1
|
Cummings JL, Teunissen CE, Fiske BK, Le Ber I, Wildsmith KR, Schöll M, Dunn B, Scheltens P. Biomarker-guided decision making in clinical drug development for neurodegenerative disorders. Nat Rev Drug Discov 2025:10.1038/s41573-025-01165-w. [PMID: 40185982 DOI: 10.1038/s41573-025-01165-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2025] [Indexed: 04/07/2025]
Abstract
Neurodegenerative disorders are characterized by complex neurobiological changes that are reflected in biomarker alterations detectable in blood, cerebrospinal fluid (CSF) and with brain imaging. As accessible proxies for processes that are difficult to measure, biomarkers are tools that hold increasingly important roles in drug development and clinical trial decision making. In the past few years, biomarkers have been the basis for accelerated approval of new therapies for Alzheimer disease and amyotrophic lateral sclerosis as surrogate end points reasonably likely to predict clinical benefit.Blood-based biomarkers are emerging for Alzheimer disease and other neurodegenerative disorders (for example, Parkinson disease, frontotemporal dementia), and some biomarkers may be informative across multiple disease states. Collection of CSF provides access to biomarkers not available in plasma, including markers of synaptic dysfunction and neuroinflammation. Molecular imaging is identifying an increasing array of targets, including amyloid plaques, neurofibrillary tangles, inflammation, mitochondrial dysfunction and synaptic density. In this Review, we consider how biomarkers can be implemented in clinical trials depending on their context of use, including providing information on disease risk and/or susceptibility, diagnosis, prognosis, pharmacodynamic outcomes, monitoring, prediction of response to therapy and safety. Informed choice of increasingly available biomarkers and rational deployment in clinical trials support drug development decision making and de-risk the drug development process for neurodegenerative disorders.
Collapse
Affiliation(s)
- Jeffrey L Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, Kirk Kerkorian School of Medicine, University of Nevada Las Vegas (UNLV), Las Vegas, NV, USA.
| | - Charlotte E Teunissen
- Neurochemistry Laboratory and Biobank, Department of Neuroscience, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Brian K Fiske
- The Michael J. Fox Foundation for Parkinson's Research, New York, NY, USA
| | - Isabelle Le Ber
- Sorbonne Université, Paris Brain Institute - Institut du Cerveau - ICM, Inserm U1127, CNRS UMR 7225, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France
| | | | - Michael Schöll
- Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and Neurochemistry, University of Gothenburg, Göteborg, Sweden
- Dementia Research Centre, Queen Square Institute of Neurology, University College London, London, UK
| | - Billy Dunn
- The Michael J. Fox Foundation for Parkinson's Research, New York, NY, USA
| | - Philip Scheltens
- Alzheimer's Center Amsterdam, Amsterdam University Medical Center, Amsterdam, the Netherlands
- EQT Group, Dementia Fund, Stockholm, Sweden
| |
Collapse
|
2
|
van Karnebeek CDM, Müller AR, Benkemoun L, Boussaad I, Cornel MC, IntHout J, de Kort M, de Oliveira Martins S, Prigione A, Rigter T, Roes KCB, Sanchez A, Schipper R, Wilkinson MD, 't Hoen PAC. SIMPATHIC: Accelerating drug repurposing for rare diseases by exploiting SIMilarities in clinical and molecular PATHology. Mol Genet Metab 2025; 144:109073. [PMID: 40086177 DOI: 10.1016/j.ymgme.2025.109073] [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: 01/08/2025] [Revised: 02/27/2025] [Accepted: 02/27/2025] [Indexed: 03/16/2025]
Abstract
Rare diseases affect over 400 million people worldwide, with approved treatment available for less than 6 % of these diseases. Drug repurposing is a key strategy in the development of therapies for rare disease patients with large unmet medical needs. The process of repurposing drugs compared to novel drug development is a time-saving and cost-efficient method potentially resulting in higher success rates. To accelerate and ensure sustainability in therapy development for rare neurometabolic, neurological, and neuromuscular diseases, an international consortium SIMilarities in clinical and molecular PATHology (SIMPATHIC) has been established where we move away from the one drug one disease concept and move towards one drug targeting a pathomechanism shared between diseases, by applying parallel preclinical and clinical drug development. Here the consortium describes accelerators of drug repurposing pursued by the consortium, including 1) co-creation, 2) patient empowerment, 3) use of standardized induced pluripotent stem cell (iPSC)-derived disease models and cellular and molecular profiling, 4) high-throughput drug screening in neurons, 5) innovative clinical trial design, and 6) selection of appropriate exploitation and patient access models. In this way, a fast and effective drug repurposing pathway for several rare diseases will be established to reduce time from discovery to patient access.
Collapse
Affiliation(s)
- Clara D M van Karnebeek
- Departments of Pediatrics and Human Genetics, Amsterdam UMC location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Emma Center for Personalized Medicine, Amsterdam UMC, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Amsterdam Reproduction and Development, Amsterdam UMC, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Annelieke R Müller
- Departments of Pediatrics and Human Genetics, Amsterdam UMC location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Emma Center for Personalized Medicine, Amsterdam UMC, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Amsterdam Public Health research institute, Personalized Medicine program, Boelelaan 1117, 1007 MB Amsterdam, the Netherlands
| | | | - Ibrahim Boussaad
- LCSB, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 7 Av. des Hauts-Fourneaux, 4362 Esch-Belval, Esch-sur-Alzette, Luxembourg
| | - Martina C Cornel
- Amsterdam Reproduction and Development, Amsterdam UMC, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Department of Human Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam, 1007 MB Amsterdam, the Netherlands
| | - Joanna IntHout
- IQ Health science department Radboudumc, Postbus 9101, 6500 HB Nijmegen, the Netherlands
| | - Martin de Kort
- EATRIS ERIC European Infrastructure for Translational Medicine, De Boelelaan 1118, 1081 HZ Amsterdam, the Netherlands
| | - Sofia de Oliveira Martins
- Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; Comprehensive Health Research Center, Evora, Portugal
| | - Alessandro Prigione
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225 Düsseldorf, Germany
| | - Tessel Rigter
- Amsterdam Public Health research institute, Personalized Medicine program, Boelelaan 1117, 1007 MB Amsterdam, the Netherlands; Department of Human Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam, 1007 MB Amsterdam, the Netherlands
| | - Kit C B Roes
- IQ Health science department Radboudumc, Postbus 9101, 6500 HB Nijmegen, the Netherlands
| | - Anna Sanchez
- EATRIS ERIC European Infrastructure for Translational Medicine, De Boelelaan 1118, 1081 HZ Amsterdam, the Netherlands
| | - Raymond Schipper
- Department of Medical BioSciences, Radboud university medical center, Geert Grooteplein Zuid 26/28, 6525GA Nijmegen, the Netherlands
| | - Mark D Wilkinson
- FAIR Data Systems S.L., C. del Corazón de María, 9, 1'D, Chamartín, 28002 Madrid, Spain; Departamento de Biotecnología-Biología Vegetal, Escuela Técnica Superior de Ingeniería Agronómica, Alimentaria y de Biosistemas, Centro de Biotecnología y Genómica de Plantas (CBGP), Universidad Politécnica de Madrid (UPM), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria-CSIC (INIA-CSIC), Pozuelo de Alarcón (Madrid), Spain
| | - Peter A C 't Hoen
- Department of Medical BioSciences, Radboud university medical center, Geert Grooteplein Zuid 26/28, 6525GA Nijmegen, the Netherlands.
| |
Collapse
|
3
|
Pullen LC, Bott N, McCanless C, Revana A, Sevinc G, Gorman C, Duncan A, Poliquin S, Pfalzer AC, Schmidt KQ, Wassman ER, Chapman C, Picone M. Use of Basket Trials to Solve Sleep Problems in Patients with Rare Diseases. Clocks Sleep 2024; 6:656-667. [PMID: 39584973 PMCID: PMC11586945 DOI: 10.3390/clockssleep6040044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/18/2024] [Accepted: 10/25/2024] [Indexed: 11/26/2024] Open
Abstract
The need for sleep is universal, and the ability to meet this need impacts the quality of life for patients, families, and caregivers. Although substantial progress has been made in treating rare diseases, many patients have unmet medical sleep needs, and current regulatory policy makes it prohibitively difficult to address those needs medically. This opinion reviews the rare disease experience with sleep disorders and explores potential solutions. First, we provide case profiles for the rare diseases Wilson's Disease, Angelman Syndrome, and Prader-Willi Syndrome. These profiles highlight challenges in rare disease diagnosis and barriers to pinpointing disease pathophysiology, including biomarkers that intersect with sleep disorders. Second, we transition to a bird's eye view of sleep disorders and rare diseases by reporting input from a stakeholder discussion with the U.S. Food and Drug Administration regarding abnormal sleep patterns in various rare diseases. Last, in response to the profound unmet medical needs of patients with rare diseases and sleep disorders, we propose adapting and using the clinical trial design known as a "basket trial". In this case, a basket trial would include patients with different rare diseases but the same debilitating symptoms. This research approach has the potential to benefit many rare disease patients who are otherwise left with profound unmet medical needs.
Collapse
Affiliation(s)
| | - Nick Bott
- Takeda Pharmaceuticals, Cambridge, MA 02139, USA;
| | | | - Amee Revana
- Texas Children’s Hospital, Houston, TX 77001, USA;
| | - Gunes Sevinc
- Ardea Outcomes, Halifax, NS B3J 0J2, Canada; (G.S.); (C.C.)
| | - Casey Gorman
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Alexandra Duncan
- COMBINEDBrain, Brentwood, TN 37027, USA; (A.D.); (S.P.); (A.C.P.); (K.Q.S.)
| | - Sarah Poliquin
- COMBINEDBrain, Brentwood, TN 37027, USA; (A.D.); (S.P.); (A.C.P.); (K.Q.S.)
| | - Anna C. Pfalzer
- COMBINEDBrain, Brentwood, TN 37027, USA; (A.D.); (S.P.); (A.C.P.); (K.Q.S.)
- Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Katie Q. Schmidt
- COMBINEDBrain, Brentwood, TN 37027, USA; (A.D.); (S.P.); (A.C.P.); (K.Q.S.)
| | | | - Chère Chapman
- Ardea Outcomes, Halifax, NS B3J 0J2, Canada; (G.S.); (C.C.)
| | - Maria Picone
- TREND Community, Philadelphia, PA 19102, USA; (E.R.W.); (M.P.)
| |
Collapse
|
4
|
Cummings JL, Osse AML, Kinney JW, Cammann D, Chen J. Alzheimer's Disease: Combination Therapies and Clinical Trials for Combination Therapy Development. CNS Drugs 2024; 38:613-624. [PMID: 38937382 PMCID: PMC11258156 DOI: 10.1007/s40263-024-01103-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/11/2024] [Indexed: 06/29/2024]
Abstract
Alzheimer's disease (AD) is a complex multifaceted disease. Recently approved anti-amyloid monoclonal antibodies slow disease progression by approximately 30%, and combination therapy appears necessary to prevent the onset of AD or produce greater slowing of cognitive and functional decline. Combination therapies may address core features, non-specific co-pathology commonly occurring in patients with AD (e.g., inflammation), or non-AD pathologies that may co-occur with AD (e.g., α-synuclein). Combination therapies may be advanced through co-development of more than one new molecular entity or through add-on strategies including an approved agent plus a new molecular entity. Addressing add-on combination therapy is currently urgent since patients on anti-amyloid monoclonal antibodies may be included in clinical trials for experimental agents. Phase 1 information must be generated for each agent in combination drug development. Phase 2 and Phase 3 of add-on therapies may contrast the new molecular entity, the approved agent as standard of care, and the combination. More complex development programs including standard or modified combinatorial designs are required for co-development of two or more new molecular entities. Biomarkers are markedly affected by anti-amyloid monoclonal antibodies, and these effects must be anticipated in add-on trials. Examining target engagement biomarkers and comparing the magnitude and sequence of biomarker changes in those receiving more than one therapy, compared with those on monotherapy, may be informative. Using network-based medicine approaches, computational strategies may identify rational combinations using disease and drug effect network mapping.
Collapse
Affiliation(s)
- Jeffrey L Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas (UNLV, Las Vegas, NV, USA.
- Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas (UNLV), Las Vegas, NV, USA.
- , 1380 Opal Valley Street, Henderson, NV, 89052, USA.
| | - Amanda M Leisgang Osse
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas (UNLV, Las Vegas, NV, USA
- Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas (UNLV), Las Vegas, NV, USA
| | - Jefferson W Kinney
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas (UNLV, Las Vegas, NV, USA
- Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas (UNLV), Las Vegas, NV, USA
| | - Davis Cammann
- Nevada Institute of Personalized Medicine, University of Nevada Las Vegas (UNLV), Las Vegas, NV, USA
| | - Jingchun Chen
- Nevada Institute of Personalized Medicine, University of Nevada Las Vegas (UNLV), Las Vegas, NV, USA
| |
Collapse
|
5
|
Ryan EG, Gao CX, Grantham KL, Thao LTP, Charles-Nelson A, Bowden R, Herschtal A, Lee KJ, Forbes AB, Heritier S, Phillipou A, Wolfe R. Advancing randomized controlled trial methodologies: The place of innovative trial design in eating disorders research. Int J Eat Disord 2024; 57:1337-1349. [PMID: 38469971 DOI: 10.1002/eat.24187] [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: 11/13/2023] [Revised: 02/26/2024] [Accepted: 02/26/2024] [Indexed: 03/13/2024]
Abstract
Randomized controlled trials can be used to generate evidence on the efficacy and safety of new treatments in eating disorders research. Many of the trials previously conducted in this area have been deemed to be of low quality, in part due to a number of practical constraints. This article provides an overview of established and more innovative clinical trial designs, accompanied by pertinent examples, to highlight how design choices can enhance flexibility and improve efficiency of both resource allocation and participant involvement. Trial designs include individually randomized, cluster randomized, and designs with randomizations at multiple time points and/or addressing several research questions (master protocol studies). Design features include the use of adaptations and considerations for pragmatic or registry-based trials. The appropriate choice of trial design, together with rigorous trial conduct, reporting and analysis, can establish high-quality evidence to advance knowledge in the field. It is anticipated that this article will provide a broad and contemporary introduction to trial designs and will help researchers make informed trial design choices for improved testing of new interventions in eating disorders. PUBLIC SIGNIFICANCE: There is a paucity of high quality randomized controlled trials that have been conducted in eating disorders, highlighting the need to identify where efficiency gains in trial design may be possible to advance the eating disorder research field. We provide an overview of some key trial designs and features which may offer solutions to practical constraints and increase trial efficiency.
Collapse
Affiliation(s)
- Elizabeth G Ryan
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Caroline X Gao
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
- Centre for Youth Mental Health, University of Melbourne, Melbourne, Victoria, Australia
- Orygen, Melbourne, Victoria, Australia
| | - Kelsey L Grantham
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Le Thi Phuong Thao
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Anaïs Charles-Nelson
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Rhys Bowden
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Alan Herschtal
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Katherine J Lee
- Clinical Epidemiology and Biostatistics Unit, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Andrew B Forbes
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Stephane Heritier
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Andrea Phillipou
- Centre for Youth Mental Health, University of Melbourne, Melbourne, Victoria, Australia
- Orygen, Melbourne, Victoria, Australia
- Department of Psychological Sciences, Swinburne University of Technology, Melbourne, Victoria, Australia
- Department of Mental Health, Austin Health, Melbourne, Victoria, Australia
- Department of Mental Health, St Vincent's Hospital, Melbourne, Victoria, Australia
| | - Rory Wolfe
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
6
|
Mitra S. Special Issue 'Advances in Neurodegenerative Diseases Research and Therapy 2.0'. Int J Mol Sci 2024; 25:4709. [PMID: 38731928 PMCID: PMC11083915 DOI: 10.3390/ijms25094709] [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: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
Neurodegenerative disorders (NDs) and the development of various therapeutic strategies to combat them have received increased attention in recent decades [...].
Collapse
Affiliation(s)
- Sumonto Mitra
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of NVS, Karolinska Institutet, 141 52 Huddinge, Sweden
| |
Collapse
|
7
|
Müller AR, den Hollander B, van de Ven PM, Roes KCB, Geertjens L, Bruining H, van Karnebeek CDM, Jansen FE, de Wit MCY, Ten Hoopen LW, Rietman AB, Dierckx B, Wijburg FA, Boot E, Brands MMG, van Eeghen AM. Cannabidiol (Epidyolex®) for severe behavioral manifestations in patients with tuberous sclerosis complex, mucopolysaccharidosis type III and fragile X syndrome: protocol for a series of randomized, placebo-controlled N-of-1 trials. BMC Psychiatry 2024; 24:23. [PMID: 38177999 PMCID: PMC10768432 DOI: 10.1186/s12888-023-05422-3] [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: 07/27/2023] [Accepted: 11/29/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND Many rare genetic neurodevelopmental disorders (RGNDs) are characterized by intellectual disability (ID), severe cognitive and behavioral impairments, potentially diagnosed as a comorbid autism spectrum disorder or attention-deficit hyperactivity disorder. Quality of life is often impaired due to irritability, aggression and self-injurious behavior, generally refractory to standard therapies. There are indications from previous (case) studies and patient reporting that cannabidiol (CBD) may be an effective treatment for severe behavioral manifestations in RGNDs. However, clear evidence is lacking and interventional research is challenging due to the rarity as well as the heterogeneity within and between disease groups and interindividual differences in treatment response. Our objective is to examine the effectiveness of CBD on severe behavioral manifestations in three RGNDs, including Tuberous Sclerosis Complex (TSC), mucopolysaccharidosis type III (MPS III), and Fragile X syndrome (FXS), using an innovative trial design. METHODS We aim to conduct placebo-controlled, double-blind, block-randomized, multiple crossover N-of-1 studies with oral CBD (twice daily) in 30 patients (aged ≥ 6 years) with confirmed TSC, MPS III or FXS and severe behavioral manifestations. The treatment is oral CBD up to a maximum of 25 mg/kg/day, twice daily. The primary outcome measure is the subscale irritability of the Aberrant Behavior Checklist. Secondary outcome measures include (personalized) patient-reported outcome measures with regard to behavioral and psychiatric outcomes, disease-specific outcome measures, parental stress, seizure frequency, and adverse effects of CBD. Questionnaires will be completed and study medication will be taken at the participants' natural setting. Individual treatment effects will be determined based on summary statistics. A mixed model analysis will be applied for analyzing the effectiveness of the intervention per disorder and across disorders combining data from the individual N-of-1 trials. DISCUSSION These N-of-1 trials address an unmet medical need and will provide information on the effectiveness of CBD for severe behavioral manifestations in RGNDs, potentially generating generalizable knowledge at an individual-, disorder- and RGND population level. TRIAL REGISTRATION EudraCT: 2021-003250-23, registered 25 August 2022, https://www.clinicaltrialsregister.eu/ctr-search/trial/2021-003250-23/NL .
Collapse
Affiliation(s)
- A R Müller
- Department of Pediatrics, Emma Children's Hospital, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
- 's Heeren Loo Care Group, Amersfoort, The Netherlands
- Emma Center for Personalized Medicine, Amsterdam UMC, Amsterdam, the Netherlands
| | - B den Hollander
- Department of Pediatrics, Emma Children's Hospital, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
- Emma Center for Personalized Medicine, Amsterdam UMC, Amsterdam, the Netherlands
- United for Metabolic Diseases, Amsterdam, The Netherlands
| | - P M van de Ven
- Department of Data Science and Biostatistics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - K C B Roes
- Department of Health Evidence, Biostatistics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - L Geertjens
- Child and Adolescent Psychiatry and Psychosocial Care, Amsterdam UMC Location Vrije Universiteit, Amsterdam, The Netherlands
- Amsterdam UMC, Amsterdam Neuroscience, Amsterdam Reproduction and Development, N=You Neurodevelopmental Precision Center, Amsterdam, The Netherlands
| | - H Bruining
- Emma Center for Personalized Medicine, Amsterdam UMC, Amsterdam, the Netherlands
- Child and Adolescent Psychiatry and Psychosocial Care, Amsterdam UMC Location Vrije Universiteit, Amsterdam, The Netherlands
- Amsterdam UMC, Amsterdam Neuroscience, Amsterdam Reproduction and Development, N=You Neurodevelopmental Precision Center, Amsterdam, The Netherlands
- Levvel, Center for Child and Adolescent Psychiatry, Amsterdam, The Netherlands
| | - C D M van Karnebeek
- Department of Pediatrics, Emma Children's Hospital, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
- Emma Center for Personalized Medicine, Amsterdam UMC, Amsterdam, the Netherlands
- United for Metabolic Diseases, Amsterdam, The Netherlands
- Department of Human Genetics, Amsterdam UMC, Amsterdam, The Netherlands
| | - F E Jansen
- Department of Pediatric Neurology, Brain, University Medical Center Utrecht, Utrecht, The Netherlands
| | - M C Y de Wit
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Neurology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - L W Ten Hoopen
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Child and Adolescent Psychiatry/Psychology, Sophia Children's Hospital, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - A B Rietman
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Child and Adolescent Psychiatry/Psychology, Sophia Children's Hospital, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - B Dierckx
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Child and Adolescent Psychiatry/Psychology, Sophia Children's Hospital, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - F A Wijburg
- Department of Pediatrics, Emma Children's Hospital, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - E Boot
- 's Heeren Loo Care Group, Amersfoort, The Netherlands
- The Dalglish Family 22Q Clinic, Toronto, ON, Canada
- Department of Psychiatry & Neuropsychology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - M M G Brands
- Department of Pediatrics, Emma Children's Hospital, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
- Emma Center for Personalized Medicine, Amsterdam UMC, Amsterdam, the Netherlands
- United for Metabolic Diseases, Amsterdam, The Netherlands
| | - A M van Eeghen
- Department of Pediatrics, Emma Children's Hospital, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands.
- 's Heeren Loo Care Group, Amersfoort, The Netherlands.
- Emma Center for Personalized Medicine, Amsterdam UMC, Amsterdam, the Netherlands.
| |
Collapse
|
8
|
Foltynie T, Gandhi S, Gonzalez-Robles C, Zeissler ML, Mills G, Barker R, Carpenter J, Schrag A, Schapira A, Bandmann O, Mullin S, Duffen J, McFarthing K, Chataway J, Parmar M, Carroll C. Towards a multi-arm multi-stage platform trial of disease modifying approaches in Parkinson's disease. Brain 2023; 146:2717-2722. [PMID: 36856727 PMCID: PMC10316775 DOI: 10.1093/brain/awad063] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 12/20/2022] [Accepted: 01/08/2023] [Indexed: 03/02/2023] Open
Abstract
An increase in the efficiency of clinical trial conduct has been successfully demonstrated in the oncology field, by the use of multi-arm, multi-stage trials allowing the evaluation of multiple therapeutic candidates simultaneously, and seamless recruitment to phase 3 for those candidates passing an interim signal of efficacy. Replicating this complex innovative trial design in diseases such as Parkinson's disease is appealing, but in addition to the challenges associated with any trial assessing a single potentially disease modifying intervention in Parkinson's disease, a multi-arm platform trial must also specifically consider the heterogeneous nature of the disease, alongside the desire to potentially test multiple treatments with different mechanisms of action. In a multi-arm trial, there is a need to appropriately stratify treatment arms to ensure each are comparable with a shared placebo/standard of care arm; however, in Parkinson's disease there may be a preference to enrich an arm with a subgroup of patients that may be most likely to respond to a specific treatment approach. The solution to this conundrum lies in having clearly defined criteria for inclusion in each treatment arm as well as an analysis plan that takes account of predefined subgroups of interest, alongside evaluating the impact of each treatment on the broader population of Parkinson's disease patients. Beyond this, there must be robust processes of treatment selection, and consensus derived measures to confirm target engagement and interim assessments of efficacy, as well as consideration of the infrastructure needed to support recruitment, and the long-term funding and sustainability of the platform. This has to incorporate the diverse priorities of clinicians, triallists, regulatory authorities and above all the views of people with Parkinson's disease.
Collapse
Affiliation(s)
- Tom Foltynie
- Department of Clinical & Movement Neurosciences, UCL Institute of Neurology, London WC1N 3BG, UK
| | - Sonia Gandhi
- Department of Clinical & Movement Neurosciences, UCL Institute of Neurology, London WC1N 3BG, UK
| | - Cristina Gonzalez-Robles
- Department of Clinical & Movement Neurosciences, UCL Institute of Neurology, London WC1N 3BG, UK
| | | | - Georgia Mills
- Department of Clinical & Movement Neurosciences, UCL Institute of Neurology, London WC1N 3BG, UK
| | - Roger Barker
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0PY, UK
| | | | - Anette Schrag
- Department of Clinical & Movement Neurosciences, UCL Institute of Neurology, London WC1N 3BG, UK
| | - Anthony Schapira
- Department of Clinical & Movement Neurosciences, UCL Institute of Neurology, London WC1N 3BG, UK
| | - Oliver Bandmann
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
| | - Stephen Mullin
- Faculty of Health, University of Plymouth, Plymouth PL4 9AA, UK
| | - Joy Duffen
- MRC Clinical Trials Unit at UCL, London WC1V 6LJ, UK
| | | | - Jeremy Chataway
- Department of Clinical & Movement Neurosciences, UCL Institute of Neurology, London WC1N 3BG, UK
| | - Mahesh Parmar
- MRC Clinical Trials Unit at UCL, London WC1V 6LJ, UK
| | - Camille Carroll
- Faculty of Health, University of Plymouth, Plymouth PL4 9AA, UK
| |
Collapse
|
9
|
Samudra N, Lane-Donovan C, VandeVrede L, Boxer AL. Tau pathology in neurodegenerative disease: disease mechanisms and therapeutic avenues. J Clin Invest 2023; 133:e168553. [PMID: 37317972 PMCID: PMC10266783 DOI: 10.1172/jci168553] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023] Open
Abstract
Tauopathies are disorders associated with tau protein dysfunction and insoluble tau accumulation in the brain at autopsy. Multiple lines of evidence from human disease, as well as nonclinical translational models, suggest that tau has a central pathologic role in these disorders, historically thought to be primarily related to tau gain of toxic function. However, a number of tau-targeting therapies with various mechanisms of action have shown little promise in clinical trials in different tauopathies. We review what is known about tau biology, genetics, and therapeutic mechanisms that have been tested in clinical trials to date. We discuss possible reasons for failures of these therapies, such as use of imperfect nonclinical models that do not predict human effects for drug development; heterogeneity of human tau pathologies which may lead to variable responses to therapy; and ineffective therapeutic mechanisms, such as targeting of the wrong tau species or protein epitope. Innovative approaches to human clinical trials can help address some of the difficulties that have plagued our field's development of tau-targeting therapies thus far. Despite limited clinical success to date, as we continue to refine our understanding of tau's pathogenic mechanism(s) in different neurodegenerative diseases, we remain optimistic that tau-targeting therapies will eventually play a central role in the treatment of tauopathies.
Collapse
|
10
|
Abdelnour C, Gonzalez MC, Gibson LL, Poston KL, Ballard CG, Cummings JL, Aarsland D. Dementia with Lewy Bodies Drug Therapies in Clinical Trials: Systematic Review up to 2022. Neurol Ther 2023; 12:727-749. [PMID: 37017910 PMCID: PMC10195935 DOI: 10.1007/s40120-023-00467-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 03/14/2023] [Indexed: 04/06/2023] Open
Abstract
INTRODUCTION Reviews of randomized clinical trials (RCTs) in dementia with Lewy bodies (DLB) are essential for informing ongoing research efforts of symptomatic therapies and potentially disease-modifying therapies (DMTs). METHODS We performed a systematic review of all clinical trials conducted until September 27, 2022, by examining 3 international registries: ClinicalTrials.gov, the European Union Drug Regulating Authorities Clinical Trials Database, and the International Clinical Trials Registry Platform, to identify drugs in trials in DLB. RESULTS We found 25 agents in 40 trials assessing symptomatic treatments and DMTs for DLB: 7 phase 3, 31 phase 2, and 2 phase 1 trials. We found an active pipeline for drug development in DLB, with most ongoing clinical trials in phase 2. We identified a recent trend towards including participants at the prodromal stages, although more than half of active clinical trials will enroll mild to moderate dementia patients. Additionally, repurposed agents are frequently tested, representing 65% of clinical trials. CONCLUSION Current challenges in DLB clinical trials include the need for disease-specific outcome measures and biomarkers, and improving representation of global and diverse populations.
Collapse
Affiliation(s)
- Carla Abdelnour
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.
| | - Maria Camila Gonzalez
- Department of Quality and Health Technology, Faculty of Health Sciences, University of Stavanger, Stavanger, Norway
- The Norwegian Centre for Movement Disorders, Stavanger University Hospital, Stavanger, Norway
- Centre for Age-Related Diseases, Stavanger University Hospital, Stavanger, Norway
| | - Lucy L Gibson
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Kathleen L Poston
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Jeffrey L Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Pam Quirk Brain Health and Biomarker Laboratory, Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas, Las Vegas, NV, USA
| | - Dag Aarsland
- Centre for Age-Related Diseases, Stavanger University Hospital, Stavanger, Norway
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| |
Collapse
|
11
|
Inagaki E, Yoshimatsu S, Okano H. Accelerated neuronal aging in vitro ∼melting watch ∼. Front Aging Neurosci 2022; 14:868770. [PMID: 36016855 PMCID: PMC9397486 DOI: 10.3389/fnagi.2022.868770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
In developed countries, the aging of the population and the associated increase in age-related diseases are causing major unresolved medical, social, and environmental matters. Therefore, research on aging has become one of the most important and urgent issues in life sciences. If the molecular mechanisms of the onset and progression of neurodegenerative diseases are elucidated, we can expect to develop disease-modifying methods to prevent neurodegeneration itself. Since the discovery of induced pluripotent stem cells (iPSCs), there has been an explosion of disease models using disease-specific iPSCs derived from patient-derived somatic cells. By inducing the differentiation of iPSCs into neurons, disease models that reflect the patient-derived pathology can be reproduced in culture dishes, and are playing an active role in elucidating new pathological mechanisms and as a platform for new drug discovery. At the same time, however, we are faced with a new problem: how to recapitulate aging in culture dishes. It has been pointed out that cells differentiated from pluripotent stem cells are juvenile, retain embryonic traits, and may not be fully mature. Therefore, attempts are being made to induce cell maturation, senescence, and stress signals through culture conditions. It has also been reported that direct conversion of fibroblasts into neurons can reproduce human neurons with an aged phenotype. Here, we outline some state-of-the-art insights into models of neuronal aging in vitro. New frontiers in which stem cells and methods for inducing differentiation of tissue regeneration can be applied to aging research are just now approaching, and we need to keep a close eye on them. These models are forefront and intended to advance our knowledge of the molecular mechanisms of aging and contribute to the development of novel therapies for human neurodegenerative diseases associated with aging.
Collapse
Affiliation(s)
- Emi Inagaki
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
- Japanese Society for the Promotion of Science (JSPS), Tokyo, Japan
| | - Sho Yoshimatsu
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
- *Correspondence: Hideyuki Okano,
| |
Collapse
|