1
|
Carlsson LM, Peltonen M, Jacobson P, Andersson-Assarsson JC, Svensson PA, Taube M, Karlsson C, Ahlin S, Kristensson FM, Perkins R, Arnetorp I, Carlsson A, Admeus L, Langegård E, Carlsson B, Sjöholm K. Possible selection bias in register-based obesity studies. Eur J Epidemiol 2025:10.1007/s10654-025-01237-6. [PMID: 40353980 DOI: 10.1007/s10654-025-01237-6] [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: 07/09/2024] [Accepted: 04/22/2025] [Indexed: 05/14/2025]
Abstract
Some studies of obesity treatments use control groups identified from real-world registers, which may differ from people with obesity in the general population. We evaluated whether such control groups affect the results. The SOS study examines long-term mortality post-bariatric surgery. Among volunteers with obesity, 2,007 individuals underwent surgery, while a control group of 2,040 individuals was matched using 18 variables. Age was 37-60 years and BMI was ≥ 34/≥38 kg/m2 for men and women, respectively. We subdivided the control group into those with an obesity diagnosis (n = 177) and those without an obesity diagnosis (n = 1,863) in the Swedish National Patient Register prior to study inclusion. Mortality was determined over a median follow-up period of 26 years. The controls with a prior obesity diagnosis had a higher mortality rate than the controls without a prior obesity diagnosis, with 19.7 (95% CI, 15.5-25.1) and 14.4 (95% CI, 13.3-15.7) deaths per 1000 person-years, respectively. This corresponds to a hazard ratio of 1.45 (95% CI, 1.12-1.89; p = 0.005) and a 3.4-year shorter life expectancy. These results were confirmed in another cohort (n = 2,759, HR = 1.82 [95% CI, 1.47-2.25; p<0.001] and a 6.1-year shorter life expectancy). Controls with obesity identified from real-world datasets may be in poorer health than those who voluntarily participate in clinical studies. Consequently, selection bias could lead to an overestimation of the survival benefits of obesity treatments in research using controls identified by prior obesity diagnosis.
Collapse
Affiliation(s)
- Lena Ms Carlsson
- Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | | - Peter Jacobson
- Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | | - Per-Arne Svensson
- Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Institute of Health and Care Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Magdalena Taube
- Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Cecilia Karlsson
- Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Late-Stage Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Sofie Ahlin
- Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Physiology, Region Västra Götaland, NU hospital group, Trollhättan, Sweden
| | - Felipe M Kristensson
- Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Dept of Surgery, Region Västra Götaland, Sahlgrenska University Hospital/Östra, Gothenburg, Sweden
| | - Rosie Perkins
- Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ida Arnetorp
- Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Alexander Carlsson
- Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lucas Admeus
- Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Elin Langegård
- Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Björn Carlsson
- Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- BioPharmaceuticals R&D, AstraZeneca, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), Gothenburg, Sweden
| | - Kajsa Sjöholm
- Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
2
|
Vidalis A, Dumoulin O, Godbole M, Proenca CC. The role and value of real-world evidence in health technology decision-making in France, Germany, Italy, Spain, and the UK: insights on external control arms. Int J Technol Assess Health Care 2025; 41:e25. [PMID: 40260460 PMCID: PMC12019763 DOI: 10.1017/s0266462324004720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 04/23/2025]
Abstract
Real-world evidence (RWE) is increasingly used and accepted by health technology assessment (HTA) bodies as supportive evidence to inform the approval of new technologies. However, the criteria driving RWE acceptance are often unclear.This study aims to improve understanding of the role and value of RWE in HTA decision-making and outline the best practices in building real-world external control arms (ECAs).A mixed approach of a targeted literature review and HTA expert interviews was applied. The HTA reports of ten selected technologies and the expert interviews from France, Germany, Italy, Spain, and the UK informed the criteria driving the acceptance of RWE. Overall, the UK and Spanish HTA bodies are more receptive to accepting RWE, whereas the French and German are the least accepting. When RWE is used to substantiate efficacy claims, the level of scrutiny from regulators and HTA bodies is considerably higher than when RWE has different intended uses. Representativeness of the data source, overall transparency in the study and robust methodologies are the key criteria driving RWE acceptance across markets.
Collapse
|
3
|
Koong AJ, Irvin VL, Narayan A, Song S, Kaplan RM. Evidence available and used by the Food and Drug Administration for the approval of orphan and nonorphan drugs. HEALTH AFFAIRS SCHOLAR 2025; 3:qxaf057. [PMID: 40190699 PMCID: PMC11970248 DOI: 10.1093/haschl/qxaf057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/25/2025] [Accepted: 03/17/2025] [Indexed: 04/09/2025]
Abstract
There are substantial financial incentives to develop orphan drugs for rare diseases, but concerns about the quality and volume of supporting evidence have emerged. We compare evidence used to evaluate orphan and nonorphan drugs approved by the Food and Drug Administration (FDA) between 2016 and 2023. This retrospective cross-sectional analysis utilizes FDA data on approvals and study information from ClinicalTrials.gov to compare characteristics of studies relevant to orphan and nonorphan drugs approved between 2016 and 2023. Of the 368 total drugs approved, 50% were orphan drugs. The FDA-approved drugs based on significantly fewer studies for orphan (1.5 studies/drug) compared to nonorphan (2.4 studies/drug). Additionally, a significantly lower proportion of studies were completed before FDA approval for orphan drugs (25% vs 41%). Orphan drugs were significantly less likely to be evaluated in randomized clinical trials (RCTs) (34% vs 63%). Of these RCTs, there were significantly fewer completed before approval (40% vs 54%) and that had results posted (35% vs 53%). There was a significant difference in the available evidence for orphan and nonorphan drugs. As new legislation like Cures 2.0 is developed, it is critical to examine the balance between an expedited approval timeline and the standard of clinical evidence.
Collapse
Affiliation(s)
- Amanda J Koong
- Department of Health Policy and Management, Fielding School of Public Health at the University of California at Los Angeles, Los Angeles 90095, USA
- McGovern Medical School at UTHealth Houston, Houston 77030, USA
| | - Veronica L Irvin
- College of Health, Oregon State University, Corvallis 97331, USA
| | - Aditya Narayan
- Stanford University Schools of Medicine and Business, Stanford 94305, USA
| | - Sujin Song
- Clinical Excellence Research Center, Stanford University School of Medicine, Stanford 94304, USA
| | - Robert M Kaplan
- Clinical Excellence Research Center, Stanford University School of Medicine, Stanford 94304, USA
| |
Collapse
|
4
|
NajafZadeh M, Fernández Oromendia A, Burcu M, Mcconnochie B, Kim E, Vaccaro T, Patorno E. Linkage of Clinical Trial Data to Routinely Collected Data Sources: A Scoping Review. JAMA Netw Open 2025; 8:e257797. [PMID: 40299382 PMCID: PMC12042059 DOI: 10.1001/jamanetworkopen.2025.7797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 02/27/2025] [Indexed: 04/30/2025] Open
Abstract
Importance Patients who participate in clinical trials generate valuable routinely collected data (eg, medical records, electronic health records, claims databases, disease registries, or vital statistics) through their routine interactions with the health care system before, during, and after the trial. When this routinely collected data is linked at the participant level, it can supplement active data collection in the trial and provide deeper insights into the benefits, risks, and costs of treatments. Objective To review clinical trials linked to routinely collected data in various countries, identifying the use cases and designs of these linkage studies. Evidence Review Research articles that reported the linkage of clinical trials to medical records, electronic health records, claims databases, disease registries, or vital statistics in their title or abstract were searched for in PubMed and MEDLINE. The search covered the period from January 1, 2016, through December 30, 2023. Opinion pieces, study protocols, or studies that involved interventions other than medications, dietary supplements, vaccines, devices, procedures, or diagnostics (eg, behavioral interventions) were excluded. Study eligibility and data extraction were performed independently by 2 reviewers to ensure the accuracy of findings. Findings Of the 990 abstracts initially screened, a full text review was conducted for 147 articles. In total, 71 studies were included in the results, including 42 medication, vaccine, and dietary supplement trials (59.2%) and 29 device, procedure, or diagnostic trials (40.8%). Of these 71 studies, 24 (32.4%) were conducted in the US. In 32 studies (45.1%), consent for linkage was obtained prospectively as part of the main trial, while 33 studies (46.5%) received a waiver of authorization from the respective ethical review boards. The most frequent use cases of linkage to were posttrial long-term follow-up (22 studies [31.0%]), capturing primary or secondary outcomes of trials (19 studies [26.8%]), validation of routinely collected data outcomes (17 studies [23.9%]), and measuring health care resource utilization and cost in trials (12 studies [16.9%]). Conclusions and Relevance This study found that the linkage of patients' clinical trial data to routinely collected data has been implemented in several trials for various use cases and that most studies obtained consent for linkage prospectively as part of the main trial or received a waiver of authorization from ethical review boards. These findings demonstrate the feasibility and provide an overview of the use cases for linking trials to routinely collected data.
Collapse
Affiliation(s)
- Mehdi NajafZadeh
- Medidata Solutions, A Dassault System Company, Boston, MABoston, Massachusetts
| | | | | | - Ben Mcconnochie
- Medidata Solutions, A Dassault System Company, Boston, MABoston, Massachusetts
| | - Ella Kim
- Medidata Solutions, A Dassault System Company, Boston, MABoston, Massachusetts
| | - Thomas Vaccaro
- Medidata Solutions, A Dassault System Company, Boston, MABoston, Massachusetts
- Now with: Datavant, Phoenix, Arizona
| | - Elisabetta Patorno
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
5
|
Antonini M, Mattar A, Pereira da Costa Pinheiro DJ, Maia IB, Teixeira MD, Amorim AG, Ferraro O, Chrispim de Oliveira L, Ramos MDNM, Cavalcante FP, Zerwes F, Madeira M, Barroso-Sousa R, de Camargo Millen E, Frasson AL, Brenelli FP, Facina G, Fenile R, Gebrim LH, Real JM. Disparities in access to anti-HER2 therapies in neoadjuvant chemotherapy: A prognostic analysis based on real-world data comparing Brazil's public and private healthcare systems. Breast 2025; 80:104417. [PMID: 39983435 PMCID: PMC11893340 DOI: 10.1016/j.breast.2025.104417] [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: 11/18/2024] [Revised: 01/08/2025] [Accepted: 02/06/2025] [Indexed: 02/23/2025] Open
Abstract
BACKGROUND Trastuzumab has significantly improved the treatment of HER2-positive breast cancer, particularly in the neoadjuvant setting, where its combination with chemotherapy increases the pathologic complete response (pCR) rate. This retrospective cohort study assesses the implications of disparities in access to trastuzumab within the Brazilian public healthcare system, focusing on pCR, overall survival (OS) and disease-free survival (DFS) in non-metastatic, HER2-positive breast cancer patients undergoing neoadjuvant chemotherapy (NAC). METHODS The study was conducted in the Hospital Pérola Byington (PEROLA), a public institution, and in the Hospital do Servidor Público Estadual (HSPE), a private institution. pCR was defined as the absence of residual invasive or in situ tumors in the breast and axillary nodes. OS and DFS were calculated by Kaplan-Meier survival analysis for a 5-year period. RESULTS From 2011 to 2020, 381 patients at PEROLA and 78 at HSPE underwent NAC. Trastuzumab availability was higher at HSPE (83.4 % vs. 60.0 %, p < 0.0001). Use of trastuzumab correlated with significantly higher pCR rates at both the PEROLA (54.3 % vs. 26.4 %, p < 0.0001) and the HSPE (52.7 % vs. 26.4 %, p < 0.0001). HER2-positive patients with pCR at HSPE also had better OS (80 % vs. 61 %, p < 0.0001) and DFS (89 % vs. 67 %, p < 0.0001) compared to those at PEROLA. CONCLUSION There were significant differences in the provision of trastuzumab between the public and private healthcare systems, adversely affecting clinical outcomes and patient survival. The current data highlight the pressing need to address equity in cancer treatment to improve prognosis for every patient.
Collapse
Affiliation(s)
- Marcelo Antonini
- Department of Breast Surgery, Hospital do Servidor Público Estadual Francisco Morato de Oliveira, São Paulo, SP, Brazil; Centro de Desenvolvimento de Ensino e Pesquisa do Instituo de Assistência Médica ao Servidor Público Estadual (CEDEP - IAMSPE), São Paulo, SP, Brazil; BBREAST - Brazilian Breast Cancer Association Team, São Paulo, SP, Brazil.
| | - André Mattar
- BBREAST - Brazilian Breast Cancer Association Team, São Paulo, SP, Brazil; Department of Breast Surgery, Women's Health Hospital, São Paulo, SP, Brazil; Breast Surgeon at Oncoclínicas, São Paulo, SP, Brazil.
| | | | - Isabela Bastos Maia
- Department of Breast Surgery, Hospital do Servidor Público Estadual Francisco Morato de Oliveira, São Paulo, SP, Brazil.
| | | | | | - Odair Ferraro
- Department of Breast Surgery, Hospital do Servidor Público Estadual Francisco Morato de Oliveira, São Paulo, SP, Brazil.
| | | | | | - Francisco Pimentel Cavalcante
- BBREAST - Brazilian Breast Cancer Association Team, São Paulo, SP, Brazil; Department of Breast Surgery, Hospital Geral de Fortaleza Fortaleza, CE, Brazil.
| | - Felipe Zerwes
- BBREAST - Brazilian Breast Cancer Association Team, São Paulo, SP, Brazil; Department of Breast Surgery, Medical School of the Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil.
| | - Marcelo Madeira
- Department of Breast Surgery, Faculdade Israelita de Ciências da Saúde Albert Einstein, São Paulo, SP, Brazil.
| | | | - Eduardo de Camargo Millen
- BBREAST - Brazilian Breast Cancer Association Team, São Paulo, SP, Brazil; Breast Surgeon at Americas Oncologia, Rio de Janeiro, RJ, Brazil.
| | - Antonio Luiz Frasson
- BBREAST - Brazilian Breast Cancer Association Team, São Paulo, SP, Brazil; Breast Surgeon at Hospital Albert Einstein, São Paulo, SP, Brazil.
| | - Fabricio Palermo Brenelli
- BBREAST - Brazilian Breast Cancer Association Team, São Paulo, SP, Brazil; Department of Breast Surgery, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil.
| | - Gil Facina
- Department of Ginecology, Universidade Federal de São Paulo, São Paulo, SP, Brazil.
| | - Rogério Fenile
- Department of Mastology of Hospital Ipiranda, São Paulo, SP, Brazil.
| | | | - Juliana Monte Real
- Department of Breast Surgery, Hospital do Servidor Público Estadual Francisco Morato de Oliveira, São Paulo, SP, Brazil; Centro de Desenvolvimento de Ensino e Pesquisa do Instituo de Assistência Médica ao Servidor Público Estadual (CEDEP - IAMSPE), São Paulo, SP, Brazil.
| |
Collapse
|
6
|
El Kababji S, Mitsakakis N, Jonker E, Beltran-Bless AA, Pond G, Vandermeer L, Radhakrishnan D, Mosquera L, Paterson A, Shepherd L, Chen B, Barlow W, Gralow J, Savard MF, Fesl C, Hlauschek D, Balic M, Rinnerthaler G, Greil R, Gnant M, Clemons M, El Emam K. Augmenting Insufficiently Accruing Oncology Clinical Trials Using Generative Models: Validation Study. J Med Internet Res 2025; 27:e66821. [PMID: 40053790 PMCID: PMC11923467 DOI: 10.2196/66821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 12/27/2024] [Accepted: 01/31/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Insufficient patient accrual is a major challenge in clinical trials and can result in underpowered studies, as well as exposing study participants to toxicity and additional costs, with limited scientific benefit. Real-world data can provide external controls, but insufficient accrual affects all arms of a study, not just controls. Studies that used generative models to simulate more patients were limited in the accrual scenarios considered, replicability criteria, number of generative models, and number of clinical trials evaluated. OBJECTIVE This study aimed to perform a comprehensive evaluation on the extent generative models can be used to simulate additional patients to compensate for insufficient accrual in clinical trials. METHODS We performed a retrospective analysis using 10 datasets from 9 fully accrued, completed, and published cancer trials. For each trial, we removed the latest recruited patients (from 10% to 50%), trained a generative model on the remaining patients, and simulated additional patients to replace the removed ones using the generative model to augment the available data. We then replicated the published analysis on this augmented dataset to determine if the findings remained the same. Four different generative models were evaluated: sequential synthesis with decision trees, Bayesian network, generative adversarial network, and a variational autoencoder. These generative models were compared to sampling with replacement (ie, bootstrap) as a simple alternative. Replication of the published analyses used 4 metrics: decision agreement, estimate agreement, standardized difference, and CI overlap. RESULTS Sequential synthesis performed well on the 4 replication metrics for the removal of up to 40% of the last recruited patients (decision agreement: 88% to 100% across datasets, estimate agreement: 100%, cannot reject standardized difference null hypothesis: 100%, and CI overlap: 0.8-0.92). Sampling with replacement was the next most effective approach, with decision agreement varying from 78% to 89% across all datasets. There was no evidence of a monotonic relationship in the estimated effect size with recruitment order across these studies. This suggests that patients recruited earlier in a trial were not systematically different than those recruited later, at least partially explaining why generative models trained on early data can effectively simulate patients recruited later in a trial. The fidelity of the generated data relative to the training data on the Hellinger distance was high in all cases. CONCLUSIONS For an oncology study with insufficient accrual with as few as 60% of target recruitment, sequential synthesis can enable the simulation of the full dataset had the study continued accruing patients and can be an alternative to drawing conclusions from an underpowered study. These results provide evidence demonstrating the potential for generative models to rescue poorly accruing clinical trials, but additional studies are needed to confirm these findings and to generalize them for other diseases.
Collapse
Affiliation(s)
- Samer El Kababji
- School of Epidemiology and Public Health, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- CHEO Research Institute, Ottawa, ON, Canada
| | | | | | | | - Gregory Pond
- Department of Oncology, McMaster University, Hamilton, ON, Canada
| | - Lisa Vandermeer
- Division of Medical Oncology, University of Ottawa, Ottawa, ON, Canada
| | | | - Lucy Mosquera
- Generate Ops & Data Science, Aetion, Ottawa, ON, Canada
| | | | - Lois Shepherd
- Public Health Sciences, Queens University, Kingston, ON, Canada
| | - Bingshu Chen
- Public Health Sciences, Queens University, Kingston, ON, Canada
| | - William Barlow
- Biostatistics, University of Washington, Seattle, WA, United States
| | - Julie Gralow
- Medical Oncology, University of Washington, Seattle, WA, United States
| | | | - Christian Fesl
- Clinical Statistics, Austrian Breast & Colorectal Cancer Study Group (ABCSG), Vienna, Austria
| | - Dominik Hlauschek
- Clinical Statistics, Austrian Breast & Colorectal Cancer Study Group (ABCSG), Vienna, Austria
| | - Marija Balic
- Division of Clinical Oncology, Medical University Graz, Graz, Austria
| | | | - Richard Greil
- Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Michael Gnant
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Mark Clemons
- Division of Medical Oncology, University of Ottawa, Ottawa, ON, Canada
| | - Khaled El Emam
- School of Epidemiology and Public Health, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- CHEO Research Institute, Ottawa, ON, Canada
| |
Collapse
|
7
|
Johnsen SP, Baas P, Sørensen JB, Chouaid C, Griesinger F, Daumont MJ, Rault C, Emanuel G, Penrod JR, Jacobs H, Muwaffak M, Schoemaker MJ, Munro REJ, Baskaran D, Durand-Zaleski I, O’Donnell JC. Advancing real-world research in thoracic malignancies: learnings from the international I-O Optimise initiative. Future Oncol 2025; 21:867-878. [PMID: 39996596 PMCID: PMC11916385 DOI: 10.1080/14796694.2025.2466416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 02/10/2025] [Indexed: 02/26/2025] Open
Abstract
In recent years, the thoracic malignancies treatment landscape has become more complex with the emergence of novel targeted and immunotherapy-based treatments. Although beneficial to patients and physicians, this fast-paced therapeutic evolution has increased the complexity of clinical decision-making and amplified the importance of real-world evidence to support data from randomized controlled trials. The international I-O Optimise initiative was established in 2016 to provide real-world insights into the thoracic malignancies treatment landscape, and has since collaborated with 14 data sources across Europe and Canada, allowing access to data from ~ 500,000 patients with non-small-cell lung cancer, small-cell lung cancer, and malignant pleural mesothelioma. This article reviews pertinent I-O Optimise research, with discussion of the methodological/data-related learnings and expectations for future insights.
Collapse
Affiliation(s)
- Søren Paaske Johnsen
- Danish Center for Health Services Research, Department of Clinical Medicine, Aalborg University and Aalborg University Hospital, Gistrup, Denmark
| | - Paul Baas
- Department of Thoracic Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Pulmonary Disease, Leiden University Medical Hospital, Leiden, The Netherlands
| | | | - Christos Chouaid
- Pneumology Unit, Centre Hospitalier Intercommunal de Créteil, Créteil, France
| | - Frank Griesinger
- Department of Haematology & Oncology, University Department Internal Medicine-Oncology, Pius-Hospital, University Medicine Oldenburg, Oldenburg, Germany
| | - Melinda J Daumont
- Worldwide Health Economics & Outcomes Research, Bristol Myers Squibb, Braine-L’Alleud, Belgium
| | | | - Gabrielle Emanuel
- Real-World Data Analytics Markets, Bristol Myers Squibb, Uxbridge, UK
| | - John R Penrod
- Worldwide Health Economics & Outcomes Research, Bristol Myers Squibb, Princeton, NJ, USA
| | | | | | | | | | | | - Isabelle Durand-Zaleski
- AP-HP Health Economics Research Unit, Hôtel-Dieu Hospital, INSERM UMR 1153 CRESS, UPEC, Paris, France
| | - John C O’Donnell
- Worldwide Health Economics & Outcomes Research, Bristol Myers Squibb, Princeton, NJ, USA
| |
Collapse
|
8
|
Hu H, Wang H, Ang L, Shi M, Wu X, Zhang C, Han M, Liu S, Li K, Zhang J, Ji Z. Patient-Focused Drug Development and Real World Study. Integr Med Res 2025; 14:101119. [PMID: 39935619 PMCID: PMC11810706 DOI: 10.1016/j.imr.2024.101119] [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: 04/16/2024] [Revised: 12/23/2024] [Accepted: 12/26/2024] [Indexed: 02/13/2025] Open
Abstract
Background Patient-focused drug development (PFDD) is an important direction in the field of medical research and is of great significance to the development of medicine. In recent years, PFDD and real-world study (RWS) have gained much interest, of which both have their advantages. This study aims to promote research methods innovation and optimize clinical research design and implementation. Methods After a brief introduction of PFDD and RWS, this review focused on the comparison of clinical trials of PFDD and RWS in terms of objectives, Population, Intervention, Comparator, Outcome (PICO) elements, research team members, data acquisition, and research key points, and clarified the feasibility and significance of "patient-focused RWS" research model. Results PFDD emphasized that patients' needs as well as the willingness and satisfaction of patients should be considered throughout the whole research process and the patient experience data should be collected during the study development and drug use. RWS emphasized the facticity of research implementation environment and the breadth of patient sources, which concerned the problem of the extrapolation of study results, the clinical localization, and patient applicability of the target drug. However, there is a connection between both of the above. Both clinical trials of PFDD and RWS bring benefits to patients. Conclusions Combining PFDD idea and RWS research method to carry out new research will maximize the benefits for patients. The study model combining the PFDD concept with RWS can facilitate drug development and dissemination, which can be popularized and applied in various research areas. This study can innovate research methods and provide new ideas for future research.
Collapse
Affiliation(s)
- Haiyin Hu
- Evidence Base Medicine Center, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, PR China
| | - Hui Wang
- Evidence Base Medicine Center, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Lin Ang
- KM Science Research Division, Korea Institute of Oriental Medicine, Daejeon, Korea
| | - Menglong Shi
- Evidence Base Medicine Center, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Xiaolei Wu
- Evidence Base Medicine Center, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Chenyao Zhang
- Evidence Base Medicine Center, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Mei Han
- Evidence Base Medicine Center, Beijing University of Traditional Chinese Medicine, Beijing, PR China
| | - Shigang Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, PR China
| | - Kai Li
- Shanxi Hospital of Integrated Traditional Chinese and Western Medicine (Shanxi Provincial Key Laboratory of classical prescription strengthening yang), Shanxi, PR China
| | - Junhua Zhang
- Evidence Base Medicine Center, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Zhaochen Ji
- Evidence Base Medicine Center, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
- School of Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| |
Collapse
|
9
|
Farnsworth MG, Khanipov K, Botnar K, Weaver SC, Barrett ADT, Golovko G. Real-world evidence of yellow Fever vaccination data-driven study. Vaccine 2025; 48:126758. [PMID: 39848130 DOI: 10.1016/j.vaccine.2025.126758] [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: 11/01/2024] [Revised: 12/16/2024] [Accepted: 01/16/2025] [Indexed: 01/25/2025]
Abstract
Yellow Fever (YF), a mosquito-borne viral disease caused by yellow fever virus (YFV), remains endemic in tropical Sub-Saharan Africa and South America. The 17D live-attenuated vaccine has significantly reduced YF incidence with minimal risk of vaccine-associated adverse events, including Yellow Fever Vaccine-fever-associated Neurotropic Disease (YEL-AND) and Yellow Fever Vaccine-Associated Viscerotropic Disease (YEL-AVD). This study investigates the potential of Real-World Evidence (RWE) to enhance vaccine surveillance by analyzing electronic health records (EHRs) from the TriNetX platform, which identified a total of 15,835 individuals who were vaccinated with the Stamaril® YF vaccine between 2017 and 2021 in the United States. We compared adverse event rates obtained from RWE with those reported by the manufacturer in a recent study of Stamaril® used in the United States during this period. Our findings were consistent with those published previously and suggest no significant increase in adverse medical outcomes post-vaccination across all age groups, particularly in long-term analysis. This proof-of-concept study underscores the value of RWE in monitoring vaccine safety and supports its potential to complement traditional surveillance methods, offering a robust tool for continuous post-marketing vaccine evaluation.
Collapse
Affiliation(s)
- Madison G Farnsworth
- Department of Human Pathophysiology and Translational Medicine, Institute for Translational Science, University of Texas Medical Branch, Galveston, TX, United States.
| | - Kamil Khanipov
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, United States
| | - Kostiantyn Botnar
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, United States
| | - Scott C Weaver
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, United States
| | - Alan D T Barrett
- Sealy Institute for Vaccine Sciences and Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - George Golovko
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
10
|
Plooij J, Delnoij DMJ. Patient-relevance of outcome measures in breast cancer clinical trials: a cross-sectional comparative analysis of patient preferences and trials conducted between 2014 and 2024. Expert Rev Pharmacoecon Outcomes Res 2025:1-6. [PMID: 39960492 DOI: 10.1080/14737167.2025.2467379] [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: 09/20/2024] [Revised: 01/22/2025] [Accepted: 01/27/2025] [Indexed: 02/21/2025]
Abstract
BACKGROUND Worldwide, many clinical trials are performed using clinical outcomes and surrogate outcomes as endpoints. Surrogate outcomes are used, for instance, if there is not enough follow-up time to measure the outcome of interest. Surrogate outcomes might not be patient-relevant, however. This study assesses to what extent patient-relevant outcomes are measured in clinical trials for breast cancer drugs. RESEARCH DESIGN AND METHODS A cross-sectional comparative analysis was conducted in which patient preferences for outcomes derived from the literature were compared to outcomes measured in phase III breast cancer trials conducted between 2014 and 2024. RESULTS Patients prefer outcomes addressing survival benefits, treatment effectiveness, adverse events and health-related quality of life. Minor improvements in survival benefits are greatly valued. The majority of patients are willing to accept some side effects for a positive outcome. The primary outcome used most frequently in trials is progression-free survival. The most common secondary outcomes are adverse events, mortality, overall response rate, and health-related quality of life. CONCLUSION Phase III trial outcomes appear to align largely with breast cancer patients' preferences. Nevertheless, patients and trial designers emphasize different outcomes. Improvement is therefore needed to enhance the relevance of trial data for patients.
Collapse
Affiliation(s)
- Jasmijn Plooij
- Erasmus School of Health Policy & Management, Erasmus University Rotterdam, Rotterdam, The Netherlands
| | - Diana M J Delnoij
- Erasmus School of Health Policy & Management, Erasmus University Rotterdam, Rotterdam, The Netherlands
- National Health Care Institute, Diemen, The Netherlands
| |
Collapse
|
11
|
Costa V, Custodio MG, Gefen E, Fregni F. The relevance of the real-world evidence in research, clinical, and regulatory decision making. Front Public Health 2025; 13:1512429. [PMID: 40041193 PMCID: PMC11878099 DOI: 10.3389/fpubh.2025.1512429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 01/21/2025] [Indexed: 03/06/2025] Open
Abstract
The scientific method has been established as the optimal approach for systematically gathering and interpreting data on various human phenomena, mainly through the adoption of strict experimental methods, such as controlled randomized trials, which is relevant for clinical decision-making and research, but also weights the regulatory processes for approval of drugs and other medical products. However, a key factor in strict methods is the generalizability of findings that may be limited to specific settings and patient characteristics. This limitation can be addressed by non-experimental methods aimed at investigating populations in naturalistic routine clinical settings, which may offer a more representative reflection of the usage, effectiveness, and safety of healthcare interventions. These approaches can generate the so-called real-world data and the resulting real-world evidence. In this narrative review, we present these concepts, explore the potential applications and advantages of real-world evidence for clinical, research, and regulatory decision-making, and discuss the challenges of employing it, the solutions to improve its generation, and lastly the current level of evidence required for the integration of real-world evidence into regulatory decision-making. We concluded that the advantages of using it, when utilized in a balanced manner, overcome the challenges and, therefore, can offer a time- and cost-saving solution for researchers, the healthcare industry, regulatory agencies, policymakers, and payers towards the patient benefit. The knowledge generated by this approach provides valuable additional insights into medical interventions in real patients under realistic daily scenarios.
Collapse
Affiliation(s)
- Valton Costa
- Neuromodulation Center and Center for Clinical Research Learning, Spaulding Rehabilitation Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Laboratory of Neuroscience and Neurological Rehabilitation, Physical Therapy Department, Federal University of Sao Carlos, São Carlos, Brazil
| | - Marcelo Graziano Custodio
- Global Innovation and Development, Established Pharmaceuticals Division, Abbott Products Operations AG, Allschwil, Switzerland
| | - Eran Gefen
- Global Innovation and Development, Established Pharmaceuticals Division, Abbott Products Operations AG, Allschwil, Switzerland
| | - Felipe Fregni
- Neuromodulation Center and Center for Clinical Research Learning, Spaulding Rehabilitation Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
12
|
Bodner E, Roth L, Wiencke K, Bischoff C, Schwarz PE. Effect of Multimodal App-Based Interventions on Glycemic Control in Patients With Type 2 Diabetes: Systematic Review and Meta-Analysis. J Med Internet Res 2025; 27:e54324. [PMID: 39854703 PMCID: PMC11806272 DOI: 10.2196/54324] [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: 11/06/2023] [Revised: 05/17/2024] [Accepted: 10/30/2024] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND Digital technologies for type 2 diabetes mellitus (T2DM) care hold great potential to improve patients' health in the long term. Only a subset of telemedicine offerings are digital interventions that meet the criteria for prescribable digitale Gesundheitsanwendung (digital health apps; DiGAs) in Germany. Digital treatments further provide vast amounts of patient data that are important to generate evidence. OBJECTIVE This systematic review aims to analyze the efficacy of multimodal digital therapies that mainly meet the DiGA criteria for patients with T2DM and to elicit the potential of such therapies. This includes evidence from randomized controlled trials (RCTs) as well as from real-world data. The outcome of interest was a reduction in glycated hemoglobin (hemoglobin A1c [HbA1c]; long-term blood glucose measurements). METHODS A systematic literature search was conducted in the literature bases PubMed, LIVIVO, and Cochrane, based on the predefined PICO (Population; Intervention; Control; Outcome) scheme. Identified studies were assessed for risk of bias, pragmatism, and overall quality of evidence. Meta-analyses were conducted for between group differences using RCTs only, and for within-group differences using RCTs and non-RCTs, to examine the effect of the interventions on HbA1c. RESULTS In total, 795 records were identified, of which 24 were eligible for this systematic review and 23 studies were eligible for the meta-analysis. The results of the meta-analyses showed significant and clinically relevant reductions in HbA1c in patients with T2DM. Regarding the between-group difference for HbA1c reduction, the pooled effect of the RCTs showed a reduction of -0.36% (95% CI -0.59% to -0.14%; P<.001), favoring app-based interventions. The average mean within-group reduction in HbA1c was -0.79 (95% CI -1.02 to -0.55), with no significant difference between RCTs (-0.69, 95% CI -1.13 to -0.24) and non-RCTs (-0.87, 95% CI -1.16 to -0.57; P<.01, differences between RCTs and RCTs P=.44). A pragmatism rating showed that both study types were on average (very) pragmatic, that is, close to usual care. However, the overall quality of evidence was low to very low. CONCLUSIONS This systematic review shows that digital therapies that mainly meet the DiGA criteria can effectively improve HbA1c in patients with T2DM. The integration of digital health care into usual care holds great potential and should be considered as a complementary option to usual care in the future. TRIAL REGISTRATION PROSPERO CRD42023440203; https://www.crd.york.ac.uk/prospero/display_record.php?RecordID=440203.
Collapse
Affiliation(s)
- Emma Bodner
- Sidekick Health Germany GmbH, Hamburg, Germany
| | - Lena Roth
- Department for Prevention and Care of Diabetes, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Kathleen Wiencke
- Sidekick Health Germany GmbH, Hamburg, Germany
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | | | - Peter Eh Schwarz
- Department for Prevention and Care of Diabetes, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
13
|
Maihöfner C, Mallick-Searle T, Vollert J, Kalita P, Sood Sethi V. Review of Challenges in Performing Real-World Evidence Studies for Nonprescription Products. Pragmat Obs Res 2025; 16:7-18. [PMID: 39873007 PMCID: PMC11771160 DOI: 10.2147/por.s504709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 01/07/2025] [Indexed: 01/30/2025] Open
Abstract
In recent years, regulatory authorities have signaled a willingness to consider real-world evidence (RWE) data to support applications for new claims and indications for pharmaceuticals. Historically, RWE studies have been the domain of prescription drugs, driven by the fact that clinical data on patients are routinely captured in medical records, claims databases, registries, etc. However, RWE reports of nonprescription drugs and supplements are relatively sparse due to methodological gaps in this area. The objective of this narrative review is to identify which RWE methodologies have been used to study nonprescription products. A total of 49 articles were included based on literature searches. Label comprehension studies, used to support prescription-to-nonprescription switches, are useful in determining how nonprescription products will be used; however, they provide no actual clinical data. The most common RWE studies of nonprescription products were cross-sectional surveys, which investigated a broad range of indications and were conducted in an array of settings, including online, by phone, point-of-sale (pharmacy), outpatient clinics, and shopping malls. However, while this type of study is effective for identifying use patterns and attitudes in the general population, recall bias limits the ability to collect safety and effectiveness data. Studies of electronic medical records and claims databases are hampered by incomplete or absent capturing of data on nonprescription products. As a result, most RWE studies to date have provided limited useful information. Although case reports and expert opinion should not be discounted, in the absence of other information they provide few actual data. Novel approaches using smartphone apps and artificial intelligence may provide new opportunities to collect RWE for nonprescription products, but these areas of research are in their infancy. Overall, there is a need to develop standards for execution of RWE studies of nonprescription products in terms of endpoints, study design, and study quality.
Collapse
Affiliation(s)
- Christian Maihöfner
- Department of Neurology, General Fürth Hospital, University of Erlangen, Fürth, Germany
| | - Theresa Mallick-Searle
- Division of Pain Medicine, Stanford Health Care Pain Management Clinic, Palo Alto, CA, USA
| | - Jan Vollert
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Pranab Kalita
- Global Category Medical Affairs, Haleon Plc, Weybridge, England, UK
| | - Vidhu Sood Sethi
- Global Medical Affairs, GSK Consumer Healthcare Singapore Pte. Ltd, Singapore
| |
Collapse
|
14
|
Whalen J, Chandra A, Kracker S, Ehl S, Seidel MG, Gulas I, Dron L, Velummailum R, Nagamuthu C, Liu S, Tutein Nolthenius J, Maccari ME. Comparative efficacy of leniolisib (CDZ173) versus standard of care on rates of respiratory tract infection and serum immunoglobulin M (IgM) levels among individuals with activated phosphoinositide 3-kinase delta (PI3Kδ) syndrome (APDS): an externally controlled study. Clin Exp Immunol 2025; 219:uxae107. [PMID: 39673396 PMCID: PMC11754865 DOI: 10.1093/cei/uxae107] [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: 07/30/2024] [Revised: 10/22/2024] [Accepted: 11/29/2024] [Indexed: 12/16/2024] Open
Abstract
Leniolisib, an oral, targeted phosphoinositide 3-kinase delta (PI3Kδ) inhibitor, was well-tolerated and efficacious versus placebo in treating individuals with activated PI3Kδ syndrome (APDS), an ultra-rare inborn error of immunity (IEI), in a 12-week randomised controlled trial. However, longer-term comparative data versus standard of care are lacking. This externally controlled study compared the long-term effects of leniolisib on annual rate of respiratory tract infections and change in serum immunoglobulin M (IgM) levels versus current standard of care, using data from the leniolisib single-arm open-label extension study 2201E1 (NCT02859727) and the European Society for Immunodeficiencies (ESID) registry. The endpoints were chosen following feasibility assessment considering comparability and availability of data from both sources. Baseline characteristics between groups were balanced through inverse probability of treatment weighting. The leniolisib-treated group included 37 participants, with 62 and 49 participants in the control group for the respiratory tract infections and serum IgM analyses, respectively. Significant reductions in the annual rate of respiratory tract infections (rate ratio: 0.34; 95% confidence interval [CI]: 0.19, 0.59) and serum IgM levels (treatment effect: -1.09 g/L; 95% CI: -1.78, -0.39, P = 0.002) were observed in leniolisib-treated individuals versus standard of care. The results were consistent across all sensitivity analyses, regardless of censoring, baseline infection rate definition, missing data handling, or covariate selection. These novel data provide an extended comparison of leniolisib treatment versus standard of care, highlighting the potential for leniolisib to deliver long-term benefits by restoring immune system function and reducing infection rate, potentially reducing complications and treatment burden.
Collapse
Affiliation(s)
- John Whalen
- Pharming Group N.V., Leiden, The Netherlands
| | - Anita Chandra
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Sven Kracker
- Université Paris Cité, Imagine Institute, Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, F-7015, Paris, France
| | - Stephan Ehl
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Markus G Seidel
- Division of Pediatric Hematology Oncology, Department of Pediatrics and Adolescent Medicine, Medical University of Graz, Austria
| | | | | | | | | | | | | | - Maria Elena Maccari
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
15
|
Inayama Y, Yamaguchi K, Mizuno K, Tanaka-Mizuno S, Koike A, Higashiyama N, Taki M, Yamanoi K, Murakami R, Hamanishi J, Yoshida S, Mandai M, Kawakami K. Changes in Physical Activity Across Cancer Diagnosis and Treatment Based on Smartphone Step Count Data Linked to a Japanese Claims Database: Retrospective Cohort Study. JMIR Cancer 2025; 11:e58093. [PMID: 39726139 PMCID: PMC11791449 DOI: 10.2196/58093] [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: 03/05/2024] [Revised: 10/24/2024] [Accepted: 12/16/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Although physical activity (PA) is recommended for patients with cancer, changes in PA across cancer diagnosis and treatment have not been objectively evaluated. OBJECTIVE This study aimed to assess the impact of cancer diagnosis and treatment on PA levels. METHODS This was a retrospective cohort study using a Japanese claims database provided by DeSC Healthcare Inc, in which daily step count data, derived from smartphone pedometers, are linked to the claims data. In this study, we included patients newly diagnosed with cancer, along with those newly diagnosed with diabetes mellitus for reference. We collected data between April 2014 and September 2021 and analyzed them. The observation period spanned from 6 months before diagnosis to 12 months after diagnosis. We applied a generalized additive mixed model with a cubic spline to describe changes in step counts before and after diagnosis. RESULTS We analyzed the step count data of 326 patients with malignant solid tumors and 1388 patients with diabetes. Patients with cancer exhibited a 9.6% (95% CI 7.1%-12.1%; P<.001) reduction in step counts from baseline at the start of the diagnosis month, which further deepened to 12.4% (95% CI 9.5%-15.2%; P<.001) at 3 months and persisted at 7.1% (95% CI 4.2%-10.0%; P<.001) at 12 months, all relative to baseline. Conversely, in patients with diabetes, step counts remained relatively stable after diagnosis, with a slight upward trend, resulting in a change of +0.6% (95% CI -0.6% to 1.9%; P=.31) from baseline at 3 months after diagnosis. At 12 months after diagnosis, step counts remained decreased in the nonendoscopic subdiaphragmatic surgery group, with an 18.0% (95% CI 9.1%-26.2%; P<.001) reduction, whereas step counts returned to baseline in the laparoscopic surgery group (+0.3%, 95% CI -6.3% to 7.5%; P=.93). CONCLUSIONS The analysis of objective pre- and postdiagnostic step count data provided fundamental information crucial for understanding changes in PA among patients with cancer. While cancer diagnosis and treatment reduced PA, the decline may have already started before diagnosis. The study findings may help tailor exercise recommendations based on lifelog data for patients with cancer in the future.
Collapse
Affiliation(s)
- Yoshihide Inayama
- Department of Gynecology and Obstetrics, Graduate School of Medicine and Faculty of Medicine, Kyoto University, Kyoto, Japan
- Department of Pharmacoepidemiology, Graduate School of Medicine and Public Health, Kyoto University, Kyoto, Japan
| | - Ken Yamaguchi
- Department of Gynecology and Obstetrics, Graduate School of Medicine and Faculty of Medicine, Kyoto University, Kyoto, Japan
| | - Kayoko Mizuno
- Department of Pharmacoepidemiology, Graduate School of Medicine and Public Health, Kyoto University, Kyoto, Japan
- Department of Digital Health and Epidemiology, Graduate School of Medicine and Public Health, Kyoto University, Kyoto, Japan
| | - Sachiko Tanaka-Mizuno
- Department of Pharmacoepidemiology, Graduate School of Medicine and Public Health, Kyoto University, Kyoto, Japan
- Department of Digital Health and Epidemiology, Graduate School of Medicine and Public Health, Kyoto University, Kyoto, Japan
- Laboratory of Epidemiology and Prevention, Kobe Pharmaceutical University, Kobe, Japan
| | - Ayami Koike
- Department of Gynecology and Obstetrics, Graduate School of Medicine and Faculty of Medicine, Kyoto University, Kyoto, Japan
| | - Nozomi Higashiyama
- Department of Gynecology and Obstetrics, Graduate School of Medicine and Faculty of Medicine, Kyoto University, Kyoto, Japan
| | - Mana Taki
- Department of Gynecology and Obstetrics, Graduate School of Medicine and Faculty of Medicine, Kyoto University, Kyoto, Japan
| | - Koji Yamanoi
- Department of Gynecology and Obstetrics, Graduate School of Medicine and Faculty of Medicine, Kyoto University, Kyoto, Japan
| | - Ryusuke Murakami
- Department of Gynecology and Obstetrics, Graduate School of Medicine and Faculty of Medicine, Kyoto University, Kyoto, Japan
| | - Junzo Hamanishi
- Department of Gynecology and Obstetrics, Graduate School of Medicine and Faculty of Medicine, Kyoto University, Kyoto, Japan
| | - Satomi Yoshida
- Department of Pharmacoepidemiology, Graduate School of Medicine and Public Health, Kyoto University, Kyoto, Japan
- Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Masaki Mandai
- Department of Gynecology and Obstetrics, Graduate School of Medicine and Faculty of Medicine, Kyoto University, Kyoto, Japan
| | - Koji Kawakami
- Department of Pharmacoepidemiology, Graduate School of Medicine and Public Health, Kyoto University, Kyoto, Japan
| |
Collapse
|
16
|
Tibble H, Alyami RA, Bush A, Cunningham S, Julious S, Price D, Quint JK, Turner S, Wang K, Wilson A, Davies GA, Mukherjee M, Chan AHY, Varghese D, Jackson T, Morgan N, Daines L, Pinnock H. Using routine primary care data in research: (in)efficient case studies and perspectives from the Asthma UK Centre for Applied Research. BMJ Health Care Inform 2025; 32:e101134. [PMID: 39788753 PMCID: PMC11751789 DOI: 10.1136/bmjhci-2024-101134] [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: 05/16/2024] [Accepted: 11/22/2024] [Indexed: 01/12/2025] Open
Abstract
AIM We aimed to identify enablers and barriers of using primary care routine data for healthcare research, to formulate recommendations for improving efficiency in knowledge discovery. BACKGROUND Data recorded routinely in primary care can be used for estimating the impact of interventions provided within routine care for all people who are clinically eligible. Despite official promotion of 'efficient trial designs', anecdotally researchers in the Asthma UK Centre for Applied Research (AUKCAR) have encountered multiple barriers to accessing and using routine data. METHODS Using studies within the AUKCAR portfolio as exemplars, we captured limitations, barriers, successes, and strengths through correspondence and discussions with the principal investigators and project managers of the case studies. RESULTS We identified 14 studies (8 trials, 2 developmental studies and 4 observational studies). Investigators agreed that using routine primary care data potentially offered a convenient collection of data for effectiveness outcomes, health economic assessment and process evaluation in one data extraction. However, this advantage was overshadowed by time-consuming processes that were major barriers to conducting efficient research. Common themes were multiple layers of information governance approvals in addition to the ethics and local governance approvals required by all health service research; lack of standardisation so that local approvals required diverse paperwork and reached conflicting conclusions as to whether a study should be approved. Practical consequences included a trial that over-recruited by 20% in order to randomise 144 practices with all required permissions, and a 5-year delay in reporting a trial while retrospectively applied regulations were satisfied to allow data linkage. CONCLUSIONS Overcoming the substantial barriers of using routine primary care data will require a streamlined governance process, standardised understanding/application of regulations and adequate National Health Service IT (Information Technology) capability. Without policy-driven prioritisation of these changes, the potential of this valuable resource will not be leveraged.
Collapse
Affiliation(s)
- Holly Tibble
- The University of Edinburgh Usher Institute of Population Health Sciences and Informatics, Edinburgh, UK
| | - Rami A Alyami
- University of Sheffield, Sheffield, Saudi Arabia
- Respiratory Therapy Department, Jazan University, Jazan, Saudi Arabia
| | - Andrew Bush
- National Heart and Lung Institute, London, UK
- Imperial College London, London, UK
| | - Steve Cunningham
- Department of Child Life and Health, University of Edinburgh, Edinburgh, UK
| | | | - David Price
- Observational and Pragmatic Research Institute, Singapore
- University of Aberdeen, Aberdeen, UK
| | | | | | - Kay Wang
- University of Oxford, Oxford, UK
| | | | | | | | | | | | | | | | - Luke Daines
- Centre for Medical Informatics, The University of Edinburgh Usher Institute of Population Health Sciences and Informatics, Edinburgh, UK
| | - Hilary Pinnock
- College of Medicine and Veterinary Medic, The University of Edinburgh Usher Institute of Population Health Sciences and Informatics, Edinburgh, UK
| |
Collapse
|
17
|
Zong J, Rojubally A, Pan X, Wolf B, Greenfeder S, Upton A, Gdovin Bergeson J. A Review and Comparative Case Study Analysis of Real-World Evidence in European Regulatory and Health Technology Assessment Decision Making for Oncology Medicines. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2025; 28:31-41. [PMID: 39393564 DOI: 10.1016/j.jval.2024.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 09/13/2024] [Accepted: 09/16/2024] [Indexed: 10/13/2024]
Abstract
OBJECTIVES Real-world evidence (RWE) is valuable in supporting regulatory and health technology assessment (HTA) decisions; however, the actual contribution to approvals remains elusive. This study aimed to review RWE approaches and use in oncology medicine approvals in Europe and understand cohesion and discrepancy in the acceptance of the RWE by the European Medicines Agency (EMA) and European HTA bodies. METHODS This scoping review involved a search of the EMA database, National Institute for Health and Care Excellence (NICE), Gemeinsamer Bundesausschuss (G-BA), and Haute Autorité de Santé (HAS) websites to identify final reports and appraisals for oncology medicines with references to RWE. The selection was guided by research terms associated with RWE study designs, data sources, and outcomes. Qualitative analysis was used to systemize the data. Case studies assessed by more than one agency were selected for comparative assessment of RWE approach, use, and acceptability. RESULTS RWE was mainly leveraged as an external control for indirect treatment comparisons or contextualization to support clinical trial results by the EMA, NICE, G-BA, and HAS. However, this approach was mostly rejected due to methodology biases. Comparative assessment of RWE acceptability for the same oncology medicines across agencies suggests discrepancies between EMA and European HTA bodies and among NICE, G-BA, and HAS. CONCLUSIONS There is diverging acceptance of RWE in EMA and European HTA bodies with no clear consensus on the most effective way to leverage RWE in approvals. With the introduction of the joint European Union Joint Clinical Assessment in 2025, it is crucial for European HTA bodies and EMA to develop synergetic standards for the use of RWE to ensure equitable and timely access to medicines.
Collapse
Affiliation(s)
- Jihong Zong
- Bayer Healthcare Pharmaceuticals Inc, Whippany, NJ, USA
| | | | - Xiaoyun Pan
- Bayer Healthcare Pharmaceuticals Inc, Boston, MA, USA
| | | | | | | | | |
Collapse
|
18
|
Wang M, Ma H, Shi Y, Ni H, Qin C, Ji C. Single-arm clinical trials: design, ethics, principles. BMJ Support Palliat Care 2024; 15:46-54. [PMID: 38834238 PMCID: PMC11874317 DOI: 10.1136/spcare-2024-004984] [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: 05/15/2024] [Accepted: 05/21/2024] [Indexed: 06/06/2024]
Abstract
Although randomised controlled trials are considered the gold standard in clinical research, they are not always feasible due to limitations in the study population, challenges in obtaining evidence, high costs and ethical considerations. As a result, single-arm trial designs have emerged as one of the methods to address these issues. Single-arm trials are commonly applied to study advanced-stage cancer, rare diseases, emerging infectious diseases, new treatment methods and medical devices. Single-arm trials have certain ethical advantages over randomised controlled trials, such as providing equitable treatment, respecting patient preferences, addressing rare diseases and timely management of adverse events. While single-arm trials do not adhere to the principles of randomisation and blinding in terms of scientific rigour, they still incorporate principles of control, balance and replication, making the design scientifically reasonable. Compared with randomised controlled trials, single-arm trials require fewer sample sizes and have shorter trial durations, which can help save costs. Compared with cohort studies, single-arm trials involve intervention measures and reduce external interference, resulting in higher levels of evidence. However, single-arm trials also have limitations. Without a parallel control group, there may be biases in interpreting the results. In addition, single-arm trials cannot meet the requirements of randomisation and blinding, thereby limiting their evidence capacity compared with randomised controlled trials. Therefore, researchers consider using single-arm trials as a trial design method only when randomised controlled trials are not feasible.
Collapse
Affiliation(s)
- Minyan Wang
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Huan Ma
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yun Shi
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Haojie Ni
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Chu Qin
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Conghua Ji
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| |
Collapse
|
19
|
Wilson BE, Booth CM. Real-world data: bridging the gap between clinical trials and practice. EClinicalMedicine 2024; 78:102915. [PMID: 39588211 PMCID: PMC11585814 DOI: 10.1016/j.eclinm.2024.102915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 11/27/2024] Open
Abstract
Real-world data (RWD) are rapidly emerging sources of information for patients, clinicians and regulators. While randomized controlled trials (RCTs) reduce bias and confounding through the randomization process and provide the highest quality of evidence regarding drug efficacy, RCTs may be impractical or unfeasible for rare diseases or disease subsets. And yet, studies attempting to replicate clinical trial results using observational datasets have failed. Given the inherent differences between observational data and clinical trial results, this discordance is not surprising. However, RWD may still have independent value as complementary tools to trial results. In this viewpoint, we explore the challenges of RWD and discuss key questions that clinicians, patients, and regulators will need to consider when faced with positive efficacy data from clinical trials, and negative effectiveness data from real world studies. Finally, we explore novel trial designs that might help bridge the gap from RCTs to RWD.
Collapse
Affiliation(s)
- Brooke E. Wilson
- Department of Oncology, Queen's University, Kingston, Ontario, Canada
- Division of Cancer Care and Epidemiology, Queen's Cancer Research Institute, Kingston, Canada
- School of Population Health, Faculty of Medicine and Health, UNSW Sydney, Australia
| | - Christopher M. Booth
- Department of Oncology, Queen's University, Kingston, Ontario, Canada
- Division of Cancer Care and Epidemiology, Queen's Cancer Research Institute, Kingston, Canada
| |
Collapse
|
20
|
Daigl M, Abogunrin S, Castro F, McGough SF, Sturrup RH, Boersma C, Abrams KR. Advancing the role of real-world evidence in comparative effectiveness research. J Comp Eff Res 2024; 13:e240101. [PMID: 39392412 DOI: 10.57264/cer-2024-0101] [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] [Indexed: 10/12/2024] Open
Abstract
Aim: Comparative effectiveness research (CER) is essential for making informed decisions about drug access. It provides insights into the effectiveness and safety of new drugs compared with existing treatments, thereby guiding better healthcare decisions and ensuring that new therapies meet the real-world needs of patients and healthcare systems. Objective: To provide a tool that assists analysts and decision-makers in identifying the most suitable analytical approach for answering a CER question, given specific data availability contexts. Methods: A systematic literature review of the scientific literature was performed and existing regulatory and health technology assessment (HTA) guidance were evaluated to identify and compare recommendations and best practices. Based on this review a methods flowchart that synthesizes current practices and requirements was proposed. Results: The review did not find any papers that clearly identified the most appropriate analytical approach for answering CER questions under various conditions. Therefore, a methods flowchart was designed to inform analyst and decision makers choices starting from a well-defined scientific question. Conclusion: The proposed methods flowchart offers clear guidance on CER methodologies across a range of settings and research needs. It begins with a well-defined research question and considers multiple feasibility aspects related to CER. This tool aims to standardize methods, ensure rigorous and consistent research quality and promote a culture of evidence-based decision-making in healthcare.
Collapse
Affiliation(s)
- Monica Daigl
- Global Access F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland
| | - Seye Abogunrin
- Global Access F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland
| | - Felipe Castro
- Data Science, F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland
| | - Sarah F McGough
- Computational Sciences, Genentech Inc., South San Franscisco, CA 94080, USA
| | | | - Cornelis Boersma
- Health-Ecore, 3704 HE Zeist, The Netherlands
- Department of Health Sciences, University Medical Center Groningen, Groningen, 9700 AB, The Netherlands
- Department of Management Sciences, Open University, Heerlen, 6419 AT, The Netherlands
| | - Keith R Abrams
- Department of Statistics & Warwick Medical School, University of Warwick, Coventry, CV4 7AL, UK
- Centre for Health Economics, University of York, York, YO10 5DD, UK
| |
Collapse
|
21
|
Takayama A, Yoshida S, Kawakami K. Tadalafil use is associated with a lower incidence of Type 2 diabetes in men with benign prostatic hyperplasia: A population-based cohort study. J Intern Med 2024; 296:422-434. [PMID: 39287476 DOI: 10.1111/joim.20012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
BACKGROUND Tadalafil, commonly prescribed for benign prostatic hyperplasia (BPH), may benefit patients with Type 2 diabetes mellitus (T2DM) for glycemic markers and complications. However, the association between the long-term use of tadalafil and the incidence of T2DM has not been investigated. METHODS We emulated a target trial of tadalafil use (5 mg/day) and the risk of T2DM using a population-based claims database in Japan. Patients who initiated tadalafil or alpha-blockers for BPH and had no history of diabetes diagnosis, no dispensing of glucose-lowering drugs, and no history of hemoglobin A1c levels of ≥6.5% (47-48 mmol/mol) were included. The primary outcome was the incidence of T2DM. Pooled logistic regression was used to estimate adjusted risk ratios (RRs) and 5-year cumulative incidence differences (CIDs). RESULTS A total of 5180 participants initiated tadalafil treatment and were compared with 20,049 patients who initiated alpha-blockers. The median follow-up time for each arm was 27.2 months (interquartile range [IQR], 12.0-47.9) in tadalafil users and 31.3 months (IQR, 13.7-57.2) in alpha-blocker users. The incidence rates of T2DM in tadalafil and alpha-blocker users were 5.4 (95% confidence interval [CI], 4.0-7.2) and 8.8 (95% CI, 7.8-9.8) per 1000-person years, respectively. Initiation of tadalafil was associated with a reduced risk of T2DM (RR, 0.47; 95% CI, 0.39-0.62; 5-year CID, -0.031; 95% CI, -0.040 to -0.019). CONCLUSION The incidence of T2DM was lower in men with BPH treated with tadalafil than in those treated with alpha-blockers. Thus, tadalafil may be more beneficial than alpha-blockers in preventing T2DM.
Collapse
Affiliation(s)
- Atsushi Takayama
- Department of Pharmacoepidemiology, Kyoto University Graduate School of Medicine and Public Health, Kyoto, Japan
| | - Satomi Yoshida
- Department of Clinical Medicine, Division of Social Medicine, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Koji Kawakami
- Department of Pharmacoepidemiology, Kyoto University Graduate School of Medicine and Public Health, Kyoto, Japan
| |
Collapse
|
22
|
Imamura K, Izumi Y, Egawa N, Ayaki T, Nagai M, Nishiyama K, Watanabe Y, Murakami T, Hanajima R, Kataoka H, Kiriyama T, Nanaura H, Sugie K, Hirayama T, Kano O, Nakamori M, Maruyama H, Haji S, Fujita K, Atsuta N, Tatebe H, Tokuda T, Takahashi N, Morinaga A, Tabuchi R, Oe M, Kobayashi M, Lobello K, Morita S, Sobue G, Takahashi R, Inoue H. Protocol for a phase 2 study of bosutinib for amyotrophic lateral sclerosis using real-world data: induced pluripotent stem cell-based drug repurposing for amyotrophic lateral sclerosis medicine (iDReAM) study. BMJ Open 2024; 14:e082142. [PMID: 39461864 PMCID: PMC11529471 DOI: 10.1136/bmjopen-2023-082142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 09/24/2024] [Indexed: 10/29/2024] Open
Abstract
INTRODUCTION Amyotrophic lateral sclerosis (ALS) is a progressive, severe neurodegenerative disease caused by motor neuron death. Development of a medicine for ALS is urgently needed, and induced pluripotent cell-based drug repurposing identified a Src/c-Abl inhibitor, bosutinib, as a candidate for molecular targeted therapy of ALS. A phase 1 study confirmed the safety and tolerability of bosutinib in a 12-week treatment of ALS patients. The objectives of this study are to evaluate the efficacy and longer-term safety of bosutinib in ALS patients. METHODS AND ANALYSIS An open-label, multicentre phase 2 study was designed. The study consisted of a 12-week observation period, a 1-week transitional period, a 24-week study treatment period and a 4-week follow-up period. Following the transitional period, patients whose total Revised ALS Functional Rating Scale (ALSFRS-R) score declined by 1 to 4 points during the 12-week observation period were to receive bosutinib for 24 weeks. In this study, 25 ALS patients will be enrolled; patients will be randomly assigned to the following groups: 12 patients in the 200 mg quaque die (QD) group and 13 patients in the 300 mg QD group of bosutinib. The safety and exploratory efficacy of bosutinib in ALS patients for 24 weeks will be assessed. Efficacy using the ALSFRS-R score will be compared with the external published data from an edaravone study (MCI186-19) and registry data from a multicentre ALS cohort study, the Japanese Consortium for Amyotrophic Lateral Sclerosis Research. ETHICS AND DISSEMINATION This study was approved by the ethics committees of Kyoto University, Tokushima University, Kitasato University, Tottori University, Nara Medical University School of Medicine, Toho University and Hiroshima University. The findings will be disseminated in peer-reviewed journals and at scientific conferences. TRIAL REGISTRATION NUMBER jRCT2051220002; Pre-results, NCT04744532; Pre-results.
Collapse
Affiliation(s)
- Keiko Imamura
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Yuishin Izumi
- Department of Neurology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Naohiro Egawa
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takashi Ayaki
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Makiko Nagai
- Department of Neurology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Kazutoshi Nishiyama
- Department of Neurology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Yasuhiro Watanabe
- Division of Neurology, Department of Brain and Neurosciences, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Takenobu Murakami
- Division of Neurology, Department of Brain and Neurosciences, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Ritsuko Hanajima
- Division of Neurology, Department of Brain and Neurosciences, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Hiroshi Kataoka
- Department of Neurology, Nara Medical University School of Medicine, Kashihara, Japan
| | - Takao Kiriyama
- Department of Neurology, Nara Medical University School of Medicine, Kashihara, Japan
| | - Hitoki Nanaura
- Department of Neurology, Nara Medical University School of Medicine, Kashihara, Japan
| | - Kazuma Sugie
- Department of Neurology, Nara Medical University School of Medicine, Kashihara, Japan
| | - Takehisa Hirayama
- Department of Neurology, Toho University Faculty of Medicine, Tokyo, Japan
| | - Osamu Kano
- Department of Neurology, Toho University Faculty of Medicine, Tokyo, Japan
| | - Masahiro Nakamori
- Department of Clinical Neuroscience and Therapeutics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hirofumi Maruyama
- Department of Clinical Neuroscience and Therapeutics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Shotaro Haji
- Department of Neurology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Koji Fujita
- Department of Neurology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Naoki Atsuta
- Department of Neurology, Aichi Medical University, Nagakute, Japan
| | - Harutsugu Tatebe
- Advanced Neuroimaging Center, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Takahiko Tokuda
- Advanced Neuroimaging Center, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Naoto Takahashi
- Department of Hematology, Nephrology, and Rheumatology, Akita University Graduate School of Medicine, Akita, Japan
| | | | | | | | | | - Kasia Lobello
- Pfizer Worldwide Research and Development, Collegeville, Pennsylvania, USA
| | - Satoshi Morita
- Department of Biomedical Statistics and Bioinformatics, Kyoto University, Kyoto, Japan
- Institute for Advancement of Clinical and Translational Science (iACT), Kyoto University Hospital, Kyoto, Japan
| | - Gen Sobue
- Aichi Medical University, Nagakute, Japan
| | - Ryosuke Takahashi
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Haruhisa Inoue
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Institute for Advancement of Clinical and Translational Science (iACT), Kyoto University Hospital, Kyoto, Japan
| |
Collapse
|
23
|
Nyström T. Key results from observational studies and real-world evidence of sodium-glucose cotransporter-2 inhibitor effectiveness and safety in reducing cardio-renal risk. Diabetes Obes Metab 2024; 26 Suppl 5:35-57. [PMID: 38859661 DOI: 10.1111/dom.15696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 05/09/2024] [Accepted: 05/20/2024] [Indexed: 06/12/2024]
Abstract
Sodium-glucose cotransporter 2 (SGLT2) inhibitors, originally designed to manage blood sugar levels in individuals with type 2 diabetes (T2D), have emerged as a crucial class of drugs for managing cardio-renal diseases. These drugs work by targeting the SGLT2 protein in the kidneys, promoting the excretion of glucose and influencing metabolic pathways beyond glucose control. The relationship between cardio-renal diseases and SGLT2 inhibitors has been explored through landmark trials and real-world evidence (RWE) studies, demonstrating significant reductions in cardio-renal complications. This review discusses the importance of RWE studies alongside randomized controlled trials in understanding the real-world effectiveness and safety of SGLT2 inhibitors. It outlines the advantages and disadvantages of RWE compared to RCTs, highlighting their complementary roles in providing comprehensive insights into treatment outcomes. By examining a range of RWE studies, the review underscores the cardio-renal benefits of SGLT2 inhibitors across various patient populations. Safety assessments indicate that SGLT2 inhibitors are generally well tolerated, with severe adverse events being rare. Common issues, such as genital mycotic infections and urinary tract infections, are acknowledged, alongside less frequent but significant adverse events including diabetic ketoacidosis, lower-limb amputations, and bone fractures. In summary, SGLT2 inhibitors show promising cardio-renal protective effects in real-world scenarios across diverse populations in T2D, indicating their potential as early intervention measures. Continued research is essential for gaining a thorough understanding of their long-term effects and safety profiles.
Collapse
Affiliation(s)
- Thomas Nyström
- Department of Clinical Science and Education, Karolinska Institutet, Södersjukhuset, Stockholm, Sweden
- Department of Internal Medicine, Section of Endocrinology and Diabetology, Södersjukhuset, Stockholm, Sweden
| |
Collapse
|
24
|
Wang P, Chow SC. The use of real-world data for clinical investigation of effectiveness in drug development. J Biopharm Stat 2024; 34:818-841. [PMID: 38519266 DOI: 10.1080/10543406.2024.2330215] [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: 07/10/2023] [Accepted: 01/08/2024] [Indexed: 03/24/2024]
Abstract
With the growing interest in leveraging real-world data (RWD) to support effectiveness evaluations for new indications, new target populations, and post-market performance, the United States Food and Drug Administration has published several guidance documents on RWD sources and real-world studies (RWS) to assist sponsors in generating credible real-world evidence (RWE). Meanwhile, the randomized controlled trial (RCT) remains the gold standard in drug evaluation. Along this line, we propose a hybrid two-stage adaptive design to evaluate effectiveness based on evidence from both RCT and RWS. At the first stage, a typical non-inferiority test is conducted using RCT data to test for not-ineffectiveness. Once not-ineffectiveness is established, the study proceeds to the second stage to conduct an RWS and test for effectiveness using integrated information from RCT and RWD. The composite likelihood approach is implemented as a down-weighing strategy to account for the impact of high variability in RWS population. An optimal sample size determination procedure for RCT and RWS is introduced, aiming to achieve the minimal expected sample size. Through extensive numerical study, the proposed design demonstrates the ability to control type I error inflation in most cases and consistently maintain statistical power above the desired level. In general, this RCT/RWS hybrid two-stage adaptive design is beneficial for effectiveness evaluations in drug development, especially for oncology and rare diseases.
Collapse
Affiliation(s)
- Peijin Wang
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Shein-Chung Chow
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, North Carolina, USA
| |
Collapse
|
25
|
Hegele A, Häußermann R, Schultheis S, Skrobek L, Vink M, Hollwegs S, Ludwig M, Huwe P, Maywurm M, Bartsch-Polle A, Weber J, Thiemer M, Varughese D. Apalutamide for non-metastatic castration-resistant prostate cancer (nmCRPC): real world data of a multicenter study. J Cancer Res Clin Oncol 2024; 150:414. [PMID: 39249593 PMCID: PMC11384626 DOI: 10.1007/s00432-024-05928-7] [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: 05/31/2024] [Accepted: 08/16/2024] [Indexed: 09/10/2024]
Abstract
PURPOSE Apalutamide plus androgen-deprivation therapy (ADT) improved outcomes in patients with non-metastatic castration-resistant prostate cancer (nmCRPC). Nevertheless real-world data are limited. The aim of this multicenter study was to generate real-world data from nmCRPC patients treated with ADT plus apalutamide. METHODS In this observational cohort based investigator initiated trial data of nmCRPC patients receiving apalutamide plus ADT were collected focusing on patient demographic data, prostate-specific antigen (PSA) declines, safety profile including dose modification/discontinuation as well as subsequent therapy and metastasis-free survival (MFS). RESULTS Data from a total of 31 nmCRPC patients were documented. Compared to the Phase III study Spartan real-world patients are older, showed a higher ECOG-PS and more aggressive tumors. In the cohort PSA decreased about 98.1%, 74% of patients showed a PSA decrease over 90% and 54.8% reached a PSA-level < 0.2ng/ml. Apalutamide was well tolerated in real world patients: adverse events occurred in 67.7% but were in the majority mild (≥ grade 3: 6.5%). Dose reduction was necessary in 38.7% and 32.2% discontinued apalutamide treatment. MFS was 43 months and majority of patients were subsequently treated with abiraterone. CONCLUSION In real world more comorbid nmCRPC patients with a higher ECOG-PS and more aggressive tumors are treated with apalutamide plus ADT. Nevertheless efficacy results as well as side effects are similar in real-world compared to Spartan trial showing also a rapid, durable and deep PSA response with a median MFS of 43 months.
Collapse
Affiliation(s)
- Axel Hegele
- Urological Center Mittelhessen, DRK Hospital Biedenkopf, Biedenkopf, Germany.
- Department of Urology, Philipps University Marburg, Marburg, Germany.
| | - Rainer Häußermann
- Urological Center Mittelhessen, DRK Hospital Biedenkopf, Biedenkopf, Germany
- Department of Urology, Philipps University Marburg, Marburg, Germany
| | | | | | - Meike Vink
- Urological Practice, Homberg/Efze, Germany
| | | | | | | | | | | | - Jost Weber
- Urological Center Wetterau, Büdingen, Germany
| | - Markus Thiemer
- Department of Radiotherapy and Radiooncology, Philipps-University Marburg, Marburg, Germany
| | | |
Collapse
|
26
|
Somolinos-Simón FJ, García-Sáez G, Tapia-Galisteo J, Corcoy R, Elena Hernando M. Cluster analysis of adult individuals with type 1 diabetes: Treatment pathways and complications over a five-year follow-up period. Diabetes Res Clin Pract 2024; 215:111803. [PMID: 39089589 DOI: 10.1016/j.diabres.2024.111803] [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: 02/23/2024] [Revised: 06/14/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024]
Abstract
AIMS To identify subgroups of adults with type 1 diabetes and analyse their treatment pathways and risk of diabetes-related complications over a 5-year follow-up. METHODS We performed a k-means cluster analysis using the T1DExchange Registry (n = 6,302) to identify subgroups based on demographic and clinical characteristics. Annual reassessments linked treatment trajectories with these clusters, considering drug and technology use. Complication risks were analysed using Cox regression. RESULTS Five clusters were identified: 1) A favourable combination of all variables (31.67 %); 2) Longer diabetes duration (22.63 %); 3) Higher HbA1c levels (13.28 %); 4) Higher BMI (15.25 %); 5) Older age at diagnosis (17.17 %). Two-thirds of patients remained in their initial cluster annually. Technology adoption showed improved glycaemic control over time. Cox proportional hazards showed different risk patterns: Cluster 1 had low complication risk; Cluster 2 had the highest risk for retinopathy, coronary artery disease and autonomic neuropathy; Cluster 3 had the highest risk for albuminuria, depression and diabetic ketoacidosis; Cluster 4 had increased risk for multiple complications; Cluster 5 had the highest risk for hypertension and severe hypoglycaemia, with elevated coronary artery disease risk. CONCLUSIONS Clinical characteristics can identify subgroups of patients with T1DM showing differences in treatment and complications during follow-up.
Collapse
Affiliation(s)
- Francisco J Somolinos-Simón
- Centre for Biomedical Technology (CTB), ETSI de Telecomunicación, Universidad Politécnica de Madrid, Madrid, Spain
| | - Gema García-Sáez
- Centre for Biomedical Technology (CTB), ETSI de Telecomunicación, Universidad Politécnica de Madrid, Madrid, Spain; CIBER-BBN, ISCIII, Madrid, Spain.
| | - Jose Tapia-Galisteo
- Centre for Biomedical Technology (CTB), ETSI de Telecomunicación, Universidad Politécnica de Madrid, Madrid, Spain; CIBER-BBN, ISCIII, Madrid, Spain
| | - Rosa Corcoy
- CIBER-BBN, ISCIII, Madrid, Spain; Departament de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain; Institut de Recerca, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - M Elena Hernando
- Centre for Biomedical Technology (CTB), ETSI de Telecomunicación, Universidad Politécnica de Madrid, Madrid, Spain; CIBER-BBN, ISCIII, Madrid, Spain
| |
Collapse
|
27
|
Vaszar LT, Sharp RR, Carter RE, Wright RS. Pragmatic Clinical Trials: The Ethics of Conducting Research in the Real World. Mayo Clin Proc 2024; 99:1369-1373. [PMID: 39101864 PMCID: PMC11831573 DOI: 10.1016/j.mayocp.2024.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 04/30/2024] [Accepted: 05/06/2024] [Indexed: 08/06/2024]
Affiliation(s)
- Laszlo T Vaszar
- Institutional Review Board, Department of Medicine, Division of Pulmonary Medicine, Mayo Clinic Arizona, Phoenix, AZ, USA
| | - Richard R Sharp
- Department of Quantitative Health Sciences, Biomedical Ethics Research Program, Mayo Clinic Rochester, Rochester MN, USA
| | - Rickey E Carter
- Department of Quantitative Health Sciences, Mayo Clinic Florida, Jacksonville, FL, USA
| | - R Scott Wright
- Institutional Review Board, Department of Cardiovascular Diseases, Mayo Clinic Rochester, Rochester MN, USA
| |
Collapse
|
28
|
Suzuki N, Taguri M. A New Criterion for Determining a Cutoff Value Based on the Biases of Incidence Proportions in the Presence of Non-differential Outcome Misclassifications. Epidemiology 2024; 35:618-627. [PMID: 38968067 PMCID: PMC11309335 DOI: 10.1097/ede.0000000000001756] [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: 05/16/2023] [Accepted: 05/22/2024] [Indexed: 07/07/2024]
Abstract
When conducting database studies, researchers sometimes use an algorithm known as "case definition," "outcome definition," or "computable phenotype" to identify the outcome of interest. Generally, algorithms are created by combining multiple variables and codes, and we need to select the most appropriate one to apply to the database study. Validation studies compare algorithms with the gold standard and calculate indicators such as sensitivity and specificity to assess their validities. As the indicators are calculated for each algorithm, selecting an algorithm is equivalent to choosing a pair of sensitivity and specificity. Therefore, receiver operating characteristic curves can be utilized, and two intuitive criteria are commonly used. However, neither was conceived to reduce the biases of effect measures (e.g., risk difference and risk ratio), which are important in database studies. In this study, we evaluated two existing criteria from perspectives of the biases and found that one of them, called the Youden index always minimizes the bias of the risk difference regardless of the true incidence proportions under nondifferential outcome misclassifications. However, both criteria may lead to inaccurate estimates of absolute risks, and such property is undesirable in decision-making. Therefore, we propose a new criterion based on minimizing the sum of the squared biases of absolute risks to estimate them more accurately. Subsequently, we apply all criteria to the data from the actual validation study on postsurgical infections and present the results of a sensitivity analysis to examine the robustness of the assumption our proposed criterion requires.
Collapse
Affiliation(s)
- Norihiro Suzuki
- From the Department of Health Data Science, Tokyo Medical University, Tokyo, Japan
| | - Masataka Taguri
- From the Department of Health Data Science, Tokyo Medical University, Tokyo, Japan
- Research Center for Medical and Health Data Science, The Institute of Statistical Mathematics, Tokyo, Japan
| |
Collapse
|
29
|
Wu YE, Zheng YY, Li QY, Yao BF, Cao J, Liu HX, Hao GX, van den Anker J, Zheng Y, Zhao W. Model-informed drug development in pediatric, pregnancy and geriatric drug development: States of the art and future. Adv Drug Deliv Rev 2024; 211:115364. [PMID: 38936664 DOI: 10.1016/j.addr.2024.115364] [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: 09/25/2023] [Revised: 06/09/2024] [Accepted: 06/19/2024] [Indexed: 06/29/2024]
Abstract
The challenges of drug development in pediatric, pregnant and geriatric populations are a worldwide concern shared by regulatory authorities, pharmaceutical companies, and healthcare professionals. Model-informed drug development (MIDD) can integrate and quantify real-world data of physiology, pharmacology, and disease processes by using modeling and simulation techniques to facilitate decision-making in drug development. In this article, we reviewed current MIDD policy updates, reflected on the integrity of physiological data used for MIDD and the effects of physiological changes on the drug PK, as well as summarized current MIDD strategies and applications, so as to present the state of the art of MIDD in pediatric, pregnant and geriatric populations. Some considerations are put forth for the future improvements of MIDD including refining regulatory considerations, improving the integrity of physiological data, applying the emerging technologies, and exploring the application of MIDD in new therapies like gene therapies for special populations.
Collapse
Affiliation(s)
- Yue-E Wu
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yuan-Yuan Zheng
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qiu-Yue Li
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Bu-Fan Yao
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jing Cao
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Hui-Xin Liu
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Guo-Xiang Hao
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - John van den Anker
- Division of Clinical Pharmacology, Children's National Medical Center, Washington, DC, USA; Departments of Pediatrics, Pharmacology & Physiology, George Washington University, School of Medicine and Health Sciences, Washington, DC, USA; Department of Paediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel, Basel, Switzerland
| | - Yi Zheng
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wei Zhao
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.
| |
Collapse
|
30
|
Alipour‐Haris G, Liu X, Acha V, Winterstein AG, Burcu M. Real-world evidence to support regulatory submissions: A landscape review and assessment of use cases. Clin Transl Sci 2024; 17:e13903. [PMID: 39092896 PMCID: PMC11295294 DOI: 10.1111/cts.13903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/24/2024] [Accepted: 07/10/2024] [Indexed: 08/04/2024] Open
Abstract
Real-world evidence (RWE) has an increasing role in preapproval settings to support the approval of new medicines and indications. The main objectives of this study were to identify and characterize regulatory use cases that utilized RWE and other related observational approaches through targeted review of publications and regulatory review documents. After screening and inclusion/exclusion, the review characterized 85 regulatory applications with RWE. A total of 31 were in oncology and 54 were in non-oncology therapeutic areas. Most were for indications in adults only (N = 42, 49.4%), while 13 were in pediatrics only (15.3%), and 30 were in both (35.3%). In terms of regulatory context, 59 cases (69.4%) were for an original marketing application, 24 (28.2%) were for label expansion, and 2 (2.4%) were for label modification. Most also received special regulatory designations (e.g., orphan indication, breakthrough therapy, fast track, conditional, and accelerated approvals). There were 42 cases that utilized RWE to support single-arm trials. External data to support single-arm trials were utilized in various ways across use cases, including direct matching, benchmarking, natural history studies as well as literature or previous trials. A variety of data sources were utilized, including electronic health records, claims, registries, site-based charts. Endpoints in oncology use cases commonly included overall survival, progression-free survival. In 13 use cases, RWE was not considered supportive/definitive in regulatory decision-making due to design issues (e.g., small sample size, selection bias, missing data). Overall, RWE is utilized in regulatory approval processes for new indications/label expansion across various therapeutic areas with wide range of approaches. Multifaceted cross-sector efforts are needed to further improve the quality and utility of RWE in regulatory decision-making.
Collapse
|
31
|
Mishra S, Venkatesh MP. Rare disease clinical trials in the European Union: navigating regulatory and clinical challenges. Orphanet J Rare Dis 2024; 19:285. [PMID: 39085891 PMCID: PMC11292868 DOI: 10.1186/s13023-024-03146-5] [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: 06/28/2023] [Accepted: 03/24/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND Clinical development for orphan drugs presents significant difficulties and challenges. There is no unique or standard design, conduct, and outcome assessment methodology and it is sometimes impractical to fit design models of rare disease trials in any practiced and well-known framework. In the European Union (EU) these challenges encompass a broad array of subjects, including trial design, study outcomes, patient recruitment, trial conduct ethics, trial cost, and chances of success. This literature-based review study aims to provide a thorough overview of the critical aspects of rare disease trials in the EU by analyzing the current landscape of rare disease trials, highlighting key challenges, delving into regulatory and research initiatives and innovation in trial designs, and proposing multi-faceted solutions to implement effective rare disease clinical trials in the region. DISCUSSION Traditional clinical trial designs, validation, and evaluation methodologies used for nonorphan drugs often prove unsuitable for orphan drugs, given the small patient populations, sometimes fewer than 1000 cases. There is an increasing need for accessible therapies and both regulators as well as industry are trying to develop affordable and effective drugs to address this need. Despite several steps that have been taken, the timely development of drugs remains a challenge. One of the reasons behind the long development timeline is the recruitment, retention, and conduct of rare disease trials. To optimize the development timelines of orphan drugs in the EU, it is important to ensure that the safety and efficacy of the product is not compromised. Industry and regulatory agencies must implement innovative trial designs, devise flexible policies, and incorporate real-world data for assessing clinical outcomes. CONCLUSION Collaboration among academic institutions, pharmaceutical companies (both small and major), patient groups, and health authorities is crucial in overcoming obstacles related to clinical trials and providing assistance and creative ideas. The ultimate objective of granting rare disease patients timely and affordable access to medications with a positive balance between benefits and risks is to be met.
Collapse
Affiliation(s)
- Sangita Mishra
- Dept. of Pharmaceutics, Centre of Excellence in Regulatory Sciences, JSS College of Pharmacy, JSS Academy of Higher Education and Research, SS Nagara, Mysore, Karnataka, 570015, India
| | - M P Venkatesh
- Dept. of Pharmaceutics, Centre of Excellence in Regulatory Sciences, JSS College of Pharmacy, JSS Academy of Higher Education and Research, SS Nagara, Mysore, Karnataka, 570015, India.
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, Malaysia.
| |
Collapse
|
32
|
Solanki N, Beck B, Labadia M, Smith K, Peterson L, King S, Micklewright S, Pennington E, Farooq S, Zhang P, Aronica M, Zein J, Khatri S, Comhair S, Erzurum S. The role of ACT score in mepolizumab discontinuation. J Asthma 2024; 61:550-560. [PMID: 38064231 PMCID: PMC11076164 DOI: 10.1080/02770903.2023.2293067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/09/2023] [Accepted: 12/03/2023] [Indexed: 12/23/2023]
Abstract
BACKGROUND Mepolizumab is a therapy for severe asthma. We have little knowledge of the characteristics of people in the US that discontinue mepolizumab in clinical care. OBJECTIVE To investigate the real-world efficacy and time to clinical discontinuation of mepolizumab, we evaluated individuals with asthma started on mepolizumab at the Cleveland Clinic. We hypothesized that individuals that discontinue mepolizumab have more severe and uncontrolled asthma at baseline. METHODS Between 2016 and 2022, patients who started on mepolizumab consented to be assessed over 18 months. At baseline, a questionnaire including demographic and medical history was collected. Laboratory findings such as ACT score, FENO (Fractional Excretion of Nitric Oxide), and spirometry were recorded. At the conclusion of the observation period, the participants were divided into two categories: Group A and Group B. RESULTS Group B [N = 28] discontinued mepolizumab (p < 0.05) at an average of 5.8 months (SD 4.2 months). Group A [N = 129] stayed on the therapy for at least 1 year. A participant with an ACT score less than 13 has an odds ratio of 6.64 (95% CI, 2.1 - 26.0) of discontinuing mepolizumab therapy. For a male, the odds of discontinuing mepolizumab therapy is 3.39 (95% CI, 1.1-11.2). CONCLUSION In this real-world study, we find that high eosinophil count may not be adequate in screening which individuals will benefit from mepolizumab. Up to 17% of patients fail therapy within 6 months, with male sex and low ACT score increasing risk of mepolizumab discontinuation at Cleveland Clinic.
Collapse
Affiliation(s)
- Neha Solanki
- Respiratory Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Brittany Beck
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Monica Labadia
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Kevin Smith
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Laura Peterson
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Stephanie King
- Respiratory Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | | | | | - Sobia Farooq
- Respiratory Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Peng Zhang
- Respiratory Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Mark Aronica
- Respiratory Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Joe Zein
- Respiratory Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Sumita Khatri
- National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| | - Suzy Comhair
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Serpil Erzurum
- Respiratory Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
33
|
Nguyen PA, Hsu MH, Chang TH, Yang HC, Huang CW, Liao CT, Lu CY, Hsu JC. Taipei Medical University Clinical Research Database: a collaborative hospital EHR database aligned with international common data standards. BMJ Health Care Inform 2024; 31:e100890. [PMID: 38749529 PMCID: PMC11097871 DOI: 10.1136/bmjhci-2023-100890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 04/29/2024] [Indexed: 05/18/2024] Open
Abstract
OBJECTIVE The objective of this paper is to provide a comprehensive overview of the development and features of the Taipei Medical University Clinical Research Database (TMUCRD), a repository of real-world data (RWD) derived from electronic health records (EHRs) and other sources. METHODS TMUCRD was developed by integrating EHRs from three affiliated hospitals, including Taipei Medical University Hospital, Wan-Fang Hospital and Shuang-Ho Hospital. The data cover over 15 years and include diverse patient care information. The database was converted to the Observational Medical Outcomes Partnership Common Data Model (OMOP CDM) for standardisation. RESULTS TMUCRD comprises 89 tables (eg, 29 tables for each hospital and 2 linked tables), including demographics, diagnoses, medications, procedures and measurements, among others. It encompasses data from more than 4.15 million patients with various medical records, spanning from the year 2004 to 2021. The dataset offers insights into disease prevalence, medication usage, laboratory tests and patient characteristics. DISCUSSION TMUCRD stands out due to its unique advantages, including diverse data types, comprehensive patient information, linked mortality and cancer registry data, regular updates and a swift application process. Its compatibility with the OMOP CDM enhances its usability and interoperability. CONCLUSION TMUCRD serves as a valuable resource for researchers and scholars interested in leveraging RWD for clinical research. Its availability and integration of diverse healthcare data contribute to a collaborative and data-driven approach to advancing medical knowledge and practice.
Collapse
Affiliation(s)
- Phung-Anh Nguyen
- Clinical Data Center, Office of Data Science, Taipei Medical University, Taipei, Taiwan
- Research Center of Health Care Industry Data Science, College of Management, Taipei Medical University, Taipei, Taiwan
- Clinical Big Data Research Center, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
| | - Min-Huei Hsu
- Office of Data Science, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Data Science, College of Management, Taipei Medical University, Taipei, Taiwan
| | - Tzu-Hao Chang
- Clinical Big Data Research Center, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- International Center for Health Information Technology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Hsuan-Chia Yang
- Clinical Big Data Research Center, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- International Center for Health Information Technology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Research Center of Big Data and Meta-Analysis, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Chih-Wei Huang
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- International Center for Health Information Technology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Chia-Te Liao
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Taipei Medical University-Research Center of Urology and Kidney, Taipei Medical University, Taipei, Taiwan
| | - Christine Y Lu
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA
- Kolling Institute, Faculty of Medicine and Health, The University of Sydney and the Northern Sydney Local Health District, Sydney, NSW, Australia
- School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Jason C Hsu
- Clinical Data Center, Office of Data Science, Taipei Medical University, Taipei, Taiwan
- Research Center of Health Care Industry Data Science, College of Management, Taipei Medical University, Taipei, Taiwan
- Clinical Big Data Research Center, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
- International Ph.D. Program in Biotech and Healthcare Management, College of Management, Taipei Medical Unversity, Taipei, Taiwan
| |
Collapse
|
34
|
Gray C, Ralphs E, Fox MP, Lash TL, Liu G, Kou TD, Rivera DR, Bosco J, Braun KVN, Grimson F, Layton D. Use of quantitative bias analysis to evaluate single-arm trials with real-world data external controls. Pharmacoepidemiol Drug Saf 2024; 33:e5796. [PMID: 38680093 DOI: 10.1002/pds.5796] [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: 05/23/2023] [Revised: 03/27/2024] [Accepted: 04/01/2024] [Indexed: 05/01/2024]
Abstract
PURPOSE Use of real-world data (RWD) for external controls added to single-arm trials (SAT) is increasingly prevalent in regulatory submissions. Due to inherent differences in the data-generating mechanisms, biases can arise. This paper aims to illustrate how to use quantitative bias analysis (QBA). METHODS Advanced non-small cell lung cancer (NSCLC) serves as an example, where many small subsets of patients with molecular tumor subtypes exist. First, some sources of bias that may occur in oncology when comparing RWD to SAT are described. Second, using a hypothetical immunotherapy agent, a dataset is simulated based on expert input for survival analysis of advanced NSCLC. Finally, we illustrate the impact of three biases: missing confounder, misclassification of exposure, and outcome evaluation. RESULTS For each simulated scenario, bias was induced by removing or adding data; hazard ratios (HRs) were estimated applying conventional analyses. Estimating the bias-adjusted treatment effect and uncertainty required carefully selecting the bias model and bias factors. Although the magnitude of each biased and bias-adjusted HR appeared moderate in all three hypothetical scenarios, the direction of bias was variable. CONCLUSION These findings suggest that QBA can provide an intuitive framework for bias analysis, providing a key means of challenging assumptions about the evidence. However, the accuracy of bias analysis is itself dependent on correct specification of the bias model and bias factors. Ultimately, study design should reduce bias, but QBA allows us to evaluate the impact of unavoidable bias to assess the quality of the evidence.
Collapse
Affiliation(s)
- Christen Gray
- Real World Data Science, Biopharmaceuticals Medical Evidence, AstraZeneca, Cambridge, UK
- Methods and Evidence Generation, Real World Solutions, IQVIA, London, UK
- Health Data Science, London School of Hygiene and Tropical Medicine, London, UK
| | - Eleanor Ralphs
- Methods and Evidence Generation, Real World Solutions, IQVIA, London, UK
| | - Matthew P Fox
- Department of Epidemiology, Department of Global Health, Boston University, Boston, Massachusetts, USA
| | - Timothy L Lash
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
- Cancer Prevention and Control Program, Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| | - Geoffrey Liu
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Universal Health Network, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- Applied Molecular Profiling Pharmacogenomic Epidemiologic Laboratory, Princess Margaret Cancer Centre, Universal Health Network, Toronto, Ontario, Canada
| | - Tzuyung Doug Kou
- Global Patient Safety, BeiGene, Ridgefield Park, New Jersey, USA
| | - Donna R Rivera
- Oncology Center of Excellence, United States Food & Drug Administration, Silver Spring, Maryland, USA
| | - Jaclyn Bosco
- Epidemiology and Database Studies, Real World Solutions, IQVIA, Boston, Massachusetts, USA
- Department of Epidemiology, Boston University, Boston, Massachusetts, USA
| | - Kim Van Naarden Braun
- Translational Epidemiology, Informatics and Predictive Sciences, BMS, Summit, New Jersey, USA
| | - Fiona Grimson
- Health Data Science, London School of Hygiene and Tropical Medicine, London, UK
- Biometrics and Quantitative Sciences, UCB Pharma, Slough, UK
| | - Deborah Layton
- PEPI Consultancy Limited, Southampton, UK
- School of Life & Medical Sciences, University of Hertfordshire, Hatfield, UK
| |
Collapse
|
35
|
Ren L, Chen Q, Gao J, Liu Y, Tao Y, Li X, Luo Q, Lv F, Min S. Clinical efficacy of adjunctive esketamine anesthesia in electroconvulsive therapy for major depressive disorders: A pragmatic, randomized, controlled trial. Psychiatry Res 2024; 335:115843. [PMID: 38461645 DOI: 10.1016/j.psychres.2024.115843] [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: 11/01/2023] [Revised: 02/27/2024] [Accepted: 02/29/2024] [Indexed: 03/12/2024]
Abstract
Electroconvulsive therapy (ECT) is an effective treatment for depression, and esketamine has been shown to have antidepressant effects. However, it is currently unclear whether adjunctive esketamine can enhance the clinical efficacy of ECT in real-world clinical practice. In this pragmatic clinical trial, patients with major depression were randomly assigned into two groups: patients received 0.25 mg/kg esketamine plus propofol (esketamine group) or the same volume of saline (control group) plus propofol. Results indicated that there was no difference in response and remission rates between the two groups. However, patients receiving esketamine had a higher remission rate of SI and lower psychotic scores. Patients receiving esketamine also required a lower electric dose, but the seizure duration and cognitive function were comparable between the two groups. Diastolic blood pressure increased after esketamine injection, but there was no increased risk of hypertension. Furthermore, incidence of delirium and confusion were comparable between the groups. Conclusively, adjunctive esketamine anesthesia does not provide any advantage in improving the response and remission rates of ECT. However, it can improve remission of SI and alleviate accompanying psychotic symptoms in depressive patients. With adjunctive usage, the adverse cardiovascular and neuropsychiatric events associated with esketamine appear to be tolerable.
Collapse
Affiliation(s)
- Li Ren
- Department of Anesthesiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Qibin Chen
- Department of Anesthesiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Jin Gao
- Department of Anesthesiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Yuanyuan Liu
- Department of Anesthesiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Yi Tao
- Department of Phase I Clinical Trial Ward, the First Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Xiao Li
- Department of Psychiatry, the First Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Qinghua Luo
- Department of Psychiatry, the First Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Feng Lv
- Department of Anesthesiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, PR China.
| | - Su Min
- Department of Anesthesiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, PR China.
| |
Collapse
|
36
|
Mikl V, Baltic D, Czypionka T, Degelsegger-Márquez A, Forgó N, Gouya-Lechner G, Herzog A, Klimek P, Lumenta DB, Mraz B, Ostermann H, Scharinger R, Stamm T, Strassnig M, Zeitlinger M, Pleiner-Duxneuner J. A national evaluation analysis and expert interview study of real-world data sources for research and healthcare decision-making. Sci Rep 2024; 14:9751. [PMID: 38679653 PMCID: PMC11056370 DOI: 10.1038/s41598-024-59475-9] [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: 10/09/2023] [Accepted: 04/11/2024] [Indexed: 05/01/2024] Open
Abstract
Real-world data (RWD) can provide intel (real-world evidence, RWE) for research and development, as well as policy and regulatory decision-making along the full spectrum of health care. Despite calls from global regulators for international collaborations to integrate RWE into regulatory decision-making and to bridge knowledge gaps, some challenges remain. In this work, we performed an evaluation of Austrian RWD sources using a multilateral query approach, crosschecked against previously published RWD criteria and conducted direct interviews with representative RWD source samples. This article provides an overview of 73 out of 104 RWD sources in a national legislative setting where major attempts are made to enable secondary use of RWD (e.g. law on the organisation of research, "Forschungsorganisationsgesetz"). We were able to detect omnipresent challenges associated with data silos, variable standardisation efforts and governance issues. Our findings suggest a strong need for a national health data strategy and data governance framework, which should inform researchers, as well as policy- and decision-makers, to improve RWD-based research in the healthcare sector to ultimately support actual regulatory decision-making and provide strategic information for governmental health data policies.
Collapse
Affiliation(s)
- Veronika Mikl
- Gesellschaft für Pharmazeutische Medizin E.V. (GPMed), 1210, Vienna, Austria
- Roche Austria GmbH, Vienna, Austria
| | - Dejan Baltic
- Gesellschaft für Pharmazeutische Medizin E.V. (GPMed), 1210, Vienna, Austria
- Amgen GmbH, Vienna, Austria
| | - Thomas Czypionka
- Institut für Höhere Studien - Institute for Advanced Studies (IHS), 1080, Vienna, Austria
| | | | - Nikolaus Forgó
- Faculty of Law, Department of Innovation and Digitalisation in Law, University of Vienna, 1010, Vienna, Austria
| | - Ghazaleh Gouya-Lechner
- Gesellschaft für Pharmazeutische Medizin E.V. (GPMed), 1210, Vienna, Austria
- Gouya Insights GmbH & CoKG, 1190, Vienna, Austria
| | - Arnold Herzog
- Austrian Medicines and Medical Devices Agency (AGES Medizinmarktaufsicht), 1220, Vienna, Austria
| | - Peter Klimek
- Supply Chain Intelligence Institute Austria (ASCII), 1080, Vienna, Austria
- Section for Science of Complex Systems, Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University Vienna, 1090, Vienna, Austria
- Complexity Science Hub Vienna, 1080, Vienna, Austria
| | - David Benjamin Lumenta
- Research Unit for Digital Surgery, Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036, Graz, Austria
| | - Bernhard Mraz
- Gesellschaft für Pharmazeutische Medizin E.V. (GPMed), 1210, Vienna, Austria
- Novartis Pharma GmbH, Vienna, Austria
| | | | - Robert Scharinger
- Federal Ministry of Social Affairs, Health, Care and Consumer Protection, 1010, Vienna, Austria
| | - Tanja Stamm
- Gesellschaft für Pharmazeutische Medizin E.V. (GPMed), 1210, Vienna, Austria
- Section for Outcomes Research, Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, Medical University Vienna, 1090, Vienna, Austria
| | - Michael Strassnig
- Wiener Wissenschafts-, Forschungs- und Technologiefonds (Vienna Science and Technology Fund), 1090, Vienna, Austria
- Expertenplattform Plattform Registerforschung, c/o WWTF, 1090, Vienna, Austria
| | - Markus Zeitlinger
- Gesellschaft für Pharmazeutische Medizin E.V. (GPMed), 1210, Vienna, Austria
- Department of Clinical Pharmacology, Medical University Vienna, 1090, Vienna, Austria
| | - Johannes Pleiner-Duxneuner
- Gesellschaft für Pharmazeutische Medizin E.V. (GPMed), 1210, Vienna, Austria.
- Roche Austria GmbH, Vienna, Austria.
| |
Collapse
|
37
|
Sabolinski ML, Archambault T. Real-World Comparative Effectiveness Assessment Study of a Native Type I Collagen Matrix Plus Polyhexamethylene Biguanide Antimicrobial and a Cryopreserved Cadaveric Skin Allograft for Use in Diabetic Foot Ulcers - A Non-inferiority Analysis. EPLASTY 2024; 24:e16. [PMID: 38685995 PMCID: PMC11056628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
Objective To determine the effectiveness of a native type I collagen matrix plus polyhexamethylene biguanide antimicrobial (PCMP) and a cryopreserved cadaveric skin allograft (CCSA) for use in diabetic foot ulcers (DFUs). Methods A real-world data study was conducted on 989 DFUs analyzed digitally. Of these, 325 and 664 DFUs were treated with PCMP and CCSA, respectively. Non-inferiority testing for equivalence of PCMP and CCSA was performed at a level of significance of P < .05. Results Cox proportional hazards regression analysis for healing for PCMP and CCSA at weeks 4, 8, 12, and 24 was 12% vs 10%, 27% vs 24%, 39 % vs 37%, and 60% vs. 64%, respectively. No statistically significant differences were shown; P = .95. The median time to healing was 18 and 17 weeks for PCMP and CCSA, respectively; P = .95. The probability of healing was statistically equivalent between PCMP and CCSA; hazard ratio = 0.99; 95% CI (0.85, 1.17). Non-inferiority statistical testing results showed P = .01. Conclusions Using non-inferiority hypothesis testing at a level of significance of P <.05, we showed that PCMP was equivalent to CCSA; P = .01. PCMP vs CCSA demonstrated no statistically significant differences in median time, percentage, and probability of healing. Data from real-world data comparative effectiveness assessment studies can help guide clinicians to limit overuse of ineffective therapies and underuse of effective therapies.
Collapse
|
38
|
Sabolinski ML, Archambault T. Real-world data analysis of bilayered living cellular construct and fetal bovine collagen dressing treatment for pressure injuries: a comparative effectiveness study. J Comp Eff Res 2024; 13:e230109. [PMID: 38348818 PMCID: PMC11044950 DOI: 10.57264/cer-2023-0109] [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: 06/27/2023] [Accepted: 01/31/2024] [Indexed: 03/23/2024] Open
Abstract
Aim: To determine the effectiveness of bilayered living cellular construct (BLCC) versus a fetal bovine collagen dressing (FBCD) in pressure injuries (PRIs). Methods: A real-world data study was conducted on 1352 PRIs analyzed digitally. 1046 and 306 PRIs were treated with BLCC and FBCD, respectively. Results: Cox healing for BLCC (n = 1046) was significantly greater (p < 0.0001) at week 4 (13 vs 7%), 8 (29 vs 17%), 12 (42 vs 27%), 24 (64 vs 45%), and 36 (73 vs 56%). The probability of healing increased by 66%, (hazard ratio = 1.66 [95% CI (1.38, 2.00)]; p < 0.0001. Time to healing was 162 days for FBCD and 103 days for BLCC showing a 36% reduction in time to healing with BLCC; (p < 0.0001). Conclusion: BLCC significantly improved healing of PRIs versus FBCD.
Collapse
Affiliation(s)
- Michael L Sabolinski
- Sabolinski LLC, Managing Member, Department of Medicine, Franklin, MA 02038, USA
| | - Tad Archambault
- Virtu Stat Ltd., Department of Statistics, North Wales, PA 19454, USA
| |
Collapse
|
39
|
Vogelberg C, Klimek L, Kruppert S, Becker S. Long-term effects of pollen allergoid tyrosine-adsorbed subcutaneous immunotherapy on allergic rhinitis and asthma. Clin Exp Allergy 2024; 54:253-264. [PMID: 38146840 DOI: 10.1111/cea.14444] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 11/28/2023] [Accepted: 11/30/2023] [Indexed: 12/27/2023]
Abstract
BACKGROUND Allergen immunotherapy (AIT) may have a long-term disease-modifying effect. The aim of this study was to demonstrate the long-term effects of pollen allergoid tyrosine-adsorbed subcutaneous AIT on allergic rhinitis (AR) and asthma (AA) in clinical practice. METHODS This retrospective study, funded by an AIT manufacturer, analysed the impact of AIT on AR progression and onset of need for AA medication, using a German database covering ~35% of national prescriptions during 2008-2020. Anonymized prescription data of AR patients aged 5-65 years treated with grass or tree pollen AIT between 2009 and 2013 and followed for at least 2 years after AIT cessation were compared with matched control patients with seasonal AR. RESULTS 181,496 patients received AIT prescriptions. 5959 fulfilled the inclusion criteria. The median AIT treatment duration was 1092 days and the follow-up duration was 6.4 years. Less patients treated with AIT received prescriptions for symptomatic AR medication in the follow-up versus controls (AIT: OR: 0.37; 95% Confidence Interval (CI) 0.34, 0.40; p < .001, tyrosine-adsorbed AIT: OR: 0.27; 95% CI 0.20, 0.35 p < .001). Less asthmatic patients under AIT received prescriptions for AA medications versus controls (AIT: OR: 0.48; 95% CI 0.41, 0.55; p < .001, tyrosine-adsorbed AIT: OR: 0.48; 95% CI 0.29, 0.79; p = .004). AR and AA medication prescriptions for AIT patients were reduced in the follow-up versus baseline and controls (AIT: AR: 20.0%; 1.5 vs. 0.2 prescriptions; AA: 29.1%; 2.0 vs. 0.6 prescriptions, p < .001; tyrosine-adsorbed AIT: AR: 24.2%, 1.4 vs. 0.2 prescriptions; AA: 35.6%, 2.1 vs. 0.6 prescriptions, p < .001). The probability of AA medication onset in non-asthmatic patients during follow-up was reduced for AIT patients compared to controls (OR: 0.77, 95% CI 0.66, 0.90; p = .001). All endpoints were significant for children/adolescents and adults in stratified analyses. CONCLUSIONS We found evidence for long-term effects up to 9.5 years for tyrosine-adsorbed AIT.
Collapse
Affiliation(s)
- Christian Vogelberg
- Department of Pediatric Pneumology and Allergology, Faculty of Medicine and University Hospital Carl Gustav Carus Dresden, Technische Universität Dresden, Dresden, Germany
| | - Ludger Klimek
- Center for Rhinology and Allergy, Wiesbaden, Germany
| | | | - Sven Becker
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Tübingen, Tübingen, Germany
| |
Collapse
|
40
|
Osborne V, Goodin A, Brown J, Winterstein AG, Bate A, Cohet C, Pont L, Moeny D, Klungel O, Pinheiro S, Seeger J, Chan KA, Edlavitch S, Tilson H, Layton D. Updated core competencies in pharmacoepidemiology to inform contemporary curricula and training for academia, government, and industry. Pharmacoepidemiol Drug Saf 2024; 33:e5789. [PMID: 38629216 DOI: 10.1002/pds.5789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 03/07/2024] [Accepted: 03/22/2024] [Indexed: 04/19/2024]
Abstract
PURPOSE The first paper to specify the core content of pharmacoepidemiology as a profession was published by an ISPE (International Society for Pharmacoepidemiology) workgroup in 2012 (Jones JK et al. PDS 2012; 21[7]:677-689). Due to the broader and evolving scope of pharmacoepidemiology, ISPE considers it important to proactively identify, update and expand the list of core competencies to inform curricula of education programs; thus, better positioning pharmacoepidemiologists across academic, government (including regulatory), and industry positions. The aim of this project was to update the list of core competencies in pharmacoepidemiology. METHODS To ensure applicability of findings to multiple areas, a working group was established consisting of ISPE members with positions in academia, industry, government, and other settings. All competencies outlined by Jones et al. were extracted from the initial manuscript and presented to the working group for review. Expert-based judgments were collated and used to identify consensus. It was noted that some competencies could contribute to multiple groups and could be directly or indirectly related to a group. RESULTS Five core domains were proposed: (1) Epidemiology, (2) Clinical Pharmacology, (3) Regulatory Science, (4) Statistics and data science, and (5) Communication and other professional skills. In total, 55 individual competencies were proposed, of which 25 were new competencies. No competencies from the original work were dropped but aggregation or amendments were made where considered necessary. CONCLUSIONS While many core competencies in pharmacoepidemiology have remained the same over the past 10 years, there have also been several updates to reflect new and emerging concepts in the field.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Lisa Pont
- University of Technology Sydney, Sydney, Australia
| | - David Moeny
- Food & Drug Administration, Silver Spring, USA
| | | | | | | | | | | | - Hugh Tilson
- University of North Carolina, Chapel Hill, USA
| | | |
Collapse
|
41
|
Muntner P, Hernandez RK, Kent ST, Browning JE, Gilbertson DT, Hurwitz KE, Jick SS, Lai EC, Lash TL, Monda KL, Rothman KJ, Bradbury BD, Brookhart MA. Staging and clean room: Constructs designed to facilitate transparency and reduce bias in comparative analyses of real-world data. Pharmacoepidemiol Drug Saf 2024; 33:e5770. [PMID: 38419140 DOI: 10.1002/pds.5770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 03/02/2024]
Abstract
PURPOSE We describe constructs designed to protect the integrity of the results from comparative analyses using real-world data (RWD): staging and clean room. METHODS Staging involves performing sequential preliminary analyses and evaluating the population size available and potential bias before conducting comparative analyses. A clean room involves restricted access to data and preliminary results, policies governing exploratory analyses and protocol deviations, and audit trail. These constructs are intended to allow decisions about protocol deviations, such as changes to design or model specification, to be made without knowledge of how they might affect subsequent analyses. We describe an example for implementing staging with a clean room. RESULTS Stage 1 may involve selecting a data source, developing and registering a protocol, establishing a clean room, and applying inclusion/exclusion criteria. Stage 2 may involve attempting to achieve covariate balance, often through propensity score models. Stage 3 may involve evaluating the presence of residual confounding using negative control outcomes. After each stage, check points may be implemented when a team of statisticians, epidemiologists and clinicians masked to how their decisions may affect study outcomes, reviews the results. This review team may be tasked with making recommendations for protocol deviations to address study precision or bias. They may recommend proceeding to the next stage, conducting additional analyses to address bias, or terminating the study. Stage 4 may involve conducting the comparative analyses. CONCLUSIONS The staging and clean room constructs are intended to protect the integrity and enhance confidence in the results of analyses of RWD.
Collapse
Affiliation(s)
- Paul Muntner
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Rohini K Hernandez
- Center for Observational Research, Amgen Inc., Thousand Oaks, California, USA
| | - Shia T Kent
- Center for Observational Research, Amgen Inc., Thousand Oaks, California, USA
| | - James E Browning
- Center for Observational Research, Amgen Inc., Thousand Oaks, California, USA
| | - David T Gilbertson
- Chronic Disease Research Group, Hennepin Healthcare Research Institute, Minneapolis, Minnesota, USA
| | | | - Susan S Jick
- Boston Collaborative Drug Surveillance Program, Boston University School of Public Health, Boston, Massachusetts, USA
| | - Edward C Lai
- School of Pharmacy, Institute of Clinical Pharmacy and Pharmaceutical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Timothy L Lash
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Keri L Monda
- Center for Observational Research, Amgen Inc., Thousand Oaks, California, USA
| | - Kenneth J Rothman
- RTI Health Solutions, Research Triangle Institute, Research Triangle Park, North Carolina, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, Massachusetts, USA
| | - Brian D Bradbury
- Center for Observational Research, Amgen Inc., Thousand Oaks, California, USA
| | - M Alan Brookhart
- Department of Population Health Sciences, Duke University, Durham, North Carolina, USA
| |
Collapse
|
42
|
Chung WK, Huh KY, Park J, Oh J, Yu KS. Establishment of Advanced Regulatory Innovation for Clinical Trials Transformation (ARICTT): a multi-stakeholder public-private partnership-based organization to accelerate the transformation of clinical trials. Transl Clin Pharmacol 2024; 32:30-40. [PMID: 38586121 PMCID: PMC10990728 DOI: 10.12793/tcp.2024.32.e1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 01/09/2024] [Accepted: 01/12/2024] [Indexed: 04/09/2024] Open
Abstract
Clinical trials have evolved with digital technologies and tend towards patient-centricity. A multi-stakeholder approach is needed to address the emerging complexities in clinical trials. In particular, the introduction of digital technologies and an emphasis on patient-centricity are the major trends in clinical trials. In response, we established a public-private partnership-based organization named Advanced Regulatory Innovation for Clinical Trials Transformation (ARICTT). Eleven organizations in total, from academia, industry, and regulatory agencies, participate in ARICTT. Based on multi-stakeholder collaboration from academia, industry, and government/regulatory bodies, we collected and prioritized current topics in clinical trials based on an internal survey. We established a three-year roadmap with axes that were termed trend, goal, structure, theme, topic, and method. In addition, we planned the development of recommendations based on real-world cases with feasibility studies. We developed appropriate organizational structure to fulfill the roadmap of ARICTT. The selected topics were decentralized clinical trials during the first year, followed by the three topics that were awarded the highest priority according to the internal survey: advances in the informed consent process, supporting sites using digital technology, and an effective recruitment strategy. We developed a case-based recommendation paper presenting an overview of the regulatory landscape and practical considerations with explanatory cases. We also designed and conducted fully decentralized trials to evaluate considerations in real-world settings for the selected topics. Overall engagement and communication were supported by the online platform and annual symposiums. In conclusion, we established a multi-stakeholder, public-private partnership-based organization to accelerate the transformation of clinical trials.
Collapse
Affiliation(s)
- Woo Kyung Chung
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Korea
| | - Ki Young Huh
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Korea
| | - Jiyeon Park
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Korea
| | - Jaeseong Oh
- Department of Pharmacology, Jeju National University College of Medicine, Jeju, Korea
| | - Kyung-Sang Yu
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Korea
| |
Collapse
|
43
|
Thorlund K, Duffield S, Popat S, Ramagopalan S, Gupta A, Hsu G, Arora P, Subbiah V. Quantitative bias analysis for external control arms using real-world data in clinical trials: a primer for clinical researchers. J Comp Eff Res 2024; 13:e230147. [PMID: 38205741 PMCID: PMC10945419 DOI: 10.57264/cer-2023-0147] [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: 09/27/2023] [Accepted: 12/14/2023] [Indexed: 01/12/2024] Open
Abstract
Development of medicines in rare oncologic patient populations are growing, but well-powered randomized controlled trials are typically extremely challenging or unethical to conduct in such settings. External control arms using real-world data are increasingly used to supplement clinical trial evidence where no or little control arm data exists. The construction of an external control arm should always aim to match the population, treatment settings and outcome measurements of the corresponding treatment arm. Yet, external real-world data is typically fraught with limitations including missing data, measurement error and the potential for unmeasured confounding given a nonrandomized comparison. Quantitative bias analysis (QBA) comprises a collection of approaches for modelling the magnitude of systematic errors in data which cannot be addressed with conventional statistical adjustment. Their applications can range from simple deterministic equations to complex hierarchical models. QBA applied to external control arm represent an opportunity for evaluating the validity of the corresponding comparative efficacy estimates. We provide a brief overview of available QBA approaches and explore their application in practice. Using a motivating example of a comparison between pralsetinib single-arm trial data versus pembrolizumab alone or combined with chemotherapy real-world data for RET fusion-positive advanced non-small cell lung cancer (aNSCLC) patients (1-2% among all NSCLC), we illustrate how QBA can be applied to external control arms. We illustrate how QBA is used to ascertain robustness of results despite a large proportion of missing data on baseline ECOG performance status and suspicion of unknown confounding. The robustness of findings is illustrated by showing that no meaningful change to the comparative effect was observed across several 'tipping-point' scenario analyses, and by showing that suspicion of unknown confounding was ruled out by use of E-values. Full R code is also provided.
Collapse
Affiliation(s)
- Kristian Thorlund
- Dept. Health Research Methods, Evidence, & Impact, McMaster University, ON, Canada
| | | | - Sanjay Popat
- Royal Marsden Hospital, Imperial College, London, UK
| | | | | | | | - Paul Arora
- Dalla Lana School of Public Health, University of Toronto, ON, Canada
| | | |
Collapse
|
44
|
Jonéus P, Johansson P, Langenskiöld S. Novel hormonal therapy versus standard of care-A registry-based comparative effectiveness evaluation for mCRPC-patients. PLoS One 2024; 19:e0290833. [PMID: 38354183 PMCID: PMC10866493 DOI: 10.1371/journal.pone.0290833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 08/15/2023] [Indexed: 02/16/2024] Open
Abstract
BACKGROUND This paper presents results from one of the few comparative effectiveness evaluations of novel antiandrogen medications (NHT) against standard of care (SoC) for patients suffering from metastatic castrate-resistant prostate cancer (mCRPC). METHODS The design and the analysis are published in a protocol before accessing outcome data. Two groups of patients are balanced on hundreds of important covariates measured before the prostate cancer diagnosis and up to the date of the prescription. While the design yields balance on the observed covariates, one cannot discard the possibility that unobserved confounders are not balanced. The unconfoundedness assumption is assessed by estimating placebo regressions on two health measures, not included in the design but added together with the outcome data after protocol publication. RESULTS We find a substantial (64 percent) increase in mortality for patients prescribed with NHT rather than SoC. However, based on the results from one of the two placebo regressions, we cannot rule out that the difference in mortality may be due to confounding. Using a bounding strategy of the effect, we can, however, rule out that NHT reduces mortality compared to SoC. Under an empirical valid assumption that most mCRPC patients who die suffer from bone metastases, we have a strong indication of increased skeleton-related events in patients if prescribed NHT against SoC. CONCLUSIONS Generally, the SoC for this group of patients is docetaxel. Given the substantially higher costs of many of the NHT, the finding of no positive effects from NHT on both mortality and SRE is important. More comparative studies, including studies analysing quality of life outcomes, are thus needed.
Collapse
Affiliation(s)
- Paulina Jonéus
- Department of Statistics, Uppsala University, Uppsala, Sweden
| | - Per Johansson
- Department of Statistics, Uppsala University, Uppsala, Sweden
- Centre for Health-Economic Research, Uppsala University, Uppsala, Sweden
- YMSC, Tsinghua University, Beijing, China
| | - Sophie Langenskiöld
- Centre for Health-Economic Research, Uppsala University, Uppsala, Sweden
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
45
|
Gross-Goupil M, Bodnar L, Campbell MT, Michael A, Venugopal B, Żołnierek J, Dutailly P, Procopio G, Albiges L. Cabozantinib in the Routine Management of Renal Cell Carcinoma: A Systematic Literature Review of Real-World Evidence. Clin Genitourin Cancer 2024; 22:84-97. [PMID: 38101983 DOI: 10.1016/j.clgc.2023.11.001] [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: 10/05/2023] [Revised: 11/01/2023] [Accepted: 11/02/2023] [Indexed: 12/17/2023]
Abstract
Real-world cabozantinib use has increased since its approval to treat patients with advanced renal cell carcinoma (RCC) in 2016. We reviewed cabozantinib use in real-world clinical practice and compared outcomes with pivotal cabozantinib randomized control trials (RCTs). This PRISMA-standard systematic literature review evaluated real-world effectiveness and tolerability of cabozantinib in patients with RCC (PROSPERO registration: CRD42021245854). Systematic MEDLINE, Embase, and Cochrane database searches were conducted on November 2, 2022. Eligible publications included ≥ 20 patients with RCC receiving cabozantinib. After double-screening for eligibility, standardized data were abstracted, qualitatively summarized, and assessed for risk of bias using the Newcastle-Ottawa Scale. Of 353 screened publications, 41 were included, representing approximately 11,000 real-world patients. Most publications reported cabozantinib monotherapy cohort studies (40/41) of retrospective (39/41) and multicenter (32/41) design; most included patients from North America and/or Europe (30/41). Baseline characteristics were demographically similar between real-world and pivotal RCT populations, but real-world populations showed greater variation in prevalence of prior nephrectomy, multiple-site/brain metastasis, and nonclear-cell RCC histology. Cabozantinib activity was reported across real-world treatment lines and tumor types. Overall survival, progression-free survival, and objective response rate values from pivotal RCTs were within the ranges reported for equivalent outcomes across real-world studies. Common real-world grade ≥ 3 adverse events were consistent with those in pivotal RCTs (fatigue, palmar-plantar erythrodysesthesia syndrome, diarrhea, hypertension), but less frequent. No new tolerability concerns were identified. Real-world RCC survival outcomes for cabozantinib monotherapy were broadly consistent with pivotal RCTs, despite greater heterogeneity in real-world populations.
Collapse
Affiliation(s)
| | - Lubomir Bodnar
- University of Natural Sciences and Humanities in Siedlce, Institute of Health Sciences, Siedlce, Poland
| | | | - Agnieszka Michael
- University of Surrey, School of Biosciences and Medicine, Guildford, UK
| | - Balaji Venugopal
- Beatson West of Scotland Cancer Centre and University of Glasgow, Glasgow, UK
| | | | | | | | | |
Collapse
|
46
|
Wicherski J, Haenisch B. [The application of real-world evidence in drug regulatory decision-making]. Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz 2024; 67:149-154. [PMID: 38214723 PMCID: PMC10834571 DOI: 10.1007/s00103-023-03830-0] [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: 08/28/2023] [Accepted: 12/18/2023] [Indexed: 01/13/2024]
Abstract
Drug regulation is a system to support and protect public health. Drugs with market access must be effective, safe and of high quality. Therefore, drug regulatory decision-making by the competent authorities is made on a scientific basis. Real-world evidence (RWE) from real-world data (RWD) has so far predominantly been taken into account in a supportive manner in drug regulatory decision-making with regard to drug safety after marketing authorisation. The extensive potential of RWE for regulatory decision-making processes along the entire product life cycle has been increasingly used and further examined in recent years.This article provides an overview of current applications of RWE in drug regulatory decision-making processes. The potentials of RWE along with the hurdles to be addressed are described and examples of current projects on RWE research for drug regulation are given. The work is based on current international literature as well as examples from international and European initiatives and regulatory practice, which aim to support an increased use of RWD/RWE in regulatory decision-making processes. In order to be able to utilise the potential of RWE even more in the future, it is important to make relevant RWD sources more readily available through research projects and initiatives, to further develop evaluative methods and to establish the significance of RWE.
Collapse
Affiliation(s)
- Julia Wicherski
- Bundesinstitut für Arzneimittel und Medizinprodukte (BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175, Bonn, Deutschland
| | - Britta Haenisch
- Bundesinstitut für Arzneimittel und Medizinprodukte (BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175, Bonn, Deutschland.
- Deutsches Zentrum für Neurodegenerative Erkrankungen e. V. (DZNE), Bonn, Deutschland.
- Zentrum für Translationale Medizin, Universität Bonn, Bonn, Deutschland.
| |
Collapse
|
47
|
Valcarcel B, Schonfeld SJ, Meyer CL, Brunson A, Cooley JJP, Abrahão R, Wun T, Auletta JJ, Gadalla SM, Engels E, Albert PS, Spellman SR, Rizzo JD, Shaw BE, Muffly L, Keegan THM, Morton LM. Comparison of Vital Status, Cause of Death, and Follow-Up after Hematopoietic Cell Transplantation in Linked Center for International Blood and Marrow Transplant Research and California Cancer Registry Data, 1991 to 2018. Transplant Cell Ther 2024; 30:239.e1-239.e11. [PMID: 37981238 PMCID: PMC10872486 DOI: 10.1016/j.jtct.2023.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 11/09/2023] [Accepted: 11/10/2023] [Indexed: 11/21/2023]
Abstract
Assessing outcomes following hematopoietic cell transplantation (HCT) poses challenges due to the necessity for systematic and often prolonged patient follow-up. Linking the HCT database of the Center for International Blood and Marrow Transplant Research (CIBMTR) with cancer registry data may improve long-term outcome ascertainment, but the reliability of mortality data in death certificates from cancer registries among HCT recipients remains unknown. We compared the classification of vital status and primary cause of death (COD), as well as the length of follow-up between the CIBMTR and California Cancer Registry (CCR) to assess the possibility of supplementing the CIBMTR with cancer registry data. This retrospective study leveraged a linked CIBMTR-CCR dataset. We included patients who were California residents at the time of HCT and received a first allogeneic (allo) or autologous (auto) HCT for a hematologic malignancy diagnosed during 1991-2016. Follow-up was through 2018. We analyzed 18,450 patients (alloHCT, n = 8232; autoHCT, n = 10,218). The Vital status agreement was 97.7% for alloHCT and 97.2% for autoHCT. Unknown COD was higher in CIBMTR (12.9%) than in CCR (1.6%). After excluding patients with unknown COD information, the overall agreement of primary COD (cancer versus noncancer) was 53.7% for alloHCT and 83.2% for autoHCT. This agreement was lower within the first 100 days post-HCT (alloHCT, 31.0%; autoHCT, 54.6%). Compared with CIBMTR, deaths due to cancer were higher in CCR (alloHCT, 90.0%; autoHCT, 90.1% versus alloHCT, 47.3%; autoHCT, 82.5% in CIBMTR). CIBMTR reports more frequently noncancer-related deaths, including graft-versus-host disease and infections. The cumulative incidence of cancer-specific mortality at 20 years differed, particularly for alloHCT (CCR, 53.7%; CIBMTR, 27.6%). The median follow-up among alive patients was longer in CCR (alloHCT, 6.0 years; autoHCT, 4.7 years) than in CIBMTR (alloHCT, 5.0 years; autoHCT, 3.8 years). Our findings highlight the completeness of vital status data in CIBMTR but reveal substantial disagreement in primary COD. Consequently, caution is required when interpreting HCT studies that use only death certificates to estimate cause-specific mortality outcomes. Improving the accuracy of COD registration and follow-up completeness by developing communication pathways between cancer registries and hospital-based cohorts may enhance our understanding of late effects and long-term outcomes among HCT survivors.
Collapse
Affiliation(s)
- Bryan Valcarcel
- Radiation Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland.
| | - Sara J Schonfeld
- Radiation Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland
| | - Christa L Meyer
- Center for International Blood and Marrow Transplant Research, National Marrow Donor Program/Be The Match, Minneapolis, Minnesota
| | - Ann Brunson
- Center for Oncology Hematology Outcomes Research and Training, Division of Hematology and Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, California
| | - Julianne J P Cooley
- California Cancer Reporting and Epidemiologic Surveillance Program, University of California Davis Comprehensive Cancer Center, Sacramento, California
| | - Renata Abrahão
- Center for Oncology Hematology Outcomes Research and Training, Division of Hematology and Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, California
| | - Ted Wun
- Center for Oncology Hematology Outcomes Research and Training, Division of Hematology and Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, California
| | - Jeffery J Auletta
- Center for International Blood and Marrow Transplant Research, National Marrow Donor Program/Be The Match, Minneapolis, Minnesota; Divisions of Hematology/Oncology/BMT and Infectious Diseases, Nationwide Children's Hospital, Columbus, Ohio
| | - Shahinaz M Gadalla
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland
| | - Eric Engels
- Infections and Immunoepidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland
| | - Paul S Albert
- Biostatistics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland
| | - Stephen R Spellman
- Center for International Blood and Marrow Transplant Research, National Marrow Donor Program/Be The Match, Minneapolis, Minnesota
| | - J Douglas Rizzo
- Center for International Blood and Marrow Transplant Research, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Bronwen E Shaw
- Center for International Blood and Marrow Transplant Research, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Lori Muffly
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, California
| | - Theresa H M Keegan
- Center for Oncology Hematology Outcomes Research and Training, Division of Hematology and Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, California
| | - Lindsay M Morton
- Radiation Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland
| |
Collapse
|
48
|
Venkatraman V, Bharmi R, Coletti F, Gellad ZF, Lempel N, Amit R, Blank N, Brown J, Kumar C, Fishman M, Vallejo R, Datta D, Reeve BB, Chakravarthy K, Sharan AD, Lad SP. Real World Characterization of Chronic Pain, Success Rates and Implant Rates: Evidence from a Digital Health Platform of Patients Undergoing Spinal Cord Stimulation Evaluations. THE JOURNAL OF PAIN 2023; 24:2228-2239. [PMID: 37541604 PMCID: PMC11225030 DOI: 10.1016/j.jpain.2023.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 06/17/2023] [Accepted: 07/06/2023] [Indexed: 08/06/2023]
Abstract
Spinal cord stimulation is an effective treatment for those experiencing chronic back and leg pain but requires a temporary evaluation period (SCSeval) before permanent implantation. We present real-world data from 7,000 patients who underwent SCSeval while utilizing a mobile digital health platform for education, feedback, and outcomes collection during their surgical journey. We analyzed preoperative patient demographics, characterized patient pain profiles using the patient-reported outcomes measurement information system-29 surveys, and calculated the rates of conversion from temporary to permanent spinal cord stimulation (SCS) implantation. Between August 1, 2021, and March 2, 2023, 7,000 patients (mean age 59.1, 59.6% female) underwent SCSeval procedures while utilizing a mobile application. Patients commonly experienced aching, sharp, stabbing, tingling, numb, and burning pain. Patients had tried multiple prior therapies and wanted to reduce their use of opioids and pain medications. Overall, 90.1% of the patients had a successful SCSeval, and 80.4% of those converted to permanent implant, with the highest rates among those who underwent SCSeval in a hospital setting. There was a significant improvement in all domains of pain as evaluated by pre and postoperative patient-reported outcomes measurement information system-29 surveys. This study supports the use of digital health technology as part of the SCS journey to improve the patient experience and allow for robust patient-reported outcomes collection. The overall rate of SCSeval to permanent SCS in our study of 72.4% was higher than national rates of 64%, suggesting that an app may allow clinicians to better quantify changes in chronic pain and provide more insight into choosing to implant SCS permanently. PERSPECTIVE: This article presents real-world evidence from a digital health platform for therapy education and outcomes collection from patients undergoing spinal cord stimulation evaluation procedures. Such tools could allow for better pain characterization and allow for more nuanced tracking of patient outcomes among those with chronic pain.
Collapse
Affiliation(s)
- Vishal Venkatraman
- Department of Neurosurgery, Duke University School of Medicine, Durham, NC, USA
| | | | | | - Ziad F. Gellad
- Division of Gastroenterology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Noa Lempel
- Nutrino Health, Medtronic, Tel Aviv, Israel
| | - Roy Amit
- Nutrino Health, Medtronic, Tel Aviv, Israel
| | | | - Jason Brown
- Medtronic Neuromodulation, Minneapolis, MN, USA
| | | | - Michael Fishman
- Center for Interventional Pain and Spine, Exton, Pennsylvania, USA
| | | | | | - Bryce B. Reeve
- Department of Population Health Sciences, Duke University School of Medicine, Durham, NC, USA
| | - Krishnan Chakravarthy
- Coastal Pain and Spinal Diagnostics, San Diego, CA, USA
- Department of Anesthesiology, UC San Diego School of Medicine, La Jolla, CA, USA
| | | | - Shivanand P. Lad
- Department of Neurosurgery, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
49
|
Dupuis-Girod S, Rivière S, Lavigne C, Fargeton AE, Gilbert-Dussardier B, Grobost V, Leguy-Seguin V, Maillard H, Mohamed S, Decullier E, Roux A, Bernard L, Saurin JC, Saroul N, Faure F, Cartier C, Altwegg R, Laccourreye L, Oberti F, Beaudoin M, Dhelens C, Desvignes C, Azzopardi N, Paintaud G, Hermann R, Chinet T. Efficacy and safety of intravenous bevacizumab on severe bleeding associated with hemorrhagic hereditary telangiectasia: A national, randomized multicenter trial. J Intern Med 2023; 294:761-774. [PMID: 37592715 DOI: 10.1111/joim.13714] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/19/2023]
Abstract
BACKGROUND Bevacizumab-a humanized monoclonal antibody-has been widely used to treat patients with hereditary hemorrhagic telangiectasia (HHT), but no randomized trial has yet been conducted. METHODS This study is a double-blind multicenter randomized phase 2 trial with a 1:1 active-treatment-to-placebo ratio. We included patients over the age of 18 with a confirmed diagnosis and the need for at least four red blood cell (RBC) units transfused in the 3 months before study enrollment. Bevacizumab was administered at a dose of 5 mg/kg every 14 days with a total of six injections. The primary efficacy criterion was a decrease of at least 50% in the cumulative number of RBC units transfused in a 3-month period before and after treatment. RESULTS A total of 24 patients (12 in each group) were included and randomized at 4 different centers. In intention-to-treat analysis, 63.6% of patients (7/11) in the bevacizumab group versus 33.3% of patients (4/12) in the placebo group decreased the number of blood transfusions by at least 50% (p = 0.22). Hemoglobin levels significantly improved at 6 months in the bevacizumab versus placebo group (p = 0.02). The pharmacokinetics study revealed that patients with high exposure to bevacizumab had a significant decrease in RBC transfusions (p = 0.03). Fifty-nine adverse events were observed, 34 in the placebo arm versus 25 in the bevacizumab arm. CONCLUSION Though the present trial was underpowered, patients with HHT receiving bevacizumab required numerically fewer red blood cell transfusions than those receiving placebo, particularly those with high exposure.
Collapse
Affiliation(s)
- Sophie Dupuis-Girod
- Service de Génétique et centre de référence de la maladie de Rendu-Osler, Hôpital Femme-Mère-Enfants, Hospices Civils de Lyon, Bron, France
- Inserm, CEA, Laboratory Biology of Cancer and Infection, Université Grenoble Alpes, Grenoble, France
| | - Sophie Rivière
- Service de Médecine Interne A, Centre Hospitalier Universitaire, Montpellier, France
| | - Christian Lavigne
- Service de médecine interne-Immunologie clinique, CHU d'Angers, Angers cedex 09, France
| | - Anne-Emmanuelle Fargeton
- Service de Génétique et centre de référence de la maladie de Rendu-Osler, Hôpital Femme-Mère-Enfants, Hospices Civils de Lyon, Bron, France
| | | | - Vincent Grobost
- Service de Médecine Interne CHU Estaing, Clermont-Ferrand University Hospital, Clermont-Ferrand, France
| | | | - Hélène Maillard
- CHU Lille, Service de Médecine Interne et Immunologie Clinique, Lille, France
| | - Shirine Mohamed
- Département de Médecine interne et Immunologie Clinique, CHRU BRABOIS, Vandoeuvre-lès-Nancy, France
| | - Evelyne Decullier
- Hospices Civils de Lyon, Pôle Santé Publique, Lyon, France
- Faculté de médecine, Université Lyon 1, Lyon, France
| | - Adeline Roux
- Hospices Civils de Lyon, Pôle Santé Publique, Lyon, France
- Faculté de médecine, Université Lyon 1, Lyon, France
| | | | - Jean-Christophe Saurin
- Faculté de médecine, Université Lyon 1, Lyon, France
- Hospices Civils de Lyon, Service d'Hépato-gastroentérologie, Hôpital E. Herriot, Lyon, France
| | - Nicolas Saroul
- CHU Clermont Ferrand, Hôpital Gabriel Montpied, Service d'ORL, Clermont-Ferrand, France
| | - Frédéric Faure
- Hospices Civils de Lyon, Hôpital E. Herriot, Service d'ORL, Lyon, France
| | - Cesar Cartier
- Service d'ORL Centre Hospitalier Universitaire, Montpellier, France
| | - Romain Altwegg
- Service Hépatogastroentérologie CHU St Eloi, Montpellier, France
| | | | - Frédéric Oberti
- Service Hépatogastroentérologie, UPRES EA 3859, Faculté de médecine, CHU Angers and Laboratoire HIFIH, Angers, France
| | - Marjolaine Beaudoin
- Service de Génétique et centre de référence de la maladie de Rendu-Osler, Hôpital Femme-Mère-Enfants, Hospices Civils de Lyon, Bron, France
| | - Carole Dhelens
- Hospices Civils de Lyon, Hôpital Edouard Herriot, Pharmacie à Usage Intérieur, Lyon, France
| | - Céline Desvignes
- CHRU de Tours, Plateforme Recherche, Centre Pilote de suivi Biologique des traitements par Anticorps (CePiBAc), Tours, France
- EA 4245 Transplantation, Immunologie, Inflammation (T2I), Université de Tours, Tours, France
| | | | - Gilles Paintaud
- EA 4245 Transplantation, Immunologie, Inflammation (T2I), Université de Tours, Tours, France
- Service de Pharmacologie Médicale, CHRU de Tours, Tours, France
| | - Ruben Hermann
- Hospices Civils de Lyon, Hôpital Femme-Mère-Enfants, Service d'ORL et centre de référence de la maladie de Rendu-Osler, Bron, France
| | - Thierry Chinet
- Centre Rendu-Osler, Hôpital Ambroise Paré, Assistance Publique-Hôpitaux de Paris, Université de Versailles SQY, Boulogne, France
| |
Collapse
|
50
|
Gonzalez SA, Chirikov VV, Wang WJ, Huang X, Jamil K, Simonetto DA. Terlipressin vs Midodrine Plus Octreotide for Hepatorenal Syndrome-Acute Kidney Injury: A Propensity Score-Matched Comparison. Clin Transl Gastroenterol 2023; 14:e00627. [PMID: 37622521 PMCID: PMC10749708 DOI: 10.14309/ctg.0000000000000627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 07/24/2023] [Indexed: 08/26/2023] Open
Abstract
INTRODUCTION Evidence on the comparison of treatments for hepatorenal syndrome-acute kidney injury (HRS-AKI) in a US population is limited. An indirect comparison of terlipressin plus albumin vs midodrine and octreotide plus albumin (MO) may provide further insight into treatment efficacy. METHODS Cohorts of patients treated for HRS-AKI characterized by inclusion of patients with serum creatinine (SCr) <5 mg/dL and baseline acute-on-chronic liver failure grades 0-2 and exclusion of patients listed for transplant if model for end-stage liver disease scores ≥35 were pooled from (i) the CONFIRM and REVERSE randomized controlled trials (N = 159 meeting eligibility criteria from N = 216 overall, treated with terlipressin) and (ii) a retrospective review of medical records from 10 US tertiary hospitals (2016-2019; N = 55 treated with MO meeting eligibility criteria from N = 200 overall). The primary end point comparing the 2 cohorts was HRS reversal defined as achieving SCr ≤1.5 mg/dL at least once during the treatment. Covariate balancing propensity scoring was used to adjust for differences in baseline characteristics. RESULTS HRS-AKI reversal was achieved in 52.35% of terlipressin-treated patients compared with 20% of MO-treated patients (adjusted mean difference 32.35%, 95% confidence interval [CI] 17.40-47.30, P < 0.0001). Terlipressin-treated patients had increased overall survival (adjusted hazard ratio 0.57, 95% CI 0.35-0.93, P = 0.02) but similar transplant-free survival (adjusted hazard ratio 0.79, 95% CI 0.53-1.17, P = 0.24). Achievement of HRS-AKI reversal was associated with increased OS and TFS regardless of treatment ( P < 0.001). DISCUSSION Consistent with prior reports, terlipressin plus albumin is more effective in improving kidney function and achieving HRS-AKI reversal than MO plus albumin based on indirect comparison in a US population.
Collapse
Affiliation(s)
- Stevan A. Gonzalez
- Division of Hepatology, Annette C. and Harold C. Simmons Transplant Institute, Baylor Scott & White All Saints Medical Center, Fort Worth, Texas, USA
- Department of Medicine, Burnett School of Medicine at TCU, Fort Worth, Texas, USA
| | | | | | - Xingyue Huang
- Mallinckrodt Pharmaceuticals, Hampton, New Jersey, USA
| | - Khurram Jamil
- Mallinckrodt Pharmaceuticals, Hampton, New Jersey, USA
| | - Douglas A. Simonetto
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, USA
| |
Collapse
|