51
|
Furth SL. Trials and Tribulations - The Challenges of Clinical Trials in Children. NEJM EVIDENCE 2023; 2:EVIDe2300280. [PMID: 38320507 DOI: 10.1056/evide2300280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
In this issue of NEJM Evidence, we see the results of a randomized clinical trial of dapagliflozin or saxagliptin in pediatric type 2 diabetes (T2D). In children and adolescents with T2D, dapagliflozin achieved significant improvements in glycemia in the trial.1 In June 2023, following another pivotal trial, the U.S. Food and Drug Administration (FDA) approved empagliflozin and the combination of empagliflozin and metformin as additions to diet and exercise to improve blood sugar control in children 10 years and older with T2D. Metformin, the only other oral therapy available for the treatment of children with T2D, was first approved for pediatric use in 2000.
Collapse
Affiliation(s)
- Susan L Furth
- Research Institute, Children's Hospital of Philadelphia, Philadelphia
- Division of Nephrology, Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| |
Collapse
|
52
|
Ahmed MA, Burnham J, Dwivedi G, AbuAsal B. Achieving big with small: quantitative clinical pharmacology tools for drug development in pediatric rare diseases. J Pharmacokinet Pharmacodyn 2023; 50:429-444. [PMID: 37140724 DOI: 10.1007/s10928-023-09863-x] [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: 03/04/2023] [Accepted: 04/26/2023] [Indexed: 05/05/2023]
Abstract
Pediatric populations represent a major fraction of rare diseases and compound the intrinsic challenges of pediatric drug development and drug development for rare diseases. The intertwined complexities of pediatric and rare disease populations impose unique challenges to clinical pharmacologists and require integration of novel clinical pharmacology and quantitative tools to overcome multiple hurdles during the discovery and development of new therapies. Drug development strategies for pediatric rare diseases continue to evolve to meet the inherent challenges and produce new medicines. Advances in quantitative clinical pharmacology research have been a key component in advancing pediatric rare disease research to accelerate drug development and inform regulatory decisions. This article will discuss the evolution of the regulatory landscape in pediatric rare diseases, the challenges encountered during the design of rare disease drug development programs and will highlight the use of innovative tools and potential solutions for future development programs.
Collapse
Affiliation(s)
- Mariam A Ahmed
- Takeda Development Center Americas Inc, 125 Binney St, Cambridge, MA, 02142-1123, USA.
| | | | - Gaurav Dwivedi
- Takeda Development Center Americas Inc, 125 Binney St, Cambridge, MA, 02142-1123, USA
| | - Bilal AbuAsal
- US Food and Drug Administration, 10903, New Hampshire Ave, Silver Spring, MD, 20993, USA
| |
Collapse
|
53
|
Yang L, Atakhanova N, Arellano MTC, Mohamed MY, Hani T, Fahdil AA, Castillo-Acobo RY, Juyal A, Hussein AK, Amin AH, Pecho RDC, Akhavan-Sigari R. Translational research of new developments in targeted therapy of colorectal cancer. Pathol Res Pract 2023; 252:154888. [PMID: 37948996 DOI: 10.1016/j.prp.2023.154888] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 10/08/2023] [Accepted: 10/11/2023] [Indexed: 11/12/2023]
Abstract
A severe global health concern is the rising incidence and mortality rate of colorectal cancer (CRC). Chemotherapy, which is typically used to treat CRC, is known to have limited specificity and can have noticeable side effects. A paradigm shift in cancer treatment has been brought about by the development of targeted therapies, which has led to the appearance of pharmacological agents with improved efficacy and decreased toxicity. Epidermal growth factor receptor (EGFR), vascular endothelial growth factor (VEGF), human epidermal growth factor receptor 2 (HER2), and BRAF are among the molecular targets covered in this review that are used in targeted therapy for CRC. The current discussion also covers advancements in targeted therapeutic approaches, such as antibody-drug conjugates, immune checkpoint inhibitors, and chimeric antigen receptor (CAR) T-cell therapy. A review of the clinical trials and application of these particular therapies in treating CRC is also done. Despite the improvements in targeted therapy for CRC, problems such as drug resistance and patient selection remain to be solved. Despite this, targeted therapies have offered fresh possibilities for identifying and treating CRC, paving the way for the development of personalized medicine and extending the life expectancy and general well-being of CRC patients.
Collapse
Affiliation(s)
- Lei Yang
- Department of Clinical Laboratory, People's Hospital of Chongqing Liangjiang New Area, Chongqing 401121, China
| | - Nigora Atakhanova
- Head of the Department of Oncology, Tashkent Medical Academy, Tashkent 100109, Uzbekistan
| | | | | | - Thamer Hani
- Dentistry Department, Al-Turath University College, Baghdad, Iraq
| | - Ali A Fahdil
- Medical technical college, Al-Farahidi University, Iraq
| | | | - Ashima Juyal
- Uttaranchal Institute of Technology, Uttaranchal University, Dehradun 248007, India
| | | | - Ali H Amin
- Deanship of Scientific Research, Umm Al-Qura University, Makkah, Saudi Arabia
| | | | - Reza Akhavan-Sigari
- Department of Neurosurgery, University Medical Center Tuebingen, Germany; Department of Health Care Management and Clinical Research, Collegium Humanum Warsaw Management University Warsaw, Poland
| |
Collapse
|
54
|
Naderalvojoud B, Shah ND, Mutanga JN, Belov A, Staiger R, Chen JH, Whitaker B, Hernandez-Boussard T. Trends in Influenza Vaccination Rates among a Medicaid Population from 2016 to 2021. Vaccines (Basel) 2023; 11:1712. [PMID: 38006044 PMCID: PMC10675465 DOI: 10.3390/vaccines11111712] [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: 09/26/2023] [Revised: 10/28/2023] [Accepted: 11/03/2023] [Indexed: 11/26/2023] Open
Abstract
Seasonal influenza is a leading cause of death in the U.S., causing significant morbidity, mortality, and economic burden. Despite the proven efficacy of vaccinations, rates remain notably low, especially among Medicaid enrollees. Leveraging Medicaid claims data, this study characterizes influenza vaccination rates among Medicaid enrollees and aims to elucidate factors influencing vaccine uptake, providing insights that might also be applicable to other vaccine-preventable diseases, including COVID-19. This study used Medicaid claims data from nine U.S. states (2016-2021], encompassing three types of claims: fee-for-service, major Medicaid managed care plan, and combined. We included Medicaid enrollees who had an in-person healthcare encounter during an influenza season in this period, excluding those under 6 months of age, over 65 years, or having telehealth-only encounters. Vaccination was the primary outcome, with secondary outcomes involving in-person healthcare encounters. Chi-square tests, multivariable logistic regression, and Fisher's exact test were utilized for statistical analysis. A total of 20,868,910 enrollees with at least one healthcare encounter in at least one influenza season were included in the study population between 2016 and 2021. Overall, 15% (N = 3,050,471) of enrollees received an influenza vaccine between 2016 and 2021. During peri-COVID periods, there was an increase in vaccination rates among enrollees compared to pre-COVID periods, from 14% to 16%. Children had the highest influenza vaccination rates among all age groups at 29%, whereas only 17% were of 5-17 years, and 10% were of the 18-64 years were vaccinated. We observed differences in the likelihood of receiving the influenza vaccine among enrollees based on their health conditions and medical encounters. In a study of Medicaid enrollees across nine states, 15% received an influenza vaccine from July 2016 to June 2021. Vaccination rates rose annually, peaking during peri-COVID seasons. The highest uptake was among children (6 months-4 years), and the lowest was in adults (18-64 years). Female gender, urban residency, and Medicaid-managed care affiliation positively influenced uptake. However, mental health and substance abuse disorders decreased the likelihood. This study, reliant on Medicaid claims data, underscores the need for outreach services.
Collapse
Affiliation(s)
- Behzad Naderalvojoud
- Department of Medicine, Stanford University, Stanford, CA 94305, USA; (B.N.); (R.S.)
- Stanford Center for Biomedical Informatics Research, Stanford, CA 94305, USA
| | - Nilpa D. Shah
- Department of Medicine, Stanford University, Stanford, CA 94305, USA; (B.N.); (R.S.)
- Stanford Center for Biomedical Informatics Research, Stanford, CA 94305, USA
| | - Jane N. Mutanga
- Center for Biologics Evaluation and Research, Office of Biostatistics and Pharmacovigilance, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA; (J.N.M.)
| | - Artur Belov
- Center for Biologics Evaluation and Research, Office of Biostatistics and Pharmacovigilance, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA; (J.N.M.)
| | - Rebecca Staiger
- Department of Medicine, Stanford University, Stanford, CA 94305, USA; (B.N.); (R.S.)
| | - Jonathan H. Chen
- Department of Medicine, Stanford University, Stanford, CA 94305, USA; (B.N.); (R.S.)
- Stanford Center for Biomedical Informatics Research, Stanford, CA 94305, USA
- Division of Hospital Medicine, Stanford, CA 94305, USA
- Clinical Excellence Research Center, Stanford, CA 94304, USA
| | - Barbee Whitaker
- Center for Biologics Evaluation and Research, Office of Biostatistics and Pharmacovigilance, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA; (J.N.M.)
| | - Tina Hernandez-Boussard
- Department of Medicine, Stanford University, Stanford, CA 94305, USA; (B.N.); (R.S.)
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
55
|
Mehtälä J, Ali M, Miettinen T, Partanen L, Laapas K, Niemelä PT, Khorlo I, Ström S, Kurki S, Vapalahti J, Abdelgawwad K, Leinonen JV. Utilization of anonymization techniques to create an external control arm for clinical trial data. BMC Med Res Methodol 2023; 23:258. [PMID: 37925415 PMCID: PMC10625188 DOI: 10.1186/s12874-023-02082-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 10/26/2023] [Indexed: 11/06/2023] Open
Abstract
BACKGROUND Subject-level real-world data (RWD) collected during daily healthcare practices are increasingly used in medical research to assess questions that cannot be addressed in the context of a randomized controlled trial (RCT). A novel application of RWD arises from the need to create external control arms (ECAs) for single-arm RCTs. In the analysis of ECAs against RCT data, there is an evident need to manage and analyze RCT data and RWD in the same technical environment. In the Nordic countries, legal requirements may require that the original subject-level data be anonymized, i.e., modified so that the risk to identify any individual is minimal. The aim of this study was to conduct initial exploration on how well pseudonymized and anonymized RWD perform in the creation of an ECA for an RCT. METHODS This was a hybrid observational cohort study using clinical data from the control arm of the completed randomized phase II clinical trial (PACIFIC-AF) and RWD cohort from Finnish healthcare data sources. The initial pseudonymized RWD were anonymized within the (k, ε)-anonymity framework (a model for protecting individuals against identification). Propensity score matching and weighting methods were applied to the anonymized and pseudonymized RWD, to balance potential confounders against the RCT data. Descriptive statistics for the potential confounders and overall survival analyses were conducted prior to and after matching and weighting, using both the pseudonymized and anonymized RWD sets. RESULTS Anonymization affected the baseline characteristics of potential confounders only marginally. The greatest difference was in the prevalence of chronic obstructive pulmonary disease (4.6% vs. 5.4% in the pseudonymized compared to the anonymized data, respectively). Moreover, the overall survival changed in anonymization by only 8% (95% CI 4-22%). Both the pseudonymized and anonymized RWD were able to produce matched ECAs for the RCT data. Anonymization after matching impacted overall survival analysis by 22% (95% CI -21-87%). CONCLUSIONS Anonymization may be a viable technique for cases where flexible data transfer and sharing are required. As anonymization necessarily affects some aspects of the original data, further research and careful consideration of anonymization strategies are needed.
Collapse
Affiliation(s)
| | - Mehreen Ali
- Veil.ai Oy, Helsinki, Finland
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Timo Miettinen
- Veil.ai Oy, Helsinki, Finland
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
56
|
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
|
57
|
Carbone A, Vitullo P, Di Gioia S, Conese M. Lung Inflammatory Genes in Cystic Fibrosis and Their Relevance to Cystic Fibrosis Transmembrane Conductance Regulator Modulator Therapies. Genes (Basel) 2023; 14:1966. [PMID: 37895314 PMCID: PMC10606852 DOI: 10.3390/genes14101966] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 10/16/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Cystic fibrosis (CF) is a monogenic syndrome determined by over 2000 mutations in the CF Transmembrane Conductance Regulator (CFTR) gene harbored on chromosome 7. In people with CF (PWCF), lung disease is the major determinant of morbidity and mortality and is characterized by a clinical phenotype which differs in the presence of equal mutational assets, indicating that genetic and environmental modifiers play an important role in this variability. Airway inflammation determines the pathophysiology of CF lung disease (CFLD) both at its onset and progression. In this narrative review, we aim to depict the inflammatory process in CF lung, with a particular emphasis on those genetic polymorphisms that could modify the clinical outcome of the respiratory disease in PWCF. The natural history of CF has been changed since the introduction of CFTR modulator therapies in the clinical arena. However, also in this case, there is a patient-to-patient variable response. We provide an overview on inflammatory/immunity gene variants that affect CFLD severity and an appraisal of the effects of CFTR modulator therapies on the inflammatory process in lung disease and how this knowledge may advance the optimization of the management of PWCF.
Collapse
Affiliation(s)
- Annalucia Carbone
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy; (A.C.); (S.D.G.)
| | - Pamela Vitullo
- Cystic Fibrosis Support Center, Ospedale “G. Tatarella”, 71042 Cerignola, Italy;
| | - Sante Di Gioia
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy; (A.C.); (S.D.G.)
| | - Massimo Conese
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy; (A.C.); (S.D.G.)
| |
Collapse
|
58
|
Larsen K, Walton RN, Elsayed M, Ipatov A, Townsend-Holyoake F, Axelsson SFA, Quinones N, Papsch R, Givens J, Bedenkov A, Seewald M. A blueprint for success in real-world evidence: "glocal" approach to building capabilities and generating impactful evidence. Front Pharmacol 2023; 14:1233617. [PMID: 37886128 PMCID: PMC10598715 DOI: 10.3389/fphar.2023.1233617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 09/27/2023] [Indexed: 10/28/2023] Open
Abstract
The past decade has seen the increasing influence and relevance of real-world data (RWD) and real-world evidence (RWE) in healthcare decision making. The value added by RWD/RWE has prompted the pharmaceutical industry to develop high performing systems and practices to harness the power of evidence generated at the global level. However, this worldwide transformation provides outstanding opportunities to support capability building within local affiliates and to impact key country-level stakeholders through resulting evidence. Therefore, we present an Evidence Blueprint Initiative, which links the global and local ("glocal") skills, and furthermore addresses the opportunities and gaps in evidence generation capabilities at the local level. Cross-functional experts were recruited at the local, regional, and global level to define best practices. A framework was developed to characterize the foundational expertise needed and to assess markets' existing capabilities. Subsequently, targeted roadmaps were developed and implemented to build capabilities in specific areas within each affiliate. The impact from the Blueprint is encouraging, resulting in improved local evidence plans, established evidence teams, enhanced RWD use and strategic implementation of patient centric science in local affiliates. The success of the Blueprint resides in empowering affiliates to realise their local evidence generation ambitions and to match them to their local context. It strengthens and expands the ties between various parts of the organisation and the external environment while building fit-for-future evidence capabilities from local affiliates.
Collapse
Affiliation(s)
| | - Ryan N. Walton
- AstraZeneca Medical, Europe and Canada Region, Baar, Switzerland
| | - Mohamed Elsayed
- AstraZeneca Medical, International Region, Dubai, United Arab Emirates
| | - Andrey Ipatov
- AstraZeneca Medical, International Region, Moscow, Russia
| | | | | | - Nacho Quinones
- IQVIA, EMEA Real World Solutions, Medical Evidence Practice, London, United Kingdom
| | - Rudiger Papsch
- IQVIA, EMEA Real World Solutions, Medical Evidence Practice, London, United Kingdom
| | - Jennifer Givens
- AstraZeneca Medical, Global, Gaithersburg, MD, United States
| | | | | |
Collapse
|
59
|
Zhu R, Vora B, Menon S, Younis I, Dwivedi G, Meng Z, Datta-Mannan A, Manchandani P, Nayak S, Tammara BK, Garhyan P, Iqbal S, Dagenais S, Chanu P, Mukherjee A, Ghobadi C. Clinical Pharmacology Applications of Real-World Data and Real-World Evidence in Drug Development and Approval-An Industry Perspective. Clin Pharmacol Ther 2023; 114:751-767. [PMID: 37393555 DOI: 10.1002/cpt.2988] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 06/19/2023] [Indexed: 07/04/2023]
Abstract
Since the 21st Century Cures Act was signed into law in 2016, real-world data (RWD) and real-world evidence (RWE) have attracted great interest from the healthcare ecosystem globally. The potential and capability of RWD/RWE to inform regulatory decisions and clinical drug development have been extensively reviewed and discussed in the literature. However, a comprehensive review of current applications of RWD/RWE in clinical pharmacology, particularly from an industry perspective, is needed to inspire new insights and identify potential future opportunities for clinical pharmacologists to utilize RWD/RWE to address key drug development questions. In this paper, we review the RWD/RWE applications relevant to clinical pharmacology based on recent publications from member companies in the International Consortium for Innovation and Quality in Pharmaceutical Development (IQ) RWD Working Group, and discuss the future direction of RWE utilization from a clinical pharmacology perspective. A comprehensive review of RWD/RWE use cases is provided and discussed in the following categories of application: drug-drug interaction assessments, dose recommendation for patients with organ impairment, pediatric plan development and study design, model-informed drug development (e.g., disease progression modeling), prognostic and predictive biomarkers/factors identification, regulatory decisions support (e.g., label expansion), and synthetic/external control generation for rare diseases. Additionally, we describe and discuss common sources of RWD to help guide appropriate data selection to address questions pertaining to clinical pharmacology in drug development and regulatory decision making.
Collapse
Affiliation(s)
- Rui Zhu
- Clinical Pharmacology, Genentech, Inc., South San Francisco, California, USA
| | - Bianca Vora
- Clinical Pharmacology, Genentech, Inc., South San Francisco, California, USA
| | - Sujatha Menon
- Clinical Pharmacology, Pfizer Inc., Groton, Connecticut, USA
| | - Islam Younis
- Clinical Pharmacology, Gilead Sciences, Inc., Foster City, California, USA
| | - Gaurav Dwivedi
- Quantitative Clinical Pharmacology, Takeda Development Center Americas, Inc., Cambridge, Massachusetts, USA
| | - Zhaoling Meng
- R&D Data and Data Science, Clinical Modeling & Evidence Integration, Sanofi, Cambridge, Massachusetts, USA
| | - Amita Datta-Mannan
- Exploratory Medicine & Pharmacology, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Pooja Manchandani
- Clinical Pharmacology and Exploratory Division, Astellas Pharma Global Development, Northbrook, Illinois, USA
| | | | | | - Parag Garhyan
- Global PK/PD/Pharmacometrics, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Shahed Iqbal
- Biomarker Sciences, Gilead Sciences, Inc., Foster City, California, USA
| | - Simon Dagenais
- Real World Evidence Center of Excellence, Pfizer, Inc., New York, New York, USA
| | - Pascal Chanu
- Clinical Pharmacology, Genentech/Roche, Inc., Lyon, France
| | - Arnab Mukherjee
- Clinical Pharmacology, Pfizer Inc., Groton, Connecticut, USA
| | - Cyrus Ghobadi
- Exploratory Medicine & Pharmacology, Eli Lilly and Company, Indianapolis, Indiana, USA
| |
Collapse
|
60
|
Morrato EH, Lennox LA, Dearing JW, Coughlan AT, Gano ES, McFadden D, Mora N, Pincus HA, Firestein GS, Toto R, Reis SE. The Evolve to Next-Gen ACT Network: An evolving open-access, real-world data resource primed for real-world evidence research across the Clinical and Translational Science Award Consortium. J Clin Transl Sci 2023; 7:e224. [PMID: 38028333 PMCID: PMC10643916 DOI: 10.1017/cts.2023.617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 08/09/2023] [Accepted: 08/21/2023] [Indexed: 12/01/2023] Open
Abstract
The ACT Network was funded by NIH to provide investigators from across the Clinical and Translational Science Award (CTSA) Consortium the ability to directly query national federated electronic health record (EHR) data for cohort discovery and feasibility assessment of multi-site studies. NIH refunded the program for expanded research application to become "Evolve to Next-Gen ACT" (ENACT). In parallel, the US Food and Drug Administration has been evaluating the use of real-world data (RWD), including EHR data, as sources of real-world evidence (RWE) for its regulatory decisions involving drug and biological products. Using insights from implementation science, six lessons learned from ACT for developing and sustaining RWD/RWE infrastructures and networks across the CTSA Consortium are presented in order to inform ENACT's development from the outset. Lessons include intentional institutional relationship management, end-user engagement, beta-testing, and customer-driven adaptation. The ENACT team is also conducting customer discovery interviews with CTSA hub and investigators using Innovation-Corps@NCATS (I-Corps™) methodology for biomedical entrepreneurs to uncover unmet RWD needs. Possible ENACT value proposition hypotheses are presented by stage of research. Developing evidence about methods for sustaining academically derived data infrastructures and support can advance the science of translation and support our nation's RWD/RWE research capacity.
Collapse
Affiliation(s)
- Elaine H. Morrato
- Parkinson School of Health Sciences and Public Health, Loyola University Chicago, Chicago, IL, USA
- Institute for Translational Medicine, Loyola University Chicago, Chicago, IL, USA
| | - Lindsay A. Lennox
- Colorado Clinical and Translational Sciences Institute, CU Anschutz Medical Campus, Aurora, CO, USA
| | - James W. Dearing
- College of Communications, Arts and Sciences, Michigan State University, East Lansing, MI, USA
| | - Anne T. Coughlan
- Kellogg School of Management, Northwestern University, Evanston, IL, USA
| | | | - Doug McFadden
- Harvard Catalyst, Harvard University, Boston, MA, USA
| | - Nallely Mora
- Parkinson School of Health Sciences and Public Health, Loyola University Chicago, Chicago, IL, USA
- Institute for Translational Medicine, Loyola University Chicago, Chicago, IL, USA
| | - Harold Alan Pincus
- Irving Institute for Clinical and Translational Research, Columbia University and New York State Psychiatric Institute, New York, NY, USA
| | - Gary S. Firestein
- Altman Clinical and Translational Research Institute at the University of California San Diego, San Diego, CA, USA
| | - Robert Toto
- Center for Translational Medicine, UT Southwestern, Dallas, TX, USA
| | - Steven E. Reis
- Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
61
|
Curtis LH, Sola-Morales O, Heidt J, Saunders-Hastings P, Walsh L, Casso D, Oliveria S, Mercado T, Zusterzeel R, Sobel RE, Jalbert JJ, Mastey V, Harnett J, Quek RGW. Regulatory and HTA Considerations for Development of Real-World Data Derived External Controls. Clin Pharmacol Ther 2023; 114:303-315. [PMID: 37078264 DOI: 10.1002/cpt.2913] [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: 04/12/2023] [Indexed: 04/21/2023]
Abstract
Regulators and Health Technology Assessment (HTA) bodies are increasingly familiar with, and publishing guidance on, external controls derived from real-world data (RWD) to generate real-world evidence (RWE). We recently conducted a systematic literature review (SLR) evaluating publicly available information on the use of RWD-derived external controls to contextualize outcomes from uncontrolled trials submitted to the European Medicines Agency (EMA), the US Food and Drug Administration (FDA), and/or select HTA bodies. The review identified several key operational and methodological aspects for which more detailed guidance and alignment within and between regulatory agencies and HTA bodies is necessary. This paper builds on the SLR findings by delineating a set of key takeaways for the responsible generation of fit-for-purpose RWE. Practical methodological and operational guidelines for designing, conducting, and reporting RWD-derived external control studies are explored and discussed. These considerations include: (i) early engagement with regulators and HTA bodies during the study planning phase; (ii) consideration of the appropriateness and comparability of external controls across multiple dimensions, including eligibility criteria, temporality, population representation, and clinical evaluation; (iii) ensuring adequate sample sizes, including hypothesis testing considerations; (iv) implementation of a clear and transparent strategy for assessing and addressing data quality, including data missingness across trials and RWD; (v) selection of comparable and meaningful endpoints that are operationalized and analyzed using appropriate analytic methods; and (vi) conduct of sensitivity analyses to assess the robustness of findings in the context of uncertainty and sources of potential bias.
Collapse
Affiliation(s)
- Lesley H Curtis
- Duke Department of Population Health Sciences and Duke Clinical Research Institute, Durham, North Carolina, USA
| | - Oriol Sola-Morales
- Fundació HiTT and Universitat Internacional de Catalunya (UIC), Barcelona, Spain
| | - 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
| | - Tiffany Mercado
- IQVIA, Regulatory Science and Strategy, Falls Church, Virginia, USA
| | | | - Rachel E Sobel
- Regeneron Pharmaceuticals Inc., Pharmacoepidemiology, Tarrytown, New York, USA
| | - Jessica J Jalbert
- Regeneron Pharmaceuticals Inc., Health Economics & Outcomes Research, Tarrytown, New York, 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
|
62
|
Shau WY, Setia S, Shinde S, Santoso H, Furtner D. Generating fit-for-purpose real-world evidence in Asia: How far are we from closing the gaps? Perspect Clin Res 2023; 14:108-113. [PMID: 37554247 PMCID: PMC10405531 DOI: 10.4103/picr.picr_193_22] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 12/29/2022] [Accepted: 01/12/2023] [Indexed: 08/10/2023] Open
Abstract
Evidence generated by randomized controlled trials (RCTs) does not often represent the patient journey and clinical outcomes in the real world due to limited external validity or generalizability. Studies based on real-world data are intended to generalize results to the broader population; however, if the influence of external factors or confounders is not effectively managed, the cause-and-effect relationship and internal validity may be challenged, resulting in flawed results. The collection of quality real-world evidence (RWE) is crucial in Asia as there is often an underrepresentation of Asian populations in RCTs. In addition, few countries in Asia are catching up with the Western world in issuing practical foundational principles and guidance for conducting and adopting evidence for regulatory and reimbursement decisions. However, privacy and data protection laws are generally lagging behind technological developments in electronic medical records. While leveraging RWE in clinical and regulatory decision-making holds excellent potential, collective efforts across industry, governments, and research institutions are required for generating standardized practices and building capabilities for developing fit-for-purpose RWE in Asia.
Collapse
Affiliation(s)
- Wen-Yi Shau
- Medical Affairs, Emerging Asia, Pfizer Corporation Hong Kong Limited, Quarry Bay, Hong Kong
| | - Sajita Setia
- Executive Office Transform Medical Communications Limited, Wanganui, New Zealand
| | - Salil Shinde
- Medical Affairs, Emerging Asia, Pfizer Corporation Hong Kong Limited, Quarry Bay, Hong Kong
| | - Handoko Santoso
- Medical Affairs, Emerging Asia, Pfizer Corporation Hong Kong Limited, Quarry Bay, Hong Kong
| | - Daniel Furtner
- Executive Office Transform Medical Communications Limited, Wanganui, New Zealand
| |
Collapse
|
63
|
Wang M, Sushil M, Miao BY, Butte AJ. Bottom-up and top-down paradigms of artificial intelligence research approaches to healthcare data science using growing real-world big data. J Am Med Inform Assoc 2023; 30:1323-1332. [PMID: 37187158 PMCID: PMC10280344 DOI: 10.1093/jamia/ocad085] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 04/03/2023] [Accepted: 05/04/2023] [Indexed: 05/17/2023] Open
Abstract
OBJECTIVES As the real-world electronic health record (EHR) data continue to grow exponentially, novel methodologies involving artificial intelligence (AI) are becoming increasingly applied to enable efficient data-driven learning and, ultimately, to advance healthcare. Our objective is to provide readers with an understanding of evolving computational methods and help in deciding on methods to pursue. TARGET AUDIENCE The sheer diversity of existing methods presents a challenge for health scientists who are beginning to apply computational methods to their research. Therefore, this tutorial is aimed at scientists working with EHR data who are early entrants into the field of applying AI methodologies. SCOPE This manuscript describes the diverse and growing AI research approaches in healthcare data science and categorizes them into 2 distinct paradigms, the bottom-up and top-down paradigms to provide health scientists venturing into artificial intelligent research with an understanding of the evolving computational methods and help in deciding on methods to pursue through the lens of real-world healthcare data.
Collapse
Affiliation(s)
- Michelle Wang
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, California, USA
| | - Madhumita Sushil
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, California, USA
| | - Brenda Y Miao
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, California, USA
| | - Atul J Butte
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, California, USA
- Department of Pediatrics, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
64
|
Alberto IRI, Alberto NRI, Ghosh AK, Jain B, Jayakumar S, Martinez-Martin N, McCague N, Moukheiber D, Moukheiber L, Moukheiber M, Moukheiber S, Yaghy A, Zhang A, Celi LA. The impact of commercial health datasets on medical research and health-care algorithms. Lancet Digit Health 2023; 5:e288-e294. [PMID: 37100543 PMCID: PMC10155113 DOI: 10.1016/s2589-7500(23)00025-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 12/26/2022] [Accepted: 02/03/2023] [Indexed: 04/28/2023]
Abstract
As the health-care industry emerges into a new era of digital health driven by cloud data storage, distributed computing, and machine learning, health-care data have become a premium commodity with value for private and public entities. Current frameworks of health data collection and distribution, whether from industry, academia, or government institutions, are imperfect and do not allow researchers to leverage the full potential of downstream analytical efforts. In this Health Policy paper, we review the current landscape of commercial health data vendors, with special emphasis on the sources of their data, challenges associated with data reproducibility and generalisability, and ethical considerations for data vending. We argue for sustainable approaches to curating open-source health data to enable global populations to be included in the biomedical research community. However, to fully implement these approaches, key stakeholders should come together to make health-care datasets increasingly accessible, inclusive, and representative, while balancing the privacy and rights of individuals whose data are being collected.
Collapse
Affiliation(s)
| | | | - Arnab K Ghosh
- Department of Medicine, Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - Bhav Jain
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | | | - Ned McCague
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA; Markforged, Watertown, MA, USA
| | - Dana Moukheiber
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Lama Moukheiber
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Mira Moukheiber
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sulaiman Moukheiber
- Department of Computer Science, Worcester Polytechnic Institute, Worcester, MA, USA
| | - Antonio Yaghy
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA; New England Eye Center, Tufts University Medical Center, Boston, MA, USA
| | - Andrew Zhang
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Leo Anthony Celi
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA; Department of Biostatistics, Harvard T H Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
65
|
Velummailum RR, McKibbon C, Brenner DR, Stringer EA, Ekstrom L, Dron L. Data Challenges for Externally Controlled Trials: Viewpoint. J Med Internet Res 2023; 25:e43484. [PMID: 37018021 PMCID: PMC10132012 DOI: 10.2196/43484] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 02/01/2023] [Accepted: 02/19/2023] [Indexed: 02/21/2023] Open
Abstract
The preferred evidence of a large randomized controlled trial is difficult to adopt in scenarios, such as rare conditions or clinical subgroups with high unmet needs, and evidence from external sources, including real-world data, is being increasingly considered by decision makers. Real-world data originate from many sources, and identifying suitable real-world data that can be used to contextualize a single-arm trial, as an external control arm, has several challenges. In this viewpoint article, we provide an overview of the technical challenges raised by regulatory and health reimbursement agencies when evaluating comparative efficacy, such as identification, outcome, and time selection challenges. By breaking down these challenges, we provide practical solutions for researchers to consider through the approaches of detailed planning, collection, and record linkage to analyze external data for comparative efficacy.
Collapse
Affiliation(s)
| | | | - Darren R Brenner
- Department of Oncology, University of Calgary, Calgary, AB, Canada
| | | | | | | |
Collapse
|
66
|
Heavner SF, Anderson W, Kashyap R, Dasher P, Mathé EA, Merson L, Guerin PJ, Weaver J, Robinson M, Schito M, Kumar VK, Nagy P. A Path to Real-World Evidence in Critical Care Using Open-Source Data Harmonization Tools. Crit Care Explor 2023; 5:e0893. [PMID: 37025303 PMCID: PMC10072311 DOI: 10.1097/cce.0000000000000893] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023] Open
Abstract
COVID-19 highlighted the need for use of real-world data (RWD) in critical care as a near real-time resource for clinical, research, and policy efforts. Analysis of RWD is gaining momentum and can generate important evidence for policy makers and regulators. Extracting high quality RWD from electronic health records (EHRs) requires sophisticated infrastructure and dedicated resources. We sought to customize freely available public tools, supporting all phases of data harmonization, from data quality assessments to de-identification procedures, and generation of robust, data science ready RWD from EHRs. These data are made available to clinicians and researchers through CURE ID, a free platform which facilitates access to case reports of challenging clinical cases and repurposed treatments hosted by the National Center for Advancing Translational Sciences/National Institutes of Health in partnership with the Food and Drug Administration. This commentary describes the partnership, rationale, process, use case, impact in critical care, and future directions for this collaborative effort.
Collapse
|
67
|
Teodorescu M, Grigorescu AC. Date de tip real world privind utilizarea nivolumabului în cancerul pulmonar nonmicrocelular avansat – dincolo de linia a doua. Experienţa noastră în România imediat după rambursarea imunoterapiei prin sistemul naţional de sănătate. ONCOLOG-HEMATOLOG.RO 2023. [DOI: 10.26416/onhe.62.1.2023.7750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
|
68
|
Abstract
Real-world evidence (RWE) is clinical evidence on a medical product's safety and efficacy that is generated using real-world data (RWD) resulting from routine healthcare delivery. There are several sources of RWD, including electronic health records (EHRs), registries, claims/billing data, and patient-generated data, as well as those from mobile health applications and wearable devices. Real-world data from these sources can be collected and analysed through different study designs such as prospective and retrospective cohort studies, case-control studies, and pragmatic clinical trials. Real-world evidence in the form of post-marketing surveillance has been extensively used to generate pharmacovigilance data. Of late, it has been realised that, apart from safety, RWE has additional applications in different stages of the drug approval cycle, and can be used to optimize the design of randomised controlled trials (RCTs). There has been an increasing awareness and acceptance of RWE from different stakeholders, including physicians, pharmaceutical companies, payers, regulators, and patients. Several regulatory authorities have also created frameworks and guidelines for efficient harnessing of RWE while acknowledging several challenges in RWD collection and analysis. The purpose of this review is to offer an outline of the current information on RWE, its advantages and disadvantages, as well as the associated challenges and ways to overcome them, while also throwing some light on the future of RWE.
Collapse
Affiliation(s)
- Amit Dang
- MarksMan Healthcare Communications, J1309, Amethyst Tower, PBEL City, Peeramcheruvu Village, Rajendra Nagar Mandal, Hyderabad, Telangana, 500091, India.
| |
Collapse
|
69
|
Song B, Nie L, Bozorov K, Kuryazov R, Aisa HA, Zhao J. Parallel synthesis of condensed pyrimidine-thiones and their antitumor activities. RESEARCH ON CHEMICAL INTERMEDIATES 2022. [DOI: 10.1007/s11164-022-04912-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
70
|
Charpignon ML, Vakulenko-Lagun B, Zheng B, Magdamo C, Su B, Evans K, Rodriguez S, Sokolov A, Boswell S, Sheu YH, Somai M, Middleton L, Hyman BT, Betensky RA, Finkelstein SN, Welsch RE, Tzoulaki I, Blacker D, Das S, Albers MW. Causal inference in medical records and complementary systems pharmacology for metformin drug repurposing towards dementia. Nat Commun 2022; 13:7652. [PMID: 36496454 PMCID: PMC9741618 DOI: 10.1038/s41467-022-35157-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 11/21/2022] [Indexed: 12/13/2022] Open
Abstract
Metformin, a diabetes drug with anti-aging cellular responses, has complex actions that may alter dementia onset. Mixed results are emerging from prior observational studies. To address this complexity, we deploy a causal inference approach accounting for the competing risk of death in emulated clinical trials using two distinct electronic health record systems. In intention-to-treat analyses, metformin use associates with lower hazard of all-cause mortality and lower cause-specific hazard of dementia onset, after accounting for prolonged survival, relative to sulfonylureas. In parallel systems pharmacology studies, the expression of two AD-related proteins, APOE and SPP1, was suppressed by pharmacologic concentrations of metformin in differentiated human neural cells, relative to a sulfonylurea. Together, our findings suggest that metformin might reduce the risk of dementia in diabetes patients through mechanisms beyond glycemic control, and that SPP1 is a candidate biomarker for metformin's action in the brain.
Collapse
Affiliation(s)
- Marie-Laure Charpignon
- Institute for Data, Systems, and Society, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Bang Zheng
- Ageing Epidemiology Research Unit, School of Public Health, Imperial College London, London, UK
| | - Colin Magdamo
- Department of Neurology, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - Bowen Su
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Kyle Evans
- Department of Neurology, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Harvard Medical School, Boston, MA, USA
| | - Steve Rodriguez
- Department of Neurology, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Harvard Medical School, Boston, MA, USA
| | - Artem Sokolov
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Harvard Medical School, Boston, MA, USA
| | - Sarah Boswell
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Harvard Medical School, Boston, MA, USA
| | - Yi-Han Sheu
- Department of Psychiatry, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - Melek Somai
- Inception Labs, Collaborative for Health Delivery Sciences, Medical College of Wisconsin, Wauwatosa, WI, USA
| | - Lefkos Middleton
- Ageing Epidemiology Research Unit, School of Public Health, Imperial College London, London, UK
- Public Health Directorate, Imperial College London NHS Healthcare Trust, London, UK
| | - Bradley T Hyman
- Department of Neurology, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - Rebecca A Betensky
- Department of Biostatistics, School of Global Public Health, New York University, New York, NY, USA
| | - Stan N Finkelstein
- Institute for Data, Systems, and Society, Massachusetts Institute of Technology, Cambridge, MA, USA
- Division of Clinical Informatics, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Roy E Welsch
- Institute for Data, Systems, and Society, Massachusetts Institute of Technology, Cambridge, MA, USA
- Sloan School of Management, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ioanna Tzoulaki
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK.
- Dementia Research Institute, Imperial College London, London, UK.
- Department of Hygiene and Epidemiology, University of Ioannina, Ioannina, Greece.
| | - Deborah Blacker
- Department of Psychiatry, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA.
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Sudeshna Das
- Department of Neurology, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA.
| | - Mark W Albers
- Department of Neurology, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA.
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
71
|
Kang J, Cairns J. Exploring uncertainty and use of real-world data in the National Institute for Health and Care Excellence single technology appraisals of targeted cancer therapy. BMC Cancer 2022; 22:1268. [PMID: 36471259 PMCID: PMC9724266 DOI: 10.1186/s12885-022-10350-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 11/22/2022] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES Dealing with uncertainty is one of the critical topics in health technology assessment. The greater decision uncertainty in appraisals, the less clear the clinical- and cost-effectiveness of the health technology. Although the development of targeted cancer therapies (TCTs) has improved patient health care, additional complexity has been introduced in drug appraisals due to targeting more specific populations. Real-world data (RWD) are expected to provide helpful information to fill the evidence gaps in appraisals. This study compared appraisals of TCTs with those of non-targeted cancer therapies (non-TCTs) regarding sources of uncertainty and reviewed how RWD have been used to supplement the information in these appraisals. METHODS This study reviews single technology appraisals (STAs) of oncology medicines performed by the National Institute for Health and Care Excellence (NICE) over 11 years up to December 2021. Three key sources of uncertainty were identified for comparison (generalisability of clinical trials, availability of direct treatment comparison, maturity of survival data in clinical trials). To measure the intensity of use of RWD in appraisals, three components were identified (overall survival, volume of treatment, and choice of comparators). RESULTS TCTs received more recommendations for provision through the Cancer Drugs Fund (27.7, 23.6% for non-TCT), whereas similar proportions were recommended for routine commissioning. With respect to sources of uncertainty, the external validity of clinical trials was greater in TCT appraisals (p = 0.026), whereas mature survival data were available in fewer TCT appraisals (p = 0.027). Both groups showed similar patterns of use of RWD. There was no clear evidence that RWD have been used more intensively in appraisals of TCT. CONCLUSIONS Some differences in uncertainty were found between TCT and non-TCT appraisals. The appraisal of TCT is generally challenging, but these challenges are neither new nor distinctive. The same sources of uncertainty were often found in the non-TCT appraisals. The uncertainty when appraising TCT stems from insufficient data rather than the characteristics of the drugs. Although RWD might be expected to play a more active role in appraisals of TCT, the use of RWD has generally been limited.
Collapse
Affiliation(s)
- Jiyeon Kang
- Department of Health Service Research and Policy, Faculty of Public Health and Policy, London School of Hygiene and Tropical Medicine, 15-17 Tavistock place, London, WC1H 9SH, UK.
- Centre for Cancer Biomarkers (CCBIO), University of Bergen, Bergen, Norway.
| | - John Cairns
- Department of Health Service Research and Policy, Faculty of Public Health and Policy, London School of Hygiene and Tropical Medicine, 15-17 Tavistock place, London, WC1H 9SH, UK
- Centre for Cancer Biomarkers (CCBIO), University of Bergen, Bergen, Norway
| |
Collapse
|
72
|
Naumann-Winter F, Wolter F, Hermes U, Malikova E, Lilienthal N, Meier T, Kalland ME, Magrelli A. Licensing of Orphan Medicinal Products—Use of Real-World Data and Other External Data on Efficacy Aspects in Marketing Authorization Applications Concluded at the European Medicines Agency Between 2019 and 2021. Front Pharmacol 2022; 13:920336. [PMID: 36034814 PMCID: PMC9413272 DOI: 10.3389/fphar.2022.920336] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 06/08/2022] [Indexed: 12/03/2022] Open
Abstract
Background: Reference to so-called real-world data is more often made in marketing authorization applications for medicines intended to diagnose, prevent or treat rare diseases compared to more common diseases. We provide granularity on the type and aim of any external data on efficacy aspects from both real-world data sources and external trial data as discussed in regulatory submissions of orphan designated medicinal products in the EU. By quantifying the contribution of external data according to various regulatory characteristics, we aimed at identifying specific opportunities for external data in the field of orphan conditions. Methods: Information on external data in regulatory documents covering 72 orphan designations was extracted. Our sample comprised public assessment reports for approved, refused, or withdrawn applications concluded from 2019–2021 at the European Medicines Agency. Products with an active orphan designation at the time of submission were scrutinized regarding the role of external data on efficacy aspects in the context of marketing authorization applications, or on the criterion of “significant benefit” for the confirmation of the orphan designation at the time of licensing. The reports allowed a broad distinction between clinical development, regulatory decision making, and intended post-approval data collection. We defined three categories of external data, administrative data, structured clinical data, and external trial data (from clinical trials not sponsored by the applicant), and noted whether external data concerned the therapeutic context of the disease or the product under review. Results: While reference to external data with respect to efficacy aspects was included in 63% of the approved medicinal products in the field of rare diseases, 37% of marketing authorization applications were exclusively based on the dedicated clinical development plan for the product under review. Purely administrative data did not play any role in our sample of reports, but clinical data collected in a structured manner (from routine care or clinical research) were often used to inform on the trial design. Two additional recurrent themes for the use of external data were the contextualization of results, especially to confirm the orphan designation at the time of licensing, and reassurance of a large difference in treatment effect size or consistency of effects observed in clinical trials and practice. External data on the product under review were restricted to either active substances already belonging to the standard of care even before authorization or to compassionate use schemes. Furthermore, external data were considered pivotal for marketing authorization only exceptionally and only for active substances already in use within the specific therapeutic indication. Applications for the rarest conditions and those without authorized treatment alternatives were especially prominent with respect to the use of external data from real-world data sources both in the pre- and post-approval setting. Conclusion: Specific opportunities for external data in the setting of marketing authorizations in the field of rare diseases were identified. Ongoing initiatives of fostering systematic data collection are promising steps for a more efficient medicinal product development in the field of rare diseases.
Collapse
Affiliation(s)
- Frauke Naumann-Winter
- Federal Institute for Drugs and Medical Devices, Bonn, Germany
- *Correspondence: Frauke Naumann-Winter,
| | | | - Ulrike Hermes
- Federal Institute for Drugs and Medical Devices, Bonn, Germany
| | - Eva Malikova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, Bratislava, Slovakia
- State Institute for Drug Control, Bratislava, Slovakia
| | - Nils Lilienthal
- Federal Institute for Drugs and Medical Devices, Bonn, Germany
| | - Tania Meier
- Federal Institute for Drugs and Medical Devices, Bonn, Germany
| | | | - Armando Magrelli
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
73
|
Barrett JS, Nicholas T, Azer K, Corrigan BW. Role of Disease Progression Models in Drug Development. Pharm Res 2022; 39:1803-1815. [PMID: 35411507 PMCID: PMC9000925 DOI: 10.1007/s11095-022-03257-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 04/05/2022] [Indexed: 12/11/2022]
Abstract
The use of Disease progression models (DPMs) in Drug Development has been widely adopted across therapeutic areas as a method for integrating previously obtained disease knowledge to elucidate the impact of novel therapeutics or vaccines on disease course, thus quantifying the potential clinical benefit at different stages of drug development programs. This paper provides a brief overview of DPMs and the evolution in data types, analytic methods, and applications that have occurred in their use by Quantitive Clinical Pharmacologists. It also provides examples of how these models have informed decisions and clinical trial design across several therapeutic areas and at various stages of development. It briefly describes potential new applications of DPMs utilizing emerging data sources, and utilizing new analytic techniques, and discuss new challenges faced such as requiring description of multiple endpoints, rapid model development, application of machine learning-based analytics, and use of high dimensional and real-world data. Considerations for the continued evolution future of DPMs to serve as community-maintained expert systems are also provided.
Collapse
Affiliation(s)
- Jeffrey S. Barrett
- Rare Disease Cures Accelerator Data Analytics Platform, Critical Path Institute, Tuscon, AZ 85718 USA
| | - Tim Nicholas
- Global Product Development, Pfizer Inc, 445 Eastern Point Rd, Groton, CT 06340 USA
| | - Karim Azer
- Axcella Therapeutics, 840 Memorial Drive, Cambridge, MA 02139 USA
| | - Brian W. Corrigan
- Global Product Development, Pfizer Inc, 445 Eastern Point Rd, Groton, CT 06340 USA
| |
Collapse
|
74
|
Clay I, Cormack F, Fedor S, Foschini L, Gentile G, van Hoof C, Kumar P, Lipsmeier F, Sano A, Smarr B, Vandendriessche B, De Luca V. Measuring Health-Related Quality of Life With Multimodal Data: Viewpoint. J Med Internet Res 2022; 24:e35951. [PMID: 35617003 PMCID: PMC9185357 DOI: 10.2196/35951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/14/2022] [Accepted: 04/25/2022] [Indexed: 11/18/2022] Open
Abstract
The ability to objectively measure aspects of performance and behavior is a fundamental pillar of digital health, enabling digital wellness products, decentralized trial concepts, evidence generation, digital therapeutics, and more. Emerging multimodal technologies capable of measuring several modalities simultaneously and efforts to integrate inputs across several sources are further expanding the limits of what digital measures can assess. Experts from the field of digital health were convened as part of a multi-stakeholder workshop to examine the progress of multimodal digital measures in two key areas: detection of disease and the measurement of meaningful aspects of health relevant to the quality of life. Here we present a meeting report, summarizing key discussion points, relevant literature, and finally a vision for the immediate future, including how multimodal measures can provide value to stakeholders across drug development and care delivery, as well as three key areas where headway will need to be made if we are to continue to build on the encouraging progress so far: collaboration and data sharing, removal of barriers to data integration, and alignment around robust modular evaluation of new measurement capabilities.
Collapse
Affiliation(s)
- Ieuan Clay
- Digital Medicine Society, Boston, MA, United States
| | | | | | | | | | | | | | | | - Akane Sano
- Department of Electrical and Computer Engineering, Rice University, Houston, TX, United States
| | - Benjamin Smarr
- Department of Bioengineering and Halicioglu Data Science Institute, University of California, San Diego, San Diego, CA, United States
| | | | - Valeria De Luca
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| |
Collapse
|