51
|
Qiu HB, Zhuang W, Wu T, Xin S, Lin CZ, Ruan HL, Zhu X, Huang M, Li JL, Hou XY, Zhou ZW, Wang XD. Imatinib-induced ophthalmological side-effects in GIST patients are associated with the variations of EGFR, SLC22A1, SLC22A5 and ABCB1. THE PHARMACOGENOMICS JOURNAL 2017; 18:460-466. [PMID: 28762371 DOI: 10.1038/tpj.2017.40] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 05/27/2017] [Accepted: 06/07/2017] [Indexed: 12/14/2022]
Abstract
Imatinib-induced ophthalmological side-effects, including conjunctiva hemorrhage and periorbital oedema, although very common and still remain relatively little understood. The present study investigated the effects of genetic polymorphisms of drug targets and membrane transporters on these side effects. We found that the minor allele of EGFR rs10258429 and SLC22A1 rs683369 were significant risk determinants of conjunctival hemorrhage with OR of 7.061 (95%CI=1.791-27.837, P=0.005 for EGFR rs10258429 CT+TT vs CC), and 4.809 (95%CI=1.267-18.431, P=0.021 for SLC22A1 rs683369 GG+CG vs CC). The minor allele of SLC22A5 rs274558 and ABCB1 rs2235040 were protective factors to periorbital oedema with OR of 0.313 (95%CI=0.149-0.656, P=0.002 for SLC22A5 rs274558 AA+AG vs GG), and 0.253 (95%CI=0.079-0.805, P=0.020 for ABCB1 rs2235040 CT vs CC). These results indicated that variants in EGFR, SLC22A1, SLC22A5 and ABCB1 influenced the incidence of Imatinib-induced ophthalmological toxicities, and polymorphism analyses in associated genes might be beneficial to optimize Imatinib treatment.
Collapse
Affiliation(s)
- H-B Qiu
- Department of Gastric Surgery, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - W Zhuang
- Institute of Clinical Pharmacology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - T Wu
- Department of Gastric Surgery, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - S Xin
- Department of Gastric Surgery, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Institute of Clinical Pharmacology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - C-Z Lin
- Institute of Clinical Pharmacology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China.,Department of Pharmacy, Huadu District People's Hospital, Guangzhou, China
| | - H-L Ruan
- School of Public Health, Guangzhou Medical University, Guangzhou, China
| | - X Zhu
- Department of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - M Huang
- Institute of Clinical Pharmacology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - J-L Li
- Institute of Clinical Pharmacology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - X-Y Hou
- Institute of Clinical Pharmacology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Z-W Zhou
- Department of Gastric Surgery, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - X-D Wang
- Institute of Clinical Pharmacology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
52
|
Shimomura A, Kondo S, Kobayashi N, Iwasa S, Kitano S, Tamura K, Fujiwara Y, Yamamoto N. Do all patients in the phase I oncology trials need to be hospitalized? Domestic but outstanding issues for globalization of drug development in Japan. Int J Clin Oncol 2017; 22:780-785. [PMID: 28293794 PMCID: PMC5533862 DOI: 10.1007/s10147-017-1108-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 02/25/2017] [Indexed: 11/25/2022]
Abstract
INTRODUCTION Most trials investigating new drugs around the world, including phase I trials, are conducted in outpatient clinics. However, in Japan, regulatory authority requirements and traditional domestic guidelines often require hospitalization of phase I study participants. PATIENTS AND METHODS Patients participating in single-agent phase I clinical trials at National Cancer Center Hospital between December 1996 and August 2014 were monitored. Toxicity requiring hospitalization is defined as toxicity that needs intensive treatment. Study designs were classified into three types: first-in-human (FIH) study, dose-escalation study (conventional dose-escalation study to determine maximum tolerated dose (MTD) in Japanese patients), and dose-finding study (to assess safety and pharmacokinetic profiles up to the MTD previously determined in the West). RESULTS A total of 945 patients who participated in a variety of single-agent phase I clinical trials between December 1996 and August 2014 were included in this study. Patients participated in one of three study types: dose-escalation (n = 582, 62%), first-in-human (n = 129, 14%), or dose-finding (n = 234, 25%). A total of 76 study drugs were evaluated as part of this pool of phase I studies. Subdivided by mechanism of action, 20 (26%) were cytotoxic, 50 (66%) were molecularly targeted, and 6 (8%) were immune checkpoint inhibitor. Thirty-six patients (3.8%) had severe toxicities requiring hospitalization during the first cycle. The overall number of toxicities requiring hospitalization and/or grade 4 toxicities during any cycle was 5.0%. CONCLUSIONS The frequency of severe toxicity that needs to be hospitalized was unexpectedly low. The data did not demonstrate the need for hospitalization in the phase I trials, suggesting that phase I trials in Japan could be conducted in outpatient settings.
Collapse
Affiliation(s)
- Akihiko Shimomura
- Department of Experimental Therapeutics, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Shunsuke Kondo
- Department of Experimental Therapeutics, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Noriko Kobayashi
- Clinical Trial Support Office, National Cancer Center Hospital, Tokyo, Japan
| | - Satoru Iwasa
- Department of Experimental Therapeutics, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Shigehisa Kitano
- Department of Experimental Therapeutics, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Kenji Tamura
- Department of Experimental Therapeutics, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yutaka Fujiwara
- Department of Experimental Therapeutics, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Noboru Yamamoto
- Department of Experimental Therapeutics, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan.
- Clinical Trial Support Office, National Cancer Center Hospital, Tokyo, Japan.
| |
Collapse
|
53
|
Lee SM, Zhang Y, Minasian LM, Unger JM, Hershman DL. Using Delayed Toxicities to Re-evaluate Tolerability in Phase 2 Trials: A Case Example using Bortezomib. Cancer Invest 2017; 35:484-489. [PMID: 28692317 DOI: 10.1080/07357907.2017.1340479] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
In advanced stage patients enrolled in dose-finding trials, it is difficult to assess delayed toxicities because frequently patients discontinue after one or two cycles of treatment. Patients enrolled in phase 2 trials are typically followed longer to assess efficacy. Thus, their data may be useful for evaluating long-term tolerability. We illustrate this using as example two phase 2 bortezomib trials (total N = 172) conducted by SWOG. While treatment-related severe toxicity rates based on cycle 1 were acceptable (23% and 31%), they were notably higher over extended administration (37% and 70%). This additional information should be considered when designing subsequent trials.
Collapse
Affiliation(s)
- Shing M Lee
- a Department of Biostatistics , Mailman School of Public Health, Columbia University , New York , NY , USA
| | - Yuan Zhang
- a Department of Biostatistics , Mailman School of Public Health, Columbia University , New York , NY , USA
| | | | - Joseph M Unger
- c SWOG Statistical Center, Fred Hutchinson Cancer Research Center , Seattle , WA , USA
| | - Dawn L Hershman
- d Herbert Irving Comprehensive Cancer Center, Department of Medicine , Columbia University , New York , NY , USA
| |
Collapse
|
54
|
Henon C, Lissa D, Paoletti X, Thibault C, Le Tourneau C, Lanoy E, Hollebecque A, Massard C, Soria JC, Postel-Vinay S. Patient-reported tolerability of adverse events in phase 1 trials. ESMO Open 2017; 2:e000148. [PMID: 28761740 PMCID: PMC5519806 DOI: 10.1136/esmoopen-2016-000148] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 02/12/2017] [Accepted: 02/15/2017] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND Phase I experts recommend revisiting dose-limiting toxicity (DLT) definition to include chronic and cumulative toxicities induced by new molecularly targeted therapies. Patient's assessment of late toxicities' tolerability is, however, unknown. MATERIALS AND METHODS A prospective survey on adverse events (AEs) tolerability on 23 National Cancer InstituteCommon Terminology Criteria for Adverse Event, Version 4 (NCI-CTCAE.v4) items was conducted at Gustave Roussy's Phase I department. Patients' maximum tolerability duration was recorded at baseline, during trial and at trial completion. Results were compared with the corresponding physicians' survey. RESULTS 52 patients enrolled in 27 Phase I trials between May 2014 and November 2015 completed 102 forms. At baseline, the most feared G2/G3 AEs were haematuria (74%), vomiting (71%) and hyperglycemia (64%)/dry mouth (94%), hyperglycemia (92%) and vomiting (92%). At trial completion, the most feared G2/G3 AEs were personality change (83.3%), haematuria (82%) and fever (80%)/dry mouth, fever and dizziness (100% each). Tolerability score did not differ over time. More previous treatments and occurrence of severe AEs were associated with better tolerability at study completion (p=0.0234 and p=0.0153, respectively, in multivariate analysis). Patient's tolerability differed from physician's assessment. CONCLUSION AEs considered intolerable by patients are toxicities that directly impact their quality of life and differ from those feared by physicians or included in DLT definition. Patient-reported tolerability of AEs may help in optimising drug development.
Collapse
Affiliation(s)
- Clémence Henon
- Drug Development department (DITEP), Gustave Roussy Cancer Campus, Paris-Saclay University, Villejuif, France
| | - Delphine Lissa
- Drug Development department (DITEP), Gustave Roussy Cancer Campus, Paris-Saclay University, Villejuif, France
| | - Xavier Paoletti
- Biostatistics and Epidemiology Department, Gustave Roussy Cancer Campus, Villejuif, France.,CESP-OncoStat, INSERM and Paris-Saclay University, Paris-Sud University, Versailles Saint-Quentin-en-Yvelines University, Villejuif, France
| | - Constance Thibault
- Drug Development department (DITEP), Gustave Roussy Cancer Campus, Paris-Saclay University, Villejuif, France
| | - Christophe Le Tourneau
- Department of Medical Oncology, Institut Curie, Paris, France.,UMR900, INSERM, Paris, France
| | - Emilie Lanoy
- Biostatistics and Epidemiology Department, Gustave Roussy Cancer Campus, Villejuif, France.,CESP-OncoStat, INSERM and Paris-Saclay University, Paris-Sud University, Versailles Saint-Quentin-en-Yvelines University, Villejuif, France
| | - Antoine Hollebecque
- Drug Development department (DITEP), Gustave Roussy Cancer Campus, Paris-Saclay University, Villejuif, France
| | - Christophe Massard
- Drug Development department (DITEP), Gustave Roussy Cancer Campus, Paris-Saclay University, Villejuif, France
| | - Jean-Charles Soria
- Drug Development department (DITEP), Gustave Roussy Cancer Campus, Paris-Saclay University, Villejuif, France.,UMR981, INSERM, Villejuif, France
| | - Sophie Postel-Vinay
- Drug Development department (DITEP), Gustave Roussy Cancer Campus, Paris-Saclay University, Villejuif, France.,UMR981, INSERM, Villejuif, France
| |
Collapse
|
55
|
Yin J, Shen S. Challenges and Innovations in Phase I Dose-Finding Designs for Molecularly Targeted Agents and Cancer Immunotherapies. JOURNAL OF BIOMETRICS & BIOSTATISTICS 2017; 7. [PMID: 28616356 PMCID: PMC5467542 DOI: 10.4172/2155-6180.1000324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Phase I oncology trials are designed to identify a safe dose with an acceptable toxicity profile. In traditional phase I dose-finding design, the dose is typically determined based on the probability of severe toxicity observed during the first treatment cycle. The recent development of molecularly targeted agents and cancer immunotherapies call for new innovations in phase I designs, because of prolonged treatment cycles often involved. Various phase I designs using toxicity and efficacy endpoints from multiple treatment cycles have been developed for these new treatment agents. Here, we will review the novel endpoints and designs for the phase I oncology clinical trials.
Collapse
Affiliation(s)
- Jun Yin
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905, USA
| | - Shihao Shen
- Department of Microbiology, Immunology, & Molecular Genetics, UCLA, Los Angeles, CA 90024, USA
| |
Collapse
|
56
|
Outcomes and prognostic factors for relapsed or refractory lymphoma patients in phase I clinical trials. Invest New Drugs 2017; 36:62-74. [PMID: 28597151 DOI: 10.1007/s10637-017-0480-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Accepted: 06/01/2017] [Indexed: 12/22/2022]
Abstract
Background Although safety and prognostic factors for overall survival (OS) have been extensively studied in Phase I clinical trials on patients with solid tumours, data on lymphoma trials are scarce. Here, we investigated safety, outcomes and prognostic factors in relapsed or refractory lymphoma patients included in a series of Phase I trials. Method and patients All consecutive adult patients with recurrent/refractory lymphoma enrolled in 26 Phase I trials at a single cancer centre in France between January 2008 and June 2016 were retrospectively assessed. Results 133 patients (males: 65%) were included in the analysis. The median (range) age was 65 (23-86). Aggressive non-Hodgkin, indolent non-Hodgkin and Hodgkin types accounted for 64%, 25% and 11% of the patients, respectively. The patients had received a median (range) of 3 (1-13) lines of treatment prior to trial entry. The median [95% confidence interval] progression-free survival and OS times were 3.0 [1.8-3.6] and 17.8 [12.7-30.4] months, respectively. High-grade toxicity (grade 3 or higher, according to the National Cancer Institute's Common Terminology Criteria for Adverse Events classification) was experienced by 56 of the 133 patients (42%) and was related to the investigational drug in 44 of these cases (79%). No toxicity-related deaths occurred. Dose-limiting toxicity (DLT) was encountered in 11 (9%) of the 116 evaluable patients. High-grade toxicity occurred during the DLT period for 34 of the 56 patients (61%) and after the DLT period in the remaining 22 (39%). The main prognostic factors for poor OS were the histological type (i.e. tumour aggressiveness), an elevated serum LDH level, and a low serum albumin level. Early withdrawal from a trial was correlated with the performance status score, the histological type and the serum LDH level. The overall objective response and disease control rates were 24% and 57%, respectively. Conclusion Performance status, LDH, albumin and histological type (tumour aggressiveness) appear to be the most relevant prognostic factors for enrolling Phase I participants with relapsed or refractory lymphoma. 39% of the patients experienced a first high-grade toxic event after the dose-limiting toxicity period, suggesting that the conventional concept of dose-limiting toxicity (designed for chemotherapy) should be redefined in the era of modern cancer therapies.
Collapse
|
57
|
Brédart A, Bodson S, Le Tourneau C, Flahault C, Bonnetain F, Beaudeau A, Coquan E, Dolbeault S, Paoletti X. Patients' perceived tolerance of side effects in phase I cancer clinical trials: A qualitative study. Eur J Cancer Care (Engl) 2016; 26. [PMID: 27734561 DOI: 10.1111/ecc.12596] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/14/2016] [Indexed: 12/18/2022]
Abstract
This qualitative study aimed to explore cancer patients' perceived tolerance of side effects in phase I drug trials. Patients with solid tumours receiving molecularly targeted agents with/without chemotherapy were eligible for inclusion. In-depth semi-structured interviews were carried out with 17 patients with a median [range] age of 63 [41-72] years. Treatment was discontinued in seven patients. Verbatim transcripts of the audio-taped interviews were analysed using a constructivist grounded theory approach. Four conceptual categories emerged from data analysis, labelled "suffering from side effects" comprising a range of symptoms, psychosocial or role disturbances; "striving to cope with side effects" reflecting psychological strategies for managing side effects; "hoping" reflecting expectations about treatment efficacy and relief from side effects; and "appraisal of care." Among patients remaining in the trial, treatment was currently perceived as fairly tolerable. For most respondents, whether still in a trial or not, treatment discontinuation could not be justified by the non-tolerance of treatment side effects. These results question the adequacy of patient-perceived tolerance reports to determine an optimal drug dose for phase II trials. Confronted with patients' hopes and inappropriate beliefs, communication is challenging in phase I trials and could benefit from facilitating psychosocial interventions.
Collapse
Affiliation(s)
- A Brédart
- Psycho-Oncology Unit, Institut Curie, Paris, France.,Psychopathology and Health Process Laboratory (EA 4057), Psychology Institute, University Paris Descartes, Boulogne-Billancourt, France
| | - S Bodson
- Psychopathology and Health Process Laboratory (EA 4057), Psychology Institute, University Paris Descartes, Boulogne-Billancourt, France
| | - C Le Tourneau
- Department of Medical Oncology, Institut Curie, Paris & Saint-Cloud, France.,EA7285, Versailles-Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux, France
| | - C Flahault
- Psycho-Oncology Unit, Institut Curie, Paris, France.,Psychopathology and Health Process Laboratory (EA 4057), Psychology Institute, University Paris Descartes, Boulogne-Billancourt, France
| | - F Bonnetain
- Methodology and Quality of Life in Oncology Unit (EA 3181) & Quality of Life and Cancer Clinical Research Platform, CHU Besançon, France
| | - A Beaudeau
- Psycho-Oncology Unit, Institut Curie, Paris, France
| | - E Coquan
- Department of Medical Oncology, Institut Curie, Paris & Saint-Cloud, France.,EA7285, Versailles-Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux, France
| | - S Dolbeault
- Psycho-Oncology Unit, Institut Curie, Paris, France.,Inserm, U 669, Paris, France
| | - X Paoletti
- Gustave Roussy Cancer Campus, Villejuif, France
| |
Collapse
|
58
|
Sharma RA, Plummer R, Stock JK, Greenhalgh TA, Ataman O, Kelly S, Clay R, Adams RA, Baird RD, Billingham L, Brown SR, Buckland S, Bulbeck H, Chalmers AJ, Clack G, Cranston AN, Damstrup L, Ferraldeschi R, Forster MD, Golec J, Hagan RM, Hall E, Hanauske AR, Harrington KJ, Haswell T, Hawkins MA, Illidge T, Jones H, Kennedy AS, McDonald F, Melcher T, O'Connor JPB, Pollard JR, Saunders MP, Sebag-Montefiore D, Smitt M, Staffurth J, Stratford IJ, Wedge SR. Clinical development of new drug-radiotherapy combinations. Nat Rev Clin Oncol 2016; 13:627-42. [PMID: 27245279 DOI: 10.1038/nrclinonc.2016.79] [Citation(s) in RCA: 233] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In countries with the best cancer outcomes, approximately 60% of patients receive radiotherapy as part of their treatment, which is one of the most cost-effective cancer treatments. Notably, around 40% of cancer cures include the use of radiotherapy, either as a single modality or combined with other treatments. Radiotherapy can provide enormous benefit to patients with cancer. In the past decade, significant technical advances, such as image-guided radiotherapy, intensity-modulated radiotherapy, stereotactic radiotherapy, and proton therapy enable higher doses of radiotherapy to be delivered to the tumour with significantly lower doses to normal surrounding tissues. However, apart from the combination of traditional cytotoxic chemotherapy with radiotherapy, little progress has been made in identifying and defining optimal targeted therapy and radiotherapy combinations to improve the efficacy of cancer treatment. The National Cancer Research Institute Clinical and Translational Radiotherapy Research Working Group (CTRad) formed a Joint Working Group with representatives from academia, industry, patient groups and regulatory bodies to address this lack of progress and to publish recommendations for future clinical research. Herein, we highlight the Working Group's consensus recommendations to increase the number of novel drugs being successfully registered in combination with radiotherapy to improve clinical outcomes for patients with cancer.
Collapse
Affiliation(s)
- Ricky A Sharma
- UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Martin D Forster
- UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
| | - Julian Golec
- Vertex Pharmaceuticals (Europe) Ltd, Abingdon, UK
| | | | - Emma Hall
- The Institute of Cancer Research/The Royal Marsden NIHR Biomedical Research Centre, London, UK
| | | | - Kevin J Harrington
- The Institute of Cancer Research/The Royal Marsden NIHR Biomedical Research Centre, London, UK
| | | | | | | | | | | | - Fiona McDonald
- The Institute of Cancer Research/The Royal Marsden NIHR Biomedical Research Centre, London, UK
| | | | | | | | | | | | | | - John Staffurth
- Cardiff University and Velindre Cancer Centre, Cardiff, UK
| | | | | |
Collapse
|
59
|
Yin J, Qin R, Ezzalfani M, Sargent DJ, Mandrekar SJ. A Bayesian dose-finding design incorporating toxicity data from multiple treatment cycles. Stat Med 2016; 36:67-80. [PMID: 27633877 DOI: 10.1002/sim.7134] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 08/16/2016] [Accepted: 08/25/2016] [Indexed: 01/05/2023]
Abstract
Phase I oncology trials are designed to identify a safe dose with an acceptable toxicity profile. The dose is typically determined based on the probability of severe toxicity observed during the first treatment cycle, although patients continue to receive treatment for multiple cycles. In addition, the toxicity data from multiple types and grades are typically summarized into a single binary outcome of dose-limiting toxicity. A novel endpoint, the total toxicity profile, was previously developed to account for the multiple toxicity types and grades. In this work, we propose to account for longitudinal repeated measures of total toxicity profile over multiple treatment cycles, accounting for cumulative toxicity during dosing-finding. A linear mixed model was utilized in the Bayesian framework, with addition of Bayesian risk functions for decision-making in dose assignment. The performance of this design is evaluated using simulation studies and compared with the previously proposed quasi-likelihood continual reassessment method (QLCRM) design. Twelve clinical scenarios incorporating four different locations of maximum tolerated dose and three different time trends (decreasing, increasing, and no effect) were investigated. The proposed repeated measures design was comparable with the QLCRM when only cycle 1 data were utilized in dose-finding; however, it demonstrated an improvement over the QLCRM when data from multiple cycles were used across all scenarios. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Jun Yin
- Department of Health Sciences Research, Mayo Clinic, 55905, Rochester, MN, U.S.A
| | - Rui Qin
- Department of Health Sciences Research, Mayo Clinic, 55905, Rochester, MN, U.S.A
| | - Monia Ezzalfani
- Biostatistics Department, Institut Gustave-Roussy, Villejuif, France
| | - Daniel J Sargent
- Department of Health Sciences Research, Mayo Clinic, 55905, Rochester, MN, U.S.A
| | - Sumithra J Mandrekar
- Department of Health Sciences Research, Mayo Clinic, 55905, Rochester, MN, U.S.A
| |
Collapse
|
60
|
Lin R, Yin G. Nonparametric overdose control with late-onset toxicity in phase I clinical trials. Biostatistics 2016; 18:180-194. [DOI: 10.1093/biostatistics/kxw038] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Revised: 07/08/2016] [Accepted: 07/11/2016] [Indexed: 11/12/2022] Open
|
61
|
Wong HH, Barton C, Acton G, McLeod R, Halford S. Trends in the characteristics, dose-limiting toxicities and efficacy of phase I oncology trials: The Cancer Research UK experience. Eur J Cancer 2016; 66:9-16. [PMID: 27514008 DOI: 10.1016/j.ejca.2016.07.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Revised: 06/28/2016] [Accepted: 07/04/2016] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Phase I oncology trials have evolved over the years, and these changes could have implications for future studies and patients. METHODS Adult trials sponsored by Cancer Research UK Centre for Drug Development between 1995 and 2013 were analysed. Forty-nine trials were divided into two groups based on the starting date for recruitment: 1995-2003 (24 trials, n = 603) and 2004-2013 (25 trials, n = 750) for comparative purposes. RESULTS From 1995-2003 to 2004-2013, there was a shift towards studying non-cytotoxic agents that are administered orally. In later trials, patients tended to have better performance status, were older, had greater disease burden, and were more likely to have received prior treatment. In 2004-2013, wider variety of dose escalation designs were used, and studies were more likely to be multicentre, target/disease specific, conducted in first-/any-line setting and to require tumour biopsy. The overall incidence of dose-limiting toxicities (DLTs) was unchanged (10.9%; risk of death 0.4%), but DLTs such as neuropathy, stomatitis and thrombocytopaenia were less frequent in the more recent trials, while elevated liver enzymes were more frequent. Non-classical DLTs emerged in the later trials, including hypertension, hypophosphataemia, cardiac and ophthalmic toxicities. Disease control rate (DCR) increased from 27.9% (1995-2003) to 36.0% (2004-2013; P = 0.0033) due to higher rates of disease stabilisation. CONCLUSION Changes in trial designs, therapeutic agents, patient characteristics and DLTs were observed. Although the nature of DLTs changed, the incidence was similar in the two time periods and DCR improved, suggesting that the benefit-risk balance for patients participating in early-phase trials remains acceptable.
Collapse
Affiliation(s)
- Han Hsi Wong
- Cancer Research UK Centre for Drug Development, Angel Building, 407 St. John Street, London EC1V 4AD, UK; Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's Hospital, Box 193, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0QQ, UK.
| | - Claire Barton
- Cancer Research UK Centre for Drug Development, Angel Building, 407 St. John Street, London EC1V 4AD, UK
| | - Gary Acton
- Cancer Research UK Centre for Drug Development, Angel Building, 407 St. John Street, London EC1V 4AD, UK
| | - Robert McLeod
- Cancer Research UK Centre for Drug Development, Angel Building, 407 St. John Street, London EC1V 4AD, UK
| | - Sarah Halford
- Cancer Research UK Centre for Drug Development, Angel Building, 407 St. John Street, London EC1V 4AD, UK
| |
Collapse
|
62
|
Narayan V, Mamtani R, Keefe S, Guzzo T, Malkowicz SB, Vaughn DJ. Cisplatin, Gemcitabine, and Lapatinib as Neoadjuvant Therapy for Muscle-Invasive Bladder Cancer. Cancer Res Treat 2016; 48:1084-91. [PMID: 26639198 PMCID: PMC4946374 DOI: 10.4143/crt.2015.405] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 11/09/2015] [Indexed: 01/19/2023] Open
Abstract
PURPOSE We sought to investigate the safety and efficacy of gemcitabine, cisplatin, and lapatinib (GCL) as neoadjuvant therapy in patients with muscle-invasive bladder cancer (MIBC) planned for radical cystectomy. MATERIALS AND METHODS Four cycles of GCL were administered as neoadjuvant therapy for patients with MIBC. Although initially designed as a phase II efficacy study with a primary endpoint of pathologic complete response at the time of radical cystectomy, the dose selected for investigation proved excessively toxic. A total of six patients were enrolled. RESULTS The initial four patients received gemcitabine 1,000 mg/m(2) intravenously on days 1 and 8 and cisplatin 70 mg/m(2) intravenously on day 1 of each 21-day treatment cycle. Lapatinib was administered as 1,000 mg orally daily starting one week prior to the initiation of cycle 1 of gemcitabine and cisplatin (GC) and continuing until the completion of cycle 4 of GC. These initial doses were poorly tolerated, and the final two enrolled patients received a reduced lapatinib dose of 750 mg orally daily. However, reduction of the lapatinib dose did not result in improved tolerance or drug-delivery, and the trial was terminated early due to excessive toxicity. Grade 3/4 toxicities included diarrhea (33%), nausea/vomiting (33%), and thrombocytopenia (33%). CONCLUSION The addition of lapatinib to GC as neoadjuvant therapy for MIBC was limited by excessive treatment-related toxicity. These findings highlight the importance of thorough dose-escalation investigation of combination therapies prior to evaluation in the neoadjuvant setting, as well as the limitations of determination of maximum tolerated dose for novel targeted combination regimens.
Collapse
Affiliation(s)
- Vivek Narayan
- Division of Medical Oncology, Hospital of the University of Pennsylvania, Abramson Cancer Center, Philadelphia, PA, USA
| | - Ronac Mamtani
- Division of Medical Oncology, Hospital of the University of Pennsylvania, Abramson Cancer Center, Philadelphia, PA, USA
| | - Stephen Keefe
- Division of Medical Oncology, Hospital of the University of Pennsylvania, Abramson Cancer Center, Philadelphia, PA, USA
| | - Thomas Guzzo
- Department of Urology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - S. Bruce Malkowicz
- Department of Urology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - David J. Vaughn
- Division of Medical Oncology, Hospital of the University of Pennsylvania, Abramson Cancer Center, Philadelphia, PA, USA
| |
Collapse
|
63
|
Assenat E, Azria D, Mollevi C, Guimbaud R, Tubiana-Mathieu N, Smith D, Delord JP, Samalin E, Portales F, Larbouret C, Robert B, Bibeau F, Bleuse JP, Crapez E, Ychou M, Pèlegrin A. Dual targeting of HER1/EGFR and HER2 with cetuximab and trastuzumab in patients with metastatic pancreatic cancer after gemcitabine failure: results of the "THERAPY"phase 1-2 trial. Oncotarget 2016; 6:12796-808. [PMID: 25918250 PMCID: PMC4494975 DOI: 10.18632/oncotarget.3473] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 01/22/2015] [Indexed: 12/12/2022] Open
Abstract
To improve treatment efficacy, we decided to simultaneously target HER1 and HER2 with trastuzumab and cetuximab. Following promising preclinical results, we conducted a phase 1-2 trial in advanced pancreatic cancer patients after first-line gemcitabine-based chemotherapy failure. In this single-arm, non-randomized, multicenter trial, patients received weekly cetuximab (400mg/m², then 250mg/m²). They were sequentially included in two trastuzumab dose levels: 3.0 or 4.0mg/kg, then 1.5 or 2.0mg/kg/weekly. Endpoints were the objective response rate, safety, progression-free (PFS) and overall survival (OS). During phase 1 (n=10 patients), toxicities were evenly distributed except for skin toxicities that frequently caused compliance issues. The higher dose level was defined as the trastuzumab recommended dose. During phase 2 (n=39 patients), toxicities were mainly cutaneous reactions and asthenia. No objective response was observed. Nine patients were stabilized but arrested treatment due to toxicity. Median PFS was 1.8 months (95%CI: 1.7-2.0 months) and median OS was 4.6 months (95%CI: 2.7–6.6 months). Both were positively correlated with skin toxicity severity (P=0.027 and P=0.001, respectively). Conventional phase 1 dose-escalation schedules are unsuitable for targeted therapies because most cutaneous toxicities are not considered dose-limiting toxicities. The compliance issues caused by skin toxicities were particularly detrimental because of the toxicity-response correlation.
Collapse
Affiliation(s)
- Eric Assenat
- Centre Hospitalier Régional Universitaire (CHU) de Montpellier, Montpellier, France.,Institut Régional du Cancer de Montpellier (ICM)-Val d'Aurelle, Montpellier, France
| | - David Azria
- Institut Régional du Cancer de Montpellier (ICM)-Val d'Aurelle, Montpellier, France.,IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France.,INSERM, U896, Montpellier, France.,Université Montpellier 1, Montpellier, France.,Institut Régional du Cancer de Montpellier, ICM, Montpellier, France
| | - Caroline Mollevi
- Institut Régional du Cancer de Montpellier (ICM)-Val d'Aurelle, Montpellier, France
| | - Rosine Guimbaud
- Centre Hospitalier Universitaire (CHU) de Toulouse, TSA, Toulouse Cedex, France
| | | | - Denis Smith
- Centre Hospitalier Universitaire (CHU) de Bordeaux, Talence Cedex, France
| | | | - Emmanuelle Samalin
- Institut Régional du Cancer de Montpellier (ICM)-Val d'Aurelle, Montpellier, France
| | - Fabienne Portales
- Institut Régional du Cancer de Montpellier (ICM)-Val d'Aurelle, Montpellier, France
| | - Christel Larbouret
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France.,INSERM, U896, Montpellier, France.,Université Montpellier 1, Montpellier, France.,Institut Régional du Cancer de Montpellier, ICM, Montpellier, France
| | - Bruno Robert
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France.,INSERM, U896, Montpellier, France.,Université Montpellier 1, Montpellier, France.,Institut Régional du Cancer de Montpellier, ICM, Montpellier, France
| | - Frédéric Bibeau
- Institut Régional du Cancer de Montpellier (ICM)-Val d'Aurelle, Montpellier, France
| | - Jean-Pierre Bleuse
- Institut Régional du Cancer de Montpellier (ICM)-Val d'Aurelle, Montpellier, France
| | - Evelyne Crapez
- Institut Régional du Cancer de Montpellier (ICM)-Val d'Aurelle, Montpellier, France
| | - Marc Ychou
- Centre Hospitalier Régional Universitaire (CHU) de Montpellier, Montpellier, France.,Institut Régional du Cancer de Montpellier (ICM)-Val d'Aurelle, Montpellier, France.,IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France.,INSERM, U896, Montpellier, France.,Université Montpellier 1, Montpellier, France.,Institut Régional du Cancer de Montpellier, ICM, Montpellier, France
| | - André Pèlegrin
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France.,INSERM, U896, Montpellier, France.,Université Montpellier 1, Montpellier, France.,Institut Régional du Cancer de Montpellier, ICM, Montpellier, France
| |
Collapse
|
64
|
Zheng W, Zhao Y, Lu Y, Miao H, Liu H. A Bayesian three-parameter logistic model for early- and late-onset DLTs in oncology Phase I studies. J Biopharm Stat 2016; 26:339-51. [DOI: 10.1080/10543406.2014.1003433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
65
|
Lee SM, Backenroth D, Cheung YKK, Hershman DL, Vulih D, Anderson B, Ivy P, Minasian L. Case Example of Dose Optimization Using Data From Bortezomib Dose-Finding Clinical Trials. J Clin Oncol 2016; 34:1395-401. [PMID: 26926682 DOI: 10.1200/jco.2015.66.0662] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE The current dose-finding methodology for estimating the maximum tolerated dose of investigational anticancer agents is based on the cytotoxic chemotherapy paradigm. Molecularly targeted agents (MTAs) have different toxicity profiles, which may lead to more long-lasting mild or moderate toxicities as well as to late-onset and cumulative toxicities. Several approved MTAs have been poorly tolerated during long-term administration, leading to postmarketing dose optimization studies to re-evaluate the optimal treatment dose. Using data from completed bortezomib dose-finding trials, we explore its toxicity profile, optimize its dose, and examine the appropriateness of current designs for identifying an optimal dose. PATIENTS AND METHODS We classified the toxicities captured from 481 patients in 14 bortezomib dose-finding studies conducted through the National Cancer Institute Cancer Therapy Evaluation Program, computed the incidence of late-onset toxicities, and compared the incidence of dose-limiting toxicities (DLTs) among groups of patients receiving different doses of bortezomib. RESULTS A total of 13,008 toxicities were captured: 46% of patients' first DLTs and 88% of dose reductions or discontinuations of treatment because of toxicity were observed after the first cycle. Moreover, for the approved dose of 1.3 mg/m(2), the estimated cumulative incidence of DLT was > 50%, and the estimated cumulative incidence of dose reduction or treatment discontinuation because of toxicity was nearly 40%. CONCLUSIONS When considering the entire course of treatment, the approved bortezomib dose exceeds the conventional ceiling DLT rate of 20% to 33%. Retrospective analysis of trial data provides an opportunity for dose optimization of MTAs. Future dose-finding studies of MTAs should take into account late-onset toxicities to ensure that a tolerable dose is identified for future efficacy and comparative trials.
Collapse
Affiliation(s)
- Shing M Lee
- Shing M. Lee, Daniel Backenroth, Ying Kuen Ken Cheung, and Dawn L. Hershman, Columbia University, New York, NY; Diana Vulih and Barry Anderson, Theradex Systems, Princeton, NJ; and Percy Ivy and Lori Minasian, National Cancer Institute, Bethesda, MD.
| | - Daniel Backenroth
- Shing M. Lee, Daniel Backenroth, Ying Kuen Ken Cheung, and Dawn L. Hershman, Columbia University, New York, NY; Diana Vulih and Barry Anderson, Theradex Systems, Princeton, NJ; and Percy Ivy and Lori Minasian, National Cancer Institute, Bethesda, MD
| | - Ying Kuen Ken Cheung
- Shing M. Lee, Daniel Backenroth, Ying Kuen Ken Cheung, and Dawn L. Hershman, Columbia University, New York, NY; Diana Vulih and Barry Anderson, Theradex Systems, Princeton, NJ; and Percy Ivy and Lori Minasian, National Cancer Institute, Bethesda, MD
| | - Dawn L Hershman
- Shing M. Lee, Daniel Backenroth, Ying Kuen Ken Cheung, and Dawn L. Hershman, Columbia University, New York, NY; Diana Vulih and Barry Anderson, Theradex Systems, Princeton, NJ; and Percy Ivy and Lori Minasian, National Cancer Institute, Bethesda, MD
| | - Diana Vulih
- Shing M. Lee, Daniel Backenroth, Ying Kuen Ken Cheung, and Dawn L. Hershman, Columbia University, New York, NY; Diana Vulih and Barry Anderson, Theradex Systems, Princeton, NJ; and Percy Ivy and Lori Minasian, National Cancer Institute, Bethesda, MD
| | - Barry Anderson
- Shing M. Lee, Daniel Backenroth, Ying Kuen Ken Cheung, and Dawn L. Hershman, Columbia University, New York, NY; Diana Vulih and Barry Anderson, Theradex Systems, Princeton, NJ; and Percy Ivy and Lori Minasian, National Cancer Institute, Bethesda, MD
| | - Percy Ivy
- Shing M. Lee, Daniel Backenroth, Ying Kuen Ken Cheung, and Dawn L. Hershman, Columbia University, New York, NY; Diana Vulih and Barry Anderson, Theradex Systems, Princeton, NJ; and Percy Ivy and Lori Minasian, National Cancer Institute, Bethesda, MD
| | - Lori Minasian
- Shing M. Lee, Daniel Backenroth, Ying Kuen Ken Cheung, and Dawn L. Hershman, Columbia University, New York, NY; Diana Vulih and Barry Anderson, Theradex Systems, Princeton, NJ; and Percy Ivy and Lori Minasian, National Cancer Institute, Bethesda, MD
| |
Collapse
|
66
|
Drilon A, Eaton AA, Schindler K, Gounder MM, Spriggs DR, Harris P, Ivy SP, Iasonos A, Lacouture ME, Hyman DM. Beyond the dose-limiting toxicity period: Dermatologic adverse events of patients on phase 1 trials of the Cancer Therapeutics Evaluation Program. Cancer 2016; 122:1228-37. [PMID: 26916138 DOI: 10.1002/cncr.29918] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 12/17/2015] [Accepted: 01/07/2016] [Indexed: 12/13/2022]
Abstract
BACKGROUND Dermatologic adverse events (AEs) can be key determinants of overall drug tolerability and of the maximum tolerated and recommended phase 2 doses in phase 1 trials. The authors present the largest dedicated analysis of dermatologic AEs on phase 1 trials to date. METHODS Data from a prospectively maintained database of patients with solid tumors who were enrolled onto Cancer Therapeutics Evaluation Program (CTEP)-sponsored phase 1 trials of cytotoxic or molecularly targeted agents (MTAs) from 2000 to 2010 were analyzed. Cumulative incidence, site, and type of drug-related dermatologic AEs were described and compared. The timing of worst drug-related dermatologic AEs was summarized. RESULTS In total, 3517 patients with solid tumors and 6165 unique, drug-related dermatologic AEs were analyzed, including 1545 patients on MTA-only trials, 671 on cytotoxic-only trials, and 1392 on combination MTA and cytotoxic trials. Of 1270 patients who had drug-related dermatologic events, the timing of the worst AE was as follows: 743 (cycle 1), 303 (cycle 2), and 224 (cycle 3 or later). Although the cumulative incidence of grade ≥3 drug-related AEs increased to 2.4% by cycle 6, it was only 1.6% at the end of cycle 1. The cumulative incidence of drug-related AEs was highest in patients who received MTA-only therapy (P < .001) and differed by dose level (P < .001). In patients who received MTA-only therapy, drug-related AEs were most common for combination kinase inhibitor-containing therapy (P < .001). CONCLUSIONS A substantial proportion of drug-related dermatologic AEs occur after the traditional dose-limiting toxicity monitoring period of phase 1 clinical trials. Future designs should account for late toxicities.
Collapse
Affiliation(s)
- Alexander Drilon
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Anne A Eaton
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Katja Schindler
- Dermatology Department, Medical University of Vienna, Vienna, Austria
| | - Mrinal M Gounder
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Medicine, Weill Cornell Medical College, New York, New York
| | - David R Spriggs
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Pamela Harris
- Cancer Therapeutics and Evaluation Program, National Cancer Institute, Bethesda, Maryland
| | - S Percy Ivy
- Cancer Therapeutics and Evaluation Program, National Cancer Institute, Bethesda, Maryland
| | - Alexia Iasonos
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Mario E Lacouture
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - David M Hyman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Medicine, Weill Cornell Medical College, New York, New York
| |
Collapse
|
67
|
Doussau A, Geoerger B, Jiménez I, Paoletti X. Innovations for phase I dose-finding designs in pediatric oncology clinical trials. Contemp Clin Trials 2016; 47:217-27. [PMID: 26825023 DOI: 10.1016/j.cct.2016.01.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 01/14/2016] [Accepted: 01/23/2016] [Indexed: 01/24/2023]
Abstract
Phase I oncology clinical trials are designed to identify the optimal dose that will be recommended for phase II trials. In pediatric oncology, the conduct of those trials raises specific challenges, as the disease is rare with limited therapeutic options. In addition, the tolerance profile is known from adult trials. This paper provides a review of the major recent developments in the design of these trials, inspired by the need to cope with the specific challenges of dose finding in cancer pediatric oncology. We reviewed simulation studies comparing designs dedicated to address these challenges. We also reviewed the design used in published dose-finding trials in pediatric oncology over the period 2009-2014. Three main fields of innovation were identified. First, designs that were developed in order to relax the rules for more flexible inclusions. Second, methods to incorporate data emerging from adult studies. Third, designs accounting for toxicity evaluation at repeated cycles in pediatric oncology. In addition to this overview, we propose some further directions for designing pediatric dose-finding trials.
Collapse
Affiliation(s)
- Adelaide Doussau
- National Institutes of Health, Clinical Center, Department of Bioethics, Bethesda, MD, USA.
| | - Birgit Geoerger
- Gustave Roussy, Pediatric and Adolescent Oncology, Villejuif, France; CNRS UMR8203, Univ. Paris-Sud, Univ. Paris-Saclay, Villejuif, France
| | - Irene Jiménez
- Institut Curie, Pediatric, Adolescent and Young Adults Department, Paris, France
| | - Xavier Paoletti
- Gustave Roussy, Biostatistics and Epidemiology unit, Villejuif, France; INSERM U1018, CESP, Univ. Paris-Sud, Univ. Paris-Saclay, Villejuif, France
| |
Collapse
|
68
|
Bruce JY, LoRusso PM, Goncalves PH, Heath EI, Sadowski E, Shalinsky DR, Zhang Y, Traynor AM, Breazna A, Ricart AD, Tortorici M, Liu G. A pharmacodynamically guided dose selection of PF-00337210 in a phase I study in patients with advanced solid tumors. Cancer Chemother Pharmacol 2016; 77:527-38. [PMID: 26791870 DOI: 10.1007/s00280-016-2958-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 01/05/2016] [Indexed: 12/29/2022]
Abstract
PURPOSE PF-00337210 is an oral, highly selective vascular endothelial growth factor receptor (VEGFR) inhibitor. We evaluated a composite of biomarkers in real time to identify the recommended phase 2 dose (RP2D) and preliminary anticancer activity of PF-00337210. PATIENTS AND METHODS Patients (Pts) with advanced cancers were treated once (QD) or twice daily (BID) with escalating doses. Acute effects on tumor perfusion and vascularity were assessed using DCE-MRI, weekly BP readings, soluble VEGFR-2, and hemoglobin levels. RESULTS Forty-six pts were treated with 0.67-9 mg QD and 4-6 mg BID of PF-00337210. Nineteen pts (41%) previously received VEGF/VEGFR inhibitors. Two pts had dose-limiting toxicity (DLT) at 9 mg QD (troponin I increase and hypertension). The MTD at QD dose was 8 mg. Common drug-related adverse events were hypertension, fatigue, proteinuria, and nausea. Hypertension incidence and intensity corresponded with dose, but was well controlled with medication. Two confirmed partial responses and minor regressions (>10 to <30% reduction in target lesions) were noted. Complete DCE-MRI was acquired in 21 pts (20 evaluable for vascular response). Ten pts were vascular responders, including 5/6 pts at BID doses. Greatest modulation of soluble VEGFR-2 was at 6 mg BID. The maximum change from baseline in diastolic BP was higher at BID doses. There were no significant differences for systolic BP and hemoglobin levels. CONCLUSIONS PF-00337210 has profound VEGFR inhibition effects at well-tolerated doses. Antitumor activity and VEGF inhibition effects were observed across BID doses. The RP2D was 6 mg BID.
Collapse
Affiliation(s)
- Justine Yang Bruce
- Wisconsin Institute for Medical Research, University of Wisconsin Carbone Cancer Center, Room 7105, 1111 Highland Avenue, Madison, WI, 53705, USA.
| | - Patricia M LoRusso
- Karmanos Cancer Institute, Wayne State University, 4100 John R Street, Detroit, MI, 48201, USA
| | - Priscila H Goncalves
- Karmanos Cancer Institute, Wayne State University, 4100 John R Street, Detroit, MI, 48201, USA
| | - Elisabeth I Heath
- Karmanos Cancer Institute, Wayne State University, 4100 John R Street, Detroit, MI, 48201, USA
| | - Elizabeth Sadowski
- E3/366 Clinical Science Center, Department of Radiology, University of Wisconsin SMPH, 600 Highland Avenue, Madison, WI, 53792-3252, USA
| | - David R Shalinsky
- Department of Pharmacology, Pfizer, Inc., 235 E. 42nd Street, New York, NY, 10017, USA
| | - Yanwei Zhang
- Department of Statistics, Pfizer Inc., 610 Main Street, Cambridge, MA, 02139, USA
| | - Anne M Traynor
- Wisconsin Institute for Medical Research, University of Wisconsin Carbone Cancer Center, Room 3103, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - Aurora Breazna
- Department of Biostatistics, Pfizer, Inc., 235 E. 42nd Street, New York, NY, 10017, USA
| | - Alejandro D Ricart
- Department of Biotechnology and Oncology Research, Pfizer, Inc., 235 E. 42nd Street, New York, NY, 10017, USA
| | - Michael Tortorici
- Department of Clinical Pharmacology, Pfizer, Inc., 235 E. 42nd Street, New York, NY, 10017, USA
| | - Glenn Liu
- Wisconsin Institute for Medical Research, University of Wisconsin Carbone Cancer Center, Room 7105, 1111 Highland Avenue, Madison, WI, 53705, USA
| |
Collapse
|
69
|
Greystoke A, Chaturvedi A. An introduction to stratified medicine. Drug Discov Today 2015; 20:1409-13. [DOI: 10.1016/j.drudis.2015.07.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 07/13/2015] [Accepted: 07/16/2015] [Indexed: 02/01/2023]
|
70
|
D'Arcangelo M, Margetts J, Greystoke A. The use of circulating biomarkers in early clinical trials in patients with cancer. Biomark Med 2015; 9:1011-23. [PMID: 26441037 DOI: 10.2217/bmm.15.51] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
The development of targeted therapies has changed the approach to early oncological clinical trial design. Identification of patient populations most likely to derive benefit and the biologically effective dose are now as important as determination of the maximum tolerated dose. Completion of the 'pharmacological audit trail' highlights drugs most likely to progress through to license, so resources can be allocated appropriately. Key to the success of this changing model is the validation/qualification of circulating biomarkers. These might provide a readily accessible and dynamic picture of drug effect, tumor response and toxicity with minimum risk to patients. This review article examines circulating biomarkers currently used in early oncological clinical trials. It considers the evidence for their employment, limitations and challenges for future development.
Collapse
Affiliation(s)
- Manolo D'Arcangelo
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, UK.,Northern Centre for Cancer Care, Freeman Hospital, Newcastle upon Tyne, UK
| | - Jane Margetts
- Northern Centre for Cancer Care, Freeman Hospital, Newcastle upon Tyne, UK
| | - Alastair Greystoke
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, UK.,Northern Centre for Cancer Care, Freeman Hospital, Newcastle upon Tyne, UK
| |
Collapse
|
71
|
Dose-limiting toxicity and maximum tolerated dose: still fit for purpose? Lancet Oncol 2015; 16:1287-8. [DOI: 10.1016/s1470-2045(15)00248-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 08/17/2015] [Accepted: 08/18/2015] [Indexed: 11/21/2022]
|
72
|
Colin P, Micallef S, Delattre M, Mancini P, Parent E. Towards using a full spectrum of early clinical trial data: a retrospective analysis to compare potential longitudinal categorical models for molecular targeted therapies in oncology. Stat Med 2015; 34:2999-3016. [PMID: 26059319 DOI: 10.1002/sim.6548] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Revised: 02/09/2015] [Accepted: 05/17/2015] [Indexed: 11/09/2022]
Abstract
Following the pattern of phase I clinical trials for cytotoxic drugs, dose-finding clinical trials in oncology of molecularly targeted agents (MTA) aim at determining the maximum tolerated dose (MTD). In classical phase I clinical trials, MTD is generally defined by the number of patients with short-term major treatment toxicities (usually called dose-limiting toxicities, DLT), occurring during the first cycle of study treatment (e.g. within the first 3weeks of treatment). However, S. Postel-Vinay (2011) highlighted that half of grade 3 to 4 toxicities, usually considered as DLT, occur after the first cycle of MTA treatment. In addition, MTAs could induce other moderate (e.g. grade 2) toxicities which could be taken into account depending on their clinical importance, chronic nature and duration. Ignoring these late toxicities may lead to an underestimation of the drug toxicity and to wrong dose recommendations for phase II and III clinical trials. Some methods have been proposed, such as the time-to-event continuous reassessment method (Cheung 2000 and Mauguen 2011), to take into account the late toxicities. We suggest approaches based on longitudinal models (Doussau 2013). We compare several models for longitudinal data, such as transitional or marginal models, to take into account all relevant toxicities occurring during the entire length of the patient treatment (and not just the events within a predefined short-term time-window). These models allow the statistician to benefit from a larger amount of safety data which could potentially improve that accuracy in MTD assessment.
Collapse
Affiliation(s)
- Pierre Colin
- Sanofi R&D, Biostatistique Oncologie, Vitry-sur-Seine, France.,UMR 518 MIA, AgroParisTech, Paris, 75005, France
| | | | - Maud Delattre
- UMR 518 MIA, AgroParisTech, Paris, 75005, France.,UMR 518 MIA, INRA, Paris, 75005, France
| | - Pierre Mancini
- Sanofi R&D, Biostatistique Oncologie, Vitry-sur-Seine, France
| | - Eric Parent
- UMR 518 MIA, AgroParisTech, Paris, 75005, France.,UMR 518 MIA, INRA, Paris, 75005, France
| |
Collapse
|
73
|
Thanarajasingam G, Hubbard JM, Sloan JA, Grothey A. The Imperative for a New Approach to Toxicity Analysis in Oncology Clinical Trials. J Natl Cancer Inst 2015; 107:djv216. [DOI: 10.1093/jnci/djv216] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 07/09/2015] [Indexed: 11/15/2022] Open
|
74
|
Coupe N, Gupta A, Lord SR. Early phase cancer clinical trials: design, ethics and future directions. Br J Hosp Med (Lond) 2015; 76:409-13. [PMID: 26140560 DOI: 10.12968/hmed.2015.76.7.409] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The main role of early phase clinical trials in cancer is to determine the dose to take forward for future clinical study. However, study design is changing in order to account for the change in focus of drug development toward molecularly targeted agents.
Collapse
Affiliation(s)
| | - Avinash Gupta
- Clinical Research Fellow in Medical Oncology in the Early Phase Trials Unit and the IGF Group, Department of Oncology, University of Oxford, Oxford
| | - Simon R Lord
- Honorary Consultant in Medical Oncology and Senior Clinical Researcher in Experimental Cancer Therapeutics in the Early Phase Trials Unit, Department of Oncology, Churchill Hospital, University of Oxford, Oxford OX3 7LE
| |
Collapse
|
75
|
Lawrence Gould A, Boye ME, Crowther MJ, Ibrahim JG, Quartey G, Micallef S, Bois FY. Joint modeling of survival and longitudinal non-survival data: current methods and issues. Report of the DIA Bayesian joint modeling working group. Stat Med 2015; 34:2181-95. [PMID: 24634327 PMCID: PMC4677775 DOI: 10.1002/sim.6141] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 02/19/2014] [Indexed: 12/25/2022]
Abstract
Explicitly modeling underlying relationships between a survival endpoint and processes that generate longitudinal measured or reported outcomes potentially could improve the efficiency of clinical trials and provide greater insight into the various dimensions of the clinical effect of interventions included in the trials. Various strategies have been proposed for using longitudinal findings to elucidate intervention effects on clinical outcomes such as survival. The application of specifically Bayesian approaches for constructing models that address longitudinal and survival outcomes explicitly has been recently addressed in the literature. We review currently available methods for carrying out joint analyses, including issues of implementation and interpretation, identify software tools that can be used to carry out the necessary calculations, and review applications of the methodology.
Collapse
Affiliation(s)
- A Lawrence Gould
- Merck Research Laboratories, 351 North Sumneytown Pike, North Wales, PA 19454, U.S.A
| | - Mark Ernest Boye
- Eli Lilly, 893 S. Delaware Street, Indianapolis, IN 46285, U.S.A
| | - Michael J Crowther
- Department of Health Sciences, University of Leicester, Adrian Building, University Road, Leicester LE1 7RH, U.K
| | - Joseph G Ibrahim
- Department of Statistics and Operations Research, University of North Carolina, 318 Hanes Hall Chapel Hill, NC 27599, U.S.A
| | | | | | - Frederic Y Bois
- Université de Technologie de Compiègne, Centre de Recherche de Royallieu, 60205 Compiègne Cedex, France
- INERIS/CRD/VIVA/METO, Verneuil en Halatte, France
| |
Collapse
|
76
|
Systematic review and meta-analysis of phase I/II targeted therapy combined with radiotherapy in patients with glioblastoma multiforme: quality of report, toxicity, and survival. J Neurooncol 2015; 123:307-14. [PMID: 25975195 DOI: 10.1007/s11060-015-1802-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 04/05/2015] [Indexed: 10/23/2022]
Abstract
To perform a systematic review and meta-analysis of severe adverse events (SAE) reported in early trials combining molecularly targeted therapies (MTT) with radiotherapy (RT), and to compare them to standard therapy. A summary data meta-analysis was performed and compared to the historical standard. Inclusion criteria were phase I and/or II trials published between 2000 and 2011, with glioblastoma multiforme patients treated with RT and MTT. Pooled incidence rates (IR) of SAE were estimated as well as the pooled median progression-free survival (PFS) and overall survival (OS). Nineteen prospective trials (9 phase I, 1 phase I/II and 9 phase II) out of 29 initially selected were included (n = 755 patients). The exact number of patients who had experienced SAE was mentioned in 37 % of the trials, concerning only 17 % of the patients. Information such as the period during which adverse events were monitored, the planned treatment duration, and late toxicity were not reported in the trials. The pooled IR of overall SAE was 131.2 (95 % CI 88.8-193.7) per 1000 person-months compared to 74.7 (63.6-87.8) for standard therapy (p < 0.01). Significant differences were observed for gastrointestinal events (p = 0.05) and treatment-related deaths (p = 0.02), in favour of standard therapy. No significant difference was observed in PFS and OS. Reporting a summary of toxicity data in early clinical trials should be stringently standardized. The use of MTT with RT compared to standard therapy increased SAE while yielded comparable survival in glioblastoma multiforme patients.
Collapse
|
77
|
Cook N, Hansen AR, Siu LL, Abdul Razak AR. Early phase clinical trials to identify optimal dosing and safety. Mol Oncol 2015; 9:997-1007. [PMID: 25160636 PMCID: PMC4329110 DOI: 10.1016/j.molonc.2014.07.025] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 07/30/2014] [Indexed: 12/13/2022] Open
Abstract
The purpose of early stage clinical trials is to determine the recommended dose and toxicity profile of an investigational agent or multi-drug combination. Molecularly targeted agents (MTAs) and immunotherapies have distinct toxicities from chemotherapies that are often not dose dependent and can lead to chronic and sometimes unpredictable side effects. Therefore utilizing a dose escalation method that has toxicity based endpoints may not be as appropriate for determination of recommended dose, and alternative parameters such as pharmacokinetic or pharmacodynamic outcomes are potentially appealing options. Approaches to enhance safety and optimize dosing include improved preclinical models and assessment, innovative model based design and dose escalation strategies, patient selection, the use of expansion cohorts and extended toxicity assessments. Tailoring the design of phase I trials by adopting new strategies to address the different properties of MTAs is required to enhance the development of these agents. This review will focus on the limitations to safety and dose determination that have occurred in the development of MTAs and immunotherapies. In addition, strategies are proposed to overcome these challenges to develop phase I trials that can more accurately define the recommended dose and identify adverse events.
Collapse
Affiliation(s)
- Natalie Cook
- Princess Margaret Cancer Centre, University Health Network, Division of Medical Oncology and Hematology, Toronto, ON, Canada; Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Aaron R Hansen
- Princess Margaret Cancer Centre, University Health Network, Division of Medical Oncology and Hematology, Toronto, ON, Canada; Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Lillian L Siu
- Princess Margaret Cancer Centre, University Health Network, Division of Medical Oncology and Hematology, Toronto, ON, Canada; Department of Medicine, University of Toronto, Toronto, ON, Canada.
| | - Albiruni R Abdul Razak
- Princess Margaret Cancer Centre, University Health Network, Division of Medical Oncology and Hematology, Toronto, ON, Canada; Department of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
78
|
Abstract
BACKGROUND Concerns have been recognized about the operating characteristics of the standard 3 + 3 dose-escalation design. Various innovative phase 1 trial designs have been proposed to address the issues and new challenges posed by molecularly targeted agents. However, in spite of these proposals, the conventional design is still the most widely utilized. METHODS A review of the literature of phase 1 trials and relevant statistical studies was performed. RESULTS Beyond statistical simulations, sparse clinical data exist to support or refute many of the shortcomings ascribed to the 3 + 3 rule method. Data from phase 1 trials demonstrate that traditional designs identified the correct dose and relevant toxicities with an acceptable level of precision in some instances; however, no single escalation method was proven superior in all circumstances. CONCLUSIONS Design selection should be guided by the principle of slow escalation in the face of toxicity and rapid dose increases in the setting of minimal or no adverse events. When the toxicity of a drug is uncertain or a narrow therapeutic window is suggested from preclinical testing, then a conservative 3 + 3 method is generally appropriate. However, if the therapeutic window is wide and the expected toxicity is low, then rapid escalation with a novel rule- or model-based design should be employed.
Collapse
Affiliation(s)
- Aaron R Hansen
- Drug Development Program, Princess Margaret Cancer Centre, Toronto, Ontario, Canada M5G 2M9.
| | | | | | | |
Collapse
|
79
|
Oh DY, Doi T, Shirao K, Lee KW, Park SR, Chen Y, Yang L, Valota O, Bang YJ. Phase I Study of Axitinib in Combination with Cisplatin and Capecitabine in Patients with Previously Untreated Advanced Gastric Cancer. Cancer Res Treat 2015; 47:687-96. [PMID: 25687867 PMCID: PMC4614203 DOI: 10.4143/crt.2014.225] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 10/15/2014] [Indexed: 12/14/2022] Open
Abstract
PURPOSE This phase I trial evaluated the question of whether the standard starting dose of axitinib could be administered in combination with therapeutic doses of cisplatin/capecitabine in patients with previously untreated advanced gastric cancer, and assessed overall safety, pharmacokinetics, and preliminary antitumor activity of this combination. MATERIALS AND METHODS Patients in dose level (DL) 1 received axitinib 5 mg twice a day (days 1 to 21) with cisplatin 80 mg/m(2) (day 1) and capecitabine 1,000 mg/m(2) twice a day (days 1 to 14) in 21-day cycles. Maximum tolerated dose (MTD) was the highest dose at which ≤ 30% of the first 12 patients experienced a dose-limiting toxicity (DLT) during cycle 1. Ten additional patients were enrolled and treated at the MTD in order to obtain additional safety and pharmacokinetic data. RESULTS Three DLTs occurred during cycle 1 in three (25%) of the first 12 patients: ruptured abdominal aortic aneurysm, acute renal failure, and > 5 consecutive days of missed axitinib due to thrombocytopenia. DL1 was established as the MTD, since higher DL cohorts were not planned. Common grade 3/4 non-hematologic adverse events in 22 patients treated at DL1 included hypertension (36.4%) and decreased appetite and stomatitis (18.2% each). Cisplatin/capecitabine slightly increased axitinib exposure; axitinib decreased capecitabine and 5-fluorouracil exposure. Eight patients (36.4%) each had partial response or stable disease. Median response duration was 9.1 months; median progression-free survival was 3.8 months. CONCLUSION In patients with advanced gastric cancer, standard doses of axitinib plus therapeutic doses of cisplatin and capecitabine could be administered in combination. Adverse events were manageable.
Collapse
Affiliation(s)
- Do-Youn Oh
- Department of Internal Medicine, Seoul National University Hospital and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Toshihiko Doi
- Division of Gastroenterology, National Cancer Center Hospital East, Chiba, Japan
| | - Kuniaki Shirao
- Department of Medical Oncology, Oita University Faculty of Medicine, Yufu, Japan
| | - Keun-Wook Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Sook Ryun Park
- Department of Internal Medicine, Research Institute and Hospital, National Cancer Center, Goyang, Korea
| | | | | | | | - Yung-Jue Bang
- Department of Internal Medicine, Seoul National University Hospital and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
80
|
Toloi DDA, Jardim DLF, Hoff PMG, Riechelmann RSP. Phase I trials of antitumour agents: fundamental concepts. Ecancermedicalscience 2015; 9:501. [PMID: 25729414 PMCID: PMC4335968 DOI: 10.3332/ecancer.2015.501] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Indexed: 01/25/2023] Open
Abstract
Phase I trials are an important step in the development of new drugs. Because of the advancing knowledge of cancer's molecular biology, these trials offer an important platform for the development of new agents and also for patient treatment. Therefore, comprehension of their peculiar terminology and methodology are increasingly important. Our objectives were to review the fundamental concepts of phase I designs and to critically contextualise this type of study as a therapeutic option for patients with refractory cancer.
Collapse
|
81
|
Spicer J, Baird R, Suder A, Cresti N, Corbacho JG, Hogarth L, Frenkel E, Matsumoto S, Kawabata I, Donaldson K, Posner J, Sarker D, Jodrell D, Plummer R. Phase 1 dose-escalation study of S-222611, an oral reversible dual tyrosine kinase inhibitor of EGFR and HER2, in patients with solid tumours. Eur J Cancer 2015; 51:137-45. [PMID: 25434923 DOI: 10.1016/j.ejca.2014.11.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 10/27/2014] [Accepted: 11/10/2014] [Indexed: 11/28/2022]
Abstract
BACKGROUND S-222611 is a reversible inhibitor of EGFR, HER2 and HER4 with preclinical activity in models expressing these proteins. We have performed a Phase 1 study to determine safety, maximum tolerated dose (MTD), pharmacokinetic profile (PK) and efficacy in patients with solid tumours expressing EGFR or HER2. PATIENTS AND METHODS Subjects had advanced tumours not suitable for standard treatment, expressing EGFR or HER2, and/or with amplified HER2. Daily oral doses of S-222611 were escalated from 100mg to 1600 mg. Full plasma concentration profiles for drug and metabolites were obtained. RESULTS 33 patients received S-222611. It was well tolerated, and the most common toxicities, almost all mild (grade 1 or 2), were diarrhoea, fatigue, rash and nausea. Only two dose-limiting toxicities occurred (diarrhoea and rash), which resolved on interruption. MTD was not reached. Plasma exposure increased with dose up to 800 mg, exceeding levels eliciting pre-clinical responses. The plasma terminal half-life was more than 24h, supporting once daily dosing. Responses were seen over a wide range of doses in oesophageal, breast and renal tumours, including a complete clinical response in a patient with HER2-positive breast carcinoma previously treated with lapatinib and trastuzumab. Four patients have remained on treatment for more than 12 months. Downregulation of pHER3 was seen in paired tumour biopsies from a responding patient. CONCLUSIONS Continuous daily oral S-222611 is well tolerated, modulates oncogenic signalling, and has significant antitumour activity. The recommended Phase 2 dose, based on PK and efficacy, is 800 mg/day.
Collapse
Affiliation(s)
- J Spicer
- King's College London, Guy's Hospital, London, UK.
| | - R Baird
- University of Cambridge, Department of Oncology, Cambridge, UK
| | - A Suder
- King's College London, Guy's Hospital, London, UK
| | - N Cresti
- Northern Centre for Cancer Care, Newcastle upon Tyne, UK
| | | | - L Hogarth
- Northern Centre for Cancer Care, Newcastle upon Tyne, UK
| | - E Frenkel
- University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | | | | | - J Posner
- Shionogi & Co. Ltd., Osaka, Japan
| | - D Sarker
- King's College London, Guy's Hospital, London, UK
| | - D Jodrell
- University of Cambridge, Department of Oncology, Cambridge, UK
| | - R Plummer
- Northern Centre for Cancer Care, Newcastle upon Tyne, UK
| |
Collapse
|
82
|
Moreno García V, Olmos D, Gomez-Roca C, Cassier PA, Morales-Barrera R, Del Conte G, Gallerani E, Brunetto AT, Schöffski P, Marsoni S, Schellens JH, Penel N, Voest E, Evans J, Plummer R, Wilson RH, Soria JC, Tabernero J, Verweij J, Kaye SB. Dose–Response Relationship in Phase I Clinical Trials: A European Drug Development Network (EDDN) Collaboration Study. Clin Cancer Res 2014; 20:5663-71. [DOI: 10.1158/1078-0432.ccr-14-0719] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
83
|
Towards new methods for the determination of dose limiting toxicities and the assessment of the recommended dose for further studies of molecularly targeted agents – Dose-Limiting Toxicity and Toxicity Assessment Recommendation Group for Early Trials of Targeted therapies, an European Organisation for Research and Treatment of Cancer-led study. Eur J Cancer 2014; 50:2040-9. [DOI: 10.1016/j.ejca.2014.04.031] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 04/04/2014] [Indexed: 11/22/2022]
|
84
|
Paoletti X, Le Tourneau C, Verweij J, Siu LL, Seymour L, Postel-Vinay S, Collette L, Rizzo E, Ivy P, Olmos D, Massard C, Lacombe D, Kaye SB, Soria JC. Defining dose-limiting toxicity for phase 1 trials of molecularly targeted agents: Results of a DLT-TARGETT international survey. Eur J Cancer 2014; 50:2050-6. [DOI: 10.1016/j.ejca.2014.04.030] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 04/04/2014] [Indexed: 10/25/2022]
|
85
|
Bedard PL, Siu LL. Tilting the Balance of Dose Modification for Oral Anticancer Drugs? J Clin Oncol 2014; 32:1537-9. [DOI: 10.1200/jco.2014.55.2372] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Philippe L. Bedard
- Princess Margaret Cancer Centre, University Health Network; University of Toronto, Toronto, ON, Canada
| | - Lillian L. Siu
- Princess Margaret Cancer Centre, University Health Network; University of Toronto, Toronto, ON, Canada
| |
Collapse
|
86
|
Sinclair K, Whitehead A. A Bayesian approach to dose-finding studies for cancer therapies: incorporating later cycles of therapy. Stat Med 2014; 33:2665-80. [PMID: 24590816 DOI: 10.1002/sim.6132] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Revised: 02/06/2014] [Accepted: 02/12/2014] [Indexed: 11/07/2022]
Abstract
We consider phase I dose-finding studies for cytotoxic drugs in cancer, where the objective is identification of a target dose (TD100δ ) associated with the probability δ of a dose-limiting toxicity (DLT). Previous authors have presented a design utilising a Bayesian decision procedure based on a logistic regression model to describe the relationship between dose and the risk of a DLT (LRDP). A cautious prior, chosen to ensure that the first cohort of patients are given the lowest dose, is combined with binary observations of DLTs to update model parameters and choose a safe dose for the next cohort. This process continues with each new cohort of patients. Typically, only DLTs occurring in the first treatment cycle are included. To incorporate data from later cycles, a new Bayesian decision procedure based on an interval-censored survival model (ICSDP) has been developed. This models the probability that the first DLT occurs in each specific cycle via the probability of a DLT during a specific cycle, conditional on having no DLT in any previous cycle. The second cohort of patients start after responses have been obtained from the first cycle of the first cohort, and subsequently, dose selection for each new cohort is based on DLTs observed across all completed cycles for all patients. A simulation study comparing the ICSDP and LRDP showed that the ICSDP induces faster updating of the current estimate of the target dose, leading to shorter trials and fewer patients, whilst keeping the same level of accuracy.
Collapse
Affiliation(s)
- Karen Sinclair
- Medical and Pharmaceutical Statistics Research Unit, Department of Mathematics and Statistics, Lancaster University, U.K
| | | |
Collapse
|
87
|
Jardim DL, Hess KR, Lorusso P, Kurzrock R, Hong DS. Predictive value of phase I trials for safety in later trials and final approved dose: analysis of 61 approved cancer drugs. Clin Cancer Res 2014; 20:281-8. [PMID: 24190980 DOI: 10.1158/1078-0432.ccr-13-2103] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Phase I trials use a small number of patients to define a maximum tolerated dose (MTD) and the safety of new agents. We compared data from phase I and registration trials to determine whether early trials predicted later safety and final dose. We searched the U.S. Food and Drug Administration (FDA) website for drugs approved in nonpediatric cancers (January 1990-October 2012). The recommended phase II dose (R2PD) and toxicities from phase I were compared with doses and safety in later trials. In 62 of 85 (73%) matched trials, the dose from the later trial was within 20% of the RP2D. In a multivariable analysis, phase I trials of targeted agents were less predictive of the final approved dose (OR, 0.2 for adopting ± 20% of the RP2D for targeted vs. other classes; P = 0.025). Of the 530 clinically relevant toxicities in later trials, 70% (n = 374) were described in phase I. A significant relationship (P = 0.0032) between increasing the number of patients in phase I (up to 60) and the ability to describe future clinically relevant toxicities was observed. Among 28,505 patients in later trials, the death rate that was related to drug was 1.41%. In conclusion, dosing based on phase I trials was associated with a low toxicity-related death rate in later trials. The ability to predict relevant toxicities correlates with the number of patients on the initial phase I trial. The final dose approved was within 20% of the RP2D in 73% of assessed trials.
Collapse
Affiliation(s)
- Denis L Jardim
- Authors' Affiliations: Phase I Clinical Trials Program, Department of Investigational Cancer Therapeutics; Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas; Department of Clinical Medicine, Hemocentro da Unicamp, University of Campinas, Sao Paulo, Brazil; Karmanos Cancer Institute/Wayne State University, Detroit, Michigan; and Department of Medicine, University of California, San Diego, La Jolla, California
| | | | | | | | | |
Collapse
|
88
|
Liu S, Yin G, Yuan Y. BAYESIAN DATA AUGMENTATION DOSE FINDING WITH CONTINUAL REASSESSMENT METHOD AND DELAYED TOXICITY. Ann Appl Stat 2013; 7:1837-2457. [PMID: 24707327 DOI: 10.1214/13-aoas661] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
A major practical impediment when implementing adaptive dose-finding designs is that the toxicity outcome used by the decision rules may not be observed shortly after the initiation of the treatment. To address this issue, we propose the data augmentation continual re-assessment method (DA-CRM) for dose finding. By naturally treating the unobserved toxicities as missing data, we show that such missing data are nonignorable in the sense that the missingness depends on the unobserved outcomes. The Bayesian data augmentation approach is used to sample both the missing data and model parameters from their posterior full conditional distributions. We evaluate the performance of the DA-CRM through extensive simulation studies, and also compare it with other existing methods. The results show that the proposed design satisfactorily resolves the issues related to late-onset toxicities and possesses desirable operating characteristics: treating patients more safely, and also selecting the maximum tolerated dose with a higher probability. The new DA-CRM is illustrated with two phase I cancer clinical trials.
Collapse
|
89
|
Kelley RK, Hwang J, Magbanua MJM, Watt L, Beumer JH, Christner SM, Baruchel S, Wu B, Fong L, Yeh BM, Moore AP, Ko AH, Korn WM, Rajpal S, Park JW, Tempero MA, Venook AP, Bergsland EK. A phase 1 trial of imatinib, bevacizumab, and metronomic cyclophosphamide in advanced colorectal cancer. Br J Cancer 2013; 109:1725-34. [PMID: 24022191 PMCID: PMC3790192 DOI: 10.1038/bjc.2013.553] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 07/14/2013] [Accepted: 08/19/2013] [Indexed: 02/08/2023] Open
Abstract
Background: This phase 1 clinical trial was conducted to determine the safety, maximum-tolerated dose (MTD), and pharmacokinetics of imatinib, bevacizumab, and metronomic cyclophosphamide in patients with advanced colorectal cancer (CRC). Methods: Patients with refractory stage IV CRC were treated with bevacizumab 5 mg kg−1 i.v. every 2 weeks (fixed dose) plus oral cyclophosphamide q.d. and imatinib q.d. or b.i.d. in 28-day cycles with 3+3 dose escalation. Response was assessed every two cycles. Pharmacokinetics of imatinib and cyclophosphamide and circulating tumour, endothelial, and immune cell subsets were measured. Results: Thirty-five patients were enrolled. Maximum-tolerated doses were cyclophosphamide 50 mg q.d., imatinib 400 mg q.d., and bevacizumab 5 mg kg−1 i.v. every 2 weeks. Dose-limiting toxicities (DLTs) included nausea/vomiting, neutropaenia, hyponatraemia, fistula, and haematuria. The DLT window required expansion to 42 days (1.5 cycles) to capture delayed toxicities. Imatinib exposure increased insignificantly after adding cyclophosphamide. Seven patients (20%) experienced stable disease for >6 months. Circulating tumour, endothelial, or immune cells were not associated with progression-free survival. Conclusion: The combination of metronomic cyclophosphamide, imatinib, and bevacizumab is safe and tolerable without significant drug interactions. A subset of patients experienced prolonged stable disease independent of dose level.
Collapse
Affiliation(s)
- R K Kelley
- Department of Medicine, University of California, San Francisco, Helen Diller Family Comprehensive Cancer Center, 1600 Divisadero Street, Box 1700, San Francisco, CA 94143, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
90
|
Doussau A, Thiébaut R, Paoletti X. Dose-finding design using mixed-effect proportional odds model for longitudinal graded toxicity data in phase I oncology clinical trials. Stat Med 2013; 32:5430-47. [PMID: 24018535 DOI: 10.1002/sim.5960] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Revised: 06/20/2013] [Accepted: 08/05/2013] [Indexed: 11/06/2022]
Abstract
Phase I oncology clinical trials are designed to identify the optimal dose that will be recommended for phase II trials. This dose is typically defined as the dose associated with a certain probability of severe toxicity during the first cycle of treatment, although toxicity is repeatedly measured over cycles on an ordinal scale. We propose a new adaptive dose-finding design using longitudinal measurements of ordinal toxic adverse events, with proportional odds mixed-effect models. Likelihood-based inference is implemented. The optimal dose is then the dose producing a target rate of severe toxicity per cycle. This model can also be used to identify cumulative or late toxicities. The performances of this approach were compared with those of the continual reassessment method in a simulation study. Operating characteristics were evaluated in terms of correct identification of the target dose, distribution of the doses allocated and power to detect trends in the risk of toxicities over time. This approach was also used to reanalyse data from a phase I oncology trial. Use of a proportional odds mixed-effect model appears to be feasible in phase I dose-finding trials, increases the ability of selecting the correct dose and provides a tool to detect cumulative effects.
Collapse
Affiliation(s)
- Adélaïde Doussau
- INSERM, ISPED, Centre INSERM U897-Epidemiologie-Biostatistique, F-33000 Bordeaux, France; CHU de Bordeaux, Pole de Santé Publique, F-33000 Bordeaux, France; Univ. Bordeaux, ISPED, CIC-EC7, F-33000 Bordeaux, France; INSERM, U900, F-75005 Paris, France
| | | | | |
Collapse
|
91
|
Liu S, Ning J. A Bayesian Dose-finding Design for Drug Combination Trials with Delayed Toxicities. BAYESIAN ANALYSIS 2013; 8:703-722. [PMID: 27924182 PMCID: PMC5136476 DOI: 10.1214/13-ba839] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
We propose a Bayesian adaptive dose-finding design for drug combination trials with delayed toxicity. We model the dose-toxicity relationship using the Finney model, a model widely used in drug-drug interaction studies. The intuitive interpretations of the Finney model facilitate incorporating the available prior dose-toxicity information from single-agent trials into combination trials through prior elicitation. We treat unobserved delayed toxicity outcomes as missing data and handle them using Bayesian data augmentation. We conduct extensive simulation studies to examine the operating characteristics of the proposed method under various practical scenarios. Results show that the proposed design is safe and able to select the target dose combinations with high probabilities.
Collapse
Affiliation(s)
- Suyu Liu
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, TX,
| | - Jing Ning
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, TX,
| |
Collapse
|
92
|
Paoletti X, Geoerger B, Doz F, Baruchel A, Lokiec F, Le Tourneau C. A comparative analysis of paediatric dose-finding trials of molecularly targeted agent with adults’ trials. Eur J Cancer 2013; 49:2392-402. [DOI: 10.1016/j.ejca.2013.02.028] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Revised: 02/01/2013] [Accepted: 02/23/2013] [Indexed: 10/27/2022]
|
93
|
Abstract
We developed a module of the MD Anderson Symptom Inventory (MDASI) for patients with chronic myeloid leukemia (CML). To develop the MDASI-CML, we identified CML-specific symptoms from qualitative interviews with 35 patients. A list of candidate symptoms was reduced by a panel of patients, caregivers, and clinicians to the 13 core MDASI symptom items and 6 CML-specific items; these items were subsequently administered to 30 patients. Cognitive debriefing confirmed that the items were clear, relevant, and easy to use. One additional CML-specific symptom item was added, for a total of 7. The refined MDASI-CML was administered to 152 patients once every 2 weeks for 1 year. The content, concurrent, known-group, and construct validity of the MDASI-CML were evaluated. The internal consistency and test-retest reliabilities of the module were adequate. Longitudinal analysis showed relatively stable symptom severity scores over time. The most severe symptoms were fatigue, drowsiness, disturbed sleep, muscle soreness and cramping, and trouble remembering things. Approximately one-third of the patients who completed the MDASI-CML reported persistent moderate-to-severe symptoms. The MDASI-CML is a valid and reliable symptom assessment instrument that can be used in clinical studies of symptom status in patients with CML.
Collapse
|
94
|
Broniscer A, Baker SD, Wetmore C, Pai Panandiker AS, Huang J, Davidoff AM, Onar-Thomas A, Panetta JC, Chin TK, Merchant TE, Baker JN, Kaste SC, Gajjar A, Stewart CF. Phase I trial, pharmacokinetics, and pharmacodynamics of vandetanib and dasatinib in children with newly diagnosed diffuse intrinsic pontine glioma. Clin Cancer Res 2013; 19:3050-8. [PMID: 23536435 DOI: 10.1158/1078-0432.ccr-13-0306] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE Testing of promising drug combinations is crucial in the treatment of diffuse intrinsic pontine glioma (DIPG). As the VEGF and platelet-derived growth factor (PDGF) pathways are critical in gliomas, we evaluated the safety, maximum tolerated dose (MTD), pharmacokinetics, and pharmacodynamics of vandetanib, a VEGFR-2 inhibitor, combined with dasatinib, a potent PDGFR inhibitor, during and after radiotherapy in children with newly diagnosed DIPG. EXPERIMENTAL DESIGN Dasatinib was started concurrently with radiotherapy. Vandetanib was started 8 days later. We tested increasing doses of vandetanib (65 and 85 mg/m(2) once daily) and dasatinib (65 and 85 mg/m(2) twice daily). Dose-limiting toxicities were evaluated during the first 6 weeks of therapy. Plasma pharmacokinetics was obtained on days 8 and 42 ± 3 in all patients and concomitantly with cerebrospinal fluid (CSF) when possible. Inhibition of targets of dasatinib in peripheral blood mononuclear cells (PBMC) was evaluated. RESULTS Twenty-five patients were treated. Treatment was well tolerated. The median duration of treatment was 184 days. Diarrhea was the most significant toxicity. Three patients experienced substantial myelosuppression. The steady-state plasma pharmacokinetics of vandetanib was comparable with previous studies. Although the plasma exposure to dasatinib decreased from days 8 to 42, it remained similar to adult studies. CSF to plasma exposure of vandetanib and dasatinib were approximately 2% in 2 patients. Phosphorylated 70S6K decreased during therapy in PBMCs. CONCLUSIONS The MTD of vandetanib and dasatinib in combination was 65 mg/m(2) for each drug. Other studies are underway to test dasatinib and other PDGFR inhibitors alone or in combination for this deadly cancer.
Collapse
Affiliation(s)
- Alberto Broniscer
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
95
|
Park SR, Davis M, Doroshow JH, Kummar S. Safety and feasibility of targeted agent combinations in solid tumours. Nat Rev Clin Oncol 2013; 10:154-68. [PMID: 23358316 DOI: 10.1038/nrclinonc.2012.245] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The plethora of novel molecular-targeted agents (MTAs) has provided an opportunity to selectively target pathways involved in carcinogenesis and tumour progression. Combination strategies of MTAs are being used to inhibit multiple aberrant pathways in the hope of optimizing antitumour efficacy and to prevent development of resistance. While the selection of specific agents in a given combination has been based on biological considerations (including the role of the putative targets in cancer) and the interactions of the agents used in combination, there has been little exploration of the possible enhanced toxicity of combinations resulting from alterations in multiple signalling pathways in normal cell biology. Owing to the complex networks and crosstalk that govern normal and tumour cell proliferation, inhibiting multiple pathways with MTA combinations can result in unpredictable disturbances in normal physiology. This Review focuses on the main toxicities and the lack of tolerability of some common MTA combinations, particularly where evidence of enhanced toxicity compared to either agent alone is documented or there is development of unexpected toxicity. Toxicities caused by MTA combinations highlight the need to introduce new preclinical testing paradigms early in the drug development process for the assessment of chronic toxicities resulting from such combinations.
Collapse
Affiliation(s)
- Sook Ryun Park
- Division of Cancer Treatment and Diagnosis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 31, Room 3A44, 31 Center Drive, Bethesda, MD 20892, USA
| | | | | | | |
Collapse
|
96
|
Principles of dose finding studies in cancer: a comparison of trial designs. Cancer Chemother Pharmacol 2013; 71:1107-14. [PMID: 23299793 PMCID: PMC3636432 DOI: 10.1007/s00280-012-2059-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Accepted: 12/12/2012] [Indexed: 10/27/2022]
Abstract
PURPOSE One key aim of Phase I cancer studies is to identify the dose of a treatment to be further evaluated in Phase II. We describe, in non-statistical language, three classes of dose-escalation trial design and compare their properties. METHODS We review three classes of dose-escalation design suitable for Phase I cancer trials: algorithmic approaches (including the popular 3 + 3 design), Bayesian model-based designs and Bayesian curve-free methods. We describe an example from each class and summarize the advantages and disadvantages of the design classes. RESULTS The main benefit of algorithmic approaches is the simplicity with which they may be communicated: it may be for this reason alone that they are still employed in the vast majority of Phase I trials. Model-based and curve-free Bayesian approaches are preferable to algorithmic methods due to their superior ability to identify the dose with the desired toxicity rate and their allocation of a greater proportion of patients to doses at, or close to, that dose. CONCLUSIONS For statistical and practical reasons, algorithmic methods cannot be recommended. The choice between a Bayesian model-based or curve-free approach depends on the previous information available about the compound under investigation. If this provides assurance about a particular model form, the model-based approach would be appropriate; if not, the curve-free method would be preferable.
Collapse
|
97
|
Abstract
Attrition rates of drugs from human entry to regulatory approval are far higher in anticancer drugs than those for nononcology indications. In the era of molecular therapeutics that results from a deeper understanding in cancer biology and advancing technologies, the number of compounds available for clinical testing is likely to continue to increase. Although the main objectives of phase I trials are to characterize toxicities of new agents and to determine the recommended dose for phase II development, most phase I studies are now designed to provide some early signal on preliminary efficacy as secondary objectives. The "go-no-go" decision to further develop a drug, or not, is now often pushed forward to the phase I setting. Thus, there is a need for investigators to be able to critically review the preclinical data available in order to determine which drugs should advance on the developmental path. This review highlights the intrinsic characteristics of a drug and the relevant data to be collected during its preclinical assessment, which may maximize the chances of success in clinical testing and eventual regulatory approval.
Collapse
Affiliation(s)
- Herbert H Loong
- From the Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | | |
Collapse
|
98
|
Le Tourneau C, Gan HK, Razak ARA, Paoletti X. Efficiency of new dose escalation designs in dose-finding phase I trials of molecularly targeted agents. PLoS One 2012; 7:e51039. [PMID: 23251419 PMCID: PMC3521009 DOI: 10.1371/journal.pone.0051039] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Accepted: 10/30/2012] [Indexed: 11/25/2022] Open
Abstract
Background Statistical simulations have consistently demonstrated that new dose-escalation designs such as accelerated titration design (ATD) and continual reassessment method (CRM)-type designs outperform the standard “3+3” design in phase I cancer clinical trials. Methods We evaluated the actual efficiency of different dose escalation methods employed in first-in-human phase I clinical trials of targeted agents administered as single agents published over the last decade. Results Forty-nine per cent of the 84 retrieved trials used the standard “3+3” design. Newer designs used included ATD in 42%, modified CRM [mCRM] in 7%, and pharmacologically guided dose escalation in 1%. The median numbers of dose levels explored in trials using “3+3”, ATD and mCRM designs were 6, 8 and 10, respectively. More strikingly, the mean MTD to starting dose ratio appeared to be at least twice as high for trials using mCRM or ATD designs as for trials using a standard “3+3” design. Despite this, the mean number of patients exposed to a dose below the MTD was similar in trials using “3+3”, ATD and mCRM designs. Conclusion Our results support a more extensive implementation of innovative dose escalation designs such as mCRM and ATD in phase I cancer clinical trials of molecularly targeted agents.
Collapse
|
99
|
Galsky MD, Hahn NM, Powles T, Hellerstedt BA, Lerner SP, Gardner TA, Yu M, O'Rourke M, Vogelzang NJ, Kocs D, McKenney SA, Melnyk AM, Hutson TE, Rauch M, Wang Y, Asmar L, Sonpavde G. Gemcitabine, Cisplatin, and sunitinib for metastatic urothelial carcinoma and as preoperative therapy for muscle-invasive bladder cancer. Clin Genitourin Cancer 2012; 11:175-81. [PMID: 23228446 DOI: 10.1016/j.clgc.2012.10.001] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Revised: 09/28/2012] [Accepted: 10/01/2012] [Indexed: 12/26/2022]
Abstract
BACKGROUND Data support chemotherapy combined with antiangiogenic therapy in metastatic urothelial cancer (mUC) and muscle-invasive bladder cancer (MIBC). We investigated the efficacy and safety of gemcitabine, cisplatin, and sunitinib (GCS) in mUC and MIBC in parallel phase II trials. PATIENTS AND METHODS Trial 1 enrolled 36 patients with mUC who were chemotherapy naive; trial 2 enrolled 9 patients with MIBC. The primary endpoints for trials 1 and 2 were response rate and pathologic complete response, respectively. GCS was given as first-line treatment for patients with mUC and as neoadjuvant therapy for patients with MIBC. The Simon minimax 2-stage design was used for an objective response rate in trial 1 and for the pathologic complete response rate in trial 2. RESULTS The initial trial 1 GCS dose was gemcitabine 1000 mg/m(2) intravenously, days 1 and 8; cisplatin 70 mg/m(2) intravenously, day 1; and sunitinib 37.5 mg orally daily, days 1 to 14 of a 21-day cycle. These doses proved intolerable. The doses of gemcitabine and cisplatin were subsequently reduced to 800 and 60 mg/m(2), respectively, without an improvement in drug delivery, and the trial was closed. This lower-dose regimen was applied in trial 2, which was stopped early due to excess toxicity. Grade 3 to 4 hematologic toxicities occurred in 70% (23/33) of patients in trial 1 and 22% (2/9) of patients in trial 2. In trial 1, the response rate was 49% (95% CI, 31%-67%); in trial 2, the pathologic complete response was 22% (2/9). Due to early closure secondary to toxicity, the sample sizes of both trials were small. CONCLUSIONS Delivery of GCS was hampered by excessive toxicity in both advanced and neoadjuvant settings.
Collapse
Affiliation(s)
- Matthew D Galsky
- US Oncology Research, McKesson Specialty Health, The Woodlands, TX, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
100
|
Penel N, Adenis A, Bonneterre J. Role of the investigator in phase 1 trials of anticancer drugs. Lancet Oncol 2012; 13:1177-9. [DOI: 10.1016/s1470-2045(12)70502-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|