ,Surgery ,Stem cell mobilization ,Female ,Multiple Myeloma ,business ,medicine.drug - Abstract
It is unknown whether clinical characteristics can successfully predict which multiple myeloma (MM) patients would be poor mobilizers with growth factor (GF) alone so they can be assigned to mobilization with chemotherapy + GF or GF + plerixafor. MM patients (N = 477) who underwent autologous mobilization with GF were retrospectively reviewed and assigned into training and validation cohorts. In multiple regression analysis, age, platelet count at time of mobilization, type of GF utilized, and extent of exposure to lenalidomide independently correlated with peripheral blood (PB)-CD34+ and were integrated in a predicting score (PS) for poor mobilizers, defined as PB-CD34+
- Published
- 2014
- Full Text
- View/download PDF
38. Ivosidenib (AG-120) in Mutant IDH1 Relapsed/Refractory Acute Myeloid Leukemia: Results of a Phase 1 Study
- Author
-
Sam Agresta, Alice S. Mims, Martin S. Tallman, Meredith Goldwasser, Harry P. Erba, Robert H. Collins, Gail J. Roboz, Will Donnellan, James L. Slack, Richard Stone, Martha Arellano, Hongfang Wang, Daniel A. Pollyea, Sung Choe, Hua Yang, Bin Fan, Ronan T. Swords, Anthony S. Stein, Christophe Willekens, Arnaud Pigneux, Mikkael A. Sekeres, Gabrielle T. Prince, Stephanie M. Kapsalis, Robert K. Stuart, James M. Foran, Elie Traer, Hagop M. Kantarjian, Vickie Zhang, Amir T. Fathi, Geoffrey L. Uy, Katharine E. Yen, Stéphane de Botton, David Dai, Eyal C. Attar, Jessica K. Altman, Gabriel N. Mannis, Bin Wu, Eytan M. Stein, Courtney D. DiNardo, and Hua Liu
- Subjects
0301 basic medicine ,Cancer Research ,IDH1 ,business.industry ,Mutant ,Myeloid leukemia ,Hematology ,03 medical and health sciences ,030104 developmental biology ,Oncology ,Phase (matter) ,Relapsed refractory ,Cancer research ,Medicine ,business - Published
- 2018
- Full Text
- View/download PDF
39. Graft-Versus-Host Disease Prophylaxis in Reduced Intensity Conditioning Allogeneic Hematopoietic Cell Transplantation: Comparing the Efficacy of Mycophenolate Mofetil and Methotrexate in Combination with Calcineurin Inhibitor
- Author
-
Amin M. Alousi, Dennis Dong Hwan Kim, Robert K. Stuart, Mukta Arora, Ying Liu, Joseph Pidala, Michael T. Hemmer, Betty K. Hamilton, Luciano J. Costa, Stephen R. Spellman, Saurabh Chhabra, Daniel R. Couriel, Olle Ringdén, Navneet S. Majhail, and Tao Wang
- Subjects
Transplantation ,Hematopoietic cell ,business.industry ,Hematology ,Pharmacology ,medicine.disease ,Mycophenolate ,Calcineurin ,Graft-versus-host disease ,Reduced Intensity Conditioning ,medicine ,Methotrexate ,business ,medicine.drug - Published
- 2018
- Full Text
- View/download PDF
40. Subanalysis of Patients with Secondary Acute Myeloid Leukemia (sAML) with Refractory Anemia with Excess of Blasts in Transformation (RAEB-T) Enrolled in a Phase 3 Study of CPX-351 Versus Conventional 7 + 3 Cytarabine and Daunorubicin
- Author
-
Qi An, Gary J. Schiller, Nikolai A. Podoltsev, Erica D. Warlick, Bruno C. Medeiros, Geoffrey L. Uy, Dale L. Bixby, S. Eric Rubenstein, Stefan Faderl, Tara L. Lin, Arthur C. Louie, Robert K. Stuart, Matthew J. Wieduwilt, Jeffrey E. Lancet, Matthew C. Foster, Laura F. Newell, and Wendy Stock
- Subjects
Transplantation ,Daunorubicin ,business.industry ,Phases of clinical research ,Hematology ,medicine.disease ,03 medical and health sciences ,Transformation (genetics) ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Cytarabine ,medicine ,Cancer research ,Secondary Acute Myeloid Leukemia ,Refractory anemia with excess of blasts ,business ,030215 immunology ,medicine.drug - Published
- 2018
- Full Text
- View/download PDF
41. The VITAL Trial: Phase II Trial of Vosaroxin and Infusional Cytarabine for Frontline Treatment of acute Myeloid Leukemia
- Author
-
Nikolai A. Podoltsev, Stephen A. Strickland, Sanjay R. Mohan, Merrida A Childress, Robert K. Stuart, Michael R. Savona, Gregory D. Ayers, Michael Byrne, Steven D. Gore, and Amer M. Zeidan
- Subjects
medicine.medical_specialty ,business.industry ,Immunology ,Complete remission ,Phases of clinical research ,Cell Biology ,Hematology ,Trial Phase ,Tp53 mutation ,Biochemistry ,Vosaroxin ,chemistry.chemical_compound ,Older patients ,chemistry ,Internal medicine ,medicine ,Cytarabine ,Allogeneic hematopoietic stem cell transplant ,business ,health care economics and organizations ,medicine.drug - Abstract
Background: AML is an aggressive malignancy with a median age of 67 yrs at diagnosis and an age-adjusted 5-yr survival of less than 25%. Most older patients (pts) with AML have poor risk cytogenetic/genetic features associated with suboptimal complete remission (CR) rates and overall survival (OS). Until recently, the anthracycline and cytarabine based induction remained unchanged for over 4 decades. Vosaroxin (Vos) is a first-in-class anticancer quinolone derivative which is thought to induce less cardiac toxicity than standard anthracycline therapy. Vos was previously administered in combination with intermediate dose cytarabine (IDAC) in relapsed/refractory (r/r) AML pts in a large randomized phase 3 trial (VALOR) and demonstrated significantly improved CR rates compared to IDAC alone but without improvement in OS. In the VITAL study, we investigated the combination of infusional cytarabine (iAC) with Vos ("7 + V") in newly diagnosed AML. Methods: VITAL (NCT02658487) is a single-arm, open-label, two-stage phase II study of 90 mg/m2, Vos on days 1 and 4 (reduced to 70 mg/m2, days 1 and 4 in the event of re-induction) and 100 mg/m2 continuous iAC on days 1-7. Oral cryotherapy was administered during Vos administration as prophylaxis for oral mucositis seen in prior studies. Enrollment in stage 1 was limited to 17 evaluable pts >55 yrs of any risk classification or pts 18-54 years with high-risk disease. The two-stage design required CR in more than 7 of the first 17 pts in order to proceed to stage 2. The operating characteristics of the study design provided a 64% probability of early termination if CR occurred in 7 or fewer pts suggesting that the true CR rate would be ≤ 40%. Stage 2 included an additional 24 pts (age 18-54 yrs with intermediate or high-risk disease, or 55 years or older with any risk disease). Response was assessed using the modified IWG-2003 criteria. Results: In total, 42 pts were enrolled and assessed for toxicity. One patient who achieved an aplastic marrow biopsy at day 14, succumbed to an AML-related sepsis event prior to formal response assessment was initially deemed unevaluable for response and replaced but was ultimately included in the response assessment. Baseline demographics of the 42 enrolled pts are seen in Table 1a. Median age was 63 yrs (range, 43-74) and 86% (36/42) were ≥ 55yrs at time of enrollment. Almost all, 40/42 (95%), had intermediate (11/40) or poor (29/40) risk AML. Adverse events (AEs) of special interest of any Grade (Gr) and Gr 3-5 AEs occurring in > 10% of pts are listed in Table 1b. Oral mucositis was observed in 16/42 (38%) pts with 12/42 (29%) having Gr 1-2 and only 4/42 (10%) having Gr 3 severity. Three Gr 5 events occurred (2-Infection/Sepsis; 1-neutropenic colitis). No acute cardiac toxicity was seen. CR was achieved in 20/42 (48%) pts, while 3 additional pts achieved CR with incomplete count recovery (CRi) for a CR/CRi rate of 55% (23/42). CR was observed in 100% (2/2) with favorable risk (both >65 yrs old). CR/CRi was seen in 73% (8/11) intermediate and 45% (13/29) poor risk pts. One poor risk pt with CRi converted to CR in the absence of additional therapy but did so outside of the protocol defined time period for response assessment (Day 57 +/- 3 days) and was recorded as CRi for this analysis. CR/CRi was noted in 40% (4/10) pts whose AML had TP53 mutations. Allogeneic hematopoietic stem cell transplant was performed in 45% (19/42) of pts. Progression-free survival was 43% and OS was 48% for all pts at 1-year of follow-up (Figure 1). Conclusion: The combination of Vos and iAC in newly diagnosed AML pts appears safe. Mucocutaneous complications were observed as noted in previous studies with Vos. Administration of oral cryotherapy during Vos administration appeared to reduce occurrence of oral mucositis during induction, but > Gr 3 neutropenic enterocolitis occurred in 7/42 (17%) pts. Still, the combination appears to be tolerable with an adverse event profile analogous to conventional induction regimens. While the VALOR trial observed Gr 3-5 cardiac AEs in < 1% of the 705 r/r AML pts with prior anthracycline exposure, no attributable cardiac events were seen within the VITAL trial population. Vosaroxin and infusional cytarabine is a clinically active induction regimen which warrants further investigation based on response rates and toxicity profile at least similar to current anthracycline based induction strategies with an apparent absence of cardiac toxicity. Disclosures Strickland: Astellas: Consultancy; Jazz: Consultancy; Kite: Consultancy; Pfizer: Consultancy; Novartis: Consultancy; Sunesis: Research Funding; AbbVie: Consultancy. Podoltsev:Alexion: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Pfizer: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Agios Pharmaceuticals: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Blueprint Medicines: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Incyte: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Novartis: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Boehringer Ingelheim: Research Funding; Kartos Therapeutics: Research Funding; Astellas Pharma: Research Funding; Daiichi Sankyo: Research Funding; Sunesis Pharmaceuticals: Research Funding; Jazz Pharmaceuticals: Research Funding; Pfizer: Research Funding; Astex Pharmaceuticals: Research Funding; CTI Biopharma: Research Funding; Celgene: Other: Grant funding, Research Funding; Genentech: Research Funding; AI Therapeutics: Research Funding; Samus Therapeutics: Research Funding; Arog Pharmaceuticals: Research Funding. Zeidan:Acceleron Pharma: Consultancy, Honoraria, Research Funding; Celgene Corporation: Consultancy, Honoraria, Research Funding; Abbvie: Consultancy, Honoraria, Research Funding; Otsuka: Consultancy, Honoraria, Research Funding; Pfizer: Consultancy, Honoraria, Research Funding; Medimmune/AstraZeneca: Research Funding; Boehringer-Ingelheim: Consultancy, Honoraria, Research Funding; Trovagene: Consultancy, Honoraria, Research Funding; Incyte: Consultancy, Honoraria, Research Funding; Takeda: Consultancy, Honoraria, Research Funding; ADC Therapeutics: Research Funding; Jazz: Honoraria; Ariad: Honoraria; Agios: Honoraria; Novartis: Honoraria; Astellas: Honoraria; Daiichi Sankyo: Honoraria; Cardinal Health: Honoraria; Seattle Genetics: Honoraria; BeyondSpring: Honoraria. Byrne:Karyopharm: Research Funding. Gore:Celgene Corporation: Consultancy, Research Funding. Savona:Celgene Corporation: Membership on an entity's Board of Directors or advisory committees; Boehringer Ingelheim: Patents & Royalties; AbbVie: Membership on an entity's Board of Directors or advisory committees; Sunesis: Research Funding; TG Therapeutics: Membership on an entity's Board of Directors or advisory committees, Research Funding; Takeda: Membership on an entity's Board of Directors or advisory committees, Research Funding; Selvita: Membership on an entity's Board of Directors or advisory committees; Karyopharm Therapeutics: Consultancy, Equity Ownership, Membership on an entity's Board of Directors or advisory committees; Incyte Corporation: Membership on an entity's Board of Directors or advisory committees, Research Funding.
- Published
- 2019
- Full Text
- View/download PDF
42. Outcomes of AML Patients Undergoing Reduced Intensity Allogeneic Stem Cell Transplantation with Adverse Risk Disease By European Leukemianet Classification: A Single Center Retrospective Analysis
- Author
-
Robert Collins, Robert K. Stuart, Brian T. Hess, Amarendra K. Neppalli, Georges J. Nahhas, Lauren Pearce, Clark Alsfeld, Zane Chiad, Brion V. Randolph, Laura Milligan, Kathy Hogan Edwards, and Irl Brian Greenwell
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Immunology ,Reduced intensity ,Cell Biology ,Hematology ,Disease ,Single Center ,Biochemistry ,Transplantation ,European LeukemiaNet ,medicine.anatomical_structure ,hemic and lymphatic diseases ,Internal medicine ,Retrospective analysis ,Medicine ,Bone marrow ,Stem cell ,business - Abstract
Background: Acute myeloid leukemia (AML) is the most common acute leukemia in adults with a median age at diagnosis of close to 70 years. (Juliusson G, et al. Blood, 2011;119(17):3890-3899) The majority of patients with AML require allogeneic stem cell transplant (Allo-SCT) in order to achieve cure. European LeukemiaNET (ELN) published risk stratifications for AML patients according to cytogenetics in 2010 and then updated risk categories in 2017 to incorporate molecular abnormalities as well. The ELN adverse risk category predicts for a higher risk of relapse and worse overall survival (OS) compared to favorable and intermediate risk patients. (Döhner H, et al. Blood, 2017;129(4):424-447) Older patients are more likely than younger patients to have adverse risk disease. (Mrozek K, et al. JCO, 2012;30(36):4515-4523). In addition, older patients are typically not considered for myeloablative conditioning (MAC) regimens due to the high risk of non-relapse mortality (NRM). Thus, a significant proportion of AML patients who are candidates for Allo-SCT have adverse risk disease by ELN criteria and are only candidates for RIC Allo-SCT, both of which increase the risk of relapse. A recent randomized trial in AML and MDS patients noted a trend toward improved OS and statistically significant improvement in relapse free survival (RFS) favoring MAC over RIC (Scott BL, et al. JCO, 2017; 35(11): 1154-1161). Despite this, studies have shown durable remissions and long term disease free survival with RIC Allo-SCT in AML patients with high risk features. (Tauro S, et al. JCO, 2005; 23(36):9387-9393) However, to our knowledge no study has investigated the use of RIC Allo-SCT exclusively in patients who have adverse risk by 2017 ELN criteria. Thus, we reviewed AML patients with adverse risk disease by 2017 ELN criteria at diagnosis who underwent RIC Allo-SCT at our institution. Methods: We conducted a single center retrospective study of high risk adult AML patients by ELN criteria who underwent RIC Allo-SCT at MUSC from 3/20/2010 to 2/20/2018. Adverse risk disease was defined by the 2017 ELN criteria. Baseline demographic, clinical, laboratory, pathology, and outcomes data were collected by retrospective chart review. Kaplan Meier was utilized for time to event analysis. Results: A total of 42 patients with adverse risk AML received RIC Allo-SCT. The median follow up was 16.6 months. Clinical characteristics were collected for each patient and are included in table 1. The median age was 62 with 16 patients (38%) ≥ 65 years of age. Twenty-three patients had a HCT-CI score ≥ 3 (54%) with 9 patients (21%) having a HCT-CI score ≥ 5. Complete remission (CR) was defined as bone marrow blasts < 5% without known molecular evidence of persistent/relapsed AML. CR and CR with incomplete count recovery (CRi) was not able to be defined. Thirty-one patients (73%) were in CR1 at transplant, 7 patients (16%) were in CR2+ defined as CR necessitating ≥ 2 treatment regimens. Four patients (9%) never achieved a CR and were labelled as primary induction failure (PIF), and one patient (3%) received prior MAC Allo-SCT before receiving RIC Allo-SCT and was labeled as Relapse. Four of the five patients with disease status of PIF or relapse were deceased within 13 months of Allo-SCT. The non-relapse mortality (NRM) at 1 year was 26.2%. Relapse-free survival (RFS) at 1, 2, and 3 years was 57.1%, 45.2%, and 35.7% respectively. The overall survival (OS) at 1, 2, and 3 years was 66.7%, 50.0%, and 40.5% respectively. A subset analysis of the patients in CR1 or CR2+ prior to RIC-Allo SCT noted a NRM at 1 year of 27% along with a RFS at 1, 2, and 3 years of 59.5%, 48.7%, and 40.5% respectively. Cause of death included relapse (33%), infection (29%), acute GVHD (25%), and chronic GVHD (3%). Conclusion: This provides evidence that although adverse risk disease by ELN criteria and reduced intensity conditioning both increase the incidence of relapse in patients with AML, that RIC Allo-SCT for this high risk patient population can provide long term disease free survival. In addition, the favorable outcomes for patients in CR1 and CR2+ prior to RIC-Allo are promising. It would be interesting to investigate a larger multi-center series of patients with adverse risk disease by ELN in CR prior to RIC Allo-SCT. Minimal residual disease by myeloid molecular panel and/or flow in this patient population prior to transplant would be of great interest as well. Disclosures Edwards: Genzyme: Consultancy.
- Published
- 2019
- Full Text
- View/download PDF
43. Abstract CT184: Gilteritinib significantly prolongs overall survival in patients with FLT3-mutated (FLT3mut+) relapsed/refractory (R/R) acute myeloid leukemia (AML): Results from the Phase III ADMIRAL trial
- Author
-
Alexander E. Perl, Richard A. Larson, Nikolai A. Podoltsev, Stefania Paolini, Antonio Di Stasi, Wen-Chien Chou, Amir T. Fathi, Andreas Neubauer, Margaret Kasner, Xuan Liu, Francesco Fabbiano, Giovanni Martinelli, Celalettin Ustun, Timothy S. Pardee, Sung-Soo Yoon, Mark J. Levis, Maria R. Baer, Je-Hwan Lee, Ellin Berman, Pau Montesinos, Hisayuki Yokoyama, Christian Recher, Chaofeng Liu, Fabio Ciceri, Erkut Bahceci, Robert K. Stuart, Rebecca L. Olin, Jorge E. Cortes, and Naoko Hosono
- Subjects
Cancer Research ,medicine.medical_specialty ,Population ,Salvage therapy ,01 natural sciences ,Gastroenterology ,010104 statistics & probability ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Idarubicin ,030212 general & internal medicine ,Midostaurin ,0101 mathematics ,education ,education.field_of_study ,business.industry ,Induction chemotherapy ,medicine.disease ,Fludarabine ,Oncology ,chemistry ,Cytarabine ,business ,Febrile neutropenia ,medicine.drug - Abstract
Introduction: Gilteritinib is a potent/selective oral inhibitor of FMS-like tyrosine kinase 3 (FLT3). Based upon interim analysis response rates from the ADMIRAL Phase III study of gilteritinib vs salvage chemotherapy (SC) in patients (pts) with R/R FLT3mut+ AML (NCT02421939), gilteritinib became the first FLT3 inhibitor approved as single agent therapy in this population. Here we present the final results of this pivotal trial. Methods: Adults with confirmed FLT3mut+ AML (FLT3-ITD and/or FLT3-TKD D835 or I836 mutations) refractory to induction chemotherapy or in untreated first relapse were randomized (2:1) to receive continuous 28-day cycles of 120 mg/day gilteritinib or pre-randomization selected SC: low-dose cytarabine (LoDAC), azacitidine (AZA), mitoxantrone/etoposide/cytarabine (MEC), or fludarabine/cytarabine/granulocyte colony-stimulating factor/idarubicin (FLAG-IDA). Prior FLT3 inhibitor use, other than midostaurin or sorafenib, was excluded. Overall survival (OS) and the combined rate of complete remission/complete remission with partial hematologic recovery (CR/CRh) were co-primary endpoints. Secondary endpoints were event-free survival (EFS) and CR rate; safety/tolerability was also examined. Results: A total of 371 pts were randomized: 247 to gilteritinib and 124 to SC (MEC, 25.7%; FLAG-IDA, 36.7%; LoDAC, 14.7%; AZA, 22.9%). Median age was 62 years (range, 19-85). Baseline FLT3 mutations were: FLT3-ITD, 88.4%; FLT3-TKD, 8.4%; both FLT3-ITD and FLT3-TKD, 1.9%; unconfirmed, 1.3%. Overall, 39.4% of pts had refractory AML and 60.6% had relapsed AML. Patients randomized to gilteritinib had significantly longer OS (9.3 months) than SC (5.6 months; hazard ratio [HR] for death = 0.637; P=0.0007); 1-year survival rates were 37.1% and 16.7%, respectively. The CR/CRh rates for gilteritinib and SC were 34.0% and 15.3%, respectively (P=0.0001); CR rates were 21.1% and 10.5% (2-sided P=0.0106). Median EFS was 2.8 months and 0.7 months in the gilteritinib and SC arms, respectively (HR 0.793, P=0.0830). Common adverse events (AEs) in all randomized pts were febrile neutropenia (43.7%), anemia (43.4%), and pyrexia (38.6%). Common grade ≥3 AEs related to gilteritinib were anemia (19.5%), febrile neutropenia (15.4%), thrombocytopenia (12.2%), and decreased platelet count (12.2%). Adjusted for exposure duration, serious treatment-emergent AEs per patient year were less common with gilteritinib (7.1%) than SC (9.2%). Conclusions: In patients with R/R FLT3mut+ AML, the potent, selective FLT3 inhibitor gilteritinib resulted in significantly longer OS and higher response rates compared with chemotherapy and had a favorable safety profile. These results change the treatment paradigm for salvage therapy of R/R FLT3mut+ AML and establish gilteritinib as the new standard of care. Citation Format: Alexander E. Perl, Giovanni Martinelli, Jorge E. Cortes, Andreas Neubauer, Ellin Berman, Stefania Paolini, Pau Montesinos, Maria R. Baer, Richard A. Larson, Celalettin Ustun, Francesco Fabbiano, Antonio Di Stasi, Robert Stuart, Rebecca Olin, Margaret Kasner, Fabio Ciceri, Wen-Chien Chou, Nikolai Podoltsev, Christian Recher, Hisayuki Yokoyama, Naoko Hosono, Sung-Soo Yoon, Je-Hwan Lee, Timothy Pardee, Amir T. Fathi, Chaofeng Liu, Xuan Liu, Erkut Bahceci, Mark J. Levis. Gilteritinib significantly prolongs overall survival in patients with FLT3-mutated (FLT3mut+) relapsed/refractory (R/R) acute myeloid leukemia (AML): Results from the Phase III ADMIRAL trial [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr CT184.
- Published
- 2019
- Full Text
- View/download PDF
44. PB1733 OUTCOMES WITH CPX-351 VERSUS 7+3 BY BASELINE BONE MARROW BLAST PERCENTAGE IN OLDER ADULTS WITH NEWLY DIAGNOSED, HIGH-RISK/SECONDARY AML: EXPLORATORY ANALYSIS OF A PHASE 3 STUDY
- Author
-
T.L. Lin, R.J. Ryan, G.J. Schiller, S.R. Solomon, Robert K. Stuart, D.H. Ryan, M.J. Wieduwilt, J.E. Cortes, M. Chiarella, and Ellen K. Ritchie
- Subjects
medicine.medical_specialty ,medicine.anatomical_structure ,business.industry ,Internal medicine ,Phases of clinical research ,Medicine ,Hematology ,Newly diagnosed ,Bone marrow ,Exploratory analysis ,business ,Secondary AML ,Baseline (configuration management) - Published
- 2019
- Full Text
- View/download PDF
45. A randomized phase II trial of CX-01 with standard therapy in elderly patients with acute myeloid leukemia (AML)
- Author
-
Stephen Marcus, Robert K. Stuart, Jonathan E. Kolitz, Matthew J. Wieduwilt, Don A. Stevens, Tibor Kovacsovics, Michael B. Maris, John L. Reagan, Paul J. Shami, Cecilia Arana-Yi, Moshe Yair Levy, Hana Safah, Michaela L. Tsai, Rachel J. Cook, Jay Yang, William B. Donnellan, and Peter Westervelt
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,medicine.drug_class ,business.industry ,Anticoagulant ,Myeloid leukemia ,Heparin ,03 medical and health sciences ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,Heparin Derivative ,business ,Standard therapy ,030215 immunology ,medicine.drug - Abstract
7001 Background: Elderly AML patients have poor outcomes irrespective of therapy. CX-01 is a low anticoagulant heparin derivative that retains heparin’s ability to alter the activity of the CXCL12/CXCR4 axis, P-selectin, extracellular histones, and Platelet Factor 4. A pilot study of CX-01 combined with standard therapy for AML led to a complete remission (CR) rate of 92% ( Blood Adv 2:381, 2018). We conducted a randomized, dose-finding study of the same combination in newly diagnosed elderly AML patients. Methods: 76 fit patients older than 59 were randomized to induction with idarubicin and cytarabine on a 7+3 schedule only (Group 1); 7+3 with a lower dose of CX-01 (0.125 mg/kg/hour) (Group 2); or 7+3 with a higher dose of CX-01 (0.25 mg/kg/hour) (Group 3). Patients in CR received consolidation therapy consisting of up to 3 cycles of intermediate dose cytarabine (1000 mg/m2 every 12 hours on Days 1, 3, 5) without or with the same dose of CX-01 for Groups 1, 2, and 3, respectively. CX-01 was given as a continuous infusion after a 4 mg/kg bolus until completion of chemotherapy. Results: 66 of 75 treated patients were evaluable for response. Ten patients were not evaluable due to withdrawal of consent (6 patients), introduction of midostaurin after its approval (3 patients), or death due to hepatic sinusoidal obstructive disease at Day 21 (1 patient in Group 2). We present results for evaluable patients and not on an intent to treat basis. Baseline characteristics were similar across groups. The composite CR rate (CR + CRi) was highest for patients in Group 3 with 89% patients achieving a composite CR as compared to 58% and 50% in Groups 1 and 2, respectively. Kaplan-Meier curves indicated a statistically significant improvement in event free survival (EFS) (P = 0.019) and a non-significant trend (P = 0.10) to improvement in OS in Group 3 as compared to Group 1. Groups 1 and 2 were comparable for EFS and OS. CX-01 was well tolerated without increased incidence of bleeding in Groups 2 and 3. Conclusions: The encouraging CR rate and EFS in elderly fit patients with newly diagnosed AML suggests that CX-01 may potentiate the efficacy of standard AML induction therapy. A randomized study to confirm these findings with the higher dose of CX-01 is warranted. Clinical trial information: NCT02873338.
- Published
- 2019
- Full Text
- View/download PDF
46. Outcomes with CPX-351 versus 7+3 by baseline bone marrow (BM) blast percentage in older adults with newly diagnosed high-risk/secondary acute myeloid leukemia (sAML)
- Author
-
Scott R. Solomon, Ellen K. Ritchie, Gary J. Schiller, Richard Stone, Robert J. Ryan, Tara L. Lin, Robert K. Stuart, Michael Chiarella, Jorge E. Cortes, Matthew J. Wieduwilt, Daniel H. Ryan, and Laura F. Newell
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Daunorubicin ,Newly diagnosed ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,Cytarabine ,Secondary Acute Myeloid Leukemia ,Bone marrow ,business ,030215 immunology ,medicine.drug - Abstract
7042 Background: CPX-351, a liposomal encapsulation of cytarabine (C) and daunorubicin (D) at a synergistic ratio, is approved as Vyxeos in the US and EU for adults with newly diagnosed therapy-related AML or AML with myelodysplasia-related changes. In a phase 3 study, CPX-351 significantly improved OS and remission rates vs 7+3 in patients (pts) aged 60-75 y with newly diagnosed high-risk/sAML. Some studies suggest a high baseline blast percentage may portend a worse prognosis in AML. This post hoc analysis of phase 3 data assessed outcomes by baseline BM blast percentage. Methods: Pts diagnosed with AML per 2008 WHO criteria (≥20% blasts in peripheral blood or BM) were randomized 1:1 to receive ≤2 inductions of CPX-351 (100 units/m2 [C 100 mg/m2 + D 44 mg/m2] on Days 1, 3, 5 [2nd induction: Days 1, 3]) or 7+3 (C 100 mg/m2/d continuously for 7 d [2nd induction: 5 d] + D 60 mg/m2 on Days 1-3 [2nd induction: Days 1-2]). Pts achieving complete remission (CR) or CR with incomplete platelet or neutrophil recovery (CRi) could receive ≤2 consolidations. Results: CPX-351 had longer median OS and higher remission rates vs 7+3 irrespective of baseline BM blast percentage; median OS was worse in higher blast groups for both treatments (Table). The incidence of grade ≥3 TEAEs was >80% for both arms; febrile neutropenia was the most common. Conclusions: Improved outcomes were observed with CPX-351 vs 7+3 irrespective of baseline BM blast percentage in older adults with newly diagnosed high-risk/sAML. Clinical trial information: NCT01696084. [Table: see text]
- Published
- 2019
- Full Text
- View/download PDF
47. Large Granular Lymphocytosis after Allogeneic Hematopoietic Stem Cell Transplantation: A Retrospective Cohort Study
- Author
-
Robert K. Stuart, Amarendra K. Neppalli, Leonard C. Alsfeld, Brian T. Hess, and Lauren Pearce
- Subjects
Transplantation ,medicine.medical_specialty ,Cyclophosphamide ,business.industry ,medicine.medical_treatment ,CD34 ,chemical and pharmacologic phenomena ,Retrospective cohort study ,Viremia ,Hematology ,Hematopoietic stem cell transplantation ,medicine.disease ,surgical procedures, operative ,Internal medicine ,Cohort ,medicine ,Etiology ,business ,medicine.drug - Abstract
Introduction Large granular lymphocytosis (LGL) is observed following hematopoietic stem cell transplantation (HSCT) in 0.5-18.4% of patients. The development of LGL has been associated with and attributed to various conditions such as immune system reconstitution, CMV viremia, and graft-versus-host disease. There is literature available that suggests the development of LGL may be associated with improved outcomes, but management varies greatly because of the different theories surrounding this condition. Thus, we aimed to review a cohort of patients from our institution who developed LGL after HSCT in order to further our understanding and guide management recommendations. Methods We performed a single-center retrospective cohort analysis of patients who had a hematopoietic stem cell transplant from April 1, 2015 to June 30, 2018 and developed large granular lymphocytosis up to one year after transplantation. We collected and reviewed various clinical aspects of each patientOs transplant course and the management of large granular lymphocytosis. Results Of the 127 patients who received an allogeneic HSCT in this time period, 13 patients (10%) were diagnosed with large granular lymphocytosis. Patient demographics, transplant information, and LGL management are listed in Table 1. LGL was diagnosed between 57 and 236 days post-transplant. None of the 13 patients developed EBV viremia by PCR after transplant. 7 of 13 (54%) had CMV viremia/reactivation by PCR after transplant but before LGL diagnosis; 3 of 13 (23%) had CMV viremia by PCR after LGL diagnosis; 3 of 13 (23%) did not develop CMV viremia by PCR before or after LGL diagnosis. 4 of 13 (31%) developed chronic GVHD, and 3 of 13 (23%) developed acute GVHD. Of note, 6 of 13 (46%) had post-transplant cyclophosphamide and 2 of 13 (15%) had CD34 selected grafts; therefore, 8 of 13 (62%) had T-cell depleted transplants. All of the patients in this cohort were alive at day 100. 11 patients had a transplant greater than 1 year ago, and all of these patients were alive at the 1-year mark. Conclusions Based upon the results from this study, we cannot definitively attribute the development of LGL after allogeneic HSCT to one etiology. More importantly, all patients were alive at day 100 and 1 year regardless of the management approach, which suggests that this process may be self-limiting. Therefore, we believe a more conservative and supportive approach to management is warranted.
- Published
- 2019
- Full Text
- View/download PDF
48. A phase 3 study of gemtuzumab ozogamicin during induction and postconsolidation therapy in younger patients with acute myeloid leukemia
- Author
-
Martin S. Tallman, Marilyn L. Slovak, Joseph M. Brandwein, Frederick R. Appelbaum, Harry P. Erba, Richard A. Larson, Roland B. Walter, Cheryl L. Willman, Patrick J. Stiff, Stephen H. Petersdorf, Robert K. Stuart, Thomas J. Nevill, Kenneth J. Kopecky, and Leif Stenke
- Subjects
Male ,medicine.medical_specialty ,Randomization ,Clinical Trials and Observations ,Daunorubicin ,Gemtuzumab ozogamicin ,Immunology ,Phases of clinical research ,Pharmacology ,Enasidenib ,Antibodies, Monoclonal, Humanized ,Biochemistry ,Gastroenterology ,hemic and lymphatic diseases ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Survival rate ,business.industry ,Induction chemotherapy ,Cell Biology ,Hematology ,Gemtuzumab ,Consolidation Chemotherapy ,Leukemia, Myeloid, Acute ,Aminoglycosides ,Cytarabine ,Female ,business ,medicine.drug - Abstract
This randomized phase 3 clinical trial evaluated the potential benefit of the addition of gemtuzumab ozogamicin (GO) to standard induction and postconsolidation therapy in patients with acute myeloid leukemia. Patients were randomly assigned to receive daunorubicin (45 mg/m2 per day on days 1, 2, and 3), cytarabine (100 mg/m2 per day by continuous infusion on days 1–7), and GO (6 mg/m2 on day 4; DA+GO) vs standard induction therapy with daunorubicin (60 mg/m2 per day on days 1, 2, and 3) and cytarabine alone (DA). Patients who achieved complete remission (CR) received 3 courses of high-dose cytarabine. Those remaining in CR after consolidation were randomly assigned to receive either no additional therapy or 3 doses of GO (5 mg/m2 every 28 days). From August 2004 until August 2009, 637 patients were registered for induction. The CR rate was 69% for DA+GO and 70% for DA (P = .59). Among those who achieved a CR, the 5-year relapse-free survival rate was 43% in the DA+GO group and 42% in the DA group (P = .40). The 5-year overall survival rate was 46% in the DA+GO group and 50% in the DA group (P = .85). One hundred seventy-four patients in CR after consolidation underwent the postconsolidation randomization. Disease-free survival was not improved with postconsolidation GO (HR, 1.48; P = .97). In this study, the addition of GO to induction or postconsolidation therapy failed to show improvement in CR rate, disease-free survival, or overall survival. This trial is registered with www.clinicaltrials.gov as #NCT00085709.
- Published
- 2013
- Full Text
- View/download PDF
49. Developmental Therapeutics in Acute Myelogenous Leukemia: Are There Any New Effective Cytotoxic Chemotherapeutic Agents Out There?
- Author
-
Robert K. Stuart and Alice S. Mims
- Subjects
Cancer Research ,medicine.medical_specialty ,medicine.medical_treatment ,Antineoplastic Agents ,Sapacitabine ,Vosaroxin ,chemistry.chemical_compound ,Myelogenous ,Drug Discovery ,medicine ,Humans ,Clofarabine ,Intensive care medicine ,Cladribine ,Protein Kinase Inhibitors ,Clinical Trials as Topic ,Chemotherapy ,business.industry ,Biological Transport ,Hematology ,medicine.disease ,Clinical trial ,Leukemia, Myeloid, Acute ,Leukemia ,Oncology ,chemistry ,business ,medicine.drug - Abstract
Therapies for AML have remained mostly unchanged since the introduction of anthracyline- and cytarabine-based regimens in the 1970s. Though some changes have been made in the dosing of anthracylines, in the choice of consolidation regimens versus allogeneic stem cell transplant, and in supportive care, clinical outcomes remain poor for most patients. As we continue to strive for better treatment options to improve upon outcomes, different agents, both chemotherapeutic and targeted therapies, are being studied. Here we discuss new chemotherapeutic agents that show promise in recent clinical trials and attempt to answer the question if there are any new effective cytotoxic chemotherapy agents out there.
- Published
- 2013
- Full Text
- View/download PDF
50. Plerixafor (a CXCR4 antagonist) following myeloablative allogeneic hematopoietic stem cell transplantation enhances hematopoietic recovery
- Author
-
Ari Horwitz, Kelly Corbet, Stefanie Sarantopoulos, Jagadish Kummetha Venkata, Yubin Kang, Zhiguo Li, David A. Rizzieri, Luciano J. Costa, Mitchell E. Horwitz, Michael Green, Anthony D. Sung, Cristina Gasparetto, Gwynn D. Long, Robert K. Stuart, Keith M. Sullivan, Alice S. Mims, Saurabh Chhabra, and Nelson J. Chao
- Subjects
0301 basic medicine ,Oncology ,Male ,Cancer Research ,Benzylamines ,Transplantation Conditioning ,Neutrophils ,medicine.medical_treatment ,Myeloablative Agonist ,Hematopoietic stem cell transplantation ,Cyclams ,CXCR4 ,Heterocyclic Compounds ,Graft Survival ,lcsh:Diseases of the blood and blood-forming organs ,Hematology ,Middle Aged ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,3. Good health ,Hematologic Neoplasms ,Absolute neutrophil count ,Female ,medicine.drug ,Adult ,medicine.medical_specialty ,Receptors, CXCR4 ,Platelet Engraftment ,Outcomes ,lcsh:RC254-282 ,03 medical and health sciences ,Young Adult ,Stromal-derived factor-1 ,Internal medicine ,medicine ,Humans ,Molecular Biology ,Aged ,Neutrophil Engraftment ,lcsh:RC633-647.5 ,business.industry ,Platelet Count ,Plerixafor ,Research ,Neutrophil engraftment ,Antagonist ,Recovery of Function ,Myeloablative Agonists ,Chemokine CXCL12 ,Hematopoiesis ,Transplantation ,030104 developmental biology ,Case-Control Studies ,business ,Hematopoietic stem cells ,Platelet engraftment - Abstract
Background The binding of CXCR4 with its ligand (stromal-derived factor-1) maintains hematopoietic stem/progenitor cells (HSPCs) in a quiescent state. We hypothesized that blocking CXCR4/SDF-1 interaction after hematopoietic stem cell transplantation (HSCT) promotes hematopoiesis by inducing HSC proliferation. Methods We conducted a phase I/II trial of plerixafor on hematopoietic cell recovery following myeloablative allogeneic HSCT. Patients with hematologic malignancies receiving myeloablative conditioning were enrolled. Plerixafor 240 μg/kg was administered subcutaneously every other day beginning day +2 until day +21 or until neutrophil recovery. The primary efficacy endpoints of the study were time to absolute neutrophil count >500/μl and platelet count >20,000/μl. The cumulative incidence of neutrophil and platelet engraftment of the study cohort was compared to that of a cohort of 95 allogeneic peripheral blood stem cell transplant recipients treated during the same period of time and who received similar conditioning and graft-versus-host disease prophylaxis. Results Thirty patients received plerixafor following peripheral blood stem cell (n = 28) (PBSC) or bone marrow (n = 2) transplantation. Adverse events attributable to plerixafor were mild and indistinguishable from effects of conditioning. The kinetics of neutrophil and platelet engraftment, as demonstrated by cumulative incidence, from the 28 study subjects receiving PBSC showed faster neutrophil (p = 0.04) and platelet recovery >20 K (p = 0.04) compared to the controls. Conclusions Our study demonstrated that plerixafor can be given safely following myeloablative HSCT. It provides proof of principle that blocking CXCR4 after HSCT enhances hematopoietic recovery. Larger, confirmatory studies in other settings are warranted. Trial registration ClinicalTrials.gov NCT01280955
- Published
- 2016
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.