48 results on '"Eric Laille"'
Search Results
2. Pharmacokinetics and Pharmacodynamics with Extended Dosing of CC-486 in Patients with Hematologic Malignancies.
- Author
-
Eric Laille, Tao Shi, Guillermo Garcia-Manero, Christopher R Cogle, Steven D Gore, Joel Hetzer, Keshava Kumar, Barry Skikne, and Kyle J MacBeth
- Subjects
Medicine ,Science - Abstract
CC-486 (oral azacitidine) is an epigenetic modifier in development for patients with myelodysplastic syndromes and acute myeloid leukemia. In part 1 of this two-part study, a 7-day CC-486 dosing schedule showed clinical activity, was generally well tolerated, and reduced DNA methylation. Extending dosing of CC-486 beyond 7 days would increase duration of azacitidine exposure. We hypothesized that extended dosing would therefore provide more sustained epigenetic activity. Reported here are the pharmacokinetic (PK) and pharmacodynamic (PD) profiles of CC-486 extended dosing schedules in patients with myelodysplastic syndromes (MDS), chronic myelomonocytic leukemia (CMML) or acute myeloid leukemia (AML) from part 2 of this study. PK and/or PD data were available for 59 patients who were sequentially assigned to 1 of 4 extended CC-486 dosing schedules: 300mg once-daily or 200mg twice-daily for 14 or 21 days per 28-day cycle. Both 300mg once-daily schedules and the 200mg twice-daily 21-day schedule significantly (all P < .05) reduced global DNA methylation in whole blood at all measured time points (days 15, 22, and 28 of the treatment cycle), with sustained hypomethylation at cycle end compared with baseline. CC-486 exposures and reduced DNA methylation were significantly correlated. Patients who had a hematologic response had significantly greater methylation reductions than non-responding patients. These data demonstrate that extended dosing of CC-486 sustains epigenetic effects through the treatment cycle.ClinicalTrials.gov NCT00528983.
- Published
- 2015
- Full Text
- View/download PDF
3. Supplementary Figure 3 from Phase I Study of CC-486 Alone and in Combination with Carboplatin or nab-Paclitaxel in Patients with Relapsed or Refractory Solid Tumors
- Author
-
Johanna C. Bendell, Eric Laille, Kejian Liu, Aaron Nguyen, Jorge DiMartino, Gordon L. Bray, Patricia M. LoRusso, Miguel Martin, W. Jeff Edenfield, Jose A. Lopez-Martin, Ron H.J. Mathijssen, Bert O'Neil, Christophe Le Tourneau, Nicolas Isambert, Jan H.M. Schellens, Pamela N. Munster, Elisabeth I. Heath, Drew W. Rasco, and Daniel D. Von Hoff
- Abstract
Correlation analysis between pharmacodynamic and pharmacokinetic findings in part 1
- Published
- 2023
- Full Text
- View/download PDF
4. Supplementary Figure 2 from Phase I Study of CC-486 Alone and in Combination with Carboplatin or nab-Paclitaxel in Patients with Relapsed or Refractory Solid Tumors
- Author
-
Johanna C. Bendell, Eric Laille, Kejian Liu, Aaron Nguyen, Jorge DiMartino, Gordon L. Bray, Patricia M. LoRusso, Miguel Martin, W. Jeff Edenfield, Jose A. Lopez-Martin, Ron H.J. Mathijssen, Bert O'Neil, Christophe Le Tourneau, Nicolas Isambert, Jan H.M. Schellens, Pamela N. Munster, Elisabeth I. Heath, Drew W. Rasco, and Daniel D. Von Hoff
- Abstract
Mean paclitaxel plasma concentrations following nab-paclitaxel administration alone or with CC-486 in part 1 arm B
- Published
- 2023
- Full Text
- View/download PDF
5. Supplementary Tables 1-8 from Phase I Study of CC-486 Alone and in Combination with Carboplatin or nab-Paclitaxel in Patients with Relapsed or Refractory Solid Tumors
- Author
-
Johanna C. Bendell, Eric Laille, Kejian Liu, Aaron Nguyen, Jorge DiMartino, Gordon L. Bray, Patricia M. LoRusso, Miguel Martin, W. Jeff Edenfield, Jose A. Lopez-Martin, Ron H.J. Mathijssen, Bert O'Neil, Christophe Le Tourneau, Nicolas Isambert, Jan H.M. Schellens, Pamela N. Munster, Elisabeth I. Heath, Drew W. Rasco, and Daniel D. Von Hoff
- Abstract
Table S1. CC-486 Plus Carboplatin Dose Levels in Part 1 Arm A Table S2. CC-486 Plus nab-Paclitaxel Dose Levels in Part 1 Arm B Table S3. CC-486 Dose Levels in Part 1 Arm C Table S4. Number of Patients With Treatment Emergent Adverse Events for Part 1 Table S5. Number of Patients With Treatment-Emergent Adverse Events in Any Treatment Arm in Part 2 Table S6. Mean CC-486 Plasma Pharmacokinetic Parameters Table S7. Efficacy of CC-486 Alone or in Combination With Carboplatin or nab-Paclitaxel in Part 2 Table S8. Prior Treatment History of NPV Patients
- Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Figure 1 from Phase I Study of CC-486 Alone and in Combination with Carboplatin or nab-Paclitaxel in Patients with Relapsed or Refractory Solid Tumors
- Author
-
Johanna C. Bendell, Eric Laille, Kejian Liu, Aaron Nguyen, Jorge DiMartino, Gordon L. Bray, Patricia M. LoRusso, Miguel Martin, W. Jeff Edenfield, Jose A. Lopez-Martin, Ron H.J. Mathijssen, Bert O'Neil, Christophe Le Tourneau, Nicolas Isambert, Jan H.M. Schellens, Pamela N. Munster, Elisabeth I. Heath, Drew W. Rasco, and Daniel D. Von Hoff
- Abstract
Mean total carboplatin plasma concentration following carboplatin administration alone or with CC-486 in part 1 arm A
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Methods Data from Phase I Study of CC-486 Alone and in Combination with Carboplatin or nab-Paclitaxel in Patients with Relapsed or Refractory Solid Tumors
- Author
-
Johanna C. Bendell, Eric Laille, Kejian Liu, Aaron Nguyen, Jorge DiMartino, Gordon L. Bray, Patricia M. LoRusso, Miguel Martin, W. Jeff Edenfield, Jose A. Lopez-Martin, Ron H.J. Mathijssen, Bert O'Neil, Christophe Le Tourneau, Nicolas Isambert, Jan H.M. Schellens, Pamela N. Munster, Elisabeth I. Heath, Drew W. Rasco, and Daniel D. Von Hoff
- Abstract
Pharmacodynamic Assessments
- Published
- 2023
- Full Text
- View/download PDF
8. Data from Phase I Study of CC-486 Alone and in Combination with Carboplatin or nab-Paclitaxel in Patients with Relapsed or Refractory Solid Tumors
- Author
-
Johanna C. Bendell, Eric Laille, Kejian Liu, Aaron Nguyen, Jorge DiMartino, Gordon L. Bray, Patricia M. LoRusso, Miguel Martin, W. Jeff Edenfield, Jose A. Lopez-Martin, Ron H.J. Mathijssen, Bert O'Neil, Christophe Le Tourneau, Nicolas Isambert, Jan H.M. Schellens, Pamela N. Munster, Elisabeth I. Heath, Drew W. Rasco, and Daniel D. Von Hoff
- Abstract
Purpose: This large two-part, three-arm phase I study examined the safety and tolerability of CC-486 (an oral formulation of azacitidine, a hypomethylating agent) alone or in combination with the cytotoxic agents, carboplatin or nab-paclitaxel, in patients with advanced unresectable solid tumors.Patients and Methods: Part 1 (n = 57) was a dose escalation of CC-486 alone (arm C) or with carboplatin (arm A) or nab-paclitaxel (arm B). The primary endpoint was safety, MTD, and recommended part 2 dose (RP2D) of CC-486. In part 2 (n = 112), the primary endpoint was the safety and tolerability of CC-486 administered at the RP2D for each treatment arm, in tumor-specific expansion cohorts. Secondary endpoints included pharmacokinetics, pharmacodynamics, and antitumor activity of CC-486.Results: At pharmacologically active doses CC-486 in combination with carboplatin or nab-paclitaxel had a tolerable safety profile and no drug–drug interactions. The CC-486 RP2D was determined as 300 mg (every day, days 1–14/21) in combination with carboplatin (arm A) or as monotherapy (arm C); and 200 mg in the same dosing regimen in combination with nab-paclitaxel (arm B). Albeit limited by the small sample size, CC-486 monotherapy resulted in partial responses (three/eight) and stable disease (four/eight) in patients with nasopharyngeal cancer. Three of the stable disease responses lasted more than 150 days.Conclusions: CC-486 is well tolerated alone or in combination with carboplatin or nab-paclitaxel. Exploratory analyses suggest clinical activity of CC-486 monotherapy in nasopharyngeal cancer and provided the basis for an ongoing phase II clinical trial (ClinicalTrials.gov identifier: NCT02269943). Clin Cancer Res; 24(17); 4072–80. ©2018 AACR.
- Published
- 2023
- Full Text
- View/download PDF
9. Enasidenib vs conventional care in older patients with late-stage mutant-IDH2 relapsed/refractory AML: a randomized phase 3 trial
- Author
-
Stéphane de Botton, Pau Montesinos, Andre C. Schuh, Cristina Papayannidis, Paresh Vyas, Andrew H. Wei, Hans Ommen, Sergey Semochkin, Hee-Je Kim, Richard A. Larson, Jaime Koprivnikar, Olga Frankfurt, Felicitas Thol, Jörg Chromik, Jenny Byrne, Arnaud Pigneux, Xavier Thomas, Olga Salamero, Maria Belen Vidriales, Vadim Doronin, Hartmut Döhner, Amir T. Fathi, Eric Laille, Xin Yu, Maroof Hasan, Patricia Martin-Regueira, Courtney D. DiNardo, Institut Català de la Salut, [de Botton S] Gustave Roussy, Université Paris-Saclay, Villejeuf, France. [Montesinos P] Hospital Universitari i Politecnic La Fe, Valencia, Spain. [Schuh AC] Princess Margaret Cancer Centre, Toronto, ON, Canada. [Papayannidis C] IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia 'Seràgnoli', Bologna, Italy. [Vyas P] Oxford Biomedical Research Centre and Oxford University Hospitals National Health Service Foundation Trust, Oxford, United Kingdom. [Wei AH] The Alfred Hospital, Melbourne, VIC, Australia. Monash University, Melbourne, VIC, Australia. [Salamero O] Vall d’Hebron Hospital Universitari, Barcelona, Spain. Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain, and Vall d'Hebron Barcelona Hospital Campus
- Subjects
Otros calificadores::/uso terapéutico [Otros calificadores] ,Immunology ,Genetic Phenomena::Genetic Variation::Mutation [PHENOMENA AND PROCESSES] ,Medicaments antineoplàstics - Ús terapèutic ,Neoplasms::Neoplasms by Histologic Type::Leukemia::Leukemia, Myeloid::Leukemia, Myeloid, Acute [DISEASES] ,Otros calificadores::Otros calificadores::/farmacoterapia [Otros calificadores] ,acciones y usos químicos::acciones farmacológicas::usos terapéuticos::antineoplásicos [COMPUESTOS QUÍMICOS Y DROGAS] ,Cell Biology ,Hematology ,Other subheadings::Other subheadings::/drug therapy [Other subheadings] ,Biochemistry ,neoplasias::neoplasias por tipo histológico::leucemia::leucemia mieloide::leucemia mieloide aguda [ENFERMEDADES] ,Anomalies cromosòmiques ,Leucèmia mieloide aguda - Aspectes genètics ,Leucèmia mieloide aguda - Tractament ,Other subheadings::/therapeutic use [Other subheadings] ,Chemical Actions and Uses::Pharmacologic Actions::Therapeutic Uses::Antineoplastic Agents [CHEMICALS AND DRUGS] ,fenómenos genéticos::variación genética::mutación [FENÓMENOS Y PROCESOS] - Abstract
Enasidenib; Conventional care Enasidenib; Atenció convencional Enasidenib; Atención convencional This open-label, randomized, phase 3 trial (NCT02577406) compared enasidenib, an oral IDH2 (isocitrate dehydrogenase 2) inhibitor, with conventional care regimens (CCRs) in patients aged ≥60 years with late-stage, mutant-IDH2 acute myeloid leukemia (AML) relapsed/refractory (R/R) to 2 or 3 prior AML-directed therapies. Patients were first preselected to a CCR (azacitidine, intermediate-dose cytarabine, low-dose cytarabine, or supportive care) and then randomized (1:1) to enasidenib 100 mg per day or CCR. The primary endpoint was overall survival (OS). Secondary endpoints included event-free survival (EFS), time to treatment failure (TTF), overall response rate (ORR), hematologic improvement (HI), and transfusion independence (TI). Overall, 319 patients were randomized to enasidenib (n = 158) or CCR (n = 161). The median age was 71 years, median (range) enasidenib exposure was 142 days (3 to 1270), and CCR was 36 days (1 to 1166). One enasidenib (0.6%) and 20 CCR (12%) patients received no randomized treatment, and 30% and 43%, respectively, received subsequent AML-directed therapies during follow-up. The median OS with enasidenib vs CCR was 6.5 vs 6.2 months (HR [hazard ratio], 0.86; P = .23); 1-year survival was 37.5% vs 26.1%. Enasidenib meaningfully improved EFS (median, 4.9 vs 2.6 months with CCR; HR, 0.68; P = .008), TTF (median, 4.9 vs 1.9 months; HR, 0.53; P < .001), ORR (40.5% vs 9.9%; P
- Published
- 2022
10. Pharmacokinetics and safety of apremilast (CC-10004) in subjects with hepatic impairment.
- Author
-
Mahmoud S. Assaf, Eric Laille, Liangang Liu, Edward O'Mara, Anfan Wu, Maria Palmisano, Thomas C. Marbury, and Richard A. Preston
- Published
- 2014
- Full Text
- View/download PDF
11. Enasidenib vs conventional care in mutant-IDH2 relapsed/refractory acute myeloidleukemia: a randomized, phase 3 trial
- Author
-
Stéphane, de Botton, Pau, Montesinos, Andre, Schuh, Cristina, Papayannidis, Paresh, Vyas, Andrew H, Wei, Hans Beier, Ommen, Sergey, Semochkin, Hee-Je, Kim, Richard A, Larson, Jamie, Koprivnikar, Olga, Frankfurt, Felicitas R, Thol, Jörg, Chromik, Jenny L, Byrne, Arnaud, Pigneux, Xavier, Thomas, Olga, Salamero, María-Belén, Vidriales, Vadim A, Doronin, Hartmut, Döhner, Amir T, Fathi, Eric, Laille, Xin, Yu, Maroof, Hasan, Patricia, Martín-Regueira, and Courtney D, DiNardo
- Abstract
This open-label, randomized, phase 3 trial (NCT02577406) compared enasidenib, an oral IDH2 inhibitor, with conventional care regimens (CCR) in patients aged =60 years with late-stage, mutant-IDH2 acute myeloid leukemia (AML) relapsed/refractory (R/R) to 2 or 3 prior AML-directed therapies. Patients were first preselected to a CCR (azacitidine, intermediate-dose cytarabine, low-dose cytarabine, or supportive care), and then randomized (1:1) to enasidenib 100 mg/day or CCR. The primary endpoint was overall survival (OS). Secondary endpoints included event-free survival (EFS), time to treatment failure (TTF), overall response rate (ORR), hematologic improvement (HI), and transfusion independence (TI). Overall, 319 patients were randomized to enasidenib (n=158) or CCR (n=161). Median age was 71 years. Median (range) enasidenib exposure was 142 days (3-1270) and CCR was 36 days (1-1166). One enasidenib (0.6%) and 20 CCR (12%) patients received no randomized treatment, and 30% and 43%, respectively, received subsequent AML-directed therapies during follow-up. Median OS with enasidenib vs CCR was 6.5 vs 6.2 months (HR [hazard ratio] 0.86; P=.23); 1-year survival was 37.5% vs 26.1%. Enasidenib meaningfully improved EFS (median 4.9 months, vs 2.6 months with CCR; HR 0.68; P=.008), TTF (median 4.9 vs 1.9 months, HR 0.53; P
- Published
- 2022
12. Evaluation of the bioequivalence and food effect on the bioavailability of CC-486 (oral azacitidine) tablets in adult patients with cancer
- Author
-
Thai H. Ho, C.L. Beach, Eric Laille, Barry S. Skikne, Michael R. Savona, Kao Tai Tsai, Dale R. Shepard, Maen Abdelrahim, Mohammed M. Milhem, Hani M. Babiker, William Jeffery Edenfield, Joseph Aisner, and Swaminathan P. Iyer
- Subjects
Pharmacology ,Cancer Research ,medicine.medical_specialty ,FOOD EFFECT ,Oral azacitidine ,Adult patients ,business.industry ,Short Communication ,Pharmacology toxicology ,Cancer ,Bioequivalence ,Toxicology ,medicine.disease ,Oral Azacitidine ,Bioavailability ,Oncology ,Pharmacokinetics ,Internal medicine ,medicine ,Pharmacology (medical) ,business ,CC-486 - Abstract
Purpose CC-486 is an oral formulation of azacitidine that allows for extended dosing schedules to prolong azacitidine exposure to malignant cells and maximize clinical activity. CC-486 300 mg daily, administered for 14 or 21 days of 28-day treatment cycles, is currently under investigation in two ongoing phase III trials. The 300-mg daily dose in these studies is administered as two 150-mg tablets (Formulation A). Methods We evaluated the bioequivalence of one 300-mg CC-486 tablet (Formulation B) with Formulation A and food effect on Formulation B, in adult patients with cancer in a 2-stage crossover design study. Results The ratios of the geometric means of the maximum azacitidine plasma concentration (Cmax) and of the area under the plasma concentration–time curve from time 0 extrapolated to infinity (AUC∞) were 101.5% and 105.7%, demonstrating the bioequivalence of Formulations A and B. Formulation B was rapidly absorbed under fasted and fed conditions. The geometric mean of Cmax was significantly decreased by ~ 21% in the fed state. Median Tmax was reached at 2 h and 1 h post-dose in fed and fasted states, respectively (P Cmax and Tmax are not expected to have a clinical impact. Conclusion The single 300-mg CC-486 tablet was bioequivalent to two 150-mg tablets, which have shown to be efficacious and generally well-tolerated in clinical trials, and can be taken with or without food.
- Published
- 2020
- Full Text
- View/download PDF
13. Oral azacitidine plus venetoclax in patients with relapsed/refractory or newly diagnosed acute myeloid leukemia: The phase 1b OMNIVERSE trial
- Author
-
Farhad Ravandi, Hetty E Carraway, Lilia Taningco, Eric Laille, Jing Gong, Thomas Prebet, Daniel Lopes de Menezes, and Andrew H. Wei
- Subjects
Cancer Research ,Oncology - Abstract
TPS7068 Background: For patients (pts) with acute myeloid leukemia (AML) who cannot undergo intensive chemotherapy (IC), lower-intensity treatment (Tx) regimens, including low-dose cytarabine (LDAC) and hypomethylating agents (HMAs; azacitidine [AZA], decitabine), are generally well tolerated but are associated with lower response rates than IC [Vey 2020]. Similarly, the BCL2 inhibitor, venetoclax (VEN), has shown antileukemic activity, although only modest clinical benefit as monotherapy [Konopleva 2016]. VEN + AZA shows synergistic activity in preclinical models, enhancing leukemic cell apoptosis in vitro and anti-tumor activity in vivo [Jin 2020]. In IC-ineligible pts with ND AML, VEN + injectable AZA significantly increased complete remission (CR)/CR with incomplete hematologic recovery (CRi) rates ( P < 0.001) and prolonged overall survival (OS; P < 0.001) vs AZA only [DiNardo 2020]. VEN, in combination with an HMA or LDAC, is approved in the US for Tx of pts with ND AML ≥ 75 years (y) of age or who cannot undergo IC due to comorbidities. Oral-AZA (CC-486) is approved for Tx of pts with newly diagnosed (ND) AML in first CR or CRi after IC who cannot receive curative therapy (eg, HSCT). In the phase 3 QUAZAR AML-001 trial, maintenance Tx with Oral-AZA 300 mg QD for 14 days (d)/28-d Tx cycle improved OS and relapse-free survival vs placebo in older pts in CR/CRi after IC [Wei 2020]. Incorporation of AZA into DNA is S-phase-restricted; thus, extending AZA exposure over a longer duration by using an oral formulation increases the opportunity for cycling tumor cells to incorporate the drug to sustain therapeutic activity [Laille 2015]. Additionally, an all-oral combination regimen allows for outpatient administration to optimize pt convenience and reduce resource utilization. Methods: OMNIVERSE (NCT04887857) is an open-label, multicenter, 2-part phase 1b trial. The main goals are to evaluate safety and establish the maximum tolerated dose of Oral-AZA + VEN in pts ≥ 18 y of age with relapsed/refractory AML who are ineligible for further IC ( part 1), and subsequently, in pts with ND AML ≥ 75 y of age, or pts 18–74 y of age with comorbidities that prevent use of IC or HSCT ( part 2). Key eligibility criteria include ECOG performance status of 0–2 (ECOG 3 is allowed for pts 18–74 y of age with comorbidities) and unfavorable-risk cytogenetics for pts with ND AML. The Oral-AZA starting dose is 300 mg QD × 14 d/28-d cycle, which can be de-escalated to 200 mg QD × 14 d depending on dose-limiting toxicities; oral VEN 400 mg QD is taken continuously (or 21 d/cycle for dose level −2). A modified toxicity probability interval-2 design is used to evaluate dose levels. Sample size depends on the dose levels utilized (≤ 18 pts/part). Enrollment began in 2021. The trial is ongoing at clinical sites in the United States and Australia. Clinical trial information: NCT04887857.
- Published
- 2022
- Full Text
- View/download PDF
14. Phase III, randomized, placebo-controlled trial of CC-486 (oral azacitidine) in patients with lower-risk myelodysplastic syndromes
- Author
-
Paresh Vyas, Dietger Niederwieser, Rena Buckstein, David Valcárcel, Valentina Giai, Gianluigi Reda, C.L. Beach, Guillermo Garcia-Manero, Ignazia La Torre, Jake Shortt, Valeria Santini, Barry S. Skikne, Moshe Mittelman, Lewis R. Silverman, Uwe Platzbecker, Jianhua Zhong, Anna Jonasova, Aristoteles Giagounidis, Luana Fianchi, Maria Diez Campelo, Pierre Fenaux, Stephen Larsen, Esther Oliva, Eric Laille, Antonio Almeida, Francesco Buccisano, Jose F Falantes, and Daniel Menezes
- Subjects
Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Anemia ,Azacitidine ,Placebo-controlled study ,Administration, Oral ,Lower risk ,Oral Azacitidine ,Internal medicine ,medicine ,Humans ,Prospective Studies ,Prospective cohort study ,Survival rate ,Aged ,Aged, 80 and over ,business.industry ,Myelodysplastic syndromes ,Middle Aged ,medicine.disease ,Prognosis ,Settore MED/15 ,Survival Rate ,Oncology ,Myelodysplastic Syndromes ,Female ,Follow-Up Studies ,business ,medicine.drug - Abstract
PURPOSE Treatment options are limited for patients with lower-risk myelodysplastic syndromes (LR-MDS). This phase III, placebo-controlled trial evaluated CC-486 (oral azacitidine), a hypomethylating agent, in patients with International Prognostic Scoring System LR-MDS and RBC transfusion–dependent anemia and thrombocytopenia. METHODS Patients were randomly assigned 1:1 to CC-486 300-mg or placebo for 21 days/28-day cycle. The primary end point was RBC transfusion independence (TI). RESULTS Two hundred sixteen patients received CC-486 (n = 107) or placebo (n = 109). The median age was 74 years, median platelet count was 25 × 109/L, and absolute neutrophil count was 1.3 × 109/L. In the CC-486 and placebo arms, 31% and 11% of patients, respectively, achieved RBC-TI ( P = .0002), with median durations of 11.1 and 5.0 months. Reductions of ≥ 4 RBC units were attained by 42.1% and 30.6% of patients, respectively, with median durations of 10.0 and 2.3 months, and more CC-486 patients had ≥ 1.5 g/dL hemoglobin increases from baseline (23.4% v 4.6%). Platelet hematologic improvement rate was higher with CC-486 (24.3% v 6.5%). Underpowered interim overall survival analysis showed no difference between CC-486 and placebo (median, 17.3 v 16.2 months; P = .96). Low-grade GI events were the most common adverse events in both arms. In the CC-486 and placebo arms, 90% and 73% of patients experienced a grade 3-4 adverse event. Overall death rate was similar between arms, but there was an imbalance in deaths during the first 56 days (CC-486, n = 16; placebo, n = 6), most related to infections; the median pretreatment absolute neutrophil count for the 16 CC-486 patients was 0.57 × 109/L. CONCLUSION CC-486 significantly improved RBC-TI rate and induced durable bilineage improvements in patients with LR-MDS and high-risk disease features. More early deaths occurred in the CC-486 arm, most related to infections in patients with significant pretreatment neutropenia. Further evaluation of CC-486 in MDS is needed.
- Published
- 2021
15. Azacitidine is removed effectively by hemodialysis
- Author
-
Gert Mayer, H Neuwirt, Eric Laille, Reinhard Stauder, and A G E Beer
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,medicine.medical_treatment ,Azacitidine ,03 medical and health sciences ,0302 clinical medicine ,Renal Dialysis ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Humans ,neoplasms ,business.industry ,Myelodysplastic syndromes ,Myeloid leukemia ,Hematology ,medicine.disease ,stomatognathic diseases ,030220 oncology & carcinogenesis ,Myelodysplastic Syndromes ,Hemodialysis ,business ,030215 immunology ,medicine.drug - Abstract
Azacitidine for injection (AZA) (Vidaza, Celgene Corp., Summit, NJ), represents a well-established and registered agent in Myelodysplastic Syndromes (MDS) and in acute myeloid leukemia (AML) [1]. A...
- Published
- 2020
16. Phase I and Pharmacokinetic Study of Romidepsin in Patients with Cancer and Hepatic Dysfunction: A National Cancer Institute Organ Dysfunction Working Group Study
- Author
-
Roisin M. Connolly, Eric Laille, Ulka Vaishampayan, Vincent Chung, Karen Kelly, Afshin Dowlati, Olatunji B. Alese, R. Donald Harvey, Paul Haluska, Lillian L. Siu, Shivaani Kummar, Richard Piekarz, S. Percy Ivy, Nicole M. Anders, Melinda Downs, Ashley O'Connor, Angela Scardina, Jacqueline Saunders, Gary L. Rosner, Michael A. Carducci, and Michelle A. Rudek
- Subjects
0301 basic medicine ,Oncology ,Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Multiple Organ Failure ,Phases of clinical research ,Antineoplastic Agents ,Lymphoma, T-Cell ,Article ,Romidepsin ,03 medical and health sciences ,0302 clinical medicine ,Pharmacokinetics ,Internal medicine ,Depsipeptides ,Medicine ,Humans ,Dosing ,Adverse effect ,Aged ,business.industry ,Liver Diseases ,Organ dysfunction ,Cancer ,Middle Aged ,medicine.disease ,National Cancer Institute (U.S.) ,United States ,030104 developmental biology ,Liver ,030220 oncology & carcinogenesis ,Cohort ,Female ,medicine.symptom ,business ,medicine.drug - Abstract
Purpose: Romidepsin dosing recommendations for patients with malignancy and varying degrees of hepatic dysfunction was lacking at the time of regulatory approval for T-cell lymphoma. We conducted a multicenter phase I clinical trial (ETCTN-9008) via the NCI Organ Dysfunction Working Group to investigate safety, first cycle MTD, and pharmacokinetic profile of romidepsin in this setting. Patients and Methods: Patients with select advanced solid tumors or hematologic malignancies were stratified according to hepatic function. Romidepsin was administered intravenously on days 1, 8, and 15 of a 28-day cycle and escalation followed a 3 + 3 design in moderate and severe impairment cohorts. Blood samples for detailed pharmacokinetic analyses were collected after the first dose. Results: Thirty-one patients received one dose of romidepsin and were evaluable for pharmacokinetic analyses in normal (n = 12), mild (n = 8), moderate (n = 5), and severe (n = 6) cohorts. Adverse events across cohorts were similar, and dose-limiting toxicity occurred in two patients (mild and severe impairment cohorts). The MTD was not determined because the geometric mean AUC values of romidepsin in moderate (7 mg/m2) and severe (5 mg/m2) impairment cohort were 114% and 116% of the normal cohort (14 mg/m2). Conclusions: Data from the ETCTN-9008 trial led to changes in the romidepsin labeling to reflect starting dose adjustment for patients with cancer and moderate and severe hepatic impairment, with no adjustment for mild hepatic impairment.
- Published
- 2020
17. OMNIVERSE: A Phase 1b/2 Study of Oral Azacitidine Plus Venetoclax in Patients with Relapsed/Refractory (R/R) or Newly Diagnosed (ND) Acute Myeloid Leukemia (AML)
- Author
-
Daniel Menezes, Andrew H. Wei, Kimberley Dilley, Lilia Taningco, Hetty E. Carraway, Eric Laille, Farhad Ravandi, and Jing Gong
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Venetoclax ,Immunology ,Myeloid leukemia ,Cell Biology ,Hematology ,Newly diagnosed ,Biochemistry ,Oral Azacitidine ,chemistry.chemical_compound ,chemistry ,Internal medicine ,Relapsed refractory ,medicine ,In patient ,business - Abstract
Background: Novel AML treatments (Tx) have improved the tolerability and clinical outcomes for older patients (pts) with AML, especially since standard intensive chemotherapy (IC) regimens are associated with risks of toxicity and mortality. Lower-intensity Tx options for IC-ineligible pts include low-dose cytarabine (LDAC) or hypomethylating agents (HMAs; azacitidine [AZA] and decitabine), which are generally well tolerated, but have been associated with inferior outcomes vs IC [Vey 2020]. As such, the development of less-toxic regimens with better efficacy has been a priority, and increased focus has been given to optimizing Tx regimens in the outpatient setting for convenience, independence, and quality of life (QoL). The oral formulation of AZA (Oral-AZA [CC-486]) is approved in the United States, European Union, and Canada for Tx of pts with ND AML in first remission after IC who are not able to complete curative therapy (eg, HSCT). In a pivotal phase 3 trial, Oral-AZA 300 mg QD for 14 days (d)/28d Tx cycle was well tolerated and improved overall survival (OS) vs placebo in pts with AML in CR/CRi after IC. Oral-AZA has also shown preliminary clinical efficacy in pts with active AML [Savona 2015]. Novel regimens based on LDAC or HMAs in combination with newer agents, including the BCL2 inhibitor, venetoclax (VEN), have demonstrated promising activity in older pts with AML. Although only modest clinical benefits were achieved with single-agent VEN in pts with R/R AML [Konopleva 2016], the combination of VEN + AZA in pts with ND AML shows synergistic anti-leukemic activity via AZA-mediated inhibition of MCL1 and BCL-XL, enhancing BCL2 dependence in leukemic cells. In IC-ineligible ND AML, VEN + injectable AZA significantly improved remission rates and OS vs AZA alone [DiNardo 2020]. Consequently, VEN, in combination with an HMA or LDAC, was approved in the US for Tx of pts with ND AML aged ≥ 75 years (y) or those with comorbidities precluding use of IC. Given S-phase-restricted DNA incorporation of AZA, extended Oral-AZA dosing regimens (> 7d/28d Tx cycle) increases the opportunity for cycling cells to incorporate the drug, thereby sustaining epigenetic activity [Laille 2015]. An outpatient VEN + AZA oral combination regimen may also increase the likelihood of pt adherence [Eek 2016]. Here, we describe the study design and objectives of the OMNIVERSE trial of Oral-AZA + VEN in older pts with ND AML or pts with AML R/R to prior Tx. Study Design and Methods: OMNIVERSE (NCT04887857) is a multicenter, open-label, phase 1b/2 trial conducted in 3 sequential study parts (Figure). Key objectives of the 2-part, dose-finding phase 1b portion are to evaluate safety and establish the maximum tolerated dose (MTD) and/or recommended phase 2 dose (RP2D) of Oral-AZA + VEN, first in pts with R/R AML (World Health Organization [WHO] 2016 criteria) ineligible to receive further IC (part 1), and subsequently in pts with ND AML (WHO 2016) aged ≥ 75 y, or those aged ≥ 18-74 y with comorbidities that preclude use of IC or HSCT (part 2). Key pt eligibility criteria include an Eastern Cooperative Oncology Group (ECOG) performance status of 0-2 (ECOG 3 is allowed for pts aged ≥ 18-74 y with comorbidities), and an unfavorable cytogenetic risk profile for pts with ND AML. The starting dose of Oral-AZA is 300 mg QD × 14d cycle, which can be escalated to 300 mg × 21d or de-escalated to 200 mg × 14d/28d cycle depending on dose-limiting toxicities experienced; oral VEN is taken QD continuously (or 21d/cycle for dose level -2). A modified toxicity probability interval-2 (mTPI-2) design is used to evaluate the planned dose levels. The sample size of the phase 1b portion depends on the dose levels utilized (≤ 18 pts in each part). In phase 2, pts with ND AML (same inclusion criteria as for phase 1b-part 2) will receive Oral-AZA + VEN at the RP2D. The primary endpoint is the rate of complete remission (CR) plus CR with partial hematologic recovery (CRh). Key secondary endpoints include safety, duration of CR/CRh, overall response rate (per European Leukemia Net criteria), and longitudinal minimal residual disease (MRD) profiling for CR/CRh responders. Key exploratory objectives are to characterize DNA methylation patterns and BCL2 family member expression profiles. The phase 2 expansion will enroll ~100 pts, which is calculated based on the response rates from the M14-358 (NCT02203773) and VIALE-A (NCT02993523) studies. Enrollment is planned to begin in 2021. Figure 1 Figure 1. Disclosures Ravandi: Amgen: Honoraria, Research Funding; Prelude: Research Funding; Syros Pharmaceuticals: Consultancy, Honoraria, Research Funding; Taiho: Honoraria, Research Funding; Xencor: Honoraria, Research Funding; Jazz: Honoraria, Research Funding; Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Astex: Honoraria, Research Funding; Novartis: Honoraria; Agios: Honoraria, Research Funding; AbbVie: Honoraria, Research Funding; AstraZeneca: Honoraria; Bristol Myers Squibb: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding. Carraway: Takeda: Other: Independent review committee; Celgene, a Bristol Myers Squibb company: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Astex: Other: Independent review committee; Stemline: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Jazz: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Agios: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; AbbVie: Other: Independent review committee; Novartis: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Bristol Myers Squibb: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau. Taningco: Bristol Myers Squibb: Current Employment. Dilley: Bristol Myers Squibb: Current Employment, Current equity holder in publicly-traded company; Abbvie: Current equity holder in publicly-traded company, Ended employment in the past 24 months. Laille: Bristol Myers Squibb: Current Employment, Current equity holder in publicly-traded company. Gong: Bristol Myers Squibb: Current Employment, Current equity holder in publicly-traded company, Divested equity in a private or publicly-traded company in the past 24 months. Lopes de Menezes: Bristol Myers Squibb: Current Employment, Current equity holder in publicly-traded company, Patents & Royalties. Wei: Abbvie, Amgen, Astellas, AstraZeneca, Celgene/BMS, Genentech, Janssen, MacroGenics, Novartis, Pfizer, and Servier: Membership on an entity's Board of Directors or advisory committees; Novartis, Abbvie, Celgene/BMS: Speakers Bureau; Former employee of Walter and Eliza Hall Institute: Patents & Royalties: Prof. Andrew Wei is a former employee of the Walter and Eliza Hall Institute and is eligible for a fraction of the royalty stream related to Venetoclax; Abbvie, Amgen, Astellas, AstraZeneca, Celgene/BMS, Genentech, Janssen, MacroGenics, Novartis, Pfizer, and Servier: Honoraria; Servier: Consultancy; Abbvie, Amgen, AstraZeneca, Celgene/BMS, Novartis, Servier and F. Hoffmann-La Roche: Research Funding.
- Published
- 2021
- Full Text
- View/download PDF
18. Romidepsin in Japanese patients with relapsed or refractory peripheral T-cell lymphoma: a phase I/II and pharmacokinetics study
- Author
-
Toru Kiguchi, Hiromi Tamakoshi, Kunihiro Tsukasaki, Michinori Ogura, Kensei Tobinai, Kiyohiko Hatake, Tokihiro Ro, Masafumi Taniwaki, Eric Laille, Naoki Kobayashi, Yoichi Tatsumi, Dai Maruyama, Takayuki Ikezoe, Kiyoshi Ando, Koji Kato, Takashi Ishida, Sanae Sakurai, Kenichi Ishizawa, Tomoko Ohtsu, and Toshiki Uchida
- Subjects
Male ,medicine.medical_specialty ,Neutropenia ,Gastroenterology ,Disease-Free Survival ,Romidepsin ,03 medical and health sciences ,0302 clinical medicine ,Refractory ,Pharmacokinetics ,Recurrence ,Depsipeptides ,Internal medicine ,medicine ,Humans ,Adverse effect ,Aged ,Aged, 80 and over ,Leukopenia ,business.industry ,Lymphoma, T-Cell, Peripheral ,Hematology ,Middle Aged ,medicine.disease ,Lymphoma ,Surgery ,Survival Rate ,030220 oncology & carcinogenesis ,Toxicity ,Female ,medicine.symptom ,business ,030215 immunology ,medicine.drug - Abstract
This phase I/II multicenter study evaluated romidepsin treatment in Japanese patients with relapsed/refractory peripheral T-cell lymphoma (PTCL) or cutaneous T-cell lymphoma (CTCL). Patients aged ≥20 years received romidepsin via a 4-h intravenous infusion on days 1, 8, and 15 of each 28-day cycle. Phase I used a 3 + 3 design to identify any dose-limiting toxicity (DLT) for regimens of romidepsin 9 and 14 mg/m2. The primary endpoints for phase I and II were DLT and overall response rate (ORR), respectively. Intent-to-treat patients were those who received ≥1 romidepsin dose (PTCL, n = 48; CTCL, n = 2). In phase I, none of the patients (n = 3, 9 mg/m2; n = 6, 14 mg/m2) exhibited DLT. In phase II, 40 patients with PTCL were treated with 14 mg/m2 romidepsin. The most common treatment-emergent grade ≥3 adverse events were lymphopenia (74%), neutropenia (54%), leukopenia (46%), and thrombocytopenia (38%). Patients in phase II showed a 43% ORR, including 25% complete responses. Median progression-free survival was 5.6 months and median duration of response was 11.1 months. This phase I/II study identified a well-tolerated dose of romidepsin, with an acceptable toxicity profile and clinically meaningful efficacy in Japanese patients with relapsed/refractory PTCL. ClinicalTrials.gov Identifier NCT01456039.
- Published
- 2017
- Full Text
- View/download PDF
19. Correction to: Evaluation of the bioequivalence and food effect on the bioavailability of CC-486 (oral azacitidine) tablets in adult patients with cancer
- Author
-
Michael R. Savona, Hani M. Babiker, Dale R. Shepard, Barry S. Skikne, Maen Abdelrahim, Kao Tai Tsai, Joseph Aisner, C.L. Beach, Thai H. Ho, Mohammed M. Milhem, William Jeffery Edenfield, Eric Laille, and Swaminathan P. Iyer
- Subjects
Pharmacology ,Cancer Research ,FOOD EFFECT ,Cancer chemotherapy ,Adult patients ,business.industry ,Cancer ,Correction ,Bioequivalence ,Toxicology ,medicine.disease ,Oral Azacitidine ,Bioavailability ,Oncology ,medicine ,Pharmacology (medical) ,business - Abstract
CC-486 is an oral formulation of azacitidine that allows for extended dosing schedules to prolong azacitidine exposure to malignant cells and maximize clinical activity. CC-486 300 mg daily, administered for 14 or 21 days of 28-day treatment cycles, is currently under investigation in two ongoing phase III trials. The 300-mg daily dose in these studies is administered as two 150-mg tablets (Formulation A).We evaluated the bioequivalence of one 300-mg CC-486 tablet (Formulation B) with Formulation A and food effect on Formulation B, in adult patients with cancer in a 2-stage crossover design study.The ratios of the geometric means of the maximum azacitidine plasma concentration (CThe single 300-mg CC-486 tablet was bioequivalent to two 150-mg tablets, which have shown to be efficacious and generally well-tolerated in clinical trials, and can be taken with or without food.
- Published
- 2020
20. Phase I Study of CC-486 Alone and in Combination with Carboplatin or nab-Paclitaxel in Patients with Relapsed or Refractory Solid Tumors
- Author
-
Aaron N. Nguyen, Johanna C. Bendell, Patricia LoRusso, Christophe Le Tourneau, Miguel Martin, Drew W. Rasco, W. Jeff Edenfield, Pamela N. Munster, Jose A. Lopez-Martin, Bert H. O'Neil, Gordon L. Bray, Elisabeth I. Heath, Eric Laille, Jorge DiMartino, Kejian Liu, Jan H.M. Schellens, Nicolas Isambert, Daniel D. Von Hoff, Ron H.J. Mathijssen, Medical Oncology, The Translational Genomics Research Institute (TGen), South Texas Accelerated Research Therapeutics [San Antonio, Texas] (START), Karmanos Cancer Institute (Detroit), Department of Medicine [San Francisco], University of California [San Francisco] (UCSF), University of California-University of California, Netherlands Cancer Institute (NKI), Antoni van Leeuwenhoek Hospital, Centre Régional de Lutte contre le cancer Georges-François Leclerc [Dijon] (UNICANCER/CRLCC-CGFL), UNICANCER, Département d'Oncologie Médicale [Institut Curie, Paris], Institut Curie [Paris], Indiana University School of Medicine, Indiana University System, Erasmus MC Cancer Institute, Rotterdam, 12 de Octubre University Hospital, Greenville Memorial Hospital [University of South Carolina, Greenville] (GHS), East Carolina University [Greenville] (ECU), University of North Carolina System (UNC)-University of North Carolina System (UNC), Hospital General Universitario 'Gregorio Marañón' [Madrid], Celgene Corporation, and Sarah Cannon Research Institute [Nashville, Tennessee]
- Subjects
0301 basic medicine ,Oncology ,Male ,Cancer Research ,oral azacitidine ,design ,Carboplatin ,chemistry.chemical_compound ,0302 clinical medicine ,Neoplasms ,Antineoplastic Combined Chemotherapy Protocols ,Clinical endpoint ,6.2 Cellular and gene therapies ,Cancer ,trial ,Middle Aged ,myelodysplastic syndromes ,3. Good health ,Treatment Outcome ,Tolerability ,030220 oncology & carcinogenesis ,6.1 Pharmaceuticals ,Azacitidine ,Female ,acute myeloid-leukemia ,safety ,medicine.medical_specialty ,Paclitaxel ,Maximum Tolerated Dose ,Clinical Trials and Supportive Activities ,Oncology and Carcinogenesis ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Article ,Disease-Free Survival ,Drug Administration Schedule ,03 medical and health sciences ,5-azacytidine ,Pharmacokinetics ,SDG 3 - Good Health and Well-being ,Clinical Research ,Internal medicine ,Albumins ,medicine ,cancer ,Humans ,Oncology & Carcinogenesis ,Aged ,therapy ,business.industry ,nasopharyngeal carcinoma ,Evaluation of treatments and therapeutic interventions ,medicine.disease ,Clinical trial ,030104 developmental biology ,Hypomethylating agent ,chemistry ,Pharmacodynamics ,business - Abstract
Purpose: This large two-part, three-arm phase I study examined the safety and tolerability of CC-486 (an oral formulation of azacitidine, a hypomethylating agent) alone or in combination with the cytotoxic agents, carboplatin or nab-paclitaxel, in patients with advanced unresectable solid tumors. Patients and Methods: Part 1 (n = 57) was a dose escalation of CC-486 alone (arm C) or with carboplatin (arm A) or nab-paclitaxel (arm B). The primary endpoint was safety, MTD, and recommended part 2 dose (RP2D) of CC-486. In part 2 (n = 112), the primary endpoint was the safety and tolerability of CC-486 administered at the RP2D for each treatment arm, in tumor-specific expansion cohorts. Secondary endpoints included pharmacokinetics, pharmacodynamics, and antitumor activity of CC-486. Results: At pharmacologically active doses CC-486 in combination with carboplatin or nab-paclitaxel had a tolerable safety profile and no drug–drug interactions. The CC-486 RP2D was determined as 300 mg (every day, days 1–14/21) in combination with carboplatin (arm A) or as monotherapy (arm C); and 200 mg in the same dosing regimen in combination with nab-paclitaxel (arm B). Albeit limited by the small sample size, CC-486 monotherapy resulted in partial responses (three/eight) and stable disease (four/eight) in patients with nasopharyngeal cancer. Three of the stable disease responses lasted more than 150 days. Conclusions: CC-486 is well tolerated alone or in combination with carboplatin or nab-paclitaxel. Exploratory analyses suggest clinical activity of CC-486 monotherapy in nasopharyngeal cancer and provided the basis for an ongoing phase II clinical trial (ClinicalTrials.gov identifier: NCT02269943). Clin Cancer Res; 24(17); 4072–80. ©2018 AACR.
- Published
- 2018
- Full Text
- View/download PDF
21. Extended dosing with CC-486 (oral azacitidine) in patients with myeloid malignancies
- Author
-
Amy Kellerman, Eric Laille, Edwin C. Kingsley, Carlos Becerra, Michael R. Savona, Barry S. Skikne, Stacey M. Ukrainskyj, Qian Dong, Paul Conkling, Kathryn S. Kolibaba, Robert M. Rifkin, and John C. Morris
- Subjects
Male ,medicine.medical_specialty ,Antimetabolites, Antineoplastic ,Myeloid ,Anemia ,Gastrointestinal Diseases ,Azacitidine ,Chronic myelomonocytic leukemia ,Administration, Oral ,Neutropenia ,Gene mutation ,Gastroenterology ,Drug Administration Schedule ,03 medical and health sciences ,Food-Drug Interactions ,0302 clinical medicine ,Internal medicine ,hemic and lymphatic diseases ,medicine ,Humans ,Drug Interactions ,Fatigue ,Research Articles ,Aged ,Aged, 80 and over ,Dose-Response Relationship, Drug ,business.industry ,Myelodysplastic syndromes ,Leukemia, Myelomonocytic, Chronic ,Proton Pump Inhibitors ,Hematology ,Gastric Acidity Determination ,DNA Methylation ,Middle Aged ,medicine.disease ,Hematologic Diseases ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,Tolerability ,Food ,030220 oncology & carcinogenesis ,Myelodysplastic Syndromes ,Female ,business ,030215 immunology ,medicine.drug ,Research Article - Abstract
CC‐486 (oral azacitidine) is an epigenetic modifier in clinical development for treatment of hematological cancers. This study of extended CC‐486 dosing included patients with myelodysplastic syndromes (MDSs), chronic myelomonocytic leukemia (CMML), or acute myeloid leukemia (AML). After a pharmacokinetic assessment period, 31 patients (MDS n = 18, CMML n = 4, and AML n = 9) entered a clinical phase in which they received CC‐486 300 mg once‐daily for 21 days of repeated 28‐day cycles. Median age was 71 years (range: 53‐93); 42% of patients were aged ≥75 years. A total of 5 patients with AML (63%) had prior MDS. Median number of CC‐486 treatment cycles was 4 (range: 1‐32). The most common treatment‐emergent adverse events (TEAEs) were gastrointestinal (84% of patients) and hematologic (81%). Most common grade 3‐4 TEAEs were neutropenia (n = 13, 42%) and anemia (n = 9, 29%). Ten patients experienced grade 4 neutropenia. Infrequently, CC‐486 dose was interrupted or reduced due to gastrointestinal (n = 5, 16%) or hematologic (n = 6, 19%) TEAEs. Overall response rate (complete remission [CR], CR with incomplete hematological recovery [CRi], partial remission [PR], marrow CR) in the MDS/CMML subgroups was 32% and in the AML subgroup (CR/CRi/PR) was 22%. Red blood cell transfusion independence rates in the MDS/CMML and AML subgroups were 33% and 25%, respectively, and 2 MDS/CMML patients attained hematologic improvement as a best response on‐study. No baseline gene mutation was predictive of response/nonresponse. CC‐486 allows flexible dosing and schedules to improve tolerability or response. Neutropenia in early treatment cycles deserves scrutiny and may warrant initiation of prophylactic antibiotics. KEY POINTS The safety profile of oral CC‐486 was comparable to that of injectable azacitidine; most adverse events were hematological and gastrointestinal. Extended (21‐day/cycle) CC‐486 dosing induced responses in patients with hematological malignancies, many of whom had prior DNMTi failure.
- Published
- 2018
22. CC-486 Maintenance after Stem Cell Transplantation in Patients with Acute Myeloid Leukemia or Myelodysplastic Syndromes
- Author
-
Betul Oran, Joel Hetzer, Eric Laille, Marcos de Lima, Sergio Giralt, Richard E. Champlin, Basem M. William, Esperanza B. Papadopoulos, Barry S. Skikne, Charles Craddock, Bart L. Scott, and Becky Hubbell
- Subjects
Oncology ,Adult ,Male ,medicine.medical_specialty ,Azacitidine ,Myelodysplastic syndromes ,Graft vs Host Disease ,Disease-Free Survival ,Article ,Maintenance Chemotherapy ,03 medical and health sciences ,0302 clinical medicine ,Maintenance therapy ,Internal medicine ,medicine ,Humans ,Dosing ,Aged ,Acute myeloid leukemia ,business.industry ,Hematopoietic Stem Cell Transplantation ,Myeloid leukemia ,Hematology ,Middle Aged ,medicine.disease ,Allografts ,Allogeneic stem cell ,Transplantation ,Survival Rate ,Leukemia, Myeloid, Acute ,030220 oncology & carcinogenesis ,Concomitant ,Female ,business ,Progressive disease ,030215 immunology ,medicine.drug ,CC-486 ,transplantation - Abstract
Relapse is the main cause of treatment failure after allogeneic stem cell transplant (alloSCT) in acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS). Injectable azacitidine can improve post-transplant outcomes but presents challenges with exposure and compliance. Oral CC-486 allows extended dosing to prolong azacitidine activity. We investigated use of CC-486 maintenance therapy after alloSCT. Adults with MDS or AML in morphologic complete remission at CC-486 initiation (42 to 84 days after alloSCT) were included. Patients received 1 of 4 CC-486 dosing schedules per 28-day cycle for up to 12 cycles. Endpoints included safety, pharmacokinetics, graft-versus-host disease (GVHD) incidence, relapse/progression rate, and survival. Of 30 patients, 7 received CC-486 once daily for 7 days per cycle (200 mg, n = 3; 300 mg, n = 4) and 23 for 14 days per cycle (150 mg, n = 4; 200 mg, n = 19 [expansion cohort]). Grades 3 to 4 adverse events were infrequent and occurred with similar frequency across regimens. Standard concomitant medications did not alter CC-486 pharmacokinetic parameters. Three patients (10%) experienced grade III acute GVHD and 9 experienced chronic GVHD. Of 28 evaluable patients, 6 (21%) relapsed or had progressive disease: 3 of 7 patients (43%) who had received 7-day dosing and 3 of 23 (13%) who had received 14-day dosing. Transplant-related mortality was 3%. At 19 months of follow-up, median overall survival was not reached. Estimated 1-year survival rates were 86% and 81% in the 7-day and 14-day dosing cohorts, respectively. CC-486 maintenance was generally well tolerated, with low rates of relapse, disease progression, and GVHD. CC-486 maintenance may permit epigenetic manipulation of the alloreactive response postallograft. Findings require confirmation in randomized trials. (ClinicalTrials.gov NCT01835587.)
- Published
- 2018
23. Efficacy and safety of extended dosing schedules of CC-486 (oral azacitidine) in patients with lower-risk myelodysplastic syndromes
- Author
-
Kyle J. MacBeth, Barry S. Skikne, Tao Shi, William Jeffery Edenfield, G. Garcia-Manero, Eric Laille, A Tefferi, Keshava Kumar, Bart L. Scott, Joel Hetzer, Steven D. Gore, Suman Kambhampati, and Christopher R. Cogle
- Subjects
Adult ,Male ,Antimetabolites, Antineoplastic ,Cancer Research ,medicine.medical_specialty ,Azacitidine ,Administration, Oral ,Pharmacology ,Lower risk ,Gastroenterology ,Drug Administration Schedule ,Oral Azacitidine ,03 medical and health sciences ,0302 clinical medicine ,Risk Factors ,Internal medicine ,medicine ,Humans ,Tissue Distribution ,Dosing ,Aged ,Neoplasm Staging ,Aged, 80 and over ,business.industry ,Myelodysplastic syndromes ,Therapeutic effect ,Hematology ,Middle Aged ,Prognosis ,medicine.disease ,Platelet transfusion ,Oncology ,International Prognostic Scoring System ,Myelodysplastic Syndromes ,030220 oncology & carcinogenesis ,Original Article ,Female ,Safety ,business ,Follow-Up Studies ,030215 immunology ,medicine.drug - Abstract
CC-486, the oral formulation of azacitidine (AZA), is an epigenetic modifier and DNA methyltransferase inhibitor in clinical development for treatment of hematologic malignancies. CC-486 administered for 7 days per 28-day treatment cycle was evaluated in a phase 1 dose-finding study. AZA has a short plasma half-life and DNA incorporation is S-phase-restricted; extending CC-486 exposure may increase the number of AZA-affected diseased target cells and maximize therapeutic effects. Patients with lower-risk myelodysplastic syndromes (MDS) received 300 mg CC-486 once daily for 14 days (n=28) or 21 days (n=27) of repeated 28-day cycles. Median patient age was 72 years (range 31-87) and 75% of patients had International Prognostic Scoring System Intermediate-1 risk MDS. Median number of CC-486 treatment cycles was 7 (range 2-24) for the 14-day dosing schedule and 6 (1-24) for the 21-day schedule. Overall response (complete or partial remission, red blood cell (RBC) or platelet transfusion independence (TI), or hematologic improvement) (International Working Group 2006) was attained by 36% of patients receiving 14-day dosing and 41% receiving 21-day dosing. RBC TI rates were similar with both dosing schedules (31% and 38%, respectively). CC-486 was generally well-tolerated. Extended dosing schedules of oral CC-486 may provide effective long-term treatment for patients with lower-risk MDS.
- Published
- 2015
- Full Text
- View/download PDF
24. Evaluation of CYP3A‐mediated drug–drug interactions with romidepsin in patients with advanced cancer
- Author
-
Hendrik-Tobias Arkenau, Liangang Liu, Manish Patel, Howard A. Burris, Charlotte Lemech, Jeffrey R. Infante, Suzanne F. Jones, and Eric Laille
- Subjects
Adult ,Male ,ketoconazole ,Cmax ,Pharmacology ,Bioequivalence ,Romidepsin ,Pharmacokinetics ,Depsipeptides ,Neoplasms ,medicine ,Cytochrome P-450 CYP3A ,Humans ,romidepsin ,Drug Interactions ,Pharmacology (medical) ,drug–drug interaction ,Aged ,Aged, 80 and over ,Volume of distribution ,Depsipeptide ,business.industry ,Cytochrome P-450 CYP3A Inducers ,Middle Aged ,Toxicity ,Cytochrome P-450 CYP3A Inhibitors ,Female ,Ketoconazole ,Rifampin ,business ,pharmacokinetics ,medicine.drug - Abstract
Two multicenter, single‐arm, single‐infusion, open‐label studies were conducted to evaluate the effect of ketoconazole (a strong CYP3A inhibitor) or rifampin (a strong CYP3A inducer) daily for 5 days on the pharmacokinetics (PK) and safety of romidepsin (8 mg/m2 intravenous 4‐hour infusion for the ketoconazole study or a 14 mg/m2 intravenous 4‐hour infusion for the rifampin study) in patients with advanced cancer. Romidepsin coadministered with ketoconazole (400 mg) or rifampin (600 mg) was not bioequivalent to romidepsin alone. With ketoconazole, the mean romidepsin AUC and Cmax were increased by approximately 25% and 10%, respectively. With rifampin, the mean romidepsin AUC and Cmax were unexpectedly increased by approximately 80% and 60%, respectively; this is likely because of inhibition of active liver uptake. For both studies, romidepsin clearance and volume of distribution were decreased, terminal half‐life was comparable, and median Tmax was similar. Overall, the safety profile of romidepsin was not altered by coadministration with ketoconazole or rifampin, except that a higher incidence and greater severity of thrombocytopenia was observed when romidepsin was given with rifampin. The use of romidepsin with rifampin and strong CYP3A inducers should be avoided. Toxicity related to romidepsin exposure should be monitored when romidepsin is given with strong CYP3A inhibitors.
- Published
- 2015
- Full Text
- View/download PDF
25. Efficacy, safety and pharmacokinetics of subcutaneous azacitidine in Chinese patients with higher risk myelodysplastic syndromes: Results from a multicenter, single-arm, open-label phase 2 study
- Author
-
C.L. Beach, Eric Laille, Qian Dong, Aining Sun, Yue-Yun Lai, Jie Jin, Zhijian Xiao, Yu Hu, Shen Zx, Xin Du, Stephen Songer, Li Yu, Xiao Li, and Ting Liu
- Subjects
0301 basic medicine ,Adult ,Male ,medicine.medical_specialty ,Antimetabolites, Antineoplastic ,Azacitidine ,Cmax ,Phases of clinical research ,Neutropenia ,Infusions, Subcutaneous ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Pharmacokinetics ,Asian People ,Internal medicine ,Medicine ,Humans ,Adverse effect ,Aged ,business.industry ,Myelodysplastic syndromes ,General Medicine ,Middle Aged ,medicine.disease ,030104 developmental biology ,Treatment Outcome ,Oncology ,030220 oncology & carcinogenesis ,Myelodysplastic Syndromes ,Cohort ,Female ,business ,medicine.drug - Abstract
Background Azacitidine safety and efficacy were established in studies of mainly Caucasian patients. Differences in drug metabolism enzymes between Caucasian and East Asian populations prevent extrapolation of drug effects between these groups. This phase 2 study evaluated azacitidine safety, efficacy and pharmacokinetics in patients with higher-risk myelodysplastic syndromes (HR-MDS) in mainland China. Methods Patients aged ≥18 years with HR-MDS were to receive subcutaneous azacitidine 75 mg/m2 /day for 7 days per 28-day cycle, for ≥6 cycles. Pharmacokinetic blood samples were collected in cycle 1 predose on days 5-7, and postdose on day 7. Pharmacokinetic outcomes are descriptively compared with those of a historical North American cohort. Results Of 72 participants, 46 (64%) completed ≥6 cycles. Response rate was 96%, driven primarily by stable disease (94%); one patient achieved complete remission. Hematologic improvement was attained by 53% of patients. Azacitidine mean plasma concentration versus time profiles were similar in shape for Chinese (n = 12) and North American (n = 45) patients. Maximum plasma concentration (Cmax ) was higher in Chinese patients; however, mean azacitidine exposure (1190 ng·h/mL) was similar to the North American cohort (1021 ng·h/mL). Most common grade 3-4 treatment-emergent adverse events (TEAEs) were thrombocytopenia (69%) and neutropenia (67%). Conclusions Azacitidine was safe and effective in Chinese patients with HR-MDS. Clinical outcomes were comparable to those for primarily Caucasian patients in the phase 3 AZA-001 study. Cmax differences between Chinese and North American patients were not associated with differences in TEAE frequency or severity. No initial azacitidine dose adjustment is required for Chinese patients with HR-MDS.
- Published
- 2017
26. Quantitative determination of azacitidine triphosphate in peripheral blood mononuclear cells using liquid chromatography coupled with high-resolution mass spectrometry
- Author
-
Eric Laille, Hilde Rosing, Michel J.X. Hillebrand, Jos H. Beijnen, Jan H.M. Schellens, Ellen J. B. Derissen, and Hans M. M. B. Otten
- Subjects
Male ,Antimetabolites, Antineoplastic ,Cytidine triphosphate ,viruses ,Clinical Biochemistry ,Azacitidine ,Pharmaceutical Science ,Mass spectrometry ,Orbitrap ,Mass Spectrometry ,Analytical Chemistry ,law.invention ,chemistry.chemical_compound ,law ,Drug Discovery ,medicine ,Humans ,heterocyclic compounds ,Spectroscopy ,Aged ,Uridine triphosphate ,Chromatography ,Cytidine ,Middle Aged ,Triple quadrupole mass spectrometer ,enzymes and coenzymes (carbohydrates) ,chemistry ,Leukocytes, Mononuclear ,Feasibility Studies ,Female ,Monoisotopic mass ,Chromatography, Liquid ,medicine.drug - Abstract
Azacitidine is a cytidine analog used in the treatment of myelodysplastic syndromes, chronic myelomonocytic leukemia and acute myeloid leukemia. The pharmacological effect of azacitidine arises after incorporation into the DNA and RNA. To this end, the drug first has to be converted into its triphosphate forms. This paper describes the development of an assay for quantitative determination of azacitidine triphosphate (aza-CTP) in peripheral blood mononuclear cells (PBMCs). To quantify aza-CTP, separation from the endogenous nucleotides cytidine triphosphate (CTP) and uridine triphosphate (UTP) is required. This was a challenge as the structures of these nucleotides are highly similar and the monoisotopic molecular masses of aza-CTP, UTP and the naturally occurring [(13)C]- and [(15)N]-isotopes of CTP differ less than 0.02 Da. Efforts to select a specific MS(2)-fragment for aza-CTP using a triple quadrupole mass spectrometer remained without success. Therefore, we investigated the feasibility to separate these highly resembling nucleotides based on accurate mass spectrometry using a linear trap quadrupole (LTQ) coupled with an Orbitrap. The LTQ-Orbitrap was able to differentiate between aza-CTP and the endogenous nucleotides UTP and [(13)C]-CTP. There was no baseline resolution between aza-CTP and [(15)N]-CTP, but the [(15)N]-CTP interference was low. For quantification, extracted ion chromatograms were obtained for the accurate m/z window of the aza-CTP product ion. The assay was able to determine aza-CTP concentrations in PBMC lysate from 40.7 to 281 nM. Assuming that an average cell suspension extracted from 16 mL blood contains 10 to 42 million PBMCs per mL, this range corresponds with 2.58/10.9-17.8/74.9 pmol aza-CTP per million PBMCs. Intra-assay accuracies were between -1.1 and 9.5% deviation and coefficient of variation values were ≤13.2%. The assay was successfully applied to quantify aza-CTP in samples from two patients treated with azacitidine. Aza-CTP concentrations up to 19.0 pmol per million PBMCs were measured. This is the first time that aza-CTP concentrations were quantified in PBMCs from patients treated with azacitidine.
- Published
- 2014
- Full Text
- View/download PDF
27. Pharmacokinetics of different formulations of oral azacitidine (CC-486) and the effect of food and modified gastric pH on pharmacokinetics in subjects with hematologic malignancies
- Author
-
Eric Laille, Bart L. Scott, Barry S. Skikne, Qian Dong, Michael R. Savona, and Thomas E. Boyd
- Subjects
Pharmacology ,medicine.drug_class ,business.industry ,Azacitidine ,Cmax ,Proton-pump inhibitor ,Capsule ,Oral Azacitidine ,Gastric ph ,Pharmacokinetics ,medicine ,Pharmacology (medical) ,business ,Omeprazole ,medicine.drug - Abstract
Parenteral azacitidine improves overall survival in higher-risk myelodysplastic syndromes. An oral azacitidine formulation would allow extended dosing schedules, potentially improving safety and/or efficacy. Two Phase 1 studies evaluated the pharmacokinetics (PK) of oral azacitidine in subjects with hematologic malignancies. Study 1 evaluated different oral formulations (immediate release tablet [IRT], enteric-coated tablet, and capsule; N = 16). Study 2 assessed the effect of food (Part 1; N = 17) and gastric pH modulation with omeprazole (Part 2; N = 14) on oral azacitidine PK. Azacitidine plasma concentration-time profiles for IRT and capsule formulations were similar, with more rapid time to maximum plasma concentration (Tmax ) than the enteric-coated tablet. Study 2 evaluated only IRT formulations of oral azacitidine. Under fed condition, Tmax was delayed ∼1.5 hours but area under the concentration-time curve (AUC∞ ) and maximum plasma concentrations (Cmax ) were comparable under fed and fasted conditions. Mean azacitidine AUC∞ and Cmax increased upon omeprazole co-administration (18.3% and 13.2%, respectively, vs. oral azacitidine alone), but not to a clinically meaningful extent. High inter-subject variability in AUC∞ and Cmax (%CV range 46.4-68.9%) was observed. Oral azacitidine is rapidly absorbed with little or no effect of food on PK parameters, and does not require dose adjustments when taking a proton-pump inhibitor.
- Published
- 2014
- Full Text
- View/download PDF
28. A Phase I Study in Patients with Solid or Hematologic Malignancies of the Dose Proportionality of Subcutaneous Azacitidine and Its Pharmacokinetics in Patients with Severe Renal Impairment
- Author
-
Eric Laille, C.L. Beach, Liangang Liu, Kevin R. Kelly, Sanjay Goel, Nashat Y. Gabrail, Alain C. Mita, and Stephen Songer
- Subjects
Adult ,Male ,Antimetabolites, Antineoplastic ,medicine.medical_specialty ,Time Factors ,Injections, Subcutaneous ,Azacitidine ,Cmax ,Urology ,Renal function ,Severity of Illness Index ,Pharmacokinetics ,Neoplasms ,Severity of illness ,Humans ,Medicine ,Pharmacology (medical) ,Renal Insufficiency ,Dosing ,Aged ,Aged, 80 and over ,Dose-Response Relationship, Drug ,business.industry ,Middle Aged ,Dose–response relationship ,Tolerability ,Hematologic Neoplasms ,Anesthesia ,Female ,business ,medicine.drug - Abstract
Study Objective To assess the dose proportionality of azacitidine pharmacokinetics (PK) after single subcutaneous (SC) doses of 25–100 mg/m2, and determine the effect of renal impairment on PK after single and multiple 75 mg/m2 SC azacitidine doses. Design Multicenter, phase I, open-label, parallel group study. Setting Community clinics and major academic centers. Patients Twenty-seven patients with solid or hematologic malignancies. Interventions Part 1 evaluated azacitidine dose proportionality in patients with normal renal function randomized to single 25, 50, 75, or 100 mg/m2 SC doses. The 75 mg/m2 dosing group received 4 additional days of SC azacitidine. In Part 2, patients with severe renal impairment (creatinine clearance
- Published
- 2013
- Full Text
- View/download PDF
29. Efficacy, safety, and pharmacokinetics of subcutaneous azacitidine in Taiwanese patients with higher-risk myelodysplastic syndromes
- Author
-
S. Songer, Anoula Galettis, Yeu Chin Chen, Liang Tsai Hsiao, Su Peng Yeh, Wen-Chien Chou, Tsai Yun Chen, Sheng Fung Lin, C.L. Beach, Qian Dong, and Eric Laille
- Subjects
0301 basic medicine ,Adult ,Male ,medicine.medical_specialty ,Antimetabolites, Antineoplastic ,Azacitidine ,Taiwan ,Phases of clinical research ,Neutropenia ,Infusions, Subcutaneous ,03 medical and health sciences ,0302 clinical medicine ,Pharmacokinetics ,Internal medicine ,medicine ,Humans ,Intensive care medicine ,Adverse effect ,Aged ,Aged, 80 and over ,Leukopenia ,business.industry ,Myelodysplastic syndromes ,General Medicine ,Middle Aged ,medicine.disease ,030104 developmental biology ,Platelet transfusion ,Treatment Outcome ,Oncology ,030220 oncology & carcinogenesis ,Myelodysplastic Syndromes ,Female ,medicine.symptom ,business ,medicine.drug - Abstract
Aim Clinical and pharmacokinetic effects of azacitidine in higher-risk myelodysplastic syndromes were established in mainly Caucasian populations. Because of inter-ethnic genotype variability of drug-metabolizing enzymes, it is important to evaluate azacitidine in populations expected to use the drug. Methods In this single-arm study, Taiwanese patients with higher-risk myelodysplastic syndromes received azacitidine 75 mg/m2/day for 7 days/28-day cycle for up to six cycles. Response-evaluable patients had baseline and cycle 6 marrow assessments. Clinical outcomes are compared descriptively with those from a phase 3 study comprising mainly Caucasian patients (N = 179). Pharmacokinetics in a subgroup of Taiwanese patients are descriptively compared with a historical control of North American patients (N = 45). Results Median age of Taiwanese patients (N = 44) was 64 years (range 36–90), and 46% had poor cytogenetics. Median number of azacitidine cycles was six (1–6). No response-evaluable patient (n = 33) achieved complete or partial remission; however, 22 patients (50%) achieved hematologic improvement, 12 of 32 patients attained RBC transfusion independence and 7 of 18 attained platelet transfusion independence. Most common grade 3–4 treatment-emergent adverse events were neutropenia (52%) and leukopenia (39%). Pharmacokinetic profiles were similar for Taiwanese (N = 12) and North American (N = 45) patients. Maximum plasma concentration was higher in Taiwanese patients; however, mean azacitidine exposure was within the range for North American patients. Conclusion These data confirm the safety and efficacy of azacitidine in Taiwanese patients with higher-risk myelodysplastic syndromes. Clinical outcomes were generally comparable with those for Caucasian patients. No meaningful differences in azacitidine pharmacokinetics were observed for Taiwanese patients, and no initial dose adjustment is necessary.
- Published
- 2016
30. Phase I Study of Oral Azacitidine in Myelodysplastic Syndromes, Chronic Myelomonocytic Leukemia, and Acute Myeloid Leukemia
- Author
-
Barry S. Skikne, Kyle J. MacBeth, Elias J. Jabbour, Renee Ward, Tao Shi, Heidi Giordano, Steven D. Gore, Guillermo Garcia-Manero, Sarah Sakoian, Hagop M. Kantarjian, Eric Laille, and Christopher R. Cogle
- Subjects
Adult ,Male ,Antimetabolites, Antineoplastic ,Cancer Research ,medicine.medical_specialty ,Neutropenia ,Maximum Tolerated Dose ,Antimetabolites ,Gastrointestinal Diseases ,Azacitidine ,Administration, Oral ,Biological Availability ,Chronic myelomonocytic leukemia ,Pharmacology ,Gastroenterology ,Oral Azacitidine ,Myelogenous ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,Internal medicine ,Original Reports ,medicine ,Humans ,Adverse effect ,Fatigue ,Aged ,Aged, 80 and over ,business.industry ,Myelodysplastic syndromes ,Myeloid leukemia ,Leukemia, Myelomonocytic, Chronic ,DNA Methylation ,Middle Aged ,medicine.disease ,Leukemia ,Oncology ,Myelodysplastic Syndromes ,Female ,business ,medicine.drug - Abstract
Purpose To determine the maximum-tolerated dose (MTD), safety, pharmacokinetic and pharmacodynamic profiles, and clinical activity of an oral formulation of azacitidine in patients with myelodysplastic syndromes (MDSs), chronic myelomonocytic leukemia (CMML), or acute myeloid leukemia (AML). Patients and Methods Patients received 1 cycle of subcutaneous (SC) azacitidine (75 mg/m2) on the first 7 days of cycle 1, followed by oral azacitidine daily (120 to 600 mg) on the first 7 days of each additional 28-day cycle. Pharmacokinetic and pharmacodynamic profiles were evaluated during cycles 1 and 2. Adverse events and hematologic responses were recorded. Cross-over to SC azacitidine was permitted for nonresponders who received ≥ 6 cycles of oral azacitidine. Results Overall, 41 patients received SC and oral azacitidine (MDSs, n = 29; CMML, n = 4; AML, n = 8). Dose-limiting toxicity (grade 3/4 diarrhea) occurred at the 600-mg dose and MTD was 480 mg. Most common grade 3/4 adverse events were diarrhea (12.2%), nausea (7.3%), vomiting (7.3%), febrile neutropenia (19.5%), and fatigue (9.8%). Azacitidine exposure increased with escalating oral doses. Mean relative oral bioavailability ranged from 6.3% to 20%. Oral and SC azacitidine decreased DNA methylation in blood, with maximum effect at day 15 of each cycle. Hematologic responses occurred in patients with MDSs and CMML. Overall response rate (ie, complete remission, hematologic improvement, or RBC or platelet transfusion independence) was 35% in previously treated patients and 73% in previously untreated patients. Conclusion Oral azacitidine was bioavailable and demonstrated biologic and clinical activity in patients with MDSs and CMML.
- Published
- 2011
- Full Text
- View/download PDF
31. In vitro assessment of cytochrome P450 inhibition and induction potential of azacitidine
- Author
-
Gondi Kumar, Eric Laille, Yong Chen, Sekhar Surapaneni, and Lisa Liu
- Subjects
Drug–drug interactions ,Antimetabolites, Antineoplastic ,Cancer Research ,Short Communication ,CYP1B1 ,Azacitidine ,Cytochrome P450 ,Pharmacology ,Toxicology ,Inhibitory postsynaptic potential ,Isozyme ,Cytochrome P-450 Enzyme System ,CYP induction ,medicine ,Cytochrome P-450 Enzyme Inhibitors ,Humans ,Pharmacology (medical) ,Enzyme Inhibitors ,Cells, Cultured ,biology ,Chemistry ,In vitro ,Oncology ,Enzyme Induction ,Hepatocytes ,Microsomes, Liver ,Microsome ,biology.protein ,CYP inhibition ,medicine.drug - Abstract
Purpose To assess the potential inhibitory and inductive effects of azacitidine on cytochrome P450 isozymes in vitro. Methods The inhibitory effects of azacitidine on various CYP isozymes were determined in human liver microsomes. In addition, the ability of azacitidine to induce CYP enzymes in cultured human hepatocytes was evaluated. Results Azacitidine did not inhibit CYP2B6-, CYP2C8-, CYP2C9-, CYP2C19-, CYP2D6-, and CYP3A4-mediated activities in human liver microsomes up to a concentration of 100 μM, while weak inhibition (
- Published
- 2010
- Full Text
- View/download PDF
32. Phase 1 study of the oral isotype specific histone deacetylase inhibitor MGCD0103 in leukemia
- Author
-
Wilson H. Miller, Marja Dubay, Tracy Patterson, Zeev Estrov, Zuomei Li, Caroline Rousseau, Eric Laille, Gregory K. Reid, Guillermo Garcia-Manero, Sarit Assouline, Robert E. Martell, Ann Kalita, Willie Newsome, Claire Bonfils, H. Yang, Hagop M. Kantarjian, Jorge E. Cortes, Mark D. Minden, and J. M. Besterman
- Subjects
Adult ,Male ,medicine.medical_specialty ,Myeloid ,Maximum Tolerated Dose ,Clinical Trials and Observations ,medicine.drug_class ,Immunology ,Administration, Oral ,Biochemistry ,Gastroenterology ,Histone Deacetylases ,Histones ,Myelogenous ,Pharmacokinetics ,Internal medicine ,medicine ,Humans ,Cells, Cultured ,Aged ,Aged, 80 and over ,Leukemia ,Hematology ,Dose-Response Relationship, Drug ,business.industry ,Myelodysplastic syndromes ,Histone deacetylase inhibitor ,Cell Biology ,Middle Aged ,medicine.disease ,Histone Deacetylase Inhibitors ,Leukemia, Myeloid, Acute ,Pyrimidines ,medicine.anatomical_structure ,Benzamides ,Female ,Bone marrow ,business - Abstract
MGCD0103 is an isotype-selective inhibitor of histone deacetylases (HDACs) targeted to isoforms 1, 2, 3, and 11. In a phase 1 study in patients with leukemia or myelodysplastic syndromes (MDS), MGCD0103 was administered orally 3 times weekly without interruption. Twenty-nine patients with a median age of 62 years (range, 32-84 years) were enrolled at planned dose levels (20, 40, and 80 mg/m2). The majority of patients (76%) had acute myelogenous leukemia (AML). In all, 24 (83%) of 29 patients had received 1 or more prior chemotherapies (range, 0-5), and 18 (62%) of 29 patients had abnormal cytogenetics. The maximum tolerated dose was determined to be 60 mg/m2, with dose-limiting toxicities (DLTs) of fatigue, nausea, vomiting, and diarrhea observed at higher doses. Three patients achieved a complete bone marrow response (blasts ≤ 5%). Pharmacokinetic analyses indicated absorption of MGCD0103 within 1 hour and an elimination half-life in plasma of 9 (± 2) hours. Exposure to MGCD0103 was proportional to dose up to 60 mg/m2. Analysis of peripheral white cells demonstrated induction of histone acetylation and dose-dependent inhibition of HDAC enzyme activity. In summary, MGCD0103 was safe and had antileukemia activity that was mechanism based in patients with advanced leukemia.
- Published
- 2008
- Full Text
- View/download PDF
33. CC-486 (Oral Azacitidine) Maintenance Therapy Is Well Tolerated after Allogeneic Hematopoietic Stem Cell Transplantation (AlloHSCT) in Patients with Myelodysplastic Syndromes (MDS) or Acute Myeloid Leukemia (AML)
- Author
-
Betul Oran, Joel Hetzer, Alessandra Tosolini, Esperanza B. Papadopoulos, Marcos de Lima, Sergio Giralt, Basem M. William, Eric Laille, Bart L. Scott, Barry S. Skikne, Richard E. Champlin, and Charles Craddock
- Subjects
Oncology ,Transplantation ,medicine.medical_specialty ,business.industry ,medicine.medical_treatment ,Myelodysplastic syndromes ,Myeloid leukemia ,Hematology ,Hematopoietic stem cell transplantation ,medicine.disease ,Oral Azacitidine ,03 medical and health sciences ,0302 clinical medicine ,Maintenance therapy ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,In patient ,business ,030215 immunology - Published
- 2016
- Full Text
- View/download PDF
34. Plasma concentrations of praziquantel after oral administration of single and multiple doses in loggerhead sea turtles ( Caretta caretta )
- Author
-
Lara K. Maxwell, Glenn R. Harman, Elliott R. Jacobson, and Eric Laille
- Subjects
Male ,medicine.medical_specialty ,Dose ,Cmax ,Administration, Oral ,Physiology ,Biology ,Multiple dosing ,Drug Administration Schedule ,Praziquantel ,Pharmacokinetics ,Oral administration ,parasitic diseases ,medicine ,Animals ,Dosing ,Skin ,Anthelmintics ,Dose-Response Relationship, Drug ,General Veterinary ,General Medicine ,Turtles ,Surgery ,Area Under Curve ,Plasma concentration ,Female ,medicine.drug - Abstract
Objective—To determine the pharmacokinetics of praziquantel following single and multiple oral dosing in loggerhead sea turtles. Animals—12 healthy juvenile loggerhead sea turtles. Procedure—Praziquantel was administered orally as a single dose (25 and 50 mg/kg) to 2 groups of turtles; a multiple-dose study was then performed in which 6 turtles received 3 doses of praziquantel (25 mg/kg, PO) at 3-hour intervals. Blood samples were collected from all turtles before and at intervals after drug administration for assessment of plasma praziquantel concentrations. Pharmacokinetic analyses included maximum observed plasma concentration (Cmax), time to maximum concentration (Tmax), area under the plasma praziquantel concentration-time curve, and mean residence time (MRTt). Results—Large interanimal variability in plasma praziquantel concentrations was observed for all dosages. One turtle that received 50 mg of praziquantel/kg developed skin lesions within 48 hours of administration. After administration of 25 or 50 mg of praziquantel/ kg, mean plasma concentrations were below the limit of quantification after 24 hours. In the multiple- dose group of turtles, mean plasma concentration was 90 ng/mL at the last sampling time-point (48 hours after the first of 3 doses). In the single-dose study, mean Cmax and Tmax with dose were not significantly different between doses. After administration of multiple doses of praziquantel, only MRTt was significantly increased, compared with values after administration of a single 25-mg dose. Conclusions and Clinical Relevance—Oral administration of 25 mg of praziquantel/kg 3 times at 3-hour intervals may be appropriate for treatment of loggerhead sea turtles with spirorchidiasis. (Am J Vet Res 2003;64:304–309)
- Published
- 2003
- Full Text
- View/download PDF
35. A phase Ia study of CC-90003, a selective extracellular signal-regulated kinase (ERK) inhibitor, in patients with relapsed or refractory BRAF or RAS-mutant tumors
- Author
-
Patricia LoRusso, Johanna C. Bendell, Xiaoling Wu, Eric Laille, Ida Aronchik, Gordon L. Bray, Nanette H Hock, Monica M. Mita, Ellen Filvaroff, Edward S. Kim, and Grant A. McArthur
- Subjects
0301 basic medicine ,Neuroblastoma RAS viral oncogene homolog ,MAPK/ERK pathway ,Cancer Research ,medicine.medical_specialty ,Hematology ,medicine.diagnostic_test ,business.industry ,Mutant ,Cardiac echo ,medicine.disease_cause ,Gastroenterology ,Peripheral blood mononuclear cell ,Transaminase ,03 medical and health sciences ,030104 developmental biology ,Oncology ,Internal medicine ,medicine ,KRAS ,business - Abstract
2577 Background: CC-90003 is an irreversible inhibitor of ERK 1/2 with potent anti-proliferative activity in KRAS and BRAF mutant tumor models. We conducted a first-in-human study of CC-90003 in patients with RAS or BRAF mutant tumors. Methods: Patients received escalating doses of oral CC-90003 on a 21/28 day cycle. Standard safety (adverse events, chemistry/hematology, physical findings, ECGs and cardiac ECHO/MUGA scans) and PK parameters were assessed. Response was assessed per RECIST 1.1. A proprietary ELISA-based assay measured ERK levels unbound to CC-90003 in peripheral blood mononuclear cells. Results: Nineteen patients (median age: 60 yrs) harboring KRAS (n = 15), NRAS (n = 1), or BRAF (n = 3) mutant tumors received CC-90003 doses from 20 to 160 mg /day. The MTD was 120 mg based on the occurrence of Grade 3 transaminase elevations (n = 2) and hypertension (n = 1) observed at 160 mg (the NTD). Patients completed a median of 2 cycles (range: 1 to 5). AEs (mostly Grade 1 or 2) reported in ≥ 3 patients included constitutional (asthenia, fatigue), gastrointestinal (anorexia, nausea/vomiting, diarrhea), hepatic (transaminase elevations) and neurologic (dizziness, gait disturbance, paresthesias) toxicities. Grade 1-3 neurotoxicity was observed primarily at doses from 80 to 160 mg/day and resolved with dose reduction/interruption. PK parameters were highly variable, with AUC and Cmax increasing overall, with increasing dose. CC-90003 accumulation was observed after multiple doses. There were no objective responses. Levels of free ERK were reduced by ≥80% compared to baseline by C1D8 at doses ≥ 80 mg/day. Conclusions: ERK inhibition may be an attractive target for the management of mutant RAS or BRAF-driven tumors, however proof-of-concept demonstration for CC-90003 was limited by a lack of objective responses, an unfavorable PK profile and unanticipated neurotoxicity. Clinical trial information: NCT02313012.
- Published
- 2017
- Full Text
- View/download PDF
36. Pharmacokinetics of different formulations of oral azacitidine (CC-486) and the effect of food and modified gastric pH on pharmacokinetics in subjects with hematologic malignancies
- Author
-
Eric, Laille, Michael R, Savona, Bart L, Scott, Thomas E, Boyd, Qian, Dong, and Barry, Skikne
- Subjects
Aged, 80 and over ,Male ,Antimetabolites, Antineoplastic ,Chemistry, Pharmaceutical ,Stomach ,Administration, Oral ,Capsules ,Hydrogen-Ion Concentration ,Middle Aged ,Food-Drug Interactions ,Delayed-Action Preparations ,Hematologic Neoplasms ,Azacitidine ,Humans ,Female ,Aged ,Tablets - Abstract
Parenteral azacitidine improves overall survival in higher-risk myelodysplastic syndromes. An oral azacitidine formulation would allow extended dosing schedules, potentially improving safety and/or efficacy. Two Phase 1 studies evaluated the pharmacokinetics (PK) of oral azacitidine in subjects with hematologic malignancies. Study 1 evaluated different oral formulations (immediate release tablet [IRT], enteric-coated tablet, and capsule; N = 16). Study 2 assessed the effect of food (Part 1; N = 17) and gastric pH modulation with omeprazole (Part 2; N = 14) on oral azacitidine PK. Azacitidine plasma concentration-time profiles for IRT and capsule formulations were similar, with more rapid time to maximum plasma concentration (Tmax ) than the enteric-coated tablet. Study 2 evaluated only IRT formulations of oral azacitidine. Under fed condition, Tmax was delayed ∼1.5 hours but area under the concentration-time curve (AUC∞ ) and maximum plasma concentrations (Cmax ) were comparable under fed and fasted conditions. Mean azacitidine AUC∞ and Cmax increased upon omeprazole co-administration (18.3% and 13.2%, respectively, vs. oral azacitidine alone), but not to a clinically meaningful extent. High inter-subject variability in AUC∞ and Cmax (%CV range 46.4-68.9%) was observed. Oral azacitidine is rapidly absorbed with little or no effect of food on PK parameters, and does not require dose adjustments when taking a proton-pump inhibitor.
- Published
- 2013
37. Phase I/II Multicenter Study of Romidepsin in Japanese Patients with Relapsed or Refractory Peripheral T-Cell Lymphoma
- Author
-
Kiyoshi Ando, Yoichi Tatsumi, Hiromi Tamakoshi, Sanae Sakurai, Takashi Ishida, Masafumi Taniwaki, Kiyohiko Hatake, Kensei Tobinai, Michinori Ogura, Eric Laille, Tomoko Ohtsu, Dai Maruyama, Kunihiro Tsukasaki, Takayuki Ikezoe, Toru Kiguchi, Kenichi Ishizawa, Toshiki Uchida, Naoki Kobayashi, Tokihiro Ro, and Koji Kato
- Subjects
education.field_of_study ,medicine.medical_specialty ,Leukopenia ,business.industry ,Immunology ,Population ,Aggressive lymphoma ,Cell Biology ,Hematology ,Neutropenia ,medicine.disease ,Biochemistry ,Gastroenterology ,Romidepsin ,Dysgeusia ,Internal medicine ,medicine ,Clinical endpoint ,medicine.symptom ,education ,business ,Progressive disease ,medicine.drug - Abstract
Introduction: Peripheral T-cell lymphoma (PTCL) is an aggressive lymphoma associated with poor prognosis. There is no consensus on standard therapy, and options are limited for patients with relapsed/refractory (R/R) disease. Romidepsin, a potent histone deacetylase inhibitor, has US FDA approval for patients with >=1 prior treatment for PTCL based on 25%-38% overall response rates (ORR) and durable responses (Piekarz et al, Blood. 2011;117:5827; Coiffier et al, J Clin Oncol. 2012;30:631). Here we report results from the phase I/II, multicenter, open-label study of romidepsin in Japanese patients with R/R PTCL or cutaneous T-cell lymphoma (CTCL) (TCL-001; NCT01456039). Methods: Patients aged >=20 years with R/R PTCL or CTCL received romidepsin via a 4-hour IV infusion on days 1, 8, and 15 of each 28-day cycle until progressive disease or unacceptable toxicity. The phase I portion of the study used a 3+3 design to identify any dose-limiting toxicity (DLT; phase I primary endpoint) with romidepsin 9 mg/m2 (cohort 1) and 14 mg/m2 (cohort 2). The phase II dose was based on the highest dose where =1 day. Assessments of the intent-to-treat (ITT) population included all patients receiving at least 1 dose of romidepsin. Results: The ITT population comprised 48 patients with PTCL and 2 with CTCL (1 each in the 9 and 14 mg/m2 cohorts). The common PTCL subtypes were angioimmunoblastic T-cell lymphoma (AITL, n=21, 44%), PTCL not otherwise specified (PTCL-NOS, n=20, 42%), and anaplastic large-cell lymphoma (ALCL ALK-1 negative, n=3, 6%). Most patients had a favorable ECOG performance status (86% 0-1) and 72% were >=65 years of age. Patients had received a median of 2 prior therapies (range, 1-9). Of 9 patients assessable in phase I (n=3 at 9 mg/m2 and n=6 at 14 mg/m2), none experienced a DLT. The recommended phase II dose was 14 mg/m2 in 40 subsequently treated patients. The overall population received a median of 10 doses (range, 1-135) for a median treatment duration of 12.9 weeks (range, 0.1-184.3; 8 patients were treated for >=36 weeks). The common all-grade adverse events (AEs) were thrombocytopenia (n=49, 98%), lymphopenia (n=44, 88%), leukopenia (n=42, 84%), neutropenia (n=40, 80%), pyrexia (n=33, 66%), dysgeusia (n=31, 62%), decreased appetite (n=28, 56%), and nausea (n=27, 54%). The common grade >=3 AEs were lymphopenia (n=37, 74%), neutropenia (n=27, 54%), leukopenia (n=23, 46%), and thrombocytopenia (n=19, 38%). Of the 39 patients whose CD4+ T-cell counts were monitored, while decreased CD4+ T-cell counts ( Among the 40 patients with PTCL (phase II), the ORR was 43% (95% CI, 27%-58%), including 10 patients (25%) with complete response (CR) or CR unconfirmed (CRu) and 7 (17%) with partial responses (PR) (Table 1). The obtained ORR was significantly higher compared with the threshold ORR of 10% (P Conclusions: Results from this phase I/II study identified a tolerable dose of romidepsin and indicated that romidepsin has an acceptable toxicity profile with clinically meaningful, efficacy in Japanese patients with R/R PTCL. The efficacy and safety data were comparable with results from other romidepsin phase II studies. Disclosures Ogura: Celltrion, Inc.: Consultancy, Honoraria; SymBio Pharmaceuticals: Consultancy, Honoraria. Maruyama:Janssen: Honoraria; Takeda: Honoraria. Tobinai:Chugai Pharma: Research Funding; Daiichi Sankyo Co., Ltd.: Consultancy; Mundipharma KK: Honoraria, Research Funding; SERVIER: Research Funding; HUYA Bioscience: Honoraria; Celgene: Research Funding; Kyowa Hakko Kirin: Research Funding; GlaxoSmithKline: Research Funding; Janssen Pharmaceuticals: Honoraria, Research Funding; Abbvie: Research Funding; Takeda: Honoraria, Research Funding; Zenyaku Kogyo: Honoraria; Eisai: Honoraria, Research Funding; Ono Pharmaceutical: Research Funding. Uchida:SymBio Pharmaceuticals: Research Funding. Hatake:Chugai: Research Funding; Meiji-Seika: Consultancy; Otsuka: Consultancy; Kyowa Kirin: Honoraria, Research Funding. Tsukasaki:Daiichi Sankyo Co., Ltd.: Consultancy; Takeda: Research Funding. Ishida:Celgene KK: Research Funding; Kyowa Hakko Kirin, Co., Ltd.: Honoraria, Research Funding; Bayer Pharma AG: Research Funding. Ishizawa:SymBio Pharmaceuticals: Research Funding. Laille:Celgene: Employment, Equity Ownership. Ro:Celgene: Employment, Equity Ownership. Tamakoshi:Celgene: Employment, Equity Ownership. Sakurai:Celgene: Employment. Ohtsu:Celgene: Employment, Equity Ownership.
- Published
- 2016
- Full Text
- View/download PDF
38. Phase I study of MGCD0103 given as a three-times-per-week oral dose in patients with advanced solid tumors
- Author
-
Eric Laille, J. M. Besterman, M. Maclean, Roberto Pili, Michael A. Carducci, Robert E. Martell, Ann Kalita, Wells A. Messersmith, Eric X. Chen, Zuomei Li, Shirley Brown, Rana Sullivan, Serina King, Gregory K. Reid, Lillian L. Siu, and Ignacio Duran
- Subjects
Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Nausea ,Administration, Oral ,Antineoplastic Agents ,Anorexia ,Gastroenterology ,Drug Administration Schedule ,Article ,Histones ,Pharmacokinetics ,Internal medicine ,Neoplasms ,medicine ,Leukocytes ,Humans ,Enzyme Inhibitors ,Aged ,Dose-Response Relationship, Drug ,business.industry ,Cancer ,Acetylation ,Middle Aged ,medicine.disease ,Surgery ,Histone Deacetylase Inhibitors ,Dose–response relationship ,Pyrimidines ,Oncology ,Tolerability ,Pharmacodynamics ,Benzamides ,Vomiting ,Patient Compliance ,Female ,medicine.symptom ,business - Abstract
Purpose MGCD0103 is a novel isotype-selective inhibitor of human histone deaceylases (HDACs) with the potential to regulate aberrant gene expression and restore normal growth control in malignancies. Patients and Methods A phase I trial of MGCD0103, given as a three-times-per-week oral dose for 2 of every 3 weeks, was performed in patients with advanced solid tumors. Primary end points were safety, tolerability, pharmacokinetics (PK), pharmacodynamic (PD) assessments of HDAC activity, and histone acetylation status in peripheral WBCs. Results Six dose levels ranging from 12.5 to 56 mg/m2/d were evaluated in 38 patients over 99 cycles (median, 2; range, 1 to 11). The recommended phase II dose was 45 mg/m2/d. Dose-limiting toxicities consisting of fatigue, nausea, vomiting, anorexia, and dehydration were observed in three (27%) of 11 and two (67%) of three patients treated at the 45 and 56 mg/m2/d dose levels, respectively. Disease stabilization for four or more cycles was observed in five (16%) of 32 patients assessable for efficacy. PK analyses demonstrated interpatient variability which was improved by coadministration with low pH beverages. Elimination half-life ranged from 6.7 to 12.2 hours, and no accumulation was observed with repeated dosing. PD evaluations confirmed inhibition of HDAC activity and induction of acetylation of H3 histones in peripheral WBCs from patients by MGCD0103. Conclusion At doses evaluated, MGCD0103 appears tolerable and exhibits favorable PK and PD profiles with evidence of target inhibition in surrogate tissues.
- Published
- 2008
39. CC-486 in Patients with Myelodysplastic Syndromes (MDS), Acute Myeloid Leukemia (AML), or Chronic Myelomonocytic Leukemia (CMML): Safety, Tolerability, and Response
- Author
-
Edwin C. Kingsley, Barry S. Skikne, Paul Conkling, Eric Laille, Carlos Becerra, Robert M. Rifkin, Kathryn S. Kolibaba, Michael R. Savona, John C. Morris, Stacey M. Ukrainskyj, Qian Dong, and Amy Kellerman
- Subjects
medicine.medical_specialty ,education.field_of_study ,business.industry ,Myelodysplastic syndromes ,MedDRA ,Immunology ,Population ,Chronic myelomonocytic leukemia ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Regimen ,Platelet transfusion ,Tolerability ,Internal medicine ,medicine ,Intensive care medicine ,education ,business ,Febrile neutropenia - Abstract
Background: CC-486 is an oral formulation of the epigenetic modifier, azacitidine, in clinical development for treatment (Tx) of hematological cancers. Part 1 of this phase I, multicenter study showed no effect on pharmacokinetics (PK) of CC-486 when taken with food or with a proton-pump inhibitor (Laille, 2014). A prospective extension phase of this study is ongoing to assess the safety and efficacy of CC-486. Objectives: To assess safety, tolerability, and hematological response associated with an extended CC-486 dosing schedule in patients (pts) with MDS, AML, or CMML. Methods: Eligible pts were age ≥18 years, had confirmed WHO-defined MDS, CMML, or AML based on bone marrow (BM) aspirate and biopsy, and an ECOG performance status score 0-2. In the PK phase, pts received 2 to 3 single CC-486 doses and in the extension phase, pts received CC-486 300 mg QD for the first 21 days of repeated 28-day cycles. Tx-emergent adverse events (TEAEs) were coded by MedDRA v15.0 and graded using NCI-CTCAE v4.0. In addition to TEAEs for all pts, because PK-phase exposure was quite limited, TEAEs of interest are reported for the subset of pts who entered the extension phase (21-day dosing). Responses (complete or partial remission [CR, PR], CR with incomplete blood count recovery [CRi], marrow CR [mCR], hematologic improvement [HI], and transfusion independence [TI]) were defined by IWG 2006 (MDS) or IWG 2003 (AML) criteria. Baseline transfusion dependence (TD) was defined as ≥4 red blood cell (RBC) units or ≥2 platelet transfusions, in the 56 days before first dose. TI was defined as no transfusion for 56 consecutive days. The safety-evaluable cohort included all pts who received ≥1 CC-486 dose. Efficacy-evaluable pts had ≥1 post-baseline efficacy measurement during the extension phase as of data cut-off (June 6, 2014). Efficacy results are reported for the combined MDS and CMML pt population and separately for pts with AML. Results: A total of 32 pts (MDS n=19 [59%], CMML n=4 [13%], and AML n=9 [28%]) received ≥1 CC-486 dose and are evaluated for safety. Median (range) age of all pts was 72 (53-93) years, with 44% of pts age ≥75 years. Most pts were male (69%) and all were Caucasian. Median number of CC-486 Tx cycles for all pts was 4 (range 1-18), and within the MDS, CMML, and AML groups was 5 (1-18), 4 (2-17), and 1 (1-9), respectively. The mean ±SD Tx cycle length was 30 ±9.0 days overall (by group: 33 ±8.3 days in MDS, 25 ±5.4 in CMML, and 27 ±10.7 in AML pts). The most frequent types of TEAE were gastrointestinal (GI) and hematological (Table 1). CC-486 dose was adjusted due to GI TEAEs for 5 pts (16%), and due to hematological TEAEs for 8 pts (25%). Of all 32 pts, 30 pts (94%) entered the 21-day dosing extension phase. Of these pts, GI TEAEs (grade ≥2) were reported for 23 pts (77%). Of these, only 4 grade 3 (vomiting and diarrhea, 2 each) and no grade 4 TEAEs were observed. Hematological TEAEs (grade ≥3) were reported for 19 pts (63%). Neutropenic grade ≥2 TEAEs occurred in 12 (40%) of 30 pts, of which 29% were grade 2, 38% grade 3, and 32% grade 4. Febrile neutropenia occurred in 3 pts and grade 4 thrombocytopenia in 3. Most TEAEs occurred during the first 2 Tx cycles: GI, 73% and hematological, 55%. The efficacy-evaluable population at data cut-off included 27 pts (84%): 20 pts with MDS or CMML, and 7 pts with AML (Table 2). One pt with MDS achieved CR, 1 pt with AML achieved CRi, and 1 pt with AML achieved PR (pt was to proceed to transplant). Of HI-evaluable pts in the MDS+CMML and AML groups, respectively, 6/18 (33%) and 2/7 (29%) attained an HI response. Few pts were TD at baseline. Of pts who were RBC TD at baseline, 1/5 pts (20%) in the MDS+CMML group and 1/5 pts (20%) in the AML group attained RBC TI. Of 3 pts who were platelet TD at baseline, 1 pt with AML attained platelet TI. Conclusions: CC-486 can provide the flexibility to adjust dose or dosing regimen to improve tolerability or efficacy as needed. The safety and tolerability of CC-486 300 mg QD taken for the first 21 days of repeated 28-day cycles were consistent with the known safety profile of injectable azacitidine in these pts with MDS, CMML, and AML. Grade 3-4 gastrointestinal TEAEs were infrequent ( Disclosures Savona: Karyopharm: Consultancy, Equity Ownership; Celgene: Consultancy; Gilead: Consultancy; Incyte: Consultancy. Kolibaba:Celgene: Research Funding. Conkling:Celgene: Research Funding. Rifkin:Celgene: Scientific Advisory Board Other; Millennium/Takeda: Scientific Advisory Board, Scientific Advisory Board Other; Onyx/Amgen: Scientific Advisory Board, Scientific Advisory Board Other. Laille:Celgene: Employment, Equity Ownership. Kellerman:Celgene: Employment. Ukrainskyj:Celgene: Employment, Equity Ownership. Dong:Celgene: Employment. Skikne:Celgene: Employment, Equity Ownership.
- Published
- 2014
- Full Text
- View/download PDF
40. CC-486 (Oral Azacitidine) Following Allogeneic Hematopoietic Stem Cell Transplantation (AlloHSCT) in Patients with Myelodysplastic Syndromes (MDS) or Acute Myeloid Leukemia (AML)
- Author
-
Barry S. Skikne, Esperanza B. Papadopoulos, Eric Laille, Richard E. Champlin, Basem M. William, Betul Oran, Joel Hetzer, Xiwei Wang, Charles Craddock, Bart L. Scott, Marcos de Lima, and Sergio Giralt
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Myelodysplastic syndromes ,medicine.medical_treatment ,Immunology ,Azacitidine ,Cell Biology ,Hematology ,Hematopoietic stem cell transplantation ,medicine.disease ,Biochemistry ,Rash ,Oral Azacitidine ,Surgery ,Graft-versus-host disease ,Internal medicine ,Concomitant ,Medicine ,medicine.symptom ,business ,Febrile neutropenia ,medicine.drug - Abstract
Background: Disease relapse and graft vs host disease (GvHD) following alloHSCT are major causes of treatment (Tx) failure in patients (pts) with MDS and AML (Paietta, 2012). Studies of parenteral azacitidine, an epigenetic modifier, have shown efficacy in preventing post-transplant relapse in MDS and AML pts, and possibly reducing GvHD severity (Platzbecker, 2012; de Lima, 2010). Azacitidine maintenance after alloHSCT may enhance graft vs leukemia (GvL) effects and reduce GvHD by expansion of regulatory T cells (Goodyear, 2010, 2012). CC-486 is a novel oral formulation of azacitidine which, as well as potentially allowing easier administration over extended schedules, could increase the duration of drug exposure to residual malignant cells. We now report preliminary results from a prospective phase I/II dose-finding study of CC-486 as maintenance Tx after alloHSCT in pts with MDS or AML. Objective: To evaluate the maximum tolerated dose (MTD), dose-limiting toxicities (DLT), pharmacokinetics (PK), and safety outcomes of CC-486 following alloHSCT in pts with MDS or AML. Methods: Pts with WHO-defined MDS or AML who have undergone alloHSCT with myeloablative (MAC) or reduced-intensity conditioning (RIC) regimens and who had sibling or unrelated donors with ≤1 antigen mismatch at the HLA-A, -B, -C, -DRB1 or -DQB1 locus, are enrolled. CC-486 Tx is initiated between days (d) 42 and 84 after alloHSCT. Pts receive prophylactic Tx for GvHD and infections per institutional standard. A standard 3x3 MTD design is being used to evaluate 4 dosing schedules: CC-486 200 mg or 300 mg QD, administered for 7 d (Cohorts 1 and 2) or 14 d (Cohorts 3 and 4) of repeated 28d Tx cycles. MTD determination is based on DLT that occur during the first 2 Tx cycles (pts are not evaluable if unable to complete 2 Tx cycles for reasons other than a DLT). Adverse events (AEs) are graded using NCI-CTCAE v4.0. Pts are followed for safety, incidence of acute and chronic GvHD, and relapse. PK of azacitidine after CC-486 administration, alone or with standard concomitant medications, are evaluated on d 1 of CC-486 Tx cycles 1 and 2. Results: At data cut-off (7/17/2014), outcome data were available for 7 pts enrolled in the first 2 cohorts (Table): 1 pt had IPSS Int-2 MDS and 6 had AML with high-risk features. Stem cell source was from bone marrow (n=2) or peripheral blood (n=5) from unrelated (n=5) or sibling (n=2) donors. Two pts had 1 antigen mismatch and 5 had a full match.Conditioning included MAC (n=3) and RIC (n=4) regimens. One pt in Cohort 2 was not evaluable for DLT or PK assessments due to early discontinuation (AML relapse) during the first CC-486 Tx cycle. Of the 6 evaluable pts, 4 completed ≥6 CC-486 Tx cycles and 2 ongoing pts had not reached 6 cycles on-study at data cut-off (Table). One pt who had GvHD before CC-486 Tx subsequently withdrew from the study after developing febrile neutropenia and diarrhea related to GvHD of the bowel; 1 pt withdrew for personal reasons; and 1 pt discontinued after 6 Tx cycles due to relapse. At data cut-off, 3 pts remained on-study, all with continued CR (Table). No pt in Cohort 2 developed GvHD. For all pts, most AEs were grade 1-2. Grade 3-4 AEs were reported for 2 pts: both had nausea and vomiting, which were controlled with antiemetic agents upon onset; 1 also had device-related infection and dyspnea, and 1 also had febrile neutropenia and rash. No DLT emerged with these CC-486 regimens and no pt experienced secondary graft failure. Overall azacitidine plasma concentration profiles (Figure) and PK parameters were similar following CC-486 given alone or with concomitant medications, and were within range of values observed following similar CC-486 doses in pts with MDS, CMML, and AML (Garcia-Manero, 2011). The AUC range for oral CC-486 is ~10% of that seen with subcutaneous azacitidine administered at 75 mg/m2 (Garcia-Manero, 2011). Conclusion: Preliminary data from this analysis suggest that CC-486 administered at doses of 200 or 300 mg QD for 7 days every 4 weeks is safe and well tolerated in the post-transplant setting. Overall, azacitidine plasma concentration profiles and PK parameters were not affected by use of concomitant medications. No DLT and a low rate of GvHD support the ongoing evaluation of 14d extended CC-486 dosing regimens in this ongoing study, as a preliminary to a prospective, randomized trial of this new agent post-transplant. Figure 1 Figure 1. Figure 2 Figure 2. Disclosures de Lima: Celgene: Consultancy. Champlin:Celgene: Consultancy. Giralt:Celgene: Consultancy, Honoraria, Research Funding, Speakers Bureau. Scott:Celgene: Consultancy, Honoraria, Research Funding. Hetzer:Celgene: Employment, Equity Ownership. Wang:Celgene: Employment, Equity Ownership. Laille:Celgene: Employment, Equity Ownership. Skikne:Celgene: Employment, Equity Ownership. Craddock:Celgene: Honoraria, Research Funding.
- Published
- 2014
- Full Text
- View/download PDF
41. Abstract B217: A Phase I study of CC-486 (oral azacitidine) to evaluate the pharmacokinetics (PK) and pharmacodynamics (PD) of azacitidine administered alone and in combination with carboplatin or ABI-007 (NAB-paclitaxel) in subjects with solid tumors
- Author
-
Michael B. Gonzalez, Anne Blackwood-Chirchir, Eric Laille, Aaron N. Nguyen, Johanna C. Bendell, Pamela N. Munster, Jorge DiMartino, Drew W. Rasco, Patricia LoRusso, Daniel D. Von Hoff, William Jeffery Edenfield, Gengxi Chen, and Ramesh K. Ramanathan
- Subjects
Volume of distribution ,Cancer Research ,business.industry ,Azacitidine ,Cmax ,Pharmacology ,Carboplatin ,Oral Azacitidine ,chemistry.chemical_compound ,Oncology ,Pharmacokinetics ,chemistry ,Pharmacodynamics ,Medicine ,Dosing ,business ,medicine.drug - Abstract
Background: CC-486 is an orally bioavailable formulation of azacitidine that has demonstrated clinical activity in subjects with Myelodysplastic Syndromes (MDS) and Acute Myleoid Leukemia (AML). In these patient populations, CC-486 in extended dosing regimens, 200 mg and 300 mg given daily on a 14 or 21 out of 28-day schedule, induces sustained global genomic hypomethylation. Methods: This Phase I, multicenter, open-label study was conducted in subjects with solid tumors to evaluate: 1) the effect of carboplatin or ABI-007 on the PK of CC-486; 2) the effect of CC-486 on the PK of carboplatin or ABI-007; and 3) the PD (DNA methylation) effects of CC[[Unable to Display Character: ‑]]486 as a single agent and in combination with carboplatin or ABI-007. Safety and efficacy information of this trial are presented in a separate abstract submission. Subjects received CC-486 on Days 1-14 with carboplatin AUC 4 on Day 8 of a 21-day cycle (Arm A), CC-486 on Days 1-14 with ABI-007 100 mg/m2 given weekly starting on Day 8 of a 21-day cycle (Arm B), or CC-486 on Days 1-21 of a 21-day cycle (Arm C). Results: High inter-subject variability was observed in PK concentrations and parameters. In subjects from Arms A and B, following administration of CC-486 alone or in combination with carboplatin or ABI-007, CC-486 was rapidly absorbed and reached Tmax within approximately 1.0 hour post-dose (median); azacitidine PK parameters (AUC, Cmax, half-life elimination, apparent clearance, and volume of distribution) were comparable following administration of CC-486 alone or in combination. Similarly, carboplatin and ABI-007 PK parameters were similar following administration of carboplatin or ABI-007 alone or in combination with CC-486. In subjects from the 3 Arms, global genomic hypomethylation was observed in PBMCs, with maximum effect on Day 15. A PK/PD (AUC/hypomethylation change on Day 15) correlation was noted (r>0.5; p Conclusion: Carboplatin and ABI-007 had minimal to no effect on CC-486 PK parameters, and vice-versa. Following administration of CC-486, global genomic hypomethylation was correlated with plasma AUC. Citation Information: Mol Cancer Ther 2013;12(11 Suppl):B217. Citation Format: Eric Laille*, Aaron N. Nguyen*, Gengxi Chen, Drew Rasco, Patricia LoRusso, Daniel D. Von Hoff, Johanna Bendell, Pamela Munster, William J. Edenfield, Ramesh Ramanathan, Michael B. Gonzalez, Anne Blackwood-Chirchir, Jorge DiMartino. A Phase I study of CC-486 (oral azacitidine) to evaluate the pharmacokinetics (PK) and pharmacodynamics (PD) of azacitidine administered alone and in combination with carboplatin or ABI-007 (NAB-paclitaxel) in subjects with solid tumors. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2013 Oct 19-23; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2013;12(11 Suppl):Abstract nr B217.
- Published
- 2013
- Full Text
- View/download PDF
42. Extended Dosing of Oral Azacitidine (CC-486) for 14 and 21 Days Provides More Effective Methylation Reversal Than a 7-Day Schedule
- Author
-
Eric Laille, Kyle J. MacBeth, Guillermo Garcia-Manero, Steven D. Gore, Christopher R. Cogle, Keshava Kumar, Barry S. Skikne, and Tao Shi
- Subjects
business.industry ,Myelodysplastic syndromes ,Immunology ,Azacitidine ,Half-life ,Cell Biology ,Hematology ,Pharmacology ,medicine.disease ,Biochemistry ,Oral Azacitidine ,Pharmacokinetics ,Pharmacodynamics ,medicine ,Dosing ,business ,medicine.drug ,Whole blood - Abstract
Abstract 1337 Background: Oral azacitidine (CC-486) is bioavailable, biologically and clinically active, and well tolerated in patients (pts) with myelodysplastic syndromes (MDS) and acute myeloid leukemia (Garcia-Manero, J Clin Oncol, 2011). Because DNA methyltransferase (DNMT) inhibitors require active cell cycling to effect methylation reversal, prolonged administration of lower doses of azacitidine may provide more extensive reversal of DNA methylation compared with shorter administration. Purpose: We conducted analyses 1) to determine the pharmacokinetic (PK) and pharmacodynamic (PD; ie, DNA demethylating activity) profiles following subcutaneous (SC) AZA and various dosing schedules of oral azacitidine; 2) to assess the correlation between the PK and PD profiles of oral azacitidine administered in extended dosing schedules; and 3) to compare PD effects observed at different time points in the 28-day (d) treatment cycle with SC AZA or oral azacitidine administered for 7 days vs. the PD effects of 300mg QD oral azacitidine administered in extended dosing schedules in pts with MDS. Methods: This multicenter, phase 1 study had 2 parts. In Part 1, 41 pts with MDS, CMML, or AML received SC AZA (75mg/m2 QDx7d of a 28d cycle) for 1 cycle, then oral azacitidine doses ranging from 120 to 600mg (QDx7d of a 28d cycle) in subsequent cycles. In Part 2, 86 pts received oral azacitidine in 1 of 4 extended dosing schedules: 300mg QD or 200mg BID, each for 14d or 21d of repeated 28d cycles. PK parameters were derived from plasma concentrations. The correlation between PK and PD was determined using data from pts who had received oral azacitidine 200mg BID or 300mg QD for 14d or 21d for whom PD data were available at day 15 of the first 28d cycle (C1D15). The PD endpoint was reduction in percentage of highly methylated (≥70%) loci of DNA in whole blood, assessed using Illumina's Infinium Methylation27 Bead Array. Results: SC AZA or oral azacitidine were rapidly absorbed and reached Tmax (median [min, max]) within 0.5 hr [0.2, 1.1] and 1.0 hr [0.3, 3.6] post-dose, respectively. Mean elimination half-life was 1.5 +/− 0.7 hr and 0.6 +/− 0.2 hr for SC and oral azacitidine, respectively. No drug accumulation was noted following multiple dose administration. Compared with SC AZA, 300mg oral azacitidine QDx14d and QDx21d provided mean cumulative exposures (AUC) per cycle of 38% and 56%, respectively. A PK/PD correlation (AUC C1D1 vs. change in methylation on C1D15 compared with baseline) was observed with oral azacitidine 300mg QD or 200mg BID administered for 14d or 21d (r2=0.659, p Conclusions: Methylation reversal was correlated with plasma AUC following oral azacitidine dosing. Consistent with the hypothesis that prolonged exposure to lower doses of DNMT inhibitors leads to improved methylation reversal, extended dosing schedules led to significantly more demethylation compared with the 7-day oral azacitidine schedule. Additionally, DNA hypomethylation was maintained at cycle end with extended oral azacitidine dosing. The oral azacitidine 300mg QDx21d dosing schedule provided the greatest level of sustained hypomethylation at cycle end of all tested dose regimens, including SC AZA. Extended dosing schedules provide the ability to achieve the PD and epigenetic activity of oral azacitidine over a longer period of time. Disclosures: Laille: Celgene: Employment, Equity Ownership. Shi:Celgene: Employment, Equity Ownership. Garcia-Manero:Celgene: Research Funding, Speakers Bureau. Gore:Celgene: Consultancy, Research Funding. Kumar:Celgene: Employment, Equity Ownership. Skikne:Celgene: Employment, Equity Ownership. MacBeth:Celgene: Employment, Equity Ownership.
- Published
- 2012
- Full Text
- View/download PDF
43. A 2-Part Phase I Study in Patients with Solid or Hematologic Malignancies: Dose Proportionality of Subcutaneous (SC) Azacitidine (AZA) and Pharmacokinetics of SC AZA in Patients with Severe Renal Impairment
- Author
-
Joseph Schwarz, Eric Laille, Alain C. Mita, C.L. Beach, Sanjay Goel, and Nashat Y. Gabrail
- Subjects
Creatinine ,medicine.medical_specialty ,Kidney ,business.industry ,Immunology ,Cmax ,Urology ,Cell Biology ,Hematology ,Urine ,Pharmacology ,Biochemistry ,chemistry.chemical_compound ,medicine.anatomical_structure ,chemistry ,Tolerability ,Pharmacokinetics ,Medicine ,Dosing ,business ,Blood urea nitrogen - Abstract
Abstract 3480 Background: The recommended starting dose for all patients receiving SC AZA is 75 mg/m2 daily for 7 days in 28-day cycles. If no response is observed, the dose may be increased to 100 mg/m2. Conversely, if cytopenias do not adequately resolve between dosing cycles, AZA dose may be reduced. Similarly, because AZA and its metabolites are primarily excreted by the kidneys, patients with renal impairment may require monitoring for elevations of BUN or serum creatinine (cr), in which case the next AZA treatment cycle should be delayed until values return to baseline and the next AZA dose should be reduced by 50% (Vidaza® prescribing information, 2011). Currently, the pharmacokinetics (PK) of SC AZA in reduced ( Objectives: To assess the dose proportionality of AZA PK after single SC doses ranging from 25 to 100 mg/m2, and to determine the effect of renal impairment on AZA PK after single and multiple (5 days) SC doses of 75 mg/m2. Also, the safety and tolerability of AZA in patients with severe renal impairment were determined. Methods: This 2-part multicenter, randomized, open-label study included patients with solid or hematologic malignancies. Part 1 was a 4-treatment, parallel-group evaluation of the dose proportionality of SC AZA in patients with normal renal function (cr clearance [CLcr] >80 mL/min/1.73 m2, Cockcroft-Gault equation adjusted for body surface area). Patients were randomized to receive a single dose of 25-, 50-, 75-, or 100-mg/m2 SC AZA. Blood and urine samples were collected before dosing and at various time points up to 8 hours post-dose. The 75 mg/m2 dosing group in Part 1 received an additional 4 days of AZA treatment and blood and urine were collected from these patients on the same schedule on Day 5. For Part 2, patients with severe renal impairment (CLcr Results: At the time of this analysis, 21 patients were enrolled and were included in safety evaluations, with PK data available for 18 patients. At baseline, median ages of patients with normal renal function (n=17) or severe renal impairment (n=4) were 61 years (range: 38–76) and 71 years (range: 54–90), respectively. Of patients with normal renal function, 12 (70%) had solid tumors, 4 had MDS (RAEB-t [n=2], RA, RARS), and 1 had multiple myeloma. Of patients with severe renal impairment, 2 (50%) had solid tumors, 1 had CMML, and 1 had MDS (RA). In Part 1, 14 patients were randomized to either 25 mg/m2 (n=4), 50 mg/m2 (n=4), 75 mg/m2 (n=3), or 100 mg/m2 (n=3). Mean [±SD] AUC0-∞ in the 25-, 50-, 75-, and 100 mg/m2 dose groups were 490 [146], 895 [300], 1270 [480], and 1410 [212] ng*hr/mL, respectively. Preliminary results show AZA is dose proportional across the 25–100 mg/m2dose range (Figure 1). In Part 2, on Days 1 and 5 of 5 consecutive days of SC AZA administration, AZA was rapidly absorbed by patients with severe renal impairment, reaching Cmax within 0.75 hours post-dose. AZA concentration decreased thereafter in a pseudobiphasic manner (Figure 2). Similar profiles were observed in patients with normal renal function who received the same dose. Mean [±SD] AUC0-∞ values after a 75 mg/m2 SC AZA dose on Day 1 were 1270 [480] ng*hr/mL in patients with normal renal function and 1630 [913] ng*hr/mL in patients with severely impaired renal function. On Day 5, mean AUC0-∞ values were 901 [92] and 1280 [728] ng*hr/mL, respectively. Similar observations were noted for Cmax. No accumulation of AZA was noted on Day 5 in either group. High inter-patient variability was noted in both groups (% coefficient of variation up to ∼82%). Patients with or without renal impairment did not show unusual or unexpected adverse events. Conclusions: AZA is dose-proportional over the 25–100 mg/m2 dosing range. PK parameters from patients with severe renal impairment treated with multiple doses of AZA 75 mg/m2 SC were comparable to those obtained from patients with normal renal function. Treatment with AZA 75 mg/m2 SC over multiple days was safe and well tolerated in this small group of patients with normal renal function or severe renal impairment. Disclosures: Laille: Celgene Corporation: Employment, Equity Ownership. Goel:Celgene: Research Funding. Schwarz:Celgene Corporation: Employment, Equity Ownership. Beach:Celgene Corporation: Employment, Equity Ownership.
- Published
- 2011
- Full Text
- View/download PDF
44. Oral Azacitidine (AZA) Activity in Patients with Acute Myelogenous Leukemia (AML)
- Author
-
Eric Laille, Christopher R. Cogle, Heidi Giordano, Barry S. Skikne, Steven D. Gore, Tao Shi, Guillermo Garcia-Manero, and Kyle J. MacBeth
- Subjects
medicine.medical_specialty ,education.field_of_study ,Acute myelogenous leukemia (AML) ,business.industry ,Immunology ,Population ,Cmax ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Gastroenterology ,Chemotherapy regimen ,Oral Azacitidine ,Surgery ,stomatognathic diseases ,Regimen ,Pharmacodynamics ,Internal medicine ,Medicine ,business ,education ,Febrile neutropenia - Abstract
Abstract 1546 Background: When administered subcutaneously (SC), the cytidine nucleoside analog azacitidine (AZA) has been shown to significantly improve overall survival in patients (pts) with low-blast count (20%–30%) AML compared with conventional care regimens (Fenaux, JCO, 2010). An oral AZA formulation would be more convenient for pts, increase access to treatment, allow extended dosing schedules, and may improve efficacy. An initial study of oral AZA administered once daily on a 7-day schedule demonstrated it to be bioavailable, safe, and clinically active in pts with myelodysplastic syndromes (MDS) or chronic myelomonocytic leukemia (CMML) (Garcia-Manero, JCO, 2011). Purpose: To present results of a multicenter phase I study of the safety, pharmacokinetic (PK) and pharmacodynamic (PD) profiles, and preliminary hematologic response rates in pts with AML receiving varying dose regimens of oral AZA. Methods: Pts aged ≥ 18 years with AML who were not candidates for other therapies (but who may have failed prior treatment) were included. In Part 1, pts received an initial cycle of SC AZA 75 mg/m2 QD x 7 days of a 28-day cycle. Thereafter, oral AZA (120–600 mg QD) was given for 7 days of repeated 28-day cycles. In Part 2, pts received only oral AZA, in 1 of 4 extended dosing schedules: 300 mg QD or 200 mg BID, each for 14 or 21 days of 28-day cycles. Oral AZA was continued until disease progression or withdrawal for other reasons. PK was assessed during the first cycle of oral AZA treatment and DNA methylation was assessed during cycles 1 and 2 of oral AZA (Infinium Methylation27 BeadArray). Pts were monitored continuously for adverse events (AEs) and assessed for treatment response (Cheson, JCO, 2003; Cheson, JCO, 2006) at the end of every second cycle. Results: In all, 23 pts (median age 68 years [range 44–85], 13 male/10 female) with AML (13 de novo, 10 transformed from MDS) have received oral AZA on 7-day QD (n=8), 14-day QD (n=4), 21-day QD (n=4), 14-day BID (n=4), or 21-day BID (n=3) treatment schedules. At baseline, 14 (61%) and 8 (35%) pts were red blood cell (RBC) or platelet transfusion dependent, respectively; 13 pts (57%) had prior AML treatment, primarily intensive chemotherapy. Eight pts had unfavorable and 12 pts had intermediate cytogenetics (data not available, n=3). At baseline, median (range) hematologic measures were WBC = 1.5 K/μL (0.4 − 21.2), ANC = 0.2 K/μL (0 − 3.5), platelets = 34.0 K/μL (6.0 − 102), and % BM blasts = 28.0% (2.0 − 92.0). Median duration of oral AZA (any regimen) was 4 cycles (range 1–14). Seven pts (30%) received ≥ 6 cycles of oral AZA and 16 pts discontinued before completing 6 cycles (Figure). PK results were consistent with recently published data (Garcia-Manero, JCO, 2011). Following a single 300 mg dose (n=8), AUC0-∞ = 230 ± 210 ng*hr/mL, Cmax = 150 ± 114 ng/mL, t1/2 = 0.54 ± 0.21 hr, and median Tmax = 0.8 hr. There was no accumulation following multiple doses. Compared with extrapolated cumulative exposure of SC AZA administered for 7 days per the approved dosing regimen, extrapolated cumulative exposures of oral AZA with extended dosing schedules ranged from 55% for 300 mg QD × 14 days to 104% for 200 mg BID × 21 days. Overall, hematologic improvement (HI) was achieved by 5 pts (22%) (Table). One pt receiving 7-day QD oral AZA achieved a CRi. Responses were seen in 3/13 pts with de novo AML and 3/10 with secondary AML (Figure), and in 2/13 pts with prior AML treatment and 4/10 pts without prior treatment. DNA methylation levels in whole blood were reduced after oral AZA in 4/7 tested pts. The most common AEs (any severity) were gastrointestinal. Most common grade 3–4 AEs were febrile neutropenia (35%), pneumonia and syncope (17% each), nausea (13%), and thrombocytopenia, diarrhea, fatigue, dehydration, headache and mental status changes (9% each). Two pts died due to causes related to AML, and 2 pts discontinued (treatment failure, withdrew consent), within the first 60 days on-study. Conclusions: Oral AZA treatment was well tolerated in this AML population of pts who were not candidates for other therapy. The oral formulation showed biological activity by reducing DNA methylation. Oral AZA also showed clinical activity in these AML pts, many of whom had complex cytogenetics and/or had failed prior therapy before study entry. Disclosures: Gore: Celgene Corporation: Consultancy, Equity Ownership, Research Funding. Off Label Use: Oral formulation for Azacitidine in AML patients. Cogle:Celgene Corporation: Research Funding. Skikne:Celgene Corporation: Employment, Equity Ownership. Laille:Celgene Corporation: Employment, Equity Ownership. Shi:Celgene Corporation: Employment, Equity Ownership. MacBeth:Celgene Corporation: Employment, Equity Ownership. Giordano:Celgene Corporation: Employment, Equity Ownership.
- Published
- 2011
- Full Text
- View/download PDF
45. Evaluation of Oral Azacitidine Using Extended Treatment Schedules: A Phase I Study
- Author
-
M. Renee Ward, Eric Laille, Barry S. Skikne, Kyle J. MacBeth, Christopher R. Cogle, Guillermo Garcia-Manero, Elias Jabbour, Heidi Giordano, Hagop M. Kantarjian, and Steven D. Gore
- Subjects
medicine.medical_specialty ,Nausea ,business.industry ,Immunology ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Oral Azacitidine ,Pharmacokinetics ,Pharmacodynamics ,Internal medicine ,medicine ,Vomiting ,Dosing ,medicine.symptom ,business ,Adverse effect ,Febrile neutropenia - Abstract
Abstract 603 Parenteral azacitidine (AZA) is approved for administration on days 1–7 of a 28-day treatment schedule. Based on the short plasma half-life of AZA, S-phase restricted incorporation into DNA, and rapid re-methylation of DNA, it is possible that chronic daily exposure could enhanced its clinical activity. An oral formulation would be convenient and allow evaluation of lower doses administered on extended schedules. The initial phase I study of oral AZA, administered daily on a 7-day schedule demonstrated that it was bioavailable, safe, and clinically active in patients with MDS and AML (Garcia-Manero G, et al. Blood 2009;114:A117). Here, we report the results of a multicenter phase I exploration of extended oral AZA schedules, including dose-limiting toxicities (DLTs), safety, pharmacokinetic (PK), pharmacodynamic (PD), and preliminary response data. Patients aged ≥ 18 years with MDS, CMML or AML (not candidates for other therapies) were enrolled in the study. Inclusion criteria were a hemoglobin level of ≤ 9.0 g/dL, and/or platelet count of ≤ 50 × 109/L, and/or be RBC transfusion-dependent; prior azanucleoside therapy was not permitted. Patients received oral AZA daily (QD) or twice daily (BID) on 14- or 21-days schedules, with starting at a dose of 300 mg for QD dosing and 200 mg for BID dosing. Patients were enrolled into cohorts of 6 and evaluated for DLTs at the end of Cycle 1. Patients were monitored continuously for adverse events (AEs) and assessed for disease response at the end of every second cycle. During Cycle 1, on the first and last day of treatment, PK parameters were derived from AZA concentrations in the plasma after the first dose of the day. PD samples were collected during the first 2 cycles and DNA methylation changes were evaluated using a LINE-1 assay. To date, 25 patients (median age 68 years [range 44–87]; 14 male and 11 female) with MDS (n = 13), AML (n = 7 de novo and n = 3 transformed), and CMML (n = 2) have received oral AZA on extended treatment schedules. Two DLTs, grade 3 nausea and grade 3 vomiting, occurred in 1 of 6 DLT-evaluable patients treated at 14-days QD (n = 7). No DLTs were observed on the 21-day QD (n = 6) or 14-day BID (n = 6) schedules; safety evaluation for the 21-day BID schedule is ongoing (n = 6). The maximum tolerated dose has not been reached on these schedules; no patient has received > 300 mg per dose. Overall rates of all grades nausea, vomiting, diarrhea, constipation, and abdominal pain with the extended schedules were similar to those observed with the oral 7-day schedule. The rate of febrile neutropenia (all grades) was higher in the 21-day QD cohort. This was observed in 4 patients with baseline ANC < 500 and/or AML diagnosis. Most common grade 3/4 AEs in the QD schedules were febrile neutropenia (14-day, 1/7; 21-day, 4/6), anemia (14-day, 1/7; 21-day, 0/6), thrombocytopenia (14-day, 1/7; 21-day, 1/6), diarrhea (14-day, 0/7; 21-day, 1/6), nausea (14-day, 1/7; 21-day, 0/6), and vomiting (14-day, 1/7; 21-day, 0/6). Extended BID schedules are under evaluation. PK data have been generated for 19 of 25 patients. For the 300 mg 14-day QD, 300 mg 21-day QD, and 200 mg 14-day BID schedules, using mean AUC (first and last day) results, extrapolated cumulative exposures per cycle were ~28%, 42% and 26%, respectively, compared with historical exposure observed following subcutaneous administration. AZA exposure increased with increasing dose, but was not dose-proportional. Clinical responses were observed for MDS/CMML patients on both extended QD schedules, with assessment ongoing for BID schedules (Table). In summary, extended (14- and 21-day) dosing of oral AZA is generally well tolerated, with no AZA accumulation, and promising clinical responses were observed, including complete remission (CR), marrow CR (mCR), and hematologic improvement (HI). Table. Parameter, n (%) Oral AZA Treatment Schedule MDS/CMML Responders/Evaluable patients, (%) 14-day QD 21-day QD Overall response* (CR, mCR, any HI) 5/6 (83) 3/3 (100) CR 0/6 2/3 (67) mCR 0/6 3/3 (100) HI 5/6 (83) 3/3 (100) HI-erythroid 3/5 (60) 1/1 (100) HI-platelet† 2/5 (40) 3/3 (100) HI-neutrophil 0/1 0/1 Transfusion independence 3/4 (75) 1/2 (50) RBC 1/2 (50) 1/1 (100) Platelet 2/2 (100) 0/1 * International Working Group 2006 criteria, patients only counted once for overall response, but may be counted more than once in individual response categories. † Includes patients achieving partial (≥ 50%) or complete platelet transfusion independence. Disclosures: Gore: Celgene: Consultancy, Equity Ownership, Research Funding. Cogle:Celgene: Research Funding, Speakers Bureau. Ward:Celgene: Equity Ownership. MacBeth:Celgene: Employment, Equity Ownership. Laille:Celgene: Employment. Giordano:Celgene: Employment, Equity Ownership. Kantarjian:Celgene: Research Funding. Skikne:Celgene: Employment.
- Published
- 2010
- Full Text
- View/download PDF
46. A Phase 1, Open-Label, Dose-Escalation Study to Evaluate the Safety, Pharmacokinetics, and Pharmacodynamics of Oral Azacitidine in Patients with Myelodysplastic Syndromes (MDS) or Acute Myelogenous Leukemia (AML)
- Author
-
Christopher R. Cogle, Steven D. Gore, Eric Laille, Kyle J. MacBeth, M. Renee Ward, Yuhong Ning, Barry S. Skikne, and Guillermo Garcia-Manero
- Subjects
Oncology ,Not evaluated ,medicine.medical_specialty ,Acute myelogenous leukemia (AML) ,business.industry ,Immunology ,Azacitidine ,Cell Biology ,Hematology ,Pharmacology ,medicine.disease ,Biochemistry ,Oral Azacitidine ,Demethylating agent ,Regimen ,chemistry.chemical_compound ,chemistry ,Internal medicine ,Pharmacodynamics ,medicine ,business ,Febrile neutropenia ,medicine.drug - Abstract
Abstract 117 Background: Parenteral azacitidine significantly improves overall survival of subjects with higher-risk MDS and WHO AML (20–30% blasts) compared to conventional care (Fenaux, Lancet Oncol 2009; 10:223). An oral formulation would be a more convenient administration route for patients and allow the evaluation of extended low-dose regimens, which could in turn lead to better treatment efficacy and lower toxicity profiles. The aim of this phase 1 study is to determine the maximum tolerated dose (MTD), dose limiting toxicities (DLTs), safety, pharmacokinetic (PK) and pharmacodynamic (PD) profiles of increasing doses of orally-administered azacitidine in subjects with MDS or AML. Methods: Subjects aged ≥ 18 years with a diagnosis of MDS or AML (not candidates for other therapies) by WHO criteria were enrolled into this multicenter, Phase 1, dose escalation study. Prior treatment with azacitidine or other demethylating agent was permitted. To allow PD/PK analyses of oral vs. subcutaneous (SC) administration, azacitidine was administered SC (75 mg/m2/day x 7 days) in Cycle 1, then orally in Cycle 2 starting at a dose of 120 mg/day x 7 days of every 28-day cycle. A standard “3+3” dose escalation design was used and an expansion cohort at the MTD was planned. Subjects were evaluated for DLT during the first cycle of oral azacitidine therapy. Azacitidine levels in plasma and urine were measured. DNA methylation was assayed using Illumina's Infinium Human Methylation27 BeadArrays. Response was assessed according to International Working Group (IWG) criteria for MDS (2006) and AML (2003). Results: To date, 45 subjects have enrolled into the study. Forty subjects received both SC and oral azacitidine; 5 subjects received SC azacitidine only. Median age is 70 years (range 31–91). Thirty-four subjects had MDS, 9 AML, and 2 CMML. Median pretreatment WBC and platelet counts of MDS subjects were 2.5 × 109/L (range 0.8–39.4) and 55.0 × 109/L (range 4.0–234.0) respectively. For subjects with AML, the median pretreatment WBC and platelet counts were 1.8 × 109/L (range 0.5–3.4) and 42.8 × 109/L (range 10.0–82.0) respectively. Baseline cytogenetics for subjects with MDS were as follows: 12 complex karyotype; 10 diploid; 1 with −7; 1 with t(11q23) and 10 whose karyotype was unknown. Doses of oral azacitidine evaluated were 120, 180, 240, 300, 360, 480 and 600 mg. The MTD of oral azacitidine on a 7-day QD treatment schedule was 480 mg. The DLT was grade 3 or 4 diarrhea in 2 of 3 subjects enrolled in the 600 mg cohort. Other grade 3 or 4 AEs reported include febrile neutropenia (6 subjects), infection (6 subjects), nausea (2 subjects), vomiting (2 subjects), fatigue (2 subjects), and thrombocytopenia (2 subjects). The bioavailability of oral azacitidine ranged from 5% to 35%. The exposure (AUCinf) of oral azacitidine was highly variable compared to SC exposure, ranging from 15% to 74% (except for one subject whose PK exposure following oral therapy was 167% of his SC exposure). Median duration of oral azacitidine was 5+ cycles (range 1–19+) for subjects with MDS and 2.5+ cycles (range 1–6+) for subjects with AML. Responses were evaluated following 6 cycles of oral therapy. To date, 14 subjects have received ≥ 6 cycles of oral azacitidine: 4 (29%) had complete response (CR), 6 (43%) had stable disease (SD), 3 (21%) had disease progression, and 1 subject (7%) was not evaluated. Among subjects with CR or SD, hematologic improvement by disease-specific IWG criteria included erythroid response (n = 5), platelet response (n = 5) and neutrophil response (n = 3). Fourteen subjects are ongoing, but have not completed 6 cycles of oral therapy; 17 subjects discontinued prior to completing 6 cycles. Changes in DNA methylation were observed in the blood of MDS and AML subjects following SC and oral azacitidine treatment, and a set of CpG loci were identified for which these changes were associated with azacitidine AUCinf and/or Cmax.. In addition, approximately 260 hypomethylated loci were common to both SC and oral therapy. Conclusions: Results to date indicate that a 7-day dosing regimen of oral azacitidine is active and well tolerated, with a manageable side effect profile in subjects with MDS or AML. Oral azacitidine exposure was highly variable and lower than that of SC azacitidine. Changes in DNA methylation were observed in the blood of MDS and AML subjects following SC and oral therapy. Evaluation of prolonged and BID treatment schedules is planned. Disclosures: Garcia-Manero: Celgene: Research Funding. Gore:Celgene: Consultancy, Equity Ownership, Research Funding; Johnson & Johnson: Research Funding. Skikne:Celgene: Employment. Cogle:Celgene: Research Funding, Speakers Bureau; Eisai: Speakers Bureau. Ning:Celgene: Employment, Equity Ownership. MacBeth:Celgene Corporation: Employment, Equity Ownership. Laille:Celgene: Employment, Equity Ownership. Ward:Celgene: Employment, Equity Ownership.
- Published
- 2009
- Full Text
- View/download PDF
47. The pharmacokinetics of azacitidine following subcutaneous treatment in patients with myelodysplastic syndromes (MDS) or acute myelogenous leukemia (AML)
- Author
-
G. Garcia-Manero, Christopher R. Cogle, Renee Ward, Abdullah Nasser, Barry S. Skikne, Eric Laille, Steven D. Gore, and M. Stoltz
- Subjects
Cancer Research ,Acute myelogenous leukemia (AML) ,business.industry ,Cellular differentiation ,Myelodysplastic syndromes ,Azacitidine ,medicine.disease ,DNA metabolism ,Oncology ,Pharmacokinetics ,Gene expression ,medicine ,Cancer research ,In patient ,business ,neoplasms ,medicine.drug - Abstract
7087 Background: 5-azacitidine (AZA), through its effects on DNA metabolism, gene expression, and cell differentiation, has proven beneficial in treatment of MDS and AML and AZA therapy significantly increases survival in higher-risk MDS and AML compared to conventional care. Few studies have evaluated the pharmacokinetics (PK) of AZA and the renal elimination of AZA has not been previously published to our knowledge. Plasma PK of AZA are herein reported in patients receiving SC doses of 75 mg/m2. This study was designed to also assess the contribution of renal elimination to the overall clearance of AZA. Methods: Adult patients with MDS or AML and ECOG status 0–2 were treated with 7 consecutive daily SC doses of 75 mg/m2 AZA during their first treatment cycle. PK parameters of AZA were derived from drug concentrations in plasma and urine collected after the first and last dose (day 7) of AZA. Safety was evaluated by adverse event reporting (NCI-CTC). Results: Currently, 18 patients have been treated with SC AZA. AZA was rapidly absorbed and reached peak plasma concentrations (concs) within 0.5 hr post dosing. The AUCinf after SC doses was 1170 hr*ng/mL. The AZA concs declined in a pseudo bi-phasic manner with an elimination half-life of 1.25 hours. The plasma PK profiles after the first and last dose were superimposable. The apparent total clearance (CL/F) and volume of distribution (Vd/F) were 143 L/hr and 318 L, respectively. AZA recovery in urine was very small relative to dose (inf after SC doses is similar to the published AUC value (1044 hr*ng/mL) after 75 mg/m2 IV doses indicating approximating 100% systemic bioavailability. After SC dosing, CL/F exceeded hepatic blood flow indicating extra-hepatic metabolism. Vd/F was 4–5 fold greater than total body water suggesting extensive AZA tissue distribution. Renal elimination appears to play a minor role in the overall clearance of AZA. [Table: see text]
- Published
- 2009
- Full Text
- View/download PDF
48. Treatment of relapsed or refractory non-hodgkin lymphoma with the oral isotype-selective histone deacetylase inhibitor MGCD0103: Interim results from a phase II study
- Author
-
Charalambos Andreadis, Robert E. Martell, Michael Crump, Eric Laille, P. McLaughlin, David A. Rizzieri, Sarit Assouline, Ahmed I. Younes, Zuomei Li, and A. Wedgwood
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Pathology ,medicine.drug_class ,business.industry ,Histone deacetylase inhibitor ,Follicular lymphoma ,Phases of clinical research ,medicine.disease ,Isotype ,Lymphoma ,Refractory ,Refractory Non-Hodgkin Lymphoma ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Stem cell ,business - Abstract
8528 Background: MGCD0103 is an orally available selective inhibitor of histone deacetylases (HDACs) with significant biological activity in preclinical models of both solid tumors and hematopoietic cancers. Methods: Open-label, Phase II trial in adults (Trial 008) with relapsed or refractory diffuse large B-cell lymphoma (DLBCL) or follicular lymphoma (FL). Eligibility included: ≥1 site of measurable disease (≥2.0 cm), ECOG 0 or 1. MGCD0103 was given 3x/week starting at 110 mg; after 32 patients (pts) the starting dose was reduced to 85 mg. Tumor responses were determined every 2 cycles. PK was evaluated on Day 1, Cycle 1. Results: 50 pts received treatment; 33 DLBCL and 17 FL (median age [range], 61.5 years [32−80]; male, n=27; prior rituximab therapy: 96%; prior stem cell transplant: 33%). The response rate in pts with DLBCL met the protocol defined criteria for expansion beyond 21 pts, and enrollment of this cohort is now complete. Of 17 DLBCL pts with tumor reassessed by CT, most had tumor reduction,...
- Published
- 2008
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.