1
|
Vaitsiakhovich T, Hartenstein A, Privitera S, Patel MR, Piccini JP, Coleman CI, Abdelgawwad K, Holberg G, Khorlo I, Mundl H, Schaefer B, Viethen T, Vogtländer K, Vowinkel A, Kleinjung F. An External Control Arm for the Oral Factor XIa Inhibitor Asundexian Phase 2 Trial in Atrial Fibrillation (PACIFIC-AF) Using Electronic Health Records. Cardiol Ther 2025:10.1007/s40119-025-00411-x. [PMID: 40377841 DOI: 10.1007/s40119-025-00411-x] [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: 02/06/2025] [Accepted: 04/08/2025] [Indexed: 05/18/2025] Open
Abstract
INTRODUCTION The aim of this study was to assess the applicability of an external control arm (ECA) approach in the clinical development of the oral factor XIa inhibitor asundexian for stroke prevention in patients with atrial fibrillation (AF), using prospectively collected data from the phase 2 PACIFIC-AF trial (NCT04218266) and real-world individual-level data from patients with AF treated with apixaban in the Optum® de-identified Electronic Health Record data set (Optum® EHR) 2013-2019. METHODS To build ECAs, real-world patients meeting trial eligibility criteria were matched to patients enrolled in PACIFIC-AF. The primary outcome was the composite of International Society on Thrombosis and Haemostasis-defined major bleeding or clinically relevant non-major bleeding. Event rates were compared between PACIFIC-AF and ECAs at 85 days of trial duration and projected up to 2 years. RESULTS Overall, 160,153 real-world patients met PACIFIC-AF eligibility criteria and were matched to patients from the PACIFIC-AF apixaban arm on 101 variables, with matching ratios of 1:10, 1:5, and 1:1. At day 85, the number of events for the primary outcome was 92 (3.68%) in the 1:10 ECA (2500 patients) and 6 (2.40%) in the PACIFIC-AF apixaban arm (250 patients), with incidence rates of 16.67 (90% confidence interval [CI] 13.92-19.63) and 11.10 (90% CI 4.83-19.45) per 100 person-years, respectively. CONCLUSIONS ECAs matching the PACIFIC-AF apixaban arm could be built from EHRs with concordant event rates for key trial endpoints. The ECA approach enabled the determination of event rates for treatment duration up to 2 years, thereby informing the asundexian pivotal phase 3 trial in AF.
Collapse
Affiliation(s)
| | | | | | - Manesh R Patel
- Duke University School of Medicine, Durham, NC, 27710, USA
| | | | - Craig I Coleman
- University of Connecticut School of Pharmacy, Storrs, CT, 06269, USA
| | | | | | - Igor Khorlo
- Global Commercialization, Bayer AG, Berlin, Germany
| | - Hardi Mundl
- Research and Development, Bayer AG, Wuppertal, Germany
| | | | | | | | | | - Frank Kleinjung
- Medical Affairs and Pharmacovigilance, Bayer AG, Berlin, Germany
| |
Collapse
|
2
|
Sarri G, Rizzo M, Upadhyaya S, Paly VF, Hernandez L. Navigating the unknown: how to best 'reflect' standard of care in indications without a dedicated treatment pathway in health technology assessment submissions. J Comp Eff Res 2024; 13:e230145. [PMID: 38226913 PMCID: PMC10842305 DOI: 10.57264/cer-2023-0145] [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: 09/26/2023] [Accepted: 12/12/2023] [Indexed: 01/17/2024] Open
Abstract
There is an urgent need for expedited approval and access for new health technologies targeting rare and very rare diseases, some of which are associated with high unmet treatment needs. Once a new technology achieves regulatory approval, the technology needs to be assessed by health technology assessment (HTA) bodies to inform coverage and reimbursement decisions. This assessment quantitatively examines the clinical effectiveness, safety and/or economic impact of the new technology relative to standard of care (SoC) in a specific market. However, in rare and very rare diseases, the patient populations are small and there is often no established treatment pathway available to define 'SoC'. In these situations, several challenges arise to assess the added benefit of a new technology - both clinically and economically - due to lack of established SoC to guide an appropriate comparator selection. These challenges include: How should 'SoC' be defined and characterized in HTA submissions for new technologies aiming to establish new treatment standards? What is usual care without an established clinical pathway? How should the evidence for the comparator 'SoC' (i.e., usual care) arm be collected in situations with low patient representation and, sometimes, limited disease-specific clinical knowledge in certain geographies? This commentary outlines the evidence generation challenges in designing clinical comparative effectiveness for a new technology when there is a lack of established SoC. The commentary also proposes considerations to facilitate the reliable integration of real-world evidence into HTA and decision-making based on the collective experience of the authors.
Collapse
Affiliation(s)
| | | | | | | | - Luis Hernandez
- Takeda Pharmaceuticals America, Inc., Lexington, MA, USA
| |
Collapse
|
3
|
Jandhyala R. Development of a definition for real-world evidence using the Jandhyala method for observing consensus opinion among medical affairs pharmaceutical physicians. Curr Med Res Opin 2023; 39:1551-1558. [PMID: 36710630 DOI: 10.1080/03007995.2023.2172261] [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: 11/30/2022] [Accepted: 01/20/2023] [Indexed: 01/31/2023]
Abstract
OBJECTIVE Low rates of inclusion of real-world evidence (RWE) during regulation may arise from lack of clarity and consensus on its definition. A conceptually mature definition of RWE may have pragmatic utility, increasing its inclusion during regulation. The aim was to develop a definition of RWE to promote inclusion in regulatory submissions and assess its conceptual maturity. METHODS Thirteen medical affairs pharmaceutical physicians completed two qualitative online surveys to generate items needed in a definition of RWE. Items that reached a consensus index of > 50% (CI > = 0.51) were retained in the final definition. The maturity of the definition was assessed using concept analysis. RESULTS After attrition, 11 participants completed the study and generated 18 items to be included in a definition of RWE. All items reached the consensus threshold and were included. The definition was conceptually mature on three of the four dimensions: the potential for a consensual definition across stakeholders, a description of its characteristics and clear preconditions and outcomes. Further research is needed to delineate the boundaries of RWE. CONCLUSIONS A definition of RWE was generated that may increase its inclusion during medicines regulation, especially with further refinement from regulators and other stakeholders.
Collapse
Affiliation(s)
- Ravi Jandhyala
- Medialis Ltd, Wolverton Mill, Milton Keynes, England, UK
- Centre for Pharmaceutical Medicine Research, King's College University, London, UK
| |
Collapse
|
4
|
Acha V, Barefoot B, Juarez Garcia A, Lehner V, Monno R, Sandler S, Spooner A, Verpillat P. Principles for Good Practice in the Conduct of Non-interventional Studies: The View of Industry Researchers. Ther Innov Regul Sci 2023; 57:1199-1208. [PMID: 37460826 PMCID: PMC10579109 DOI: 10.1007/s43441-023-00544-y] [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: 08/05/2022] [Accepted: 06/02/2023] [Indexed: 10/17/2023]
Abstract
This reflection paper presents a consolidated view of EFPIA on the need for principles for good practice in the generation and use of non-interventional studies (NIS), including overarching principles such as the registration of hypothesis evaluating treatment effect (HETE) studies. We first define NIS and the important adjacencies to clinical trials and relationship with real-world evidence (RWE). We then outline the principles for good practice with respect to appropriate research design, study protocol, fit-for-purpose variables and data quality, analytical methods, bias reduction, transparency in conduct and use, privacy management and ethics review. We conclude with recommendations for action for the research community to promote trust and credibility in the use of NIS.
Collapse
|
5
|
Argotti U, Leyens L, Lisbona C, López P, Alonso-Orgaz S, Nevado A, Cozzi V. Comparison of the Latin America Regulation Landscape and International Reference Health Authorities to Hasten Drug Registration and Clinical Research Applications. Ther Innov Regul Sci 2023; 57:1287-1297. [PMID: 37682461 PMCID: PMC10579156 DOI: 10.1007/s43441-023-00565-7] [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: 01/26/2023] [Accepted: 07/28/2023] [Indexed: 09/09/2023]
Abstract
INTRODUCTION Promptly providing new drugs to fulfill unmet medical needs requires changes in drug development and registration processes. Health Authorities (HAs) considered as reference due to their experience and acknowledgement (Food and Drug Administration [FDA] among others) already consider innovative clinical trial (CT) designs and flexible approval procedures, but Latin America (LATAM) regulations are still far. A comparison was performed to identify gaps. MATERIALS AND METHODS CT requirements for drug Marketing Authorization Application (MAA) and CT approval regulations were compared between LATAM and reference HAs (FDA/European Medicines Agency [EMA]/Health-Canada/Swissmedic/Therapeutic Goods Administration [TGA]/Pharmaceuticals and Medical Devices Agency [PMDA]), as of August 2022. Procedure included reference HAs regulations review, item selection, identification in LATAM regulations, and International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH) guidelines (ICH-E6[R2]/ICH-E8[R1]) implementation revision. RESULTS For MAA, specific application requirements or ICH guideline M4(R4) on common technical document (CTD) adoption are generally stated, and phase-I/III performance is mandatory (explicitly/implicitly). Faster patient access procedures are infrequent: Priority-drug programs, conditional authorizations, or expedited procedures are scarce or non-existent. Regulatory reliance procedures are adopted through different pathways. Regarding CT approval, innovative/complex CT designs are not prohibited but usually omitted. Some countries implemented adapted CT conducting during the COVID-19 pandemic. Early scientific advice meetings (HA-sponsor) are occasionally considered. Most countries are not formally ICH-joined. CONCLUSIONS LATAM regulations must adapt to new regulatory standards (FDA/EMA/ICH) through implementation of frequent updates, reliance/expedited procedures, early HA-sponsor interactions, innovative/complex CTs, mandatory phase-III reaching elimination, and decentralized elements for CT conducting.
Collapse
Affiliation(s)
- Urimara Argotti
- International Regulatory Policy Department, Latin America Productos Roche, S.A. de C.V., Mexico City, Mexico
| | - Lada Leyens
- Product Development Regulatory, F. Hoffmann-La Roche AG, Basel, Switzerland
| | | | - Pilar López
- Medical Writing Department, LIDESEC S.L, Madrid, Spain
| | | | - Angel Nevado
- Medical Writing Department, LIDESEC S.L, Madrid, Spain
| | - Virginia Cozzi
- Medical Affairs Department, Roche Central America, Venezuela, and the Caribbean, Heredia, Costa Rica.
| |
Collapse
|
6
|
khalidi-idrissi A, Madinzi A, Anouzla A, Pala A, Mouhir L, Kadmi Y, Souabi S. Recent advances in the biological treatment of wastewater rich in emerging pollutants produced by pharmaceutical industrial discharges. INTERNATIONAL JOURNAL OF ENVIRONMENTAL SCIENCE AND TECHNOLOGY : IJEST 2023; 20:1-22. [PMID: 37360558 PMCID: PMC10019435 DOI: 10.1007/s13762-023-04867-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 12/19/2022] [Accepted: 02/22/2023] [Indexed: 06/28/2023]
Abstract
Pharmaceuticals and personal care products present potential risks to human health and the environment. In particular, wastewater treatment plants often detect emerging pollutants that disrupt biological treatment. The activated sludge process is a traditional biological method with a lower capital cost and limited operating requirements than more advanced treatment methods. In addition, the membrane bioreactor combines a membrane module and a bioreactor, widely used as an advanced method for treating pharmaceutical wastewater with good pollution performance. Indeed, the fouling of the membrane remains a major problem in this process. In addition, anaerobic membrane bioreactors can treat complex pharmaceutical waste while recovering energy and producing nutrient-rich wastewater for irrigation. Wastewater characterizations have shown that wastewater's high organic matter content facilitates the selection of low-cost, low-nutrient, low-surface-area, and effective anaerobic methods for drug degradation and reduces pollution. However, to improve the biological treatment, researchers have turned to hybrid processes in which all physical, chemical, and biological treatment methods are integrated to remove various emerging contaminants effectively. Hybrid systems can generate bioenergy, which helps reduce the operating costs of the pharmaceutical waste treatment system. To find the most effective treatment technique for our research, this work lists the different biological treatment techniques cited in the literature, such as activated sludge, membrane bioreactor, anaerobic treatment, and hybrid treatment, combining physicochemical and biological techniques.
Collapse
Affiliation(s)
- A. khalidi-idrissi
- Laboratory of Process Engineering and Environment, Faculty of Science and Technology, Mohammedia, University Hassan II of Casablanca, BP. 146, Mohammedia, Morocco
| | - A. Madinzi
- Laboratory of Process Engineering and Environment, Faculty of Science and Technology, Mohammedia, University Hassan II of Casablanca, BP. 146, Mohammedia, Morocco
| | - A. Anouzla
- Laboratory of Process Engineering and Environment, Faculty of Science and Technology, Mohammedia, University Hassan II of Casablanca, BP. 146, Mohammedia, Morocco
| | - A. Pala
- Environmental Research and Development Center (CEVMER), Dokuz Eylul University, Izmir, Turkey
| | - L. Mouhir
- Laboratory of Process Engineering and Environment, Faculty of Science and Technology, Mohammedia, University Hassan II of Casablanca, BP. 146, Mohammedia, Morocco
| | - Y. Kadmi
- CNRS, UMR 8516 - LASIR, University Lille, 59000 Lille, France
| | - S. Souabi
- Laboratory of Process Engineering and Environment, Faculty of Science and Technology, Mohammedia, University Hassan II of Casablanca, BP. 146, Mohammedia, Morocco
| |
Collapse
|
7
|
Heikkinen I, Eskola S, Acha V, Morrison A, Walker C, Weil C, Bril A, Wegner M, Metcalfe T, Chibout SD, Chlebus M. Role of innovation in pharmaceutical regulation: A proposal for principles to evaluate EU General Pharmaceutical Legislation from the innovator perspective. Drug Discov Today 2023; 28:103526. [PMID: 36792005 DOI: 10.1016/j.drudis.2023.103526] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 01/29/2023] [Accepted: 02/08/2023] [Indexed: 02/15/2023]
Abstract
Because the EU General Pharmaceutical Legislation is under review, the EFPIA Innovation Board developed evaluation principles for the policy proposals and key considerations on how the regulatory framework can support innovation while ensuring only safe, efficacious and quality medicines are authorized. The evaluation principles are anchored on actions to promote: agile adoption of new methodologies with soft law tools; continued emphasis on regulatory science to inform policies; a cost/benefit assessment of the new regulation to ensure they have an overall positive impact; and mitigation of any negative externalities or unintended effects for any type of innovation or products. The evaluation principles are intended to guide the impact assessment of the pharmaceutical legislation in the EU but the principles can be applied globally.
Collapse
Affiliation(s)
| | - Sini Eskola
- Director Regulatory Strategy, EFPIA, Brussels, Belgium
| | - Virginia Acha
- Associate Vice President, Global Regulatory Policy, MSD, UK
| | - Alan Morrison
- Vice President, Regulatory Affairs International, MSD, UK
| | - Chris Walker
- Vice President, Head of Regulatory Affairs International, Amgen, UK
| | - Catherine Weil
- Head of Global Regulatory Sciences, Europe & Canada, BMS, Switzerland
| | - Antoine Bril
- Scientific Director Public Affairs, Servier, France
| | - Max Wegner
- Senior Vice President, Global Head of Regulatory Affairs, Bayer, Germany
| | - Thomas Metcalfe
- Personalised Healthcare Ecosystems, Roche Pharma, Switzerland
| | - Salah-Dine Chibout
- Global Head External Partnerships & TA Head Oncology Preclinical Safety, Novartis, Switzerland
| | - Magda Chlebus
- Executive Director, Science Policy and Regulatory Affairs, EFPIA, Belgium
| |
Collapse
|
8
|
Pharmacokinetic and Pharmacodynamic Modeling of siRNA Therapeutics - a Minireview. Pharm Res 2022; 39:1749-1759. [PMID: 35819583 DOI: 10.1007/s11095-022-03333-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 07/04/2022] [Indexed: 10/17/2022]
Abstract
The approval of four small interfering RNA (siRNA) products in the past few years has demonstrated unequivocally the therapeutic potential of this novel modality. Three such products (givosiran, lumasiran and inclisiran) are liver-targeted, using tris N-acetylgalactosamine (GalNAc)3 as the targeting ligand. Upon subcutaneous administration, GalNAc-conjugated siRNAs rapidly distribute into the liver via asialoglycoprotein receptor (ASGPR) mediated uptake in the hepatocytes, resulting in fast elimination from the systemic circulation. Patisiran, on the other hand, has been formulated in a lipid nanoparticle for optimal delivery to the liver. While several publications have described preclinical and clinical pharmacokinetic (PK) and pharmacodynamic (PD) results, including absorption, distribution, metabolism, and elimination (ADME) profiles in selected species as well as limited modeling efforts for siRNA therapeutics, there is no systematic review of the PK and PD models developed for these agents or work summarizing the utility and application(s) of such models in drug development and regulatory review. Here, we provide a mini-review of the current state of modeling efforts for siRNA therapeutics within the early preclinical, translational, and clinical stages of siRNA development. Diverse modeling methods including simple compartmental, mechanistic and systems PK/PD, physiologically-based PK (PBPK), population PK/PD, and dose-response-time models are introduced and reviewed. The utility of such models in development and regulatory review for siRNA therapeutics is also discussed with examples. Finally, the current knowledge gaps in mechanism of action of siRNA and resulting challenges in model development are summarized. The goal of this minireview is to trigger cross-functional discussion amongst all key stakeholders to generate key experimental datasets and align on current assumptions, model structures, and approaches to facilitate development and application of robust PK/PD models for siRNA therapeutics.
Collapse
|
9
|
Zong N, Wen A, Moon S, Fu S, Wang L, Zhao Y, Yu Y, Huang M, Wang Y, Zheng G, Mielke MM, Cerhan JR, Liu H. Computational drug repurposing based on electronic health records: a scoping review. NPJ Digit Med 2022; 5:77. [PMID: 35701544 PMCID: PMC9198008 DOI: 10.1038/s41746-022-00617-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 05/19/2022] [Indexed: 11/30/2022] Open
Abstract
Computational drug repurposing methods adapt Artificial intelligence (AI) algorithms for the discovery of new applications of approved or investigational drugs. Among the heterogeneous datasets, electronic health records (EHRs) datasets provide rich longitudinal and pathophysiological data that facilitate the generation and validation of drug repurposing. Here, we present an appraisal of recently published research on computational drug repurposing utilizing the EHR. Thirty-three research articles, retrieved from Embase, Medline, Scopus, and Web of Science between January 2000 and January 2022, were included in the final review. Four themes, (1) publication venue, (2) data types and sources, (3) method for data processing and prediction, and (4) targeted disease, validation, and released tools were presented. The review summarized the contribution of EHR used in drug repurposing as well as revealed that the utilization is hindered by the validation, accessibility, and understanding of EHRs. These findings can support researchers in the utilization of medical data resources and the development of computational methods for drug repurposing.
Collapse
Affiliation(s)
- Nansu Zong
- Department of Artificial Intelligence and Informatics Research, Mayo Clinic, Rochester, MN, USA.
| | - Andrew Wen
- Department of Artificial Intelligence and Informatics Research, Mayo Clinic, Rochester, MN, USA
| | - Sungrim Moon
- Department of Artificial Intelligence and Informatics Research, Mayo Clinic, Rochester, MN, USA
| | - Sunyang Fu
- Department of Artificial Intelligence and Informatics Research, Mayo Clinic, Rochester, MN, USA
| | - Liwei Wang
- Department of Artificial Intelligence and Informatics Research, Mayo Clinic, Rochester, MN, USA
| | - Yiqing Zhao
- Department of Preventive Medicine, Northwestern Medicine, Northwestern University, Chicago, IL, USA
| | - Yue Yu
- Department of Artificial Intelligence and Informatics Research, Mayo Clinic, Rochester, MN, USA
| | - Ming Huang
- Department of Artificial Intelligence and Informatics Research, Mayo Clinic, Rochester, MN, USA
| | - Yanshan Wang
- Department of Health Information Management, School of Health and Rehabilitation Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Gang Zheng
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | | | - James R Cerhan
- Department of Artificial Intelligence and Informatics Research, Mayo Clinic, Rochester, MN, USA
| | - Hongfang Liu
- Department of Artificial Intelligence and Informatics Research, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
10
|
Jandhyala R. The effect of adding real-world evidence to regulatory submissions on the breadth of population indicated for rare disease medicine treatment by the European Medicines Agency. J Pharm Policy Pract 2022; 15:36. [PMID: 35509059 PMCID: PMC9069778 DOI: 10.1186/s40545-022-00433-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 04/23/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Despite calls for the use of additional real-world evidence (RWE) during drug development, rates of inclusion at the regulatory stage remain low. The medicine adoption model suggests that providing additional RWE to regulators would result in a wider indicated population than providing randomised-controlled trial evidence (RCTE) alone. Here, we tested this hypothesis. METHODS All engagements concerning the 88 orphan drugs approved between 2009 and 2019 on the European Medicines Agency Orphan Register were reviewed between September and December 2019. Engagements were grouped as containing either randomised-controlled trial evidence (RCTE) or RCTE with real-world evidence (RWE). The data on indicatable population (the therapeutic indication requested by an engagement) and indicated population (the therapeutic indication ultimately granted) as well as the median number of criteria limiting the indicated population in each study type (RCTE/RWE) was extracted. A chi-square test assessed the association between the indicated population (as a proportion of the indicatable population) and type of evidence (RCTE with or without RWE) and a Wilcoxon rank sum test assessed the difference between the median number of limiting criteria between RCTE and RWE studies. Prediction modelling extrapolated the results of a power analysis to a level expected to deliver significance and the time this would take. RESULTS The review identified 103 engagements, of which three were excluded (one contained only RWE; two contained only systematic literature reviews), leaving 100 engagements for 87 orphan medicines in the final analysis. Only 13% of engagements contained RWE. Although the difference was statistically insignificant, 76.92% of engagements containing RCTE and RWE resulted in a broader indicated population as compared to only 56.32% of those that contained RCTE alone. The median number of limiting criteria from RCTE (37 (28, 43)) and RWE (5 (2, 9)) studies varied significantly (p = 0.005). Modelling suggested that the analysis would achieve sufficient power by 2033-37 at the current RWE adoption rate. CONCLUSION The proportion of the disease population studied in RWE was greater than that in RCTE. The analysis testing the relationship between additional RWE and broader indicated population would achieve adequate power between 2032 and 2037 at the current RWE adoption rate.
Collapse
Affiliation(s)
- Ravi Jandhyala
- Medialis Ltd, 13 Horse Fair, Banbury, OX16 0AH, UK. .,Centre for Pharmaceutical Medicine Research, King's College University, London, UK.
| |
Collapse
|
11
|
Eskola SM, Leufkens HGM, Bate A, De Bruin ML, Gardarsdottir H. Use of Real-World Data and Evidence in Drug Development of Medicinal Products Centrally Authorized in Europe in 2018-2019. Clin Pharmacol Ther 2021; 111:310-320. [PMID: 34689334 PMCID: PMC9299055 DOI: 10.1002/cpt.2462] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 10/08/2021] [Indexed: 11/05/2022]
Abstract
Real‐world data/real‐world evidence (RWD/RWE) are considered to have a great potential to complement, in some cases, replace the evidence generated through randomized controlled trials. By tradition, use of RWD/RWE in the postauthorization phase is well‐known, whereas published evidence of use in the pre‐authorization phase of medicines development is lacking. The primary aim of this study was to identify and quantify the role of potential use of RWD/RWE (RWE signatures) during the pre‐authorization phase, as presented in the initial marketing authorization applications of new medicines centrally evaluated with a positive opinion in 2018–2019 (n = 111) by the European Medicines Agency (EMA). Data for the study was retrieved from the evaluation overviews of the European Public Assessment Reports (EPARs), which reflect the scientific conclusions of the assessment process and are accessible through the EMA website. RWE signatures were extracted into an RWE Data Matrix, including 11 categories divided over 5 stages of the drug development lifecycle. Nearly all EPARs included RWE signatures for the discovery (98.2%) and life‐cycle management (100.0%). Half of them included RWE signatures for the full development phase (48.6%) and for supporting regulatory decisions at the registration (46.8%), whereas over a third (35.1%) included RWE signatures for the early development. RWE signatures were more often seen for orphan and conditionally approved medicines. Oncology, hematology, and anti‐infectives stood out as therapeutic areas with most RWE signatures in their full development phase. The findings bring unprecedented insights about the vast use of RWD/RWE in drug development supporting the regulatory decision making.
Collapse
Affiliation(s)
- Sini Marika Eskola
- Utrecht Centre for Pharmaceutical Policy and Regulation, Division of Pharmacoepidemiology and Clinical Pharmacology, Faculty of Science, Utrecht University, Utrecht, The Netherlands.,European Federation of Pharmaceutical Industries and Associations, Brussels, Belgium
| | - Hubertus Gerardus Maria Leufkens
- Utrecht Centre for Pharmaceutical Policy and Regulation, Division of Pharmacoepidemiology and Clinical Pharmacology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Andrew Bate
- Global Safety, GSK, Brentford, Middlesex, UK.,Department of Non-Communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK.,Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Marie Louise De Bruin
- Utrecht Centre for Pharmaceutical Policy and Regulation, Division of Pharmacoepidemiology and Clinical Pharmacology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Helga Gardarsdottir
- Utrecht Centre for Pharmaceutical Policy and Regulation, Division of Pharmacoepidemiology and Clinical Pharmacology, Faculty of Science, Utrecht University, Utrecht, The Netherlands.,Department of Clinical Pharmacy, University Medical Center, Utrecht, The Netherlands.,Department of Pharmaceutical Sciences, School of Health Sciences, University of Iceland, Reykjavík, Iceland
| |
Collapse
|
12
|
Venkatakrishnan K, van der Graaf PH. Model-Informed Drug Development: Connecting the Dots With a Totality of Evidence Mindset to Advance Therapeutics. Clin Pharmacol Ther 2021; 110:1147-1154. [PMID: 34658027 DOI: 10.1002/cpt.2422] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 09/09/2021] [Indexed: 01/01/2023]
Affiliation(s)
- Karthik Venkatakrishnan
- EMD Serono Research & Development Inc., Billerica, MA, USA.,A Business of Merck KGaA, Darmstadt, Germany
| | | |
Collapse
|