1
|
Jang DG, Federico A, Savelieff MG, Grisold W, Mancuso M, Molnar MJ, Feldman EL, Reynolds EL. Awareness and care practices for rare neurologic diseases among senior neurologists: A global survey. J Neurol Sci 2025; 470:123395. [PMID: 39855013 PMCID: PMC11846688 DOI: 10.1016/j.jns.2025.123395] [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: 07/25/2024] [Revised: 01/09/2025] [Accepted: 01/12/2025] [Indexed: 01/27/2025]
Abstract
OBJECTIVE Rare neurologic diseases (RNDs) are difficult to diagnose and treat due to their low prevalence and complex nature. This survey evaluated awareness and current care status of RNDs among esteemed neurologists affiliated with the World Federation of Neurology (WFN). METHODS A 34-question survey was distributed to renowned neurologists, including delegates from national neurology societies in the WFN Assembly, various WFN committees, and members of the Rare Neurologic Diseases Specialist group. Responses were stratified by geographical regions, including Africa, the Americas, Asia/Oceania, and Europe, and into four income groups based on the World Bank Indicator. Descriptive statistics summarized responses, stratified by geographical regions or income groups, and significant differences were assessed by Fisher's exact test. RESULTS Of 190 invited neurologists, 64 responded (34 % response rate). Among respondents, 89 % agreed that RND patients should receive timely and effective care on par with more common neurological conditions. Additionally, 77 % of respondents overall thought most RNDs could be accurately diagnosed in their country. However, there were significant differences in the perceived ability of respondents' country of practice to diagnose RNDs by region, specifically in Africa (25 %), and by income of country of practice, specifically in the lower-income group (17 %). CONCLUSIONS This global survey highlights varying RND diagnosis and care by country socioeconomic status, suggesting potential disparities in resources and preparedness. To improve outcomes and quality-of-life for RND patients, efforts should focus on improving diagnostic capabilities, fostering collaboration among neurology centers, and promoting education on the unique challenges and treatment options of RNDs.
Collapse
Affiliation(s)
- Dae-Gyu Jang
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA; NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Antonio Federico
- Chair, Subspecialist Group of Rare Neurologic Diseases, World Federation of Neurology, UK; Department Medicine, Surgery and Neurosciences, Medical School, University of Siena, Viale Bracci 2, 53100 Siena, Italy.
| | - Masha G Savelieff
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, ND 58202, USA.
| | - Wolfgang Grisold
- World Federation of Neurology, UK; Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Donaueschingenstraße 13, A-1200 Vienna, Austria.
| | - Michelangelo Mancuso
- Neurological Institute, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy.
| | - Maria J Molnar
- Institute of Genomic Medicine and Rare Disorders, Semmelweis University, 1085 Budapest, Hungary.
| | - Eva L Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA; NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Evan L Reynolds
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA; NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI 48109, USA; Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI 48824, USA.
| |
Collapse
|
2
|
Wiśniewska K, Wolski J, Anikiej-Wiczenbach P, Żabińska M, Węgrzyn G, Pierzynowska K. Behavioural disorders and sleep problems in Sanfilippo syndrome: overlaps with some other conditions and importance indications. Eur Child Adolesc Psychiatry 2025:10.1007/s00787-025-02661-5. [PMID: 40087177 DOI: 10.1007/s00787-025-02661-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 02/07/2025] [Indexed: 03/17/2025]
Abstract
Sanfilippo syndrome (MPS III) is one of the types of mucopolysaccharidoses (MPS), a group of inherited metabolic diseases in which the accumulation of glycosaminoglycans (GAGs) results from deficiency of different lysosomal enzymes. The hallmarks of MPS III are relatively minor somatic abnormalities with severe and progressive central nervous system (CNS) symptoms. An analysis of the literature showed that the biggest problems for carers of people with MPS III are behavioural disorders and sleep disorders. Despite extensive discussions on improving the quality of life of patients, little attention was paid to the families/carers of patients. The families/carers are providing appropriate medical and palliative care to the patient every day due to their loss of mobility, self-care skills, tube feeding, airway clearance and other supports continue to have an adverse effect on the quality of life of families/carers. However, a literature review of possible solutions showed that effective methods (both pharmacological and non-pharmacological) exist. The needs of carers of MPS III patients should receive as much attention as the search for new treatments. There are many options for dealing with such problems. The key issue is to identify the source of the problem and choose the most effective therapy. Alleviating behavioural disorders, pain complaints and sleep problems will have a positive impact not only on the quality of life of carers/families, but also on the patients themselves.
Collapse
Affiliation(s)
- Karolina Wiśniewska
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, 80-308, Gdansk, Poland
| | - Jakub Wolski
- Psychiatry Ward, 7, Navy Hospital in Gdańsk, Polanki 117, 80-305, Gdańsk, Poland
| | | | - Magdalena Żabińska
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, 80-308, Gdansk, Poland
| | - Grzegorz Węgrzyn
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, 80-308, Gdansk, Poland
| | - Karolina Pierzynowska
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, 80-308, Gdansk, Poland.
| |
Collapse
|
3
|
Gangwal A, Lavecchia A. AI-Driven Drug Discovery for Rare Diseases. J Chem Inf Model 2025; 65:2214-2231. [PMID: 39689164 DOI: 10.1021/acs.jcim.4c01966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
Rare diseases (RDs), affecting 300 million people globally, present a daunting public health challenge characterized by complexity, limited treatment options, and diagnostic hurdles. Despite legislative efforts, such as the 1983 US Orphan Drug Act, more than 90% of RDs lack effective therapies. Traditional drug discovery models, marked by lengthy development cycles and high failure rates, struggle to meet the unique demands of RDs, often yielding poor returns on investment. However, the advent of artificial intelligence (AI), encompassing machine learning (ML) and deep learning (DL), offers groundbreaking solutions. This review explores AI's potential to revolutionize drug discovery for RDs by overcoming these challenges. It discusses AI-driven advancements, such as drug repurposing, biomarker discovery, personalized medicine, genetics, clinical trial optimization, corporate innovations, and novel drug target identification. By synthesizing current knowledge and recent breakthroughs, this review provides crucial insights into how AI can accelerate therapeutic development for RDs, ultimately improving patient outcomes. This comprehensive analysis fills a critical gap in the literature, enhancing understanding of AI's pivotal role in transforming RD research and guiding future research and development efforts in this vital area of medicine.
Collapse
Affiliation(s)
- Amit Gangwal
- Department of Natural Product Chemistry, Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule 424001, Maharashtra, India
| | - Antonio Lavecchia
- "Drug Discovery" Laboratory, Department of Pharmacy, University of Naples Federico II, I-80131 Naples, Italy
| |
Collapse
|
4
|
Wiesinger A, Bigger B, Giugliani R, Lampe C, Scarpa M, Moser T, Kampmann C, Zimmermann G, Lagler FB. Development of a novel tool for individual treatment trials in mucopolysaccharidosis. J Inherit Metab Dis 2025; 48:e12816. [PMID: 39572375 PMCID: PMC11670214 DOI: 10.1002/jimd.12816] [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: 08/13/2024] [Revised: 10/04/2024] [Accepted: 11/05/2024] [Indexed: 12/28/2024]
Abstract
Mucopolysaccharidosis (MPS) encompasses a group of genetic lysosomal storage disorders, linked to reduced life expectancy and a significant lack of effective treatment options. Immunomodulatory drugs could have the potential to be a relevant medical approach, as the accumulation of undegraded substances initiates an innate immune response, which leads to inflammation and clinical deterioration. However, immunomodulators are not licensed for this indication. Consequently, we aim to provide evidence advocating fast access to innovative individual treatment trials (ITTs) with immunomodulatory drugs and high-quality evaluation of drug effects by implementing a risk-benefit model tailored for MPS. The iterative methodology of our novel decision analysis framework (DAF) involves three key steps: (i) literature review on promising treatment targets and immunomodulators in MPS; (ii) quantitative risk-benefit assessment (RBA) of selected molecules; (iii) assigning phenotypic profiles and quantitative evaluations. The results facilitate a personalized application of the model and are based on published evidence as well as interdisciplinary experts' consensus and patient perspectives. Four promising immunomodulators have been identified: adalimumab, abatacept, anakinra, and cladribine. An improvement in mobility is most likely with adalimumab, while anakinra is anticipated as a treatment of choice for neuronopathic MPS patients. Nevertheless, a comprehensive RBA should always be completed on an individual basis. Our evidence-based DAF tool for ITTs directly addresses the substantial unmet medical need in MPS and characterizes an initial stride toward precision medicine with immunomodulators.
Collapse
Affiliation(s)
- Anna‐Maria Wiesinger
- Institute of Congenital Metabolic DiseasesParacelsus Medical UniversitySalzburgAustria
- European Reference Network for Hereditary Metabolic DiseasesMetabERNUdineItaly
| | - Brian Bigger
- European Reference Network for Hereditary Metabolic DiseasesMetabERNUdineItaly
- Institute for Regeneration and RepairThe University of EdinburghEdinburghUK
| | - Roberto Giugliani
- Department of Genetics, UFRGS, Medical Genetics Service and Biodiscovery LaboratoryHCPA, INAGEMP, DASA, and Casa dos RarosPorto AlegreBrazil
| | - Christina Lampe
- European Reference Network for Hereditary Metabolic DiseasesMetabERNUdineItaly
- Department of Child Neurology, Epileptology and Social Pediatrics, Center of Rare DiseasesUniversity Hospital GiessenMarburgGermany
| | - Maurizio Scarpa
- European Reference Network for Hereditary Metabolic DiseasesMetabERNUdineItaly
- Regional Coordinating Center for Rare DiseasesUniversity Hospital UdineUdineItaly
| | - Tobias Moser
- Department of NeurologyChristian Doppler University Hospital, Paracelsus Medical UniversitySalzburgAustria
| | | | - Georg Zimmermann
- Team Biostatistics and Big Medical DataIDA Lab Salzburg, Paracelsus Medical UniversitySalzburgAustria
- Department of Artificial Intelligence and Human Interfaces, Faculty of Digital and Analytical SciencesParis Lodron UniversitySalzburgAustria
| | - Florian B. Lagler
- Institute of Congenital Metabolic DiseasesParacelsus Medical UniversitySalzburgAustria
- European Reference Network for Hereditary Metabolic DiseasesMetabERNUdineItaly
| |
Collapse
|
5
|
Francisco S, Lamacchia L, Turco A, Ermondi G, Caron G, Rossi Sebastiano M. Restoring adapter protein complex 4 function with small molecules: an in silico approach to spastic paraplegia 50. Protein Sci 2025; 34:e70006. [PMID: 39723768 DOI: 10.1002/pro.70006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 11/22/2024] [Accepted: 12/06/2024] [Indexed: 12/28/2024]
Abstract
This study focuses on spastic paraplegia type 50 (SPG50), an adapter protein complex 4 deficiency syndrome caused by mutations in the adapter protein complex 4 subunit mu-1 (AP4M1) gene, and on the downstream alterations of the AP4M1 protein. We applied a battery of heterogeneous computational resources, encompassing two in-house tools described here for the first time, to (a) assess the druggability potential of AP4M1, (b) characterize SPG50-associated mutations and their 3D scenario, (c) identify mutation-tailored drug candidates for SPG50, and (d) elucidate their mechanisms of action by means of structural considerations on homology models of the adapter protein complex 4 core. Altogether, the collected results indicate R367Q as the mutation with the most promising potential of being corrected by small-molecule drugs, and the flavonoid rutin as best candidate for this purpose. Rutin shows promise in rescuing the interaction between the AP4M1 and adapter protein complex subunit beta-1 (AP4B1) subunits by means of a glue-like mode of action. Overall, this approach offers a framework that could be systematically applied to the investigation of mutation-wise molecular mechanisms in different hereditary spastic paraplegias, too.
Collapse
Affiliation(s)
- Serena Francisco
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Lorenzo Lamacchia
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Attilio Turco
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Giuseppe Ermondi
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Giulia Caron
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Matteo Rossi Sebastiano
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| |
Collapse
|
6
|
Stolwijk NN, Häberle J, Huidekoper HH, Wagenmakers MAEM, Hollak CEM, Bosch AM. Mapping challenges in the accessibility of treatment products for urea cycle disorders: A survey of European healthcare professionals. J Inherit Metab Dis 2025; 48:e12815. [PMID: 39625308 PMCID: PMC11670152 DOI: 10.1002/jimd.12815] [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: 08/30/2024] [Revised: 10/17/2024] [Accepted: 11/04/2024] [Indexed: 12/28/2024]
Abstract
Current management guidelines for urea cycle disorders (UCDs) offer clear strategies, incorporating both authorized and non-authorized medicinal products (including intravenous formulations and products regulated as food). These varying product categories are subject to specific accessibility challenges related to availability, reimbursement, and pricing. The aim of this study is to identify potential obstacles to optimal UCD treatment implementation in European clinical practice. A survey aimed at metabolic healthcare professionals (HCPs) managing patients with UCDs in Europe was disseminated through the European Reference Network for Hereditary Metabolic Disorders and the European registry and network for intoxication type metabolic diseases. Forty-eight survey responses were collected from 21 European countries. In 16 of these countries, at least one metabolic HCP reported challenges in accessing UCD products. Reimbursement issues were reported for most products (8/10), including both authorized and non-authorized products. Availability-related challenges were also reported for 8/10 products, although unavailability was limited to non-authorized products. Prices impacted accessibility for all authorized products (3/3) and one non-authorized IV product. The accessibility of UCD treatment products varied across Europe, although no clear regional variations could be discerned. Survey data revealed that metabolic HCPs experience challenges in accessing both authorized and non-authorized products for UCD management in the majority of European countries. This indicates that registering unauthorized products may not resolve all issues. Improved reimbursement policies and fair pricing models, as well as (adjusted) authorization procedures may help address these concerns, thereby optimizing treatment access for UCD patients.
Collapse
Affiliation(s)
- Nina N. Stolwijk
- Medicines for Society (Medicijn voor de Maatschappij)Platform at Amsterdam UMC ‐ University of AmsterdamAmsterdamThe Netherlands
- Department of Endocrinology and Metabolism, Amsterdam UMC, Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Research Institute, Expertise Center for inborn errors of Metabolism, MetabERNUniversity of AmsterdamAmsterdamThe Netherlands
| | - Johannes Häberle
- Division of Metabolism and Children's Research CenterUniversity Children's Hospital ZurichZurichSwitzerland
| | - Hidde H. Huidekoper
- Department of Pediatrics, Center for Lysosomal and Metabolic Diseases, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| | - Margreet A. E. M. Wagenmakers
- Department of Internal Medicine, Center for Lysosomal and Metabolic Diseases, Erasmus MCErasmus University Medical Centre RotterdamRotterdamNetherlands
| | - Carla E. M. Hollak
- Medicines for Society (Medicijn voor de Maatschappij)Platform at Amsterdam UMC ‐ University of AmsterdamAmsterdamThe Netherlands
- Department of Endocrinology and Metabolism, Amsterdam UMC, Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Research Institute, Expertise Center for inborn errors of Metabolism, MetabERNUniversity of AmsterdamAmsterdamThe Netherlands
| | - Annet M. Bosch
- Department of Pediatrics, Division of Metabolic Diseases, Emma Children's Hospital, Amsterdam Gastroenterology Endocrinology Metabolism (AGEM)Amsterdam UMC location University of AmsterdamAmsterdamThe Netherlands
| |
Collapse
|
7
|
Jonker AH, Tataru EA, Graessner H, Dimmock D, Jaffe A, Baynam G, Davies J, Mitkus S, Iliach O, Horgan R, Augustine EF, Bateman-House A, Pasmooij AMG, Yu T, Synofzik M, Douville J, Lapteva L, Brooks PJ, O'Connor D, Aartsma-Rus A. The state-of-the-art of N-of-1 therapies and the IRDiRC N-of-1 development roadmap. Nat Rev Drug Discov 2025; 24:40-56. [PMID: 39496921 DOI: 10.1038/s41573-024-01059-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/19/2024] [Indexed: 11/06/2024]
Abstract
In recent years, a small number of people with rare diseases caused by unique genetic variants have been treated with therapies developed specifically for them. This pioneering field of genetic N-of-1 therapies is evolving rapidly, giving hope for the individualized treatment of people living with very rare diseases. In this Review, we outline the concept of N-of-1 individualized therapies, focusing on genetic therapies, and illustrate advances and challenges in the field using cases for which therapies have been successfully developed. We discuss why the traditional drug development and reimbursement pathway is not fit for purpose in this field, and outline the pragmatic, regulatory and ethical challenges this poses for future access to N-of-1 therapies. Finally, we provide a roadmap for N-of-1 individualized therapy development.
Collapse
Affiliation(s)
| | - Elena-Alexandra Tataru
- French National Institute of Health and Medical Research (INSERM), Paris, France
- International Rare Diseases Research Consortium (IRDiRC), Paris, France
| | - Holm Graessner
- Centre for Rare Diseases, University Hospital Tübingen, Tübingen, Germany
| | | | - Adam Jaffe
- School of Clinical Medicine, University of New South Wales, Faculty of Medicine, Sydney, New South Wales, Australia
| | - Gareth Baynam
- Rare Care Centre, Perth Children's Hospital, Perth, Western Australia, Australia
- University of Western Australia, Faculty of Health and Medical Sciences, Division of Paediatrics and Telethon Kids Institute, Perth, Western Australia, Australia
- University of Notre Dame, Medical Faculty, Sydney, New South Wales, Australia
| | - James Davies
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- NIHR Blood and Transplant Research Unit in Precision Cellular Therapeutics, University of Oxford, Oxford, UK
| | - Shruti Mitkus
- Patient Services, Global Genes, Aliso Viejo, CA, USA
| | - Oxana Iliach
- Regulatory Strategy and Policy, Certara, Toronto, Ontario, Canada
- Canadian Organization for Rare Disorders (CORD), Toronto, Ontario, Canada
| | | | | | - Alison Bateman-House
- Division of Medical Ethics, Department of Population Health, NYU Grossman School of Medicine, New York, NY, USA
| | - Anna Maria Gerdina Pasmooij
- Science Department, Dutch Medicines Evaluation Board, Utrecht, the Netherlands
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| | - Tim Yu
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA
| | - Matthis Synofzik
- Research Division Translational Genomics of Neurodegenerative Diseases, Hertie-Institute of Clinical Brain Research, University of Tübingen, Tübingen, Germany
- Division of Translational Genomics of Neurodegenerative Diseases, German Center for Neurodegenerative Diseases, Tübingen, Germany
| | - Julie Douville
- ASO Discovery and Development, n-Lorem Foundation, Carlsbad, CA, USA
| | - Larissa Lapteva
- Division of Clinical Evaluation Pharmacology and Toxicology, Food and Drug Administration, Silver Spring, MD, USA
| | - Philip John Brooks
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Daniel O'Connor
- Regulatory Policy & Early Access, The Association of the British Pharmaceutical Industry (ABPI), London, UK
| | - Annemieke Aartsma-Rus
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
8
|
Zaph S, Leiser RJ, Tao M, Kaddi C, Xu C. Application of Quantitative Systems Pharmacology Approaches to Support Pediatric Labeling in Rare Diseases. Handb Exp Pharmacol 2024. [PMID: 39673036 DOI: 10.1007/164_2024_734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2024]
Abstract
Quantitative Systems Pharmacology (QSP) models offer a promising approach to extrapolate drug efficacy across different patient populations, particularly in rare diseases. Unlike conventional empirical models, QSP models can provide a mechanistic understanding of disease progression and therapeutic response by incorporating current disease knowledge into the descriptions of biomarkers and clinical endpoints. This allows for a holistic representation of the disease and drug response. The mechanistic nature of QSP models is well suited to pediatric extrapolation concepts, providing a quantitative method to assess disease and drug response similarity between adults and pediatric patients. The application of a QSP-based assessment of the disease and drug similarity in adult and pediatric patients in the clinical development program of olipudase alfa, a treatment for Acid Sphingomyelinase Deficiency (ASMD), illustrates the potential of this approach.
Collapse
Affiliation(s)
- Susana Zaph
- Translational Disease Modeling - Translational Medicine, Research & Development, Sanofi-US, Bridgewater, NJ, USA.
| | - Randolph J Leiser
- Translational Disease Modeling - Translational Medicine, Research & Development, Sanofi-US, Bridgewater, NJ, USA
| | - Mengdi Tao
- Translational Disease Modeling - Translational Medicine, Research & Development, Sanofi-US, Bridgewater, NJ, USA
| | - Chanchala Kaddi
- Translational Disease Modeling - Translational Medicine, Research & Development, Sanofi-US, Bridgewater, NJ, USA
| | - Christine Xu
- Pharmacokinetics, Dynamics, Metabolism - Translational Medicine, Research & Development, Sanofi-US, Bridgewater, NJ, USA
| |
Collapse
|
9
|
Neves-Zaph S, Kaddi C. Quantitative Systems Pharmacology Models: Potential Tools for Advancing Drug Development for Rare Diseases. Clin Pharmacol Ther 2024; 116:1442-1451. [PMID: 39340225 DOI: 10.1002/cpt.3451] [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: 06/09/2024] [Accepted: 09/08/2024] [Indexed: 09/30/2024]
Abstract
Rare diseases, affecting millions globally, present significant drug development challenges. This is due to the limited patient populations and the unique pathophysiology of these diseases, which can make traditional clinical trial designs unfeasible. Quantitative Systems Pharmacology (QSP) models offer a promising approach to expedite drug development, particularly in rare diseases. QSP models provide a mechanistic representation of the disease and drug response in virtual patients that can complement routinely applied empirical modeling and simulation approaches. QSP models can generate digital twins of actual patients and mechanistically simulate the disease progression of rare diseases, accounting for phenotypic heterogeneity. QSP models can also support drug development in various drug modalities, such as gene therapy. Impactful QSP models case studies are presented here to illustrate their value in supporting various aspects of drug development in rare indications. As these QSP model applications continue to mature, there is a growing possibility that they could be more widely integrated into routine drug development steps. This integration could provide a robust framework for addressing some of the inherent challenges in rare disease drug development.
Collapse
Affiliation(s)
- Susana Neves-Zaph
- Translational Disease Modeling, Translational Medicine and Early Development, Sanofi US, Bridgewater, New Jersey, USA
| | - Chanchala Kaddi
- Translational Disease Modeling, Translational Medicine and Early Development, Sanofi US, Bridgewater, New Jersey, USA
| |
Collapse
|
10
|
Subramanian S, Jain M, Misra R, Jain R. Peptide-based therapeutics targeting genetic disorders. Drug Discov Today 2024; 29:104209. [PMID: 39419376 DOI: 10.1016/j.drudis.2024.104209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 09/25/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024]
Abstract
Genetic disorders (GDs) are challenging to treat owing to a lack of optimal treatment regimens and intricate and often difficult-to-understand underlying biological processes. Limited therapeutic approaches, which mostly provide symptomatic relief, are available. To date, a limited number of peptide-based drugs for the treatment of GDs are available, and several candidates are under clinical study. This review provides mechanistic insights into GDs and potential target areas where peptide-based drugs are beneficial. In addition, it emphasizes the usefulness of peptides as carriers for gene delivery, biomarkers for mutation detection and peptide-based vaccines for treating GDs.
Collapse
Affiliation(s)
- Shweta Subramanian
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Sector 67, S. A. S. Nagar, Punjab 160 062, India
| | - Meenakshi Jain
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Sector 67, S. A. S. Nagar, Punjab 160 062, India
| | - Rajkumar Misra
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Sector 67, S. A. S. Nagar, Punjab 160 062, India
| | - Rahul Jain
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Sector 67, S. A. S. Nagar, Punjab 160 062, India.
| |
Collapse
|
11
|
Gonzalez-Rivera JC, Galvan A, Ryder T, Milman M, Agarwal K, Kandari L, Khetan A. A high-titer scalable Chinese hamster ovary transient expression platform for production of biotherapeutics. Biotechnol Bioeng 2024; 121:3454-3470. [PMID: 39101569 DOI: 10.1002/bit.28817] [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: 01/31/2024] [Revised: 07/08/2024] [Accepted: 07/18/2024] [Indexed: 08/06/2024]
Abstract
Transient gene expression (TGE) in Chinese hamster ovary (CHO) cells offers a route to accelerate biologics development by delivering material weeks to months earlier than what is possible with conventional cell line development. However, low productivity, inconsistent product quality profiles, and scalability challenges have prevented its broader adoption. In this study, we develop a scalable CHO-based TGE system achieving 1.9 g/L of monoclonal antibody in an unmodified host. We integrated continuous flow-electroporation and alternate tangential flow (ATF) perfusion to enable an end-to-end closed system from N-1 perfusion to fed-batch 50-L bioreactor production. Optimization of both the ATF operation for three-in-one application-cell growth, buffer exchange, and cell mass concentration-and the flow-electroporation process, led to a platform for producing biotherapeutics using transiently transfected cells. We demonstrate scalability up to 50-L bioreactor, maintaining a titer over 1 g/L. We also show comparable quality between both transiently and stably produced material, and consistency across batches. The results confirm that purity, charge variants and N-glycan profiles are similar. Our study demonstrates the potential of CHO-based TGE platforms to accelerate biologics process development timelines and contributes evidence supporting its feasibility for manufacturing early clinical material, aiming to strengthen endorsement for TGE's wider implementation.
Collapse
Affiliation(s)
| | - Alberto Galvan
- Biologics Development, Bristol Myers Squibb, New Brunswick, New Jersey, USA
| | - Todd Ryder
- Biologics Development, Bristol Myers Squibb, New Brunswick, New Jersey, USA
| | - Monica Milman
- Biologics Development, Bristol Myers Squibb, New Brunswick, New Jersey, USA
| | - Kitty Agarwal
- Biologics Development, Bristol Myers Squibb, New Brunswick, New Jersey, USA
| | - Lakshmi Kandari
- Biologics Development, Bristol Myers Squibb, New Brunswick, New Jersey, USA
| | - Anurag Khetan
- Biologics Development, Bristol Myers Squibb, New Brunswick, New Jersey, USA
| |
Collapse
|
12
|
Caferra P, Fraisse T, Trincavelli ML, Marchetti L, Piras AM. Evaluation of orphan maintained biological medicinal products in the European Union between 2018 to 2023: a regulatory perspective. Expert Opin Biol Ther 2024; 24:1279-1297. [PMID: 39460383 DOI: 10.1080/14712598.2024.2422360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 10/18/2024] [Accepted: 10/24/2024] [Indexed: 10/28/2024]
Abstract
OBJECTIVES Orphan medicinal products (OMPs) authorized by the European Union (EU) benefit from market exclusivity, fee waivers, and national incentives. Maintaining orphan status during a marketing authorization application requires meeting eligibility criteria, especially demonstrating significant benefit (SB), which is challenging. This study identifies key features linked to successful orphan status maintenance for biological OMPs approved in the EU between 2018 and 2023. METHODS Data from European public assessment reports and orphan maintenance assessment reports were analyzed. RESULTS Among the 50 biological OMP maintained orphan designations, 68.0% had to demonstrate SB over existing treatments, with 91.2% leveraging the clinically relevant advantage area, utilizing better clinical efficacy (83.9%) and efficacy in subpopulations (38.7%) subdomains. However, 32.0% did not need to demonstrate SB due to a lack of alternative treatments, most of which were ultra-orphan drugs. Advanced therapy medicinal products and monoclonal antibodies were the most numerous OMP categories, whereas oncology and immunomodulation were the preferred therapeutic areas. CONCLUSION The Orphan Regulation is essential in advancing treatments for rare diseases, fostering innovation while addressing unmet medical needs. Nonetheless, the insufficient return on investment criterion remains underused, whereas refining major contribution to patient care guidelines and incorporating real-world evidence may enhance regulatory evaluations.
Collapse
Affiliation(s)
- Paolo Caferra
- Department of Pharmacy, University of Pisa, Pisa, Italy
- Reseach & Development, Sanofi, Amsterdam, the Netherlands
| | - Thomas Fraisse
- Reseach & Development, Sanofi, Amsterdam, the Netherlands
- Faculty of Pharmacy, Montpellier University, Montpellier, France
| | | | | | | |
Collapse
|
13
|
Yoo HW. Development of orphan drugs for rare diseases. Clin Exp Pediatr 2024; 67:315-327. [PMID: 37402468 PMCID: PMC11222908 DOI: 10.3345/cep.2023.00535] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/12/2023] [Accepted: 06/28/2023] [Indexed: 07/06/2023] Open
Abstract
Most rare diseases (orphan diseases) still lack approved treatment options despite major advances in research providing the necessary tools to understand their molecular basis and legislation providing regulatory and economic incentives to expedite the development of specific therapies. Addressing this translational gap is a multifaceted challenge, a key aspect of which is the selection of an optimal therapeutic modality to translate advances in rare disease knowledge to potential medicines known as orphan drugs. There are several strategies for developing orphan drugs for rare genetic disorders, including protein replacement therapies, small-molecule therapies (e.g., substrate reduction, chemical chaperone, cofactor, expression modification, and read-through therapies), monoclonal antibodies, antisense oligonucleotides, small interfering RNA or exon skipping therapies, gene replacement and direct genome-editing therapies, mRNA therapy, cell therapy, and drug repurposing. Each strategy has its own strengths and limitations in orphan drug development. Furthermore, numerous hurdles are present in clinical trials of rare genetic diseases because of difficulty with patient recruitment, unknown molecular physiology, the natural history of the disease, ethical concerns regarding pediatric patients, and regulatory challenges. To address these barriers, the rare genetic diseases community, including academic institutions, industry, patient advocacy groups, foundations, payers, and government regulatory and research organizations, must become engaged in discussions about these issues.
Collapse
Affiliation(s)
- Han-Wook Yoo
- Department of Pediatrics, Bundang CHA Medical Center, CHA University School of Medicine, Seongnam, Korea
| |
Collapse
|
14
|
Pimentel-Vera LN, Rodríguez-López A, Espejo-Mojica AJ, Ramírez AM, Cardona C, Reyes LH, Tomatsu S, Jaroentomeechai T, DeLisa MP, Sánchez OF, Alméciga-Díaz CJ. Novel human recombinant N-acetylgalactosamine-6-sulfate sulfatase produced in a glyco-engineered Escherichia coli strain. Heliyon 2024; 10:e32555. [PMID: 38952373 PMCID: PMC11215262 DOI: 10.1016/j.heliyon.2024.e32555] [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: 02/19/2024] [Revised: 05/14/2024] [Accepted: 06/05/2024] [Indexed: 07/03/2024] Open
Abstract
Mucopolysaccharidosis IVA (MPS IVA) is a lysosomal storage disease caused by mutations in the gene encoding the lysosomal enzyme N-acetylgalactosamine-6-sulfate sulfatase (GALNS), resulting in the accumulation of keratan sulfate (KS) and chondroitin-6-sulfate (C6S). Previously, it was reported the production of an active human recombinant GALNS (rGALNS) in E. coli BL21(DE3). However, this recombinant enzyme was not taken up by HEK293 cells or MPS IVA skin fibroblasts. Here, we leveraged a glyco-engineered E. coli strain to produce a recombinant human GALNS bearing the eukaryotic trimannosyl core N-glycan, Man3GlcNAc2 (rGALNSoptGly). The N-glycosylated GALNS was produced at 100 mL and 1.65 L scales, purified and characterized with respect to pH stability, enzyme kinetic parameters, cell uptake, and KS clearance. The results showed that the addition of trimannosyl core N-glycans enhanced both protein stability and substrate affinity. rGALNSoptGly was capture through a mannose receptor-mediated process. This enzyme was delivered to the lysosome, where it reduced KS storage in human MPS IVA fibroblasts. This study demonstrates the potential of a glyco-engineered E. coli for producing a fully functional GALNS enzyme. It may offer an economic approach for the biosynthesis of a therapeutic glycoprotein that could prove useful for MPS IVA treatment. This strategy could be extended to other lysosomal enzymes that rely on the presence of mannose N-glycans for cell uptake.
Collapse
Affiliation(s)
- Luisa N. Pimentel-Vera
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá, D.C., 110231, Colombia
| | - Alexander Rodríguez-López
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá, D.C., 110231, Colombia
- Dogma Biotech, Bogotá, D.C., 110111, Colombia
| | - Angela J. Espejo-Mojica
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá, D.C., 110231, Colombia
- Dogma Biotech, Bogotá, D.C., 110111, Colombia
| | - Aura María Ramírez
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá, D.C., 110231, Colombia
| | - Carolina Cardona
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá, D.C., 110231, Colombia
- Grupo de Investigaciones Biomédicas y de Genética Humana Aplicada GIBGA, Facultad de Ciencias de la Salud, Universidad de Ciencias Aplicadas y Ambientales U.D.C.A, Bogotá, D.C., Colombia
| | - Luis H. Reyes
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá, D.C., 110231, Colombia
- Grupo de Diseño de Productos y Procesos (GDPP), Department of Chemical and Food Engineering, Universidad de los Andes, Bogotá, D.C., Colombia
| | - Shunji Tomatsu
- Nemours Children's Health, Wilmington, DE, 19803, USA
- Faculty of Arts and Sciences, University of Delaware, Newark, DE, 19716, USA
- Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu, 501-1193, Japan
- Department of Pediatrics, Thomas Jefferson University, Philadelphia, PA, 19144, USA
| | - Thapakorn Jaroentomeechai
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY, USA
| | - Matthew P. DeLisa
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY, USA
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
- Cornell Institute of Biotechnology, Cornell University, Ithaca, NY, USA
| | - Oscar F. Sánchez
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, USA
| | - Carlos J. Alméciga-Díaz
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá, D.C., 110231, Colombia
| |
Collapse
|
15
|
Saffari A, Brechmann B, Böger C, Saber WA, Jumo H, Whye D, Wood D, Wahlster L, Alecu JE, Ziegler M, Scheffold M, Winden K, Hubbs J, Buttermore ED, Barrett L, Borner GHH, Davies AK, Ebrahimi-Fakhari D, Sahin M. High-content screening identifies a small molecule that restores AP-4-dependent protein trafficking in neuronal models of AP-4-associated hereditary spastic paraplegia. Nat Commun 2024; 15:584. [PMID: 38233389 PMCID: PMC10794252 DOI: 10.1038/s41467-023-44264-1] [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: 06/07/2023] [Accepted: 12/06/2023] [Indexed: 01/19/2024] Open
Abstract
Unbiased phenotypic screens in patient-relevant disease models offer the potential to detect therapeutic targets for rare diseases. In this study, we developed a high-throughput screening assay to identify molecules that correct aberrant protein trafficking in adapter protein complex 4 (AP-4) deficiency, a rare but prototypical form of childhood-onset hereditary spastic paraplegia characterized by mislocalization of the autophagy protein ATG9A. Using high-content microscopy and an automated image analysis pipeline, we screened a diversity library of 28,864 small molecules and identified a lead compound, BCH-HSP-C01, that restored ATG9A pathology in multiple disease models, including patient-derived fibroblasts and induced pluripotent stem cell-derived neurons. We used multiparametric orthogonal strategies and integrated transcriptomic and proteomic approaches to delineate potential mechanisms of action of BCH-HSP-C01. Our results define molecular regulators of intracellular ATG9A trafficking and characterize a lead compound for the treatment of AP-4 deficiency, providing important proof-of-concept data for future studies.
Collapse
Affiliation(s)
- Afshin Saffari
- Department of Neurology & F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Division of Child Neurology and Inherited Metabolic Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Barbara Brechmann
- Department of Neurology & F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Cedric Böger
- Department of Neurology & F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Wardiya Afshar Saber
- Department of Neurology & F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Hellen Jumo
- Department of Neurology & F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Dosh Whye
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Delaney Wood
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Lara Wahlster
- Department of Hematology & Oncology, Boston Children's Hospital & Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Julian E Alecu
- Department of Neurology & F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Marvin Ziegler
- Department of Neurology & F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Marlene Scheffold
- Department of Neurology & F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Kellen Winden
- Department of Neurology & F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Jed Hubbs
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Elizabeth D Buttermore
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Lee Barrett
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Georg H H Borner
- Department of Proteomics and Signal Transduction, Max-Planck-Institute of Biochemistry, Martinsried, 82152, Germany
| | - Alexandra K Davies
- Department of Proteomics and Signal Transduction, Max-Planck-Institute of Biochemistry, Martinsried, 82152, Germany
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, M13 9PT, UK
| | - Darius Ebrahimi-Fakhari
- Department of Neurology & F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
- Movement Disorders Program, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| | - Mustafa Sahin
- Department of Neurology & F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
16
|
D'Aloia A, Pastori V, Blasa S, Campioni G, Peri F, Sacco E, Ceriani M, Lecchi M, Costa B. A new advanced cellular model of functional cholinergic-like neurons developed by reprogramming the human SH-SY5Y neuroblastoma cell line. Cell Death Discov 2024; 10:24. [PMID: 38216593 PMCID: PMC10786877 DOI: 10.1038/s41420-023-01790-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 12/11/2023] [Accepted: 12/20/2023] [Indexed: 01/14/2024] Open
Abstract
Modeling human neuronal properties in physiological and pathological conditions is essential to identify novel potential drugs and to explore pathological mechanisms of neurological diseases. For this purpose, we generated a three-dimensional (3D) neuronal culture, by employing the readily available human neuroblastoma SH-SY5Y cell line, and a new differentiation protocol. The entire differentiation process occurred in a matrix and lasted 47 days, with 7 days of pre-differentiation phase and 40 days of differentiation, and allowed the development of a 3D culture in conditions consistent with the physiological environment. Neurons in the culture were electrically active, were able to establish functional networks, and showed features of cholinergic neurons. Hence here we provide an easily accessible, reproducible, and suitable culture method that might empower studies on synaptic function, vesicle trafficking, and metabolism, which sustain neuronal activity and cerebral circuits. Moreover, this novel differentiation protocol could represent a promising cellular tool to study physiological cellular processes, such as migration, differentiation, maturation, and to develop novel therapeutic approaches.
Collapse
Affiliation(s)
- Alessia D'Aloia
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126, Milano, Italy.
| | - Valentina Pastori
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126, Milano, Italy
| | - Stefania Blasa
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126, Milano, Italy
- Milan Center for Neuroscience (NeuroMI), University of Milano-Bicocca, Piazza dell'Ateneo Nuovo 1, 20126, Milano, Italy
| | - Gloria Campioni
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126, Milano, Italy
- SYSBIO-ISBE-IT, Europe, 20126, Milano, Italy
| | - Francesco Peri
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126, Milano, Italy
| | - Elena Sacco
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126, Milano, Italy
- Milan Center for Neuroscience (NeuroMI), University of Milano-Bicocca, Piazza dell'Ateneo Nuovo 1, 20126, Milano, Italy
- SYSBIO-ISBE-IT, Europe, 20126, Milano, Italy
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research, Pisa, Italy
| | - Michela Ceriani
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126, Milano, Italy
- Milan Center for Neuroscience (NeuroMI), University of Milano-Bicocca, Piazza dell'Ateneo Nuovo 1, 20126, Milano, Italy
| | - Marzia Lecchi
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126, Milano, Italy
- Milan Center for Neuroscience (NeuroMI), University of Milano-Bicocca, Piazza dell'Ateneo Nuovo 1, 20126, Milano, Italy
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research, Pisa, Italy
| | - Barbara Costa
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126, Milano, Italy
- Milan Center for Neuroscience (NeuroMI), University of Milano-Bicocca, Piazza dell'Ateneo Nuovo 1, 20126, Milano, Italy
| |
Collapse
|
17
|
Yokoshiki S, Arato T. Implementation status of pharmacological studies in the development of orphan drugs. Orphanet J Rare Dis 2024; 19:5. [PMID: 38167464 PMCID: PMC10763054 DOI: 10.1186/s13023-023-03000-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 12/15/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND The nonclinical as well as clinical development of orphan drugs is difficult, owing to unknown pathophysiology and the absence of animal models. Both, the U.S. Food and Drug Administration (FDA) Guidance and European Medicines Agency (EMA) Guidelines, for orphan drug development describe non-clinical studies, but lack specific information, such as animal species and study design. Against this background, this study aimed to elucidate efficient methods for evaluating nonclinical efficacy based on a review report of orphan drugs approved in Japan. RESULTS A total of 184 orphan drugs, including 84 anticancer and 100 non-anticancer drugs, approved in Japan from January 2010 to December 2019 were investigated. Some anticancer drugs progressed to clinical development without distinct efficacy data in nonclinical studies. Patient-derived cells have been used for some drugs due to a lack of established cell lines. Cells used for non-clinical studies were devised for drugs indicated for cancers resistant to prior therapies, tumours with specific amino acid mutations in the target molecules, and solid tumours with specific biomarkers. For some non-anticancer drugs, similar disease animal models and normal animals were used for evaluation, since animal models did not exist. Biomarkers have been used specifically for evaluation in normal animals and as endpoints in some clinical trials. CONCLUSIONS It was possible to evaluate drug efficacy by flexibly designing nonclinical studies according to disease characteristics for potentials orphan drugs. These approaches, which are not described in detail in the EMA Guideline or FDA Guidance, may thus lead to approval.
Collapse
Affiliation(s)
- Saki Yokoshiki
- Graduate School of Medicine, Hokkaido University, Kita 15, Nishi 7, Kita-ku, Sapporo, 060-8638, Japan
- Institute of Health Science Innovation for Medical Care (HELIOS), Hokkaido University Hospital, Kita 14, Nishi 5, Kita-ku, Sapporo, Hokkaido, 060-8648, Japan
| | - Teruyo Arato
- Graduate School of Medicine, Hokkaido University, Kita 15, Nishi 7, Kita-ku, Sapporo, 060-8638, Japan.
- Institute of Health Science Innovation for Medical Care (HELIOS), Hokkaido University Hospital, Kita 14, Nishi 5, Kita-ku, Sapporo, Hokkaido, 060-8648, Japan.
| |
Collapse
|
18
|
Wu K, Karapetyan E, Schloss J, Vadgama J, Wu Y. Advancements in small molecule drug design: A structural perspective. Drug Discov Today 2023; 28:103730. [PMID: 37536390 PMCID: PMC10543554 DOI: 10.1016/j.drudis.2023.103730] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 07/19/2023] [Accepted: 07/27/2023] [Indexed: 08/05/2023]
Abstract
In this review, we outline recent advancements in small molecule drug design from a structural perspective. We compare protein structure prediction methods and explore the role of the ligand binding pocket in structure-based drug design. We examine various structural features used to optimize drug candidates, including functional groups, stereochemistry, and molecular weight. Computational tools such as molecular docking and virtual screening are discussed for predicting and optimizing drug candidate structures. We present examples of drug candidates designed based on their molecular structure and discuss future directions in the field. By effectively integrating structural information with other valuable data sources, we can improve the drug discovery process, leading to the identification of novel therapeutics with improved efficacy, specificity, and safety profiles.
Collapse
Affiliation(s)
- Ke Wu
- Division of Cancer Research and Training, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, David Geffen UCLA School of Medicine and UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095, USA
| | - Eduard Karapetyan
- Division of Cancer Research and Training, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, David Geffen UCLA School of Medicine and UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095, USA
| | - John Schloss
- Division of Cancer Research and Training, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, David Geffen UCLA School of Medicine and UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095, USA; School of Pharmacy, American University of Health Sciences, Signal Hill, CA 90755, USA
| | - Jaydutt Vadgama
- Division of Cancer Research and Training, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, David Geffen UCLA School of Medicine and UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095, USA; School of Pharmacy, American University of Health Sciences, Signal Hill, CA 90755, USA.
| | - Yong Wu
- Division of Cancer Research and Training, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, David Geffen UCLA School of Medicine and UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095, USA.
| |
Collapse
|
19
|
Rosenberg N, Stolwijk NN, van den Berg S, Heus JJ, van der Wel V, van Gelder T, Bosch AM, de Visser SJ, Hollak CEM. Development of medicines for rare diseases and inborn errors of metabolism: Toward novel public-private partnerships. J Inherit Metab Dis 2023; 46:806-816. [PMID: 36938792 DOI: 10.1002/jimd.12605] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 03/12/2023] [Accepted: 03/16/2023] [Indexed: 03/21/2023]
Abstract
Medicine development for rare diseases, including inborn errors of metabolism (IEMs) is challenging. Many academic innovations fail to reach the patient, either by stranding in the translational stage or due to suboptimal patient access related to pricing or uncertain effectiveness. Expanding and solidifying the role of the academic in public-private partnerships (PPPs) may present an innovative solution to help overcome these complexities. This narrative review explores the literature on traditional and novel collaborative approaches to medicine development for rare diseases and analyzes examples of PPPs, with a specific focus on IEMs. Several academic institutions have introduced guidelines for socially responsible licensing of innovations for private development. The PPP model offers a more integrative approach toward academic involvement of medicine development. By sharing risks and rewards, failures in the translational stage can be mutually absorbed. If socially responsible terms are not included, however, high pricing can impede patient access. Therefore, we propose a framework for socially responsible PPPs aimed at medicine development for metabolic disorders. This socially responsible PPP framework could stimulate successful and accessible medicine development for IEMs as well as other rare diseases if the establishment of such collaborations includes terms securing joint data ownership and evidence generation, fast access, and socially responsible pricing.
Collapse
Affiliation(s)
- Noa Rosenberg
- Medicines for Society (Medicijn voor de Maatschappij), Platform at Amsterdam UMC - University of Amsterdam, Amsterdam, The Netherlands
- Department of Endocrinology and Metabolism, Amsterdam UMC, Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Research Institute, Expertise Center for Inborn Errors of Metabolism, MetabERN, University of Amsterdam, Amsterdam, The Netherlands
| | - Nina N Stolwijk
- Medicines for Society (Medicijn voor de Maatschappij), Platform at Amsterdam UMC - University of Amsterdam, Amsterdam, The Netherlands
- Department of Endocrinology and Metabolism, Amsterdam UMC, Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Research Institute, Expertise Center for Inborn Errors of Metabolism, MetabERN, University of Amsterdam, Amsterdam, The Netherlands
| | - Sibren van den Berg
- Medicines for Society (Medicijn voor de Maatschappij), Platform at Amsterdam UMC - University of Amsterdam, Amsterdam, The Netherlands
- Department of Endocrinology and Metabolism, Amsterdam UMC, Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Research Institute, Expertise Center for Inborn Errors of Metabolism, MetabERN, University of Amsterdam, Amsterdam, The Netherlands
| | - Joris J Heus
- Innovation eXchange Amsterdam (IXA) Office Amsterdam UMC, Amsterdam, The Netherlands
| | - Vincent van der Wel
- Medicines for Society (Medicijn voor de Maatschappij), Platform at Amsterdam UMC - University of Amsterdam, Amsterdam, The Netherlands
- Orfenix B.V., Leiden, The Netherlands
| | - Teun van Gelder
- Department of Clinical Pharmacy & Toxicology, LUMC, Leiden University Medical Center, Leiden, The Netherlands
| | - Annet M Bosch
- Department of Pediatrics, Division of Metabolic Diseases, Emma Children's Hospital, Amsterdam Gastroenterology Endocrinology Metabolism (AGEM), Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - Saco J de Visser
- Medicines for Society (Medicijn voor de Maatschappij), Platform at Amsterdam UMC - University of Amsterdam, Amsterdam, The Netherlands
- Centre for Future Affordable & Sustainable Therapy development (FAST), The Hague, The Netherlands
| | - Carla E M Hollak
- Medicines for Society (Medicijn voor de Maatschappij), Platform at Amsterdam UMC - University of Amsterdam, Amsterdam, The Netherlands
- Department of Endocrinology and Metabolism, Amsterdam UMC, Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Research Institute, Expertise Center for Inborn Errors of Metabolism, MetabERN, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
20
|
Saffari A, Brechmann B, Boeger C, Saber WA, Jumo H, Whye D, Wood D, Wahlster L, Alecu J, Ziegler M, Scheffold M, Winden K, Hubbs J, Buttermore E, Barrett L, Borner G, Davies A, Sahin M, Ebrahimi-Fakhari D. High-Content Small Molecule Screen Identifies a Novel Compound That Restores AP-4-Dependent Protein Trafficking in Neuronal Models of AP-4-Associated Hereditary Spastic Paraplegia. RESEARCH SQUARE 2023:rs.3.rs-3036166. [PMID: 37398196 PMCID: PMC10312991 DOI: 10.21203/rs.3.rs-3036166/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Unbiased phenotypic screens in patient-relevant disease models offer the potential to detect novel therapeutic targets for rare diseases. In this study, we developed a high-throughput screening assay to identify molecules that correct aberrant protein trafficking in adaptor protein complex 4 (AP-4) deficiency, a rare but prototypical form of childhood-onset hereditary spastic paraplegia, characterized by mislocalization of the autophagy protein ATG9A. Using high-content microscopy and an automated image analysis pipeline, we screened a diversity library of 28,864 small molecules and identified a lead compound, C-01, that restored ATG9A pathology in multiple disease models, including patient-derived fibroblasts and induced pluripotent stem cell-derived neurons. We used multiparametric orthogonal strategies and integrated transcriptomic and proteomic approaches to delineate putative molecular targets of C-01 and potential mechanisms of action. Our results define molecular regulators of intracellular ATG9A trafficking and characterize a lead compound for the treatment of AP-4 deficiency, providing important proof-of-concept data for future Investigational New Drug (IND)-enabling studies.
Collapse
Affiliation(s)
| | | | | | | | | | - Dosh Whye
- Boston Children's Hospital, Harvard Medical School
| | - Delaney Wood
- Boston Children's Hospital, Harvard Medical School
| | | | - Julian Alecu
- Boston Children's Hospital, Harvard Medical School
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Wiesinger AM, Bigger B, Giugliani R, Lampe C, Scarpa M, Moser T, Kampmann C, Zimmermann G, Lagler FB. An Innovative Tool for Evidence-Based, Personalized Treatment Trials in Mucopolysaccharidosis. Pharmaceutics 2023; 15:1565. [PMID: 37242808 PMCID: PMC10221776 DOI: 10.3390/pharmaceutics15051565] [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: 04/28/2023] [Revised: 05/16/2023] [Accepted: 05/19/2023] [Indexed: 05/28/2023] Open
Abstract
Mucopolysaccharidosis (MPS) is a group of rare metabolic diseases associated with reduced life expectancy and a substantial unmet medical need. Immunomodulatory drugs could be a relevant treatment approach for MPS patients, although they are not licensed for this population. Therefore, we aim to provide evidence justifying fast access to innovative individual treatment trials (ITTs) with immunomodulators and a high-quality evaluation of drug effects by implementing a risk-benefit model for MPS. The iterative methodology of our developed decision analysis framework (DAF) consists of the following steps: (i) a comprehensive literature analysis on promising treatment targets and immunomodulators for MPS; (ii) a quantitative risk-benefit assessment (RBA) of selected molecules; and (iii) allocation phenotypic profiles and a quantitative assessment. These steps allow for the personalized use of the model and are in accordance with expert and patient representatives. The following four promising immunomodulators were identified: adalimumab, abatacept, anakinra, and cladribine. An improvement in mobility is most likely with adalimumab, while anakinra might be the treatment of choice for patients with neurocognitive involvement. Nevertheless, a RBA should always be completed on an individual basis. Our evidence-based DAF model for ITTs directly addresses the substantial unmet medical need in MPS and characterizes a first approach toward precision medicine with immunomodulatory drugs.
Collapse
Affiliation(s)
- Anna-Maria Wiesinger
- Institute of Congenital Metabolic Diseases, Paracelsus Medical University, 5020 Salzburg, Austria;
- European Reference Network for Hereditary Metabolic Diseases, MetabERN, 33100 Udine, Italy; (B.B.); (C.L.); (M.S.)
| | - Brian Bigger
- European Reference Network for Hereditary Metabolic Diseases, MetabERN, 33100 Udine, Italy; (B.B.); (C.L.); (M.S.)
- Stem Cell and Neurotherapies, Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK
| | - Roberto Giugliani
- Department of Genetics, Medical Genetics Service and Biodiscovery Laboratory, Portal Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul (UFRGS), Casa dos Raros, Porto Alegre 90610-261, Brazil;
| | - Christina Lampe
- European Reference Network for Hereditary Metabolic Diseases, MetabERN, 33100 Udine, Italy; (B.B.); (C.L.); (M.S.)
- Department of Child Neurology, Epilepetology and Social Pediatrics, Center of Rare Diseases, University Hospital Giessen/Marburg, 35392 Giessen, Germany
| | - Maurizio Scarpa
- European Reference Network for Hereditary Metabolic Diseases, MetabERN, 33100 Udine, Italy; (B.B.); (C.L.); (M.S.)
- Regional Coordinating Center for Rare Diseases, University Hospital Udine, 33100 Udine, Italy
| | - Tobias Moser
- Department of Neurology, Christian Doppler University Hospital, Paracelsus Medical University, 5020 Salzburg, Austria;
| | - Christoph Kampmann
- Department of Pediatric Cardiology, University Hospital Mainz, 55131 Mainz, Germany;
| | - Georg Zimmermann
- Team Biostatistics and Big Medical Data, IDA Lab Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria;
- Research and Innovation Management, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Florian B. Lagler
- Institute of Congenital Metabolic Diseases, Paracelsus Medical University, 5020 Salzburg, Austria;
- European Reference Network for Hereditary Metabolic Diseases, MetabERN, 33100 Udine, Italy; (B.B.); (C.L.); (M.S.)
| |
Collapse
|
22
|
A Proof of Principle Proteomic Study Detects Dystrophin in Human Plasma: Implications in DMD Diagnosis and Clinical Monitoring. Int J Mol Sci 2023; 24:ijms24065215. [PMID: 36982290 PMCID: PMC10049465 DOI: 10.3390/ijms24065215] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/03/2023] [Accepted: 03/06/2023] [Indexed: 03/12/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a rare neuromuscular disease caused by pathogenic variations in the DMD gene. There is a need for robust DMD biomarkers for diagnostic screening and to aid therapy monitoring. Creatine kinase, to date, is the only routinely used blood biomarker for DMD, although it lacks specificity and does not correlate with disease severity. To fill this critical gap, we present here novel data about dystrophin protein fragments detected in human plasma by a suspension bead immunoassay using two validated anti-dystrophin-specific antibodies. Using both antibodies, a reduction of the dystrophin signal is detected in a small cohort of plasma samples from DMD patients when compared to healthy controls, female carriers, and other neuromuscular diseases. We also demonstrate the detection of dystrophin protein by an antibody-independent method using targeted liquid chromatography mass spectrometry. This last assay detects three different dystrophin peptides in all healthy individuals analysed and supports our finding that dystrophin protein is detectable in plasma. The results of our proof-of-concept study encourage further studies in larger sample cohorts to investigate the value of dystrophin protein as a low invasive blood biomarker for diagnostic screening and clinical monitoring of DMD.
Collapse
|
23
|
Chen A, Pan Y, Chen J. Clinical, genetic, and experimental research of hyperphenylalaninemia. Front Genet 2023; 13:1051153. [PMID: 36685931 PMCID: PMC9845280 DOI: 10.3389/fgene.2022.1051153] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 12/06/2022] [Indexed: 01/06/2023] Open
Abstract
Hyperphenylalaninemia (HPA) is the most common amino acid metabolism defect in humans. It is an autosomal-recessive disorder of the phenylalanine (Phe) metabolism, in which high Phe concentrations and low tyrosine (Tyr) concentrations in the blood cause phenylketonuria (PKU), brain dysfunction, light pigmentation and musty odor. Newborn screening data of HPA have revealed that the prevalence varies worldwide, with an average of 1:10,000. Most cases of HPA result from phenylalanine hydroxylase (PAH) deficiency, while a small number of HPA are caused by defects in the tetrahydrobiopterin (BH4) metabolism and DnaJ heat shock protein family (Hsp40) member C12 (DNAJC12) deficiency. Currently, the molecular pathophysiology of the neuropathology associated with HPA remains incompletely understood. Dietary restriction of Phe has been highly successful, although outcomes are still suboptimal and patients find it difficult to adhere to the treatment. Pharmacological treatments, such as BH4 and phenylalanine ammonia lyase, are available. Gene therapy for HPA is still in development.
Collapse
Affiliation(s)
- Anqi Chen
- Department of Forensic Medicine, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yukun Pan
- Barbell Therapeutics Co. Ltd., Shanghai, China,*Correspondence: Yukun Pan, ; Jinzhong Chen,
| | - Jinzhong Chen
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, China,*Correspondence: Yukun Pan, ; Jinzhong Chen,
| |
Collapse
|
24
|
Ambrosini C, Destefanis E, Kheir E, Broso F, Alessandrini F, Longhi S, Battisti N, Pesce I, Dassi E, Petris G, Cereseto A, Quattrone A. Translational enhancement by base editing of the Kozak sequence rescues haploinsufficiency. Nucleic Acids Res 2022; 50:10756-10771. [PMID: 36165847 PMCID: PMC9561285 DOI: 10.1093/nar/gkac799] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 09/01/2022] [Accepted: 09/22/2022] [Indexed: 11/28/2022] Open
Abstract
A variety of single-gene human diseases are caused by haploinsufficiency, a genetic condition by which mutational inactivation of one allele leads to reduced protein levels and functional impairment. Translational enhancement of the spare allele could exert a therapeutic effect. Here we developed BOOST, a novel gene-editing approach to rescue haploinsufficiency loci by the change of specific single nucleotides in the Kozak sequence, which controls translation by regulating start codon recognition. We evaluated for translational strength 230 Kozak sequences of annotated human haploinsufficient genes and 4621 derived variants, which can be installed by base editing, by a high-throughput reporter assay. Of these variants, 149 increased the translation of 47 Kozak sequences, demonstrating that a substantial proportion of haploinsufficient genes are controlled by suboptimal Kozak sequences. Validation of 18 variants for 8 genes produced an average enhancement in an expression window compatible with the rescue of the genetic imbalance. Base editing of the NCF1 gene, whose monoallelic loss causes chronic granulomatous disease, resulted in the desired increase of NCF1 (p47phox) protein levels in a relevant cell model. We propose BOOST as a fine-tuned approach to modulate translation, applicable to the correction of dozens of haploinsufficient monogenic disorders independently of the causing mutation.
Collapse
Affiliation(s)
- Chiara Ambrosini
- Laboratory of Translational Genomics, Department of Cellular, Computational and Integrative Biology - CIBIO, University of Trento, Trento 38123, Italy
| | - Eliana Destefanis
- Laboratory of Translational Genomics, Department of Cellular, Computational and Integrative Biology - CIBIO, University of Trento, Trento 38123, Italy
| | - Eyemen Kheir
- Laboratory of Molecular Virology, Department of Cellular, Computational and Integrative Biology - CIBIO, University of Trento, Trento 38123, Italy
| | - Francesca Broso
- Laboratory of Translational Genomics, Department of Cellular, Computational and Integrative Biology - CIBIO, University of Trento, Trento 38123, Italy
| | - Federica Alessandrini
- Laboratory of Translational Genomics, Department of Cellular, Computational and Integrative Biology - CIBIO, University of Trento, Trento 38123, Italy
| | - Sara Longhi
- Laboratory of Translational Genomics, Department of Cellular, Computational and Integrative Biology - CIBIO, University of Trento, Trento 38123, Italy
| | - Nicolò Battisti
- Laboratory of Translational Genomics, Department of Cellular, Computational and Integrative Biology - CIBIO, University of Trento, Trento 38123, Italy
| | - Isabella Pesce
- Cell Analysis and Separation Core Facility, Department of Cellular, Computational and Integrative Biology - CIBIO, University of Trento, Trento 38123, Italy
| | - Erik Dassi
- Laboratory of RNA Regulatory Networks, Department of Cellular, Computational and Integrative Biology - CIBIO, University of Trento, Trento 38123, Italy
| | - Gianluca Petris
- Medical Research Council Laboratory of Molecular Biology (MRC LMB), Cambridge CB2 0QH, UK
| | - Anna Cereseto
- Laboratory of Molecular Virology, Department of Cellular, Computational and Integrative Biology - CIBIO, University of Trento, Trento 38123, Italy
| | - Alessandro Quattrone
- Laboratory of Translational Genomics, Department of Cellular, Computational and Integrative Biology - CIBIO, University of Trento, Trento 38123, Italy
| |
Collapse
|
25
|
Making Sure That Orphan Incentives Tip the Right Way in Europe. Healthcare (Basel) 2022; 10:healthcare10091600. [PMID: 36141212 PMCID: PMC9498781 DOI: 10.3390/healthcare10091600] [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: 07/21/2022] [Revised: 08/16/2022] [Accepted: 08/19/2022] [Indexed: 12/03/2022] Open
Abstract
The delicate balance of funding research and development of treatments for rare disease is only imperfectly achieved in Europe, and even the current provisional equilibrium is under a new threat from well-intentioned policy changes now in prospect that could—in addition to the intrinsic complexities of research—reduce the incentives on which commercial activity in this area is dependent. The European Union review of its pharmaceutical legislation, for which proposals are scheduled to appear before the end of 2022, envisages adjusting the decade-old incentives to meet objectives that are more precisely targeted. However, researchers, physicians, patients and industry have expressed concerns that ill-considered modifications could have unintended consequences in disrupting the current balance and could reduce rather than increase the flow of innovative treatments for rare diseases.
Collapse
|
26
|
Monticelli M, Liguori L, Allocca M, Bosso A, Andreotti G, Lukas J, Monti MC, Morretta E, Cubellis MV, Hay Mele B. Drug Repositioning for Fabry Disease: Acetylsalicylic Acid Potentiates the Stabilization of Lysosomal Alpha-Galactosidase by Pharmacological Chaperones. Int J Mol Sci 2022; 23:ijms23095105. [PMID: 35563496 PMCID: PMC9105905 DOI: 10.3390/ijms23095105] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/01/2022] [Accepted: 05/02/2022] [Indexed: 12/10/2022] Open
Abstract
Fabry disease is caused by a deficiency of lysosomal alpha galactosidase and has a very large genotypic and phenotypic spectrum. Some patients who carry hypomorphic mutations can benefit from oral therapy with a pharmacological chaperone. The drug requires a very precise regimen because it is a reversible inhibitor of alpha-galactosidase. We looked for molecules that can potentiate this pharmacological chaperone, among drugs that have already been approved for other diseases. We tested candidate molecules in fibroblasts derived from a patient carrying a large deletion in the gene GLA, which were stably transfected with a plasmid expressing hypomorphic mutants. In our cell model, three drugs were able to potentiate the action of the pharmacological chaperone. We focused our attention on one of them, acetylsalicylic acid. We expect that acetylsalicylic acid can be used in synergy with the Fabry disease pharmacological chaperone and prolong its stabilizing effect on alpha-galactosidase.
Collapse
Affiliation(s)
- Maria Monticelli
- Department Biology, University of Napoli « Federico II », Complesso Universitario Monte Sant’Angelo, Via Cinthia, 80126 Napoli, Italy; (M.M.); (A.B.); (B.H.M.)
- Department Environmental, Biological and Pharmaceutical Sciences and Technologies (DiSTABiF), University of Campania “Luigi Vanvitelli”, Via Vivaldi 43, 81100 Caserta, Italy; (L.L.); (M.A.)
- Institute of Biomolecular Chemistry ICB, CNR, Via Campi Flegrei 34, 80078 Pozzuoli, Italy;
| | - Ludovica Liguori
- Department Environmental, Biological and Pharmaceutical Sciences and Technologies (DiSTABiF), University of Campania “Luigi Vanvitelli”, Via Vivaldi 43, 81100 Caserta, Italy; (L.L.); (M.A.)
| | - Mariateresa Allocca
- Department Environmental, Biological and Pharmaceutical Sciences and Technologies (DiSTABiF), University of Campania “Luigi Vanvitelli”, Via Vivaldi 43, 81100 Caserta, Italy; (L.L.); (M.A.)
- Institute of Biomolecular Chemistry ICB, CNR, Via Campi Flegrei 34, 80078 Pozzuoli, Italy;
| | - Andrea Bosso
- Department Biology, University of Napoli « Federico II », Complesso Universitario Monte Sant’Angelo, Via Cinthia, 80126 Napoli, Italy; (M.M.); (A.B.); (B.H.M.)
- Institute of Biochemistry and Cellular Biology, National Research Council, Via Pietro Castellino 111, 80131 Napoli, Italy
| | - Giuseppina Andreotti
- Institute of Biomolecular Chemistry ICB, CNR, Via Campi Flegrei 34, 80078 Pozzuoli, Italy;
| | - Jan Lukas
- Translational Neurodegeneration Section “Albrecht-Kossel”, Department of Neurology, University Medical Center Rostock, 18147 Rostock, Germany;
- Center for Transdisciplinary Neurosciences Rostock (CTNR), University Medical Center Rostock, 18147 Rostock, Germany
| | - Maria Chiara Monti
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy; (M.C.M.); (E.M.)
| | - Elva Morretta
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy; (M.C.M.); (E.M.)
| | - Maria Vittoria Cubellis
- Department Biology, University of Napoli « Federico II », Complesso Universitario Monte Sant’Angelo, Via Cinthia, 80126 Napoli, Italy; (M.M.); (A.B.); (B.H.M.)
- Institute of Biomolecular Chemistry ICB, CNR, Via Campi Flegrei 34, 80078 Pozzuoli, Italy;
- Correspondence: ; Tel.: +39-081-679152
| | - Bruno Hay Mele
- Department Biology, University of Napoli « Federico II », Complesso Universitario Monte Sant’Angelo, Via Cinthia, 80126 Napoli, Italy; (M.M.); (A.B.); (B.H.M.)
| |
Collapse
|
27
|
Genetically modified mice for research on human diseases: A triumph for Biotechnology or a work in progress? THE EUROBIOTECH JOURNAL 2022. [DOI: 10.2478/ebtj-2022-0008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2022] Open
Abstract
Abstract
Genetically modified mice are engineered as models for human diseases. These mouse models include inbred strains, mutants, gene knockouts, gene knockins, and ‘humanized’ mice. Each mouse model is engineered to mimic a specific disease based on a theory of the genetic basis of that disease. For example, to test the amyloid theory of Alzheimer’s disease, mice with amyloid precursor protein genes are engineered, and to test the tau theory, mice with tau genes are engineered. This paper discusses the importance of mouse models in basic research, drug discovery, and translational research, and examines the question of how to define the “best” mouse model of a disease. The critiques of animal models and the caveats in translating the results from animal models to the treatment of human disease are discussed. Since many diseases are heritable, multigenic, age-related and experience-dependent, resulting from multiple gene-gene and gene-environment interactions, it will be essential to develop mouse models that reflect these genetic, epigenetic and environmental factors from a developmental perspective. Such models would provide further insight into disease emergence, progression and the ability to model two-hit and multi-hit theories of disease. The summary examines the biotechnology for creating genetically modified mice which reflect these factors and how they might be used to discover new treatments for complex human diseases such as cancers, neurodevelopmental and neurodegenerative diseases.
Collapse
|
28
|
Priyarega S, Natarajan R. An overview of biosimilars for cancer, diabetes mellitus, rheumatoid arthritis and other immune-mediated diseases approved between 2016 and 2021. RESULTS IN CHEMISTRY 2022. [DOI: 10.1016/j.rechem.2022.100356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
29
|
Alves VM, Korn D, Pervitsky V, Thieme A, Capuzzi SJ, Baker N, Chirkova R, Ekins S, Muratov EN, Hickey A, Tropsha A. Knowledge-based approaches to drug discovery for rare diseases. Drug Discov Today 2022; 27:490-502. [PMID: 34718207 PMCID: PMC9124594 DOI: 10.1016/j.drudis.2021.10.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 09/13/2021] [Accepted: 10/21/2021] [Indexed: 02/03/2023]
Abstract
The conventional drug discovery pipeline has proven to be unsustainable for rare diseases. Herein, we discuss recent advances in biomedical knowledge mining applied to discovering therapeutics for rare diseases. We summarize current chemogenomics data of relevance to rare diseases and provide a perspective on the effectiveness of machine learning (ML) and biomedical knowledge graph mining in rare disease drug discovery. We illustrate the power of these methodologies using a chordoma case study. We expect that a broader application of knowledge graph mining and artificial intelligence (AI) approaches will expedite the discovery of viable drug candidates against both rare and common diseases.
Collapse
Affiliation(s)
- Vinicius M Alves
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA; UNC Catalyst for Rare Diseases, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Daniel Korn
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Vera Pervitsky
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Andrew Thieme
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Stephen J Capuzzi
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Nancy Baker
- ParlezChem, 123 W Union Street, Hillsborough, NC 27278, USA
| | - Rada Chirkova
- Department of Computer Science, North Carolina State University, Raleigh, NC 27695-8206, USA
| | - Sean Ekins
- Collaborations Pharmaceuticals Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC 27606, USA
| | - Eugene N Muratov
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Pharmaceutical Sciences, Federal University of Paraiba, Joao Pessoa, PB, Brazil
| | - Anthony Hickey
- UNC Catalyst for Rare Diseases, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA.
| | - Alexander Tropsha
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|
30
|
Personalized medicine for rare neurogenetic disorders: can we make it happen? Cold Spring Harb Mol Case Stud 2022; 8:mcs.a006200. [PMID: 35332073 PMCID: PMC8958924 DOI: 10.1101/mcs.a006200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Rare neurogenetic disorders are collectively common, affecting 3% of the population, and often manifest with complex multiorgan comorbidity. With advances in genetic, -omics, and computational analysis, more children can be diagnosed and at an earlier age. Innovations in translational research facilitate the identification of treatment targets and development of disease-modifying drugs such as gene therapy, nutraceuticals, and drug repurposing. This increasingly allows targeted therapy to prevent the often devastating manifestations of rare neurogenetic disorders. In this perspective, successes in diagnosis, prevention, and treatment are discussed with a focus on inherited disorders of metabolism. Barriers for the identification, development, and implementation of rare disease-specific therapies are discussed. New methodologies, care networks, and collaborative frameworks are proposed to optimize the potential of personalized genomic medicine to decrease morbidity and improve lives of these vulnerable patients.
Collapse
|
31
|
Braga LAM, Conte Filho CG, Mota FB. Future of genetic therapies for rare genetic diseases: what to expect for the next 15 years? THERAPEUTIC ADVANCES IN RARE DISEASE 2022; 3:26330040221100840. [PMID: 37180410 PMCID: PMC10032453 DOI: 10.1177/26330040221100840] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 04/22/2022] [Indexed: 05/16/2023]
Abstract
Introduction Rare genetic diseases affect millions of people worldwide. Most of them are caused by defective genes that impair quality of life and can lead to premature death. As genetic therapies aim to fix or replace defective genes, they are considered the most promising treatment for rare genetic diseases. Yet, as these therapies are still under development, it is still unclear whether they will be successful in treating these diseases. This study aims to address this gap by assessing researchers' opinions on the future of genetic therapies for the treatment of rare genetic diseases. Methods We conducted a global cross-sectional web-based survey of researchers who recently authored peer-reviewed articles related to rare genetic diseases. Results We assessed the opinions of 1430 researchers with high and good knowledge about genetic therapies for the treatment of rare genetic diseases. Overall, the respondents believed that genetic therapies would be the standard of care for rare genetic diseases before 2036, leading to cures after this period. CRISPR-Cas9 was considered the most likely approach to fixing or replacing defective genes in the next 15 years. The respondents with good knowledge believed that genetic therapies would only have long-lasting effects after 2036, while those with high knowledge were divided on this issue. The respondents with good knowledge on the subject believed that non-viral vectors are more likely to be successful in fixing or replacing defective genes in the next 15 years, while most of the respondents with high knowledge believed viral vectors would be more successful. Conclusion Overall, the researchers who participated in this study expect that in the future genetic therapies will greatly benefit the treatment of patients with rare genetic diseases.
Collapse
Affiliation(s)
| | | | - Fabio Batista Mota
- Laboratory of Cellular Communication, Oswaldo
Cruz Institute, Oswaldo Cruz Foundation, Av. Brasil, 4.365, Pavilhão 108,
Manguinhos, Rio de Janeiro RJ 21040-360, Brazil
| |
Collapse
|
32
|
Sanchez P, Farkhondeh A, Pavlinov I, Baumgaertel K, Rodems S, Zheng W. Therapeutics Development for Alagille Syndrome. Front Pharmacol 2021; 12:704586. [PMID: 34497511 PMCID: PMC8419306 DOI: 10.3389/fphar.2021.704586] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 08/09/2021] [Indexed: 12/25/2022] Open
Abstract
Advancements in treatment for the rare genetic disorder known as Alagille Syndrome (ALGS) have been regrettably slow. The large variety of mutations to the JAG1 and NOTCH2 genes which lead to ALGS pose a unique challenge for developing targeted treatments. Due to the central role of the Notch signaling pathway in several cancers, traditional treatment modalities which compensate for the loss in activity caused by mutation are rightly excluded. Unfortunately, current treatment plans for ALGS focus on relieving symptoms of the disorder and do not address the underlying causes of disease. Here we review several of the current and potential key technologies and strategies which may yield a significant leap in developing targeted therapies for this disorder.
Collapse
Affiliation(s)
- Phillip Sanchez
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, United States
| | - Atena Farkhondeh
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, United States
| | - Ivan Pavlinov
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, United States
| | | | | | - Wei Zheng
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
33
|
Chong LC, Gandhi G, Lee JM, Yeo WWY, Choi SB. Drug Discovery of Spinal Muscular Atrophy (SMA) from the Computational Perspective: A Comprehensive Review. Int J Mol Sci 2021; 22:8962. [PMID: 34445667 PMCID: PMC8396480 DOI: 10.3390/ijms22168962] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 01/27/2021] [Indexed: 01/02/2023] Open
Abstract
Spinal muscular atrophy (SMA), one of the leading inherited causes of child mortality, is a rare neuromuscular disease arising from loss-of-function mutations of the survival motor neuron 1 (SMN1) gene, which encodes the SMN protein. When lacking the SMN protein in neurons, patients suffer from muscle weakness and atrophy, and in the severe cases, respiratory failure and death. Several therapeutic approaches show promise with human testing and three medications have been approved by the U.S. Food and Drug Administration (FDA) to date. Despite the shown promise of these approved therapies, there are some crucial limitations, one of the most important being the cost. The FDA-approved drugs are high-priced and are shortlisted among the most expensive treatments in the world. The price is still far beyond affordable and may serve as a burden for patients. The blooming of the biomedical data and advancement of computational approaches have opened new possibilities for SMA therapeutic development. This article highlights the present status of computationally aided approaches, including in silico drug repurposing, network driven drug discovery as well as artificial intelligence (AI)-assisted drug discovery, and discusses the future prospects.
Collapse
Affiliation(s)
- Li Chuin Chong
- Centre for Bioinformatics, School of Data Sciences, Perdana University, Suite 9.2, 9th Floor, Wisma Chase Perdana, Changkat Semantan, Kuala Lumpur 50490, Malaysia; (L.C.C.); (J.M.L.)
| | - Gayatri Gandhi
- Perdana University Graduate School of Medicine, Perdana University, Suite 9.2, 9th Floor, Wisma Chase Perdana, Changkat Semantan, Kuala Lumpur 50490, Malaysia; (G.G.); (W.W.Y.Y.)
| | - Jian Ming Lee
- Centre for Bioinformatics, School of Data Sciences, Perdana University, Suite 9.2, 9th Floor, Wisma Chase Perdana, Changkat Semantan, Kuala Lumpur 50490, Malaysia; (L.C.C.); (J.M.L.)
| | - Wendy Wai Yeng Yeo
- Perdana University Graduate School of Medicine, Perdana University, Suite 9.2, 9th Floor, Wisma Chase Perdana, Changkat Semantan, Kuala Lumpur 50490, Malaysia; (G.G.); (W.W.Y.Y.)
| | - Sy-Bing Choi
- Centre for Bioinformatics, School of Data Sciences, Perdana University, Suite 9.2, 9th Floor, Wisma Chase Perdana, Changkat Semantan, Kuala Lumpur 50490, Malaysia; (L.C.C.); (J.M.L.)
| |
Collapse
|
34
|
Sieberg CB, Lebel A, Silliman E, Holmes S, Borsook D, Elman I. Left to themselves: Time to target chronic pain in childhood rare diseases. Neurosci Biobehav Rev 2021; 126:276-288. [PMID: 33774086 PMCID: PMC8738995 DOI: 10.1016/j.neubiorev.2021.03.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/02/2021] [Accepted: 03/04/2021] [Indexed: 11/24/2022]
Abstract
BACKGROUND Chronic pain is prevalent among patients with rare diseases (RDs). However, little is understood about how biopsychosocial mechanisms may be integrated in the unique set of clinical features and therapeutic challenges inherent in their pain conditions. METHODS This review presents examples of major categories of RDs with particular pain conditions. In addition, we provide translational evidence on clinical and scientific rationale for psychosocially- and neurodevelopmentally-informed treatment of pain in RD patients. RESULTS Neurobiological and functional overlap between various RD syndromes and pain states suggests amalgamation and mutual modulation of the respective conditions. Emotional sequelae could be construed as an emotional homologue of physical pain mediated via overlapping brain circuitry. Given their clearly defined genetic and molecular etiologies, RDs may serve as heuristic models for unraveling pathophysiological processes inherent in chronic pain. CONCLUSIONS Systematic evaluation of chronic pain in patients with RD contributes to sophisticated insight into both pain and their psychosocial correlates, which could transform treatment.
Collapse
Affiliation(s)
- Christine B Sieberg
- Biobehavioral Pediatric Pain Lab, Department of Psychiatry & Behavioral Sciences, Boston Children's Hospital, Boston, MA, 02115, USA; Center for Pain and the Brain (P.A.I.N Group), Department of Anesthesiology, Critical Care & Pain Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, 02115, USA
| | - Alyssa Lebel
- Center for Pain and the Brain (P.A.I.N Group), Department of Anesthesiology, Critical Care & Pain Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Department of Anesthesiology, Harvard Medical School, Boston, MA, 02115, USA
| | - Erin Silliman
- Biobehavioral Pediatric Pain Lab, Department of Psychiatry & Behavioral Sciences, Boston Children's Hospital, Boston, MA, 02115, USA; Division of Graduate Medical Sciences, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Scott Holmes
- Center for Pain and the Brain (P.A.I.N Group), Department of Anesthesiology, Critical Care & Pain Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Department of Anesthesiology, Harvard Medical School, Boston, MA, 02115, USA
| | - David Borsook
- Center for Pain and the Brain (P.A.I.N Group), Department of Anesthesiology, Critical Care & Pain Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Department of Anesthesiology, Harvard Medical School, Boston, MA, 02115, USA; Department of Psychiatry, Massachusetts General Hospital, Boston, MA, 02114, USA.
| | - Igor Elman
- Center for Pain and the Brain (P.A.I.N Group), Department of Anesthesiology, Critical Care & Pain Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Cambridge Health Alliance, Harvard Medical School, Cambridge, MA, 02139, USA
| |
Collapse
|
35
|
Bang JS, Lee JH. The national drug formulary listing process for orphan drugs in South Korea: narrative review focused on pricing and reimbursement pathways. Expert Opin Orphan Drugs 2021. [DOI: 10.1080/21678707.2021.1913121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Joon Seok Bang
- College of Pharmacy, Sookmyung Women’s University, Seoul, Republic of Korea
| | - Jong Hyuk Lee
- Department of Pharmaceutical Engineering, Hoseo University, Asan, Republic of Korea
| |
Collapse
|
36
|
Jain P, Jain SK, Jain M. Harnessing Drug Repurposing for Exploration of New Diseases: An Insight to Strategies and Case Studies. Curr Mol Med 2021; 21:111-132. [PMID: 32560606 DOI: 10.2174/1566524020666200619125404] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 05/12/2020] [Accepted: 05/13/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Traditional drug discovery is time consuming, costly, and risky process. Owing to the large investment, excessive attrition, and declined output, drug repurposing has become a blooming approach for the identification and development of new therapeutics. The method has gained momentum in the past few years and has resulted in many excellent discoveries. Industries are resurrecting the failed and shelved drugs to save time and cost. The process accounts for approximately 30% of the new US Food and Drug Administration approved drugs and vaccines in recent years. METHODS A systematic literature search using appropriate keywords were made to identify articles discussing the different strategies being adopted for repurposing and various drugs that have been/are being repurposed. RESULTS This review aims to describe the comprehensive data about the various strategies (Blinded search, computational approaches, and experimental approaches) used for the repurposing along with success case studies (treatment for orphan diseases, neglected tropical disease, neurodegenerative diseases, and drugs for pediatric population). It also inculcates an elaborated list of more than 100 drugs that have been repositioned, approaches adopted, and their present clinical status. We have also attempted to incorporate the different databases used for computational repurposing. CONCLUSION The data presented is proof that drug repurposing is a prolific approach circumventing the issues poised by conventional drug discovery approaches. It is a highly promising approach and when combined with sophisticated computational tools, it also carries high precision. The review would help researches in prioritizing the drugrepositioning method much needed to flourish the drug discovery research.
Collapse
Affiliation(s)
- Priti Jain
- Department of Pharmaceutical Chemistry and Computational Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Dhule (425405) Maharashtra, India
| | - Shreyans K Jain
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, Uttar Pradesh, India
| | - Munendra Jain
- SVKM's Department of Sciences, Narsee Monjee Institute of Management Studies, Indore, Madhya Pradesh, India
| |
Collapse
|
37
|
Pricing and Reimbursement Pathways of New Orphan Drugs in South Korea: A Longitudinal Comparison. Healthcare (Basel) 2021; 9:healthcare9030296. [PMID: 33800373 PMCID: PMC8000795 DOI: 10.3390/healthcare9030296] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/05/2021] [Accepted: 03/05/2021] [Indexed: 11/17/2022] Open
Abstract
This study aimed to analyze four current pathways affecting the listing and post-listing prices of new orphan drugs (ODs) in South Korea. These mechanisms were: (1) essential OD, (2) pharmacoeconomic evaluation (PE) waiver OD, (3) weighted average price OD, and (4) PE OD. We analyzed the ratio of the listing price of 48 new ODs to the average adjusted price (AAP) of seven advanced countries and examined the change in the post-listing price. Descriptive statistics were used to analyze the listing and post-listing price changes. The mean and median ratios of the listing price of total new OD to AAP were calculated to be 69.4% and 65.4%, respectively. Essential OD showed the highest mean (93.8%) and median (80.8%) ratios. The mean cumulative price discount rate of the new OD was 7.2% in the third year and 5.7% in the fifth year. The rarity of diseases impacts the listing price of OD, but the political effects of the benefits of OD on the post-listing price of these drugs could not be verified. Further research should be conducted to develop measures that facilitate the practical sharing of budget risks and increase patient access to new ODs.
Collapse
|
38
|
Kumar D, Talluri TR, Selokar NL, Hyder I, Kues WA. Perspectives of pluripotent stem cells in livestock. World J Stem Cells 2021; 13:1-29. [PMID: 33584977 PMCID: PMC7859985 DOI: 10.4252/wjsc.v13.i1.1] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 09/28/2020] [Accepted: 11/09/2020] [Indexed: 02/06/2023] Open
Abstract
The recent progress in derivation of pluripotent stem cells (PSCs) from farm animals opens new approaches not only for reproduction, genetic engineering, treatment and conservation of these species, but also for screening novel drugs for their efficacy and toxicity, and modelling of human diseases. Initial attempts to derive PSCs from the inner cell mass of blastocyst stages in farm animals were largely unsuccessful as either the cells survived for only a few passages, or lost their cellular potency; indicating that the protocols which allowed the derivation of murine or human embryonic stem (ES) cells were not sufficient to support the maintenance of ES cells from farm animals. This scenario changed by the innovation of induced pluripotency and by the development of the 3 inhibitor culture conditions to support naïve pluripotency in ES cells from livestock species. However, the long-term culture of livestock PSCs while maintaining the full pluripotency is still challenging, and requires further refinements. Here, we review the current achievements in the derivation of PSCs from farm animals, and discuss the potential application areas.
Collapse
Affiliation(s)
- Dharmendra Kumar
- Animal Physiology and Reproduction Division, ICAR-Central Institute for Research on Buffaloes, Hisar 125001, India.
| | - Thirumala R Talluri
- Equine Production Campus, ICAR-National Research Centre on Equines, Bikaner 334001, India
| | - Naresh L Selokar
- Animal Physiology and Reproduction Division, ICAR-Central Institute for Research on Buffaloes, Hisar 125001, India
| | - Iqbal Hyder
- Department of Physiology, NTR College of Veterinary Science, Gannavaram 521102, India
| | - Wilfried A Kues
- Department of Biotechnology, Friedrich-Loeffler-Institute, Federal Institute of Animal Health, Neustadt 31535, Germany
| |
Collapse
|
39
|
Uçkan-Çetinkaya D, Haider KH. Induced Pluripotent Stem Cells in Pediatric Research and Clinical Translation. Stem Cells 2021. [DOI: 10.1007/978-3-030-77052-5_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
40
|
Stockler‐Ipsiroglu S, Potter BK, Yuskiv N, Tingley K, Patterson M, van Karnebeek C. Developments in evidence creation for treatments of inborn errors of metabolism. J Inherit Metab Dis 2021; 44:88-98. [PMID: 32944978 PMCID: PMC7891579 DOI: 10.1002/jimd.12315] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 09/13/2020] [Accepted: 09/14/2020] [Indexed: 12/13/2022]
Abstract
Inborn errors of metabolism (IEM) represent the first group of genetic disorders, amenable to causal therapies. In addition to traditional medical diet and cofactor treatments, new treatment strategies such as enzyme replacement and small molecule therapies, solid organ transplantation, and cell-and gene-based therapies have become available. Inherent to the rare nature of the single conditions, generating high-quality evidence for these treatments in clinical trials and under real-world conditions has been challenging. Guidelines developed with standardized methodologies have contributed to improve the practice of care and long-term clinical outcomes. Adaptive trial designs allow for changes in sample size, group allocation and trial duration as the trial proceeds. n-of-1 studies may be used in small sample sized when participants are clinically heterogeneous. Multicenter observational and registry-based clinical trials are promoted via international research networks. Core outcome and standard data element sets will enhance comparative analysis of clinical trials and observational studies. Patient-centered outcome-research as well as patient-led research initiatives will further accelerate the development of therapies for IEM.
Collapse
Affiliation(s)
- Sylvia Stockler‐Ipsiroglu
- Division of Biochemical Genetics, Department of Pediatrics, and BC Children's Hospital Research InstituteUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Beth K. Potter
- School of Epidemiology and Public HealthUniversity of OttawaOttawaOntarioCanada
| | - Nataliya Yuskiv
- Division of Biochemical Genetics, Department of Pediatrics, and BC Children's Hospital Research InstituteUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Kylie Tingley
- School of Epidemiology and Public HealthUniversity of OttawaOttawaOntarioCanada
| | - Marc Patterson
- Division of Child and Adolescent Neurology, Departments of Neurology Pediatrics and Medical GeneticsMayo Clinic Children's CenterRochesterMinnesotaUSA
| | - Clara van Karnebeek
- Departments of Pediatrics and Clinical GeneticsAmsterdam University Medical CentresAmsterdamThe Netherlands
- Department of PediatricsRadboud University Medical CentreNijmegenThe Netherlands
- Department of PediatricsBC Children's Hospital Research Institute, Centre for Molecular Medicine and TherapeuticsVancouverBritish ColumbiaCanada
| |
Collapse
|
41
|
Giorgio E, Pesce E, Pozzi E, Sondo E, Ferrero M, Morerio C, Borrelli G, Della Sala E, Lorenzati M, Cortelli P, Buffo A, Pedemonte N, Brusco A. A high-content drug screening strategy to identify protein level modulators for genetic diseases: A proof-of-principle in autosomal dominant leukodystrophy. Hum Mutat 2020; 42:102-116. [PMID: 33252173 DOI: 10.1002/humu.24147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 10/09/2020] [Accepted: 11/24/2020] [Indexed: 11/07/2022]
Abstract
In genetic diseases, the most prevalent mechanism of pathogenicity is an altered expression of dosage-sensitive genes. Drugs that restore physiological levels of these genes should be effective in treating the associated conditions. We developed a screening strategy, based on a bicistronic dual-reporter vector, for identifying compounds that modulate protein levels, and used it in a pharmacological screening approach. To provide a proof-of-principle, we chose autosomal dominant leukodystrophy (ADLD), an ultra-rare adult-onset neurodegenerative disorder caused by lamin B1 (LMNB1) overexpression. We used a stable Chinese hamster ovary (CHO) cell line that simultaneously expresses an AcGFP reporter fused to LMNB1 and a Ds-Red normalizer. Using high-content imaging analysis, we screened a library of 717 biologically active compounds and approved drugs, and identified alvespimycin, an HSP90 inhibitor, as a positive hit. We confirmed that alvespimycin can reduce LMNB1 levels by 30%-80% in five different cell lines (fibroblasts, NIH3T3, CHO, COS-7, and rat primary glial cells). In ADLD fibroblasts, alvespimycin reduced cytoplasmic LMNB1 by about 50%. We propose this approach for effectively identifying potential drugs for treating genetic diseases associated with deletions/duplications and paving the way toward Phase II clinical trials.
Collapse
Affiliation(s)
- Elisa Giorgio
- Department of Medical Sciences, Medical Genetics Unit, University of Torino, Turin, Italy.,Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Emanuela Pesce
- UOC Genetica Medica, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Elisa Pozzi
- Department of Medical Sciences, Medical Genetics Unit, University of Torino, Turin, Italy
| | - Elvira Sondo
- UOC Genetica Medica, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Marta Ferrero
- Department of Medical Sciences, Medical Genetics Unit, University of Torino, Turin, Italy
| | - Cristina Morerio
- UOC Laboratorio di Genetica Umana, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Giusy Borrelli
- Department of Medical Sciences, Medical Genetics Unit, University of Torino, Turin, Italy
| | - Edoardo Della Sala
- Department of Medical Sciences, Medical Genetics Unit, University of Torino, Turin, Italy
| | - Martina Lorenzati
- Department of Neuroscience Rita Levi Montalcini and Neuroscience Institute Cavalieri Ottolenghi, University of Torino, Orbassano, Torino, Italy
| | - Pietro Cortelli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.,IRCCS Istituto delle Scienze Neurologiche di Bologna, Bellaria Hospital, Bologna, Italy
| | - Annalisa Buffo
- Department of Neuroscience Rita Levi Montalcini and Neuroscience Institute Cavalieri Ottolenghi, University of Torino, Orbassano, Torino, Italy
| | | | - Alfredo Brusco
- Department of Medical Sciences, Medical Genetics Unit, University of Torino, Turin, Italy.,Medical Genetics Unit, Città della Salute e della Scienza University Hospital, Turin, Italy
| |
Collapse
|
42
|
Zhang Y, Xie X, Hu J, Afreen KS, Zhang CL, Zhuge Q, Yang J. Prospects of Directly Reprogrammed Adult Human Neurons for Neurodegenerative Disease Modeling and Drug Discovery: iN vs. iPSCs Models. Front Neurosci 2020; 14:546484. [PMID: 33328842 PMCID: PMC7710799 DOI: 10.3389/fnins.2020.546484] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 10/12/2020] [Indexed: 12/11/2022] Open
Abstract
A reliable disease model is critical to the study of specific disease mechanisms as well as for the discovery and development of new drugs. Despite providing crucial insights into the mechanisms of neurodegenerative diseases, translation of this information to develop therapeutics in clinical trials have been unsuccessful. Reprogramming technology to convert adult somatic cells to induced Pluripotent Stem Cells (iPSCs) or directly reprogramming adult somatic cells to induced Neurons (iN), has allowed for the creation of better models to understand the molecular mechanisms and design of new drugs. In recent times, iPSC technology has been commonly used for modeling neurodegenerative diseases and drug discovery. However, several technological challenges have limited the application of iN. As evidence suggests, iN for the modeling of neurodegenerative disorders is advantageous compared to those derived from iPSCs. In this review, we will compare iPSCs and iN models for neurodegenerative diseases and their potential applications in the future.
Collapse
Affiliation(s)
- Ying Zhang
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xinyang Xie
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,International Department of The Affiliated High School of South China Normal University (HFI), Guangzhou, China
| | - Jiangnan Hu
- Department of Pharmaceutical Sciences, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Kazi Sabrina Afreen
- Department of Microbiology & Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| | - Chun-Li Zhang
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX, United States
| | - Qichuan Zhuge
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jianjing Yang
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
43
|
Schmidt D, Thompson C. Case studies in rare disease small molecule discovery and development. Bioorg Med Chem Lett 2020; 30:127462. [PMID: 32791196 DOI: 10.1016/j.bmcl.2020.127462] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 07/21/2020] [Accepted: 08/01/2020] [Indexed: 12/26/2022]
Abstract
This review covers case studies in rare disease small molecule drug discovery with an emphasis on the use of new technologies and innovative target approaches. Case studies include examples of covalent modification, inducement of alternative splicing, stop codon readthrough, allosteric activation, and a repurposing example. The review highlights effective use of rare disease animal models, inducible pluripotent stem cells, and biomarkers.
Collapse
Affiliation(s)
- Darby Schmidt
- Inzen Therapeutics, 790 Memorial Drive, Suite 201, Cambridge, MA 02139, United States.
| | | |
Collapse
|
44
|
Qiu T, Wang Y, Dabbous M, Hanna E, Han R, Liang S, Toumi M. Current state of developing advanced therapies for rare diseases in the European Union. Expert Opin Orphan Drugs 2020. [DOI: 10.1080/21678707.2020.1835640] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Tingting Qiu
- Department of Public Health, Aix-Marseille University, Marseille, France
| | - Yitong Wang
- Department of Public Health, Aix-Marseille University, Marseille, France
| | - Monique Dabbous
- Department of Public Health, Aix-Marseille University, Marseille, France
| | - Eve Hanna
- Department of Price, Reimbursement and Market Access, Creativ-ceutical, Paris, France
| | - Ru Han
- Department of Public Health, Aix-Marseille University, Marseille, France
| | - Shuyao Liang
- Department of Public Health, Aix-Marseille University, Marseille, France
| | - Mondher Toumi
- Department of Public Health, Aix-Marseille University, Marseille, France
| |
Collapse
|
45
|
Aballéa S, Thokagevistk K, Velikanova R, Simoens S, Annemans L, Antonanzas F, Auquier P, François C, Fricke FU, Malone D, Millier A, Persson U, Petrou S, Dabbous O, Postma M, Toumi M. Health economic evaluation of gene replacement therapies: methodological issues and recommendations. JOURNAL OF MARKET ACCESS & HEALTH POLICY 2020; 8:1822666. [PMID: 33144927 PMCID: PMC7580851 DOI: 10.1080/20016689.2020.1822666] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 08/31/2020] [Accepted: 09/04/2020] [Indexed: 05/08/2023]
Abstract
Objective: To provide recommendations for addressing previously identified key challenges in health economic evaluations of Gene Replacement Therapies (GRTs), including: 1) the assessment of clinical effectiveness; 2) the valuation of health outcomes; 3) the time horizon and extrapolation of effects beyond trial duration; 4) the estimation of costs; 5) the selection of appropriate discount rates; 6) the incorporation of broader elements of value; and 7) affordability. Methods: A literature review on economic evaluations of GRT was performed. Interviews were conducted with 8 European and US health economic experts with experience in evaluations of GRT. Targeted literature reviews were conducted to investigate further potential solutions to specific challenges. Recommendations: Experts agreed on factors to be considered to ensure the acceptability of historical cohorts by HTA bodies. Existing prospective registries or, if not available, retrospective registries, may be used to analyse different disease trajectories and inform extrapolations. The importance of expert opinion due to limited data was acknowledged. Expert opinion should be obtained using structured elicitation techniques. Broader elements of value, beyond health gains directly related to treatment, can be considered through the application of a factor to inflate the quality-adjusted life years (QALYs) or a higher cost-effectiveness threshold. Additionally, the use of cost-benefit analysis and saved young life equivalents (SAVE) were proposed as alternatives to QALYs for the valuations of outcomes of GRT as they can incorporate broader elements of value and avoid problems of eliciting utilities for paediatric diseases. Conclusions: While some of the limitations of economic evaluations of GRT are inherent to limited clinical data and lack of experience with these treatments, others may be addressed by methodological research to be conducted by health economists.
Collapse
Affiliation(s)
| | | | - Rimma Velikanova
- Groningen Research Institute of Pharmacy, PharmacoTherapy, Epidemiology & Economics, University of Groningen, Groningen, Netherlands
| | - Steven Simoens
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Lieven Annemans
- Department of Public Health, Ghent University, Ghent, Belgium
| | | | - Pascal Auquier
- CEReSS - Health Service Research and Quality of Life Center, Aix-Marseille University, Marseille, France
| | - Clément François
- Creativ-Ceutical, HEOR, Paris, France
- Public Health Department - Research Unit, Aix-Marseille University, Marseille, France
| | | | - Daniel Malone
- Pharmacotherapy Faculty, College of Pharmacy, University of Utah, Salt Lake City, USA
| | | | - Ulf Persson
- The Swedish Institute for Health Economics (IHE), Lund, Sweden
| | - Stavros Petrou
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Omar Dabbous
- Global Health Economics and Outcomes Research and Real World EvidenceAveXis Inc, Novartis Gene Therapies, Bannockburn, IL, USA
| | - Maarten Postma
- Groningen Research Institute of Pharmacy, PharmacoTherapy, Epidemiology & Economics, University of Groningen, Groningen, Netherlands
| | - Mondher Toumi
- Creativ-Ceutical, HEOR, Paris, France
- Public Health Department - Research Unit, Aix-Marseille University, Marseille, France
| |
Collapse
|
46
|
Boada CM, French JA, Dumanis SB. Proceedings of the 15th Antiepileptic Drug and Device Trials Meeting: State of the Science. Epilepsy Behav 2020; 111:107189. [PMID: 32563052 DOI: 10.1016/j.yebeh.2020.107189] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 05/23/2020] [Indexed: 12/11/2022]
Abstract
On May 22-24, 2019, the 15th Antiepileptic Drug and Device (AEDD) Trials Conference was held, which focused on current issues related to AEDD development from preclinical models to clinical prognostication. The conference featured regulatory agencies, academic laboratories, and healthcare companies involved in emerging epilepsy therapies and research. The program included discussions around funding and support for investigations in epilepsy and neurologic research, clinical trial design and integrated outcome measures for people with epilepsy, and drug development and upcoming disease-modifying therapies. Finally, the conference included updates from the preclinical, clinical, and device pipeline. Summaries of the talks are provided in this paper, with the various pipeline therapeutics in the listed tables to be outlined in a subsequent publication.
Collapse
Affiliation(s)
- Christina M Boada
- Department of Neurology, New York University Langone Medical Center, New York, NY, USA
| | - Jacqueline A French
- Department of Neurology, New York University Langone Medical Center, New York, NY, USA
| | | |
Collapse
|
47
|
Abstract
PURPOSE OF THE REVIEW Significant numbers of patients worldwide are affected by various rare diseases, but the effective treatment options to these individuals are limited. Rare diseases remain underfunded compared to more common diseases, leading to significant delays in research progress and ultimately, to finding an effective cure. Here, we review the use of genome-editing tools to understand the pathogenesis of rare diseases and develop additional therapeutic approaches with a high degree of precision. RECENT FINDINGS Several genome-editing approaches, including CRISPR/Cas9, TALEN and ZFN, have been used to generate animal models of rare diseases, understand the disease pathogenesis, correct pathogenic mutations in patient-derived somatic cells and iPSCs, and develop new therapies for rare diseases. The CRISPR/Cas9 system stands out as the most extensively used method for genome editing due to its relative simplicity and superior efficiency compared to TALEN and ZFN. CRISPR/Cas9 is emerging as a feasible gene-editing option to treat rare monogenic and other genetically defined human diseases. SUMMARY Less than 5% of ~7000 known rare diseases have FDA-approved therapies, providing a compelling need for additional research and clinical trials to identify efficient treatment options for patients with rare diseases. Development of efficient genome-editing tools capable to correct or replace dysfunctional genes will lead to novel therapeutic approaches in these diseases.
Collapse
Affiliation(s)
- Arun Pradhan
- Center for Lung Regenerative Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, USA
| | - Tanya V. Kalin
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, USA
| | - Vladimir V. Kalinichenko
- Center for Lung Regenerative Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, USA
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, USA
| |
Collapse
|
48
|
Leal AF, Espejo-Mojica AJ, Sánchez OF, Ramírez CM, Reyes LH, Cruz JC, Alméciga-Díaz CJ. Lysosomal storage diseases: current therapies and future alternatives. J Mol Med (Berl) 2020; 98:931-946. [PMID: 32529345 DOI: 10.1007/s00109-020-01935-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 05/28/2020] [Accepted: 06/03/2020] [Indexed: 02/07/2023]
Abstract
Lysosomal storage disorders (LSDs) are a group of monogenic diseases characterized by progressive accumulation of undegraded substrates into the lysosome, due to mutations in genes that encode for proteins involved in normal lysosomal function. In recent years, several approaches have been explored to find effective and successful therapies, including enzyme replacement therapy, substrate reduction therapy, pharmacological chaperones, hematopoietic stem cell transplantation, and gene therapy. In the case of gene therapy, genome editing technologies have opened new horizons to accelerate the development of novel treatment alternatives for LSD patients. In this review, we discuss the current therapies for this group of disorders and present a detailed description of major genome editing technologies, as well as the most recent advances in the treatment of LSDs. We will further highlight the challenges and current bioethical debates of genome editing.
Collapse
Affiliation(s)
- Andrés Felipe Leal
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Cra. 7 No. 43-82 Building 54, Room 305A, Bogotá D.C, 110231, Colombia
| | - Angela Johana Espejo-Mojica
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Cra. 7 No. 43-82 Building 54, Room 305A, Bogotá D.C, 110231, Colombia
| | - Oscar F Sánchez
- Neurobiochemistry and Systems Physiology, Biochemistry and Nutrition Department, Faculty of Science, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
| | - Carlos Manuel Ramírez
- Department of Chemical and Food Engineering, Universidad de los Andes, Bogotá D.C., Colombia
| | - Luis Humberto Reyes
- Department of Chemical and Food Engineering, Universidad de los Andes, Bogotá D.C., Colombia
| | - Juan C Cruz
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá D.C., Colombia
| | - Carlos Javier Alméciga-Díaz
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Cra. 7 No. 43-82 Building 54, Room 305A, Bogotá D.C, 110231, Colombia.
| |
Collapse
|
49
|
Abdelhakim M, McMurray E, Syed AR, Kafkas S, Kamau AA, Schofield PN, Hoehndorf R. DDIEM: drug database for inborn errors of metabolism. Orphanet J Rare Dis 2020; 15:146. [PMID: 32527280 PMCID: PMC7291537 DOI: 10.1186/s13023-020-01428-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 05/28/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Inborn errors of metabolism (IEM) represent a subclass of rare inherited diseases caused by a wide range of defects in metabolic enzymes or their regulation. Of over a thousand characterized IEMs, only about half are understood at the molecular level, and overall the development of treatment and management strategies has proved challenging. An overview of the changing landscape of therapeutic approaches is helpful in assessing strategic patterns in the approach to therapy, but the information is scattered throughout the literature and public data resources. RESULTS We gathered data on therapeutic strategies for 300 diseases into the Drug Database for Inborn Errors of Metabolism (DDIEM). Therapeutic approaches, including both successful and ineffective treatments, were manually classified by their mechanisms of action using a new ontology. CONCLUSIONS We present a manually curated, ontologically formalized knowledgebase of drugs, therapeutic procedures, and mitigated phenotypes. DDIEM is freely available through a web interface and for download at http://ddiem.phenomebrowser.net.
Collapse
Affiliation(s)
- Marwa Abdelhakim
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology, 4700 KAUST, Thuwal, 23955 Kingdom of Saudi Arabia
- Computer, Electrical and Mathematical Sciences & Engineering Division (CEMSE), King Abdullah University of Science and Technology, 4700 KAUST, Thuwal, PO 23955 Saudi Arabia
| | - Eunice McMurray
- Department of Physiology, Development & Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3EG United Kingdom
| | - Ali Raza Syed
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology, 4700 KAUST, Thuwal, 23955 Kingdom of Saudi Arabia
- Computer, Electrical and Mathematical Sciences & Engineering Division (CEMSE), King Abdullah University of Science and Technology, 4700 KAUST, Thuwal, PO 23955 Saudi Arabia
| | - Senay Kafkas
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology, 4700 KAUST, Thuwal, 23955 Kingdom of Saudi Arabia
- Computer, Electrical and Mathematical Sciences & Engineering Division (CEMSE), King Abdullah University of Science and Technology, 4700 KAUST, Thuwal, PO 23955 Saudi Arabia
| | - Allan Anthony Kamau
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology, 4700 KAUST, Thuwal, 23955 Kingdom of Saudi Arabia
| | - Paul N Schofield
- Department of Physiology, Development & Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3EG United Kingdom
| | - Robert Hoehndorf
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology, 4700 KAUST, Thuwal, 23955 Kingdom of Saudi Arabia
- Computer, Electrical and Mathematical Sciences & Engineering Division (CEMSE), King Abdullah University of Science and Technology, 4700 KAUST, Thuwal, PO 23955 Saudi Arabia
| |
Collapse
|
50
|
Asai A, Konno M, Kawamoto K, Isotani A, Mori M, Eguchi H, Doki Y, Arai T, Ishii H. Hereditary pancreatitis model by blastocyst complementation in mouse. Oncotarget 2020; 11:2061-2073. [PMID: 32547704 PMCID: PMC7275788 DOI: 10.18632/oncotarget.27595] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 04/03/2020] [Indexed: 12/15/2022] Open
Abstract
The application of pluripotent stem cells is expected to contribute to the elucidation of unknown mechanism of human diseases. However, in vitro induction of organ-specific cells, such as pancreas and liver, is still difficult and the reproduction of their disorders in a model has been unfeasible. To study the mechanism of human hereditary pancreatitis (HP), we here performed the blastocyst complementation (BC) method. In the BC method, mouse embryonic stem (ES) cells harboring CRISPR/CAS9-mediated mutations in the Prss1 gene were injected into blastocysts with deficient Pdx1 gene, which is a critical transcription factor in the development of pancreas. The results showed that trypsin was activated extremely in Prss1-mutant mice. This implied that the mouse phenotype mimics that of human HP and that the BC method was useful for the reproduction and study of pancreatic disorders. The present study opens the possibility of investigating uncharacterized human diseases by utilizing the BC method.
Collapse
Affiliation(s)
- Ayumu Asai
- Department of Frontier Science for Cancer and Chemotherapy, Graduate School of Medicine, Osaka University, Suita 565-0871 Japan.,Department of Medical Data Science, Graduate School of Medicine, Osaka University, Suita 565-0871 Japan.,Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871 Japan
| | - Masamitsu Konno
- Department of Frontier Science for Cancer and Chemotherapy, Graduate School of Medicine, Osaka University, Suita 565-0871 Japan.,Department of Medical Data Science, Graduate School of Medicine, Osaka University, Suita 565-0871 Japan.,Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871 Japan
| | - Koichi Kawamoto
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871 Japan
| | - Ayako Isotani
- Organ Developmental Engineering, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma 630-0192 Japan
| | - Masaki Mori
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871 Japan.,Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582 Japan
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871 Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871 Japan
| | | | - Hideshi Ishii
- Department of Frontier Science for Cancer and Chemotherapy, Graduate School of Medicine, Osaka University, Suita 565-0871 Japan.,Department of Medical Data Science, Graduate School of Medicine, Osaka University, Suita 565-0871 Japan
| |
Collapse
|