1
|
Ashton KE, Price C, Fleming L, Blom AW, Culliford L, Evans RN, Foster NE, Hollingworth W, Jameson C, Jeynes N, Moore AJ, Orpen N, Palmer C, Reeves BC, Rogers CA, Wylde V. Effectiveness and cost-effectiveness of radiofrequency denervation versus placebo for chronic and moderate to severe low back pain: study protocol for the RADICAL randomised controlled trial. BMJ Open 2024; 14:e079173. [PMID: 39067879 DOI: 10.1136/bmjopen-2023-079173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/30/2024] Open
Abstract
INTRODUCTION Low back pain (LBP) is the leading global cause of disability. Patients with moderate to severe LBP who respond positively to a diagnostic medial nerve branch block can be offered radiofrequency denervation (RFD). However, high-quality evidence on the effectiveness of RFD is lacking. METHODS AND ANALYSIS RADICAL (RADIofrequenCy denervAtion for Low back pain) is a double-blind, parallel-group, superiority randomised controlled trial. A total of 250 adults listed for RFD will be recruited from approximately 20 National Health Service (NHS) pain and spinal clinics. Recruitment processes will be optimised through qualitative research during a 12-month internal pilot phase. Participants will be randomised in theatre using a 1:1 allocation ratio to RFD or placebo. RFD technique will follow best practice guidelines developed for the trial. Placebo RFD will follow the same protocol, but the electrode tip temperature will not be raised. Participants who do not experience a clinically meaningful improvement in pain 3 months after randomisation will be offered the alternative intervention to the one provided at the outset without disclosing the original allocation. The primary clinical outcome will be pain severity, measured using a pain Numeric Rating Scale, at 3 months after randomisation. Secondary outcomes will be assessed up to 2 years after randomisation and include disability, health-related quality of life, psychological distress, time to pain recovery, satisfaction, adverse events, work outcomes and healthcare utilisation. The primary statistical analyses will be by intention to treat and will follow a prespecified analysis plan. The primary economic evaluation will take an NHS and social services perspective and estimate the discounted cost per quality-adjusted life-year and incremental net benefit of RFD over the 2-year follow-up period. ETHICS AND DISSEMINATION Ethics approval was obtained from the London-Fulham Research Ethics Committee (21/LO/0471). Results will be disseminated in open-access publications and plain language summaries. TRIAL REGISTRATION NUMBER ISRCTN16473239.
Collapse
Affiliation(s)
- Kate E Ashton
- Bristol Trials Centre, Bristol Medical School, University of Bristol, Bristol, UK
| | | | - Leah Fleming
- Bristol Trials Centre, Bristol Medical School, University of Bristol, Bristol, UK
| | - Ashley W Blom
- Faculty of Health, The University of Sheffield, Sheffield, UK
| | - Lucy Culliford
- Bristol Trials Centre, Bristol Medical School, University of Bristol, Bristol, UK
| | - Rebecca Nicole Evans
- Bristol Trials Centre, Bristol Medical School, University of Bristol, Bristol, UK
| | - Nadine E Foster
- STARS Education and Research Alliance, Surgical Treatment and Rehabilitation Service (STARS), The University of Queensland, Saint Lucia, Queensland, Australia
| | - William Hollingworth
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Catherine Jameson
- Musculoskeletal Research Unit, Bristol Medical School, University of Bristol, Bristol, UK
- NIHR Bristol Biomedical Research Centre, University Hospitals Bristol and Weston NHS Foundation Trust and University of Bristol, Bristol, UK
| | - Nouf Jeynes
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Andrew J Moore
- Musculoskeletal Research Unit, Bristol Medical School, University of Bristol, Bristol, UK
| | - Neil Orpen
- BMI Healthcare, The Ridgeway Hospital, Swindon, UK
| | - Cecily Palmer
- Musculoskeletal Research Unit, Bristol Medical School, University of Bristol, Bristol, UK
| | - Barnaby C Reeves
- Bristol Trials Centre, Bristol Medical School, University of Bristol, Bristol, UK
| | - Chris A Rogers
- Bristol Trials Centre, Bristol Medical School, University of Bristol, Bristol, UK
| | - Vikki Wylde
- Musculoskeletal Research Unit, Bristol Medical School, University of Bristol, Bristol, UK
- NIHR Bristol Biomedical Research Centre, University Hospitals Bristol and Weston NHS Foundation Trust and University of Bristol, Bristol, UK
| |
Collapse
|
2
|
Motzer RJ, Porta C, Eto M, Powles T, Grünwald V, Hutson TE, Alekseev B, Rha SY, Merchan J, Goh JC, Lalani AKA, De Giorgi U, Melichar B, Hong SH, Gurney H, Méndez-Vidal MJ, Kopyltsov E, Tjulandin S, Gordoa TA, Kozlov V, Alyasova A, Winquist E, Maroto P, Kim M, Peer A, Procopio G, Takagi T, Wong S, Bedke J, Schmidinger M, Rodriguez-Lopez K, Burgents J, He C, Okpara CE, McKenzie J, Choueiri TK. Lenvatinib Plus Pembrolizumab Versus Sunitinib in First-Line Treatment of Advanced Renal Cell Carcinoma: Final Prespecified Overall Survival Analysis of CLEAR, a Phase III Study. J Clin Oncol 2024; 42:1222-1228. [PMID: 38227898 PMCID: PMC11095851 DOI: 10.1200/jco.23.01569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/07/2023] [Accepted: 11/12/2023] [Indexed: 01/18/2024] Open
Abstract
Clinical trials frequently include multiple end points that mature at different times. The initial report, typically based on the primary end point, may be published when key planned co-primary or secondary analyses are not yet available. Clinical trial updates provide an opportunity to disseminate additional results from studies, published in JCO or elsewhere, for which the primary end point has already been reported.We present the final prespecified overall survival (OS) analysis of the open-label, phase III CLEAR study in treatment-naïve patients with advanced renal cell carcinoma (aRCC). With an additional follow-up of 23 months from the primary analysis, we report results from the lenvatinib plus pembrolizumab versus sunitinib comparison of CLEAR. Treatment-naïve patients with aRCC were randomly assigned to receive lenvatinib (20 mg orally once daily in 21-day cycles) plus pembrolizumab (200 mg intravenously once every 3 weeks) or sunitinib (50 mg orally once daily [4 weeks on/2 weeks off]). At this data cutoff date (July 31, 2022), the OS hazard ratio (HR) was 0.79 (95% CI, 0.63 to 0.99). The median OS (95% CI) was 53.7 months (95% CI, 48.7 to not estimable [NE]) with lenvatinib plus pembrolizumab versus 54.3 months (95% CI, 40.9 to NE) with sunitinib; 36-month OS rates (95% CI) were 66.4% (95% CI, 61.1 to 71.2) and 60.2% (95% CI, 54.6 to 65.2), respectively. The median progression-free survival (95% CI) was 23.9 months (95% CI, 20.8 to 27.7) with lenvatinib plus pembrolizumab and 9.2 months (95% CI, 6.0 to 11.0) with sunitinib (HR, 0.47 [95% CI, 0.38 to 0.57]). Objective response rate also favored the combination over sunitinib (71.3% v 36.7%; relative risk 1.94 [95% CI, 1.67 to 2.26]). Treatment-emergent adverse events occurred in >90% of patients who received either treatment. In conclusion, lenvatinib plus pembrolizumab achieved consistent, durable benefit with a manageable safety profile in treatment-naïve patients with aRCC.
Collapse
Affiliation(s)
| | - Camillo Porta
- University of Bari “A. Moro,” Bari, Italy
- University of Pavia, Pavia, Italy
| | | | | | | | | | - Boris Alekseev
- P.A. Herzen Moscow Oncological Research Institute, Moscow, Russia
| | - Sun Young Rha
- Yonsei Cancer Center, Yonsei University Health System, Seoul, South Korea
| | - Jaime Merchan
- University of Miami Sylvester Comprehensive Cancer Center, Miami, FL
| | - Jeffrey C. Goh
- ICON Research, South Brisbane & Queensland University of Technology, Brisbane, Queensland, Australia
| | - Aly-Khan A. Lalani
- Juravinski Cancer Centre, McMaster University, Hamilton, Ontario, Canada
| | - Ugo De Giorgi
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) Dino Amadori, Meldola, Italy
| | - Bohuslav Melichar
- Palacky University, and University Hospital Olomouc, Olomouc, Czech Republic
| | - Sung-Hoo Hong
- Seoul St Mary's Hospital, The Catholic University of Korea, Seoul, South Korea
| | | | - María José Méndez-Vidal
- Maimonides Institute for Biomedical research of Cordoba (IMIBIC) Hospital Universitario Reina Sofía, Medical Oncology Department, Córdoba, Spain
| | - Evgeny Kopyltsov
- State Institution of Healthcare “Regional Clinical Oncology Dispensary,” Omsk, Russia
| | - Sergei Tjulandin
- N N Blokhin National Medical Research Center for Oncology, Ministry of Health of the Russian Federation, Moscow, Russia
| | | | - Vadim Kozlov
- State budgetary Health Care Institution “Novosibirsk Regional Clinical Oncology Dispensary,” Novosibirsk, Russia
| | | | | | - Pablo Maroto
- Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Miso Kim
- Seoul National University Hospital, Seoul, South Korea
| | | | | | | | | | - Jens Bedke
- Department of Urology and Transplantation Surgery, Klinikum Stuttgart, Stuttgart, Germany
| | | | | | | | | | | | | | | |
Collapse
|
3
|
Latimer NR, Dewdney A, Campioni M. A cautionary tale: an evaluation of the performance of treatment switching adjustment methods in a real world case study. BMC Med Res Methodol 2024; 24:17. [PMID: 38253996 PMCID: PMC10802004 DOI: 10.1186/s12874-024-02140-6] [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: 04/11/2023] [Accepted: 01/02/2024] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND Treatment switching in randomised controlled trials (RCTs) is a problem for health technology assessment when substantial proportions of patients switch onto effective treatments that would not be available in standard clinical practice. Often statistical methods are used to adjust for switching: these can be applied in different ways, and performance has been assessed in simulation studies, but not in real-world case studies. We assessed the performance of adjustment methods described in National Institute for Health and Care Excellence Decision Support Unit Technical Support Document 16, applying them to an RCT comparing panitumumab to best supportive care (BSC) in colorectal cancer, in which 76% of patients randomised to BSC switched onto panitumumab. The RCT resulted in intention-to-treat hazard ratios (HR) for overall survival (OS) of 1.00 (95% confidence interval [CI] 0.82-1.22) for all patients, and 0.99 (95% CI 0.75-1.29) for patients with wild-type KRAS (Kirsten rat sarcoma virus). METHODS We tested several applications of inverse probability of censoring weights (IPCW), rank preserving structural failure time models (RPSFTM) and simple and complex two-stage estimation (TSE) to estimate treatment effects that would have been observed if BSC patients had not switched onto panitumumab. To assess the performance of these analyses we ascertained the true effectiveness of panitumumab based on: (i) subsequent RCTs of panitumumab that disallowed treatment switching; (ii) studies of cetuximab that disallowed treatment switching, (iii) analyses demonstrating that only patients with wild-type KRAS benefit from panitumumab. These sources suggest the true OS HR for panitumumab is 0.76-0.77 (95% CI 0.60-0.98) for all patients, and 0.55-0.73 (95% CI 0.41-0.93) for patients with wild-type KRAS. RESULTS Some applications of IPCW and TSE provided treatment effect estimates that closely matched the point-estimates and CIs of the expected truths. However, other applications produced estimates towards the boundaries of the expected truths, with some TSE applications producing estimates that lay outside the expected true confidence intervals. The RPSFTM performed relatively poorly, with all applications providing treatment effect estimates close to 1, often with extremely wide confidence intervals. CONCLUSIONS Adjustment analyses may provide unreliable results. How each method is applied must be scrutinised to assess reliability.
Collapse
Affiliation(s)
- Nicholas R Latimer
- Sheffield Centre for Health and Related Research (SCHARR), University of Sheffield, Regent Court, 30 Regent Street, Sheffield, South Yorkshire, S1 4DA, UK.
- Delta Hat Limited, Nottingham, UK.
| | - Alice Dewdney
- Weston Park Cancer Centre, Sheffield Teaching Hospital, Sheffield, UK
| | | |
Collapse
|
4
|
Chen P, Liu M, Li GY, Sun F, Li T. Misadjustment of post-trial life-prolonging therapies in the second interim analysis of the MAGNITUDE trial. Ann Oncol 2024; 35:140-141. [PMID: 37871700 DOI: 10.1016/j.annonc.2023.10.128] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 10/10/2023] [Accepted: 10/17/2023] [Indexed: 10/25/2023] Open
Affiliation(s)
- P Chen
- Department of Urology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - M Liu
- Department of Urology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - G Y Li
- Department of Urology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - F Sun
- Department of Urology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - T Li
- Department of Urology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China.
| |
Collapse
|
5
|
Goldkuhle M, Guyatt GH, Kreuzberger N, Akl EA, Dahm P, van Dalen EC, Hemkens LG, Klugar M, Mustafa RA, Nonino F, Schünemann HJ, Trivella M, Skoetz N. GRADE concept 4: rating the certainty of evidence when study interventions or comparators differ from PICO targets. J Clin Epidemiol 2023; 159:40-48. [PMID: 37146659 DOI: 10.1016/j.jclinepi.2023.04.018] [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: 01/11/2023] [Revised: 04/13/2023] [Accepted: 04/26/2023] [Indexed: 05/07/2023]
Abstract
OBJECTIVES This Grading of Recommendations Assessment, Development and Evaluation (GRADE) concept article offers systematic reviewers, guideline authors, and other users of evidence assistance in addressing randomized trial situations in which interventions or comparators differ from those in the target people, interventions, comparators, and outcomes. To clarify what GRADE considers under indirectness of interventions and comparators, we focus on a particular example: when comparator arm participants receive some or all aspects of the intervention management strategy (treatment switching). STUDY DESIGN AND SETTING An interdisciplinary panel of the GRADE working group members developed this concept article through an iterative review of examples in multiple teleconferences, small group sessions, and e-mail correspondence. After presentation at a GRADE working group meeting in November 2022, attendees approved the final concept paper, which we support with examples from systematic reviews and individual trials. RESULTS In the presence of safeguards against risk of bias, trials provide unbiased estimates of the effect of an intervention on the people as enrolled, the interventions as implemented, the comparators as implemented, and the outcomes as measured. Within the GRADE framework, differences in the people, interventions, comparators, and outcomes elements between the review or guideline recommendation targets and the trials as implemented constitute issues of indirectness. The intervention or comparator group management strategy as implemented, when it differs from the target comparator, constitutes one potential source of indirectness: Indirectness of interventions and comparators-comparator group receipt of the intervention constitutes a specific subcategory of said indirectness. The proportion of comparator arm participants that received the intervention and the apparent magnitude of effect bear on whether one should rate down, and if one does, to what extent. CONCLUSION Treatment switching and other differences between review or guideline recommendation target interventions and comparators vs. interventions and comparators as implemented in otherwise relevant trials are best considered issues of indirectness.
Collapse
Affiliation(s)
- Marius Goldkuhle
- Evidence-based Medicine, Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany.
| | - Gordon H Guyatt
- Department of Health Research Methods, Evidence, and Impact, Michael G DeGroote Cochrane Canada Centre, Cochrane Canada, McMaster GRADE Centre and Department of Medicine, McMaster University, 1280 Main St. W., Hamilton, ON L8S 4K1, Canada
| | - Nina Kreuzberger
- Evidence-based Medicine, Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Elie A Akl
- Department of Internal Medicine, American University of Beirut, Lebanon, P.O.Box 11-0236 and Department of Health Research Methods, Evidence, and Impact, McMaster University, 1280 Main St. W., Hamilton, ON L8S 4K1, Canada
| | - Philipp Dahm
- Minneapolis VA Health Care System, Urology Section 112D, One Veterans Drive, Minneapolis, Minnesota 55417
| | - Elvira C van Dalen
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584CS Utrecht, the Netherlands
| | - Lars G Hemkens
- Department of Clinical Research, University Hospital Basel, University of Basel, Basel, Switzerland; Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel, Switzerland; Meta-Research Innovation Center at Stanford (METRICS), Stanford University, Stanford, CA, USA; Meta-Research Innovation Center Berlin (METRIC-B), Berlin Institute of Health, Berlin, Germany
| | - Miloslav Klugar
- Czech National Centre for Evidence-Based Healthcare and Knowledge Translation (Cochrane Czech Republic, Czech EBHC: JBI Centre of Excellence, Masaryk University GRADE Centre), Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic; Institute of Health Information and Statistics of the Czech Republic, 100 00 Prague, Czech Republic
| | - Reem A Mustafa
- Department of Medicine and Population Health, University of Kansas Health System, 3901 Rainbow Blvd, MS3002, Kansas City, KS 66160, USA; Department of Health Research Methods, Evidence, and Impact, McMaster University, 1280 Main St. W., Hamilton, Ontario L8S 4K1, Canada
| | - Francesco Nonino
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Unit of Epidemiology and Statistics, Cochrane Review Group Multiple Sclerosis and Rare Diseases of the CNS, Via Altura 3, 40139 Bologna, Italy
| | - Holger J Schünemann
- Department of Health Research Methods, Evidence, and Impact, Michael G DeGroote Cochrane Canada Centre, Cochrane Canada and McMaster GRADE Centre, McMaster University, Hamilton, Ontario, Canada; Department of Medicine, McMaster University, Hamilton, Ontario, Canada; Department of Biomedical Sciences, Humanitas University, Milan, Italy; Cochrane Canada, Hamilton, Ontario, Canada
| | - Marialene Trivella
- Department of Cardiovascular Medicine, John Radcliffe Hospital, University of Oxford, UK; Department of Population Health, London School of Hygiene and Tropical Medicine, London
| | - Nicole Skoetz
- Evidence-based Medicine, Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| |
Collapse
|
6
|
Atkins MB, Abu-Sbeih H, Ascierto PA, Bishop MR, Chen DS, Dhodapkar M, Emens LA, Ernstoff MS, Ferris RL, Greten TF, Gulley JL, Herbst RS, Humphrey RW, Larkin J, Margolin KA, Mazzarella L, Ramalingam SS, Regan MM, Rini BI, Sznol M. Maximizing the value of phase III trials in immuno-oncology: A checklist from the Society for Immunotherapy of Cancer (SITC). J Immunother Cancer 2022; 10:jitc-2022-005413. [PMID: 36175037 PMCID: PMC9528604 DOI: 10.1136/jitc-2022-005413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/20/2022] [Indexed: 11/03/2022] Open
Abstract
The broad activity of agents blocking the programmed cell death protein 1 and its ligand (the PD-(L)1 axis) revolutionized oncology, offering long-term benefit to patients and even curative responses for tumors that were once associated with dismal prognosis. However, only a minority of patients experience durable clinical benefit with immune checkpoint inhibitor monotherapy in most disease settings. Spurred by preclinical and correlative studies to understand mechanisms of non-response to the PD-(L)1 antagonists and by combination studies in animal tumor models, many drug development programs were designed to combine anti-PD-(L)1 with a variety of approved and investigational chemotherapies, tumor-targeted therapies, antiangiogenic therapies, and other immunotherapies. Several immunotherapy combinations improved survival outcomes in a variety of indications including melanoma, lung, kidney, and liver cancer, among others. This immunotherapy renaissance, however, has led to many combinations being advanced to late-stage development without definitive predictive biomarkers, limited phase I and phase II data, or clinical trial designs that are not optimized for demonstrating the unique attributes of immune-related antitumor activity-for example, landmark progression-free survival and overall survival. The decision to activate a study at an individual site is investigator-driven, and generalized frameworks to evaluate the potential for phase III trials in immuno-oncology to yield positive data, particularly to increase the number of curative responses or otherwise advance the field have thus far been lacking. To assist in evaluating the potential value to patients and the immunotherapy field of phase III trials, the Society for Immunotherapy of Cancer (SITC) has developed a checklist for investigators, described in this manuscript. Although the checklist focuses on anti-PD-(L)1-based combinations, it may be applied to any regimen in which immune modulation is an important component of the antitumor effect.
Collapse
Affiliation(s)
- Michael B Atkins
- Georgetown Lombardi Comprehensive Cancer Center, Washington, District of Columbia, USA
| | | | - Paolo A Ascierto
- Istituto Nazionale Tumori IRCCS Fondazione "G Pascale", Napoli, Italy
| | - Michael R Bishop
- The David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago, Illinois, USA
| | - Daniel S Chen
- Engenuity Life Sciences, Burlingame, California, USA
| | - Madhav Dhodapkar
- Center for Cancer Immunology, Winship Cancer Institute at Emory University, Atlanta, Georgia, USA
| | - Leisha A Emens
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Marc S Ernstoff
- DCTD/DTP-IOB, ImmunoOncology Branch, NCI, Bethesda, Maryland, USA
| | | | - Tim F Greten
- Gastrointestinal Malignancies Section, National Cancer Institue CCR Liver Program, Bethesda, Maryland, USA
| | - James L Gulley
- Center for Immuno-Oncology, National Cancer Institute, Bethesda, Maryland, USA
| | | | | | | | - Kim A Margolin
- St. John's Cancer Institute, Santa Monica, California, USA
| | - Luca Mazzarella
- Experimental Oncology, New Drug Development, European Instititue of Oncology IRCCS, Milan, Italy
| | | | - Meredith M Regan
- Dana-Farber/Harvard Cancer Center, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | | | - Mario Sznol
- Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
7
|
Di Scala L, Bacchi M, Bayer B, Turricchia S. Adjusting Overall Survival Estimates of Macitentan in Pulmonary Arterial Hypertension After Treatment Switching: Results from the SERAPHIN Study. Adv Ther 2022; 39:4346-4358. [PMID: 35917059 PMCID: PMC9402487 DOI: 10.1007/s12325-022-02253-8] [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: 06/01/2022] [Accepted: 06/30/2022] [Indexed: 11/30/2022]
Abstract
Introduction Evaluating overall survival in randomized controlled trials (RCTs) can often be confounded by bias introduced by treatment switching. SERAPHIN was a large RCT that evaluated the effects of long-term treatment with the endothelin receptor antagonist macitentan in patients with pulmonary arterial hypertension. In an intent-to-treat (ITT) analysis, a non-significant decrease in the risk of all-cause mortality up to study closure was reported with macitentan 10 mg versus placebo. As patients could switch treatment when experiencing symptoms of disease progression, this analysis attempts to adjust for the confounding effects on overall survival. Methods The inverse probability of censoring weighted (IPCW) and rank-preserving structural failure time (RPSFT) models were used to estimate the treatment effect on overall mortality had there been no treatment switching in SERAPHIN. Time to all-cause death was evaluated up to study closure. Treatment switching was defined as patients in the placebo group switching to open-label macitentan 10 mg, and patients in the macitentan 10 mg group prematurely discontinuing macitentan. Results By study closure, 73.2% (183/250) of patients in the placebo group had switched to macitentan 10 mg. Among these patients, exposure time to macitentan 10 mg represented 28.2% of total study treatment exposure (cumulative exposure 134.6 patient-years). At study closure, 24.8% (60/242) of patients in the macitentan 10 mg group were not receiving open-label macitentan; mean time not receiving macitentan was 44.3 weeks. The adjusted hazard ratios (HR) for overall survival using the IPCW and RPSFT methods were lower (HR 0.42, 95% confidence interval [CI] 0.22, 0.81; p = 0.009, and HR 0.33, 95% CI 0.04, 2.83, respectively) than the ITT unadjusted HR (0.80, 95% CI 0.51, 1.24). Conclusion These results from the current analyses indicate that in SERAPHIN, the standard ITT analysis was confounded by treatment switching resulting in an underestimation of the benefit of macitentan 10 mg on overall survival. By adjusting for switching, the IPCW and RPSFT models estimated a 58% and 67% reduction in risk of mortality, respectively, with macitentan 10 mg versus placebo. Trial registration ClinicalTrials.gov identifier: NCT00660179. Supplementary Information The online version contains supplementary material available at 10.1007/s12325-022-02253-8.
Collapse
Affiliation(s)
- Lilla Di Scala
- Market Access, Janssen, Actelion Pharmaceuticals Ltd, 4123, Allschwil, Switzerland.
| | - Marisa Bacchi
- Statistics and Decision Sciences, Global Development, Actelion Pharmaceuticals Ltd, 4123, Allschwil, Switzerland
| | - Bjørn Bayer
- Global Market Access and Pricing, Actelion Pharmaceuticals Ltd, 4123, Allschwil, Switzerland
| | - Stefano Turricchia
- Global Medical Affairs, Actelion Pharmaceuticals Ltd, 4123, Allschwil, Switzerland
| |
Collapse
|
8
|
Kuehne F, Rochau U, Paracha N, Yeh JM, Sabate E, Siebert U. Estimating Treatment-Switching Bias in a Randomized Clinical Trial of Ovarian Cancer Treatment: Combining Causal Inference with Decision-Analytic Modeling. Med Decis Making 2021; 42:194-207. [PMID: 34666553 DOI: 10.1177/0272989x211026288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Bevacizumab is efficacious in delaying ovarian cancer progression and controlling ascites. The ICON7 trial showed a significant benefit in overall survival for bevacizumab, whereas the GOG-218 trial did not. GOG-218 allowed control group patients to switch to bevacizumab upon progression, which may have biased the results. Lack of data on switching behavior prevented the application of g-methods to adjust for switching. The objective of this study was to apply decision-analytic modeling to estimate the impact of switching bias on causal treatment-effect estimates. METHODS We developed a causal decision-analytic Markov model (CDAMM) to emulate the GOG-218 trial and estimate overall survival. CDAMM input parameters were based on data from randomized clinical trials and the published literature. Overall switching proportion was based on GOG-218 trial information, whereas the proportion switching with and without ascites was estimated using calibration. We estimated the counterfactual treatment effect that would have been observed had no switching occurred by denying switching in the CDAMM. RESULTS The survival curves generated by the CDAMM matched well with the ones reported in the GOG-218 trial. The survival curve correcting for switching showed an estimated bias such that 79% of the true treatment effect could not be observed in the GOG-218 trial. Results were most sensitive to changes in the proportion progressing with severe ascites and mortality. LIMITATIONS We used a simplified model structure and based model parameters on published data and assumptions. Robustness of the CDAMM was tested and model assumptions transparently reported. CONCLUSIONS Medical-decision science methods may be merged with empirical methods of causal inference to integrate data from other sources where empirical data are not sufficient. We recommend collecting sufficient information on switching behavior when switching cannot be avoided.
Collapse
Affiliation(s)
- Felicitas Kuehne
- Department of Public Health, Health Services Research and Health Technology Assessment, Institute of Public Health, Medical Decision Making and Health Technology Assessment, UMIT-University for Health Sciences, Medical Informatics and Technology, Hall in Tirol, Austria
| | - Ursula Rochau
- Department of Public Health, Health Services Research and Health Technology Assessment, Institute of Public Health, Medical Decision Making and Health Technology Assessment, UMIT-University for Health Sciences, Medical Informatics and Technology, Hall in Tirol, Austria
| | - Noman Paracha
- Bayer Consumer Care AG, Pharmaceuticals, Oncology SBU, Basel, Basel-Stadt, Switzerland
| | - Jennifer M Yeh
- Department of Pediatrics, Harvard Medical School & Boston Children's Hospital
| | | | - Uwe Siebert
- Department of Public Health, Health Services Research and Health Technology Assessment, Institute of Public Health, Medical Decision Making and Health Technology Assessment, UMIT-University for Health Sciences, Medical Informatics and Technology, Hall in Tirol, Austria.,Division of Health Technology Assessment, ONCOTYROL-Center for Personalized Cancer Medicine, Innsbruck, Austria.,Center for Health Decision Science, Departments of Epidemiology and Health Policy & Management, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Institute for Technology Assessment and Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
9
|
Manitz J, Kan-Dobrosky N, Buchner H, Casadebaig ML, Degtyarev E, Dey J, Haddad V, Jie F, Martin E, Mo M, Rufibach K, Shentu Y, Stalbovskaya V, Sammi Tang R, Yung G, Zhou J. Estimands for overall survival in clinical trials with treatment switching in oncology. Pharm Stat 2021; 21:150-162. [PMID: 34605168 DOI: 10.1002/pst.2158] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 04/28/2021] [Accepted: 07/10/2021] [Indexed: 11/09/2022]
Abstract
An addendum of the ICH E9 guideline on Statistical Principles for Clinical Trials was released in November 2019 introducing the estimand framework. This new framework aims to align trial objectives and statistical analyses by requiring a precise definition of the inferential quantity of interest, that is, the estimand. This definition explicitly accounts for intercurrent events, such as switching to new anticancer therapies for the analysis of overall survival (OS), the gold standard in oncology. Traditionally, OS in confirmatory studies is analyzed using the intention-to-treat (ITT) approach comparing treatment groups as they were initially randomized regardless of whether treatment switching occurred and regardless of any subsequent therapy (treatment-policy strategy). Regulatory authorities and other stakeholders often consider ITT results as most relevant. However, the respective estimand only yields a clinically meaningful comparison of two treatment arms if subsequent therapies are already approved and reflect clinical practice. We illustrate different scenarios where subsequent therapies are not yet approved drugs and thus do not reflect clinical practice. In such situations the hypothetical strategy could be more meaningful from patient's and prescriber's perspective. The cross-industry Oncology Estimand Working Group (www.oncoestimand.org) was initiated to foster a common understanding and consistent implementation of the estimand framework in oncology clinical trials. This paper summarizes the group's recommendations for appropriate estimands in the presence of treatment switching, one of the key intercurrent events in oncology clinical trials. We also discuss how different choices of estimands may impact study design, data collection, trial conduct, analysis, and interpretation.
Collapse
Affiliation(s)
- Juliane Manitz
- Global Biostatistics, EMD Serono, Billerica, Massachusetts, USA
| | | | - Hannes Buchner
- Biostatistics and Data Science, Staburo GmbH, Munich, Germany
| | | | - Evgeny Degtyarev
- Clinical Development and Analytics, Novartis, Basel, Switzerland
| | - Jyotirmoy Dey
- Data and Statistical Sciences, AbbVie Inc., North Chicago, Illinois, USA
| | | | - Fei Jie
- Biostatistics and Data Management, Daiichi Sankyo Inc, Basking Ridge, New Jersey, USA
| | - Emily Martin
- Global Biostatistics, EMD Serono, Billerica, Massachusetts, USA
| | - Mindy Mo
- Oncology Clinical Statistics US, Bayer, Whippany, New Jersey, USA
| | - Kaspar Rufibach
- Methods, Collaboration, and Outreach, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Yue Shentu
- Biostatistics and Research Decision Sciences, Merck & Co., Inc., Kenilworth, New Jersey, USA
| | | | - Rui Sammi Tang
- Global Biometric, Servier Pharmaceuticals, Boston, Massachusetts, USA
| | - Godwin Yung
- Methods, Collaboration, and Outreach, Genentech, San Francisco, California, USA
| | | |
Collapse
|
10
|
Exploring the Impact of Treatment Switching on Overall Survival from the PROfound Study in Homologous Recombination Repair (HRR)-Mutated Metastatic Castration-Resistant Prostate Cancer (mCRPC). Target Oncol 2021; 16:613-623. [PMID: 34478046 PMCID: PMC8484203 DOI: 10.1007/s11523-021-00837-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/20/2021] [Indexed: 11/29/2022]
Abstract
Background In oncology trials, treatment switching from the comparator to the experimental regimen is often allowed but may lead to underestimating overall survival (OS) of an experimental therapy. Objective This study evaluates the impact of treatment switching from control to olaparib on OS using the final survival data from the PROfound study and compares validated adjustment methods to estimate the magnitude of OS benefit with olaparib. Patients and methods The primary population from PROfound (Cohort A) was included, alongside two populations approved for treatment with olaparib by the European Medicines Agency and US Food and Drug Administration: BRCAm and Cohort A+B (excluding the PPP2R2A gene). Five methods were explored to adjust for switching: excluding or censoring patients in the control arm who receive subsequent olaparib, Rank Preserving Structural Failure Time Model (RPSFTM), Inverse Probability of Censoring Weights, and Two-Stage Estimation. Results The RPSFTM was considered the most appropriate approach for PROfound as the results were robust to sensitivity analysis testing of the common treatment effect assumption. For Cohort A, the final OS hazard ratio reduced from 0.69 (95% CI 0.5–0.97) to between 0.42 (0.18–0.90) and 0.52 (0.31–1.00) for olaparib versus control, depending on the RPSFTM selected. Median OS reduced from 14.7 months to between 11.73 and 12.63 months for control. Conclusions The magnitude of the statistically significant (P < 0.05) survival benefit of olaparib versus control observed in Cohort A of PROfound is likely to be underestimated if adjustment for treatment switching from control to olaparib is not conducted. The RPSFTM was considered the most plausible method, although further development and validation of robust methods to estimate the magnitude of impact of treatment switching is needed. Supplementary Information The online version contains supplementary material available at 10.1007/s11523-021-00837-y.
Collapse
|
11
|
Xu Y, Wu M, He W, Liao Q, Mai Y. Teasing Out the Overall Survival Benefit With Adjustment for Treatment Switching to Multiple Treatments. Stat Biopharm Res 2021. [DOI: 10.1080/19466315.2021.1914716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Yuqing Xu
- Sanofi U.S. Biostatistics and Programming, Bridgewater, NJ
| | - Meijing Wu
- AbbVie Inc. Data and Statistical Sciences, North Chicago, IL
| | - Weili He
- AbbVie Inc. Data and Statistical Sciences, North Chicago, IL
| | - Qiming Liao
- Department of Statistics, ViiV Healthcare, Raleigh-Durham, NC
| | - Yabing Mai
- Biostatistics Asia, Boehringer Ingelheim, Shanghai, China
| |
Collapse
|
12
|
Ananthakrishnan R, Green S, Previtali A, Liu R, Li D, LaValley M. Critical review of oncology clinical trial design under non-proportional hazards. Crit Rev Oncol Hematol 2021; 162:103350. [PMID: 33989767 DOI: 10.1016/j.critrevonc.2021.103350] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 05/03/2021] [Accepted: 05/08/2021] [Indexed: 12/16/2022] Open
Abstract
In trials of novel immuno-oncology drugs, the proportional hazards (PH) assumption often does not hold for the primary time-to-event (TTE) efficacy endpoint, likely due to the unique mechanism of action of these drugs. In practice, when it is anticipated that PH may not hold for the TTE endpoint with respect to treatment, the sample size is often still calculated under the PH assumption, and the hazard ratio (HR) from the Cox model is still reported as the primary measure of the treatment effect. Sensitivity analyses of the TTE data using methods that are suitable under non-proportional hazards (non-PH) are commonly pre-planned. In cases where a substantial deviation from the PH assumption is likely, we suggest designing the trial, calculating the sample size and analyzing the data, using a suitable method that accounts for non-PH, after gaining alignment with regulatory authorities. In this comprehensive review article, we describe methods to design a randomized oncology trial, calculate the sample size, analyze the trial data and obtain summary measures of the treatment effect in the presence of non-PH. For each method, we provide examples of its use from the recent oncology trials literature. We also summarize in the Appendix some methods to conduct sensitivity analyses for overall survival (OS) when patients in a randomized trial switch or cross-over to the other treatment arm after disease progression on the initial treatment arm, and obtain an adjusted or weighted HR for OS in the presence of cross-over. This is an example of the treatment itself changing at a specific point in time - this cross-over may lead to a non-PH pattern of diminishing treatment effect.
Collapse
Affiliation(s)
| | | | | | - Rong Liu
- Bristol-Myers Squibb (BMS), 300 Connell Drive, Berkeley Heights, NJ, 07922, United States
| | - Daniel Li
- BMS, Seattle, Washington, 98109, United States
| | - Michael LaValley
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, 02118, United States
| |
Collapse
|
13
|
Latimer NR, White IR, Tilling K, Siebert U. Improved two-stage estimation to adjust for treatment switching in randomised trials: g-estimation to address time-dependent confounding. Stat Methods Med Res 2020; 29:2900-2918. [PMID: 32223524 PMCID: PMC7436445 DOI: 10.1177/0962280220912524] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
In oncology trials, control group patients often switch onto the experimental treatment during follow-up, usually after disease progression. In this case, an intention-to-treat analysis will not address the policy question of interest - that of whether the new treatment represents an effective and cost-effective use of health care resources, compared to the standard treatment. Rank preserving structural failure time models (RPSFTM), inverse probability of censoring weights (IPCW) and two-stage estimation (TSE) have often been used to adjust for switching to inform treatment reimbursement policy decisions. TSE has been applied using a simple approach (TSEsimp), assuming no time-dependent confounding between the time of disease progression and the time of switch. This is problematic if there is a delay between progression and switch. In this paper we introduce TSEgest, which uses structural nested models and g-estimation to account for time-dependent confounding, and compare it to TSEsimp, RPSFTM and IPCW. We simulated scenarios where control group patients could switch onto the experimental treatment with and without time-dependent confounding being present. We varied switching proportions, treatment effects and censoring proportions. We assessed adjustment methods according to their estimation of control group restricted mean survival times that would have been observed in the absence of switching. All methods performed well in scenarios with no time-dependent confounding. TSEgest and RPSFTM continued to perform well in scenarios with time-dependent confounding, but TSEsimp resulted in substantial bias. IPCW also performed well in scenarios with time-dependent confounding, except when inverse probability weights were high in relation to the size of the group being subjected to weighting, which occurred when there was a combination of modest sample size and high switching proportions. TSEgest represents a useful addition to the collection of methods that may be used to adjust for treatment switching in trials in order to address policy-relevant questions.
Collapse
Affiliation(s)
- NR Latimer
- School of Health and Related Research, University of Sheffield, Sheffield, UK
| | - IR White
- MRC Clinical Trials Unit, University College London, London, UK
| | - K Tilling
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
| | - U Siebert
- Department of Public Health, Health Services Research and Health Technology Assessment, UMIT -- University for Health Sciences, Medical Informatics and Technology, Hall i.T., Austria
- ONCOTYROL -- Center for Personalized Cancer Medicine, Innsbruck, Austria
- Harvard T.H. Chan School of Public Health and Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
14
|
Adjusting Overall Survival Estimates for Treatment Switching in Metastatic, Castration-Sensitive Prostate Cancer: Results from the LATITUDE Study. Target Oncol 2020; 14:681-688. [PMID: 31754962 PMCID: PMC6875513 DOI: 10.1007/s11523-019-00685-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Background LATITUDE was the first phase 3 trial examining the survival benefit of adding abiraterone acetate (AA) + prednisone (P) to androgen-deprivation therapy (ADT) in newly diagnosed metastatic, castration-sensitive prostate cancer (mCSPC). Due to significant improvement in overall survival after the first interim analysis, patients in the placebos + ADT arm could switch to AA + P + ADT during an open-label extension. As in other studies where switching is allowed, statistical adjustments are needed to assess the real benefit of new drugs. Patients and Methods This was a post hoc analysis to estimate the true survival benefit of AA + P + ADT in patients with newly diagnosed mCSPC by applying statistical adjustments commonly used to adjust for treatment switching. Results Of 112 patients still receiving placebos + ADT at the first interim analysis, 72 switched to AA + P + ADT during the open-label extension. Final analysis was conducted after median follow-up of 51.8 months. Compared to the placebos + ADT arm, the risk of death in the AA + P + ADT arm was 34% lower [hazard ratio (HR) = 0.663 (95% confidence interval 0.566–0.778)] by unadjusted intent-to-treat analysis, 37% lower [HR = 0.629 (95% confidence interval 0.526–0.753)] by rank preserving structure failure time modeling, and 38% lower [HR = 0.616 (95% confidence interval 0.524–0.724)] by inverse probability of censoring weights. Conclusions Analyses adjusting for treatment switching using two different statistical approaches confirm the improved survival benefit of adding AA + P to ADT in patients with newly diagnosed mCSPC. Trial Registration ClinicalTrials.gov identifier NCT01715285. Electronic supplementary material The online version of this article (10.1007/s11523-019-00685-x) contains supplementary material, which is available to authorized users.
Collapse
|
15
|
Sullivan TR, Latimer NR, Gray J, Sorich MJ, Salter AB, Karnon J. Adjusting for Treatment Switching in Oncology Trials: A Systematic Review and Recommendations for Reporting. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2020; 23:388-396. [PMID: 32197735 DOI: 10.1016/j.jval.2019.10.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 10/09/2019] [Accepted: 10/26/2019] [Indexed: 06/10/2023]
Abstract
OBJECTIVES To systematically review the quality of reporting on the application of switching adjustment approaches in published oncology trials and industry submissions to the National Institute for Health and Care Excellence Although methods such as the rank preserving structural failure time model (RPSFTM) and inverse probability of censoring weights (IPCW) have been developed to address treatment switching, the approaches are not widely accepted within health technology assessment. This limited acceptance may partly be a consequence of poor reporting on their application. METHODS Published trials and industry submissions were obtained from searches of PubMed and nice.org.uk, respectively. The quality of reporting in these studies was judged against a checklist of reporting recommendations, which was developed by the authors based on detailed considerations of the methods. RESULTS Thirteen published trials and 8 submissions to nice.org.uk satisfied inclusion criteria. The quality of reporting around the implementation of the RPSFTM and IPCW methods was generally poor. Few studies stated whether the adjustment approach was prespecified, more than a third failed to provide any justification for the chosen method, and nearly half neglected to perform sensitivity analyses. Further, it was often unclear how the RPSFTM and IPCW methods were implemented. CONCLUSIONS Inadequate reporting on the application of switching adjustment methods increases uncertainty around results, which may contribute to the limited acceptance of these methods by decision makers. The proposed reporting recommendations aim to support the improved interpretation of analyses undertaken to adjust for treatment switching.
Collapse
Affiliation(s)
- Thomas R Sullivan
- SAHMRI Women & Kids, South Australian Health & Medical Research Institute, Adelaide, Australia; School of Public Health, The University of Adelaide, Adelaide, Australia.
| | - Nicholas R Latimer
- School of Health and Related Research, The University of Sheffield, Sheffield, England, UK
| | - Jodi Gray
- Australian Institute of Health Innovation, Macquarie University, Sydney, Australia; College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Michael J Sorich
- College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Amy B Salter
- School of Public Health, The University of Adelaide, Adelaide, Australia
| | - Jonathan Karnon
- College of Medicine and Public Health, Flinders University, Adelaide, Australia
| |
Collapse
|
16
|
Assessing the Long-Term Effectiveness of Cladribine vs. Placebo in the Relapsing-Remitting Multiple Sclerosis CLARITY Randomized Controlled Trial and CLARITY Extension Using Treatment Switching Adjustment Methods. Adv Ther 2020; 37:225-239. [PMID: 31701485 DOI: 10.1007/s12325-019-01140-z] [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] [Received: 06/12/2019] [Indexed: 10/25/2022]
Abstract
OBJECTIVES Treatment switching adjustment methods are often used to adjust for switching in oncology randomized controlled trials (RCTs). In this exploratory analysis, we apply these methods to adjust for treatment changes in the setting of an RCT followed by an extension study in relapsing-remitting multiple sclerosis. METHODS The CLARITY trial evaluated cladribine tablets versus placebo over 96 weeks. In the 96-week CLARITY Extension, patients who received placebo in CLARITY received cladribine tablets; patients who received cladribine tablets in CLARITY were re-randomized to placebo or cladribine tablets. End points were time to first qualifying relapse (FQR) and time to 3- and 6-month confirmed disability progression (3mCDP, 6mCDP). We aimed to compare the effectiveness of cladribine tablets with placebo over CLARITY and the extension. The rank-preserving structural failure time model (RPSFTM) and iterative parameter estimation (IPE) were used to estimate what would have happened if patients had received placebo in CLARITY and the extension versus patients that received cladribine tablets and switched to placebo. To gauge whether treatment effect waned after the 96 weeks of CLARITY, we compared hazard ratios (HRs) from the adjustment analysis with HRs from CLARITY. RESULTS The RPSFTM resulted in an HR of 0.48 [95% confidence interval (CI) 0.36-0.62] for FQR, 0.62 (95% CI 0.46-0.84) for 3mCDP and 0.62 (95% CI 0.44-0.88) for 6mCDP. IPE algorithm results were similar. CLARITY HRs were 0.44 (95% CI 0.34-0.58), 0.60 (95% CI 0.41-0.87) and 0.58 (95% CI 0.40-0.83) for FQR, 3mCDP and 6mCDP, respectively. CONCLUSIONS Treatment switching adjustment methods are applicable in non-oncology settings. Adjusted CLARITY plus CLARITY Extension HRs were similar to the CLARITY HRs, demonstrating significant treatment benefits associated with cladribine tablets versus placebo. FUNDING EMD Serono, Inc. (a business of Merck KGaA, Darmstadt, Germany).
Collapse
|
17
|
Nomura S, Shinozaki T, Hamada C. Performance of randomization-based causal methods with and without integrating external data sources for adjusting overall survival in case of extensive treatment switches in placebo-controlled randomized oncology phase 3 trials. J Biopharm Stat 2019; 30:377-401. [PMID: 31820674 DOI: 10.1080/10543406.2019.1695625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
In recent placebo-controlled randomized phase 3 oncology trials, evaluation of overall survival with frequent crossover is crucial for regulatory and pricing decisions. The problem is that an intention-to-treat based analysis causes a substantial loss of power to detect causal survival effect without crossover, and performance of existing methods is not satisfactory. In this article, our aims were to evaluate properties of the existing and a proposed Bayesian power prior method where data from an external trial is available. Simulation results suggested that proposed method was the most powerful under typical scenarios where patients with better prognosis are likely to crossover.
Collapse
Affiliation(s)
- Shogo Nomura
- Center for Research and Administration and Support, National Cancer Center, Chiba, Japan
| | - Tomohiro Shinozaki
- Department of Information and Computer Technology, Graduate School of Engineering, Tokyo University of Science, Tokyo, Japan
| | - Chikuma Hamada
- Department of Information and Computer Technology, Graduate School of Engineering, Tokyo University of Science, Tokyo, Japan
| |
Collapse
|
18
|
Gorrod HB, Latimer NR, Damian D, Hettle R, Harty GT, Wong SL. Impact of Nonrandomized Dropout on Treatment Switching Adjustment in the Relapsing-Remitting Multiple Sclerosis CLARITY Trial and the CLARITY Extension Study. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2019; 22:772-776. [PMID: 31277823 DOI: 10.1016/j.jval.2018.11.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 11/16/2018] [Accepted: 11/26/2018] [Indexed: 06/09/2023]
Abstract
OBJECTIVES Statistical methods to adjust for treatment switching are commonly applied to randomized controlled trials (RCTs) in oncology. Nevertheless, RCTs with extension studies incorporating nonrandomized dropout require consideration of alternative adjustment methods. The current study used a recognized method and a novel method to adjust for treatment switching in relapsing-remitting multiple sclerosis (MS). METHODS The Cladribine Tablets Treating Multiple Sclerosis Orally (CLARITY) RCT evaluated the efficacy of cladribine versus placebo over 96 weeks. Many (but not all) CLARITY participants enrolled in the 96-week CLARITY extension study; placebo-treated patients from CLARITY received cladribine (PP→LL), and cladribine-treated patients were re-randomized to placebo (LL→PP) or continued cladribine (LL→LL). End points were time to first qualifying relapse (FQR) and time to 3-month and 6-month confirmed disability progression (3mCDP, 6mCDP). We aimed to estimate the effectiveness of the LL→PP treatment strategy compared with a counterfactual (unobserved) PP→PP strategy. We applied the commonly used rank-preserving structural failure time model (RPSFTM) and a novel approach that combined propensity score matching (PSM) with inverse probability of censoring weights (IPCW). RESULTS The RPSFTM resulted in LL→PP versus PP→PP hazard ratios (HRs) of 0.48 (95% confidence interval [CI] 0.36-0.62) for FQR, 0.62 (95% CI 0.46-0.84) for 3mCDP, and 0.62 (95% CI 0.44-0.88) for 6mCDP. The PSM+IPCW resulted in HRs of 0.47 (95% CI 0.38-0.63) for FQR, 0.61 (95% CI 0.43-0.86) for 3mCDP, and 0.63 (95% CI 0.40-0.87) for 6mCDP. CONCLUSIONS The PSM+IPCW HRs were consistent with those from the RPSFTM, suggesting that the results were not substantially biased by informative dropout, assuming that all relevant confounders were controlled for. There was no statistical evidence of a reduction in the cladribine treatment effect during the extension period.
Collapse
Affiliation(s)
- Helen Bell Gorrod
- School for Health and Related Research (ScHARR), University of Sheffield, Sheffield, England, United Kingdom.
| | - Nicholas R Latimer
- School for Health and Related Research (ScHARR), University of Sheffield, Sheffield, England, United Kingdom
| | | | - Robert Hettle
- PAREXEL International, London, England, United Kingdom
| | | | | |
Collapse
|
19
|
Latimer NR, Abrams KR, Siebert U. Two-stage estimation to adjust for treatment switching in randomised trials: a simulation study investigating the use of inverse probability weighting instead of re-censoring. BMC Med Res Methodol 2019; 19:69. [PMID: 30935369 PMCID: PMC6444622 DOI: 10.1186/s12874-019-0709-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 03/14/2019] [Indexed: 01/09/2023] Open
Abstract
Background Treatment switching is common in randomised trials of oncology treatments, with control group patients switching onto the experimental treatment during follow-up. This distorts an intention-to-treat comparison of the treatments under investigation. Two-stage estimation (TSE) can be used to estimate counterfactual survival times for patients who switch treatments – that is, survival times that would have been observed in the absence of switching. However, when switchers do not die during the study, counterfactual censoring times are estimated, inducing informative censoring. Re-censoring is usually applied alongside TSE to resolve this problem, but results in lost longer-term information – a major concern if the objective is to estimate long-term treatment effects, as is usually the case in health technology assessment. Inverse probability of censoring weights (IPCW) represents an alternative technique for addressing informative censoring but has not before been combined with TSE. We aim to determine whether combining TSE with IPCW (TSEipcw) represents a valid alternative to re-censoring. Methods We conducted a simulation study to compare TSEipcw to TSE with and without re-censoring. We simulated 48 scenarios where control group patients could switch onto the experimental treatment, with switching affected by prognosis. We investigated various switching proportions, treatment effects, survival function shapes, disease severities and switcher prognoses. We assessed the alternative TSE applications according to their estimation of control group restricted mean survival (RMST) that would have been observed in the absence of switching up to the end of trial follow-up. Results TSEipcw performed well when its weights had a low coefficient of variation, but performed poorly when the coefficient of variation was high. Re-censored analyses usually under-estimated control group RMST, whereas non-re-censored analyses usually produced over-estimates, with bias more serious when the treatment effect was high. In scenarios where TSEipcw performed well, it produced low bias that was often between the two extremes associated with the re-censoring and non-recensoring options. Conclusions Treatment switching adjustment analyses using TSE should be conducted with re-censoring, without re-censoring, and with IPCW to explore the sensitivity in results to these application options. This should allow analysts and decision-makers to better interpret the results of adjustment analyses. Electronic supplementary material The online version of this article (10.1186/s12874-019-0709-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- N R Latimer
- School of Health and Related Research, University of Sheffield, Regent Court, 30 Regent Street, Sheffield, S1 4DA, UK.
| | - K R Abrams
- Biostatistics Research Group, Department of Health Sciences, Centre for Medicine, University of Leicester, University Road, Leicester, LE1 7RH, UK
| | - U Siebert
- UMIT - University for Health Sciences, Medical Informatics and Technology, Eduard-Wallnöfer-Zentrum 1, 6060, Hall in Tirol, Austria.,Oncotyrol - Center for Personalized Cancer Medicine, Innsbruck, Austria.,Harvard T.H. Chan School of Public Health and Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
20
|
Srivastava T, Prabhu VS, Li H, Xu R, Zarabi N, Zhong Y, Pellissier JM, Perini RF, de Wit R, Mamtani R. Cost-effectiveness of Pembrolizumab as Second-line Therapy for the Treatment of Locally Advanced or Metastatic Urothelial Carcinoma in Sweden. Eur Urol Oncol 2018; 3:663-670. [PMID: 31412001 DOI: 10.1016/j.euo.2018.09.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 08/30/2018] [Accepted: 09/25/2018] [Indexed: 11/25/2022]
Abstract
BACKGROUND Urothelial carcinoma (UC) is the most common subtype of bladder cancer. The randomized phase 3 KEYNOTE-045 trial showed that pembrolizumab, used as second-line therapy significantly prolonged overall survival with fewer treatment-related adverse events than chemotherapy for advanced UC. Pembrolizumab has been approved by the European Medicines Agency for the treatment of locally advanced or metastatic UC in adults who have received platinum-containing chemotherapy. Many European countries use cost-effectiveness analysis to inform reimbursement decisions. OBJECTIVE To assess the cost-effectiveness of pembrolizumab as second-line therapy for the treatment of advanced UC from a Swedish health care perspective. DESIGN, SETTING, AND PARTICIPANTS We developed a partitioned-survival model to assess the costs and effectiveness of pembrolizumab compared with vinflunine (base case), paclitaxel, or docetaxel monotherapy in patients with advanced UC over a 15-yr time horizon. We obtained Kaplan-Meier estimates for survival endpoints, adverse events, and utility data from KEYNOTE-045. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS We performed parametric extrapolations to estimate overall and progression-free survival beyond the clinical trial period. Swedish costs and utility weights were used to estimate total costs, quality-adjusted life years (QALYs), and incremental cost-effectiveness ratios (ICERs). We performed deterministic and probabilistic sensitivity analyses to assess the robustness of the model results. RESULTS AND LIMITATIONS In the base-case analysis, pembrolizumab resulted in a mean survival gain of 1.66 years (1.38 QALYs) at an incremental cost of €69852 and an ICER of €50529/QALY gained versus vinflunine monotherapy. ICERs for other chemotherapies were €81356/QALY for pembrolizumab versus paclitaxel or docetaxel monotherapy, and €71924/QALY for pembrolizumab versus paclitaxel, docetaxel, or vinflunine monotherapy. Long-term follow-up from KEYNOTE-045 and real-world data are needed to validate the extrapolations. CONCLUSIONS The results indicate that pembrolizumab improves survival, increases QALYs, and is cost-effective as second-line therapy at a willingness-to-pay threshold of €100000/QALY for the treatment of advanced UC. PATIENT SUMMARY To date, pembrolizumab is the only treatment associated with a significant overall survival benefit compared with chemotherapy in a randomized controlled trial as second-line therapy for advanced urothelial carcinoma. Our trial-based cost-effectiveness analysis suggests that pembrolizumab is a cost-effective option over chemotherapy in patients with advanced urothelial carcinoma after platinum-based therapy in Sweden.
Collapse
Affiliation(s)
| | | | - Haojie Li
- Merck & Co, Inc., Kenilworth, NJ, USA
| | | | | | | | | | | | - Ronald de Wit
- Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Ronac Mamtani
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
21
|
Bennett I, Paracha N, Abrams K, Ray J. Accounting for Uncertainty in Decision Analytic Models Using Rank Preserving Structural Failure Time Modeling: Application to Parametric Survival Models. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2018; 21:105-109. [PMID: 29304934 DOI: 10.1016/j.jval.2017.07.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 07/04/2017] [Accepted: 07/16/2017] [Indexed: 06/07/2023]
Abstract
OBJECTIVES Rank Preserving Structural Failure Time models are one of the most commonly used statistical methods to adjust for treatment switching in oncology clinical trials. The method is often applied in a decision analytic model without appropriately accounting for additional uncertainty when determining the allocation of health care resources. The aim of the study is to describe novel approaches to adequately account for uncertainty when using a Rank Preserving Structural Failure Time model in a decision analytic model. METHODS Using two examples, we tested and compared the performance of the novel Test-based method with the resampling bootstrap method and with the conventional approach of no adjustment. In the first example, we simulated life expectancy using a simple decision analytic model based on a hypothetical oncology trial with treatment switching. In the second example, we applied the adjustment method on published data when no individual patient data were available. RESULTS Mean estimates of overall and incremental life expectancy were similar across methods. However, the bootstrapped and test-based estimates consistently produced greater estimates of uncertainty compared with the estimate without any adjustment applied. Similar results were observed when using the test based approach on a published data showing that failing to adjust for uncertainty led to smaller confidence intervals. CONCLUSIONS Both the bootstrapping and test-based approaches provide a solution to appropriately incorporate uncertainty, with the benefit that the latter can implemented by researchers in the absence of individual patient data.
Collapse
Affiliation(s)
| | | | | | - Joshua Ray
- F. Hoffmann-La Roche AG, Basel, Switzerland
| |
Collapse
|
22
|
Skaltsa K, Ivanescu C, Naidoo S, Phung D, Holmstrom S, Latimer NR. Adjusting Overall Survival Estimates after Treatment Switching: a Case Study in Metastatic Castration-Resistant Prostate Cancer. Target Oncol 2017; 12:111-121. [PMID: 27981431 PMCID: PMC5253154 DOI: 10.1007/s11523-016-0472-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND If patients in oncology trials receive subsequent therapy, standard intention-to-treat (ITT) analyses may inaccurately estimate the overall survival (OS) effect of the investigational product. In this context, a post-hoc analysis of the phase 3 PREVAIL study was performed with the aim to compare enzalutamide with placebo in terms of OS, adjusting for potential confounding from switching to antineoplastic therapies that are not part of standard metastatic castration-resistant prostate cancer (mCRPC) treatment pathways in some jurisdictions. METHODS The PREVAIL study, which included 1717 chemotherapy-naïve men with mCRPC randomized to treatment with enzalutamide 160 mg/day or placebo, was stopped after a planned interim survival analysis revealed a benefit in favor of enzalutamide. Data from this cutoff point were confounded by switching from both arms and so were evaluated in terms of OS using two switching adjustment methods: the two-stage accelerated failure time model (two-stage method) and inverse probability of censoring weights (IPCW). RESULTS Following adjustment for switching to nonstandard antineoplastic therapies by 14.8 (129/872 patients) and 21.3% (180/845 patients) of patients initially randomized to enzalutamide and placebo, respectively, the two-stage and IPCW methods both resulted in numerical reductions in the hazard ratio (HR) for OS [HR 0.66, 95% confidence interval (CI) 0.57-0.81 and HR 0.63, 95% CI 0.52-0.75, respectively] for enzalutamide compared to placebo versus the unadjusted ITT analysis (HR 0.71, 95% CI 0.60-0.84). These results suggest a slightly greater effect of enzalutamide on OS than originally reported. CONCLUSION In the PREVAIL study, switching to nonstandard antineoplastic mCRPC therapies resulted in the ITT analysis of primary data underestimating the benefit of enzalutamide on OS.
Collapse
Affiliation(s)
| | | | - Shevani Naidoo
- Astellas Medical Affairs Global Health Economics and Outcomes Research, 2000 Hillswood Dr, Chertsey, KT16 0PS, Surrey, UK.
| | - De Phung
- Astellas Pharma Global Development, Sylviusweg 62, 2333 BE, Leiden, Netherlands
| | - Stefan Holmstrom
- Astellas Medical Affairs Global Health Economics and Outcomes Research, Sylviusweg 62, 2300 AH, Leiden, Netherlands
| | - Nicholas R Latimer
- School of Health and Related Research, University of Sheffield, Regent Court, 30 Regent Street, Sheffield, S1 4DA, UK
| |
Collapse
|