1
|
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
|
2
|
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
|
3
|
Anastasi F, Dilillo M, Pellegrini D, McDonnell LA. Isolation and Proteomic Analysis of Mouse Serum Small Extracellular Vesicles for Individual Subject Analysis. Methods Mol Biol 2022; 2504:41-54. [PMID: 35467278 DOI: 10.1007/978-1-0716-2341-1_4] [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: 06/14/2023]
Abstract
Proteomics characterization of blood and circulating material has been extensively explored for the study of pathological states. In particular, circulating small extracellular vesicles (sEV, diameter: 30-150 nm) are known to play an important role in intercellular communication processes, and proteomics profiling has been explored to develop minimally invasive assays for disease monitoring and diagnosis. Due to the genetic and physiological similarities between the two species, and also on account of their shorter life span and rapid disease progression, rodent models are the most commonly used animal model for many human diseases. Such models have provided invaluable insight into the molecular mechanisms of disease progression, candidate drug efficacy, therapy monitoring, and biomarkers research.Longitudinal investigations, in which individuals are monitored over periods of time, are more able to resolve molecular changes during disease progression because they circumvent the inter-individual variation. Longitudinal investigations of rodent models are challenging because of the limited amount of blood that can be withdrawn at each time; the American Association of Veterinary Science stipulates that fortnightly sampling should be limited to a maximum of 10% of the total blood volume. For adult mice this corresponds to approximately 75 μL of serum. We developed an approach for the isolation and characterization of serum sEV proteins from just 50 μL of serum, for longitudinal studies of disease mouse models. This chapter describes in detail the steps and considerations involved in the sEV isolation, morphological characterization, and proteome profiling by mass spectrometry.
Collapse
Affiliation(s)
- Federica Anastasi
- NEST Laboratories, Scuola Normale Superiore, Pisa, Italy
- Fondazione Pisana per la Scienza ONLUS, Pisa, Italy
| | | | - Davide Pellegrini
- NEST Laboratories, Scuola Normale Superiore, Pisa, Italy
- Fondazione Pisana per la Scienza ONLUS, Pisa, Italy
| | | |
Collapse
|
4
|
Tao Y, Chen L, Pan M, Zhu F, Zhu D. Tailored Biosensors for Drug Screening, Efficacy Assessment, and Toxicity Evaluation. ACS Sens 2021; 6:3146-3162. [PMID: 34516080 DOI: 10.1021/acssensors.1c01600] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Biosensors have been flourishing in the field of drug discovery with pronounced developments in the past few years. They facilitate the screening and discovery of innovative drugs. However, there is still a lack of critical reviews that compare the merits and shortcomings of these biosensors from a pharmaceutical point of view. This contribution presents a critical and up-to-date overview on the recent progress of tailored biosensors, including surface plasmon resonance, fluorescent, photoelectrochemical, and electrochemical systems with emphasis on their mechanisms and applications in drug screening, efficacy assessment, and toxicity evaluation. Multiple functional nanomaterials have also been incorporated into the biosensors. Representative examples of each type of biosensors are discussed in terms of design strategy, response mechanism, and potential applications. In the end, we also compare the results and summarize the major insights gained from the works, demonstrating the challenges and prospects of biosensors-assisted drug discovery.
Collapse
Affiliation(s)
- Yi Tao
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Lin Chen
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Meiling Pan
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Fei Zhu
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Dong Zhu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| |
Collapse
|
5
|
Tsigkos S, Mariz S, Sheean ME, Larsson K, Magrelli A, Stoyanova-Beninska V. Regulatory Standards in Orphan Medicinal Product Designation in the EU. Front Med (Lausanne) 2021; 8:698534. [PMID: 34249982 PMCID: PMC8268149 DOI: 10.3389/fmed.2021.698534] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 06/01/2021] [Indexed: 11/21/2022] Open
Abstract
Twenty years of orphan regulation in Europe have now elapsed, with almost 2,400 orphan designated medicinal products and more than 190 orphan products authorised in the EU. Alongside the evolution in understanding of rare diseases, considerable regulatory knowledge has also been accumulated regarding the level of evidence that would support inclusion of products into the framework. This article reviews publications and regulatory documents pertaining to orphan medicinal product designation in the EU and discusses the general expectations in submitted applications as reflected in the current regulatory practise. Important elements to recommend granting a European orphan designation are the key considerations of orphan condition, medical plausibility, seriousness, and prevalence, while significant benefit is also assessed when there are authorised medicinal products for the sought indication. This review attempts to clarify the specific concepts currently used in that regard and discusses how the available data can be used to justify the criteria for designation. Moving away from theoretical expectations or assumptions, it stresses that the applications have to be complemented with nosological and epidemiological justifications pertaining to the proposed condition, as well as relevant data in specific non-clinical in vivo models or in affected patients to support inclusion into the orphan scheme.
Collapse
Affiliation(s)
- Stelios Tsigkos
- Orphan Medicines Office, European Medicines Agency, Amsterdam, Netherlands
| | - Segundo Mariz
- Orphan Medicines Office, European Medicines Agency, Amsterdam, Netherlands
| | | | - Kristina Larsson
- Orphan Medicines Office, European Medicines Agency, Amsterdam, Netherlands
| | - Armando Magrelli
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy.,Committee of Orphan Medicinal Products, European Medicines Agency, Amsterdam, Netherlands
| | - Violeta Stoyanova-Beninska
- Committee of Orphan Medicinal Products, European Medicines Agency, Amsterdam, Netherlands.,Medicines Evaluation Board, Utrecht, Netherlands
| |
Collapse
|
6
|
Stewart JB. Current progress with mammalian models of mitochondrial DNA disease. J Inherit Metab Dis 2021; 44:325-342. [PMID: 33099782 DOI: 10.1002/jimd.12324] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 10/19/2020] [Accepted: 10/21/2020] [Indexed: 12/16/2022]
Abstract
Mitochondrial disorders make up a large class of heritable diseases that cause a broad array of different human pathologies. They can affect many different organ systems, or display very specific tissue presentation, and can lead to illness either in childhood or later in life. While the over 1200 genes encoded in the nuclear DNA play an important role in human mitochondrial disease, it has been known for over 30 years that mutations of the mitochondria's own small, multicopy DNA chromosome (mtDNA) can lead to heritable human diseases. Unfortunately, animal mtDNA has resisted transgenic and directed genome editing technologies until quite recently. As such, animal models to aid in our understanding of these diseases, and to explore preclinical therapeutic research have been quite rare. This review will discuss the unusual properties of animal mitochondria that have hindered the generation of animal models. It will also discuss the existing mammalian models of human mtDNA disease, describe the methods employed in their generation, and will discuss recent advances in the targeting of DNA-manipulating enzymes to the mitochondria and how these may be employed to generate new models.
Collapse
Affiliation(s)
- James Bruce Stewart
- Max Planck Institute for Biology of Ageing, Cologne, Germany
- Wellcome Centre for Mitochondrial Research, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
7
|
Linn E, Ghanem L, Bhakta H, Greer C, Avella M. Genes Regulating Spermatogenesis and Sperm Function Associated With Rare Disorders. Front Cell Dev Biol 2021; 9:634536. [PMID: 33665191 PMCID: PMC7921155 DOI: 10.3389/fcell.2021.634536] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 01/20/2021] [Indexed: 12/26/2022] Open
Abstract
Spermatogenesis is a cell differentiation process that ensures the production of fertilizing sperm, which ultimately fuse with an egg to form a zygote. Normal spermatogenesis relies on Sertoli cells, which preserve cell junctions while providing nutrients for mitosis and meiosis of male germ cells. Several genes regulate normal spermatogenesis, some of which are not exclusively expressed in the testis and control multiple physiological processes in an organism. Loss-of-function mutations in some of these genes result in spermatogenesis and sperm functionality defects, potentially leading to the insurgence of rare genetic disorders. To identify genetic intersections between spermatogenesis and rare diseases, we screened public archives of human genetic conditions available on the Genetic and Rare Diseases Information Center (GARD), the Online Mendelian Inheritance in Man (OMIM), and the Clinical Variant (ClinVar), and after an extensive literature search, we identified 22 distinct genes associated with 21 rare genetic conditions and defective spermatogenesis or sperm function. These protein-coding genes regulate Sertoli cell development and function during spermatogenesis, checkpoint signaling pathways at meiosis, cellular organization and shape definition during spermiogenesis, sperm motility, and capacitation at fertilization. A number of these genes regulate folliculogenesis and oogenesis as well. For each gene, we review the genotype–phenotype association together with associative or causative polymorphisms in humans, and provide a description of the shared molecular mechanisms that regulate gametogenesis and fertilization obtained in transgenic animal models.
Collapse
Affiliation(s)
- Emma Linn
- Department of Biological Science, College of Engineering and Natural Sciences, University of Tulsa, Tulsa, OK, United States
| | - Lillian Ghanem
- Department of Biological Science, College of Engineering and Natural Sciences, University of Tulsa, Tulsa, OK, United States
| | - Hanisha Bhakta
- Department of Biological Science, College of Engineering and Natural Sciences, University of Tulsa, Tulsa, OK, United States
| | - Cory Greer
- Department of Biological Science, College of Engineering and Natural Sciences, University of Tulsa, Tulsa, OK, United States
| | - Matteo Avella
- Department of Biological Science, College of Engineering and Natural Sciences, University of Tulsa, Tulsa, OK, United States
| |
Collapse
|
8
|
Parenti G, Medina DL, Ballabio A. The rapidly evolving view of lysosomal storage diseases. EMBO Mol Med 2021; 13:e12836. [PMID: 33459519 PMCID: PMC7863408 DOI: 10.15252/emmm.202012836] [Citation(s) in RCA: 131] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 12/17/2022] Open
Abstract
Lysosomal storage diseases are a group of metabolic disorders caused by deficiencies of several components of lysosomal function. Most commonly affected are lysosomal hydrolases, which are involved in the breakdown and recycling of a variety of complex molecules and cellular structures. The understanding of lysosomal biology has progressively improved over time. Lysosomes are no longer viewed as organelles exclusively involved in catabolic pathways, but rather as highly dynamic elements of the autophagic-lysosomal pathway, involved in multiple cellular functions, including signaling, and able to adapt to environmental stimuli. This refined vision of lysosomes has substantially impacted on our understanding of the pathophysiology of lysosomal disorders. It is now clear that substrate accumulation triggers complex pathogenetic cascades that are responsible for disease pathology, such as aberrant vesicle trafficking, impairment of autophagy, dysregulation of signaling pathways, abnormalities of calcium homeostasis, and mitochondrial dysfunction. Novel technologies, in most cases based on high-throughput approaches, have significantly contributed to the characterization of lysosomal biology or lysosomal dysfunction and have the potential to facilitate diagnostic processes, and to enable the identification of new therapeutic targets.
Collapse
Affiliation(s)
- Giancarlo Parenti
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy.,Department of Translational Medical Sciences, Section of Pediatrics, Federico II University, Naples, Italy
| | - Diego L Medina
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy.,Department of Translational Medical Sciences, Section of Pediatrics, Federico II University, Naples, Italy
| | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy.,Department of Translational Medical Sciences, Section of Pediatrics, Federico II University, Naples, Italy.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.,Jan and Dan Duncan Neurological Research Institute, Texas Children Hospital, Houston, TX, USA.,SSM School for Advanced Studies, Federico II University, Naples, Italy
| |
Collapse
|
9
|
Murillo-Cuesta S, Artuch R, Asensio F, de la Villa P, Dierssen M, Enríquez JA, Fillat C, Fourcade S, Ibáñez B, Montoliu L, Oliver E, Pujol A, Salido E, Vallejo M, Varela-Nieto I. The Value of Mouse Models of Rare Diseases: A Spanish Experience. Front Genet 2020; 11:583932. [PMID: 33173540 PMCID: PMC7591746 DOI: 10.3389/fgene.2020.583932] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 09/14/2020] [Indexed: 11/13/2022] Open
Abstract
Animal models are invaluable for biomedical research, especially in the context of rare diseases, which have a very low prevalence and are often complex. Concretely mouse models provide key information on rare disease mechanisms and therapeutic strategies that cannot be obtained by using only alternative methods, and greatly contribute to accelerate the development of new therapeutic options for rare diseases. Despite this, the use of experimental animals remains controversial. The combination of respectful management, ethical laws and transparency regarding animal experimentation contributes to improve society’s opinion about biomedical research and positively impacts on research quality, which eventually also benefits patients. Here we present examples of current advances in preclinical research in rare diseases using mouse models, together with our perspective on future directions and challenges.
Collapse
Affiliation(s)
- Silvia Murillo-Cuesta
- Biomedical Research Networking Center on Rare Diseases (CIBERER), Institute of Health Carlos III, Madrid, Spain.,Instituto de Investigaciones Biomédicas Alberto Sols (IIBM), Consejo Superior de Investigaciones Científicas/Universidad Autónoma de Madrid, Madrid, Spain.,Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| | - Rafael Artuch
- Biomedical Research Networking Center on Rare Diseases (CIBERER), Institute of Health Carlos III, Madrid, Spain.,Institut de Recerca Sant Joan de Déu (IRSJD), Barcelona, Spain
| | - Fernando Asensio
- Gregorio Marañón Institute for Health Research (IISGM), Madrid, Spain
| | - Pedro de la Villa
- Faculty of Medicine, University of Alcalá (UAH), Alcalá de Henares, Spain
| | - Mara Dierssen
- Biomedical Research Networking Center on Rare Diseases (CIBERER), Institute of Health Carlos III, Madrid, Spain.,Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Jose Antonio Enríquez
- Spanish National Center for Cardiovascular Research (CNIC), Institute of Health Carlos III, Madrid, Spain.,Biomedical Research Networking Center on Frailty and Healthy Ageing (CIBERFES), Institute of Health Carlos III, Madrid, Spain
| | - Cristina Fillat
- Biomedical Research Networking Center on Rare Diseases (CIBERER), Institute of Health Carlos III, Madrid, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Stéphane Fourcade
- Biomedical Research Networking Center on Rare Diseases (CIBERER), Institute of Health Carlos III, Madrid, Spain.,Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Borja Ibáñez
- Spanish National Center for Cardiovascular Research (CNIC), Institute of Health Carlos III, Madrid, Spain.,Biomedical Research Networking Center on Cardiovascular Diseases (CIBERCV), Institute of Health Carlos III, Madrid, Spain.,Cardiology Department, Fundación Jiménez Díaz University Hospital Health Research Institute (IIS-FJD), Madrid, Spain
| | - Lluis Montoliu
- Biomedical Research Networking Center on Rare Diseases (CIBERER), Institute of Health Carlos III, Madrid, Spain.,National Center for Biotechnology (CNB), Spanish National Research Council, Madrid, Spain
| | - Eduardo Oliver
- Spanish National Center for Cardiovascular Research (CNIC), Institute of Health Carlos III, Madrid, Spain.,Biomedical Research Networking Center on Cardiovascular Diseases (CIBERCV), Institute of Health Carlos III, Madrid, Spain
| | - Aurora Pujol
- Biomedical Research Networking Center on Rare Diseases (CIBERER), Institute of Health Carlos III, Madrid, Spain.,Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain.,Catalan Institution of Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Eduardo Salido
- Biomedical Research Networking Center on Rare Diseases (CIBERER), Institute of Health Carlos III, Madrid, Spain.,Unidad de Investigación, Hospital Universitario de Canarias, Instituto de Tecnologías Biomédicas (ITB), La Laguna, Spain
| | - Mario Vallejo
- Instituto de Investigaciones Biomédicas Alberto Sols (IIBM), Consejo Superior de Investigaciones Científicas/Universidad Autónoma de Madrid, Madrid, Spain.,Biomedical Research Networking Center on Diabetes and Metabolic Diseases (CIBERDEM), Institute of Health Carlos III, Madrid, Spain
| | - Isabel Varela-Nieto
- Biomedical Research Networking Center on Rare Diseases (CIBERER), Institute of Health Carlos III, Madrid, Spain.,Instituto de Investigaciones Biomédicas Alberto Sols (IIBM), Consejo Superior de Investigaciones Científicas/Universidad Autónoma de Madrid, Madrid, Spain.,Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| |
Collapse
|
10
|
Duarte DM, Beatriz da Silva Lima M, Sepodes B. The translational value of animal models in orphan medicines designations for rare paediatric neurological diseases. Regul Toxicol Pharmacol 2020; 118:104810. [PMID: 33122047 DOI: 10.1016/j.yrtph.2020.104810] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 10/14/2020] [Accepted: 10/23/2020] [Indexed: 12/17/2022]
Abstract
Rare diseases are characterized by a substantial unmet need mostly because the majority have limited, or no treatment options and a large number also affect children. Appropriate animal models, based on the knowledge of the molecular pathology of the human disease, are a significant element to support the medical plausibility of an orphan designation during the development of orphan medicines for rare neurological diseases. This observational, retrospective study aims to investigate the clinical or nonclinical nature of data submitted to support medical plausibility of orphan designations in the EU (2001-2019), for a group of rare and paediatric neurological diseases. From our sample of 30 diseases, 70% are rare with paediatric onset and 37% have approved orphan designations. The use of nonclinical data was significantly higher than clinical data (65% vs. 35%, p = 0.013) to support medical plausibility. Examples of diseases, with orphan designations based only in nonclinical data, are also discussed: Aicardi-Goutières syndrome and Centronuclear myopathy animal disease models, potentially used to support medical plausibility of medicines. Nonclinical appropriate models, assessing disease relevant endpoints, may contribute to increase the translational value of animal models, in paediatric and rare neurological area, to accelerate research and the effective development of treatment options.
Collapse
Affiliation(s)
| | | | - Bruno Sepodes
- Universidade de Lisboa, Faculdade de Farmácia, Lisbon, Portugal
| |
Collapse
|
11
|
Molecular Diagnosis and Novel Therapies for Neuromuscular Diseases. J Pers Med 2020; 10:jpm10030129. [PMID: 32947786 PMCID: PMC7564006 DOI: 10.3390/jpm10030129] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 09/14/2020] [Indexed: 02/07/2023] Open
Abstract
With the development of novel targeted therapies, including exon skipping/inclusion and gene replacement therapy, the field of neuromuscular diseases has drastically changed in the last several years. Until 2016, there had been no FDA-approved drugs to treat Duchenne muscular dystrophy (DMD), the most common muscular dystrophy. However, several new personalized therapies, including antisense oligonucleotides eteplirsen for DMD exon 51 skipping and golodirsen and viltolarsen for DMD exon 53 skipping, have been approved in the last 4 years. We are witnessing the start of a therapeutic revolution in neuromuscular diseases. However, the studies also made clear that these therapies are still far from a cure. Personalized genetic medicine for neuromuscular diseases faces several key challenges, including the difficulty of obtaining appropriate cell and animal models and limited its applicability. This Special Issue “Molecular Diagnosis and Novel Therapies for Neuromuscular/Musculoskeletal Diseases” highlights key areas of research progress that improve our understanding and the therapeutic outcomes of neuromuscular diseases in the personalized medicine era.
Collapse
|
12
|
A putative silencer variant in a spontaneous canine model of retinitis pigmentosa. PLoS Genet 2020; 16:e1008659. [PMID: 32150541 PMCID: PMC7082071 DOI: 10.1371/journal.pgen.1008659] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 03/19/2020] [Accepted: 02/06/2020] [Indexed: 01/19/2023] Open
Abstract
Retinitis pigmentosa (RP) is the leading cause of blindness with nearly two million people affected worldwide. Many genes have been implicated in RP, yet in 30–80% of the RP patients the genetic cause remains unknown. A similar phenotype, progressive retinal atrophy (PRA), affects many dog breeds including the Miniature Schnauzer. We performed clinical, genetic and functional experiments to identify the genetic cause of PRA in the breed. The age of onset and pattern of disease progression suggested that at least two forms of PRA, types 1 and 2 respectively, affect the breed, which was confirmed by genome-wide association study that implicated two distinct genomic loci in chromosomes 15 and X, respectively. Whole-genome sequencing revealed a fully segregating recessive regulatory variant in type 1 PRA. The associated variant has a very recent origin based on haplotype analysis and lies within a regulatory site with the predicted binding site of HAND1::TCF3 transcription factor complex. Luciferase assays suggested that mutated regulatory sequence increases expression. Case-control retinal expression comparison of six best HAND1::TCF3 target genes were analyzed with quantitative reverse-transcriptase PCR assay and indicated overexpression of EDN2 and COL9A2 in the affected retina. Defects in both EDN2 and COL9A2 have been previously associated with retinal degeneration. In summary, our study describes two genetically different forms of PRA and identifies a fully penetrant variant in type 1 form with a possible regulatory effect. This would be among the first reports of a regulatory variant in retinal degeneration in any species, and establishes a new spontaneous dog model to improve our understanding of retinal biology and gene regulation while the affected breed will benefit from a reliable genetic testing. Retinitis pigmentosa (RP) is a blinding eye disease that affects nearly two million people worldwide. Several genes and variants have been associated with the disease, but still 30–80% of the patients lack genetic diagnosis. There is currently no standard treatment for RP, and much is expected from gene therapy. A similar disease, called progressive retinal atrophy (PRA), affects many dog breeds. We performed clinical, genetic and functional analyses to find the genetic cause for PRA in Miniature Schnauzers. We discovered two forms of PRA in the breed, named type 1 and 2, and show that they are genetically distinct as they map to different chromosomes, 15 and X, respectively. Further genetic, bioinformatic and functional analyses discovered a fully penetrant recessive variant in a putative silencer region for type 1 PRA. Silencer regions are important for gene regulation and we found that two of its predicted target genes, EDN2 and COL9A2, were overexpressed in the retina of the affected dog. Defects in both EDN2 and COL9A2 have been associated with retinal degeneration. This study provides new insights to retinal biology while the genetic test guides better breeding choices.
Collapse
|
13
|
Rosenberg JB, Chen A, Kaminsky SM, Crystal RG, Sondhi D. Advances in the Treatment of Neuronal Ceroid Lipofuscinosis. Expert Opin Orphan Drugs 2019; 7:473-500. [PMID: 33365208 PMCID: PMC7755158 DOI: 10.1080/21678707.2019.1684258] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 10/21/2019] [Indexed: 12/27/2022]
Abstract
Neuronal ceroid lipofuscinoses (NCL) represent a class of neurodegenerative disorders involving defective lysosomal processing enzymes or receptors, leading to lysosomal storage disorders, typically characterized by observation of cognitive and visual impairments, epileptic seizures, ataxia, and deterioration of motor skills. Recent success of a biologic (Brineura®) for the treatment of neurologic manifestations of the central nervous system (CNS) has led to renewed interest in therapeutics for NCL, with the goal of ablating or reversing the impact of these devastating disorders. Despite complex challenges associated with CNS therapy, many treatment modalities have been evaluated, including enzyme replacement therapy, gene therapy, stem cell therapy, and small molecule pharmacotherapy. Because the clinical endpoints for the evaluation of candidate therapies are complex and often reliant on subjective clinical scales, the development of quantitative biomarkers for NCLs has become an apparent necessity for the validation of potential treatments. We will discuss the latest findings in the search for relevant biomarkers for assessing disease progression. For this review, we will focus primarily on recent pre-clinical and clinical developments for treatments to halt or cure these NCL diseases. Continued development of current therapies and discovery of newer modalities will be essential for successful therapeutics for NCL. AREAS COVERED The reader will be introduced to the NCL subtypes, natural histories, experimental animal models, and biomarkers for NCL progression; challenges and different therapeutic approaches, and the latest pre-clinical and clinical research for therapeutic development for the various NCLs. This review corresponds to the literatures covering the years from 1968 to mid-2019, but primarily addresses pre-clinical and clinical developments for the treatment of NCL disease in the last decade and as a follow-up to our 2013 review of the same topic in this journal. EXPERT OPINION Much progress has been made in the treatment of neurologic diseases, such as the NCLs, including better animal models and improved therapeutics with better survival outcomes. Encouraging results are being reported at symposiums and in the literature, with multiple therapeutics reaching the clinical trial stage for the NCLs. The potential for a cure could be at hand after many years of trial and error in the preclinical studies. The clinical development of enzyme replacement therapy (Brineura® for CLN2), immunosuppression (CellCept® for CLN3), and gene therapy vectors (for CLN1, CLN2, CLN3, and CLN6) are providing encouragement to families that have a child afflicted with NCL. We believe that successful therapies in the future may involve the combination of two or more therapeutic modalities to provide therapeutic benefit especially as the patients grow older.
Collapse
Affiliation(s)
- Jonathan B Rosenberg
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Alvin Chen
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Stephen M Kaminsky
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Ronald G Crystal
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Dolan Sondhi
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| |
Collapse
|
14
|
Kaukonen M, Woods S, Ahonen S, Lemberg S, Hellman M, Hytönen MK, Permi P, Glaser T, Lohi H. Maternal Inheritance of a Recessive RBP4 Defect in Canine Congenital Eye Disease. Cell Rep 2019; 23:2643-2652. [PMID: 29847795 PMCID: PMC6546432 DOI: 10.1016/j.celrep.2018.04.118] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 04/16/2018] [Accepted: 04/26/2018] [Indexed: 01/20/2023] Open
Abstract
Maternally skewed transmission of traits has been associated with genomic imprinting and oocyte-derived mRNA. We report canine congenital eye malformations, caused by an amino acid deletion (K12del) near the N terminus of retinol-binding protein (RBP4). The disease is only expressed when both dam and offspring are deletion homozygotes. RBP carries vitamin A (retinol) from hepatic stores to peripheral tissues, including the placenta and developing eye, where it is required to synthesize retinoic acid. Gestational vitamin A deficiency is a known risk factor for ocular birth defects. The K12del mutation disrupts RBP folding in vivo, decreasing its secretion from hepatocytes to serum. The maternal penetrance effect arises from an impairment in the sequential transfer of retinol across the placenta, via RBP encoded by maternal and fetal genomes. Our results demonstrate a mode of recessive maternal inheritance, with a physiological basis, and they extend previous observations on dominant-negative RBP4 alleles in humans.
Collapse
Affiliation(s)
- Maria Kaukonen
- Department of Veterinary Biosciences, University of Helsinki, 00014 Helsinki, Finland; Research Programs Unit, Molecular Neurology, University of Helsinki, 00014 Helsinki, Finland; The Folkhälsan Institute of Genetics, 00290 Helsinki, Finland
| | - Sean Woods
- Department of Cell Biology and Human Anatomy, University of California, Davis School of Medicine, Davis, CA 95616, USA
| | - Saija Ahonen
- Department of Veterinary Biosciences, University of Helsinki, 00014 Helsinki, Finland; Research Programs Unit, Molecular Neurology, University of Helsinki, 00014 Helsinki, Finland; The Folkhälsan Institute of Genetics, 00290 Helsinki, Finland
| | - Seppo Lemberg
- Department of Eye Diseases, Helsinki University Hospital, 00029 The Hospital District of Helsinki and Uusimaa, Finland
| | - Maarit Hellman
- Department of Chemistry, Nanoscience Center, University of Jyväskylä, 40014 Jyväskylä, Finland
| | - Marjo K Hytönen
- Department of Veterinary Biosciences, University of Helsinki, 00014 Helsinki, Finland; Research Programs Unit, Molecular Neurology, University of Helsinki, 00014 Helsinki, Finland; The Folkhälsan Institute of Genetics, 00290 Helsinki, Finland
| | - Perttu Permi
- Department of Chemistry, Nanoscience Center, University of Jyväskylä, 40014 Jyväskylä, Finland; Department of Biological and Environmental Science, Nanoscience Center, University of Jyväskylä, 40014 Jyväskylä, Finland
| | - Tom Glaser
- Department of Cell Biology and Human Anatomy, University of California, Davis School of Medicine, Davis, CA 95616, USA.
| | - Hannes Lohi
- Department of Veterinary Biosciences, University of Helsinki, 00014 Helsinki, Finland; Research Programs Unit, Molecular Neurology, University of Helsinki, 00014 Helsinki, Finland; The Folkhälsan Institute of Genetics, 00290 Helsinki, Finland.
| |
Collapse
|
15
|
Nonclinical data supporting orphan medicinal product designations: lessons from rare neurological conditions. Drug Discov Today 2018; 23:26-48. [DOI: 10.1016/j.drudis.2017.09.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 09/01/2017] [Accepted: 09/27/2017] [Indexed: 12/14/2022]
|
16
|
Kirkegaard T, Gray J, Priestman DA, Wallom KL, Atkins J, Olsen OD, Klein A, Drndarski S, Petersen NHT, Ingemann L, Smith DA, Morris L, Bornæs C, Jørgensen SH, Williams I, Hinsby A, Arenz C, Begley D, Jäättelä M, Platt FM. Heat shock protein-based therapy as a potential candidate for treating the sphingolipidoses. Sci Transl Med 2017; 8:355ra118. [PMID: 27605553 DOI: 10.1126/scitranslmed.aad9823] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 08/18/2016] [Indexed: 12/17/2022]
Abstract
Lysosomal storage diseases (LSDs) often manifest with severe systemic and central nervous system (CNS) symptoms. The existing treatment options are limited and have no or only modest efficacy against neurological manifestations of disease. We demonstrate that recombinant human heat shock protein 70 (HSP70) improves the binding of several sphingolipid-degrading enzymes to their essential cofactor bis(monoacyl)glycerophosphate in vitro. HSP70 treatment reversed lysosomal pathology in primary fibroblasts from 14 patients with eight different LSDs. HSP70 penetrated effectively into murine tissues including the CNS and inhibited glycosphingolipid accumulation in murine models of Fabry disease (Gla(-/-)), Sandhoff disease (Hexb(-/-)), and Niemann-Pick disease type C (Npc1(-/-)) and attenuated a wide spectrum of disease-associated neurological symptoms in Hexb(-/-) and Npc1(-/-) mice. Oral administration of arimoclomol, a small-molecule coinducer of HSPs that is currently in clinical trials for Niemann-Pick disease type C (NPC), recapitulated the effects of recombinant human HSP70, suggesting that heat shock protein-based therapies merit clinical evaluation for treating LSDs.
Collapse
Affiliation(s)
| | - James Gray
- Department of Pharmacology, University of Oxford, Oxford, U.K
| | | | | | - Jennifer Atkins
- Department of Pharmacology, University of Oxford, Oxford, U.K
| | - Ole Dines Olsen
- Orphazyme ApS, Copenhagen, Denmark. Cell Death and Metabolism Unit, Center for Autophagy, Recycling, and Metabolism, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Alexander Klein
- Institut für Chemie der Humboldt-Universität zu Berlin, Berlin, Germany
| | | | | | | | - David A Smith
- Department of Pharmacology, University of Oxford, Oxford, U.K
| | - Lauren Morris
- Department of Pharmacology, University of Oxford, Oxford, U.K
| | | | | | - Ian Williams
- Department of Pharmacology, University of Oxford, Oxford, U.K
| | | | - Christoph Arenz
- Institut für Chemie der Humboldt-Universität zu Berlin, Berlin, Germany
| | - David Begley
- Institute of Pharmaceutical Science, King's College London, London, U.K
| | - Marja Jäättelä
- Cell Death and Metabolism Unit, Center for Autophagy, Recycling, and Metabolism, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Frances M Platt
- Department of Pharmacology, University of Oxford, Oxford, U.K
| |
Collapse
|
17
|
Sukoff Rizzo SJ, Crawley JN. Behavioral Phenotyping Assays for Genetic Mouse Models of Neurodevelopmental, Neurodegenerative, and Psychiatric Disorders. Annu Rev Anim Biosci 2017; 5:371-389. [PMID: 28199172 DOI: 10.1146/annurev-animal-022516-022754] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Animal models offer heuristic research tools to understand the causes of human diseases and to identify potential treatments. With rapidly evolving genetic engineering technologies, mutations identified in a human disorder can be generated in the mouse genome. Phenotypic outcomes of the mutation are then explicated to confirm hypotheses about causes and to discover effective therapeutics. Most neurodevelopmental, neurodegenerative, and psychiatric disorders are diagnosed primarily by their prominent behavioral symptoms. Mouse behavioral assays analogous to the human symptoms have been developed to analyze the consequences of mutations and to evaluate proposed therapeutics preclinically. Here we describe the range of mouse behavioral tests available in the established behavioral neuroscience literature, along with examples of their translational applications. Concepts presented have been successfully used in other species, including flies, worms, fish, rats, pigs, and nonhuman primates. Identical strategies can be employed to test hypotheses about environmental causes and gene × environment interactions.
Collapse
Affiliation(s)
| | - Jacqueline N Crawley
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California, Davis School of Medicine, Sacramento, California 95817;
| |
Collapse
|
18
|
Lochmüller H, Torrent I Farnell J, Le Cam Y, Jonker AH, Lau LP, Baynam G, Kaufmann P, Dawkins HJ, Lasko P, Austin CP, Boycott KM. The International Rare Diseases Research Consortium: Policies and Guidelines to maximize impact. Eur J Hum Genet 2017; 25:1293-1302. [PMID: 29158551 PMCID: PMC5865169 DOI: 10.1038/s41431-017-0008-z] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 08/03/2017] [Accepted: 08/23/2017] [Indexed: 12/21/2022] Open
Abstract
The International Rare Diseases Research Consortium (IRDiRC) has agreed on IRDiRC Policies and Guidelines, following extensive deliberations and discussions in 2012 and 2013, as a first step towards improving coordination of research efforts worldwide. The 25 funding members and 3 patient umbrella organizations (as of early 2013) of IRDiRC, a consortium of research funders that focuses on improving diagnosis and therapy for rare disease patients, agreed in Dublin, Ireland in April 2013 on the Policies and Guidelines that emphasize collaboration in rare disease research, the involvement of patients and their representatives in all relevant aspects of research, as well as the sharing of data and resources. The Policies and Guidelines provide guidance on ontologies, diagnostics, biomarkers, patient registries, biobanks, natural history, therapeutics, models, publication, intellectual property, and communication. Most IRDiRC members-currently nearly 50 strong-have since incorporated its policies in their funding calls and some have chosen to exceed the requirements laid out, for instance in relation to data sharing. The IRDiRC Policies and Guidelines are the first, detailed agreement of major public and private funding organizations worldwide to govern rare disease research, and may serve as a template for other areas of international research collaboration. While it is too early to assess their full impact on research productivity and patient benefit, the IRDiRC Policies and Guidelines have already contributed significantly to improving transparency and collaboration in rare disease research.
Collapse
Affiliation(s)
- Hanns Lochmüller
- Institute of Genetic Medicine, Newcastle University, Newcastle Upon Tyne, UK.
| | | | - Yann Le Cam
- EURORDIS-Rare Diseases Europe, Paris, France
| | | | - Lilian Pl Lau
- IRDiRC Scientific Secretariat, Inserm US-14, Paris, France
| | - Gareth Baynam
- Genetic Services of Western Australia, King Edward Memorial Hospital, Perth, WA, Australia
- Western Australian Register of Developmental Anomalies, WA, Perth, Australia
| | - Petra Kaufmann
- National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, Bethesda, MD, USA
| | - Hugh Js Dawkins
- Office of Population Health Genomics, Public Health Division, Department of Health, Government of Western Australia, Perth, WA, Australia
| | - Paul Lasko
- Department of Biology, McGill University, Montréal, QC, Canada
| | - Christopher P Austin
- National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, Bethesda, MD, USA
| | - Kym M Boycott
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
| |
Collapse
|
19
|
Sun W, Zheng W, Simeonov A. Drug discovery and development for rare genetic disorders. Am J Med Genet A 2017; 173:2307-2322. [PMID: 28731526 DOI: 10.1002/ajmg.a.38326] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 05/17/2017] [Indexed: 12/14/2022]
Abstract
Approximately 7,000 rare diseases affect millions of individuals in the United States. Although rare diseases taken together have an enormous impact, there is a significant gap between basic research and clinical interventions. Opportunities now exist to accelerate drug development for the treatment of rare diseases. Disease foundations and research centers worldwide focus on better understanding rare disorders. Here, the state-of-the-art drug discovery strategies for small molecules and biological approaches for orphan diseases are reviewed. Rare diseases are usually genetic diseases; hence, employing pharmacogenetics to develop treatments and using whole genome sequencing to identify the etiologies for such diseases are appropriate strategies to exploit. Beginning with high throughput screening of small molecules, the benefits and challenges of target-based and phenotypic screens are discussed. Explanations and examples of drug repurposing are given; drug repurposing as an approach to quickly move programs to clinical trials is evaluated. Consideration is given to the category of biologics which include gene therapy, recombinant proteins, and autologous transplants. Disease models, including animal models and induced pluripotent stem cells (iPSCs) derived from patients, are surveyed. Finally, the role of biomarkers in drug discovery and development, as well as clinical trials, is elucidated.
Collapse
Affiliation(s)
- Wei Sun
- National Center for Advancing Translational Sciences, National Institutes of Health, Medical Center Drive, Bethesda, Maryland
| | - Wei Zheng
- National Center for Advancing Translational Sciences, National Institutes of Health, Medical Center Drive, Bethesda, Maryland
| | - Anton Simeonov
- National Center for Advancing Translational Sciences, National Institutes of Health, Medical Center Drive, Bethesda, Maryland
| |
Collapse
|
20
|
Jin SE, Hwang SJ. Ocular delivery systems for the administration of antibody therapeutics. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2017. [DOI: 10.1007/s40005-017-0336-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
21
|
Yu JH, Tang HJ, Zhang WG, Zhu ZY, Ruan XX, Lu BC. Catheterization of the gallbladder: A novel mouse model of severe acute cholangitis. World J Gastroenterol 2017; 23:1771-1779. [PMID: 28348482 PMCID: PMC5352917 DOI: 10.3748/wjg.v23.i10.1771] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 01/12/2017] [Accepted: 02/08/2017] [Indexed: 02/07/2023] Open
Abstract
AIM To establish a severe acute cholangitis (SAC) model in mice.
METHODS Cholecystic catheterization was performed under the condition of bile duct ligation (BDL). Trans-cholecystic injection of lipopolysaccharide (LPS) was defined as the SAC animal model. Sham operation group, intraperitoneal injection of LPS without BDL group, intraperitoneal injection of LPS with BDL group and trans-cholecystic injection of normal saline with BDL group were defined as control groups. The survival rates and tissue injuries in liver, lungs and kidney were evaluated.
RESULTS Mice in the SAC group showed a time-dependent mortality and much more severe tissue injuries in liver, lungs and kidney, compared with other groups. However, relieving biliary obstruction could effectively reduce mortality and attenuate liver injury in the SAC mouse model.
CONCLUSION Trans-cholecystic injection of LPS under the condition of biliary obstruction could establish a repeatable and reversible mouse model of SAC.
Collapse
|
22
|
Integrative Systems Biology Investigation of Fabry Disease. Diseases 2016; 4:diseases4040035. [PMID: 28933415 PMCID: PMC5456327 DOI: 10.3390/diseases4040035] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 11/02/2016] [Accepted: 11/10/2016] [Indexed: 02/06/2023] Open
Abstract
Fabry disease (FD) is a rare X-linked recessive genetic disorder caused by a deficient activity of the lysosomal enzyme alpha-galactosidase A (GLA) and is characterised by intra-lysosomal accumulation of globotriaosylceramide (Gb3). We performed a meta-analysis of peer-reviewed publications including high-throughput omics technologies including naïve patients and those undergoing enzyme replacement therapy (ERT). This study describes FD on a systems level using a systems biology approach, in which molecular data sourced from multi-omics studies is extracted from the literature and integrated as a whole in order to reveal the biochemical processes and molecular pathways potentially affected by the dysregulation of differentially expressed molecules. In this way new insights are provided that describe the pathophysiology of this rare disease. Using gene ontology and pathway term clustering, FD displays the involvement of major biological processes such as the acute inflammatory response, regulation of wound healing, extracellular matrix (ECM) remodelling, regulation of peptidase activity, and cellular response to reactive oxygen species (ROS). Differential expression of acute-phase response proteins in the groups of naïve (up-regulation of ORM1, ORM2, ITIH4, SERPINA3 and FGA) and ERT (down-regulation of FGA, ORM1 and ORM2) patients could be potential hallmarks for distinction of these two patient groups.
Collapse
|
23
|
Xu M, Motabar O, Ferrer M, Marugan JJ, Zheng W, Ottinger EA. Disease models for the development of therapies for lysosomal storage diseases. Ann N Y Acad Sci 2016; 1371:15-29. [PMID: 27144735 DOI: 10.1111/nyas.13052] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 03/02/2016] [Accepted: 03/02/2016] [Indexed: 12/11/2022]
Abstract
Lysosomal storage diseases (LSDs) are a group of rare diseases in which the function of the lysosome is disrupted by the accumulation of macromolecules. The complexity underlying the pathogenesis of LSDs and the small, often pediatric, population of patients make the development of therapies for these diseases challenging. Current treatments are only available for a small subset of LSDs and have not been effective at treating neurological symptoms. Disease-relevant cellular and animal models with high clinical predictability are critical for the discovery and development of new treatments for LSDs. In this paper, we review how LSD patient primary cells and induced pluripotent stem cell-derived cellular models are providing novel assay systems in which phenotypes are more similar to those of the human LSD physiology. Furthermore, larger animal disease models are providing additional tools for evaluation of the efficacy of drug candidates. Early predictors of efficacy and better understanding of disease biology can significantly affect the translational process by focusing efforts on those therapies with the higher probability of success, thus decreasing overall time and cost spent in clinical development and increasing the overall positive outcomes in clinical trials.
Collapse
Affiliation(s)
- Miao Xu
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland.,Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Omid Motabar
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Marc Ferrer
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Juan J Marugan
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Wei Zheng
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Elizabeth A Ottinger
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| |
Collapse
|
24
|
Joshi R, Pankova N, Wang H, Baek DSH, Zhao X, Reyad M, Boyd SR. Spontaneously occurring fundus findings observed using confocal scanning laser ophthalmoscopy in wild type Sprague Dawley rats. Regul Toxicol Pharmacol 2016; 77:160-6. [PMID: 26873774 DOI: 10.1016/j.yrtph.2016.02.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 02/02/2016] [Indexed: 11/17/2022]
Abstract
PURPOSE Non-invasive in vivo imaging is an increasingly used component of pre-clinical research. However, to reliably interpret data, it may be necessary to identify and document pre-existent findings prior to initiating long-term or intensive protocols, particularly where toxicity or efficacy is under investigation. Here we report here spontaneously occurring findings from the Sprague Dawley (SD) rat eye using multi-modal confocal scanning laser ophthalmoscopy (cSLO). METHODS As part of ongoing studies, with the goal of excluding animals with abnormalities from further investigation, a total of 165 wild type SD rats (312 eyes) were assessed using cSLO imaging at baseline prior to initiating experiments to detect, describe, and determine the prevalence of spontaneous fundus findings. RESULTS Using fundus autofluorescence (FAF) as the primary screening modality, over 30% of analyzed eyes possessed some fundus finding that differed from the normal composite reference image. Unexpectedly, 100% of eyes demonstrated a diffuse hyperfluorescent region in the posterior pole that was ultimately considered normal, and formed part of the reference. Evaluated by three independent reviewers, five groups of FAF abnormalities were defined, based primarily on shape and size of the lesion. Of these, the most extensive lesions were further analyzed using infrared reflectance (IR) and red free (RF) imaging. White light and autofluorescent microscopy of excised tissue confirmed that the extensive lesions were derived from abnormalities in both the isolated retina and posterior eyecups. CONCLUSIONS Given the newly described hyperfluorescent glow that appears in all eyes, and the high basal rate of spontaneous lesions in the outbred SD rat, we suggest that investigators be aware of the variants of normal, and that baseline in vivo screening be considered prior to initiating intensive or expensive investigation.
Collapse
Affiliation(s)
- Rahul Joshi
- Keenan Research Centre for Biomedical Science, St Michael's Hospital, 209 Victoria Street, Toronto, ON, Canada, M5B 1W8
| | - Natalie Pankova
- University of Toronto, Department of Laboratory Medicine and Pathobiology, 1 King's College Circle, Toronto, ON, Canada, M5S 1A8; University of Toronto, Department of Ophthalmology and Vision Sciences, 340 College Street, Toronto, ON, Canada, M5T 3A9; Keenan Research Centre for Biomedical Science, St Michael's Hospital, 209 Victoria Street, Toronto, ON, Canada, M5B 1W8
| | - Hai Wang
- Keenan Research Centre for Biomedical Science, St Michael's Hospital, 209 Victoria Street, Toronto, ON, Canada, M5B 1W8
| | - David Sung Hyeon Baek
- University of Toronto, Department of Laboratory Medicine and Pathobiology, 1 King's College Circle, Toronto, ON, Canada, M5S 1A8; Keenan Research Centre for Biomedical Science, St Michael's Hospital, 209 Victoria Street, Toronto, ON, Canada, M5B 1W8
| | - Xu Zhao
- Keenan Research Centre for Biomedical Science, St Michael's Hospital, 209 Victoria Street, Toronto, ON, Canada, M5B 1W8
| | - Matthew Reyad
- Keenan Research Centre for Biomedical Science, St Michael's Hospital, 209 Victoria Street, Toronto, ON, Canada, M5B 1W8
| | - Shelley R Boyd
- University of Toronto, Department of Laboratory Medicine and Pathobiology, 1 King's College Circle, Toronto, ON, Canada, M5S 1A8; University of Toronto, Department of Ophthalmology and Vision Sciences, 340 College Street, Toronto, ON, Canada, M5T 3A9; Keenan Research Centre for Biomedical Science, St Michael's Hospital, 209 Victoria Street, Toronto, ON, Canada, M5B 1W8; Department of Ophthalmology, St. Michael's Hospital, 30 Bond Street, Toronto, ON, Canada, M5B 1W8; Biomedical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON, Canada, L8S 4K1.
| |
Collapse
|
25
|
Nicolson GL, de Mattos GF, Settineri R, Costa C, Ellithorpe R, Rosenblatt S, La Valle J, Jimenez A, Ohta S. Clinical Effects of Hydrogen Administration: From Animal and Human Diseases to Exercise Medicine. ACTA ACUST UNITED AC 2016. [DOI: 10.4236/ijcm.2016.71005] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
26
|
Plantié E, Migocka-Patrzałek M, Daczewska M, Jagla K. Model organisms in the fight against muscular dystrophy: lessons from drosophila and Zebrafish. Molecules 2015; 20:6237-53. [PMID: 25859781 PMCID: PMC6272363 DOI: 10.3390/molecules20046237] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 03/31/2015] [Accepted: 04/01/2015] [Indexed: 01/01/2023] Open
Abstract
Muscular dystrophies (MD) are a heterogeneous group of genetic disorders that cause muscle weakness, abnormal contractions and muscle wasting, often leading to premature death. More than 30 types of MD have been described so far; those most thoroughly studied are Duchenne muscular dystrophy (DMD), myotonic dystrophy type 1 (DM1) and congenital MDs. Structurally, physiologically and biochemically, MDs affect different types of muscles and cause individual symptoms such that genetic and molecular pathways underlying their pathogenesis thus remain poorly understood. To improve our knowledge of how MD-caused muscle defects arise and to find efficacious therapeutic treatments, different animal models have been generated and applied. Among these, simple non-mammalian Drosophila and zebrafish models have proved most useful. This review discusses how zebrafish and Drosophila MD have helped to identify genetic determinants of MDs and design innovative therapeutic strategies with a special focus on DMD, DM1 and congenital MDs.
Collapse
Affiliation(s)
- Emilie Plantié
- GReD (Genetics, Reproduction and Development laboratory), INSERM U1103, CNRS UMR6293, University of Clermont-Ferrand, 28 place Henri-Dunant, 63000 Clermont-Ferrand, France; E-Mail:
| | - Marta Migocka-Patrzałek
- Department of Animal Developmental Biology, Institute of Experimental Biology, University of Wroclaw, 21 Sienkiewicza Street, 50-335 Wroclaw, Poland; E-Mails: (M.M.-P.); (M.D.)
| | - Małgorzata Daczewska
- Department of Animal Developmental Biology, Institute of Experimental Biology, University of Wroclaw, 21 Sienkiewicza Street, 50-335 Wroclaw, Poland; E-Mails: (M.M.-P.); (M.D.)
| | - Krzysztof Jagla
- GReD (Genetics, Reproduction and Development laboratory), INSERM U1103, CNRS UMR6293, University of Clermont-Ferrand, 28 place Henri-Dunant, 63000 Clermont-Ferrand, France; E-Mail:
| |
Collapse
|
27
|
Animal Models with a Genetic Alteration of the ACE2/Ang-(1-7)/Mas Axis. THE PROTECTIVE ARM OF THE RENIN ANGIOTENSIN SYSTEM (RAS) 2015. [PMCID: PMC7150279 DOI: 10.1016/b978-0-12-801364-9.00022-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The aim of this chapter is to describe the animal models generated by transgenic technology for the functional analysis of the protective axis of the renin–angiotensin system, consisting of angiotensin-converting enzyme 2 (ACE2), angiotensin (Ang)-(1-7), and Mas. Transgenic overexpression of the components of this axis in general led to an ameliorated cardiac and vascular damage in disease states and to an improved metabolic profile. Knockout models for ACE2 and Mas, however, show aggravated cardiovascular pathologies and a metabolic syndrome-like state. In particular, the local production of Ang-(1-7) in the vascular wall, in the heart, and in the brain was found to be of high physiological relevance by the use of transgenic animals overexpressing ACE2 or Ang-(1-7) in these tissues.
Collapse
|
28
|
Tsigkos S, Mariz S, Llinares J, Fregonese L, Aarum S, Naumann-Winter F, Frauke NW, Westermark K, Sepodes B. Establishing medical plausibility in the context of orphan medicines designation in the European Union. Orphanet J Rare Dis 2014; 9:175. [PMID: 25475155 PMCID: PMC4264401 DOI: 10.1186/s13023-014-0175-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 10/29/2014] [Indexed: 01/07/2023] Open
Abstract
In the European Union, sponsors have the responsibility to demonstrate the “intention to diagnose, prevent or treat” a serious and rare condition before the Committee of Orphan Medicinal Products (COMP), for a medicinal product to meet the criteria for Orphan Designation. This requirement is commonly referred to as “medical plausibility” and the justification of this intention is assessed on the merits of each application by the COMP, which deliberates over the scientific evaluation of the evidence submitted. The scientific assessment of the applications for orphan designation by the Committee is based on the review of non-clinical (such as in vitro and in vivo) and/or clinical data submitted by the sponsor. Several challenges regarding the evidence provided emerge when the sponsor is applying for a designation at an early stage of development. Herein we discuss specific examples from the experience of the COMP, in order to elaborate on the type and level of evidence generally considered necessary for the purpose of justification of the intention to treat an orphan condition. Importantly, it is pointed out that bridging of data from other products, irrespectively of how comparable they may be, or from settings not directly associated with the condition as applied for designation, is by and large not a successful exercise and may only be exceptionally considered. It is further exemplified that, as reflected in the updated ‘Guideline on the format and context of the applications for designation’ and the guidance document ‘Recommendation on elements required to support the medical plausibility and the assumption of significant benefit for an orphan designation’ available on the EMA website, the sponsor should provide data with the specific product as applied for in specific models of the condition or in patients affected by the same condition subject of each application.
Collapse
Affiliation(s)
- Stelios Tsigkos
- Orphan Medicines Office, European Medicines Agency, 7 Westferry Circus, Canary Wharf, E144HB, London, UK.
| | - Segundo Mariz
- Orphan Medicines Office, European Medicines Agency, 7 Westferry Circus, Canary Wharf, E144HB, London, UK.
| | - Jordi Llinares
- Orphan Medicines Office, European Medicines Agency, 7 Westferry Circus, Canary Wharf, E144HB, London, UK.
| | - Laura Fregonese
- Orphan Medicines Office, European Medicines Agency, 7 Westferry Circus, Canary Wharf, E144HB, London, UK.
| | - Stiina Aarum
- Orphan Medicines Office, European Medicines Agency, 7 Westferry Circus, Canary Wharf, E144HB, London, UK.
| | | | - Naumann-Winter Frauke
- Bundesinstitut für Arzneimittel und Medizinprodukte, Kurt-Georg-Kiesinger-Allee 3, 53175, Bonn, Germany.
| | | | - Bruno Sepodes
- Research Institute for Medicines (iMED.ULisboa), Faculty of Pharmacy, University of Lisbon, 1649-003, Lisboa, Portugal.
| |
Collapse
|
29
|
Wynne GM, Russell AJ. Drug Discovery Approaches for Rare Neuromuscular Diseases. ORPHAN DRUGS AND RARE DISEASES 2014. [DOI: 10.1039/9781782624202-00257] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Rare neuromuscular diseases encompass many diverse and debilitating musculoskeletal disorders, ranging from ultra-orphan conditions that affect only a few families, to the so-called ‘common’ orphan diseases like Duchenne muscular dystrophy (DMD) and spinal muscular atrophy (SMA), which affect several thousand individuals worldwide. Increasingly, pharmaceutical and biotechnology companies, in an effort to improve productivity and rebuild dwindling pipelines, are shifting their business models away from the formerly popular ‘blockbuster’ strategy, with rare diseases being an area of increased focus in recent years. As a consequence of this paradigm shift, coupled with high-profile campaigns by not-for-profit organisations and patient advocacy groups, rare neuromuscular diseases are attracting considerable attention as new therapeutic areas for improved drug therapy. Much pioneering work has taken place to elucidate the underlying pathological mechanisms of many rare neuromuscular diseases. This, in conjunction with the availability of new screening technologies, has inspired the development of several truly innovative therapeutic strategies aimed at correcting the underlying pathology. A survey of medicinal chemistry approaches and the resulting clinical progress for new therapeutic agents targeting this devastating class of degenerative diseases is presented, using DMD and SMA as examples. Complementary strategies using small-molecule drugs and biological agents are included.
Collapse
Affiliation(s)
- Graham M. Wynne
- Chemistry Research Laboratory, University of Oxford 12 Mansfield Road Oxford OX1 3TA UK
| | - Angela J. Russell
- Chemistry Research Laboratory, University of Oxford 12 Mansfield Road Oxford OX1 3TA UK
| |
Collapse
|
30
|
Glucocerebrosidase inhibitors: future drugs for the treatment of Gaucher disease? Future Med Chem 2014; 6:975-8. [DOI: 10.4155/fmc.14.41] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
|
31
|
Sondhi D, Rosenberg JB, Van de Graaf BG, Kaminsky SM, Crystal RG. Advances in the treatment of neuronal ceroid lipofuscinosis. Expert Opin Orphan Drugs 2013. [DOI: 10.1517/21678707.2013.852081] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
32
|
Pantoja M, Ruohola-Baker H. Drosophila as a starting point for developing therapeutics for the rare disease Duchenne Muscular Dystrophy. ACTA ACUST UNITED AC 2013; 1:e24995. [PMID: 25002997 PMCID: PMC3932943 DOI: 10.4161/rdis.24995] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Accepted: 05/09/2013] [Indexed: 11/19/2022]
Abstract
Progress into developing therapeutics for rare diseases can be accelerated for those diseases that can be modeled in genetically tractable organisms. Here we comment on one disease, Duchenne Muscular Dystrophy (DMD), modeled in Drosophila that brought together disparate lines of research toward the goal of developing a therapeutic. Though the bioactive lipid sphingosine 1-phosphate (S1P) has been implicated in many anabolic processes in many cell types and tissues, including muscle, this work confirmed the therapeutic potential of assessing this pathway for DMD. Genetic dissection of sphingolipid metabolism showed the suppression of muscle structural and functional defects in flies. Moreover, improvement of muscle defects using known pharmacological agents that raise S1P levels in vivo highlight the potential of Drosophila as a drug-screening tool for DMD. We and others have extended S1P studies into the mouse model of DMD and have shown a partial amelioration of symptoms associated with DMD. Translation of this work to mammals makes the sphingolipid metabolism pathway a promising target for further drug development that may benefit the human condition.
Collapse
Affiliation(s)
- Mario Pantoja
- Department of Biochemistry; Institute for Stem Cell and Regenerative Medicine; University of Washington; Seattle, WA USA
| | - Hannele Ruohola-Baker
- Department of Biochemistry; Institute for Stem Cell and Regenerative Medicine; University of Washington; Seattle, WA USA
| |
Collapse
|