1
|
Rippin G, Sanz H, Hoogendoorn WE, Ballarini NM, Largent JA, Demas E, Postmus D, Framke T, Dávila LMA, Quinten C, Pignatti F. Examining the Effect of Missing Data and Unmeasured Confounding on External Comparator Studies: Case Studies and Simulations. Drug Saf 2024; 47:1245-1263. [PMID: 39102176 PMCID: PMC11554740 DOI: 10.1007/s40264-024-01467-9] [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] [Accepted: 07/04/2024] [Indexed: 08/06/2024]
Abstract
BACKGROUND AND OBJECTIVE Missing data and unmeasured confounding are key challenges for external comparator studies. This work evaluates bias and other performance characteristics depending on missingness and unmeasured confounding by means of two case studies and simulations. METHODS Two case studies were constructed by taking the treatment arms from two randomised controlled trials and an external real-world data source that exhibited substantial missingness. The indications of the randomised controlled trials were multiple myeloma and metastatic hormone-sensitive prostate cancer. Overall survival was taken as the main endpoint. The effects of missing data and unmeasured confounding were assessed for the case studies by reporting estimated external comparator versus randomised controlled trial treatment effects. Based on the two case studies, simulations were performed broadening the settings by varying the underlying hazard ratio, the sample size, the sample size ratio between the experimental arm and the external comparator, the number of missing covariates and the percentage of missingness. Thereby, bias and other performance metrics could be quantified dependent on these factors. RESULTS For the multiple myeloma external comparator study, results were in line with the randomised controlled trial, despite missingness and potential unmeasured confounding, while for the metastatic hormone-sensitive prostate cancer case study missing data led to a low sample size, leading overall to inconclusive results. Furthermore, for the metastatic hormone-sensitive prostate cancer study, missing data in important eligibility criteria led to further limitations. Simulations were successfully applied to gain a quantitative understanding of the effects of missing data and unmeasured confounding. CONCLUSIONS This exploratory study confirmed external comparator strengths and limitations by quantifying the impact of missing data and unmeasured confounding using case studies and simulations. In particular, missing data in key eligibility criteria were seen to limit the ability to derive the external comparator target analysis population accurately, while simulations demonstrated the magnitude of bias to expect for various settings.
Collapse
Affiliation(s)
- Gerd Rippin
- IQVIA, Unterschweinstiege 2-14, 60549, Frankfurt, Germany.
| | - Héctor Sanz
- IQVIA, Unterschweinstiege 2-14, 60549, Frankfurt, Germany
| | | | | | - Joan A Largent
- IQVIA, Unterschweinstiege 2-14, 60549, Frankfurt, Germany
| | - Eleni Demas
- IQVIA, Unterschweinstiege 2-14, 60549, Frankfurt, Germany
| | - Douwe Postmus
- European Medicines Agency, Domenico Scarlattilaan 6, Amsterdam, 1083 HS, The Netherlands
| | - Theodor Framke
- European Medicines Agency, Domenico Scarlattilaan 6, Amsterdam, 1083 HS, The Netherlands
- Institute for Biostatistics, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | | | - Chantal Quinten
- European Medicines Agency, Domenico Scarlattilaan 6, Amsterdam, 1083 HS, The Netherlands
| | - Francesco Pignatti
- European Medicines Agency, Domenico Scarlattilaan 6, Amsterdam, 1083 HS, The Netherlands
| |
Collapse
|
2
|
Choi S, Byun JM, Park SS, Han J, Oh S, Jung S, Park H, Han S, Lee JY, Koh Y, Jeon YW, Yahng SA, Shin SH, Yoon SS, Min CK. Efficacy and Safety of Bispecific T-Cell Engagers in Relapsed/Refractory Multiple Myeloma: A Real-World Data-Based Case-Controlled Study. Transplant Cell Ther 2024:S2666-6367(24)00779-6. [PMID: 39608453 DOI: 10.1016/j.jtct.2024.11.010] [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: 08/29/2024] [Revised: 10/16/2024] [Accepted: 11/19/2024] [Indexed: 11/30/2024]
Abstract
Although bispecific T-cell engager (BiTE) is a promising treatment for relapsed/refractory multiple myeloma (RRMM), it needs to be evaluated in a real-world setting. This study aimed to evaluate the efficacy and safety of BiTEs compared with a synthetic standard of care (SOC). From a multicenter registry database of 474 patients with RRMM who received third- or more advanced-line treatments between January 2021 and October 2023, 1:1 propensity score-matched BiTE cohort (n = 71) and SOC cohort (n = 71) were established. Matching was based on age, sex, number of prior therapies, international staging system at diagnosis, and baseline biochemical characteristics. Compared with the matched SOC cohort, the matched BiTE cohort demonstrated a significant improvement in median progression-free survival (PFS, 19.2 vs 5.4 months, hazard ratio (HR) = .50 [95% CI, .33 to .78], p < .01). However, the overall survival (OS) was not significantly different between the two cohorts. Safety profiles showed that 37 (52%) patients in the matched BiTE cohort experienced cytokine release syndrome, mostly grade 1 (n = 29, 41%), with rare occurrences of neurotoxicity (n = 4, 5.6%). Infections were significantly more common in the matched BiTE cohort compared with the matched SOC cohort (81% vs. 49%, p < .01). Non-B-cell mutation antigen (BCMA)-targeted BiTEs improved 6-month OS rates compared with BCMA-targeted BiTEs in monotherapy (94% [95% CI, 84 to 100] vs. 65% [95% CI, 45 to 95], p = .04) and combination with daratumumab (100% [95% CI, 100 to 100] vs. 77% [95% CI, 57 to 100], p = .20). Non-BCMA-targeted BiTEs also provided benefit for 6-month PFS rate compared with the BCMA-targeted BiTE cohort in monotherapy (76% [95% CI, 59 to 100] vs. 50% [95% CI, 31 to 82], p = .11) and combination with daratumumab (100% [95% CI, 100 to 100] vs. 69% [95% CI, 48 to 99], p = .10). Quantitative bias and sensitivity analyses confirmed the robustness of these results. This real-world data-based study underscores the potential of BiTEs to significantly enhance survival outcomes in patients with heavily treated RRMM and manageable safety profiles. The difference in clinical outcomes by BiTE targets warrants further investigation in larger clinical trials (ClinicalTrials.gov identifier: NCT06205823).
Collapse
Affiliation(s)
- Suein Choi
- Department of Pharmacology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Pharmacometrics Institute for Practical Education and Training (PIPET), College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ja Min Byun
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea; Cancer Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Sung-Soo Park
- Catholic Research Network for Multiple Myeloma, Republic of Korea; Catholic Hematology Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Hematology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| | - Jinsun Han
- Department of Pharmacology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Pharmacometrics Institute for Practical Education and Training (PIPET), College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sieun Oh
- Department of Hematology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seungpil Jung
- Department of Pharmacology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Pharmacometrics Institute for Practical Education and Training (PIPET), College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyejoon Park
- Department of Pharmacology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Pharmacometrics Institute for Practical Education and Training (PIPET), College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seunghoon Han
- Department of Pharmacology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Pharmacometrics Institute for Practical Education and Training (PIPET), College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jung Yeon Lee
- Catholic Research Network for Multiple Myeloma, Republic of Korea; Catholic Hematology Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Hematology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Youngil Koh
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea; Cancer Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Young-Woo Jeon
- Catholic Research Network for Multiple Myeloma, Republic of Korea; Catholic Hematology Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Hematology, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seung-Ah Yahng
- Catholic Research Network for Multiple Myeloma, Republic of Korea; Catholic Hematology Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Hematology, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Incheon, Republic of Korea
| | - Seung-Hwan Shin
- Catholic Research Network for Multiple Myeloma, Republic of Korea; Catholic Hematology Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Hematology, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sung-Soo Yoon
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea; Cancer Research Institute, Seoul National University Hospital, Seoul, Republic of Korea; Catholic Research Network for Multiple Myeloma, Republic of Korea
| | - Chang-Ki Min
- Catholic Research Network for Multiple Myeloma, Republic of Korea; Catholic Hematology Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Hematology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
3
|
Tabatabai G, Platten M, Preusser M, Weller M, Wick W, van den Bent M. Treatment of glioblastoma patients with personalized vaccines outside clinical trials: Lessons ignored? Neuro Oncol 2024:noae225. [PMID: 39561243 DOI: 10.1093/neuonc/noae225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2024] Open
Affiliation(s)
- Ghazaleh Tabatabai
- Department of Neurology and Interdisciplinary Neuro-Oncology, Center for Neuro-Oncology, Comprehensive Cancer Center, University Hospital Tübingen, Hertie Institute for Clinical Brain Research, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Michael Platten
- Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neurosciences (MCTN), University of Heidelberg, Mannheim, Germany
- Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Matthias Preusser
- Division of Oncology, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - Michael Weller
- Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Wolfgang Wick
- Department of Neurology, University Hospital and Heidelberg University, Clinical Cooperation Unit Neurooncology, German Cancer Research Center, Heidelberg, Germany
| | - Martin van den Bent
- The Brain Tumor Center, Erasmus Medical Center Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
4
|
Medford AJ, Carmeli AB, Ritchie A, Wagle N, Garraway L, Lander ES, Parikh A. A standing platform for cancer drug development using ctDNA-based evidence of recurrence. Nat Rev Cancer 2024; 24:810-821. [PMID: 39349822 DOI: 10.1038/s41568-024-00742-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/09/2024] [Indexed: 10/26/2024]
Abstract
The time required to conduct clinical trials limits the rate at which we can evaluate and deliver new treatment options to patients with cancer. New approaches to increase trial efficiency while maintaining rigor would benefit patients, especially in oncology, in which adjuvant trials hold promise for intercepting metastatic disease, but typically require large numbers of patients and many years to complete. We envision a standing platform - an infrastructure to support ongoing identification and trial enrolment of patients with cancer with early molecular evidence of disease (MED) after curative-intent therapy for early-stage cancer, based on the presence of circulating tumour DNA. MED strongly predicts subsequent recurrence, with the vast majority of patients showing radiographic evidence of disease within 18 months. Such a platform would allow efficient testing of many treatments, from small exploratory studies to larger pivotal trials. Trials enrolling patients with MED but without radiographic evidence of disease have the potential to advance drug evaluation because they can be smaller (given high probability of recurrence) and faster (given short time to recurrence) than conventional adjuvant trials. Circulating tumour DNA may also provide a valuable early biomarker of treatment effect, which would allow small signal-finding trials. In this Perspective, we discuss how such a platform could be established.
Collapse
Affiliation(s)
- Arielle J Medford
- Department of Medicine, Division of Hematology and Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | | | | | | | - Eric S Lander
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Science for America, Cambridge, MA, USA.
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA.
| | - Aparna Parikh
- Department of Medicine, Division of Hematology and Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
5
|
Subramaniam D, Anderson-Smits C, Rubinstein R, Thai ST, Purcell R, Girman C. A Framework for the Use and Likelihood of Regulatory Acceptance of Single-Arm Trials. Ther Innov Regul Sci 2024; 58:1214-1232. [PMID: 39285061 PMCID: PMC11530569 DOI: 10.1007/s43441-024-00693-8] [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: 04/18/2024] [Accepted: 08/23/2024] [Indexed: 11/03/2024]
Abstract
BACKGROUND Single-arm clinical trials (SAT) are common in drug and biologic submissions for rare or life-threatening conditions, especially when no therapeutic options exist. External control arms (ECAs) improve interpretation of SATs but pose methodological and regulatory challenges. OBJECTIVE Through narrative reviews and expert input, we developed a framework for considerations that might influence regulatory use and likelihood of regulatory acceptance of an SAT, identifying non-oncology first indication approvals as an area of interest. We systematically analyzed FDA and EMA approvals using SATs as pivotal evidence. The framework guided outcome abstraction on regulatory responses. METHODS We examined all non-oncology FDA and EMA drug and biologic approvals for first indications from 2019 to 2022 to identify those with SAT as pivotal safety or efficacy evidence. We abstracted outcomes, key study design features, regulator responses to SAT and (where applicable) ECA design, and product label content. RESULTS Among 20 SAT-based FDA approvals and 17 SAT-based EMA approvals, most common indications were progressive rare diseases with high unmet need/limited therapeutic options and a natural history without spontaneous improvement. Of the types of comparators, most were natural history cohorts (45% FDA; 47% EMA) and baseline controls (40% FDA; 47% EMA). Common critiques were of non-contemporaneous ECAs, subjective endpoints, and baseline covariate imbalance between arms. CONCLUSION Based on recent FDA and EMA approvals, the likelihood of regulatory success for SATs with ECAs depends on many design, analytic, and data quality considerations. Our framework is useful in early drug development when considering SAT strategies for evidence generation.
Collapse
Affiliation(s)
- Disha Subramaniam
- Real World Evidence and Patient Outcomes, CERobs Consulting, Wrightsville Beach, NC, USA.
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | | | - Rebecca Rubinstein
- Real World Evidence and Patient Outcomes, CERobs Consulting, Wrightsville Beach, NC, USA
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sydney T Thai
- Real World Evidence and Patient Outcomes, CERobs Consulting, Wrightsville Beach, NC, USA
| | | | - Cynthia Girman
- Real World Evidence and Patient Outcomes, CERobs Consulting, Wrightsville Beach, NC, USA
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
6
|
Dyachkova Y, Dunger-Baldauf C, Barbier N, Devenport J, Franzén S, Kazeem G, Künzel T, Mancini P, Mordenti G, Richert K, Ridolfi A, Saure D. Do You Want to Stay Single? Considerations on Single-Arm Trials in Drug Development and the Postregulatory Space. Pharm Stat 2024; 23:1206-1217. [PMID: 38923796 DOI: 10.1002/pst.2412] [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: 12/06/2023] [Revised: 04/03/2024] [Accepted: 05/23/2024] [Indexed: 06/28/2024]
Abstract
Single-arm trials (SATs), while not preferred, remain in use throughout the drug development cycle. They may be accepted by regulators in particular contexts (e.g., in oncology or rare diseases) when the potential effects of new treatments are very large and placebo treatment is unethical. However, in the postregulatory space, SATs are common, and perhaps even more poorly suited to address the questions of interest. In this manuscript, we review regulatory and HTA positions on SATs; challenges posed by SATs to address research questions beyond regulators, evolving statistical methods to provide context for SATs, case studies where SATs could and could not address questions of interest, and communication strategies to influence decision making and optimize study design to address evidence needs.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Daniel Saure
- Boehringer Ingelheim Europe GmbH, Ingelheim, Germany
| |
Collapse
|
7
|
Lau CY, Rawson NSB. Is Canada Moving towards a More Agile Regulatory Approval and Reimbursement Process with a Shifting Role for Real-World Evidence (RWE) for Oncology Drugs? Curr Oncol 2024; 31:5599-5607. [PMID: 39330042 PMCID: PMC11430900 DOI: 10.3390/curroncol31090414] [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: 08/23/2024] [Revised: 09/15/2024] [Accepted: 09/16/2024] [Indexed: 09/28/2024] Open
Abstract
Canada is known to have a complex pathway for new drug approval and reimbursement, resulting in delayed access for patients with serious and life-threatening diseases, such as cancer. Several recent publications from key stakeholders, including patients, physicians and policymakers, highlight patient helplessness, physician frustrations and policymakers entangled in a massive network of bureaucracy unable to make headway. Several quantitative and qualitative assessments using time from regulatory approvals to successful reimbursements confirm long review times and high rejection rates for oncology drugs, especially those receiving conditional approvals. A consensus forum of 18 Canadian oncology clinicians recently voiced frustration with the process and inability to deliver guideline-supported efficacious therapies to their patients. This manuscript compares data extracted from publicly available data sources from 2019 to June 2024 to previous publications. Methods: Public databases from Health Canada, the Canadian Agency for Drugs and Technologies in Health (CADTH), which is in the process of changing to Canada's Drug Agency, and the pan-Canadian Pharmaceutical Alliance (pCPA) were reviewed and the data collected were analyzed with descriptive statistics. Results: From the data, three trends emerge, (i) an increasing number of oncology drugs are receiving conditional approvals from Health Canada, (ii) the percentage of conditionally approved oncology drugs receiving positive reimbursement recommendations from CADTH is still low but appears to be improving, but delays in access are now contingent upon pCPA deciding whether to negotiate price and then the duration of any negotiation, and (iii) real-world evidence is no longer part of the decision-making for conditional approvals. A slight increase in the positive endorsement of RWE used to support CADTH recommendations was observed. Conclusions: The lack of timely access to oncology drugs hurts Canadian patients. While a small trend of improvement appears to be emerging, longer-term data collection is required to ensure sustained patient benefits.
Collapse
Affiliation(s)
- Catherine Y. Lau
- Independent Researcher, 158 Front Street E, Toronto, ON M5A 0K9, Canada
| | - Nigel S. B. Rawson
- Macdonald-Laurier Institute, Ottawa, ON K1N 7Z2, Canada;
- Canadian Health Policy Institute, Toronto, ON M5V 2Y5, Canada
- Centre for Health Policy Studies, Fraser Institute, Vancouver, BC V6J 3G7, Canada
| |
Collapse
|
8
|
Hogervorst MA, Soman KV, Gardarsdottir H, Goettsch WG, Bloem LT. Analytical Methods for Comparing Uncontrolled Trials With External Controls From Real-World Data: A Systematic Literature Review and Comparison With European Regulatory and Health Technology Assessment Practice. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2024:S1098-3015(24)02842-0. [PMID: 39241824 DOI: 10.1016/j.jval.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 07/04/2024] [Accepted: 08/16/2024] [Indexed: 09/09/2024]
Abstract
OBJECTIVES This study aimed to provide an overview of analytical methods in scientific literature for comparing uncontrolled medicine trials with external controls from individual patient data real-world data (IPD-RWD) and to compare these methods with recommendations made in guidelines from European regulatory and health technology assessment (HTA) organizations and with their evaluations described in assessment reports. METHODS A systematic literature review (until March 1, 2023) in PubMed and Connected Papers was performed to identify analytical methods for comparing uncontrolled trials with external controls from IPD-RWD. These methods were compared descriptively with methods recommended in method guidelines and encountered in assessment reports of the European Medicines Agency (2015-2020) and 4 European HTA organizations (2015-2023). RESULTS Thirty-four identified scientific articles described analytical methods for comparing uncontrolled trial data with IPD-RWD-based external controls. The various methods covered controlling for confounding and/or dependent censoring, correction for missing data, and analytical comparative modeling methods. Seven guidelines also focused on research design, RWD quality, and transparency aspects, and 4 of those recommended analytical methods for comparisons with IPD-RWD. The methods discussed in regulatory (n = 15) and HTA (n = 35) assessment reports were often based on aggregate data and lacked transparency owing to the few details provided. CONCLUSIONS Literature and guidelines suggest a methodological approach to comparing uncontrolled trials with external controls from IPD-RWD similar to target trial emulation, using state-of-the-art methods. External controls supporting regulatory and HTA decision making were rarely in line with this approach. Twelve recommendations are proposed to improve the quality and acceptability of these methods.
Collapse
Affiliation(s)
- Milou A Hogervorst
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, The Netherlands
| | - Kanaka V Soman
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, The Netherlands
| | - Helga Gardarsdottir
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, The Netherlands; Division Laboratory and Pharmacy, Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht, The Netherlands; Faculty of Pharmaceutical Sciences, University of Iceland, Reykjavik, Iceland
| | - Wim G Goettsch
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, The Netherlands; National Health Care Institute (ZIN), Diemen, The Netherlands
| | - Lourens T Bloem
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
9
|
Alipour‐Haris G, Liu X, Acha V, Winterstein AG, Burcu M. Real-world evidence to support regulatory submissions: A landscape review and assessment of use cases. Clin Transl Sci 2024; 17:e13903. [PMID: 39092896 PMCID: PMC11295294 DOI: 10.1111/cts.13903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/24/2024] [Accepted: 07/10/2024] [Indexed: 08/04/2024] Open
Abstract
Real-world evidence (RWE) has an increasing role in preapproval settings to support the approval of new medicines and indications. The main objectives of this study were to identify and characterize regulatory use cases that utilized RWE and other related observational approaches through targeted review of publications and regulatory review documents. After screening and inclusion/exclusion, the review characterized 85 regulatory applications with RWE. A total of 31 were in oncology and 54 were in non-oncology therapeutic areas. Most were for indications in adults only (N = 42, 49.4%), while 13 were in pediatrics only (15.3%), and 30 were in both (35.3%). In terms of regulatory context, 59 cases (69.4%) were for an original marketing application, 24 (28.2%) were for label expansion, and 2 (2.4%) were for label modification. Most also received special regulatory designations (e.g., orphan indication, breakthrough therapy, fast track, conditional, and accelerated approvals). There were 42 cases that utilized RWE to support single-arm trials. External data to support single-arm trials were utilized in various ways across use cases, including direct matching, benchmarking, natural history studies as well as literature or previous trials. A variety of data sources were utilized, including electronic health records, claims, registries, site-based charts. Endpoints in oncology use cases commonly included overall survival, progression-free survival. In 13 use cases, RWE was not considered supportive/definitive in regulatory decision-making due to design issues (e.g., small sample size, selection bias, missing data). Overall, RWE is utilized in regulatory approval processes for new indications/label expansion across various therapeutic areas with wide range of approaches. Multifaceted cross-sector efforts are needed to further improve the quality and utility of RWE in regulatory decision-making.
Collapse
|
10
|
Farah E, Kenney M, Warkentin MT, Cheung WY, Brenner DR. Examining external control arms in oncology: A scoping review of applications to date. Cancer Med 2024; 13:e7447. [PMID: 38984669 PMCID: PMC11234289 DOI: 10.1002/cam4.7447] [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: 01/04/2024] [Revised: 06/11/2024] [Accepted: 06/24/2024] [Indexed: 07/11/2024] Open
Abstract
OBJECTIVES Randomized controlled trials (RCTs) are the gold standard for evaluating the comparative efficacy and safety of new cancer therapies. However, enrolling patients in control arms of clinical trials can be challenging for rare cancers, particularly in the context of precision oncology and targeted therapies. External Control Arms (ECAs) are a potential solution to address these challenges in clinical research design. We conducted a scoping review to explore the use of ECAs in oncology. METHODS We systematically searched four databases, namely MEDLINE, EMBASE, Web of Science, and Scopus. We screened titles, abstracts, and full texts for eligible articles focusing on patients undergoing therapy for cancer, employing ECAs, and reporting clinical outcomes. RESULTS Of the 629 articles screened, 23 were included in this review. The earliest included studies were published in 1996, while most studies were published in the past 5 years. 44% (10/23) of ECAs were employed in blood-related cancer studies. Geographically, 30% (7/23) of studies were conducted in the United States, 22% (5/23) in Japan, and 9% (2/23) in South Korea. The primary data sources used to construct the ECAs involved pooled data from previous trials (35%, 8/23), administrative health databases (17%, 4/23) and electronic medical records (17%, 4/23). While 52% (12/23) of the studies employed methods to align treatment and ECAs characteristics, 48% (11/23) lacked explicit strategies. CONCLUSION ECAs offer a valuable approach in oncology research, particularly when alternative designs are not feasible. However, careful methodological planning and detailed reporting are essential for meaningful and reliable results.
Collapse
Affiliation(s)
- Eliya Farah
- Department of Oncology, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
- Department of Community Health Sciences, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
| | - Matthew Kenney
- Department of Oncology, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
- Department of Community Health Sciences, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
| | - Matthew T. Warkentin
- Department of Oncology, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
- Department of Community Health Sciences, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
| | - Winson Y. Cheung
- Department of Oncology, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
- Department of Community Health Sciences, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
| | - Darren R. Brenner
- Department of Oncology, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
- Department of Community Health Sciences, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
| |
Collapse
|
11
|
Villacampa G, Dennett S, Mello E, Holton J, Lai X, Kilburn L, Bliss J, Rekowski J, Yap C. Accrual and statistical power failure in published adjuvant phase III oncology trials: a comprehensive analysis from 2013 to 2023. ESMO Open 2024; 9:103603. [PMID: 38925083 PMCID: PMC11255358 DOI: 10.1016/j.esmoop.2024.103603] [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: 02/25/2024] [Revised: 05/19/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND In a competitive landscape with many ongoing adjuvant randomised controlled trials (RCTs), the prevalence of trials that failed to recruit their targeted sample size and were inadequately powered is unclear. The aims of the study are (i) to determine the percentage of trials with accrual and statistical power failure and (ii) to evaluate their potential impact on the drug development process. MATERIALS AND METHODS A systematic review was carried out to identify adjuvant phase III oncology RCTs reported between 2013 and 2023 across all solid tumours. No restrictions were applied regarding the type of intervention or journal of publication. The percentage of trials with accrual failure and power failure was estimated as well as their association with the efficacy endpoints. Logistic regression models were used to estimate the odds ratio (OR) and its 95% confidence interval (CI). RESULTS A total of 282 RCTs met the inclusion criteria with a median sample size of 661 patients and a median accrual period of 4.3 years. Most of these studies were superiority trials (83.0%). Accrual failure was observed in 22.0% of the studies, finishing recruitment without achieving the targeted sample size. Overall, 39.7% of the studies experienced power failure, having less power than specified in the protocol at the date of the read-out. Among superiority RCTs evaluating intermediate survival endpoints, only 31.1% presented statistically significant results. Trials with power failure were less likely to present statistically significant results (37.9% versus 21.9%, P = 0.04). The association was consistent across all cancer types. In the subset of non-inferiority trials, 35.0% formally demonstrated non-inferiority of the experimental arm. CONCLUSIONS Nearly 40% of adjuvant phase III RCTs experienced power failure, and the reduction in power significantly impacted the final study results. There is a need for procedural refinements in the design and implementation of future adjuvant RCTs to mitigate these fallacies.
Collapse
Affiliation(s)
- G Villacampa
- Clinical Trials and Statistics Unit at The Institute of Cancer Research (ICR-CTSU), London, UK; Statistics Unit, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain. https://twitter.com/G_Villacampa
| | - S Dennett
- Clinical Trials and Statistics Unit at The Institute of Cancer Research (ICR-CTSU), London, UK
| | - E Mello
- Clinical Trials and Statistics Unit at The Institute of Cancer Research (ICR-CTSU), London, UK
| | - J Holton
- Clinical Trials and Statistics Unit at The Institute of Cancer Research (ICR-CTSU), London, UK
| | - X Lai
- Clinical Trials and Statistics Unit at The Institute of Cancer Research (ICR-CTSU), London, UK
| | - L Kilburn
- Clinical Trials and Statistics Unit at The Institute of Cancer Research (ICR-CTSU), London, UK
| | - J Bliss
- Clinical Trials and Statistics Unit at The Institute of Cancer Research (ICR-CTSU), London, UK
| | - J Rekowski
- Clinical Trials and Statistics Unit at The Institute of Cancer Research (ICR-CTSU), London, UK
| | - C Yap
- Clinical Trials and Statistics Unit at The Institute of Cancer Research (ICR-CTSU), London, UK.
| |
Collapse
|
12
|
Stacchiotti S, Bouche G, Herold R, Pantziarka P, Schuster K, Wilson R, Pignatti F, Kasper B. How to develop new systemic treatments in ultra-rare cancers with high unmet needs? The case of alveolar soft-part sarcoma. Eur J Cancer 2024; 202:114003. [PMID: 38479120 DOI: 10.1016/j.ejca.2024.114003] [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: 12/07/2023] [Revised: 02/27/2024] [Accepted: 02/29/2024] [Indexed: 04/21/2024]
Abstract
Developing new drugs or generating evidence for existing drugs in new indications for ultra-rare cancers is complex and carries a high-risk of failure. This gets even harder in ultra-rare tumours, which have an annual incidence of 1 per 1,000,000 population or less. Here, we illustrate the problem of adequate evidence generation in ultra-rare tumours, using Alveolar Soft-Part Sarcomas (ASPS) - an ultra-rare sarcoma newly diagnosed in approximately 60 persons a year in the European Union - as an exemplar case showing challenges in development despite being potentially relevant for classes of agents. We discuss some possible approaches for addressing such challenges, especially focussing on constructive collaboration between academic groups, patients and advocates, drug manufacturers, and regulators to optimise drug development in ultra-rare cancers. This article, written by various European stakeholders, proposes a way forward to ultimately get better options for patients with ultra-rare cancers.
Collapse
Affiliation(s)
- Silvia Stacchiotti
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy.
| | | | | | | | | | - Roger Wilson
- Sarcoma Patient Advocacy Global Network, Wölfersheim, Germany
| | | | - Bernd Kasper
- Sarcoma Unit, Mannheim Cancer Center (MCC), Mannheim University Medical Center, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
13
|
Zou KH, Vigna C, Talwai A, Jain R, Galaznik A, Berger ML, Li JZ. The Next Horizon of Drug Development: External Control Arms and Innovative Tools to Enrich Clinical Trial Data. Ther Innov Regul Sci 2024; 58:443-455. [PMID: 38528279 PMCID: PMC11043157 DOI: 10.1007/s43441-024-00627-4] [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: 10/17/2023] [Accepted: 02/04/2024] [Indexed: 03/27/2024]
Abstract
Conducting clinical trials (CTs) has become increasingly costly and complex in terms of designing and operationalizing. These challenges exist in running CTs on novel therapies, particularly in oncology and rare diseases, where CTs increasingly target narrower patient groups. In this study, we describe external control arms (ECA) and other relevant tools, such as virtualization and decentralized clinical trials (DCTs), and the ability to follow the clinical trial subjects in the real world using tokenization. ECAs are typically constructed by identifying appropriate external sources of data, then by cleaning and standardizing it to create an analysis-ready data file, and finally, by matching subjects in the external data with the subjects in the CT of interest. In addition, ECA tools also include subject-level meta-analysis and simulated subjects' data for analyses. By implementing the recent advances in digital health technologies and devices, virtualization, and DCTs, realigning of CTs from site-centric designs to virtual, decentralized, and patient-centric designs can be done, which reduces the patient burden to participate in the CTs and encourages diversity. Tokenization technology allows linking the CT data with real-world data (RWD), creating more comprehensive and longitudinal outcome measures. These tools provide robust ways to enrich the CT data for informed decision-making, reduce the burden on subjects and costs of trial operations, and augment the insights gained for the CT data.
Collapse
Affiliation(s)
| | - Chelsea Vigna
- Medidata Solutions, a Dassault Systèmes Company, Boston, MA, USA
| | - Aniketh Talwai
- Medidata Solutions, a Dassault Systèmes Company, Boston, MA, USA
| | - Rahul Jain
- Medidata Solutions, a Dassault Systèmes Company, Boston, MA, USA
| | - Aaron Galaznik
- Medidata Solutions, a Dassault Systèmes Company, Boston, MA, USA
| | - Marc L Berger
- Medidata Solutions, a Dassault Systèmes Company, Boston, MA, USA
| | | |
Collapse
|
14
|
Cornillet M, Villard C, Rorsman F, Molinaro A, Nilsson E, Kechagias S, von Seth E, Bergquist A. The Swedish initiative for the st udy of Primary sclerosing cholangitis (SUPRIM). EClinicalMedicine 2024; 70:102526. [PMID: 38500838 PMCID: PMC10945116 DOI: 10.1016/j.eclinm.2024.102526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 02/19/2024] [Accepted: 02/20/2024] [Indexed: 03/20/2024] Open
Abstract
Background Despite more than 50 years of research and parallel improvements in hepatology and oncology, there is still today neither a treatment to prevent disease progression in primary sclerosing cholangitis (PSC), nor reliable early diagnostic tools for the associated hepatobiliary cancers. Importantly, the limited understanding of the underlying biological mechanisms in PSC and its natural history not only affects the identification of new drug targets but implies a lack of surrogate markers that hampers the design of clinical trials and the evaluation of drug efficacy. The lack of easy access to large representative well-characterised prospective resources is an important contributing factor to the current situation. Methods We here present the SUPRIM cohort, a national multicentre prospective longitudinal study of unselected PSC patients capturing the representative diversity of PSC phenotypes. We describe the 10-year effort of inclusion and follow-up, an intermediate analysis report including original results, and the associated research resource. All included patients gave written informed consent (recruitment: November 2011-April 2016). Findings Out of 512 included patients, 452 patients completed the five-year follow-up without endpoint outcomes. Liver transplantation was performed in 54 patients (10%) and hepatobiliary malignancy was diagnosed in 15 patients (3%). We draw a comprehensive landscape of the multidimensional clinical and biological heterogeneity of PSC illustrating the diversity of PSC phenotypes. Performances of available predictive scores are compared and perspectives on the continuation of the SUPRIM cohort are provided. Interpretation We envision the SUPRIM cohort as an open-access collaborative resource to accelerate the generation of new knowledge and independent validations of promising ones with the aim to uncover reliable diagnostics, prognostic tools, surrogate markers, and new treatment targets by 2040. Funding This work was supported by the Swedish Cancer Society, Stockholm County Council, and the Cancer Research Funds of Radiumhemmet.
Collapse
Affiliation(s)
- Martin Cornillet
- Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Christina Villard
- Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
- Department of Transplantation Surgery, Karolinska University Hospital, Stockholm, Sweden
| | - Fredrik Rorsman
- Department of Gastroenterology and Hepatology, Akademiska University Hospital, Uppsala, Sweden
| | - Antonio Molinaro
- Department of Hepatology, Sahlgrenska University Hospital, Göteborg, Sweden
| | - Emma Nilsson
- Gastroenterology Clinic, Skåne University Hospital, Sweden
| | - Stergios Kechagias
- Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Erik von Seth
- Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
- Department of Upper Abdominal Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Annika Bergquist
- Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
- Department of Upper Abdominal Diseases, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
15
|
Chen J, Li XN, Lu CC, Yuan S, Yung G, Ye J, Tian H, Lin J. Considerations for master protocols using external controls. J Biopharm Stat 2024:1-23. [PMID: 38363805 DOI: 10.1080/10543406.2024.2311248] [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: 05/03/2023] [Accepted: 01/24/2024] [Indexed: 02/18/2024]
Abstract
There has been an increasing use of master protocols in oncology clinical trials because of its efficiency to accelerate cancer drug development and flexibility to accommodate multiple substudies. Depending on the study objective and design, a master protocol trial can be a basket trial, an umbrella trial, a platform trial, or any other form of trials in which multiple investigational products and/or subpopulations are studied under a single protocol. Master protocols can use external data and evidence (e.g. external controls) for treatment effect estimation, which can further improve efficiency of master protocol trials. This paper provides an overview of different types of external controls and their unique features when used in master protocols. Some key considerations in master protocols with external controls are discussed including construction of estimands, assessment of fit-for-use real-world data, and considerations for different types of master protocols. Similarities and differences between regular randomized controlled trials and master protocols when using external controls are discussed. A targeted learning-based causal roadmap is presented which constitutes three key steps: (1) define a target statistical estimand that aligns with the causal estimand for the study objective, (2) use an efficient estimator to estimate the target statistical estimand and its uncertainty, and (3) evaluate the impact of causal assumptions on the study conclusion by performing sensitivity analyses. Two illustrative examples for master protocols using external controls are discussed for their merits and possible improvement in causal effect estimation.
Collapse
Affiliation(s)
- Jie Chen
- Data Sciences, ECR Global, Shanghai, China
| | | | | | - Sammy Yuan
- Oncology Statistics, GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | - Godwin Yung
- Product Development Data and Statistical Sciences, Genentech/Roche, South San Francisco, Cambridge, USA
| | - Jingjing Ye
- Global Statistics and Data Sciences, BeiGene, Fulton, Maryland, USA
| | - Hong Tian
- Global Statistics, BeiGene, Ridgefield Park, New Jersy, USA
| | - Jianchang Lin
- Statistical & Quantitative Sciences, Takeda, Cambridge, Massachusetts, USA
| |
Collapse
|
16
|
Zhou Y, Liang M, Li Y, Chen X, Yang J, Bai H, Long Y, Zhang X, Lin C. Comparative efficacy and safety of Sofosbuvir/Velpatasvir and Danoprevir for the treatment of chronic hepatitis C: the real-world data in China. BMC Gastroenterol 2024; 24:73. [PMID: 38355447 PMCID: PMC10868050 DOI: 10.1186/s12876-024-03147-5] [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] [Received: 05/05/2023] [Accepted: 01/24/2024] [Indexed: 02/16/2024] Open
Abstract
BACKGROUND Sofosbuvir/Velpatasvir (Epclusa, ECS) is the first pan-genotype direct-acting antiviral agent (DAA) for hepatitis C virus (HCV) infection, and Danoprevir (DNV) is the first DAA developed by a Chinese local enterprise, which is suitable for combined use with other drugs to treat genotype 1b chronic hepatitis C. However, previous reports have never compared the real-world data of ECS and DNV. PATIENTS AND METHODS 178 chronic hepatitis C patients were retrospectively recruited, and 94cases were accepted with Sofosbuvir/Velpatasvir ± Ribavirin (ECS group), and others (n = 84 treated with DNV combination therapy (DNV group). The HCV genotype, virological response, adverse effects and some laboratory biochemical indexes were contrasted between above two groups in the real world study. RESULTS DNV group had significantly lower level of alpha-fetoprotein (AFP), lower rates of decompensated cirrhosis ( P < 0.05). ECS group possessed more 6a (31.91% vs.13.10%) while DNV group was provided with more 1b (48.81% vs. 22.34%) patients. Significantly poor liver function was detected in ECS group at 4-week treatment (ALT and AST) and 12-week follow-up (AST) (all P < 0.05). The SVR12 undetectable rates of both groups were 100%, and no serious event was observed during the treatment and follow-up in both groups. CONCLUSION In this retrospective real-world study, the efficacy of DNV combined therapy is similar to Sofosbuvir/Velpatasvir ± Ribavirin for chronic HCV infection, and the safety is comparable. DNV based therapy is a promising regimen for chronic hepatitis C.
Collapse
Affiliation(s)
- Yunjing Zhou
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-Sen University, No. 600 Tianhe Road , 510630, Guangzhou City, Guangdong Province, China
| | - Minfeng Liang
- Department of Infectious disease, The First people's hospital of Foshan, Foshan City, Guangdong Province, China
| | - Yiting Li
- Department of General Practice, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou City, Guangdong Province, China
| | - Xing Chen
- Department of Infectious Diseases, Yangjiang Public Health Hospital, Yangjiang City, Guangdong Province, China
| | - Jie Yang
- Clinical Medicine Department of Southwest Medical University, Luzhou City, Sichuan Province, China
| | - Honglian Bai
- Department of Infectious disease, The First people's hospital of Foshan, Foshan City, Guangdong Province, China
| | - Yingzi Long
- Department of Infectious Diseases, The Eighth Hospital of Guangzhou Medical University, Guangzhou City, Guangdong Province, China
| | - Xiaohong Zhang
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-Sen University, No. 600 Tianhe Road , 510630, Guangzhou City, Guangdong Province, China
| | - Chaoshuang Lin
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-Sen University, No. 600 Tianhe Road , 510630, Guangzhou City, Guangdong Province, China.
| |
Collapse
|
17
|
Appiah K, Rizzo M, Sarri G, Hernandez L. Justifying the source of external comparators in single-arm oncology health technology submissions: a review of NICE and PBAC assessments. J Comp Eff Res 2024; 13:e230140. [PMID: 38174576 PMCID: PMC10842296 DOI: 10.57264/cer-2023-0140] [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: 09/06/2023] [Accepted: 12/06/2023] [Indexed: 01/05/2024] Open
Abstract
Background: The drive to expedite patient access for diseases with high unmet treatment needs has come with an increasing use of single-arm trials (SATs), especially in oncology. However, the lack of control arms in such trials creates challenges to assess and demonstrate comparative efficacy. External control (EC) arms can be used to bridge this gap, with various types of sources available to obtain relevant data. Objective: To examine the source of ECs in single-arm oncology health technology assessment (HTA) submissions to the National Institute for Health and Care Excellence (NICE) and the Pharmaceutical Benefits Advisory Committee (PBAC) and how this selection was justified by manufacturers and assessed by the respective HTA body. Methods: Single-arm oncology HTA submission reports published by NICE (England) and PBAC (Australia) from January 2011 to August 2021 were reviewed, with data qualitatively synthesized to identify themes. Results: Forty-eight oncology submissions using EC arms between 2011 and 2021 were identified, with most submissions encompassing blood and bone marrow cancers (52%). In HTA submissions to NICE and PBAC, the EC arm was typically constructed from a combination of data sources, with the company's justification in data source selection infrequently provided (PBAC [2 out of 19]; NICE [6 out of 29]), although this lack of justification was not heavily criticized by either HTA body. Conclusion: Although HTA bodies such as NICE and PBAC encourage that EC source justification should be provided in submissions, this review found that this is not typically implemented in practice. Guidance is needed to establish best practices as to how EC selection should be documented in HTA submissions.
Collapse
Affiliation(s)
| | | | | | - Luis Hernandez
- Takeda Pharmaceuticals America, Inc., Lexington, MA, USA
| |
Collapse
|
18
|
Loureiro H, Roller A, Schneider M, Talavera-López C, Becker T, Bauer-Mehren A. Matching by OS Prognostic Score to Construct External Controls in Lung Cancer Clinical Trials. Clin Pharmacol Ther 2024; 115:333-341. [PMID: 37975320 DOI: 10.1002/cpt.3109] [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: 08/09/2023] [Accepted: 11/08/2023] [Indexed: 11/19/2023]
Abstract
External controls (eControls) leverage historical data to create non-randomized control arms. The lack of randomization can result in confounding between the experimental and eControl cohorts. To balance potentially confounding variables between the cohorts, one of the proposed methods is to match on prognostic scores. Still, the performance of prognostic scores to construct eControls in oncology has not been analyzed yet. Using an electronic health record-derived de-identified database, we constructed eControls using one of three methods: ROPRO, a state-of-the-art prognostic score, or either a propensity score composed of five (5Vars) or 27 covariates (ROPROvars). We compared the performance of these methods in estimating the overall survival (OS) hazard ratio (HR) of 11 recent advanced non-small cell lung cancer. The ROPRO eControls had a lower OS HR error (median absolute deviation (MAD), 0.072, confidence interval (CI): 0.036-0.185), than the 5Vars (MAD 0.081, CI: 0.025-0.283) and ROPROvars eControls (MAD 0.087, CI: 0.054-0.383). Notably, the OS HR errors for all methods were even lower in the phase III studies. Moreover, the ROPRO eControl cohorts included, on average, more patients than the 5Vars (6.54%) and ROPROvars cohorts (11.7%). The eControls matched with the prognostic score reproduced the controls more reliably than propensity scores composed of the underlying variables. Additionally, prognostic scores could allow eControls to be built on many prognostic variables without a significant increase in the variability of the propensity score, which would decrease the number of matched patients.
Collapse
Affiliation(s)
- Hugo Loureiro
- Data and Analytics, Pharma Research and Early Development, Roche Innovation Center Munich (RICM), Penzberg, Germany
- Computational Health Center, Helmholtz Munich, Munich, Germany
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Andreas Roller
- Early Development Oncology, Pharma Research and Early Development, Roche Innovation Center Basel (RICB), Basel, Switzerland
| | - Meike Schneider
- Early Development Oncology, Pharma Research and Early Development, Roche Innovation Center Basel (RICB), Basel, Switzerland
| | | | - Tim Becker
- Data and Analytics, Pharma Research and Early Development, Roche Innovation Center Munich (RICM), Penzberg, Germany
| | - Anna Bauer-Mehren
- Data and Analytics, Pharma Research and Early Development, Roche Innovation Center Munich (RICM), Penzberg, Germany
| |
Collapse
|
19
|
Schwartz DE, Essaouabi H, Trippa L. Clinical trials that leverage external data: Do we need more transparent protocols and statistical plans? Eur J Cancer 2024; 196:113443. [PMID: 38008030 DOI: 10.1016/j.ejca.2023.113443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/06/2023] [Accepted: 11/10/2023] [Indexed: 11/28/2023]
Affiliation(s)
- Daniel Evan Schwartz
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, United States.
| | | | - Lorenzo Trippa
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, United States
| |
Collapse
|
20
|
Siu DHW, Lin FPY, Cho D, Lord SJ, Heller GZ, Simes RJ, Lee CK. Framework for the Use of External Controls to Evaluate Treatment Outcomes in Precision Oncology Trials. JCO Precis Oncol 2024; 8:e2300317. [PMID: 38190581 DOI: 10.1200/po.23.00317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 09/03/2023] [Accepted: 10/13/2023] [Indexed: 01/10/2024] Open
Abstract
Advances in genomics have enabled anticancer therapies to be tailored to target specific genomic alterations. Single-arm trials (SATs), including those incorporated within umbrella, basket, and platform trials, are widely adopted when it is not feasible to conduct randomized controlled trials in rare biomarker-defined subpopulations. External controls (ECs), defined as control arm data derived outside the clinical trial, have gained renewed interest as a strategy to supplement evidence generated from SATs to allow comparative analysis. There are increasing examples demonstrating the application of EC in precision oncology trials. The prospective application of EC in conducting comparative studies is associated with distinct methodological challenges, the specific considerations for EC use in biomarker-defined subpopulations have not been adequately discussed, and a formal framework is yet to be established. In this review, we present a framework for conducting a prospective comparative analysis using EC. Key steps are (1) defining the purpose of using EC to address the study question, (2) determining if the external data are fit for purpose, (3) developing a transparent study protocol and a statistical analysis plan, and (iv) interpreting results and drawing conclusions on the basis of a prespecified hypothesis. We specify the considerations required for the biomarker-defined subpopulations, which include (1) specifying the comparator and biomarker status of the comparator group, (2) defining lines of treatment, (3) assessment of the biomarker testing panels used, and (4) assessment of cohort stratification in tumor-agnostic studies. We further discuss novel clinical trial designs and statistical techniques leveraging EC to propose future directions to advance evidence generation and facilitate drug development in precision oncology.
Collapse
Affiliation(s)
- Derrick H W Siu
- National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
- Department of Medical Oncology, Illawarra Cancer Care Centre, Wollongong, NSW, Australia
| | - Frank P Y Lin
- National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- School of Clinical Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Doah Cho
- National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
| | - Sarah J Lord
- National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
- School of Medicine, University of Notre Dame, Sydney, NSW, Australia
| | - Gillian Z Heller
- National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
- Mathematics and Statistics, Macquarie University, Macquarie Park, NSW, Australia
| | - R John Simes
- National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
| | - Chee Khoon Lee
- National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
- Cancer Care Centre, St George Hospital, Kogarah, NSW, Australia
| |
Collapse
|
21
|
Sferruzza G, Malcangi M, Bosco L, Finocchiaro G. Reassessing the efficacy of bevacizumab in newly diagnosed glioblastoma: A systematic review and external pseudodata-based analysis. Neurooncol Adv 2024; 6:vdad174. [PMID: 38390032 PMCID: PMC10883711 DOI: 10.1093/noajnl/vdad174] [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: 02/24/2024] Open
Abstract
Background First-line use of bevacizumab for glioblastoma (GBM) was evaluated in 2 phase 3 randomized controlled trials (RCT), demonstrating an impact on progression-free survival but not overall survival (OS). However, the crossover events of these trials raised concerns regarding the reliability of this latter analysis. In this study, we conducted an external control-based reassessment of the bevacizumab efficacy in newly diagnosed GBM (ndGBM) against the standard Stupp protocol. Methods A systematic review of the literature was conducted to identify the phase 3 RCTs in ndGBM incorporating the Stupp protocol as an arm. For the selected studies, we extracted individual patient survival pseudodata of the Stupp protocol arm by digitizing the Kaplan-Meier plots. A comprehensive pipeline was established to select suitable control studies as external benchmarks. Results Among the 13 identified studies identified in our systematic review, 4 studies resulted as comparable with the AVAglio trial and 2 with the RTOG 0825. Pooled individual patient pseudodata analysis showed no differences in terms of OS when bevacizumab was added to the Stupp protocol. Conclusions The external-controlled-based reassessment of the bevacizumab treatment in ndGBM confirmed its lack of efficacy in extending OS. Our study includes a summary table of individual patient survival pseudodata from all phase 3 RCTs in ndGBM employing the Stupp protocol and provides a pipeline that offers comprehensive guidance for conducting external control-based assessments in ndGBM.
Collapse
Affiliation(s)
- Giacomo Sferruzza
- Vita-Salute San Raffaele University, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Massimo Malcangi
- Vita-Salute San Raffaele University, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Luca Bosco
- Vita-Salute San Raffaele University, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurophysiology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | |
Collapse
|
22
|
Ahmed S, Wedekind MF, Del Rivero J, Raygada M, Lockridge R, Glod JW, Flowers C, Thomas BJ, Bernstein DB, Kapustina OB, Jain A, Miettinen M, Raffeld M, Xi L, Tyagi M, Kim J, Aldape K, Malayeri AA, Kaplan RN, Allen T, Vivelo CA, Sandler AB, Widemann BC, Reilly KM. Longitudinal Natural History Study of Children and Adults with Rare Solid Tumors: Initial Results for First 200 Participants. CANCER RESEARCH COMMUNICATIONS 2023; 3:2468-2482. [PMID: 37966258 PMCID: PMC10699159 DOI: 10.1158/2767-9764.crc-23-0247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 11/03/2023] [Accepted: 11/07/2023] [Indexed: 11/16/2023]
Abstract
Understanding of tumor biology and identification of effective therapies is lacking for many rare tumors. My Pediatric and Adult Rare Tumor (MyPART) network was established to engage patients, advocates, and researchers and conduct a comprehensive longitudinal Natural History Study of Rare Solid Tumors. Through remote or in-person enrollment at the NIH Clinical Center, participants with rare solid tumors ≥4 weeks old complete standardized medical and family history forms, patient reported outcomes, and provide tumor, blood and/or saliva samples. Medical records are extracted for clinical status and treatment history, and tumors undergo genomic analysis. A total of 200 participants (65% female, 35% male, median age at diagnosis 43 years, range = 2-77) enrolled from 46 U.S. states and nine other countries (46% remote, 55% in-person). Frequent diagnoses were neuroendocrine neoplasms (NEN), adrenocortical carcinomas (ACC), medullary thyroid carcinomas (MTC), succinate dehydrogenase (SDH)-deficient gastrointestinal stromal tumors (sdGIST), and chordomas. At enrollment, median years since diagnosis was 3.5 (range = 0-36.6), 63% participants had metastatic disease and 20% had no evidence of disease. Pathogenic germline and tumor mutations included SDHA/B/C (sdGIST), RET (MTC), TP53 and CTNNB1 (ACC), MEN1 (NEN), and SMARCB1 (poorly-differentiated chordoma). Clinically significant anxiety was observed in 20%-35% of adults. Enrollment of participants and comprehensive data collection were feasible. Remote enrollment was critical during the COVID-19 pandemic. Over 30 patients were enrolled with ACC, NEN, and sdGIST, allowing for clinical/genomic analyses across tumors. Longitudinal follow-up and expansion of cohorts are ongoing to advance understanding of disease course and establish external controls for interventional trials. SIGNIFICANCE This study demonstrates that comprehensive, tumor-agnostic data and biospecimen collection is feasible to characterize different rare tumors, and speed progress in research. The findings will be foundational to developing external controls groups for single-arm interventional trials, where randomized control trials cannot be conducted because of small patient populations.
Collapse
Affiliation(s)
- Shadin Ahmed
- Pediatric Oncology Branch, Center for Cancer Research, NCI, Bethesda, Maryland
| | | | - Jaydira Del Rivero
- Developmental Therapeutics Branch, Center for Cancer Research, NCI, Bethesda, Maryland
| | - Margarita Raygada
- Pediatric Oncology Branch, Center for Cancer Research, NCI, Bethesda, Maryland
| | - Robin Lockridge
- Clinical Research Directorate (CRD), Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - John W. Glod
- Pediatric Oncology Branch, Center for Cancer Research, NCI, Bethesda, Maryland
| | - Crystal Flowers
- Pediatric Oncology Branch, Center for Cancer Research, NCI, Bethesda, Maryland
| | - BJ Thomas
- Pediatric Oncology Branch, Center for Cancer Research, NCI, Bethesda, Maryland
| | - Donna B. Bernstein
- Pediatric Oncology Branch, Center for Cancer Research, NCI, Bethesda, Maryland
| | - Oxana B. Kapustina
- Pediatric Oncology Branch, Center for Cancer Research, NCI, Bethesda, Maryland
| | - Ashish Jain
- Pediatric Oncology Branch, Center for Cancer Research, NCI, Bethesda, Maryland
- Research Computing, Department of Information Technology, Boston Children's Hospital, Boston, Massachusetts
| | - Markku Miettinen
- Laboratory of Pathology, Center for Cancer Research, NCI, Bethesda, Maryland
| | - Mark Raffeld
- Laboratory of Pathology, Center for Cancer Research, NCI, Bethesda, Maryland
| | - Liqiang Xi
- Laboratory of Pathology, Center for Cancer Research, NCI, Bethesda, Maryland
| | - Manoj Tyagi
- Laboratory of Pathology, Center for Cancer Research, NCI, Bethesda, Maryland
| | - Jung Kim
- Laboratory of Pathology, Center for Cancer Research, NCI, Bethesda, Maryland
| | - Kenneth Aldape
- Laboratory of Pathology, Center for Cancer Research, NCI, Bethesda, Maryland
| | - Ashkan A. Malayeri
- Department of Radiology and Imaging Sciences, Clinical Center, NIH, Bethesda, Maryland
| | - Rosandra N. Kaplan
- Pediatric Oncology Branch, Center for Cancer Research, NCI, Bethesda, Maryland
| | - Taryn Allen
- Pediatric Oncology Branch, Center for Cancer Research, NCI, Bethesda, Maryland
- Clinical Research Directorate (CRD), Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Christina A. Vivelo
- Pediatric Oncology Branch, Center for Cancer Research, NCI, Bethesda, Maryland
- Kelly Government Solutions, Bethesda, Maryland
| | - Abby B. Sandler
- Pediatric Oncology Branch, Center for Cancer Research, NCI, Bethesda, Maryland
| | | | - Karlyne M. Reilly
- Pediatric Oncology Branch, Center for Cancer Research, NCI, Bethesda, Maryland
| | | |
Collapse
|
23
|
Børø S, Thoresen S, Boge Brant S, Helland Å. Initial investigation of using Norwegian health data for the purpose of external comparator arms - an example for non-small cell lung cancer. Acta Oncol 2023; 62:1642-1648. [PMID: 37801361 DOI: 10.1080/0284186x.2023.2264484] [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: 03/20/2023] [Accepted: 09/22/2023] [Indexed: 10/07/2023]
Abstract
BACKGROUND Using real-world (RW) data from registries to mimic or substitute a comparator arm of clinical trials is increasingly investigated. The feasibility of using data sources for this purpose depends on the source's completeness of the wide spectrum of a disease characteristics and relevant endpoints, which could allow for proper matching of important variables. MATERIALS AND METHODS This is a Norwegian study using data from three population-based registries, the Cancer Registry (CRN), the National Patient Registry (NPR), and the Norwegian Prescribed Drug Registry (LMR). We assessed if the registries contained the information necessary for selecting a RW cohort of patients with characteristics that mimicked the inclusion and exclusion criteria from the control arm of the phase 3 trial (KeyNote-407) on patients with stage IV, squamous NSCLC. We did this both on an aggregated - and individual data level. We also described the survival in the RW-cohorts and compared it with the survival in the control arm of the KN-407 trial. RESULTS Using aggregated data from the CRN allowed us to find the patients based on some clinically relevant inclusion criteria, but only to a limited extent could we apply the exclusion criteria of KN-407. When we used individual data from the CRN, NPR and the LMR we could create a patient cohort that shared more criteria corresponding to the eligibility criteria from KN-407, including exclusion criteria. Compared to the 11.3 months (CI 95% 9.5, 14.8) median survival in the control arm of KN-407, both RW-cohorts had a shorter median survival, 7.07 months (CI 95% 6.7, 9.5) (individual) and 8.0 months (CI 95% 5.8, 10.5) (aggregated). CONCLUSION Even if we demonstrated that the registries contain clinically relevant information necessary to mimic the eligibility criteria of a selected RCT, the survival is shorter for RW patients compared to their control arm counterpart in the RCT.
Collapse
Affiliation(s)
- Siri Børø
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Merck AB Norway, an affiliate of Merck KGaA, Oslo, Norway
| | - Steinar Thoresen
- Merck AB Norway, an affiliate of Merck KGaA, Oslo, Norway
- Nordic RWE, Oslo, Norway
| | | | - Åslaug Helland
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Oncology and Cancer Genetics, Institute for Cancer Research, Oslo University Hospital
| |
Collapse
|
24
|
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
|
25
|
Serrano C, Rothschild S, Villacampa G, Heinrich MC, George S, Blay JY, Sicklick JK, Schwartz GK, Rastogi S, Jones RL, Rutkowski P, Somaiah N, Navarro V, Evans D, Trent JC. Rethinking placebos: embracing synthetic control arms in clinical trials for rare tumors. Nat Med 2023; 29:2689-2692. [PMID: 37828359 DOI: 10.1038/s41591-023-02578-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Affiliation(s)
- César Serrano
- Department of Medical Oncology, Vall d'Hebron University Hospital, Barcelona, Spain.
- Sarcoma Translational Research Program, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.
| | | | - Guillermo Villacampa
- Oncology Data Science, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
- The Institute of Cancer Research, London, UK
| | - Michael C Heinrich
- Portland VA Health Care System and Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Suzanne George
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Jean-Yves Blay
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France
- Université Claude Bernard, Lyon, France
| | - Jason K Sicklick
- Department of Surgery, Division of Surgical Oncology, University of California San Diego, La Jolla, CA, USA
- Department of Pharmacology, University of California San Diego, San Diego, CA, USA
| | - Gary K Schwartz
- Case Comprehensive Cancer Center in Cleveland, Cleveland, OH, USA
| | - Sameer Rastogi
- Department of Medical Oncology, All India Institute of Medical Science (AIIMS), New Delhi, India
| | - Robin L Jones
- Division of Clinical Studies, The Royal Marsden NHS Foundation Trust, The Institute of Cancer Research, London, UK
| | - Piotr Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Neeta Somaiah
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Víctor Navarro
- Oncology Data Science, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | | | - Jonathan C Trent
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| |
Collapse
|
26
|
Campbell UB, Honig N, Gatto NM. SURF: A Screening Tool (for Sponsors) to Evaluate Whether Using Real-World Data to Support an Effectiveness Claim in an FDA Application Has Regulatory Feasibility. Clin Pharmacol Ther 2023; 114:981-993. [PMID: 37550832 DOI: 10.1002/cpt.3021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 07/24/2023] [Indexed: 08/09/2023]
Abstract
Based on recent guidance and publicly available approvals, the US Food and Drug Administration (FDA) has demonstrated its openness to considering evidence of effectiveness from real-world data (RWD) and nonrandomized studies (or "real-world evidence (RWE)") in its decision making. Through analysis of the FDA approvals, several authors have identified methodologic issues commonly discussed by FDA reviewers. However, in our analysis of FDA guidance and use cases, acceptability of RWE also critically depends on whether the characteristics of the clinical development program align with circumstances in which the FDA may have flexibility in considering evidence from real-world study designs relative to more robust designs. Successful use of RWD requires advance planning to allocate the necessary resources and time to undertake principled epidemiology approaches to study design, data selection, and implementation of analyses as well as address regulatory feedback. Thus, sponsors benefit from early identification of programs in which successful RWD use is more likely, to ensure efficient use of resources required for the next steps of scientific feasibility assessment. We developed SURF, a screening tool intended to help a sponsor decide whether to prioritize resources for further exploring the feasibility of using an RWD approach to meet the FDA's effectiveness evidentiary standards for a particular clinical development program. Here, we provide an analysis of FDA guidance, highlighting the circumstances in which RWD approaches may be most acceptable, and demonstrate the use of this screening tool, including the rationale for its construction, types of evidence needed, and application to publicly available approvals as support of concept.
Collapse
Affiliation(s)
- Ulka B Campbell
- Aetion, Inc., New York, New York, USA
- Columbia Mailman School of Public Health, New York, New York, USA
| | | | - Nicolle M Gatto
- Aetion, Inc., New York, New York, USA
- Columbia Mailman School of Public Health, New York, New York, USA
- Tulane School of Public Health and Tropical Medicine, New Orleans, Louisiana, USA
| |
Collapse
|
27
|
Jannin A, Giudici F, de la Fouchardière C, Al Ghuzlan A, Wassermann J, Chougnet CN, Drui D, Godbert Y, Ilouz F, Bardet S, Zanetta S, Roudaut N, Batisse Lignier M, Groussin L, Klein M, Zerdoud S, Lamartina L, Baudin E, Decaussin-Petrucci M, Leteurtre E, Borson Chazot F, Do Cao C, Borget I, Hadoux J. Factors Associated with Survival in Anaplastic Thyroid Carcinoma: A Multicenter Study from the ENDOCAN-TUTHYREF Network. Thyroid 2023; 33:1190-1200. [PMID: 37855745 DOI: 10.1089/thy.2023.0164] [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: 10/20/2023]
Abstract
Background: Anaplastic thyroid carcinoma (ATC) is a rare and frequently fatal type of thyroid cancer. The degree of heterogeneity in survival rates for ATC is incompletely studied. This study evaluated the factors associated with overall survival (OS) of patients with ATC using multicenter real-world data from a national tertiary care center network in France. Methods: In this multicenter, retrospective cohort study, all patients with ATC diagnosed between 2010 and 2020 were identified from the national database of the French ENDOCAN-TUTHYREF network. Factors associated with OS were examined in multivariable analyses using Cox proportional hazards models. Results: The study included 360 patients. Of these, 220 (61%) were female and the median age was 72 years (interquartile range: 62-80). The percentages of patients with pure and mixed (synchronously-transformed) ATC (p-ATC and st-ATC) were 62.5% and 26.7%, respectively. The median OS was 6.8 months [confidence interval, CI: 5.5-8.1]: not reached for stage IVa, 11.4 months [8.2-17.8] for IVb, and 4.6 months [3.5-5.7] for IVc. Surgery, radiation therapy to the neck, chemotherapy, and best supportive care were administered to 69 (19.2%), 214 (59.4%), 254 (70.6%), and 66 (18.3%) patients, respectively. In a multivariable analysis, including stage IVb-IVc patients, significantly higher OS was observed in patients with Eastern Cooperative Oncology Group performance-status of 0-1 (hazard ratio [HR], 0.6; [CI, 0.4-0.9], p < 0.02), stage IVb [HR, 0.5; CI, 0.4-0.8, p < 0.001], and multimodal treatment (surgery and chemoradiotherapy) [HR, 0.07; CI, 0.04-0.1, p < 0.001]. Variables associated with significantly worse OS included: p-ATC (vs. st-ATC) [HR, 1.83; CI, 1.33-2.51, p = 0.001] and a neutrophil-to-lymphocyte ratio (NLR) >5.05 [HR, 2.05, CI, 1.39-3.05, p < 0.001]. Conclusions: Factors independently associated with improved OS in ATC included: European Cooperative Oncology Group performance status, disease stage, multimodality treatment, synchronously transformed ATC, and lower NLR. Long-term OS was observed in selected patients with ATC who underwent multimodal treatment.
Collapse
Affiliation(s)
- Arnaud Jannin
- CHU Lille, Department of Endocrinology, Diabetology, and Metabolism, Lille, France
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277-CANTHER-Cancer-Heterogeneity Plasticity and Resistance to Therapies, Lille, France
| | - Fabiola Giudici
- Department of Biostatistics and Epidemiology, Gustave Roussy, Paris-Saclay University, Villejuif, France
- Oncostat, U1018, CESP, Inserm, Paris-Saclay University, "Ligue Contre le Cancer" labeled team, Villejuif, France
| | | | - Abir Al Ghuzlan
- Department of Biology and Pathology, Gustave Roussy Cancer Campus and University Paris-Saclay, Villejuif, France
| | - Johanna Wassermann
- Medical Oncology Department and Thyroid and Endocrine Tumors Department, Pitié-Salpêtrière University Hospital, AP-HP, IUC, Sorbonne University, Paris, France
| | - Cecile N Chougnet
- Endocrine Oncology Department, Saint-Louis Hospital (AP-HP), Université Paris, Paris, France
| | - Delphine Drui
- Nantes Université, CHU Nantes, Service d'Endocrinologie-Diabétologie et Nutrition, l'institut du thorax, Nantes, France
| | - Yann Godbert
- Nuclear Medicine and Endocrine Oncology Department, Institute of Bergonié, Bordeaux, France
| | | | - Stéphane Bardet
- Department of Nuclear Medicine and Thyroid Unit, Center François Baclesse, Caen, France
| | - Sylvie Zanetta
- Nuclear Medicine Department, Center Georges François Leclerc, Dijon, France
| | - Nathalie Roudaut
- Department of Endocrinology, University Hospital of Brest, Brest, France
| | | | - Lionel Groussin
- Endocrine Department, AP-HP, Hôpital Cochin, Université Paris Cité, CNRS, INSERM, Institut Cochin, Paris, France
| | - Marc Klein
- Department of Endocrinology, CHU de Nancy, Hôpital de Brabois, Vandoeuvre-lès-Nancy, France
| | - Slimane Zerdoud
- Department of Nuclear Medicine, Institute Claudius Regaud, Oncology University Institute, IUCT Oncopole, Toulouse, France
| | - Livia Lamartina
- Département d'imagerie, Service d'Oncologie Endocrinienne, Gustave Roussy, Villejuif, France
| | - Eric Baudin
- Département d'imagerie, Service d'Oncologie Endocrinienne, Gustave Roussy, Villejuif, France
| | - Myriam Decaussin-Petrucci
- Research on Healthcare Performance (RESHAPE), INSERM U1290, EA 3738 CICLY, Université Lyon 1, Claude Bernard, Lyon, France
- Service d'Anatomopathologie, Hospices Civils de Lyon, Groupement Hospitalier Sud, Pierre Bénite, France
| | - Emmanuelle Leteurtre
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277-CANTHER-Cancer-Heterogeneity Plasticity and Resistance to Therapies, Lille, France
| | - Francoise Borson Chazot
- Research on Healthcare Performance (RESHAPE), INSERM U1290, EA 3738 CICLY, Université Lyon 1, Claude Bernard, Lyon, France
- Endocrinology Department, Hospice Civils de Lyon, Hospital Louis-Pradel, Bron, France
| | - Christine Do Cao
- CHU Lille, Department of Endocrinology, Diabetology, and Metabolism, Lille, France
| | - Isabelle Borget
- Department of Biostatistics and Epidemiology, Gustave Roussy, Paris-Saclay University, Villejuif, France
- Oncostat, U1018, CESP, Inserm, Paris-Saclay University, "Ligue Contre le Cancer" labeled team, Villejuif, France
| | - Julien Hadoux
- Département d'imagerie, Service d'Oncologie Endocrinienne, Gustave Roussy, Villejuif, France
| |
Collapse
|
28
|
Pasqualetti F, Zanotti S. Nonrandomised controlled trial in recurrent glioblastoma patients: the promise of autologous tumour lysate-loaded dendritic cell vaccination. Br J Cancer 2023; 129:895-896. [PMID: 36792723 PMCID: PMC10491746 DOI: 10.1038/s41416-023-02194-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 01/24/2023] [Accepted: 01/27/2023] [Indexed: 02/17/2023] Open
Abstract
Recently, Liau et al. reported the results of Phase 3 clinical trial testing DCVax-L vaccines on patients with glioblastoma. Despite the promising and significant results obtained, the study design and the long-lasting period of recruitment of this work deserve some reflection.
Collapse
Affiliation(s)
- Francesco Pasqualetti
- Department of Oncology, University of Oxford, OX3 7DQ, Oxford, UK.
- Radiation Oncology, Azienda Ospedaliero Universitaria Pisana, Pisa, Italy.
| | - Sofia Zanotti
- Anatomic Pathology Unit, IRCCS Humanitas University Research Hospital, Milan, Italy.
| |
Collapse
|
29
|
Turkoz I, Wong J, Chee B, Siddiqui U, Knight RK, Richarz U, Correll CU. Comparative effectiveness study of paliperidone palmitate 6-month with a real-world external comparator arm of paliperidone palmitate 1-month or 3-month in patients with schizophrenia. Ther Adv Psychopharmacol 2023; 13:20451253231200258. [PMID: 37786804 PMCID: PMC10541743 DOI: 10.1177/20451253231200258] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 08/21/2023] [Indexed: 10/04/2023] Open
Abstract
Background The paliperidone palmitate 6-month (PP6M) long-acting injectable formulation is currently the longest dosing interval available for schizophrenia treatment. Objective To compare treatment outcomes between a real-world external comparator arm (ECA; NeuroBlu database) and the PP6M open-label extension (OLE) clinical trial arm. Methods The ECA comprised patients receiving PP 1-month (PP1M) or PP 3-month (PP3M) for ⩾12 months without a relapse. The PP6M OLE arm included patients with PP1M treatment prior to randomization who completed the 12-month double-blind PP6M study on either PP3M or PP6M relapse-free. Inverse probability treatment weighting (IPTW) was used to study time-to-relapse (primary outcome) and change in Clinical Global Impressions-Severity (CGI-S) score (secondary outcome). Results At 24 months, 3.9% (7/178) of patients in the PP6M cohort experienced a relapse versus 15.6% (26/167) in the ECA. Time-to-relapse was longer in the PP6M cohort versus the ECA at 12-, 18-, and 24-months across the different weighting methods; median time-to-relapse was not reached in both cohorts. Hazard ratio (HR) for relapse was significantly lower for the PP6M cohort versus the ECA throughout the duration of the study [HR at 24 months: 0.18 (95% CI: 0.08-0.42), p < 0.001]. At 24 months, change in CGI-S score for the PP6M cohort was 0.76 points lower than the ECA (p < 0.001). Results were similar in a sensitivity analysis using propensity score matching (PSM); IPTW resulted in larger sample sizes in balanced dataset than PSM. Conclusion Consistent findings across weighting and matching methods suggest PP6M efficacy in reducing and delaying relapses and long-term symptom control compared to PP1M/PP3M in usual-care settings. Additional confounds, such as greater illness severity and more frequent comorbidities and comedications in the ECA, were not fully controlled by the applied statistical methods. Future real-world studies directly comparing PP6M with PP3M/PP1M and adjusting for these confounders are warranted.
Collapse
Affiliation(s)
- Ibrahim Turkoz
- Janssen Research & Development, LLC, 1125 Trenton-Harbourton Road, Titusville, NJ 08560-0200, USA
| | | | | | - Uzma Siddiqui
- Janssen Research & Development, LLC, Titusville, NJ, USA
| | - R. Karl Knight
- Janssen Research & Development, LLC, Titusville, NJ, USA
| | - Ute Richarz
- Janssen Research & Development, Cilag Int., Zurich, Switzerland
| | - Christoph U. Correll
- The Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, NY, USA
- The Zucker Hillside Hospital, Department of Psychiatry, Northwell Health, Glen Oaks, NY, USA
- Charité – Universitätsmedizin Berlin, Department of Child and Adolescent Psychiatry, Berlin, Germany
| |
Collapse
|
30
|
Becker JC, Ugurel S, Leiter U, Meier F, Gutzmer R, Haferkamp S, Zimmer L, Livingstone E, Eigentler TK, Hauschild A, Kiecker F, Hassel JC, Mohr P, Fluck M, Thomas I, Garzarolli M, Grimmelmann I, Drexler K, Spillner AN, Eckhardt S, Schadendorf D. Adjuvant immunotherapy with nivolumab versus observation in completely resected Merkel cell carcinoma (ADMEC-O): disease-free survival results from a randomised, open-label, phase 2 trial. Lancet 2023; 402:798-808. [PMID: 37451295 DOI: 10.1016/s0140-6736(23)00769-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 04/02/2023] [Accepted: 04/06/2023] [Indexed: 07/18/2023]
Abstract
BACKGROUND Merkel cell carcinoma (MCC) is an immunogenic but aggressive skin cancer. Even after complete resection and radiation, relapse rates are high. PD-1 and PD-L1 checkpoint inhibitors showed clinical benefit in advanced MCC. We aimed to assess efficacy and safety of adjuvant immune checkpoint inhibition in completely resected MCC (ie, a setting without an established systemic standard-of-care treatment). METHODS In this multicentre phase 2 trial, patients (any stage, Eastern Cooperative Oncology Group performance status 0-1) at 20 academic medical centres in Germany and the Netherlands with completely resected MCC lesions were randomly assigned 2:1 to receive nivolumab 480 mg every 4 weeks for 1 year, or observation, stratified by stage (American Joint Committee on Cancer stages 1-2 vs stages 3-4), age (<65 vs ≥65 years), and sex. Landmark disease-free survival (DFS) at 12 and 24 months was the primary endpoint, assessed in the intention-to-treat populations. Overall survival and safety were secondary endpoints. This planned interim analysis was triggered when the last-patient-in was followed up for more than 1 year. This study is registered with ClinicalTrials.gov (NCT02196961) and with the EU Clinical Trials Register (2013-000043-78). FINDINGS Between Oct 1, 2014, and Aug 31, 2020, 179 patients were enrolled (116 [65%] stage 3-4, 122 [68%] ≥65 years, 111 [62%] male). Stratification factors (stage, age, sex) were balanced across the nivolumab (n=118) and internal control group (observation, n=61); adjuvant radiotherapy was more common in the control group. At a median follow-up of 24·3 months (IQR 19·2-33·4), median DFS was not reached (between-groups hazard ratio 0·58, 95% CI 0·30-1·12); DFS rates in the nivolumab group were 85% at 12 months and 84% at 24 months, and in the observation group were 77% at 12 months and 73% at 24 months. Overall survival results were not yet mature. Grade 3-4 adverse events occurred in 48 [42%] of 115 patients who received at least one dose of nivolumab and seven [11%] of 61 patients in the observation group. No treatment-related deaths were reported. INTERPRETATION Adjuvant therapy with nivolumab resulted in an absolute risk reduction of 9% (1-year DFS) and 10% (2-year DFS). The present interim analysis of ADMEC-O might suggest clinical use of nivolumab in this area of unmet medical need. However, overall survival events rates, with ten events in the active treatment group and six events in the half-the-size observation group, are not mature enough to draw conclusions. The explorative data of our trial support the continuation of ongoing, randomised trials in this area. ADMEC-O suggests that adjuvant immunotherapy is clinically feasible in this area of unmet medical need. FUNDING Bristol Myers Squibb.
Collapse
Affiliation(s)
- Jürgen C Becker
- Department of Dermatology, University Hospital Essen, Essen, Germany; Translational Skin Cancer Research, Department of Dermatology and West German Cancer Center, University of Medicine Duisburg-Essen, Essen, Germany; German Cancer Consortium, Partner Site Essen, Essen, Germany
| | - Selma Ugurel
- Department of Dermatology, University Hospital Essen, Essen, Germany; German Cancer Consortium, Partner Site Essen, Essen, Germany
| | - Ulrike Leiter
- Centre for Dermatooncology, Department of Dermatology, University Hospital Tübingen, Tübingen, Germany; German Cancer Consortium, Partner Site Tübingen, Tübingen, Germany
| | - Friedegund Meier
- Department of Dermatology, University Hospital Dresden, Dresden, Germany; German Cancer Consortium, Partner Site Dresden, Dresden, Germany
| | - Ralf Gutzmer
- Skin Cancer Center Hannover, Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany; Department of Dermatology, Johannes Wesling Medical Center, Ruhr University Bochum, Minden, Germany
| | - Sebastian Haferkamp
- Department of Dermatology, University Hospital Regensburg, Regensburg, Germany
| | - Lisa Zimmer
- Department of Dermatology, University Hospital Essen, Essen, Germany; German Cancer Consortium, Partner Site Essen, Essen, Germany
| | - Elisabeth Livingstone
- Department of Dermatology, University Hospital Essen, Essen, Germany; German Cancer Consortium, Partner Site Essen, Essen, Germany
| | - Thomas K Eigentler
- Centre for Dermatooncology, Department of Dermatology, University Hospital Tübingen, Tübingen, Germany; Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Axel Hauschild
- Department of Dermatology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Felix Kiecker
- Department of Dermatology and Venereology, Vivantes Klinikum Berlin Neukölln, Berlin, Germany; Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Jessica C Hassel
- Department of Dermatology, University Hospital Heidelberg, Heidelberg, Germany; German Cancer Consortium, Partner Site Heidelberg, Heidelberg, Germany
| | - Peter Mohr
- Department of Dermatology, Elbe-Kliniken, Buxtehude, Germany
| | - Michael Fluck
- Department of Oncology Hornheide, Fachklinik Hornheide, Münster, Germany
| | - Ioannis Thomas
- Centre for Dermatooncology, Department of Dermatology, University Hospital Tübingen, Tübingen, Germany
| | - Marlene Garzarolli
- Department of Dermatology, University Hospital Dresden, Dresden, Germany
| | - Imke Grimmelmann
- Skin Cancer Center Hannover, Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | - Konstantin Drexler
- Department of Dermatology, University Hospital Regensburg, Regensburg, Germany
| | | | | | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, Essen, Germany; German Cancer Consortium, Partner Site Essen, Essen, Germany.
| |
Collapse
|
31
|
Dagogo-Jack I. Targeted Approaches to Treatment of Pleural Mesothelioma: A Review. JCO Precis Oncol 2023; 7:e2300344. [PMID: 37992257 PMCID: PMC10681489 DOI: 10.1200/po.23.00344] [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: 06/30/2023] [Revised: 08/16/2023] [Accepted: 09/11/2023] [Indexed: 11/24/2023] Open
Abstract
Pleural mesothelioma is an aggressive disease that is enriched for inactivating alterations in tumor suppressor genes. Systemic therapeutic strategies for pleural mesothelioma generally involve chemotherapies and immunotherapies that are chosen without consideration of the tumor's molecular profile. As this generalized approach to treatment rarely yields durable responses, alternative therapeutic regimens are urgently indicated. Preclinical studies have identified synthetic lethal protein and metabolic interactions, recurrently overexpressed proteins, and frequent pathway perturbations that may be therapeutically exploited in mesothelioma. This review discusses the mechanism of action of emerging investigational therapies and summarizes findings from phase I-II clinical trials exploring selective, biomarker-driven therapeutic strategies for mesothelioma, with a focus on five common targets. Finally, using lessons learned from these clinical trials, imperatives for successful implementation of targeted therapy in mesothelioma are discussed.
Collapse
Affiliation(s)
- Ibiayi Dagogo-Jack
- Department of Medicine, Massachusetts General Hospital Cancer Center, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| |
Collapse
|
32
|
Allemang-Trivalle A, Leducq S, Maruani A, Giraudeau B. Designs used in published therapeutic studies of rare superficial vascular anomalies: a systematic literature search. BMC Med Res Methodol 2023; 23:196. [PMID: 37648985 PMCID: PMC10466846 DOI: 10.1186/s12874-023-02017-0] [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: 04/07/2022] [Accepted: 08/09/2023] [Indexed: 09/01/2023] Open
Abstract
BACKGROUND Rare superficial vascular anomalies represent a wide range of diseases. Their management is difficult given the broad spectrum and the lack of clinical trials assessing treatment efficacy. A randomized clinical trial of vascular anomalies is difficult because of the rarity of the diseases and is enhanced by the population of interest often being children. Therefore, suitable designs are needed. We conducted a methodological systematic literature search to identify designs implemented for investigating the treatment of rare superficial vascular anomalies. METHODS We conducted a literature search on January 25, 2021, of the PubMed, Cochrane Central Register of Controlled Trials (CENTRAL), Embase, ClinicalTrials.gov and European Union Clinical Trials Register databases. This systematic methodological literature search was registered at the Prospective Register of Systematic Reviews (PROSPERO: CRD42021232449). Randomized and non-randomized studies were included if they met the following criteria: were prospective studies of rare superficial vascular anomaly therapies, dealt with humans (adults and children) and were published in English from 2000. We excluded case reports/case series reporting fewer than 10 patients, reviews, retrospective studies, animal studies, studies of systemic or common vascular anomalies and non-therapeutic studies. We did not assess risk of bias in the included studies because our review was a methodological one focused on the design used. The review provided a descriptive analysis of relevant features of eligible research studies. RESULTS From 2046 articles identified, we included 97 studies (62 reports and 35 ongoing studies): 25 randomized controlled studies, 7 non-randomized comparative studies, 64 prospective cohorts and 1 case series. Among the 32 comparative studies included, 21 used a parallel-group design. The 11 other studies used different designs such as cross-over, randomized placebo phase, delayed-start, within-person, or challenge-dechallenge-rechallenge or used a historical control group or an observational run-in period. CONCLUSIONS Our systematic literature search highlights the lack of randomized control trials in superficial vascular anomalies due to the rarity of patients and their heterogeneity. New designs are emerging and can overcome the limitations of testing treatments in parallel groups.
Collapse
Affiliation(s)
- Aude Allemang-Trivalle
- Université de Tours, Université de Nantes, INSERM, SPHERE U1246, Tours, France.
- INSERM CIC1415, CHRU de Tours, Tours, France.
| | - Sophie Leducq
- Université de Tours, Université de Nantes, INSERM, SPHERE U1246, Tours, France
- Department of Dermatology, Reference Center for Genodermatoses and Rare Skin Diseases (Maladies Génétiques rares à Expression Cutanée-Tours), CHRU de Tours, Tours, France
| | - Annabel Maruani
- Université de Tours, Université de Nantes, INSERM, SPHERE U1246, Tours, France
- INSERM CIC1415, CHRU de Tours, Tours, France
- Department of Dermatology, Reference Center for Genodermatoses and Rare Skin Diseases (Maladies Génétiques rares à Expression Cutanée-Tours), CHRU de Tours, Tours, France
| | - Bruno Giraudeau
- Université de Tours, Université de Nantes, INSERM, SPHERE U1246, Tours, France
- INSERM CIC1415, CHRU de Tours, Tours, France
| |
Collapse
|
33
|
Arfè A, Narang C, DuBois SG, Reaman G, Bourgeois FT. Clinical development of new drugs for adults and children with cancer, 2010-2020. J Natl Cancer Inst 2023; 115:917-925. [PMID: 37171887 PMCID: PMC10407707 DOI: 10.1093/jnci/djad082] [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: 10/26/2022] [Revised: 01/30/2023] [Accepted: 05/09/2023] [Indexed: 05/14/2023] Open
Abstract
BACKGROUND Many new molecular entities enter clinical development to evaluate potential therapeutic benefits for oncology patients. We characterized adult and pediatric development of the set of new molecular entities that started clinical testing in 2010-2015 worldwide. METHODS We extracted data from AdisInsight, an extensive database of global pharmaceutical development, and the FDA.gov website. We followed the cohort of new molecular entities initiating first-in-human phase I clinical trials in 2010-2015 to the end of 2020. For each new molecular entity, we determined whether it was granted US Food and Drug Administration (FDA) approval, studied in a trial open to pediatric enrollment, or stalled during development. We characterized the cumulative incidence of these endpoints using statistical methods for censored data. RESULTS The 572 new molecular entities starting first-in-human studies in 2010-2015 were studied in 6142 trials by the end of 2020. Most new molecular entities were small molecules (n = 316, 55.2%), antibodies (n = 148, 25.9%), or antibody-drug conjugates (n = 44, 7.7%). After a mean follow-up of 8.0 years, 173 new molecular entities did not advance beyond first-in-human trials, and 39 were approved by the FDA. New molecular entities had a 10.4% estimated probability (95% confidence interval = 6.6% to 14.1%) of being approved by the FDA within 10 years of first-in-human trials. After a median of 4.6 years since start of first-in-human trials, 67 (11.7%) new molecular entities were tested in trials open to pediatric patients, and 5 (0.9%) were approved for pediatric indications. CONCLUSIONS More efficient clinical development strategies are needed to evaluate new cancer therapies, especially for children, and incorporate approaches to ensure knowledge gain from investigational products that stall in development.
Collapse
Affiliation(s)
- Andrea Arfè
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Claire Narang
- Pediatric Therapeutics and Regulatory Science Initiative, Computational Health Informatics Program (CHIP), Boston Children’s Hospital, Boston, MA, USA
| | - Steven G DuBois
- Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Gregory Reaman
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Florence T Bourgeois
- Pediatric Therapeutics and Regulatory Science Initiative, Computational Health Informatics Program (CHIP), Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
34
|
Sola-Morales O, Curtis LH, Heidt J, Walsh L, Casso D, Oliveria S, Saunders-Hastings P, Song Y, Mercado T, Zusterzeel R, Mastey V, Harnett J, Quek RGW. Effectively Leveraging RWD for External Controls: A Systematic Literature Review of Regulatory and HTA Decisions. Clin Pharmacol Ther 2023; 114:325-355. [PMID: 37079433 DOI: 10.1002/cpt.2914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 04/14/2023] [Indexed: 04/21/2023]
Abstract
Real-world data (RWD)-derived external controls can be used to contextualize efficacy findings for investigational therapies evaluated in uncontrolled trials. As the number of submissions to regulatory and health technology assessment (HTA) bodies using external controls rises, and in light of recent regulatory and HTA guidance on the appropriate use of RWD, there is a need to address the operational and methodological challenges impeding the quality of real-world evidence (RWE) generation and the consistency in evaluation of RWE across agencies. This systematic review summarizes publicly available information on the use of external controls to contextualize outcomes from uncontrolled trials for all indications from January 1, 2015, through August 20, 2021, that were submitted to the European Medicines Agency, the US Food and Drug Administration, and/or select major HTA bodies (National Institute for Health and Care Excellence (NICE), Haute Autorité de Santé (HAS), Institut für Qualität und Wirtschaftlichkeit im Gesundheitswesen (IQWiG), and Gemeinsamer Bundesausschuss (G-BA)). By systematically reviewing submissions to regulatory and HTA bodies in the context of recent guidance, this study provides quantitative and qualitative insights into how external control design and analytic choices may be viewed by different agencies in practice. The primary operational and methodological aspects identified for discussion include, but are not limited to, engagement of regulators and HTA bodies, approaches to handling missing data (a component of data quality), and selection of real-world endpoints. Continued collaboration and guidance to address these and other aspects will inform and assist stakeholders attempting to generate evidence using external controls.
Collapse
Affiliation(s)
- Oriol Sola-Morales
- Fundació Health Innovation Technology Transfer and International, University of Catalonia, Barcelona, Spain
| | - Lesley H Curtis
- Duke Clinical Research Institute, Durham, North Carolina, USA
| | - Julien Heidt
- IQVIA, Regulatory Science and Strategy, Falls Church, Virginia, USA
| | - Laura Walsh
- IQVIA, Epidemiology and Drug Safety Practice, Boston, Massachusetts, USA
| | - Deborah Casso
- IQVIA, Epidemiology and Drug Safety Practice, Seattle, Washington, USA
| | - Susan Oliveria
- IQVIA, Epidemiology and Drug Safety Practice, New York, New York, USA
| | | | - Yufei Song
- IQVIA, Epidemiology and Drug Safety Practice, Beijing, China
| | - Tiffany Mercado
- IQVIA, Regulatory Science and Strategy, Falls Church, Virginia, USA
| | | | - Vera Mastey
- Regeneron Pharmaceuticals Inc., Health Economics & Outcomes Research, Tarrytown, New York, USA
| | - James Harnett
- Regeneron Pharmaceuticals Inc., Health Economics & Outcomes Research, Tarrytown, New York, USA
| | - Ruben G W Quek
- Regeneron Pharmaceuticals Inc., Health Economics & Outcomes Research, Tarrytown, New York, USA
| |
Collapse
|
35
|
Silva P, Janjan N, Ramos KS, Udeani G, Zhong L, Ory MG, Smith ML. External control arms: COVID-19 reveals the merits of using real world evidence in real-time for clinical and public health investigations. Front Med (Lausanne) 2023; 10:1198088. [PMID: 37484840 PMCID: PMC10359981 DOI: 10.3389/fmed.2023.1198088] [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: 03/31/2023] [Accepted: 05/31/2023] [Indexed: 07/25/2023] Open
Abstract
Randomized controlled trials are considered the 'gold standard' to reduce bias by randomizing patients to an experimental intervention, versus placebo or standard of care cohort. There are inherent challenges to enrolling a standard of care or cohorts: costs, site engagement logistics, socioeconomic variability, patient willingness, ethics of placebo interventions, cannibalizing the treatment arm population, and extending study duration. The COVID-19 pandemic has magnified aspects of constraints in trial recruitment and logistics, spurring innovative approaches to reducing trial sizes, accelerating trial accrual while preserving statistical rigor. Using data from medical records and databases allows for construction of external control arms that reduce the costs of an external control arm (ECA) randomized to standard of care. Simultaneously examining covariates of the clinical outcomes in ECAs that are being measured in the interventional arm can be particularly useful in phase 2 trials to better understand social and genetic determinants of clinical outcomes that might inform pivotal trial design. The FDA and EMA have promulgated a number of publicly available guidance documents and qualification reports that inform the use of this regulatory science tool to streamline clinical development, of phase 4 surveillance, and policy aspects of clinical outcomes research. Availability and quality of real-world data (RWD) are a prevalent impediment to the use of ECAs given such data is not collected with the rigor and deliberateness that characterizes prospective interventional control arm data. Conversely, in the case of contemporary control arms, a clinical trial outcome can be compared to a contemporary standard of care in cases where the standard of care is evolving at a fast pace, such as the use of checkpoint inhibitors in cancer care. Innovative statistical methods are an essential aspect of an ECA strategy and regulatory paths for these innovative approaches have been navigated, qualified, and in some cases published.
Collapse
Affiliation(s)
- Patrick Silva
- Institute of Bioscience and Technology and Department of Translational Medical Sciences, College Station, TX, United States
| | - Nora Janjan
- Center for Community Health and Aging, School of Public Health, Texas A&M University, College Station, TX, United States
| | - Kenneth S. Ramos
- Institute of Bioscience and Technology and Department of Translational Medical Sciences, College Station, TX, United States
| | - George Udeani
- Department of Clinical Pharmacy, School of Pharmacy, Texas A&M University, College Station, TX, United States
| | - Lixian Zhong
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas A&M University, College Station, TX, United States
| | - Marcia G. Ory
- Center for Community Health and Aging, School of Public Health, Texas A&M University, College Station, TX, United States
| | - Matthew Lee Smith
- Center for Community Health and Aging, School of Public Health, Texas A&M University, College Station, TX, United States
| |
Collapse
|
36
|
Hao Y, Hsu WC, Parzynski CS, Degtyarev E, Hampson LV, Masood A, Wu WH. Effectiveness of tisagenlecleucel versus real-world standard of care in relapsed/refractory follicular lymphoma. J Comp Eff Res 2023; 12:e220173. [PMID: 37345672 PMCID: PMC10508300 DOI: 10.57264/cer-2022-0173] [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: 09/27/2022] [Accepted: 04/06/2023] [Indexed: 06/23/2023] Open
Abstract
Aim: To contextualize the effectiveness of tisagenlecleucel versus real-world standard of care (SoC) in relapsed/refractory follicular lymphoma. Materials & methods: A retrospective indirect matched comparison study using data from the phase II ELARA trial and the US Flatiron Health Research Database. Results: Complete response rate was 69.1 versus 17.7% and the overall response rate was 85.6 versus 58.1% in tisagenlecleucel versus SoC, post weighting by odds. For overall survival, an estimated reduction in the risk of death was observed in favor of tisagenlecleucel over SoC. The hazard ratio for progression-free survival was 0.45 (95% CI: 0.26, 0.88), and for time-to-next treatment was 0.34 (95% CI: 0.15, 0.78) with tisagenlecleucel versus SoC. Conclusion: A consistent trend toward improved efficacy end points was observed in favor of tisagenlecleucel versus SoC.
Collapse
Affiliation(s)
- Yanni Hao
- Novartis Pharmaceuticals Corporation, East Hanover, NJ 07936, USA
- Current affiliation: Gilead Sciences, Foster City, CA 94404, USA
| | | | | | | | | | - Aisha Masood
- Novartis Pharmaceuticals Corporation, East Hanover, NJ 07936, USA
| | | |
Collapse
|
37
|
Gatto L, Di Nunno V, Tosoni A, Bartolini S, Ranieri L, Franceschi E. DCVax-L Vaccination in Patients with Glioblastoma: Real Promise or Negative Trial? The Debate Is Open. Cancers (Basel) 2023; 15:3251. [PMID: 37370860 DOI: 10.3390/cancers15123251] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/09/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023] Open
Abstract
The lack of significant improvement in the prognosis of patients with GB over the last decades highlights the need for innovative treatments aimed at fighting this malignancy and increasing survival outcomes. The results of the phase III clinical trial of DCVax-L (autologous tumor lysate-loaded dendritic cell vaccination), which has been shown to increase both median survival and long-term survival in newly diagnosed and relapsed glioblastoma, have been enthusiastically received by the scientific community. However, this study deserves some reflections regarding methodological issues related to the primary endpoint change, the long accrual period, and the suboptimal validity of the external control population used as the comparison arm.
Collapse
Affiliation(s)
- Lidia Gatto
- Department of Oncology, Azienda Unità Sanitaria Locale (AUSL) Bologna, 40139 Bologna, Italy
| | - Vincenzo Di Nunno
- Department of Oncology, Azienda Unità Sanitaria Locale (AUSL) Bologna, 40139 Bologna, Italy
| | - Alicia Tosoni
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy
| | - Stefania Bartolini
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy
| | - Lucia Ranieri
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy
| | - Enrico Franceschi
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy
| |
Collapse
|
38
|
Rahman R, Ventz S, Redd R, Cloughesy T, Alexander BM, Wen PY, Trippa L. Accessible Data Collections for Improved Decision Making in Neuro-Oncology Clinical Trials. Clin Cancer Res 2023; 29:2194-2198. [PMID: 36939557 PMCID: PMC10681026 DOI: 10.1158/1078-0432.ccr-22-3524] [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: 11/13/2022] [Revised: 01/22/2023] [Accepted: 03/15/2023] [Indexed: 03/21/2023]
Abstract
Drug development can be associated with slow timelines, particularly for rare or difficult-to-treat solid tumors such as glioblastoma. The use of external data in the design and analysis of trials has attracted significant interest because it has the potential to improve the efficiency and precision of drug development. A recurring challenge in the use of external data for clinical trials, however, is the difficulty in accessing high-quality patient-level data. Academic research groups generally do not have access to suitable datasets to effectively leverage external data for planning and analyses of new clinical trials. Given the need for resources to enable investigators to benefit from existing data assets, we have developed the Glioblastoma External (GBM-X) Data Platform which will allow investigators in neuro-oncology to leverage our data collection and obtain analyses. GBM-X strives to provide an uncomplicated process to use external data, contextualize single-arm trials, and improve inference on treatment effects early in drug development. The platform is designed to welcome interested collaborators and integrate new data into the platform, with the expectation that the data collection can continue to grow and remain updated. With such features, GBM-X is designed to help to accelerate evaluation of therapies, to grow with collaborations, and to serve as a model to improve drug discovery for rare and difficult-to-treat tumors in oncology.
Collapse
Affiliation(s)
- Rifaquat Rahman
- Rifaquat Rahman, MD. Department of Radiation Oncology, Dana-Farber/Brigham and Women’s Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Steffen Ventz
- Steffen Ventz, PhD. Division of Biostatistics, University of Minnesota, Minnesota, MN, USA
| | - Robert Redd
- Robert Redd, MS Department of Data Sciences, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Timothy Cloughesy
- Timothy Cloughesy, MD. Neuro-Oncology Program and Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Brian M. Alexander
- Brian M. Alexander, MD. Department of Radiation Oncology, Dana-Farber/Brigham and Women’s Cancer Center, Harvard Medical School, Boston, MA, USA; Foundation Medicine, Inc., Cambridge, MA, USA
| | - Patrick Y. Wen
- Patrick Y. Wen, MD. Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Lorenzo Trippa
- Lorenzo Trippa, PhD. Department of Data Sciences, Dana-Farber Cancer Institute, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
39
|
de Godoy LL, Chawla S, Brem S, Wang S, O'Rourke DM, Nasrallah MP, Desai A, Loevner LA, Liau LM, Mohan S. Assessment of treatment response to dendritic cell vaccine in patients with glioblastoma using a multiparametric MRI-based prediction model. J Neurooncol 2023; 163:173-183. [PMID: 37129737 DOI: 10.1007/s11060-023-04324-4] [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: 02/15/2023] [Accepted: 04/24/2023] [Indexed: 05/03/2023]
Abstract
PURPOSE Autologous tumor lysate-loaded dendritic cell vaccine (DCVax-L) is a promising treatment modality for glioblastomas. The purpose of this study was to investigate the potential utility of multiparametric MRI-based prediction model in evaluating treatment response in glioblastoma patients treated with DCVax-L. METHODS Seventeen glioblastoma patients treated with standard-of-care therapy + DCVax-L were included. When tumor progression (TP) was suspected and repeat surgery was being contemplated, we sought to ascertain the number of cases correctly classified as TP + mixed response or pseudoprogression (PsP) from multiparametric MRI-based prediction model using histopathology/mRANO criteria as ground truth. Multiparametric MRI model consisted of predictive probabilities (PP) of tumor progression computed from diffusion and perfusion MRI-derived parameters. A comparison of overall survival (OS) was performed between patients treated with standard-of-care therapy + DCVax-L and standard-of-care therapy alone (external controls). Additionally, Kaplan-Meier analyses were performed to compare OS between two groups of patients using PsP, Ki-67, and MGMT promoter methylation status as stratification variables. RESULTS Multiparametric MRI model correctly predicted TP + mixed response in 72.7% of cases (8/11) and PsP in 83.3% (5/6) with an overall concordance rate of 76.5% with final diagnosis as determined by histopathology/mRANO criteria. There was a significant concordant correlation coefficient between PP values and histopathology/mRANO criteria (r = 0.54; p = 0.026). DCVax-L-treated patients had significantly prolonged OS than those treated with standard-of-care therapy (22.38 ± 12.8 vs. 13.8 ± 9.5 months, p = 0.040). Additionally, glioblastomas with PsP, MGMT promoter methylation status, and Ki-67 values below median had longer OS than their counterparts. CONCLUSION Multiparametric MRI-based prediction model can assess treatment response to DCVax-L in patients with glioblastoma.
Collapse
Affiliation(s)
- Laiz Laura de Godoy
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Sanjeev Chawla
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| | - Steven Brem
- Department of Neurosurgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Glioblastoma Translational Center of Excellence, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Sumei Wang
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Donald M O'Rourke
- Department of Neurosurgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Glioblastoma Translational Center of Excellence, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - MacLean P Nasrallah
- Glioblastoma Translational Center of Excellence, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Clinical Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Arati Desai
- Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Glioblastoma Translational Center of Excellence, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Laurie A Loevner
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Linda M Liau
- Department of Neurosurgery, University of California Los Angeles David Geffen School of Medicine & Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Suyash Mohan
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
40
|
Li L, Jemielita T. Confounding adjustment in the analysis of augmented randomized controlled trial with hybrid control arm. Stat Med 2023. [PMID: 37186394 DOI: 10.1002/sim.9753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 03/03/2023] [Accepted: 04/16/2023] [Indexed: 05/17/2023]
Abstract
The augmented randomized controlled trial (RCT) with hybrid control arm includes a randomized treatment group (RT), a smaller randomized control group (RC), and a large synthetic control (SC) group from real-world data. This kind of trial is useful when there is logistics and ethics hurdle to conduct a fully powered RCT with equal allocation, or when it is necessary to increase the power of the RCT by incorporating real-world data. A difficulty in the analysis of augmented RCT is that the SC and RC may be systematically different in the distribution of observed and unmeasured confounding factors, causing bias when the two control groups are analyzed together as hybrid controls. We propose to use propensity score (PS) analysis to balance the observed confounders between SC and RC. The possible bias caused by unmeasured confounders can be estimated and tested by analyzing propensity score adjusted outcomes from SC and RC. We also propose a partial bias correction (PBC) procedure to reduce bias from unmeasured confounding. Extensive simulation studies show that the proposed PS + PBC procedures can improve the efficiency and statistical power by effectively incorporating the SC into the RCT data analysis, while still control the estimation bias and Type I error inflation that might arise from unmeasured confounding. We illustrate the proposed statistical procedures with data from an augmented RCT in oncology.
Collapse
Affiliation(s)
- Liang Li
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Thomas Jemielita
- Early Oncology Statistics, Merck & Co., Inc., Rahway, New Jersey, USA
| |
Collapse
|
41
|
Mair MJ, Bartsch R, Le Rhun E, Berghoff AS, Brastianos PK, Cortes J, Gan HK, Lin NU, Lassman AB, Wen PY, Weller M, van den Bent M, Preusser M. Understanding the activity of antibody-drug conjugates in primary and secondary brain tumours. Nat Rev Clin Oncol 2023; 20:372-389. [PMID: 37085569 DOI: 10.1038/s41571-023-00756-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2023] [Indexed: 04/23/2023]
Abstract
Antibody-drug conjugates (ADCs), a class of targeted cancer therapeutics combining monoclonal antibodies with a cytotoxic payload via a chemical linker, have already been approved for the treatment of several cancer types, with extensive clinical development of novel constructs ongoing. Primary and secondary brain tumours are associated with high mortality and morbidity, necessitating novel treatment approaches. Pharmacotherapy of brain tumours can be limited by restricted drug delivery across the blood-brain or blood-tumour barrier, although data from phase II studies of the HER2-targeted ADC trastuzumab deruxtecan indicate clinically relevant intracranial activity in patients with brain metastases from HER2+ breast cancer. However, depatuxizumab mafodotin, an ADC targeting wild-type EGFR and EGFR variant III, did not provide a definitive overall survival benefit in patients with newly diagnosed or recurrent EGFR-amplified glioblastoma in phase II and III trials, despite objective radiological responses in some patients. In this Review, we summarize the available data on the central nervous system activity of ADCs from trials involving patients with primary and secondary brain tumours and discuss their clinical implications. Furthermore, we explore pharmacological determinants of intracranial activity and discuss the optimal design of clinical trials to facilitate development of ADCs for the treatment of gliomas and brain metastases.
Collapse
Affiliation(s)
- Maximilian J Mair
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
- Christian Doppler Laboratory for Personalized Immunotherapy, Medical University of Vienna, Vienna, Austria
| | - Rupert Bartsch
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Emilie Le Rhun
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Anna S Berghoff
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
- Christian Doppler Laboratory for Personalized Immunotherapy, Medical University of Vienna, Vienna, Austria
| | - Priscilla K Brastianos
- Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Neuro-Oncology, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Javier Cortes
- International Breast Cancer Center (IBCC), Pangaea Oncology, Quirónsalud Group, Madrid and Barcelona, Spain
- Faculty of Biomedical and Health Sciences, Department of Medicine, Universidad Europea de Madrid, Madrid, Spain
- Medical Scientia Innovation Research (MEDSIR), Barcelona, Spain
| | - Hui K Gan
- Cancer Therapies and Biology Group, Centre of Research Excellence in Brain Tumours, Olivia Newton-John Cancer Wellness and Research Centre, Austin Hospital, Heidelberg, VIC, Australia
- La Trobe University School of Cancer Medicine, Heidelberg, VIC, Australia
- Department of Medicine, University of Melbourne, Heidelberg, VIC, Australia
| | - Nancy U Lin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Andrew B Lassman
- Division of Neuro-Oncology, Department of Neurology, Herbert Irving Comprehensive Cancer Center, Columbia University Vagelos College of Physicians and Surgeons and New York-Presbyterian Hospital, New York, NY, USA
| | - Patrick Y Wen
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Michael Weller
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Martin van den Bent
- The Brain Tumour Center, Erasmus Medical Center Cancer Institute, Rotterdam, Netherlands
| | - Matthias Preusser
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria.
- Christian Doppler Laboratory for Personalized Immunotherapy, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
42
|
Mulder J, Teerenstra S, van Hennik PB, Pasmooij AMG, Stoyanova-Beninska V, Voest EE, de Boer A. Single-arm trials supporting the approval of anticancer medicinal products in the European Union: contextualization of trial results and observed clinical benefit. ESMO Open 2023; 8:101209. [PMID: 37054504 PMCID: PMC10163162 DOI: 10.1016/j.esmoop.2023.101209] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 02/28/2023] [Accepted: 03/06/2023] [Indexed: 04/15/2023] Open
Abstract
BACKGROUND Single-arm trials (SATs) can sometimes be used to support marketing authorization of anticancer medicinal products in the European Union. The level and durability of antitumor activity of the product as well as context are important aspects to determine the relevance of trial results. The aim of this study is to provide details on the contextualization of trial results and to evaluate the magnitude of benefit of medicinal products approved based on SATs. MATERIALS AND METHODS We focused on anticancer medicinal products for solid tumors approved on the basis of SAT results (2012-2021). Data were retrieved from European public assessment reports and/or published literature. The benefit of these medicinal products was evaluated via the European Society for Medical Oncology (ESMO)-Magnitude of Clinical Benefit Scale (MCBS). RESULTS Eighteen medicinal products were approved based on 21 SATs-few medicinal products were supported by >1 SAT. For the majority of clinical trials, a clinically relevant treatment effect was (pre)specified (71.4%) and most often an accompanying sample size calculation was provided. For 10 studies, each testing a different medicinal product, a justification for the threshold for a clinically relevant treatment effect could be identified. At least 12 out of 18 applications included information to facilitate the contextualization of trial results, including six supportive studies. Of the pivotal SATs analyzed (n = 21), three were assigned an ESMO-MCBS score of 4, which corresponds to 'substantial' benefit. CONCLUSIONS The clinical relevance of the treatment effects shown by medicinal products for solid tumors tested in SATs is dependent on the effect size and context. To better facilitate regulatory decision making, prespecifying and motivating a clinically relevant effect and aligning the sample size to that effect is important. External controls may facilitate in the contextualization process, but the associated limitations must be addressed.
Collapse
Affiliation(s)
- J Mulder
- Dutch Medicines Evaluation Board, Utrecht, The Netherlands.
| | - S Teerenstra
- Dutch Medicines Evaluation Board, Utrecht, The Netherlands; Department for Health Evidence, Biostatistics Section, Radboud University Medical Center, Nijmegen, The Netherlands
| | - P B van Hennik
- Dutch Medicines Evaluation Board, Utrecht, The Netherlands
| | - A M G Pasmooij
- Dutch Medicines Evaluation Board, Utrecht, The Netherlands
| | | | - E E Voest
- The Netherlands Cancer Institute, Amsterdam, The Netherlands; Oncode Institute, Amsterdam, The Netherlands
| | - A de Boer
- Dutch Medicines Evaluation Board, Utrecht, The Netherlands; Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
43
|
Preusser M, van den Bent M. Autologous tumor lysate-loaded dendritic cell vaccination (DCVax-L) in glioblastoma: Breakthrough or fata morgana? Neuro Oncol 2023; 25:631-634. [PMID: 36562460 PMCID: PMC10076936 DOI: 10.1093/neuonc/noac281] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Indexed: 12/24/2022] Open
Affiliation(s)
- Matthias Preusser
- Division of Oncology, Department of Medicine 1, Medical University, Vienna, Austria
| | - Martin J van den Bent
- Department of Neuro-oncology, The Brain Tumour Center at the Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| |
Collapse
|
44
|
Rahman R, Polley MYC, Alder L, Brastianos PK, Anders CK, Tawbi HA, Mehta M, Wen PY, Geyer S, de Groot J, Zadeh G, Piantadosi S, Galanis E, Khasraw M. Current drug development and trial designs in neuro-oncology: report from the first American Society of Clinical Oncology and Society for Neuro-Oncology Clinical Trials Conference. Lancet Oncol 2023; 24:e161-e171. [PMID: 36990614 PMCID: PMC10401610 DOI: 10.1016/s1470-2045(23)00005-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 12/11/2022] [Accepted: 01/05/2023] [Indexed: 03/29/2023]
Abstract
Successful drug development for people with cancers of the CNS has been challenging. There are multiple barriers to successful drug development including biological factors, rarity of the disease, and ineffective use of clinical trials. Based upon a series of presentations at the First Central Nervous System Clinical Trials Conference hosted by the American Society of Clinical Oncology and the Society for Neuro-Oncology, we provide an overview on drug development and novel trial designs in neuro-oncology. This Review discusses the challenges of therapeutic development in neuro-oncology and proposes strategies to improve the drug discovery process by enriching the pipeline of promising therapies, optimising trial design, incorporating biomarkers, using external data, and maximising efficacy and reproducibility of clinical trials.
Collapse
Affiliation(s)
- Rifaquat Rahman
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Mei-Yin C Polley
- Department of Public Health Sciences, University of Chicago, Chicago, IL, USA
| | - Laura Alder
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| | - Priscilla K Brastianos
- Massachusetts General Hospital, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Carey K Anders
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| | | | - Minesh Mehta
- Miami Cancer Institute, Baptist Hospital, Miami, FL, USA
| | - Patrick Y Wen
- Centre for Neuro-Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Susan Geyer
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - John de Groot
- University of California San Francisco Brain Tumor Center, San Francisco, CA, USA
| | - Gelareh Zadeh
- Department of Neurological Surgery University of Toronto, Toronto, ON, Canada
| | - Steven Piantadosi
- Department of Surgery, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Evanthia Galanis
- Department of Oncology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Rochester, MN, USA
| | - Mustafa Khasraw
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
45
|
Olivier T, Migliorini D. Autologous tumor lysate-loaded dendritic cell vaccination in glioblastoma: What happened to the evidence? Rev Neurol (Paris) 2023; 179:502-505. [PMID: 37012085 DOI: 10.1016/j.neurol.2023.03.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
Abstract
In patients with glioblastoma, the "DCVax-L" trial reported a survival benefit with the addition of autologous tumor lysate-loaded denditric cell vaccination to the standard-of-care (SoC) in patients with glioblastoma. The trial presented as a phase 3 externally controlled trial is showing an improvement in overall survival (OS) in patients receiving the vaccine therapy as compared to externally controlled patients, both in the newly diagnosed setting (median OS = 19.3 months versus 16.5 months; HR = 0.80; 98% CI, 0.00-0.94; P = 0.002) and in the recurrent setting (median OS = 13.2 months versus 7.8 months; HR = 0.58; 98% CI, 0.00-0.76; P < 0.001). Interestingly, the original endpoint, progression-free survival (PFS), was not improved by the experimental therapy. While we praise efforts to improve outcomes in a population representing a true unmet need, the trial's design, methods and report raise several issues undermining the ability to derive meaningful conclusion. These limitations are mainly driven by multiple changes occurring years after the trial ended. External controls were used in a trial originally randomizing patients, the primary endpoint was modified (OS instead of PFS), a new study population (recurrent glioblastoma) was added, and unplanned analyses were conducted, among several other changes. Additionally, due to inclusion criteria, the external controls likely selected patients with less favorable outcome as compared with patients enrolled in the trial, potentially biasing the reported survival benefit. In the absence of data sharing, these shortcomings will not be clarified. Dendritic cell vaccination remains a promising approach for GBM. It is therefore disappointing that due to key methodological limitations, the DCVax-L trial ultimately failed to provide sound conclusions about the potential efficacy of such approach for patients with glioblastoma.
Collapse
|
46
|
Sengupta S, Ntambwe I, Tan K, Liang Q, Paulucci D, Castellanos E, Fiore J, Lane S, Micsinai Balan M, Viraswami-Apanna K, Sethuraman V, Samant M, Tiwari R. Emulating Randomized Controlled Trials with Hybrid Control Arms in Oncology: A Case Study. Clin Pharmacol Ther 2023; 113:867-877. [PMID: 36606735 DOI: 10.1002/cpt.2841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 12/27/2022] [Indexed: 01/07/2023]
Abstract
This proof-of-concept study retrospectively assessed the feasibility of applying a hybrid control arm design to a completed phase III randomized controlled trial (RCT; CheckMate-057) in advanced non-small cell lung cancer using a real-world data (RWD) source. The emulated trial consists of an experimental arm (patients from the RCT experimental cohort) and a hybrid control arm (patients from the RCT and RWD control cohorts). For the RWD control cohort, this study used a nationwide electronic health record-derived de-identified database. Three frequentist statistical borrowing methods were evaluated: a two-step Cox model, a fixed Cox model, and propensity score-integrated composite likelihood ("Methods 1-3"). The experimental treatment effect for hybrid control designs were evaluated using hazard ratios (HRs) with 95% confidence interval (CI) estimated from the Cox models accounting for covariate differences. The reduction in study duration compared to the RCT was also evaluated. All three statistical borrowing methods achieved comparable experimental treatment effects to that observed in the CheckMate-057 clinical trial, with HRs of 0.73 (95% CI: 0.59, 0.92), 0.74 (95% CI: 0.61, 0.91), 0.72 (95% CI: 0.59, 0.88) for Methods 1-3, respectively. Reduction in study duration time was 99-115 days when borrowing 30-38 events for Methods 1-3, respectively. This study demonstrated that it is feasible to emulate an RCT using a hybrid control arm design using three frequentist propensity-score based statistical borrowing methods. Selection of an appropriate, fit-for-use RWD cohort is critical to minimizing bias in experimental treatment effect.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Ram Tiwari
- Bristol Myers Squibb, New York, New York, USA
| |
Collapse
|
47
|
Walter T, Lievre A, Coriat R, Malka D, Elhajbi F, Di Fiore F, Hentic O, Smith D, Hautefeuille V, Roquin G, Perrier M, Dahan L, Granger V, Sobhani I, Mineur L, Niccoli P, Assenat E, Scoazec JY, Le Malicot K, Lepage C, Lombard-Bohas C. Bevacizumab plus FOLFIRI after failure of platinum-etoposide first-line chemotherapy in patients with advanced neuroendocrine carcinoma (PRODIGE 41-BEVANEC): a randomised, multicentre, non-comparative, open-label, phase 2 trial. Lancet Oncol 2023; 24:297-306. [PMID: 36739879 DOI: 10.1016/s1470-2045(23)00001-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/26/2022] [Accepted: 01/03/2023] [Indexed: 02/05/2023]
Abstract
BACKGROUND There is no standard second-line treatment after platinum-etoposide chemotherapy for gastroenteropancreatic neuroendocrine carcinoma. We aimed to evaluate the efficacy of FOLFIRI plus bevacizumab, and FOLFIRI alone, in this setting. METHODS We did a randomised, non-comparative, open-label, phase 2 trial (PRODIGE 41-BEVANEC) at 26 hospitals in France. We included patients aged 18 years or older with locally advanced or metastatic gastroenteropancreatic neuroendocrine carcinoma or neuroendocrine carcinoma of unknown primary origin, documented progressive disease during or after first-line platinum-etoposide chemotherapy, and an Eastern Cooperative Oncology Group performance status of 0-2. Patients were randomly assigned (1:1; block size of three), without stratification, to receive FOLFIRI (irinotecan 180 mg/m2, calcium folinate 400 mg/m2 or levofolinate 200 mg/m2, and fluorouracil 400 mg/m2 bolus then 2400 mg/m2 over 46 h) plus bevacizumab 5 mg/kg or FOLFIRI alone, intravenously, every 2 weeks until disease progression or unacceptable toxicity. Neither patients nor investigators were masked to group assignment. The primary outcome was overall survival at 6 months after randomisation, evaluated in the modified intention-to-treat population (all enrolled and randomly assigned patients who received at least one cycle of FOLFIRI). This study is now complete and is registered with ClinicalTrials.gov, NCT02820857. FINDINGS Between Sept 5, 2017, and Feb 8, 2022, 150 patients were assessed for eligibility and 133 were enrolled and randomly assigned: 65 to the FOLFIRI plus bevacizumab group and 68 to the FOLFIRI group. 126 patients (59 in the FOLFIRI plus bevacizumab group and 67 in the FOLFIRI group) received at least one cycle of FOLFIRI and were included in the modified intention-to-treat population, 83 (66%) of whom were male and 43 (34%) were female, and the median age of the patients was 67 years (IQR 58-73). The primary tumour location was colorectal in 38 (30%) of 126 patients, pancreatic in 34 (27%), gastro-oesophageal in 22 (17%), and unknown in 23 (18%). After a median follow-up of 25·7 months (95% CI 22·0-38·2), 6-month overall survival was 53% (80% CI 43-61) in the FOLFIRI plus bevacizumab group and 60% (51-68) in the FOLFIRI group. Grade 3-4 adverse events that occurred in at least 5% of patients were neutropenia (eight [14%] patients), diarrhoea (six [10%]), and asthenia (five [8%]) in the FOLFIRI plus bevacizumab group, and neutropenia (seven [10%]) in the FOLFIRI group. One treatment-related death (ischaemic stroke) occurred in the FOLFIRI plus bevacizumab group. INTERPRETATION The addition of bevacizumab did not seem to increase the benefit of FOLFIRI with regard to overall survival. FOLFIRI could be considered as a standard second-line treatment in patients with gastroenteropancreatic neuroendocrine carcinoma. FUNDING French Ministry of Health and Roche SAS.
Collapse
Affiliation(s)
- Thomas Walter
- Department of Medical Oncology, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France; Gastroenterology and Technologies for Health, Research Unit INSERM UMR 1052 CNRS UMR 5286, Cancer Research Center of Lyon, Lyon, France.
| | - Astrid Lievre
- Digestive Unit, Hôpital Universitaire de Pontchaillou, Rennes, France
| | - Romain Coriat
- Gastroenterology Department, Hôpital Cochin, Paris, France
| | - David Malka
- Gastrointestinal Oncology Department, Gustave Roussy, Villejuif, France
| | - Farid Elhajbi
- Oncology Department, Centre Oscar Lambret, Lille, France
| | - Fréderic Di Fiore
- Gastroenterology Department, Hôpital Universitaire de Rouen, Rouen, France
| | - Olivia Hentic
- Gastroenterology-Pancreatology Department, Hôpital Beaujon, Clichy, France
| | - Denis Smith
- Hepatogastroenterology and Digestive Oncology, Hôpital Universitaire de Bordeaux, Pessac, France
| | - Vincent Hautefeuille
- Gastroenterology and Digestive Oncology, Hôpital Universitaire d'Amiens, Amiens, France
| | - Guillaume Roquin
- Gastroenterology and Digestive Oncology, Hôpital Universitaire d'Angers, Angers, France
| | - Marine Perrier
- Department of Hepatogastroenterology and Digestive Oncology, Hôpital Robert Debré, Reims, France
| | - Laetitia Dahan
- Digestive Oncology Department, Hôpital Universitaire La Timone, Marseille, France
| | - Victoire Granger
- Hepatogastroenterology Department, Hôpital Universitaire Michallon, Grenoble, France
| | - Iradj Sobhani
- Department of Hepatogastroenterology and Digestive Oncology, Hôpital Henry Mondor, Creteil, France
| | - Laurent Mineur
- Oncology Department, Clinique Sainte-Catherine, Avignon, France
| | | | - Eric Assenat
- Medical Oncology Department, Hôpital St Eloi, Montpellier, France
| | - Jean-Yves Scoazec
- Department of Surgical and Molecular Pathology, Gustave Roussy, Villejuif, France
| | - Karine Le Malicot
- Fédération Francophone de Cancérologie Digestive, EPICAD INSERM LNC-UMR 1231, University of Burgundy and Franche Comté, Dijon, France
| | - Côme Lepage
- Gastroenterology and Digestive Oncology, Hôpital Universitaire Le Bocage, Dijon, France
| | | |
Collapse
|
48
|
Kotecha G, Ventz S, Trippa L. Prospectively shared control data across concurrent randomised clinical trials. Eur J Cancer 2023; 181:18-20. [PMID: 36621117 PMCID: PMC9925400 DOI: 10.1016/j.ejca.2022.11.038] [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: 11/25/2022] [Accepted: 11/29/2022] [Indexed: 12/14/2022]
Abstract
Sharing data from control groups across concurrent randomised clinical trials with identical enrolment criteria and the same control therapy can translate into efficiencies for the drug development process. We discuss potential benefits and risks of prospective data-sharing plans for concurrent randomised trials.
Collapse
Affiliation(s)
- Gopal Kotecha
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA.
| | - Steffen Ventz
- Division of Biostatistics, School of Public Health, University of Minnesota, MN, USA
| | - Lorenzo Trippa
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
49
|
[Association of autologous tumor lysate-loaded dendritic cell vaccination with extension of survival among patients with newly diagnosed and recurrent glioblastoma]. Strahlenther Onkol 2023; 199:327-329. [PMID: 36662238 PMCID: PMC9938012 DOI: 10.1007/s00066-023-02049-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/16/2023] [Indexed: 01/21/2023]
|
50
|
Christopoulos P, Schlenk R, Kazdal D, Blasi M, Lennerz J, Shah R, Budczies J, Malek N, Fröhling S, Rosenquist R, Schirmacher P, Bozorgmehr F, Kuon J, Reck M, Thomas M, Stenzinger A. Real-world data for precision cancer medicine-A European perspective. Genes Chromosomes Cancer 2023. [PMID: 36852573 DOI: 10.1002/gcc.23135] [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: 01/13/2023] [Revised: 02/14/2023] [Accepted: 02/22/2023] [Indexed: 03/01/2023] Open
Abstract
Leveraging real-world data (RWD) for drug access is necessary to overcome a key challenge of modern precision oncology: tackling numerous low-prevalence oncogenic mutations across cancers. Withholding a potentially active medication in patients with rare mutations for the sake of control chemotherapy or "best" supportive care is neither practicable nor ethically justifiable anymore, particularly as RWD could meanwhile be used instead, according to scientific principles outlined by the US Food and Drug Administration, European Medicines Agency and other stakeholders. However, practical implementation varies, with occasionally opposite recommendations based on the same evidence in different countries. In the face of growing need for precision drugs, more transparency of evaluation, a priori availability of guidance for the academia and industry, as well as a harmonized framework for health technology assessment across the European Union (EU) are imperative. These could in turn trigger infrastructural changes in national and pan-European registries, cancer management guidelines (e.g., frequency of routine radiologic restaging, inclusion of patient-reported outcomes), and the health data space, to ensure conformity with declared standards and facilitate extraction of RWD sets (including patient-level data) suitable for approval and pricing with minimal effort. For an EU-wide unification of precision cancer medicine, collective negotiation of drug supply contracts and funding solidarity would additionally be required to handle the financial burden. According to experience from pivotal European programs, off-label use could potentially also be harmonized across EU-states to accelerate availability of novel drugs, streamline collection of valuable RWD, and mitigate related costs through wider partnerships with pharmaceutical companies.
Collapse
Affiliation(s)
- Petros Christopoulos
- Department of Oncology, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany.,Department of Medical Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital, Heidelberg, Germany.,German Center for Lung Research (DZL), Gießen, Germany.,Centers for Personalized Medicine (ZPM), Germany
| | - Richard Schlenk
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital, Heidelberg, Germany.,NCT Trial Center, National Center of Tumor Diseases, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Daniel Kazdal
- German Center for Lung Research (DZL), Gießen, Germany.,Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Miriam Blasi
- Department of Oncology, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany
| | - Jochen Lennerz
- Machachussets General Hospital, Harvard University, Boston, USA
| | - Rajiv Shah
- Department of Oncology, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany.,German Center for Lung Research (DZL), Gießen, Germany
| | - Jan Budczies
- Centers for Personalized Medicine (ZPM), Germany.,Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Nisar Malek
- Centers for Personalized Medicine (ZPM), Germany.,Department of Gastroenterology, Tübingen University Hospital, Tübingen, Germany
| | - Stefan Fröhling
- Centers for Personalized Medicine (ZPM), Germany.,Department of Translational Medical Oncology, National Center for Tumor Diseases, Heidelberg, Germany.,German Cancer Consortium (DKTK), Germany
| | - Richard Rosenquist
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Peter Schirmacher
- Centers for Personalized Medicine (ZPM), Germany.,Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany.,German Cancer Consortium (DKTK), Germany
| | - Farastuk Bozorgmehr
- Department of Oncology, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany.,German Center for Lung Research (DZL), Gießen, Germany
| | - Jonas Kuon
- Department of Oncology, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany.,German Center for Lung Research (DZL), Gießen, Germany.,Department of Oncology, Lungenklinik Löwenstein, Löwenstein, Germany
| | - Martin Reck
- German Center for Lung Research (DZL), Gießen, Germany.,Department of Thoracic Oncology, Lungenclinic Großhansdorf, Großhansdorf, Germany
| | - Michael Thomas
- Department of Oncology, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany.,Department of Medical Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital, Heidelberg, Germany.,German Center for Lung Research (DZL), Gießen, Germany
| | - Albrecht Stenzinger
- German Center for Lung Research (DZL), Gießen, Germany.,Centers for Personalized Medicine (ZPM), Germany.,Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany.,German Cancer Consortium (DKTK), Germany
| |
Collapse
|