1
|
[Disease-modifying treatment approaches in Huntington disease : Past and future]. DER NERVENARZT 2021; 93:179-190. [PMID: 34762178 PMCID: PMC8825394 DOI: 10.1007/s00115-021-01224-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 10/06/2021] [Indexed: 11/18/2022]
Abstract
Die Huntington-Krankheit (HK) ist die häufigste monogenetische neurodegenerative Erkrankung und kann bereits im präklinischen Stadium zweifelsfrei diagnostiziert werden, zumindest in allen Fällen, bei denen die CAG-Expansionsmutation im Huntingtin-Gen (HTT) im Bereich der vollen Penetranz liegt. Wichtige Voraussetzungen für eine früh im Krankheitsprozess einsetzende und deshalb den weiteren Verlauf der Krankheit in klinisch relevanter Weise modifizierende Therapie sind damit gegeben und machen die HK zu einer Modellerkrankung für neuroprotektive Behandlungsansätze. In der Vergangenheit lag der Schwerpunkt auf dem Ausgleich vermuteter Neurotransmitterdefizite (GABA) analog zur Parkinson-Erkrankung und auf klassischen neuroprotektiven Strategien zur Beeinflussung hypothetischer gemeinsamer Endstrecken neurodegenerativer Erkrankungen (z. B. Exzitotoxizität, mitochondriale Dysfunktion, oxidativer Stress etc.). Mit der Entdeckung der krankheitsverursachenden HTT-Mutation im Jahr 1993 fokussierte sich die Therapieforschung zunehmend darauf, soweit proximal wie möglich in die pathophysiologische Ereigniskette einzugreifen. Ein wichtiger Ansatzpunkt ist hier die HTT-mRNA mit dem Ziel, die Nachproduktion mutierter Huntingtin-Genprodukte zu senken und damit den Körper von deren schädigenden Auswirkungen zu entlasten; zu diesem Zweck sind verschiedene Behandlungsmodalitäten (einzelsträngige DNA und RNA, divalente RNA und Zinkfinger-Repressorkomplexe, oral verfügbare Spleißmodulatoren) entwickelt worden, die sich in der klinischen Prüfung (Phase I–III) oder in späten Stadien der präklinischen Entwicklung befinden. Zudem zeichnet sich ab, dass es möglich sein könnte, die Länge der somatisch instabilen, d. h. über die Lebenszeit v. a. im Hirngewebe zunehmende CAG-Mutation selbst zu beeinflussen und die Progression der HK hierdurch zu bremsen.
Collapse
|
2
|
Wang X, Ma C, Rodríguez Labrada R, Qin Z, Xu T, He Z, Wei Y. Recent advances in lentiviral vectors for gene therapy. SCIENCE CHINA-LIFE SCIENCES 2021; 64:1842-1857. [PMID: 34708326 DOI: 10.1007/s11427-021-1952-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/19/2021] [Indexed: 02/05/2023]
Abstract
Lentiviral vectors (LVs), derived from human immunodeficiency virus, are powerful tools for modifying the genes of eukaryotic cells such as hematopoietic stem cells and neural cells. With the extensive and in-depth studies on this gene therapy vehicle over the past two decades, LVs have been widely used in both research and clinical trials. For instance, third-generation and self-inactive LVs have been used to introduce a gene with therapeutic potential into the host genome and achieve targeted delivery into specific tissue. When LVs are employed in leukemia, the transduced T cells recognize and kill the tumor B cells; in β-thalassemia, the transduced CD34+ cells express normal β-globin; in adenosine deaminase-deficient severe combined immunodeficiency, the autologous CD34+ cells express adenosine deaminase and realize immune reconstitution. Overall, LVs can perform significant roles in the treatment of primary immunodeficiency diseases, hemoglobinopathies, B cell leukemia, and neurodegenerative diseases. In this review, we discuss the recent developments and therapeutic applications of LVs. The safe and efficient LVs show great promise as a tool for human gene therapy.
Collapse
Affiliation(s)
- Xiaoyu Wang
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Cuicui Ma
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Roberto Rodríguez Labrada
- Department Clinical Neurophysiology, Centre for the Research and Rehabilitation of Hereditary Ataxias, Holguín, 80100, Cuba
| | - Zhou Qin
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ting Xu
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Zhiyao He
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China.
| | - Yuquan Wei
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
3
|
Liu P, Huang X, Wu H, Yin G, Shen L. LncRNA-H19 gene plays a significant role in regulating glioma cell function. Mol Genet Genomic Med 2021; 9:e1480. [PMID: 34477331 PMCID: PMC8580082 DOI: 10.1002/mgg3.1480] [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: 02/12/2020] [Revised: 06/08/2020] [Accepted: 08/03/2020] [Indexed: 12/15/2022] Open
Abstract
Background Glioma is an aggressive adult primary cancer, and is characterized by low cure rate, poor prognosis, and high recurrence. The present study aimed to investigate the effect of lncRNA‐H19 gene silencing on glioma cell function. Methods lncRNA‐H19 interference vector (LV3‐si‐H19) and negative control vector (LV3‐NC) were stably transfected into U251 and U87‐MG cells, respectively. Quantitative real‐time PCR (qRT‐PCR) was performed to investigate the expression of lncRNA‐H19. Cell proliferation capacity was tested by adopting cell counting kit (CCK8), and propidium iodide (PI) was used for cell cycle analysis. Meanwhile, flow cytometry (FCM) method was used to investigate cell apoptosis, cell migration capacity was detected via wound healing and transwell experiments, and sphere‐forming ability was examined in serum‐free suspension culture. Additionally, glioma animal models were conducted through injecting U251 cells to estimate the effects of lncRNA‐H19 on glioma growth in vivo. Results Knocking down lncRNA‐H19 gene could effectively suppress the proliferation of U251 and U87‐MG cells. The knockdown of lncRNA‐H19 remarkably inhibited the migration and blocked cycle progressions of U251 and U87‐MG cells, yet, no obvious changes were observed in cell apoptosis. Besides, inhibiting lncRNA‐H19 expression could attenuate sphere‐forming function of U251 and U87‐MG cells. Additionally, tumor volume and weight were significantly reduced in rats injected with U251 LV‐si‐H19 cell line compared to untransfected and negative controls, when survival time was obviously prolonged in U251 LV‐si‐H19 injection groups. Conclusion LncRNA‐H19 gene plays a carcinogenic role in glioma progression via enhancing aggressive behavior of glioma cells.
Collapse
Affiliation(s)
- Ping Liu
- Department of Oncology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, China
| | - Xinqiong Huang
- Department of Oncology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, China
| | - Haijun Wu
- Department of Oncology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, China
| | - Guoling Yin
- Department of Oncology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, China
| | - Liangfang Shen
- Department of Oncology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, China
| |
Collapse
|
4
|
Mühlbäck A, Lindenberg KS, Saft C, Priller J, Landwehrmeyer GB. [Gene-selective treatment approaches for Huntington's disease]. DER NERVENARZT 2020; 91:303-311. [PMID: 32179957 DOI: 10.1007/s00115-020-00882-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In Germany at least 8000 and probably up to ca. 14,000 people currently suffer from clinically manifest Huntington's disease (HD). In addition, an estimated 24,000 Germans carry the HD mutation in the huntingtin (HTT) gene and will develop HD during their lifetime. Although HD is a rare neurodegenerative disease, it is currently in the focus of general medical interest: clinical trials have begun that provide a rational basis for hope to slow down the so far relentless progression of the disease, ultimately resulting in patients becoming entirely dependent on nursing care. If treatment is started early enough it may be possible to mitigate the clinical manifestation of HD. These innovative therapeutic approaches aim at inhibiting the de novo production of mutant HTT gene products. A first clinical drug trial to demonstrate the efficacy (phase III) of intrathecal antisense oligonucleotides (ASO, active substance RG6042) was started in 2019. Additional clinical studies on alternative treatment approaches with allele-selective ASOs as well as gene therapeutic approaches using RNA molecules and zinc finger repressor complexes are imminent. This article gives an overview of the current gene-selective therapeutic approaches in HD under discussion.
Collapse
Affiliation(s)
- A Mühlbäck
- Abteilung Neurologie, Universitätsklinikum Ulm, Oberer Eselsberg 45/1, 89081, Ulm, Deutschland.,Klinik für Neurologie und Zentrum für klinische Neurowissenschaften, 1. Medizinische Fakultät, Karlsuniversität, Prag, Tschechien
| | - K S Lindenberg
- Abteilung Neurologie, Universitätsklinikum Ulm, Oberer Eselsberg 45/1, 89081, Ulm, Deutschland
| | - C Saft
- Huntington-Zentrum NRW, Neurologische Klinik der Ruhr-Universität Bochum, St. Josef-Hospital, Bochum, Deutschland
| | - J Priller
- Klinik für Psychiatrie und Psychotherapie, Charité Universitätsmedizin Berlin, Berlin, Deutschland
| | - G B Landwehrmeyer
- Abteilung Neurologie, Universitätsklinikum Ulm, Oberer Eselsberg 45/1, 89081, Ulm, Deutschland.
| |
Collapse
|
5
|
Maximizing lentiviral vector gene transfer in the CNS. Gene Ther 2020; 28:75-88. [PMID: 32632267 PMCID: PMC7902268 DOI: 10.1038/s41434-020-0172-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 05/20/2020] [Accepted: 06/25/2020] [Indexed: 12/19/2022]
Abstract
Gene transfer is a widely developed technique for studying and treating genetic diseases. However, the development of therapeutic strategies is challenging, due to the cellular and functional complexity of the central nervous system (CNS), its large size and restricted access. We explored two parameters for improving gene transfer efficacy and capacity for the selective targeting of subpopulations of cells with lentiviral vectors (LVs). We first developed a second-generation LV specifically targeting astrocytes for the efficient expression or silencing of genes of interest, and to better study the importance of cell subpopulations in neurological disorders. We then made use of the retrograde transport properties of a chimeric envelope to target brain circuits affected in CNS diseases and achieve a broad distribution. The combination of retrograde transport and specific tropism displayed by this LV provides opportunities for delivering therapeutic genes to specific cell populations and ensuring high levels of transduction in interconnected brain areas following local administration. This new LV and delivery strategy should be of greater therapeutic benefit and opens up new possibilities for the preclinical development of gene therapy for neurodegenerative diseases.
Collapse
|
6
|
Gonzalez-Alegre P. Recent advances in molecular therapies for neurological disease: triplet repeat disorders. Hum Mol Genet 2020; 28:R80-R87. [PMID: 31227833 DOI: 10.1093/hmg/ddz138] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 04/03/2019] [Accepted: 06/11/2019] [Indexed: 02/06/2023] Open
Abstract
Triplet repeat diseases (TRDs) are caused by pathogenic expansions of trinucleotide sequence repeats within coding and non-coding regions of different genes. They are typically progressive, very disabling and frequently involve the nervous system. Currently available symptomatic therapies provide modest benefit at best. The development of interventions that interfere with the natural history of these diseases is a priority. A common pathogenic process shared by most TRDs is the presence of toxicity from the messenger RNA or protein encoded by the gene harboring the abnormal expansion. Strategies to interfere with the expression of these genes using different molecular approaches are being pursued and have reached the clinical stage. This review will summarize the significant progress made in this field in the last few years, focusing on three main areas: the discovery of biomarkers of disease progression and target engagement, advances in preclinical studies for the polyglutamine ataxias and the initial clinical application in myotonic dystrophy type 1 and Huntington's disease.
Collapse
Affiliation(s)
- Pedro Gonzalez-Alegre
- Department of Neurology, the University of Pennsylvania, Philadelphia, PA 19104, USA.,Raymond G. Perelman Center for Cellular and Molecular Therapy, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| |
Collapse
|
7
|
Testa CM, Jankovic J. Huntington disease: A quarter century of progress since the gene discovery. J Neurol Sci 2019; 396:52-68. [DOI: 10.1016/j.jns.2018.09.022] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 09/14/2018] [Accepted: 09/18/2018] [Indexed: 01/21/2023]
|
8
|
Abstract
The 25 years since the identification of the gene responsible for Huntington disease (HD) have stood witness to profound discoveries about the nature of the disease and its pathogenesis. Despite this progress, however, the development of disease-modifying therapies has thus far been slow. Preclinical validation of the therapeutic potential of disrupted pathways in HD has led to the advancement of pharmacological agents, both novel and repurposed, for clinical evaluation. The most promising therapeutic approaches include huntingtin (HTT) lowering and modification as well as modulation of neuroinflammation and synaptic transmission. With clinical trials for many of these approaches imminent or currently ongoing, the coming years are promising not only for HD but also for more prevalent neurodegenerative disorders, such as Alzheimer and Parkinson disease, in which many of these pathways have been similarly implicated.
Collapse
|
9
|
Evers MM, Miniarikova J, Juhas S, Vallès A, Bohuslavova B, Juhasova J, Skalnikova HK, Vodicka P, Valekova I, Brouwers C, Blits B, Lubelski J, Kovarova H, Ellederova Z, van Deventer SJ, Petry H, Motlik J, Konstantinova P. AAV5-miHTT Gene Therapy Demonstrates Broad Distribution and Strong Human Mutant Huntingtin Lowering in a Huntington's Disease Minipig Model. Mol Ther 2018; 26:2163-2177. [PMID: 30007561 PMCID: PMC6127509 DOI: 10.1016/j.ymthe.2018.06.021] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 06/18/2018] [Accepted: 06/20/2018] [Indexed: 02/07/2023] Open
Abstract
Huntington’s disease (HD) is a fatal neurodegenerative disorder caused by a CAG trinucleotide repeat expansion in the huntingtin gene. Previously, we showed strong huntingtin reduction and prevention of neuronal dysfunction in HD rodents using an engineered microRNA targeting human huntingtin, delivered via adeno-associated virus (AAV) serotype 5 vector with a transgene encoding an engineered miRNA against HTT mRNA (AAV5-miHTT). One of the challenges of rodents as a model of neurodegenerative diseases is their relatively small brain, making successful translation to the HD patient difficult. This is particularly relevant for gene therapy approaches, where distribution achieved upon local administration into the parenchyma is likely dependent on brain size and structure. Here, we aimed to demonstrate the translation of huntingtin-lowering gene therapy to a large-animal brain. We investigated the feasibility, efficacy, and tolerability of one-time intracranial administration of AAV5-miHTT in the transgenic HD (tgHD) minipig model. We detected widespread dose-dependent distribution of AAV5-miHTT throughout the tgHD minipig brain that correlated with the engineered microRNA expression. Both human mutant huntingtin mRNA and protein were significantly reduced in all brain regions transduced by AAV5-miHTT. The combination of widespread vector distribution and extensive huntingtin lowering observed with AAV5-miHTT supports the translation of a huntingtin-lowering gene therapy for HD from preclinical studies into the clinic.
Collapse
Affiliation(s)
- Melvin M Evers
- Department of Research & Development, uniQure biopharma B.V., Amsterdam, the Netherlands.
| | - Jana Miniarikova
- Department of Research & Development, uniQure biopharma B.V., Amsterdam, the Netherlands
| | - Stefan Juhas
- Institute of Animal Physiology and Genetics, Libechov, Czech Republic
| | - Astrid Vallès
- Department of Research & Development, uniQure biopharma B.V., Amsterdam, the Netherlands
| | | | - Jana Juhasova
- Institute of Animal Physiology and Genetics, Libechov, Czech Republic
| | | | - Petr Vodicka
- Institute of Animal Physiology and Genetics, Libechov, Czech Republic
| | - Ivona Valekova
- Institute of Animal Physiology and Genetics, Libechov, Czech Republic
| | - Cynthia Brouwers
- Department of Research & Development, uniQure biopharma B.V., Amsterdam, the Netherlands
| | - Bas Blits
- Department of Research & Development, uniQure biopharma B.V., Amsterdam, the Netherlands
| | - Jacek Lubelski
- Department of Research & Development, uniQure biopharma B.V., Amsterdam, the Netherlands
| | - Hana Kovarova
- Institute of Animal Physiology and Genetics, Libechov, Czech Republic
| | - Zdenka Ellederova
- Institute of Animal Physiology and Genetics, Libechov, Czech Republic
| | - Sander J van Deventer
- Department of Research & Development, uniQure biopharma B.V., Amsterdam, the Netherlands
| | - Harald Petry
- Department of Research & Development, uniQure biopharma B.V., Amsterdam, the Netherlands
| | - Jan Motlik
- Institute of Animal Physiology and Genetics, Libechov, Czech Republic
| | - Pavlina Konstantinova
- Department of Research & Development, uniQure biopharma B.V., Amsterdam, the Netherlands
| |
Collapse
|
10
|
Pfister EL, DiNardo N, Mondo E, Borel F, Conroy F, Fraser C, Gernoux G, Han X, Hu D, Johnson E, Kennington L, Liu P, Reid SJ, Sapp E, Vodicka P, Kuchel T, Morton AJ, Howland D, Moser R, Sena-Esteves M, Gao G, Mueller C, DiFiglia M, Aronin N. Artificial miRNAs Reduce Human Mutant Huntingtin Throughout the Striatum in a Transgenic Sheep Model of Huntington's Disease. Hum Gene Ther 2018; 29:663-673. [PMID: 29207890 DOI: 10.1089/hum.2017.199] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Huntington's disease (HD) is a fatal neurodegenerative disease caused by a genetic expansion of the CAG repeat region in the huntingtin (HTT) gene. Studies in HD mouse models have shown that artificial miRNAs can reduce mutant HTT, but evidence for their effectiveness and safety in larger animals is lacking. HD transgenic sheep express the full-length human HTT with 73 CAG repeats. AAV9 was used to deliver unilaterally to HD sheep striatum an artificial miRNA targeting exon 48 of the human HTT mRNA under control of two alternative promoters: U6 or CβA. The treatment reduced human mutant (m) HTT mRNA and protein 50-80% in the striatum at 1 and 6 months post injection. Silencing was detectable in both the caudate and putamen. Levels of endogenous sheep HTT protein were not affected. There was no significant loss of neurons labeled by DARPP32 or NeuN at 6 months after treatment, and Iba1-positive microglia were detected at control levels. It is concluded that safe and effective silencing of human mHTT protein can be achieved and sustained in a large-animal brain by direct delivery of an AAV carrying an artificial miRNA.
Collapse
Affiliation(s)
- Edith L Pfister
- 1 Department of Medicine, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Natalie DiNardo
- 1 Department of Medicine, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Erica Mondo
- 2 Department of Neurobiology, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Florie Borel
- 3 Horae Gene Therapy Center, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Faith Conroy
- 1 Department of Medicine, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Cara Fraser
- 4 Preclinical, Imaging, and Research Laboratories, South Australian Health and Medical Research Institute , Gilles Plains, South Australia
| | - Gwladys Gernoux
- 3 Horae Gene Therapy Center, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Xin Han
- 5 West China School of Medicine, West China Hospital, Sichuan University. Chengdu , China
| | - Danjing Hu
- 5 West China School of Medicine, West China Hospital, Sichuan University. Chengdu , China
| | - Emily Johnson
- 1 Department of Medicine, University of Massachusetts Medical School , Worcester, Massachusetts.,6 Geisel School of Medicine, Dartmouth College , Hanover, New Hampshire
| | - Lori Kennington
- 1 Department of Medicine, University of Massachusetts Medical School , Worcester, Massachusetts
| | - PengPeng Liu
- 5 West China School of Medicine, West China Hospital, Sichuan University. Chengdu , China
| | - Suzanne J Reid
- 7 School of Biological Sciences, University of Auckland , Auckland, New Zealand
| | - Ellen Sapp
- 8 MassGeneral Institute for Neurodegenerative Disease , Charlestown, Massachusetts
| | - Petr Vodicka
- 8 MassGeneral Institute for Neurodegenerative Disease , Charlestown, Massachusetts.,9 Research Center PIGMOD, Institute of Animal Physiology and Genetics of the Czech Academy of Sciences , Libechov, Czech Republic
| | - Tim Kuchel
- 4 Preclinical, Imaging, and Research Laboratories, South Australian Health and Medical Research Institute , Gilles Plains, South Australia
| | - A Jennifer Morton
- 10 Department of Physiology, Development and Neuroscience, University of Cambridge , Cambridge, United Kingdom
| | - David Howland
- 11 CHDI Foundation/CHDI Management, Princeton, New Jersey
| | - Richard Moser
- 12 Department of Neurosurgery, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Miguel Sena-Esteves
- 3 Horae Gene Therapy Center, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Guangping Gao
- 3 Horae Gene Therapy Center, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Christian Mueller
- 3 Horae Gene Therapy Center, University of Massachusetts Medical School , Worcester, Massachusetts.,13 Department of Pediatrics, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Marian DiFiglia
- 8 MassGeneral Institute for Neurodegenerative Disease , Charlestown, Massachusetts
| | - Neil Aronin
- 1 Department of Medicine, University of Massachusetts Medical School , Worcester, Massachusetts.,14 RNA Therapeutics Institute, University of Massachusetts Medical School , Worcester, Massachusetts
| |
Collapse
|
11
|
Miniarikova J, Evers MM, Konstantinova P. Translation of MicroRNA-Based Huntingtin-Lowering Therapies from Preclinical Studies to the Clinic. Mol Ther 2018; 26:947-962. [PMID: 29503201 DOI: 10.1016/j.ymthe.2018.02.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 01/30/2018] [Accepted: 02/05/2018] [Indexed: 12/21/2022] Open
Abstract
The single mutation underlying the fatal neuropathology of Huntington's disease (HD) is a CAG triplet expansion in exon 1 of the huntingtin (HTT) gene, which gives rise to a toxic mutant HTT protein. There have been a number of not yet successful therapeutic advances in the treatment of HD. The current excitement in the HD field is due to the recent development of therapies targeting the culprit of HD either at the DNA or RNA level to reduce the overall mutant HTT protein. In this review, we briefly describe short-term and long-term HTT-lowering strategies targeting HTT transcripts. One of the most advanced HTT-lowering strategies is a microRNA (miRNA)-based gene therapy delivered by a single administration of an adeno-associated viral (AAV) vector to the HD patient. We outline the outcome measures for the miRNA-based HTT-lowering therapy in the context of preclinical evaluation in HD animal and cell models. We highlight the strengths and ongoing queries of the HTT-lowering gene therapy as an HD intervention with a potential disease-modifying effect. This review provides a perspective on the fast-developing HTT-lowering therapies for HD and their translation to the clinic based on existing knowledge in preclinical models.
Collapse
Affiliation(s)
- Jana Miniarikova
- Department of Research and Development, uniQure, Amsterdam, the Netherlands; Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, the Netherlands
| | - Melvin M Evers
- Department of Research and Development, uniQure, Amsterdam, the Netherlands
| | | |
Collapse
|
12
|
Merienne N, Vachey G, de Longprez L, Meunier C, Zimmer V, Perriard G, Canales M, Mathias A, Herrgott L, Beltraminelli T, Maulet A, Dequesne T, Pythoud C, Rey M, Pellerin L, Brouillet E, Perrier AL, du Pasquier R, Déglon N. The Self-Inactivating KamiCas9 System for the Editing of CNS Disease Genes. Cell Rep 2017; 20:2980-2991. [DOI: 10.1016/j.celrep.2017.08.075] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 07/14/2017] [Accepted: 08/23/2017] [Indexed: 10/18/2022] Open
|