1
|
Chen J, Nie L, Lee S, Chu H, Tian H, Wang Y, He W, Jemielita T, Gruber S, Song Y, Tamura R, Tian L, Zhao Y, Chen Y, van der Laan M, Lee H. Challenges and Possible Strategies to Address Them in Rare Disease Drug Development: A Statistical Perspective. Clin Pharmacol Ther 2025. [PMID: 40079686 DOI: 10.1002/cpt.3631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 02/21/2025] [Indexed: 03/15/2025]
Abstract
Developing drugs for rare diseases presents unique challenges from a statistical perspective. These challenges may include slowly progressive diseases with unmet medical needs, poorly understood natural history, small population size, diversified phenotypes and genotypes within a disorder, and lack of appropriate surrogate endpoints to measure clinical benefits. The Real-World Evidence (RWE) Scientific Working Group of the American Statistical Association Biopharmaceutical Section has assembled a research team to assess the landscape including challenges and possible strategies to address these challenges and the role of real-world data (RWD) and RWE in rare disease drug development. This paper first reviews the current regulations by regulatory agencies worldwide and then discusses in more detail the challenges from a statistical perspective in the design, conduct, and analysis of rare disease clinical trials. After outlining an overall development pathway for rare disease drugs, corresponding strategies to address the challenges are presented. Other considerations are also discussed for generating relevant evidence for regulatory decision-making on drugs for rare diseases. The accompanying paper discusses how RWD and RWE can be used to improve the efficiency of rare disease drug development.
Collapse
Affiliation(s)
| | - Lei Nie
- US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Shiowjen Lee
- US Food and Drug Administration, Silver Spring, Maryland, USA
| | | | | | - Yan Wang
- US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Weili He
- AbbVie, North Chicago, Illinois, USA
| | | | | | - Yang Song
- Vertex Pharmaceuticals, Boston, Massachusetts, USA
| | - Roy Tamura
- University of South Florida, Tampa, Florida, USA
| | - Lu Tian
- Stanford University, Stanford, California, USA
| | - Yihua Zhao
- Flatiron Health, San Francisco, California, USA
| | - Yong Chen
- University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | - Hana Lee
- US Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
2
|
Wilson E, Leventer R, Cunningham C, de Silva MG, Hodgson J, Uebergang E. Anything is better than nothing': exploring attitudes towards novel therapies in leukodystrophy clinical trials. Orphanet J Rare Dis 2024; 19:322. [PMID: 39237961 PMCID: PMC11378604 DOI: 10.1186/s13023-024-03320-9] [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: 03/11/2024] [Accepted: 08/08/2024] [Indexed: 09/07/2024] Open
Abstract
BACKGROUND/AIM Leukodystrophies comprise a group of genetic white matter disorders that lead to progressive motor and cognitive impairment. Recent development of novel therapies has led to an increase in clinical trials for leukodystrophies. To enable recruitment of individuals with a leukodystrophy into clinical trials, clinical trial acceptability should be ascertained. We sought therefore, to identify the motivations for and barriers to clinical trial participation in addition to clinical trial features that may be of concern to individuals with a leukodystrophy and/or their carers. METHODS Adults with a leukodystrophy and parents/carers of individuals with a leukodystrophy were recruited through the Australian Leukodystrophy Registry and through online advertisements. Qualitative semi-structured interviews were used to explore participants views on what clinical trials involve, the perceived risks and benefits of clinical trials, their desire to participate in clinical trials and their personal experience with leukodystrophy. Thematic analysis of data was performed with co-coding of interview transcripts. RESULTS 5 interviews were held with parents of children with leukodystrophy, 4 with parents of adults with leukodystrophy and 3 with adults diagnosed with leukodystrophy. Motivations for clinical trial enrolment include access to potentially lifesaving novel treatments and improved prognostic outcomes. Participants were concerned about adverse clinical trial outcomes, including side effects and exacerbation of illness. Despite this, majority of participants were willing to try anything in clinical trials, demonstrating a high tolerance for first in human trials and trials utilising invasive treatment options. CONCLUSIONS Interviewees communicated a strong desire to participate in interventional clinical trials involving novel therapies. To support enrolment into future leukodystrophy clinical trials we suggest the provision of transparent information regarding clinical trial treatments, consideration of alternative trial control measures, and inclusion of treating clinicians in the trial recruitment process. Clinicians play an integral role in initiating transparent conversations regarding trial risks and adverse outcomes.
Collapse
Affiliation(s)
- Ella Wilson
- The University of Melbourne, Melbourne, VIC, Australia.
| | - Richard Leventer
- Murdoch Children's Research Institute, Melbourne, VIC, Australia
- Australian Genomics, Melbourne, VIC, Australia
- Royal Children's Hospital, Melbourne, VIC, Australia
- The University of Melbourne, Melbourne, VIC, Australia
| | - Chloe Cunningham
- Murdoch Children's Research Institute, Melbourne, VIC, Australia
- Royal Children's Hospital, Melbourne, VIC, Australia
- Victorian Clinical Genetics Services, Melbourne, VIC, Australia
- The University of Melbourne, Melbourne, VIC, Australia
| | - Michelle G de Silva
- Murdoch Children's Research Institute, Melbourne, VIC, Australia
- Victorian Clinical Genetics Services, Melbourne, VIC, Australia
- The University of Melbourne, Melbourne, VIC, Australia
| | - Jan Hodgson
- The University of Melbourne, Melbourne, VIC, Australia
| | - Eloise Uebergang
- Murdoch Children's Research Institute, Melbourne, VIC, Australia
- Australian Genomics, Melbourne, VIC, Australia
| |
Collapse
|
3
|
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
|
4
|
Signorovitch J, Zhang J, Brown D, Dunnmon P, Xiu L, Done N, Hsu K, Barbachano Y, Lousada I. Pathway for Development and Validation of Multi-domain Endpoints for Amyloid Light Chain (AL) Amyloidosis. Ther Innov Regul Sci 2024; 58:600-609. [PMID: 38632158 PMCID: PMC11169055 DOI: 10.1007/s43441-024-00641-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 03/08/2024] [Indexed: 04/19/2024]
Abstract
Immunoglobin light chain (AL) amyloidosis is a rare disease in which a plasma cell dyscrasia leads to deposition of insoluble amyloid fibrils in multiple organs. To facilitate development of new therapies for this heterogenous disease, a public-private partnership was formed between the nonprofit Amyloidosis Research Consortium and the US Food and Drug Administration Center for Drug Evaluation and Research. In 2020, the Amyloidosis Forum launched an initiative to identify clinical trial endpoints and analytic strategies across affected organ systems and life impacts via specialized working groups. This review summarizes the proceedings of the Statistical Group and proposes a pathway for development and validation of multi-domain endpoints (MDEs) for potential use in AL amyloidosis clinical trials. Specifically, drawing on candidate domain-specific endpoints recommended by each organ-specific working group, different approaches to constructing MDEs were considered. Future studies were identified to assess the validity, meaningfulness and performance of MDEs through use of natural history and clinical trial data. Ultimately, for drug development, the context of use in a regulatory evaluation, the specific patient population, and the investigational therapeutic mechanism should drive selection of appropriate endpoints. MDEs for AL amyloidosis, once developed and validated, will provide important options for advancing patient-focused drug development in this multi-system disease.
Collapse
Affiliation(s)
| | - Jialu Zhang
- US Food and Drug Administration, Silver Spring, USA
| | - David Brown
- UK Medicines & Healthcare Products Regulatory Agency, London, UK
| | | | - Liang Xiu
- Janssen Research & Development, Raritan, USA
| | | | - Kristen Hsu
- Amyloidosis Research Consortium, 320 Nevada Street, Suite 210, Newton, MA, 02460, USA
| | | | - Isabelle Lousada
- Amyloidosis Research Consortium, 320 Nevada Street, Suite 210, Newton, MA, 02460, USA.
| |
Collapse
|
5
|
Bouwman L, Sepodes B, Leufkens H, Torre C. Trends in orphan medicinal products approvals in the European Union between 2010-2022. Orphanet J Rare Dis 2024; 19:91. [PMID: 38413985 PMCID: PMC10900541 DOI: 10.1186/s13023-024-03095-z] [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: 11/13/2023] [Accepted: 02/21/2024] [Indexed: 02/29/2024] Open
Abstract
BACKGROUND Over the last twenty years of orphan drug regulation in Europe, the regulatory framework has increased its complexity, with different regulatory paths and tools engineered to facilitate the innovation and accelerate approvals. Recently, the proposal of the new Pharmaceutical Legislation for the European Union, which will replace at least three Regulations and one Directive, was released and its new framework is raising many questions. The aim of this study was to present a characterisation of the Orphan Medicinal Products (OMPs) authorised by the European Commission (EC), between 2010 and 2022, looking into eighteen variables, contributing to the ongoing discussion on the proposal and implementation of the new Pharmaceutical Legislation proposed. METHODS Data of the OMPs identified and approved between 2010 and 2022 were extracted from the European Public Assessment Reports (EPARs) produced by the European Medicines Agency. Information regarding legal basis of the application, applicant, protocol assistance received, type of authorization, registration status, type of molecule, ATC code, therapeutic area, target age, disease prevalence, number of pivotal clinical trials supporting the application, clinical trial designs, respective efficacy endpoints and number of patients enrolled in the pivotal clinical trials were extracted. A descriptive statistical analysis was applied. RESULTS We identified 192 OMPs approved in the period between 2010 and 2022. 89% of the OMPs have legal basis of "full application". 86% of the sponsors received protocol assistance whereas 64% of the MAA benefited from the accelerated assessment. 53% of the active substances are small molecules; about 1 in 5 molecules are repurposed. 40% of the OMPs have oncological therapeutic indications and 56% of the OMPs are intended to treat only adults. 71% of the products were approved based on a single pivotal trial. CONCLUSIONS This analysis of OMPs approved between 2010 and 2022 shows that a shift has occurred in the rare disease medicine development space. Through the period studied we observe an increase of non-small molecules approved, accelerated assessment received and non-standard MA's granted.
Collapse
Affiliation(s)
- Luísa Bouwman
- Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal.
- Laboratory of Systems Integration Pharmacology, Clinical and Regulatory Science, Research Institute for Medicines of the University of Lisbon (iMED.ULisboa), Lisbon, Portugal.
| | - Bruno Sepodes
- Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
- Laboratory of Systems Integration Pharmacology, Clinical and Regulatory Science, Research Institute for Medicines of the University of Lisbon (iMED.ULisboa), Lisbon, Portugal
| | - Hubert Leufkens
- Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, The Netherlands
| | - Carla Torre
- Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
- Laboratory of Systems Integration Pharmacology, Clinical and Regulatory Science, Research Institute for Medicines of the University of Lisbon (iMED.ULisboa), Lisbon, Portugal
| |
Collapse
|
6
|
Jandhyala R. Neutral theory: applicability and neutrality of clinical study endpoints where a disease-specific instrument is available. BMC Med Res Methodol 2023; 23:121. [PMID: 37210484 DOI: 10.1186/s12874-023-01947-z] [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: 06/13/2022] [Accepted: 05/11/2023] [Indexed: 05/22/2023] Open
Abstract
BACKGROUND There is a pressing need to improve the accuracy of rare disease clinical study endpoints. Neutral theory, first described here, can be used to assess the accuracy of endpoints and improve their selection in rare disease clinical studies, reducing the risk of patient misclassification. METHODS Neutral theory was used to assess the accuracy of rare disease clinical study endpoints and the resulting probability of false positive and false negative classifications at different disease prevalence rates. Search strings were extracted from the Orphanet Register of Rare Diseases using a proprietary algorithm to conduct a systematic review of studies published until January 2021. Overall, 11 rare diseases with one disease-specific disease severity scale (133 studies) and 12 rare diseases with more than one disease-specific disease severity scale (483 studies) were included. All indicators from clinical studies were extracted, and Neutral theory was used to calculate their match to disease-specific disease severity scales, which were used as surrogates for the disease phenotype. For those with more than one disease-severity scale, endpoints were compared with the first disease-specific disease severity scale and a composite of all later scales. A Neutrality score of > 1.50 was considered acceptable. RESULTS Around half the clinical studies for half the rare diseases with one disease-specific disease severity score (palmoplantar psoriasis, achalasia, systemic lupus erythematosus, systemic sclerosis and Fournier's gangrene) met the threshold for an acceptable match to the disease phenotype, one rare disease (Guillain-Barré syndrome) had one study with an acceptable match, and four diseases (Behcet's syndrome, Creutzfeldt-Jakob disease, atypical hemolytic uremic syndrome and Prader-Willi syndrome) had no studies. Clinical study endpoints in almost half the rare diseases with more than one disease-specific DSS (acromegaly, amyotrophic lateral sclerosis, cystic fibrosis, Fabry disease and juvenile rheumatoid arthritis) were a better match to the composite, while endpoints in the remaining rare diseases (Charcot Marie Tooth disease, Gaucher disease Type I, Huntington's disease, Sjogren's syndrome and Tourette syndrome) were a worse match. Misclassifications varied with increasing disease prevalence. CONCLUSIONS Neutral theory confirmed that disease-severity measurement needs improvement in rare disease clinical studies, especially for some diseases, and suggested that the potential for accuracy increases as the body of knowledge on a disease increases. Using Neutral theory to benchmark disease-severity measurement in rare disease clinical studies may reduce the risk of misclassification, ensuring that recruitment and treatment effect assessment optimise medicine adoption and benefit patients.
Collapse
Affiliation(s)
- Ravi Jandhyala
- Medialis Ltd, 3 Warren Yard, Wolverton Mill, Milton Keynes, MK12 5NW, UK.
- Centre for Pharmaceutical Medicine Research, King's College University, London, UK.
| |
Collapse
|
7
|
Marcellusi A, Raimondo P, Galeone C, Canonico PL. Time to market access in Italia: durata del processo di P&R per i farmaci per le malattie rare. GLOBAL & REGIONAL HEALTH TECHNOLOGY ASSESSMENT 2023; 10:79-88. [PMID: 37942299 PMCID: PMC10628501 DOI: 10.33393/grhta.2023.2610] [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: 05/25/2023] [Accepted: 09/26/2023] [Indexed: 11/10/2023] Open
Abstract
Objective: This paper aims to investigate the duration of the pricing & reimbursement (P&R) procedures submitted in Italy by pharmaceutical marketing authorization holders (MAH) for drugs indicated for rare diseases. Methods: All the data used in this analysis were publicly available on different sources of the Italian Ministry of Health, the Italian Medicines Agency (AIFA) and other official websites. The information was systematically collected to investigate the timeline (days) needed to complete the P&R process. The process was divided into 6 simplified steps and the median and range of days needed for each phase were estimated based on data reported in official/published documents. The analysis was stratified considering every single step of the assessment phase and included segmentation of drugs into indications for rare diseases, Orphan designation, Innovation assessment and Managed entry agreements (MEAs). Results: Overall, 181 first indication procedures were submitted to AIFA in the period considered and, of these, 167 (92.3%) were completed and 129 procedures were considered for the final analysis and the median duration of the entire process (MAH submission to final Gazette publication) was 434 days (range 176.0-918.0). The duration of procedures for rare diseases (n = 53) was longer than those for non-rare-disease procedures (n = 76) (463.0 days vs 407.5 days respectively). Among rare disease procedures, orphan designation and MEAs represent predictors for time prolongation while innovation is associated with a shorter assessment time. Conclusion: The study describes the time spent in each phase of the assessment and the appraisal process and demonstrates that uncertainty represents the main driver for the increment in the overall time.
Collapse
Affiliation(s)
- Andrea Marcellusi
- Economic Evaluation and HTA (EEHTA-CEIS), Università degli studi di Roma “Tor Vergata”, Roma - Italy
| | | | - Carlotta Galeone
- Bicocca Applied Statistics Center (B-ASC), Università degli Studi di Milano-Bicocca, Milano - Italy
- Biostatistics & Outcome Research, Statinfo, Milano - Italy
| | - Pier Luigi Canonico
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara - Italy
| |
Collapse
|
8
|
Denton N, Mulberg AE, Molloy M, Charleston S, Fajgenbaum DC, Marsh ED, Howard P. Sharing is caring: a call for a new era of rare disease research and development. Orphanet J Rare Dis 2022; 17:389. [PMID: 36303170 PMCID: PMC9612604 DOI: 10.1186/s13023-022-02529-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 08/05/2022] [Accepted: 10/02/2022] [Indexed: 01/25/2023] Open
Abstract
Scientific advances in the understanding of the genetics and mechanisms of many rare diseases with previously unknown etiologies are inspiring optimism in the patient, clinical, and research communities and there is hope that disease-specific treatments are on the way. However, the rare disease community has reached a critical point in which its increasingly fragmented structure and operating models are threatening its ability to harness the full potential of advancing genomic and computational technologies. Changes are therefore needed to overcome these issues plaguing many rare diseases while also supporting economically viable therapy development. In "Data silos are undermining drug development and failing rare disease patients (Orphanet Journal of Rare Disease, Apr 2021)," we outlined many of the broad issues underpinning the increasingly fragmented and siloed nature of the rare disease space, as well as how the issues encountered by this community are representative of biomedical research more generally. Here, we propose several initiatives for key stakeholders - including regulators, private and public foundations, and research institutions - to reorient the rare disease ecosystem and its incentives in a way that we believe would cultivate and accelerate innovation. Specifically, we propose supporting non-proprietary patient registries, greater data standardization, global regulatory harmonization, and new business models that encourage data sharing and research collaboration as the default mode. Leadership needs to be integrated across sectors to drive meaningful change between patients, industry, sponsors, and academic medical centers. To transform the research and development landscape and unlock its vast healthcare, economic, and scientific potential for rare disease patients, a new model is ultimately the goal for all.
Collapse
Affiliation(s)
- Nathan Denton
- grid.25879.310000 0004 1936 8972Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA ,grid.427771.00000 0004 0619 7027Amicus Therapeutics, Philadelphia, PA 19104 USA
| | | | - Monique Molloy
- grid.25879.310000 0004 1936 8972Department of Medicine, Orphan Disease Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Samantha Charleston
- grid.25879.310000 0004 1936 8972Department of Medicine, Orphan Disease Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - David C. Fajgenbaum
- grid.25879.310000 0004 1936 8972Department of Medicine, Orphan Disease Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA ,grid.25879.310000 0004 1936 8972Translational Medicine & Human Genetics, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, PA 19104 USA
| | - Eric D. Marsh
- grid.25879.310000 0004 1936 8972Department of Medicine, Orphan Disease Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA ,grid.25879.310000 0004 1936 8972Departments of Neurology and Pediatrics, Perelman School of Medicine, Children’s Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA 19104 USA ,grid.239552.a0000 0001 0680 8770Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104 USA ,grid.427771.00000 0004 0619 7027Amicus Therapeutics, Philadelphia, PA 19104 USA
| | - Paul Howard
- grid.427771.00000 0004 0619 7027Amicus Therapeutics, Philadelphia, PA 19104 USA
| |
Collapse
|
9
|
Accrual-Monitoring Practices for Various Disease Trials among AACI Member Cancer Centers. Clin Pract 2022; 12:692-700. [PMID: 36136866 PMCID: PMC9498538 DOI: 10.3390/clinpract12050072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 08/26/2022] [Accepted: 08/30/2022] [Indexed: 11/19/2022] Open
Abstract
Progress in the management of rare diseases, including rare cancers, is dependent upon clinical trials; however, as many as 32% of rare-disease trials go uncompleted or unpublished due to insufficient accrual. Monitoring practices may differ between institutions. We sought to survey the regulatory standards for various trial types among major U.S. cancer centers. A 10-question survey was designed using Qualtrics assessment software. The survey was sent via email to an internal server of member institutions of the Association of American Cancer Institutes (AACI). Of 103 AACI centers, 31% completed the survey (n = 32). Respondents differed in their definitions of a rare disease, minimum expectations for rare tumor studies, and frequency of accrual monitoring by their institutional Protocol Review and Monitoring Committee. Seventy-three percent of respondents did not close trials based on low accrual. Strategies to optimize accrual included investigator incentives for high accrual and penalties for low accrual in 37% and 13% of respondents, respectively.
Collapse
|
10
|
Kubota Y, Narukawa M. Randomized Controlled Trial Data for New Drug Application for Rare Diseases in Japan. Ther Innov Regul Sci 2022; 56:659-666. [PMID: 35478399 DOI: 10.1007/s43441-022-00404-1] [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: 09/28/2021] [Accepted: 04/07/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND High-quality evidence is often not obtained in the clinical trials of rare diseases because these trials tend to be smaller in size and non-controlled. We investigated the potential factors associated with the need for randomized controlled trials (RCTs) in the clinical data package for new drug applications for rare diseases in Japan. METHODS This study focused on 130 drugs with orphan drug designation approved in Japan between April 2004 and March 2020. RESULTS Multivariable regression analysis showed that the prevalence (odds ratio [OR] 3.21, 95% confidence interval [CI] 1.18-8.6) and the type of primary endpoint (OR 6.66, 95% CI 2.41-18.37) were associated with the need for RCTs in the clinical data package in Japan. CONCLUSIONS Our findings highlight the importance of adequate understanding of the target disease in new drug development for rare diseases.
Collapse
Affiliation(s)
- Yosuke Kubota
- Department of Clinical Medicine (Pharmaceutical Medicine), Graduate School of Pharmaceutical Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan.
- Development, Astellas Pharma Inc., Tokyo, 103-0023, Japan.
| | | |
Collapse
|
11
|
Chang X, Zhang J, Jiang Y, Shang M, Wu Y. A survey of registered pharmacological clinical trials on rare neurological diseases in children in 2010-2020. Front Pediatr 2022; 10:963601. [PMID: 36405841 PMCID: PMC9672510 DOI: 10.3389/fped.2022.963601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022] Open
Abstract
OBJECTIVE To clarify the current state of methodology of clinical trials for rare neurological diseases in children, and to provide a basis for the further optimization of the trial design. METHODS Data of clinical trials for the rare neurological diseases with childhood onset (searched through https://rarediseases.info.nih.gov/diseases and www.Orpha.net) registered on the Clinicaltrils.gov from January 2010 to June 2020 was collected. Analysis on the methodology of the clinical trials were performed, focusing on initiator of the studies, multi or single research center, study design, sample size, and the endpoint using in the trial. RESULTS A total of 162 clinical trials were included, covering only 7.3% (61/835) of rare neurological diseases in children. 101 (62.3%) were initiated by pharmaceutical companies, and 61 (37.7%) by investigators. Most (95.4%) of global multicenter studies were initiated by pharmaceutical companies, whereas most (70.0%) of single-center studies were initiated by investigators (χ 2 = 61.635, P < 0.001). Of the 162 trials, 74 (45.7%) were open-label single-arm trials, 68 (42.0%) were randomized double-blind parallel controlled trials (RCT), 12 (7.4%) were randomized crossover trials. Most of RCTs (73.5%) and 54.1% of open-label single-arm trials were initiated by pharmaceutical companies. The proportion of RCTs in clinical trials for diseases with a prevalence of ≥1/10,000 (62.5%) was higher than that in diseases with prevalence ≤1/1,000,000 (12.0%) or 1/1,000,000~1/10,000 (43.1%) (χ 2 = 14.790, P = 0.001). The median expected sample size of the studies was 34 (4-500). 132 (132/162, 81.5%) studies enrolled fewer than 100 cases. Diseases with a prevalence of ≥1/10,000 had significantly larger sample sizes than other prevalence classes (P < 0.001, P = 0.003). CONCLUSIONS There were few clinical trials targeting on treatment of rare neurological diseases in children. Trials on rare diseases used fewer participants, and high-quality randomized controlled trials were less common. It is necessary to conduct global multicenter recruitment and choose optimal study designs to improve the level of evidence in clinical trials on rare diseases.
Collapse
Affiliation(s)
- Xuting Chang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Jie Zhang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Yuwu Jiang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Meixia Shang
- Department of Medical Statistics, Peking University First Hospital, Beijing, China
| | - Ye Wu
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| |
Collapse
|
12
|
Personalized Medicine to Improve Treatment of Dopa-Responsive Dystonia-A Focus on Tyrosine Hydroxylase Deficiency. J Pers Med 2021; 11:jpm11111186. [PMID: 34834538 PMCID: PMC8625014 DOI: 10.3390/jpm11111186] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 11/25/2022] Open
Abstract
Dopa-responsive dystonia (DRD) is a rare movement disorder associated with defective dopamine synthesis. This impairment may be due to the fact of a deficiency in GTP cyclohydrolase I (GTPCHI, GCH1 gene), sepiapterin reductase (SR), tyrosine hydroxylase (TH), or 6-pyruvoyl tetrahydrobiopterin synthase (PTPS) enzyme functions. Mutations in GCH1 are most frequent, whereas fewer cases have been reported for individual SR-, PTP synthase-, and TH deficiencies. Although termed DRD, a subset of patients responds poorly to L-DOPA. As this is regularly observed in severe cases of TH deficiency (THD), there is an urgent demand for more adequate or personalized treatment options. TH is a key enzyme that catalyzes the rate-limiting step in catecholamine biosynthesis, and THD patients often present with complex and variable phenotypes, which results in frequent misdiagnosis and lack of appropriate treatment. In this expert opinion review, we focus on THD pathophysiology and ongoing efforts to develop novel therapeutics for this rare disorder. We also describe how different modeling approaches can be used to improve genotype to phenotype predictions and to develop in silico testing of treatment strategies. We further discuss the current status of mathematical modeling of catecholamine synthesis and how such models can be used together with biochemical data to improve treatment of DRD patients.
Collapse
|
13
|
McIntosh A, Sverdlov O, Yu L, Kaufmann P. Clinical Design and Analysis Strategies for the Development of Gene Therapies: Considerations for Quantitative Drug Development in the Age of Genetic Medicine. Clin Pharmacol Ther 2021; 110:1207-1215. [PMID: 33666225 DOI: 10.1002/cpt.2224] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 03/01/2021] [Indexed: 12/19/2022]
Abstract
Cell and gene therapies have shown enormous promise across a range of diseases in recent years. Numerous adoptive cell therapy modalities as well as systemic and direct-to-target tissue gene transfer administrations are currently in clinical development. The clinical trial design, development, reporting, and analysis of novel cell and gene therapies can differ significantly from established practices for small molecule drugs and biologics. Here, we discuss important quantitative considerations and key competencies for drug developers in preclinical requirements, trial design, and lifecycle planning for gene therapies. We argue that the unique development path of gene therapies requires practicing quantitative drug developers-statisticians, pharmacometricians, pharmacokineticists, epidemiologists, and medical and translational science leads-to exercise active collaboration and cross-functional learning across development stages.
Collapse
Affiliation(s)
| | | | - Li Yu
- Novartis Gene Therapies, Bannockburn, Illinois, USA
| | | |
Collapse
|
14
|
Willgoss T, Cassater D, Connor S, Krishnan ML, Miller MT, Dias-Barbosa C, Phillips D, McCormack J, Bird LM, Burdine RD, Claridge S, Bichell TJ. Measuring What Matters to Individuals with Angelman Syndrome and Their Families: Development of a Patient-Centered Disease Concept Model. Child Psychiatry Hum Dev 2021; 52:654-668. [PMID: 32880036 PMCID: PMC8238699 DOI: 10.1007/s10578-020-01051-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Angelman syndrome (AS) is a complex, heterogeneous, and life-long neurodevelopmental disorder. Despite the considerable impact on individuals and caregivers, no disease-modifying treatments are available. To support holistic clinical management and the development of AS-specific outcome measures for clinical studies, we conducted primary and secondary research identifying the impact of symptoms on individuals with AS and their unmet need. This qualitative research adopted a rigorous step-wise approach, aggregating information from published literature, then evaluating it via disease concept elicitation interviews with clinical experts and caregivers. We found that the AS-defining concepts most relevant for treatment included: impaired expressive communication, seizures, maladaptive behavior, cognitive impairment, motor function difficulties, sleep disturbance, and limited self-care abilities. We highlight the relevance of age in experiencing these key AS concepts, and the difference between the perceptions of clinicians and caregivers towards the syndrome. Finally, we outline the impact of AS on individuals, caregivers, and families.
Collapse
Affiliation(s)
- Tom Willgoss
- Roche Products Limited, Hexagon Place, 6 Falcon Way, Shire Park, Welwyn Garden City, AL7 1TW, UK.
| | - Daiana Cassater
- grid.417570.00000 0004 0374 1269Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Siobhan Connor
- grid.419227.bRoche Products Limited, Hexagon Place, 6 Falcon Way, Shire Park, Welwyn Garden City, AL7 1TW UK
| | - Michelle L. Krishnan
- grid.417570.00000 0004 0374 1269Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Meghan T. Miller
- grid.417570.00000 0004 0374 1269Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | | | - Dawn Phillips
- grid.410711.20000 0001 1034 1720Division of Physical Therapy, School of Medicine, University of North Carolina, Chapel Hill, NC USA
| | - Julie McCormack
- grid.423257.50000 0004 0510 2209Evidera, Patient-Centered Research, Bethesda, MD USA
| | - Lynne M. Bird
- grid.266100.30000 0001 2107 4242Department of Pediatrics, University of California San Diego, San Diego, CA USA
| | - Rebecca D. Burdine
- grid.16750.350000 0001 2097 5006Department of Molecular Biology, Princeton University, Princeton, NJ USA
| | - Sharon Claridge
- Foundation for Angelman Syndrome Research (FAST), Downers Grove, IL USA
| | - Terry Jo Bichell
- Consortium for Outcome Measures and Biomarkers for Neurodevelopmental Disorders, Nashville, TN USA
| |
Collapse
|
15
|
Hartman H, Tamura RN, Schipper MJ, Kidwell KM. Design and analysis considerations for utilizing a mapping function in a small sample, sequential, multiple assignment, randomized trials with continuous outcomes. Stat Med 2020; 40:312-326. [PMID: 33111381 DOI: 10.1002/sim.8776] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 08/11/2020] [Accepted: 09/29/2020] [Indexed: 12/19/2022]
Abstract
Small sample, sequential, multiple assignment, randomized trials (snSMARTs) are multistage trials with the overall goal of determining the best treatment after a fixed amount of time. In snSMART trials, patients are first randomized to one of three treatments and a binary (e.g. response/nonresponse) outcome is measured at the end of the first stage. Responders to first stage treatment continue their treatment. Nonresponders to first stage treatment are rerandomized to one of the remaining treatments. The same binary outcome is measured at the end of the first and second stages, and data from both stages are pooled together to find the best first stage treatment. However, in many settings the primary endpoint may be continuous, and dichotomizing this continuous variable may reduce statistical efficiency. In this article, we extend the snSMART design and methods to allow for continuous outcomes. Instead of requiring a binary outcome at the first stage for rerandomization, the probability of staying on the same treatment or switching treatment is a function of the first stage outcome. Rerandomization based on a mapping function of a continuous outcome allows for snSMART designs without requiring a binary outcome. We perform simulation studies to compare the proposed design with continuous outcomes to standard snSMART designs with binary outcomes. The proposed design results in more efficient treatment effect estimates and similar outcomes for trial patients.
Collapse
Affiliation(s)
- Holly Hartman
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, USA
| | - Roy N Tamura
- Health Informatics Institute, University of South Florida, Tampa, Florida, USA
| | - Matthew J Schipper
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, USA.,Department of Radiation Oncology, University of Michigan
| | - Kelley M Kidwell
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
16
|
Leonardi A, Dupuis-Deniaud M, Bremond-Gignac D. Clinical efficacy assessment in severe vernal keratoconjunctivitis: preliminary validation of a new penalties-adjusted corneal fluorescein staining score. JOURNAL OF MARKET ACCESS & HEALTH POLICY 2020; 8:1748492. [PMID: 32341771 PMCID: PMC7170307 DOI: 10.1080/20016689.2020.1748492] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 02/13/2020] [Accepted: 02/17/2020] [Indexed: 06/11/2023]
Abstract
Introduction and objective: Vernal keratoconjunctivitis (VKC) is a rare allergic eye condition that occurs in children and is characterised by a combination of debilitating symptoms. Repeated use of topical corticosteroid rescue therapy is often necessary in severe forms. This study aims to assess the validity of a new composite endpoint: the penalties-adjusted corneal staining score (PACS-S) proposed as primary endpoint in VEKTIS trial evaluating the efficacy of a new corticosteroid-sparing treatment, VERKAZIA® (ciclosporin 1 mg/ml eye drops), in severe VKC patients. Methodology: This research comprised a systematic literature review to identify efficacy endpoints being proposed in clinical trials for pediatric patients with severe VKC, followed by a remote expert advisory board assessing the validity of the PACS-S. Results: While no agreed or validated endpoint for assessing efficacy in VKC was identified when VEKTIS trial started, the experts' board acknowledged a high face validity of PACS-S as a subjective integrated measure matching the current clinical practice. A fair external validity was considered with regards to VEKTIS trial secondary endpoints. Conclusion: PACS-S appears to be a reliable, valid and clinically meaningful primary endpoint that allows significant improvement over existing endpoints in severe VKC trials. Additional research is needed to validate this endpoint.
Collapse
Affiliation(s)
- Andrea Leonardi
- Ophthalmology Unit, Department of Neuroscience, University of Padua, Padua, Italy
| | | | - Dominique Bremond-Gignac
- Hospital Necker Enfants Malades, APHP, and CNRS Unit FR 3636, Paris Descartes University, Paris, France
| |
Collapse
|
17
|
Tambuyzer E, Vandendriessche B, Austin CP, Brooks PJ, Larsson K, Miller Needleman KI, Valentine J, Davies K, Groft SC, Preti R, Oprea TI, Prunotto M. Therapies for rare diseases: therapeutic modalities, progress and challenges ahead. Nat Rev Drug Discov 2019; 19:93-111. [PMID: 31836861 DOI: 10.1038/s41573-019-0049-9] [Citation(s) in RCA: 203] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2019] [Indexed: 12/26/2022]
Abstract
Most rare diseases still lack approved treatments despite major advances in research providing the tools to understand their molecular basis, as well as legislation providing regulatory and economic incentives to catalyse the development of specific therapies. Addressing this translational gap is a multifaceted challenge, for which a key aspect is the selection of the optimal therapeutic modality for translating advances in rare disease knowledge into potential medicines, known as orphan drugs. With this in mind, we discuss here the technological basis and rare disease applicability of the main therapeutic modalities, including small molecules, monoclonal antibodies, protein replacement therapies, oligonucleotides and gene and cell therapies, as well as drug repurposing. For each modality, we consider its strengths and limitations as a platform for rare disease therapy development and describe clinical progress so far in developing drugs based on it. We also discuss selected overarching topics in the development of therapies for rare diseases, such as approval statistics, engagement of patients in the process, regulatory pathways and digital tools.
Collapse
Affiliation(s)
- Erik Tambuyzer
- BioPontis Alliance for Rare Diseases Foundation fup/son, Brussels, Belgium. .,BioPontis Alliance Rare Disease Foundation, Inc, Raleigh, NC, USA.
| | - Benjamin Vandendriessche
- Byteflies, Antwerp, Belgium.,Department of Electrical, Computer, and Systems Engineering (ECSE), Case Western Reserve University, Cleveland, OH, USA
| | - Christopher P Austin
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Philip J Brooks
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Kristina Larsson
- Orphan Medicines Office, European Medicines Agency, Amsterdam, Netherlands
| | | | | | - Kay Davies
- MDUK Oxford Neuromuscular Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Stephen C Groft
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Robert Preti
- Hitachi Chemical Regenerative Medicine Business Sector, Allendale, NJ, USA
| | - Tudor I Oprea
- Translational Informatics Division, Department of Internal Medicine, University of New Mexico Albuquerque, Albuquerque, NM, USA.,UNM Comprehensive Cancer Center, University of New Mexico Health Science Center, Albuquerque, NM, USA
| | - Marco Prunotto
- School of Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
18
|
Shoffner JM. Concerning "Triheptanoin vs trioctanoin for long-chain fatty acid oxidation disorders: A double blinded, randomized controlled trial" by Gillingham et al. J Inherit Metab Dis 2019; 42:394-395. [PMID: 30838661 DOI: 10.1002/jimd.12078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 02/20/2019] [Indexed: 12/19/2022]
Affiliation(s)
- John M Shoffner
- Neurology, Biochemical Genetics, Molecular Genetics, Atlanta, GA
| |
Collapse
|
19
|
Mulberg AE, Bucci-Rechtweg C, Giuliano J, Jacoby D, Johnson FK, Liu Q, Marsden D, McGoohan S, Nelson R, Patel N, Romero K, Sinha V, Sitaraman S, Spaltro J, Kessler V. Regulatory strategies for rare diseases under current global regulatory statutes: a discussion with stakeholders. Orphanet J Rare Dis 2019; 14:36. [PMID: 30736861 PMCID: PMC6368795 DOI: 10.1186/s13023-019-1017-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 01/31/2019] [Indexed: 12/20/2022] Open
Abstract
Rare or orphan diseases often are inherited and overwhelmingly affect children. Many of these diseases have no treatments, are incurable, and have a devastating impact on patients and their families. Regulatory standards for drug approval for rare diseases must ensure that patients receive safe and efficacious treatments. However, regulatory bodies have shown flexibility in applying these standards to drug development in rare diseases, given the unique challenges that hinder efficient and effective traditional clinical trials, including low patient numbers, limited understanding of disease pathology and progression, variability in disease presentation, and a lack of established endpoints. To take steps toward improving rare disease clinical development strategies under current global regulatory statutes, Amicus Therapeutics, Inc. and BioNJ convened a 1-day meeting that included representatives from the Food and Drug Administration (FDA), biopharmaceutical industry, and not-for-profit agencies. The meeting focused on orphan diseases in pediatric and adult patients and was intended to identify potential strategies to overcome regulatory hurdles through open collaboration. During this meeting, several strategies were identified to minimize the limitations associated with low patient numbers in rare diseases, including the use of natural history to generate historical control data in comparisons, simulations, and identifying inclusion/exclusion criteria and appropriate endpoints. Novel approaches to clinical trial design were discussed to minimize patient exposure to placebo and to reduce the numbers of patients and clinical trials needed for providing substantial evidence. Novel statistical analysis approaches were also discussed to address the inherent challenges of small patient numbers. Areas of urgent unmet need were identified, including the need to develop registries that protect patient identities, to establish close collaboration and communication between the sponsor and regulatory bodies to address methodological and statistical challenges, to collaborate in pre-competitive opportunities within multiple sponsors and in conjunction with academia and disease-specific patient advocacy groups for optimal data sharing, and to develop harmonized guidelines for data extrapolation from source to target pediatric populations. Ultimately, these innovations will help in solving many regulatory challenges in rare disease drug development and encourage the availability of new treatments for patients with rare diseases.
Collapse
Affiliation(s)
- Andrew E Mulberg
- Amicus Therapeutics, Inc., 1 Cedar Brook Drive, Cranbury, NJ, 08512, USA.
| | | | - Joseph Giuliano
- Amicus Therapeutics, Inc., 1 Cedar Brook Drive, Cranbury, NJ, 08512, USA
| | - David Jacoby
- BioMarin Pharmaceutical Inc., 770 Lindaro St, San Rafael, CA, 94901, USA
| | - Franklin K Johnson
- Amicus Therapeutics, Inc., 1 Cedar Brook Drive, Cranbury, NJ, 08512, USA
| | - Qing Liu
- Amicus Therapeutics, Inc., 1 Cedar Brook Drive, Cranbury, NJ, 08512, USA
| | - Deborah Marsden
- Ultragenyx Pharmaceutical, Inc., 60 Leveroni Ct, Novato, CA, 94949, USA
| | - Scott McGoohan
- Vertex Pharmaceuticals, 50 Northern Ave, Boston, MA, 02210, USA
| | - Robert Nelson
- Johnson & Johnson, 920 Route 202 South, Raritan, NJ, 08869, USA
| | - Nita Patel
- Amicus Therapeutics, Inc., 1 Cedar Brook Drive, Cranbury, NJ, 08512, USA
| | - Klaus Romero
- Critical Path Institute, 1730 East River Rd, Tucson, AZ, 85718, USA
| | - Vikram Sinha
- Merck & Co, Inc. 351 North Sumneytown Pike, North Wales, PA, 19454, USA
| | - Sheela Sitaraman
- Amicus Therapeutics, Inc., 1 Cedar Brook Drive, Cranbury, NJ, 08512, USA
| | - John Spaltro
- Amicus Therapeutics, Inc., 1 Cedar Brook Drive, Cranbury, NJ, 08512, USA
| | - Vivian Kessler
- Amicus Therapeutics, Inc., 1 Cedar Brook Drive, Cranbury, NJ, 08512, USA
| |
Collapse
|
20
|
Koene S, van Bon L, Bertini E, Jimenez-Moreno C, van der Giessen L, de Groot I, McFarland R, Parikh S, Rahman S, Wood M, Zeman J, Janssen A, Smeitink J. Outcome measures for children with mitochondrial disease: consensus recommendations for future studies from a Delphi-based international workshop. J Inherit Metab Dis 2018; 41:1267-1273. [PMID: 30027425 PMCID: PMC6326961 DOI: 10.1007/s10545-018-0229-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 06/20/2018] [Accepted: 07/02/2018] [Indexed: 12/19/2022]
Abstract
Although there are no effective disease-modifying therapies for mitochondrial diseases, an increasing number of trials are being conducted in this rare disease group. The use of sensitive and valid endpoints is essential to test the effectiveness of potential treatments. There is no consensus on which outcome measures to use in children with mitochondrial disease. The aims of this two-day Delphi-based workshop were to (i) define the protocol for an international, multi-centre natural history study in children with mitochondrial myopathy and (ii) to select appropriate outcome measures for a validation study in children with mitochondrial encephalopathy. We suggest two sets of outcome measures for a natural history study in children with mitochondrial myopathy and for a proposed validation study in children with mitochondrial encephalopathy.
Collapse
Affiliation(s)
- Saskia Koene
- Radboud Center for Mitochondrial Medicine, Department of Paediatrics, Radboudumc, Nijmegen, The Netherlands.
| | - Lara van Bon
- Radboud Center for Mitochondrial Medicine, Department of Paediatrics, Radboudumc, Nijmegen, The Netherlands
| | - Enrico Bertini
- Unit of Neuromuscular and Neurodegenerative Disorders, Bambino Gesù Children's Research Hospital, Rome, Italy
| | - Cecilia Jimenez-Moreno
- Wellcome Centre for Mitochondrial Research, Newcastle University, Newcastle upon Tyne, UK
| | - Lianne van der Giessen
- Center for Lysosomal and Metabolic Diseases and Department of Pediatric Physiotherapy, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Imelda de Groot
- Radboud Center for Mitochondrial Medicine, Department of Paediatrics, Radboudumc, Nijmegen, The Netherlands
- Donders Center for Neuroscience, Department of Rehabilitation, Radboudumc, Nijmegen, The Netherlands
| | - Robert McFarland
- Wellcome Centre for Mitochondrial Research, Newcastle University, Newcastle upon Tyne, UK
| | - Sumit Parikh
- Mitochondrial Medicine Center, Neuroscience Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Shamima Rahman
- Mitochondrial Research Group, UCL Great Ormond Street Institute of Child Health and Metabolic Unit, Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Michelle Wood
- Mitochondrial Research Group, UCL Great Ormond Street Institute of Child Health and Metabolic Unit, Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Jiri Zeman
- Department of Paediatrics, First Faculty of Medicine and General Faculty Hospital, Prague, Czech Republic
| | - Anjo Janssen
- Radboud Center for Mitochondrial Medicine, Department of Paediatrics, Radboudumc, Nijmegen, The Netherlands
- Department of Rehabilitation, Pediatric Physical Therapy, Radboudumc, Nijmegen, The Netherlands
| | - Jan Smeitink
- Radboud Center for Mitochondrial Medicine, Department of Paediatrics, Radboudumc, Nijmegen, The Netherlands
| |
Collapse
|