1
|
Martínez-Barros H, Pousada-Fonseca Á, Pedreira-Bouzas J, Clopés-Estela A. [Translated article] Characteristics, clinical benefit, and reimbursement of new authorisations for oncohaematology drugs in Spain between 2017 and 2020. FARMACIA HOSPITALARIA 2024:S1130-6343(24)00121-1. [PMID: 39069449 DOI: 10.1016/j.farma.2024.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 04/25/2024] [Accepted: 04/26/2024] [Indexed: 07/30/2024] Open
Abstract
OBJECTIVE To describe the authorisations and funding resolutions for new onco-haematological drugs in Spain between 2017 and 2020, as well as the results of their main trials. METHODS Observational, cross-sectional, descriptive study conducted between October and December 2022. Onco-haematology drugs approved by the European Medicines Agency between 2017 and 2020 were included, according to EFPIA patients W.A.I.T Indicator 2021 Survey. Authorisation information was obtained from the main study of the European Public Assessment Report. Data were collected on medicines, their authorisation and main study, benefit shown, cost, and status and time to reimbursement. RESULTS Forty-one new drugs authorised for 49 indications were identified. More than half (58.5%) were targeted therapies, and 61.2% were for the treatment of solid tumours (61.2%). Most had palliative intent (71.4%) and were indicated in relapsed or refractory disease (55.1%). Of the clinical trials, 57.1% were phase III and 63.3% were randomised. The primary endpoint was overall survival in 16.3%, increasing to 25.8% among randomised clinical trials. Regarding licensed drugs based on response rate, the median response rate was 56.4% [IQI 40-66.3]. In those authorised on the basis of surrogate time-to-event endpoints, the median hazard ratio was 0.54 [IQI 0.38-0.57], and among those using overall survival was 0.71 [IQI 0.59-0.77]. Globally, 22.4% had shown benefit in overall survival, with a median gain of 4 months [IQI 3.6-16.7]. One-third (33.3%) of the indications evaluable according to the European Society for Medical Oncology Magnitude of Clinical Benefit Scale showed substantial clinical benefit. Of the indications, 75.5% were funded, half (48.6%; 36.7% of the total) with restrictions. The median time to funding was 19.5 months [IQI 11.4-29.3]. CONCLUSIONS Most main clinical trials of new onco-haematology drugs approved in Spain used surrogate primary endpoint and, at the time of authorisation, few had shown to prolong overall survival. More than a third were uncontrolled clinical trials.
Collapse
Affiliation(s)
| | | | | | - Ana Clopés-Estela
- Servicio de Farmacia, Instituto Catalán de Oncología (ICO) t, Instituto de Investigación Biomédica de Bellvitge (IDIBELL), Universidad Ramon Llull, Barcelona, Spain
| |
Collapse
|
2
|
Martínez-Barros H, Pousada-Fonseca Á, Pedreira-Bouzas J, Clopés-Estela A. Characteristics, clinical benefit and reimbursement of new authorisations for oncohaematology drugs in Spain between 2017 and 2020. FARMACIA HOSPITALARIA 2024:S1130-6343(24)00079-5. [PMID: 38797624 DOI: 10.1016/j.farma.2024.04.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 04/25/2024] [Accepted: 04/26/2024] [Indexed: 05/29/2024] Open
Abstract
OBJECTIVE To describe the authorisations and funding resolutions for new onco-hematological drugs in Spain between 2017 and 2020, as well as the results of their main trials. METHODS Observational, cross-sectional, descriptive study conducted between October and December 2022. Onco-hematology drugs approved by the European Medicines Agency between 2017 and 2020 were included, according to EFPIA patients W.A.I.T Indicator 2021 Survey. Authorisation information was obtained from the main study of the European Public Assessment Report (EPAR). Data were collected on medicines, their authorisation and main study, benefit shown, cost, and status and time to reimbursement. RESULTS Forty-one new drugs authorised for 49 indications were identified. More than half (58.5%) were targeted therapies, and 61.2% were for the treatment of solid tumors (61.2%). Most had palliative intent (71.4%) and were indicated in relapsed or refractory disease (55.1%). Of the clinical trials, 57.1% were phase III and 63.3% were randomised. The primary endpoint was overall survival in 16.3%, increasing to 25.8% among randomised clinical trials. Regarding licensed drugs based on response rate, the median response rate was 56.4% (IQI 40.0-66.3). In those authorised on the basis of surrogate time-to-event endpoints, the median Hazard Ratio was 0.54 (IQI 0.38-0.57), and among those using overall survival was 0.71 (IQI 0.59-0.77). Globally, 22.4% had shown benefit in overall survival, with a median gain of 4 months (IQI 3.6-16.7). One third (33.3%) of the indications evaluable according to the European Society for Medical Oncology Magnitude of Clinical Benefit Scale showed substantial clinical benefit. Of the indications, 75.5% were funded, half (48.6%; 36.7% of the total) with restrictions. The median time to funding was 19.5 months (IQI 11.4-29.3). CONCLUSIONS Most main clinical trials of new onco-haematology drugs approved in Spain used surrogate primary endpoint and, at the time of authorisation, few had shown to prolong overall survival. More than a third were uncontrolled clinical trials.
Collapse
Affiliation(s)
| | | | | | - Ana Clopés-Estela
- Servicio de Farmacia, Instituto Catalán de Oncología (ICO), Instituto de Investigación Biomédica de Bellvitge (IDIBELL), Universidad Ramon Llull, Barcelona, España
| |
Collapse
|
3
|
Arrillaga-Romany I, Lassman A, McGovern SL, Mueller S, Nabors B, van den Bent M, Vogelbaum MA, Allen JE, Melemed AS, Tarapore RS, Wen PY, Cloughesy T. ACTION: a randomized phase 3 study of ONC201 (dordaviprone) in patients with newly diagnosed H3 K27M-mutant diffuse glioma. Neuro Oncol 2024; 26:S173-S181. [PMID: 38445964 PMCID: PMC11066938 DOI: 10.1093/neuonc/noae031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Indexed: 03/07/2024] Open
Abstract
BACKGROUND H3 K27M-mutant diffuse glioma primarily affects children and young adults, is associated with a poor prognosis, and no effective systemic therapy is currently available. ONC201 (dordaviprone) has previously demonstrated efficacy in patients with recurrent disease. This phase 3 trial evaluates ONC201 in patients with newly diagnosed H3 K27M-mutant glioma. METHODS ACTION (NCT05580562) is a randomized, double-blind, placebo-controlled, parallel-group, international phase 3 study of ONC201 in newly diagnosed H3 K27M-mutant diffuse glioma. Patients who have completed standard frontline radiotherapy are randomized 1:1:1 to receive placebo, once-weekly dordaviprone, or twice-weekly dordaviprone on 2 consecutive days. Primary efficacy endpoints are overall survival (OS) and progression-free survival (PFS); PFS is assessed by response assessment in neuro-oncology high-grade glioma criteria (RANO-HGG) by blind independent central review. Secondary objectives include safety, additional efficacy endpoints, clinical benefit, and quality of life. Eligible patients have histologically confirmed H3 K27M-mutant diffuse glioma, a Karnofsky/Lansky performance status ≥70, and completed first-line radiotherapy. Eligibility is not restricted by age; however, patients must be ≥10 kg at time of randomization. Patients with a primary spinal tumor, diffuse intrinsic pontine glioma, leptomeningeal disease, or cerebrospinal fluid dissemination are not eligible. ACTION is currently enrolling in multiple international sites.
Collapse
Affiliation(s)
- Isabel Arrillaga-Romany
- Mass General Cancer Center, Neuro-Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Andrew Lassman
- Columbia University Vagelos College of Physicians and Surgeons, Herbert Irving Comprehensive Cancer Center, New York-Presbyterian Hospital, New York City, New York, USA
| | - Susan L McGovern
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sabine Mueller
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California, USA
| | - Burt Nabors
- Department of Neuro-Oncology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Martin van den Bent
- Brain Tumor Center at Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | | | | | | | | | - Patrick Y Wen
- Center For Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Timothy Cloughesy
- Bowyer Oncology Center, University of California Los Angeles, Los Angeles, California, USA
| |
Collapse
|
4
|
Cooner F, Ye J, Reaman G. Clinical trial considerations for pediatric cancer drug development. J Biopharm Stat 2023; 33:859-874. [PMID: 36749066 DOI: 10.1080/10543406.2023.2172424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 01/20/2023] [Indexed: 02/08/2023]
Abstract
Oncology has been one of the most active therapeutic areas in medicinal products development. Despite this fact, few drugs have been approved for use in pediatric cancer patients when compared to the number approved for adults with cancer. This disparity could be attributed to the fact that many oncology drugs have had orphan drug designation and were exempt from Pediatric Research Equity Act (PREA) requirements. On August 18, 2017, the RACE for Children Act, i.e. Research to Accelerate Cures and Equity Act, was signed into law as Title V of the 2017 FDA Reauthorization Act (FDARA) to amend the PREA. Pediatric investigation is now required if the drug or biological product is intended for the treatment of an adult cancer and directed at a molecular target that FDA determines to be "substantially relevant to the growth or progression of a pediatric cancer." This paper discusses the specific considerations in clinical trial designs and statistical methodologies to be implemented in oncology pediatric clinical programs.
Collapse
Affiliation(s)
- Freda Cooner
- Global Biostatistics, Amgen Inc, Thousand Oaks, CA, USA
| | - Jingjing Ye
- Global Statistics and Data Sciences (GSDS), BeiGene USA, Fulton, MD, USA
| | - Gregory Reaman
- Oncology Center of Excellence, Office of the Commissioner, U.S. FDA, Silver Spring, MD, USA
| |
Collapse
|
5
|
Olivier T, Haslam A, Prasad V. Post-progression treatment in cancer randomized trials: a cross-sectional study of trials leading to FDA approval and published trials between 2018 and 2020. BMC Cancer 2023; 23:448. [PMID: 37198564 DOI: 10.1186/s12885-023-10917-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 05/04/2023] [Indexed: 05/19/2023] Open
Abstract
BACKGROUND Suboptimal treatment upon progression may affect overall survival (OS) results in oncology randomized controlled trials (RCTs). We aim to assess the proportion of trials reporting post-progression treatment. METHODS This cross-sectional analysis included two concurrent analyses. The first one examined all published RCTs of anti-cancer drugs in six high impact medical/oncology journals between January 2018 and December 2020. The second studied all US Food and Drug Administration (FDA) approved anti-cancer drugs during the same period. Included trials needed to study an anti-cancer drug in the advanced or metastatic setting. Data abstracted included the tumor type, characteristics of trials, and reporting and assessment of post-progression treatment. RESULTS There were 275 published trials and 77 US FDA registration trials meeting inclusion criteria. Assessable post-progression data were reported in 100/275 publications (36.4%) and 37/77 approvals (48.1%). Treatment was considered substandard in 55 publications (n = 55/100, 55.0%) and 28 approvals (n = 28/37, 75.7%). Among trials with assessable post-progression data and positive OS results, a subgroup analysis identified substandard post-progression treatment in 29 publications (n = 29/42, 69.0%) and 20 approvals (n = 20/26, 76.9%). Overall, 16.4% of publications (45/275) and 11.7% of registration trials (9/77) had available post-progression data assessed as appropriate. CONCLUSION We found that most anti-cancer RCTs do not report assessable post-progression treatment. When reported, post-progression treatment was substandard in most trials. In trials reporting positive OS results and with assessable post-progression data, the proportion of trials with subpar post-progression treatment was even higher. Discrepancies between post-progression therapy in trials and the standard of care can limit RCT results' applicability. Regulatory rules should enforce higher requirements regarding post-progression treatment access and reporting.
Collapse
Affiliation(s)
- Timothée Olivier
- Department of Oncology, Geneva University Hospital, 4 Gabrielle-Perret-Gentil Street, Geneva, 1205, Switzerland.
- Department of Epidemiology and Biostatistics, University of California San Francisco, 550 16th St, 2nd Fl, San Francisco, CA, 94158, USA.
| | - Alyson Haslam
- Department of Epidemiology and Biostatistics, University of California San Francisco, 550 16th St, 2nd Fl, San Francisco, CA, 94158, USA
| | - Vinay Prasad
- Department of Epidemiology and Biostatistics, University of California San Francisco, 550 16th St, 2nd Fl, San Francisco, CA, 94158, USA
| |
Collapse
|
6
|
Olivier T, Haslam A, Prasad V. Sotorasib in KRAS G12C mutated lung cancer: Can we rule out cracking KRAS led to worse overall survival? Transl Oncol 2022; 28:101591. [PMID: 36577165 PMCID: PMC9803768 DOI: 10.1016/j.tranon.2022.101591] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 11/09/2022] [Accepted: 11/17/2022] [Indexed: 12/27/2022] Open
Abstract
The KRAS oncogene is present in up to 25% of solid tumors and for decades had been undruggable. Sotorasib was the first-in-class KRAS inhibitor to reach the US and European market, and its pharmacological inhibition is restricted to the KRAS p.G12C mutation. Sotorasib showed activity (tumor shrinkage) in patients with non-small cell lung cancer harboring this specific mutation, and efficacy was tested in the CodeBreaK 200, open-label, phase 3 trial (NCT04303780). The results were presented in the ESMO 2022 meeting. CodeBreaK 200 found an improvement in the primary endpoint of progression-free survival (PFS), but overall survival, a key secondary endpoint, was not improved. However, critical questions about the trial's design may limit inferences regarding the reported results. The control arm treatment was inferior to the best standard of care. A late protocol modification (which lowered the sample size and allowed a problematic crossover) prohibited the trial from making a determination regarding overall survival. Imbalance in censoring rates, with potential informative censoring, makes PFS estimates unreliable. Quality-of-life data were also limited. Ultimately, CodeBreaK 200 does not clarify how this therapy should be used in practice, and while we maintain cautious enthusiasm for this and other Ras inhibitors, we await more informative trials.
Collapse
Affiliation(s)
- Timothée Olivier
- Department of Oncology, Geneva University Hospital, 4 Gabrielle-Perret-Gentil Street, 1205, Geneva, Switzerland; Department of Epidemiology and Biostatistics, University of California San Francisco, 550 16th St, 2nd Fl, San Francisco, CA 94158, USA.
| | - Alyson Haslam
- Department of Epidemiology and Biostatistics, University of California San Francisco, 550 16th St, 2nd Fl, San Francisco, CA 94158, USA
| | - Vinay Prasad
- Department of Epidemiology and Biostatistics, University of California San Francisco, 550 16th St, 2nd Fl, San Francisco, CA 94158, USA
| |
Collapse
|
7
|
Strohbehn GW, Kacew AJ, Goldstein DA, Feldman RC, Ratain MJ. Combination therapy patents: a new front in evergreening. Nat Biotechnol 2021; 39:1504-1510. [PMID: 34880460 DOI: 10.1038/s41587-021-01137-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Garth W Strohbehn
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA.,Optimal Cancer Care Alliance, Ann Arbor, MI, USA.,Veterans Affairs Center for Clinical Management and Research, Ann Arbor, MI, USA
| | - Alec J Kacew
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Daniel A Goldstein
- Optimal Cancer Care Alliance, Ann Arbor, MI, USA.,Tel Aviv University, Tel Aviv, Israel.,Davidoff Cancer Center, Rabin Medical Center, Petah Tikva, Israel.,Department of Health Policy and Management, Gillings School of Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Robin C Feldman
- Center for Innovation, University of California Hastings College of the Law, San Francisco, CA, USA.
| | - Mark J Ratain
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA. .,Optimal Cancer Care Alliance, Ann Arbor, MI, USA.
| |
Collapse
|
8
|
Doussau A, Agarwal I, Fojo T, Tannock IF, Grady C. Design of placebo-controlled randomized trials of anticancer agents: Ethical considerations based on a review of published trials. Clin Trials 2021; 18:690-698. [PMID: 34693757 DOI: 10.1177/17407745211052474] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Limited information exists about the design of placebo-controlled cancer trials. Through a systematic review of trials published in 2013, we describe placebo use in randomized trials testing anticancer agents and analyze strategies that increase exposure to the experimental regimen. METHODS Trials were classified as add-on (placebo in combination with standard treatment) or placebo-only. Strategies to allow more than half of the participants to receive the experimental regimen were reviewed. The risk-benefit ratio of receiving the experimental agent was considered favorable if the difference in primary outcome was significant (p ≤ 0.05), neutral if there was no significant difference in the primary outcome and the experimental agent did not add substantial toxicity, and unfavorable otherwise. RESULTS Eighty trials were included (32,694 participants). Most trials were add-on (69%). The risk-benefit outcome was favorable, neutral, and unfavorable to the experimental agent in 52%, 32%, and 16% of placebo-only trials and 25%, 53%, and 22%, respectively, of add-on trials. Four strategies increased exposure to the experimental regimen: one-way crossover (23%), uneven randomization (21%), three-arms (13%), and randomized discontinuation design (4%); these strategies were used more often in placebo-only trials. CONCLUSION A minority of participants received placebo alone and strategies to increase experimental exposure were used commonly. Fewer than half of the studies had favorable outcomes, thus defending the use of placebo controls, when there is no established treatment. Strategies that increase patient exposure to experimental agents rather than placebo may expose them to non-beneficial, sometimes toxic, experimental agents.
Collapse
Affiliation(s)
- Adélaïde Doussau
- Department of Bioethics, Clinical Center, National Institutes of Health, Bethesda, MD, USA.,St. Mary's Research Centre, Montreal, QC, Canada
| | - Isha Agarwal
- Department of Bioethics, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Tito Fojo
- Department of Medicine, Division of Hematology/Oncology, Columbia University in the City of New York, New York, NY, USA
| | - Ian F Tannock
- Princess Margaret Cancer Center and University of Toronto, Toronto, ON, Canada
| | - Christine Grady
- Department of Bioethics, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
9
|
Gyawali B, de Vries EGE, Dafni U, Amaral T, Barriuso J, Bogaerts J, Calles A, Curigliano G, Gomez-Roca C, Kiesewetter B, Oosting S, Passaro A, Pentheroudakis G, Piccart M, Roitberg F, Tabernero J, Tarazona N, Trapani D, Wester R, Zarkavelis G, Zielinski C, Zygoura P, Cherny NI. Biases in study design, implementation, and data analysis that distort the appraisal of clinical benefit and ESMO-Magnitude of Clinical Benefit Scale (ESMO-MCBS) scoring. ESMO Open 2021; 6:100117. [PMID: 33887690 PMCID: PMC8086024 DOI: 10.1016/j.esmoop.2021.100117] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/16/2021] [Accepted: 03/22/2021] [Indexed: 12/15/2022] Open
Abstract
Background The European Society for Medical Oncology-Magnitude of Clinical Benefit Scale (ESMO-MCBS) is a validated, widely used tool developed to score the clinical benefit from cancer medicines reported in clinical trials. ESMO-MCBS scores assume valid research methodologies and quality trial implementation. Studies incorporating flawed design, implementation, or data analysis may generate outcomes that exaggerate true benefit and are not generalisable. Failure to either indicate or penalise studies with bias undermines the intention and diminishes the integrity of ESMO-MCBS scores. This review aimed to evaluate the adequacy of the ESMO-MCBS to address bias generated by flawed design, implementation, or data analysis and identify shortcomings in need of amendment. Methods As part of a refinement of the ESMO-MCBS, we reviewed trial design, implementation, and data analysis issues that could bias the results. For each issue of concern, we reviewed the ESMO-MCBS v1.1 approach against standards derived from Helsinki guidelines for ethical human research and guidelines from the International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use, the Food and Drugs Administration, the European Medicines Agency, and European Network for Health Technology Assessment. Results Six design, two implementation, and two data analysis and interpretation issues were evaluated and in three, the ESMO-MCBS provided adequate protections. Seven shortcomings in the ability of the ESMO-MCBS to identify and address bias were identified. These related to (i) evaluation of the control arm, (ii) crossover issues, (iii) criteria for non-inferiority, (iv) substandard post-progression treatment, (v) post hoc subgroup findings based on biomarkers, (vi) informative censoring, and (vii) publication bias against quality-of-life data. Conclusion Interpretation of the ESMO-MCBS scores requires critical appraisal of trials to understand caveats in trial design, implementation, and data analysis that may have biased results and conclusions. These will be addressed in future iterations of the ESMO-MCBS. We reviewed trial design, implementation, and data analysis issues that could bias the results of trials. These issues could skew the results of ESMO-MCBS scores. Six design, two implementation, and two analysis issues were reviewed, and seven shortcomings were identified. These issues will be addressed in future versions of the MCBS scale. Interpretation of MCBS scores requires critical appraisal of trials.
Collapse
Affiliation(s)
- B Gyawali
- Department of Oncology, Queen's University, Kingston, Ontario, Canada; Department of Public Health Sciences, Queen's University, Kingston, Ontario, Canada; Division of Cancer Care and Epidemiology, Queen's University, Kingston, Ontario, Canada.
| | - E G E de Vries
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - U Dafni
- Laboratory of Biostatistics, School of Health Sciences, National and Kapodistrian University of Athens, Athens; Frontier Science Foundation-Hellas, Athens, Greece
| | - T Amaral
- Skin Cancer Center, Department of Dermatology, Eberhard Karls University, Tuebingen, Germany
| | - J Barriuso
- The Christie NHS Foundation Trust and Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - J Bogaerts
- European Organisation for Research and Treatment of Cancer, Brussels, Belgium
| | - A Calles
- Medical Oncology Department, Hospital General Universitario Gregorio Marañon, Madrid, Spain
| | - G Curigliano
- Department of Oncology and Hemato-Oncology, University of Milan, Milan; European Institute of Oncology, IRCCS, Milan, Italy
| | - C Gomez-Roca
- Institut Universitaire du Cancer de Toulouse (IUCT), Toulouse, France
| | - B Kiesewetter
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - S Oosting
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - A Passaro
- Division of Thoracic Oncology, European Institute of Oncology, IRCCS, Milan, Italy
| | | | - M Piccart
- Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium
| | - F Roitberg
- WHO Cancer Management Consultant, Geneva, Switzerland; Instituto do Cancer do Estado de São Paulo (ICESP HCFMUSP), São Paulo, Brazil
| | - J Tabernero
- Vall d'Hebron Hospital Campus and Institute of Oncology (VHIO), UVic-UCC, IO-Quiron, Barcelona, Spain
| | - N Tarazona
- Department of Medical Oncology, Biomedical Research Institute INCLIVA, CIBERONC, University of Valencia, Valencia, Spain
| | - D Trapani
- European Institute of Oncology, IRCCS, Milan, Italy
| | - R Wester
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - G Zarkavelis
- University of Ioannina-Department of Medical Oncology, Ioannina, Greece
| | - C Zielinski
- Central European Cooperative Oncology Group and Central European Cancer Center, Wiener Privatklinik, Vienna, Austria
| | - P Zygoura
- Frontier Science Foundation-Hellas, Athens, Greece
| | - N I Cherny
- Cancer Pain and Palliative Medicine Service, Department of Medical Oncology, Shaare Zedek Medical Center, Jerusalem, Israel
| |
Collapse
|
10
|
Yeh J, Gupta S, Patel SJ, Kota V, Guddati AK. Trends in the crossover of patients in phase III oncology clinical trials in the USA. Ecancermedicalscience 2020; 14:1142. [PMID: 33343701 PMCID: PMC7738270 DOI: 10.3332/ecancer.2020.1142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Indexed: 11/24/2022] Open
Abstract
Background The incorporation of crossover in randomised controlled trials is accepted as an ethical obligation, especially in cancer clinical trials. The more common type of crossover is crossover allowance, which allows patients assigned to one arm to switch to another arm if there is an established benefit in the crossover arm. In contrast, crossover-designed studies involve switching patients from all arms to a different arm as part of the study design. Crossover allowance may have advantages in patient recruitment and incorporating crossover after initial positive results fulfil ethical requirements. However, crossover can also contribute to confounding major endpoints of studies, such as overall survival or the second progression-free survival interval. For this reason, it is important to investigate and identify potential trends of crossover in clinical trials testing novel therapies. Methods Data about cancer clinical trials were extracted from clinicaltrials.gov. The search query was limited to completed phase III studies in adult populations. Location was limited to the USA. Date range extended from 1990 to 2019. Search query included the terms: cancer; completed- recruitment status; age: 18–65+ years; sex: all; location: USA; and study phase: phase 3. Studies were then excluded if they were not randomised controlled trials (RCTs) with the primary purpose of treatment and if they did not test cancer-related interventions. Results A total of 744 clinical trials were identified. There were 459 RCTs aimed at treatment, and of those, 35 utilised crossover. The start dates of these crossover trials ranged from 1997 to 2012. Thirty studies utilised crossover allowance. Prostate, breast and gastrointestinal stromal tumour cancers were the most represented cancer types in crossover studies. Among the 30 studies, the median proportion of patients who crossed over relative to the original arm assignment ranged from 2% to 88%, with a median of 57.5%. Conclusions The proportion of identified clinical trials with crossover compared to those without is extremely small. Crossover in clinical trials studying cancer treatment does not appear to be a widespread practice. Even though statistical approaches to mitigate confounding exist, crossover can still skew accurate reporting of the impact of experimental therapies on overall survival.
Collapse
Affiliation(s)
- Justin Yeh
- Medical College of Georgia, Augusta University, Augusta, GA 30909, USA
| | - Shruti Gupta
- Medical College of Georgia, Augusta University, Augusta, GA 30909, USA
| | - Sunny J Patel
- Medical College of Georgia, Augusta University, Augusta, GA 30909, USA
| | - Vamsi Kota
- Division of Hematology/Oncology, Georgia Cancer Center, Augusta University, Augusta, GA 30909, USA
| | - Achuta K Guddati
- Division of Hematology/Oncology, Georgia Cancer Center, Augusta University, Augusta, GA 30909, USA
| |
Collapse
|
11
|
Pasalic D, McGinnis GJ, Fuller CD, Grossberg AJ, Verma V, Mainwaring W, Miller AB, Lin TA, Jethanandani A, Espinoza AF, Diefenhardt M, Das P, Subbiah V, Subbiah IM, Jagsi R, Garden AS, Fokas E, Rödel C, Thomas CR, Minsky BD, Ludmir EB. Progression-free survival is a suboptimal predictor for overall survival among metastatic solid tumour clinical trials. Eur J Cancer 2020; 136:176-185. [PMID: 32702645 DOI: 10.1016/j.ejca.2020.06.015] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 06/01/2020] [Accepted: 06/11/2020] [Indexed: 01/17/2023]
Abstract
BACKGROUND The use of overall survival (OS) as the gold standard primary end-point (PEP) in metastatic oncologic randomised controlled trials (RCTs) has declined in favour of progression-free survival (PFS) without a complete understanding of the degree to which PFS reliably predicts for OS. METHODS Using ClinicalTrials.gov, we identified 1239 phase III oncologic RCTs, 260 of which were metastatic solid tumour trials with a superiority-design investigating a therapeutic intervention by using either a PFS or OS PEP. Each individual trial was reviewed to quantify RCT design factors and disease-related outcomes. RESULTS A total of 172,133 patients were enrolled from the year 1999 to 2015 in RCTs that used PFS (56.2%, 146/260) or OS (43.8%, 114/260) as the PEP. PFS trials were more likely to restrict patient eligibility by using molecular criteria (15.1% versus 4.4%, p = 0.005) use targeted therapy (80.1% versus 67.5%, p = 0.048), accrue fewer patients (median 495 versus 619, p = 0.03), and successfully meet the trial PEP (66.9% versus 33.3%, p < 0.0001). On multiple binary logistic regression analysis, factors that predicted for PFS or OS PEP trial success included choice of PFS PEP (p < 0.0001), molecular profile restriction (p = 0.02) and single agent therapy (p = 0.02). Notably, there was only a 38% (31/82) conversion rate of positive PFS-to-OS benefit; lack of industry sponsorship predicted for PFS-to-OS signal conversion (80.0% without industry sponsorship versus 35.1% with industry sponsorship, p = 0.045). CONCLUSIONS A PFS PEP has suboptimal positive predictive value for OS among phase III metastatic solid tumour RCTs. Regulatory agency decisions should be judicious in using PFS results as the primary basis for approval.
Collapse
Affiliation(s)
- Dario Pasalic
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - C David Fuller
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Vivek Verma
- Allegheny Health Network Cancer Institute, Pittsburgh, PA, USA
| | | | - Austin B Miller
- The University of Texas Health Science Center, McGovern Medical School, Houston, TX, USA
| | - Timothy A Lin
- The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | | | - Markus Diefenhardt
- University of Frankfurt, Frankfurt, Germany; Frankfurt Cancer Institute, Frankfurt, Germany
| | - Prajnan Das
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vivek Subbiah
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | - Adam S Garden
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Emmanouil Fokas
- University of Frankfurt, Frankfurt, Germany; Frankfurt Cancer Institute, Frankfurt, Germany; German Cancer Research Center, Heidelberg, Germany; German Cancer Consortium, Frankfurt, Germany
| | - Claus Rödel
- University of Frankfurt, Frankfurt, Germany; Frankfurt Cancer Institute, Frankfurt, Germany; German Cancer Research Center, Heidelberg, Germany; German Cancer Consortium, Frankfurt, Germany
| | | | - Bruce D Minsky
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ethan B Ludmir
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
12
|
McNicol E, Ferguson M, Bungay K, Rowe EL, Eldabe S, Gewandter JS, Hayek SM, Katz N, Kopell BH, Markman J, Rezai A, Taylor RS, Turk DC, Dworkin RH, North RB, Thomson S. Systematic Review of Research Methods and Reporting Quality of Randomized Clinical Trials of Spinal Cord Stimulation for Pain. THE JOURNAL OF PAIN 2020; 22:127-142. [PMID: 32574787 DOI: 10.1016/j.jpain.2020.05.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 04/21/2020] [Accepted: 05/04/2020] [Indexed: 12/18/2022]
Abstract
This systematic review assessed design characteristics and reporting quality of published randomized clinical trials of spinal cord stimulation (SCS) for treatment of pain in adults and adolescents. The study protocol was registered with PROSPERO (CRD42018090412). Relevant articles were identified by searching the following databases through December 31, 2018: MEDLINE, Embase, WikiStim, The Cochrane Database of Systematic Reviews, and The Cochrane Central Register of Controlled Trials. Forty-six studies were included. Eighty-seven percent of articles identified a pain-related primary outcome. Secondary outcomes included physical functioning, health-related quality of life, and reductions in opioid use. Nineteen of the 46 studies prespecified adverse events as an outcome, with 4 assessing them as a primary outcome. Eleven studies stated that they blinded participants. Of these, only 5 were assessed as being adequately blinded. The number of participants enrolled was generally low (median 38) and study durations were short (median 12 weeks), particularly in studies of angina. Fifteen studies employed an intention-to-treat analysis, of which only seven specified a method to accommodate missing data. Review of these studies identified deficiencies in both reporting and methodology. The review's findings suggest areas for improving the design of future studies and increasing transparency of reporting. PERSPECTIVE: This article presents a systematic review of research methods and reporting quality of randomized clinical trials of SCS for the treatment of various pain complaints. The review identifies deficiencies in both methodology and reporting, which may inform the design of future studies and improve reporting standards.
Collapse
Affiliation(s)
- Ewan McNicol
- Department of Pharmacy Practice, MCPHS University, Boston, Massachusetts.
| | - McKenzie Ferguson
- Department of Pharmacy Practice, Southern Illinois University Edwardsville, Edwardsville, Illinois
| | | | | | - Sam Eldabe
- University of Exeter, Exeter, UK; Durham University, Durham, UK
| | - Jennifer S Gewandter
- Department of Anesthesiology and Perioperative Medicine, University of Rochester, Rochester, New York
| | - Salim M Hayek
- Case Western Reserve University, University Hospitals of Cleveland, Cleveland, Ohio
| | - Nathaniel Katz
- Analgesic Solutions, Wayland, Massachusetts; Tufts University School of Medicine, Boston, Massachusetts
| | - Brian H Kopell
- Departments of Neurosurgery, Neurology, Psychiatry and Neuroscience, The Icahn School of Medicine at Mount Sinai, NY, New York
| | - John Markman
- Translational Pain Research Program, Department of Neurosurgery, University of Rochester, New York
| | - Ali Rezai
- Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Rod S Taylor
- Institute of Health and Well Being, University of Glasgow, Glasgow, UK; College of Medicine and Health, University of Exeter, Exeter, UK
| | - Dennis C Turk
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, Washington
| | - Robert H Dworkin
- Department of Anesthesiology and Perioperative Medicine, University of Rochester, Rochester, New York
| | | | - Simon Thomson
- Basildon and Thurrock University Hospitals, Essex, UK
| |
Collapse
|
13
|
Haslam A, Prasad V. When is crossover desirable in cancer drug trials and when is it problematic? Ann Oncol 2019; 29:1079-1081. [PMID: 29648572 PMCID: PMC5961160 DOI: 10.1093/annonc/mdy116] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Affiliation(s)
- A Haslam
- Division of Hematology Oncology, Knight Cancer Institute
| | - V Prasad
- Division of Hematology Oncology, Knight Cancer Institute; Department of Public Health and Preventive Medicine; Center for Health Care Ethics, Oregon Health & Science University, Portland, USA.
| |
Collapse
|
14
|
Affiliation(s)
- E Y Chen
- Division of Hematology Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, USA
| | - V Prasad
- Division of Hematology Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, USA; Department of Public Health and Preventive Medicine, Oregon Health & Science University, Portland, USA; Center for Health Care Ethics, Oregon Health & Science University, Portland, USA.
| |
Collapse
|
15
|
Wayant C, Vassar M. A comparison of matched interim analysis publications and final analysis publications in oncology clinical trials. Ann Oncol 2018; 29:2384-2390. [DOI: 10.1093/annonc/mdy447] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
16
|
Brodowicz T, Mir O, Wallet J, Italiano A, Blay JY, Bertucci F, Eisterer W, Chevreau C, Piperno-Neumann S, Bompas E, Ryckewaert T, Liegl-Antzwager B, Thery J, Penel N, Le Cesne A, Le Deley MC. Efficacy and safety of regorafenib compared to placebo and to post-cross-over regorafenib in advanced non-adipocytic soft tissue sarcoma. Eur J Cancer 2018; 99:28-36. [PMID: 29902612 DOI: 10.1016/j.ejca.2018.05.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 04/24/2018] [Accepted: 05/13/2018] [Indexed: 11/30/2022]
Abstract
INTRODUCTION The placebo-controlled phase-2 REGOSARC trial demonstrated the efficacy of regorafenib in patients with leiomyosarcoma, synovial sarcoma and other non-adipocytic sarcoma but not in liposarcoma. Patients initially allocated to placebo were allowed to receive regorafenib after progression. We report here an updated analysis of the trial including evaluation of regorafenib activity after cross-over. METHODS From June 2013 to December 2014, 139 patients were enrolled in the non-adipocytic sarcoma cohorts. Median follow-up is now 32.4 months. Benefit of regorafenib versus placebo in terms of progression-free survival (PFS) and overall survival (OS) from randomisation was estimated by hazard ratio (HR) in Cox models. In the placebo arm, intra-patient benefit of regorafenib after cross-over was evaluated by the growth modulation index (GMI) (GMI was here, for each patient, PFS after cross-over regorafenib divided by PFS with placebo). Furthermore, the activity of delayed (after cross-over) versus early (at study entry) regorafenib was evaluated by comparing PFS after cross-over to regorafenib to PFS after randomisation in the regorafenib arm. RESULTS PFS benefit of regorafenib as compared to placebo was confirmed with longer follow-up (HR = 0.50; 95% CI: 0.35-0.71; p < .0001). OS was not statistically significant different (HR = 0.78; 0.54-1.12; p = .18). This finding may partially be explained by the fact that 55/68 patients who progressed on placebo (81%) received cross-over Regorafenib after progression: 59% of them had a GMI ≥ 1.3 (95% CI, 45-71%). Delayed start of regorafenib was associated with a statistically non-significant shorter PFS as compared to early treatment (HR = 1.21; 0.84-1.73; p = .30) without impact on OS. CONCLUSIONS Observed PFS confirms that regorafenib warrants further clinical investigation in refractory non-adipocytic sarcomas.
Collapse
Affiliation(s)
- Thomas Brodowicz
- Department of Medicine /Oncology, Medical University of Vienna, General Hospital, Comprehensive Cancer Center - MusculoSkeletalTumorUnit, Währinger Gürtel 18-20, 1090 Vienna, Austria; Sarcoma Platform Austria, Austria.
| | - Olivier Mir
- Department of Medical Oncology, Gustave Roussy Cancer Campus, 114 rue Edouard Vaillant, 94800 Villejuif, France.
| | - Jennifer Wallet
- Department of Clinical Research and Innovation, Centre Oscar Lambret, 3 rue Combemale, 59000 Lille, France.
| | - Antoine Italiano
- Department of Medical Oncology, Institut Bergonié, 229 Cours de l'Argonne, 33000 Bordeaux, France; Department of Medicine, Institut Bergonié, 229 Cours de l'Argonne, 33000 Bordeaux, France.
| | - Jean-Yves Blay
- Department of Medical Oncology, Centre Léon Bérard, 28 rue Laennec, 69008, Lyon, France; Claude Bernard University Lyon 1, 43 boulevard du 11 novembre 1918, 69622, Villeurbanne cedex, France.
| | - François Bertucci
- Department of Medical Oncology, Institut Paoli Calmette, 232 Boulevard de Sainte-Marguerite, 13009 Marseille, France.
| | - Wolfgang Eisterer
- Department of Internal Medicine, Klinikum Klagenfurt am Wörthersee, Feschnigstraße 11, 9020 Klagenfurt am Wörthersee, Austria; Sarcoma Platform Austria, Austria.
| | - Christine Chevreau
- Department of Medical Oncology, IUCT Oncopole, 1 Av. Irène Joliot-Curie, 31100 Toulouse, France.
| | | | - Emmanuelle Bompas
- Department of Medical Oncology, Centre René Gauducheau, St. Herblain, Saint-Herblain, France.
| | - Thomas Ryckewaert
- Department of Medical Oncology, Centre Oscar Lambret, Lille, France.
| | - Bernadette Liegl-Antzwager
- Institute of Pathology, Medical University of Graz, 8036 Graz, Austria; Sarcoma Platform Austria, Austria.
| | - Julien Thery
- Department of Clinical Research and Innovation, Centre Oscar Lambret, 3 rue Combemale, 59000 Lille, France.
| | - Nicolas Penel
- Department of Clinical Research and Innovation, Centre Oscar Lambret, 3 rue Combemale, 59000 Lille, France; Department of Medical Oncology, Centre Oscar Lambret, Lille, France; Department of Medical Oncology, University Hospital, 3 rue Combemale, 59000 Lille, France.
| | - Axel Le Cesne
- Department of Medical Oncology, Gustave Roussy Cancer Campus, 114 rue Edouard Vaillant, 94800 Villejuif, France.
| | - Marie-Cécile Le Deley
- Department of Clinical Research and Innovation, Centre Oscar Lambret, 3 rue Combemale, 59000 Lille, France; Paris-Saclay University, Univ. Paris-Sud, CESP, INSERM, Gustave Roussy, 94805 Villejuif, France.
| |
Collapse
|
17
|
Isbary G, Staab TR, Amelung VE, Dintsios CM, Iking-Konert C, Nesurini SM, Walter M, Ruof J. Effect of Crossover in Oncology Clinical Trials on Evidence Levels in Early Benefit Assessment in Germany. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2018; 21:698-706. [PMID: 29909875 DOI: 10.1016/j.jval.2017.09.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 09/17/2017] [Indexed: 06/08/2023]
Abstract
BACKGROUND In oncology clinical trials, crossover is used frequently but may lead to uncertainties regarding treatment effects. OBJECTIVE To investigate the handling of evidence from crossover trials by the European Medicines Agency (EMA) and the German Federal Joint Committee (G-BA). METHODS For oncology medicines with early benefit assessments before January 2015, presence of crossover, clinical data, EMA requests for additional data, and G-BA benefit ratings/evidence levels were analyzed from manufacturers' dossiers, G-BA appraisals, European Public Assessment Reports, and original publications. RESULTS Eleven of 21 benefit assessments included crossover trials. Significant intergroup differences (P < 0.05) in overall survival (OS) were noted in 7 of 11 trials with and 7 of 10 without crossover. For 6 of 11 medicines with crossover, these were demonstrated before crossover. Treatment effects generally worsened with increasing proportions of crossover. The EMA requested additional data more frequently if crossover was performed, particularly if no OS data were available before crossover. The G-BA granted a considerable benefit to 73% of medicines with crossover and 40% of those without. Evidence levels were intermediate for 50% and 75%, respectively. None of the medicines received the highest evidence level. CONCLUSIONS In G-BA appraisals, oncology medicines with crossover received better additional benefit ratings, but were assigned lower evidence levels, than those without. The five medicines with crossover after progression were assigned lower evidence levels than the six medicines with crossover after demonstration of superior OS, indicating that the way in which crossover is implemented may be one factor influencing the assignment of evidence levels by the G-BA.
Collapse
Affiliation(s)
| | - Thomas R Staab
- Roche Pharma AG, Grenzach-Wyhlen, Germany; Medical School of Hanover, Hanover, Germany
| | | | | | | | | | | | - Jörg Ruof
- Medical School of Hanover, Hanover, Germany; r-connect ltd, Basel, Switzerland.
| |
Collapse
|
18
|
|
19
|
Sarfaty M, Leshno M, Gordon N, Moore A, Neiman V, Rosenbaum E, Goldstein DA. Cost Effectiveness of Nivolumab in Advanced Renal Cell Carcinoma. Eur Urol 2017; 73:628-634. [PMID: 28807351 DOI: 10.1016/j.eururo.2017.07.041] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Accepted: 07/28/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND In recent years, new drugs have been introduced for second-line treatment of advanced renal cell carcinoma (RCC). Nivolumab increases overall survival and is associated with less toxicity compared to everolimus in this setting according to the CheckMate 025 study. However, because of the high cost of nivolumab, there is a need to define its value by considering both efficacy and cost. OBJECTIVE To estimate the cost effectiveness of nivolumab for second-line treatment of advanced RCC from the US payer perspective. DESIGN, SETTING, AND PARTICIPANTS A Markov model was developed to compare the costs and effectiveness of nivolumab with those of everolimus and placebo in second-line treatment of advanced RCC. Health outcomes were measured in life-years (LYs) and quality-adjusted LYs (QALYs). Drug costs were based on 2016 Medicare reimbursement rates. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Model robustness was assessed in univariable and probabilistic sensitivity analyses. We addressed the issue of the extensive duration of immunotherapy treatment among long-term survivors, which may or may not be approved by payers. RESULTS AND LIMITATIONS The total mean cost per patient was $101 070 for nivolumab and $50 935 for everolimus. Nivolumab generated a gain of 0.24 LYs (0.34 QALYs) compared to everolimus. The incremental cost-effectiveness ratio (ICER) for nivolumab was $146 532/QALY versus everolimus and $226 197/QALY versus placebo. Limiting the maximal treatment duration of nivolumab to 2 yr reduced the ICER to $121 788/QALY versus everolimus. The analysis is limited by data availability and our assumptions. CONCLUSIONS Our analysis established that with a willingness-to-pay threshold of $100 000 to $150 000 per QALY, nivolumab is estimated to be cost-effective versus everolimus, but not cost-effective versus placebo. PATIENT SUMMARY We assessed the cost effectiveness of nivolumab in previously treated metastatic kidney cancer. In the USA, it would cost $146 532 to gain one quality-adjusted life-year with nivolumab versus everolimus, or $226 197 versus placebo. Nivolumab is considered cost-effective versus everolimus, but not versus placebo.
Collapse
Affiliation(s)
- Michal Sarfaty
- Institute of Oncology, Davidoff Cancer Center, Rabin Medical Center, Petach Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Moshe Leshno
- Coller School of Management, Tel Aviv University, Tel Aviv, Israel
| | - Noa Gordon
- Institute of Oncology, Davidoff Cancer Center, Rabin Medical Center, Petach Tikva, Israel
| | - Assaf Moore
- Institute of Oncology, Davidoff Cancer Center, Rabin Medical Center, Petach Tikva, Israel
| | - Victoria Neiman
- Institute of Oncology, Davidoff Cancer Center, Rabin Medical Center, Petach Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Eli Rosenbaum
- Institute of Oncology, Davidoff Cancer Center, Rabin Medical Center, Petach Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Daniel A Goldstein
- Institute of Oncology, Davidoff Cancer Center, Rabin Medical Center, Petach Tikva, Israel; Winship Cancer Institute, Emory University, Atlanta, GA, USA
| |
Collapse
|
20
|
Five years of EMA-approved systemic cancer therapies for solid tumours-a comparison of two thresholds for meaningful clinical benefit. Eur J Cancer 2017. [PMID: 28648700 DOI: 10.1016/j.ejca.2017.05.029] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
OBJECTIVE Several societies have proposed frameworks to evaluate the benefit of oncology drugs; one prominent tool is the European Society for Medical Oncology Magnitude of Clinical Benefit Scale (ESMO-MCBS). Our objectives were to investigate the extent of European Medicines Agency (EMA)-approved cancer drugs that meet the threshold for 'meaningful clinical benefit' (MCB), defined by the framework, and determine the change in the distribution of grades when an adapted version that addresses the scale's limitations is applied. METHODS We identified cancer drugs approved by the EMA (2011-2016). We previously proposed adaptations to the ESMO-MCBS addressing its main limitations, including the use of the lower limit of the 95% confidence interval in assessing the hazard ratio. To assess the MCB, both the original and adapted ESMO-MCBS were applied to the respective approval studies. RESULTS In total, we identified 70 approval studies for 38 solid cancer drugs. 21% of therapies met the MCB threshold by the original ESMO-MCBS criteria. In contrast, only 11% of therapies met the threshold for MCB when the adapted ESMO-MCBS was applied. Thus 89% and 79% of therapies did not meet the MCB threshold in the adapted and original ESMO-MCBS, respectively. CONCLUSIONS In most of the cancer drugs, the MCB threshold is not met at the time of approval when measured using both ESMO-MCBS scales. Since approval status does not translate into a MCB, stakeholders and decision makers should focus on the benefit/risk ratio of anticancer drugs to assure an appropriate allocation of resources in health care systems.
Collapse
|
21
|
Abstract
OBJECTIVES Treatment switching occurs when patients in a randomized clinical trial switch from the treatment initially assigned to them to another treatment, typically from the control to experimental treatment. This study discusses the issues this raises and possible approaches to addressing them in trials of cancer drugs. METHODS Stakeholders from around the world were invited to a 1.5-day Workshop in Adelaide, Australia. This study attempts to capture the key points from the discussion and the perspectives of the various stakeholder groups, but is not a formal consensus statement. RESULTS Treatment switching raises challenging ethical issues with arguments for and against allowing it. It is increasingly common in cancer drug trials and presents challenges for the interpretation of results by regulators, clinicians, patients, and payers. Proposals are offered for good practice in the design, management, and analysis of trials and wider development programs for cancer drugs in which treatment switching has occurred or is likely to. Recommendations are also offered for further action to improve understanding of the importance and challenges of treatment switching and to promote agreement between key stakeholders on guidelines and other steps to address these challenges. CONCLUSIONS The handling of treatment switching in trials is of concern to all stakeholders. On the basis of the discussions at the Adelaide International Workshop, there would appear to be common ground on approaches to addressing treatment switching in cancer trials and scope for the development of formal guidelines to inform the work of regulators, payers, industry, trial designers and other stakeholders.
Collapse
|
22
|
Crossover Designs in Nutrition and Dietetics Research. J Acad Nutr Diet 2017; 117:1023-1030. [PMID: 28479137 DOI: 10.1016/j.jand.2017.03.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 03/21/2017] [Indexed: 01/22/2023]
Abstract
This article is the 12th installment in a statistical series exploring the importance of research design, epidemiologic methods, and statistical analysis as applied to nutrition and dietetics research. The purpose of this series is to assist registered dietitian nutritionists in interpreting nutrition research and aid nutrition researchers in applying scientific principles to produce high-quality nutrition research. This article focuses on the use of crossover designs in nutrition and dietetics research. The purpose is to distinguish the crossover design from the randomized clinical trial, define important terms, illustrate a 2×2 crossover design, discuss potential confounding variables in the crossover design, describe the analysis and interpretation of crossover data, present sample size considerations, provide examples of the use of the crossover design in nutrition and dietetics, and discuss additional considerations when the independent variable has more than two levels.
Collapse
|
23
|
Omer N, Le Deley MC, Piperno-Neumann S, Marec-Berard P, Italiano A, Corradini N, Bellera C, Brugières L, Gaspar N. Phase-II trials in osteosarcoma recurrences: A systematic review of past experience. Eur J Cancer 2017; 75:98-108. [DOI: 10.1016/j.ejca.2017.01.005] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 01/04/2017] [Indexed: 01/17/2023]
|
24
|
Rivalland G, Scott AM, John T. Standard of care in immunotherapy trials: Challenges and considerations. Hum Vaccin Immunother 2017; 13:2164-2178. [PMID: 28267397 DOI: 10.1080/21645515.2016.1277845] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The success of immunotherapeutics over the past decade has fundamentally altered the therapeutic landscape in melanoma and non-small cell lung (NSCLC) cancer care. Multiple clinical trials have confirmed significant improvements in survival with a variety of immunotherapeutic strategies. The careful and appropriate selection of standard of care (SOC) therapies is key to the successful design and interpretation of these trials. To date immunotherapeutic trials have used best supportive care, matched placebo, chemotherapy, targeted therapy or, more recently, established immunotherapeutics in melanoma clinical trials as SOCs. Each of these SOC choices has a fundamental impact on the selection and validity of response assessment criteria and clinical endpoints. As yet there is no established approach, thus new data must be interpreted with an understanding of the limitations of the current paradigm. Additionally, the pace of development has mandated the use of novel clinical trial designs, answering multiple therapeutic questions simultaneously and designed to expedite regulatory approval. This review addresses the most important challenges in the selection of SOC in immunotherapeutic trials and the current and future challenges in trial design.
Collapse
Affiliation(s)
- Gareth Rivalland
- a Department of Medical Oncology , Austin Health, Olivia-Newton John Cancer and Wellness Centre , Victoria , Australia.,b University of Melbourne , Victoria , Australia.,c The Olivia Newton-John Cancer Research Institute , Victoria , Australia
| | - Andrew M Scott
- b University of Melbourne , Victoria , Australia.,c The Olivia Newton-John Cancer Research Institute , Victoria , Australia.,d School of Cancer Medicine, La Trobe University , Victoria , Australia
| | - Thomas John
- a Department of Medical Oncology , Austin Health, Olivia-Newton John Cancer and Wellness Centre , Victoria , Australia.,b University of Melbourne , Victoria , Australia.,c The Olivia Newton-John Cancer Research Institute , Victoria , Australia.,d School of Cancer Medicine, La Trobe University , Victoria , Australia
| |
Collapse
|
25
|
Kim C, Prasad V. Strength of Validation for Surrogate End Points Used in the US Food and Drug Administration's Approval of Oncology Drugs. Mayo Clin Proc 2016; 91:S0025-6196(16)00125-7. [PMID: 27236424 PMCID: PMC5104665 DOI: 10.1016/j.mayocp.2016.02.012] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Revised: 01/21/2016] [Accepted: 02/09/2016] [Indexed: 01/09/2023]
Abstract
OBJECTIVE To determine the strength of the surrogate-survival correlation for cancer drug approvals based on a surrogate. PARTICIPANTS AND METHODS We performed a retrospective study of the US Food and Drug Administration (FDA) database, with focused searches of MEDLINE and Google Scholar. Among cancer drugs approved based on a surrogate end point, we examined previous publications assessing the strength of the surrogate-survival correlation. Specifically, we identified the percentage of surrogate approvals lacking any formal analysis of the strength of the surrogate-survival correlation, and when conducted, the strength of such correlations. RESULTS Between January 1, 2009, and December 31, 2014, the FDA approved marketing applications for 55 indications based on a surrogate, of which 25 were accelerated approvals and 30 were traditional approvals. We could not find any formal analyses of the strength of the surrogate-survival correlation in 14 out of 25 accelerated approvals (56%) and 11 out of 30 traditional approvals (37%). For accelerated approvals, just 4 approvals (16%) were made where a level 1 analysis (the most robust way to validate a surrogate) had been performed, with all 4 studies reporting low correlation (r≤0.7). For traditional approvals, a level 1 analysis had been performed for 15 approvals (50%): 8 (53%) reported low correlation (r≤0.7), 4 (27%) medium correlation (r>0.7 to r<0.85), and 3 (20%) high correlation (r≥0.85) with survival. CONCLUSIONS The use of surrogate end points for drug approval often lacks formal empirical verification of the strength of the surrogate-survival association.
Collapse
Affiliation(s)
- Chul Kim
- Medical Oncology Service, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Vinay Prasad
- Department of Medicine, Division of Hematology Oncology/Knight Cancer Institute, Oregon Health & Science University, Portland.
| |
Collapse
|
26
|
Martin JH, Phillips E, Thomas D, Somogyi AA. Adding the 'medicines' back into personalized medicine to improve cancer treatment outcomes. Br J Clin Pharmacol 2015; 80:929-31. [PMID: 26507561 DOI: 10.1111/bcp.12690] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 05/29/2015] [Indexed: 12/16/2022] Open
Affiliation(s)
- Jennifer H Martin
- School of Medicine and Public Health, University of Newcastle, New South Wales, Australia.,The University of Queensland Diamantina Institute, 37 Kent St, Woollongabba Queensland
| | - Elizabeth Phillips
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, Western, Australia
| | - David Thomas
- Garvan Institute of Medical Research, The Kinghorn Cancer Centre, 370 Victoria Street, Darlinghurst, NSW, 2010.,St Vincent's Clinical School, Faculty of Medicine, University of New South Wales
| | - Andrew A Somogyi
- Discipline of Pharmacology, Faculty of Health Sciences, University of Adelaide, Adelaide, 5001.,Department of Clinical Pharmacology, Royal Adelaide Hospital, Adelaide, Australia
| |
Collapse
|
27
|
[Challenges for clinical trials in oncology within the scope of early benefit assessment of drugs]. ZEITSCHRIFT FUR EVIDENZ FORTBILDUNG UND QUALITAET IM GESUNDHEITSWESEN 2015; 109:417-30. [PMID: 26474646 DOI: 10.1016/j.zefq.2015.08.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 08/14/2015] [Accepted: 08/14/2015] [Indexed: 12/22/2022]
Abstract
Until May 31, 2015 the German Institute for Quality and Efficiency in Health Care (IQWiG) conducted 108 assessments for various diseases on the basis of 103 dossiers within the scope of the early benefit assessment of drugs pursuant to the Act on the Reform of the Market for Medicinal Products (AMNOG). 29 of these assessments (28 dossiers) referred to advanced stages of oncologic (including neoplastic-hematologic) diseases. In 21 of these 29 assessments (72%), IQWiG found an added benefit for at least one subpopulation or subgroup, compared to 33% with non-oncologic diseases. For oncologic diseases, the extent of benefit was classified as "major" in six assessments (21%), compared to 5% for non-oncologic disorders. In contrast, the conclusions of the oncologic studies were less certain: only one assessment provided proof (of an added benefit); for non-oncologic diseases, this was the case in eight assessments. A distinctive methodological feature of the available oncologic studies is that, as a rule, treatment switching was planned in the event of progression (normally on the basis of imaging or laboratory findings) and that shortly afterwards the follow-up of important endpoints (adverse events and patient-reported outcomes) was normally discontinued. In particular, the pre-specified option in the study protocol allowing the control group to switch treatment to the experimental intervention after progression ("protocol-permitted treatment switches") makes it extremely difficult to interpret the results beyond the outcome "progression" (or progression-free survival). This treatment switching is mostly justified by reference to ethical necessity. This, however, alleges that the experimental intervention (i. e., the new drug) is superior to the control intervention, which means that circular reasoning is unavoidable. But despite this, oncologic studies are better than their reputation. Hence, so far the results of the early benefit assessment of new drugs (regarding the existence and extent of an added benefit) are clearly better than expected. In this context, the IQWiG methods have not been shown to be extremely conservative. On the contrary: in all cases where IQWiG rated the extent of benefit as "major" for the oncologic indications, the Federal Joint Committee (G-BA) has so far not shared this assessment, and instead rated the extent of benefit to be smaller.
Collapse
|
28
|
Prasad V, Berger VW. Hard-Wired Bias: How Even Double-Blind, Randomized Controlled Trials Can Be Skewed From the Start. Mayo Clin Proc 2015; 90:1171-5. [PMID: 26277702 PMCID: PMC4567484 DOI: 10.1016/j.mayocp.2015.05.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 05/03/2015] [Accepted: 05/12/2015] [Indexed: 12/11/2022]
Affiliation(s)
- Vinay Prasad
- Medical Oncology Branch, National Cancer Institute, National Institutes of Health, 10 Center Dr. 10/12N226, Bethesda, MD 20892, Phone: (219) 22900170; Fax (301) 402-1608
| | - Vance W. Berger
- National Cancer Institute and University of Maryland Baltimore County, Biometry Research Group, Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, 9609 Medical Center Drive, Rockville, MD 20850, (240) 276-7142 (voice)
| |
Collapse
|
29
|
Cherny NI, Sullivan R, Dafni U, Kerst JM, Sobrero A, Zielinski C, de Vries EGE, Piccart MJ. A standardised, generic, validated approach to stratify the magnitude of clinical benefit that can be anticipated from anti-cancer therapies: the European Society for Medical Oncology Magnitude of Clinical Benefit Scale (ESMO-MCBS). Ann Oncol 2015; 26:1547-73. [PMID: 26026162 DOI: 10.1093/annonc/mdv249] [Citation(s) in RCA: 595] [Impact Index Per Article: 66.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 05/22/2015] [Indexed: 12/12/2022] Open
Abstract
The value of any new therapeutic strategy or treatment is determined by the magnitude of its clinical benefit balanced against its cost. Evidence for clinical benefit from new treatment options is derived from clinical research, in particular phase III randomised trials, which generate unbiased data regarding the efficacy, benefit and safety of new therapeutic approaches. To date, there is no standard tool for grading the magnitude of clinical benefit of cancer therapies, which may range from trivial (median progression-free survival advantage of only a few weeks) to substantial (improved long-term survival). Indeed, in the absence of a standardised approach for grading the magnitude of clinical benefit, conclusions and recommendations derived from studies are often hotly disputed and very modest incremental advances have often been presented, discussed and promoted as major advances or 'breakthroughs'. Recognising the importance of presenting clear and unbiased statements regarding the magnitude of the clinical benefit from new therapeutic approaches derived from high-quality clinical trials, the European Society for Medical Oncology (ESMO) has developed a validated and reproducible tool to assess the magnitude of clinical benefit for cancer medicines, the ESMO Magnitude of Clinical Benefit Scale (ESMO-MCBS). This tool uses a rational, structured and consistent approach to derive a relative ranking of the magnitude of clinically meaningful benefit that can be expected from a new anti-cancer treatment. The ESMO-MCBS is an important first step to the critical public policy issue of value in cancer care, helping to frame the appropriate use of limited public and personal resources to deliver cost-effective and affordable cancer care. The ESMO-MCBS will be a dynamic tool and its criteria will be revised on a regular basis.
Collapse
Affiliation(s)
- N I Cherny
- Cancer Pain and Palliative Medicine Service, Department of Medical Oncology, Shaare Zedek Medical Center, Jerusalem, Israel
| | - R Sullivan
- Kings Health Partners Integrated Cancer Centre, King's College London, Institute of Cancer Policy, London, UK
| | - U Dafni
- University of Athens and Frontiers of Science Foundation-Hellas, Athens, Greece
| | - J M Kerst
- Department of Medical Oncology, Antoni van Leeuwenhoek Hospital
| | - A Sobrero
- Department of Medical Oncology, IRCCS San Martino IST, Genova, Italy
| | - C Zielinski
- Division of Oncology, Medical University Vienna, Vienna, Austria
| | - E G E de Vries
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - M J Piccart
- Jules Bordet Institute, UniversitéLibre de Bruxelles, Brussels, Belgium Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
30
|
Hoskins PJ, Fung-Kee-Fung M, Miller D, Gotlieb W. Time for a Level Playing Field: Inequalities in Regulatory/Approval Processes—The Example of Bevacizumab in Epithelial Ovarian Cancer. J Clin Oncol 2015; 33:1539-1542. [DOI: 10.1200/jco.2014.58.3690] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
Affiliation(s)
- Paul J. Hoskins
- British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | | | - Dianne Miller
- Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | | |
Collapse
|
31
|
García M, Navarro V, Clopés A. Clinical End Points and Relevant Clinical Benefits in Advanced Colorectal Cancer Trials. CURRENT COLORECTAL CANCER REPORTS 2014. [DOI: 10.1007/s11888-014-0227-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|