Widespread and sustained target engagement in Huntington’s disease minipigs upon intrastriatal microRNA-based gene therapy

Striatal delivery of microRNA-gene therapy results in widespread brain huntingtin protein lowering in Huntington’s disease minipigs up to 1 year. Targeting HTT in pigs Huntington’s disease (HD) is a genetic neurodegenerative disorder caused by mutated huntingtin (HTT) gene. Reducing the expression of the aberrant HTT has been shown to be effective in preclinical models. Now, Vallès et al. evaluated the effects of an adeno-associated viral vector (AAV)–mediated strategy delivering microRNA (miRNA) targeting human mutant HTT (mHTT) in a pig model of HD that closely resembles the human condition. Intracerebral delivery of the miRNA into the striatum resulted in widespread distribution and reduced mHTT for up to a year after injection. The results suggest that the approach could be effective in patients with HD. Huntingtin (HTT)–lowering therapies hold promise to slow down neurodegeneration in Huntington’s disease (HD). Here, we assessed the translatability and long-term durability of recombinant adeno-associated viral vector serotype 5 expressing a microRNA targeting human HTT (rAAV5-miHTT) administered by magnetic resonance imaging–guided convention-enhanced delivery in transgenic HD minipigs. rAAV5-miHTT (1.2 × 1013 vector genome (VG) copies per brain) was successfully administered into the striatum (bilaterally in caudate and putamen), using age-matched untreated animals as controls. Widespread brain biodistribution of vector DNA was observed, with the highest concentration in target (striatal) regions, thalamus, and cortical regions. Vector DNA presence and transgene expression were similar at 6 and 12 months after administration. Expression of miHTT strongly correlated with vector DNA, with a corresponding reduction of mutant HTT (mHTT) protein of more than 75% in injected areas, and 30 to 50% lowering in distal regions. Translational pharmacokinetic and pharmacodynamic measures in cerebrospinal fluid (CSF) were largely in line with the effects observed in the brain. CSF miHTT expression was detected up to 12 months, with CSF mHTT protein lowering of 25 to 30% at 6 and 12 months after dosing. This study demonstrates widespread biodistribution, strong and durable efficiency of rAAV5-miHTT in disease-relevant regions in a large brain, and the potential of using CSF analysis to determine vector expression and efficacy in the clinic.

[1]  C. Olanow,et al.  Long-term post-mortem studies following neurturin gene therapy in patients with advanced Parkinson’s disease , 2020, Brain : a journal of neurology.

[2]  Daniel G. Anderson,et al.  Reduction of the therapeutic dose of silencing RNA by packaging it in extracellular vesicles via a pre-microRNA backbone , 2020, Nature Biomedical Engineering.

[3]  J. Kleinjans,et al.  Circulating microRNAs as potential biomarkers for psychiatric and neurodegenerative disorders , 2019, Progress in Neurobiology.

[4]  M. Hayden,et al.  Potent and sustained huntingtin lowering via AAV5 encoding miRNA preserves striatal volume and cognitive function in a humanized mouse model of Huntington disease , 2019, Nucleic acids research.

[5]  J. Carette,et al.  GPR108 Is a Highly Conserved AAV Entry Factor. , 2019, Molecular therapy : the journal of the American Society of Gene Therapy.

[6]  J. Klempír,et al.  Longitudinal study revealing motor, cognitive and behavioral decline in a transgenic minipig model of Huntington's disease , 2019, Disease Models & Mechanisms.

[7]  S. V. van Deventer,et al.  AAV5-miHTT Lowers Huntingtin mRNA and Protein without Off-Target Effects in Patient-Derived Neuronal Cultures and Astrocytes , 2019, Molecular therapy. Methods & clinical development.

[8]  L. Shihabuddin,et al.  Astrocyte transduction is required for rescue of behavioral phenotypes in the YAC128 mouse model with AAV-RNAi mediated HTT lowering therapeutics , 2019, Neurobiology of Disease.

[9]  M. DiFiglia,et al.  A divalent siRNA chemical scaffold for potent and sustained modulation of gene expression throughout the central nervous system , 2019, Nature Biotechnology.

[10]  Michele Simonato,et al.  Gene Therapy Tools for Brain Diseases , 2019, Front. Pharmacol..

[11]  J. Zeman,et al.  Deterioration of mitochondrial bioenergetics and ultrastructure impairment in skeletal muscle of a transgenic minipig model in the early stages of Huntington's disease , 2019, Disease Models & Mechanisms.

[12]  K. Blennow,et al.  NFL is a marker of treatment response in children with SMA treated with nusinersen , 2019, Journal of Neurology.

[13]  D. Surmeier,et al.  Allele-selective transcriptional repression of mutant HTT for the treatment of Huntington’s disease , 2019, Nature Medicine.

[14]  S. Tabrizi,et al.  Huntingtin Lowering Strategies for Disease Modification in Huntington’s Disease , 2019, Neuron.

[15]  H. Petry,et al.  AAV5-miHTT Gene Therapy Demonstrates Sustained Huntingtin Lowering and Functional Improvement in Huntington Disease Mouse Models , 2019, Molecular therapy. Methods & clinical development.

[16]  L. Kappos,et al.  Blood neurofilament light chain as a biomarker of MS disease activity and treatment response , 2019, Neurology.

[17]  M. Sedláčková,et al.  Transgenic minipig model of Huntington's disease exhibiting gradually progressing neurodegeneration , 2019, Disease Models & Mechanisms.

[18]  D. Alexander,et al.  Evaluation of mutant huntingtin and neurofilament proteins as potential markers in Huntington’s disease , 2018, Science Translational Medicine.

[19]  B. Ravina,et al.  Gene therapy for neurological disorders: progress and prospects , 2018, Nature Reviews Drug Discovery.

[20]  H. Petry,et al.  AAV5-miHTT Gene Therapy Demonstrates Broad Distribution and Strong Human Mutant Huntingtin Lowering in a Huntington’s Disease Minipig Model , 2018, Molecular therapy : the journal of the American Society of Gene Therapy.

[21]  J. Motlík,et al.  Gradual Phenotype Development in Huntington Disease Transgenic Minipig Model at 24 Months of Age , 2018, Neurodegenerative Diseases.

[22]  P. Konstantinova,et al.  Translation of MicroRNA-Based Huntingtin-Lowering Therapies from Preclinical Studies to the Clinic. , 2018, Molecular therapy : the journal of the American Society of Gene Therapy.

[23]  P. McColgan,et al.  Huntington's disease: a clinical review , 2018, European journal of neurology.

[24]  A. Morton,et al.  Artificial miRNAs Reduce Human Mutant Huntingtin Throughout the Striatum in a Transgenic Sheep Model of Huntington's Disease. , 2017, Human gene therapy.

[25]  B. Leavitt,et al.  Validation of Ultrasensitive Mutant Huntingtin Detection in Human Cerebrospinal Fluid by Single Molecule Counting Immunoassay , 2017, Journal of Huntington's disease.

[26]  N. Déglon,et al.  AAV5-miHTT gene therapy demonstrates suppression of mutant huntingtin aggregation and neuronal dysfunction in a rat model of Huntington’s disease , 2017, Gene Therapy.

[27]  H. Petry,et al.  Successful Repeated Hepatic Gene Delivery in Mice and Non-human Primates Achieved by Sequential Administration of AAV5ch and AAV1. , 2017, Molecular therapy : the journal of the American Society of Gene Therapy.

[28]  K. Blennow,et al.  Neurofilament light protein in blood as a potential biomarker of neurodegeneration in Huntington's disease: a retrospective cohort analysis , 2017, The Lancet Neurology.

[29]  P. Dietrich,et al.  Elimination of huntingtin in the adult mouse leads to progressive behavioral deficits, bilateral thalamic calcification, and altered brain iron homeostasis , 2017, PLoS genetics.

[30]  P. Grandi,et al.  Viral vectors for therapy of neurologic diseases , 2017, Neuropharmacology.

[31]  J. Zeman,et al.  Mitochondrial Metabolism in a Large-Animal Model of Huntington Disease: The Hunt for Biomarkers in the Spermatozoa of Presymptomatic Minipigs , 2017, Neurodegenerative Diseases.

[32]  C. Mueller,et al.  Safe and Efficient Silencing with a Pol II, but Not a Pol lII, Promoter Expressing an Artificial miRNA Targeting Human Huntingtin , 2017, Molecular therapy. Nucleic acids.

[33]  P. Dayalu,et al.  Huntington’s Disease—Update on Treatments , 2017, Current Neurology and Neuroscience Reports.

[34]  H. Petry,et al.  MR-guided parenchymal delivery of adeno-associated viral vector serotype 5 in non-human primate brain , 2017, Gene Therapy.

[35]  J. McBride,et al.  Gene suppression strategies for dominantly inherited neurodegenerative diseases: lessons from Huntington's disease and spinocerebellar ataxia. , 2016, Human molecular genetics.

[36]  S. Humbert,et al.  The Biology of Huntingtin , 2016, Neuron.

[37]  M. Hayden,et al.  Design, Characterization, and Lead Selection of Therapeutic miRNAs Targeting Huntingtin for Development of Gene Therapy for Huntington's Disease , 2016, Molecular therapy. Nucleic acids.

[38]  L. Arckens,et al.  Evaluation of the expression pattern of rAAV2/1, 2/5, 2/7, 2/8, and 2/9 serotypes with different promoters in the mouse visual cortex , 2015, The Journal of comparative neurology.

[39]  M. Hayden,et al.  Ultrasensitive measurement of huntingtin protein in cerebrospinal fluid demonstrates increase with Huntington disease stage and decrease following brain huntingtin suppression , 2015, Scientific Reports.

[40]  Zeger Debyser,et al.  Serotype-dependent transduction efficiencies of recombinant adeno-associated viral vectors in monkey neocortex , 2015, Neurophotonics.

[41]  S. Tabrizi,et al.  Quantification of mutant huntingtin protein in cerebrospinal fluid from Huntington's disease patients. , 2015, The Journal of clinical investigation.

[42]  L. Kappos,et al.  Fingolimod and CSF neurofilament light chain levels in relapsing-remitting multiple sclerosis , 2015, Neurology.

[43]  Do P. M. Tromp,et al.  Titer and Product Affect the Distribution of Gene Expression after Intraputaminal Convection-Enhanced Delivery , 2014, Stereotactic and Functional Neurosurgery.

[44]  Jane S. Paulsen,et al.  Huntington disease: natural history, biomarkers and prospects for therapeutics , 2014, Nature Reviews Neurology.

[45]  T. Taksir,et al.  Silencing mutant huntingtin by adeno-associated virus-mediated RNA interference ameliorates disease manifestations in the YAC128 mouse model of Huntington's disease. , 2014, Human gene therapy.

[46]  S. Gill,et al.  Convection-enhanced delivery of AAV2 in white matter—A novel method for gene delivery to cerebral cortex , 2013, Journal of Neuroscience Methods.

[47]  A. O’Connor,et al.  Selective extracellular vesicle-mediated export of an overlapping set of microRNAs from multiple cell types , 2012, BMC Genomics.

[48]  L. Shihabuddin,et al.  Sustained Therapeutic Reversal of Huntington's Disease by Transient Repression of Huntingtin Synthesis , 2012, Neuron.

[49]  J. Prieto,et al.  Transient and intensive pharmacological immunosuppression fails to improve AAV-based liver gene transfer in non-human primates , 2012, Journal of Translational Medicine.

[50]  P. Starr,et al.  Long-term evaluation of a phase 1 study of AADC gene therapy for Parkinson's disease. , 2012, Human gene therapy.

[51]  P. Nelson,et al.  Six-month partial suppression of Huntingtin is well tolerated in the adult rhesus striatum , 2012, Brain : a journal of neurology.

[52]  B. Davidson,et al.  Preclinical safety of RNAi-mediated HTT suppression in the rhesus macaque as a potential therapy for Huntington's disease. , 2011, Molecular therapy : the journal of the American Society of Gene Therapy.

[53]  H. Zetterberg,et al.  Light subunit of neurofilament triplet protein in the cerebrospinal fluid after subthalamic nucleus stimulation for Parkinson’s disease , 2011, Acta neurologica Scandinavica.

[54]  Matthew T. Kaufman,et al.  An optogenetic toolbox designed for primates , 2011, Nature Neuroscience.

[55]  P. Pivirotto,et al.  Eight years of clinical improvement in MPTP-lesioned primates after gene therapy with AAV2-hAADC. , 2010, Molecular therapy : the journal of the American Society of Gene Therapy.

[56]  M. MacDonald,et al.  An ovine transgenic Huntington's disease model. , 2010, Human molecular genetics.

[57]  Kenneth P Vives,et al.  Comparative transduction efficiency of AAV vector serotypes 1-6 in the substantia nigra and striatum of the primate brain. , 2010, Molecular therapy : the journal of the American Society of Gene Therapy.

[58]  Megan M. Romer,et al.  Levels of the light subunit of neurofilament triplet protein in cerebrospinal fluid in Huntington's disease. , 2009, Parkinsonism & related disorders.

[59]  A. K. Hansen,et al.  The use of pigs in neuroscience: Modeling brain disorders , 2007, Neuroscience & Biobehavioral Reviews.

[60]  R. Price,et al.  Antiretroviral treatment reduces increased CSF neurofilament protein (NFL) in HIV-1 infection , 2007, Neurology.

[61]  Zeger Debyser,et al.  Comparative analysis of adeno-associated viral vector serotypes 1, 2, 5, 7, and 8 in mouse brain. , 2007, Human gene therapy.

[62]  N. Hackett,et al.  AAV2-mediated CLN2 gene transfer to rodent and non-human primate brain results in long-term TPP-I expression compatible with therapy for LINCL , 2005, Gene Therapy.

[63]  P. Reier,et al.  Recombinant AAV viral vectors pseudotyped with viral capsids from serotypes 1, 2, and 5 display differential efficiency and cell tropism after delivery to different regions of the central nervous system. , 2004, Molecular therapy : the journal of the American Society of Gene Therapy.

[64]  R. Kotin,et al.  Insect cells as a factory to produce adeno-associated virus type 2 vectors. , 2002, Human gene therapy.

[65]  I. Martins,et al.  Recombinant adeno-associated virus type 2, 4, and 5 vectors: transduction of variant cell types and regions in the mammalian central nervous system. , 2000, Proceedings of the National Academy of Sciences of the United States of America.

[66]  S. Leff,et al.  Long-term restoration of striatal l-aromatic amino acid decarboxylase activity using recombinant adeno-associated viral vector gene transfer in a rodent model of Parkinson's disease , 1999, Neuroscience.

[67]  P F Morrison,et al.  Convection-enhanced delivery of macromolecules in the brain. , 1994, Proceedings of the National Academy of Sciences of the United States of America.

[68]  G. Fraedrich,et al.  Juvenile recurrent respiratory papillomatosis: Still a mystery disease with difficult management , 2007, Head & neck.

[69]  M. Gray Astrocytes in Huntington's Disease. , 2019, Advances in experimental medicine and biology.

[70]  J. Zeman,et al.  Skeletal muscle in an early manifest transgenic minipig model of Huntington's disease revealed deterioration of mitochondrial bioenergetics and ultrastructure impairment. , 2019, Disease models & mechanisms.

[71]  E. Wild,et al.  Biofluid Biomarkers in Huntington's Disease. , 2018, Methods in molecular biology.

[72]  N. Déglon,et al.  AAV 5-miHTT gene therapy demonstrates suppression of mutant huntingtin aggregation and neuronal dysfunction in a rat model of Huntington ’ s disease , 2017 .

[73]  Eric H Kim,et al.  The Neuropathology of Huntington's Disease. , 2015, Current topics in behavioral neurosciences.

[74]  M. Hayden,et al.  Huntington disease , 2015, Nature Reviews Disease Primers.

[75]  M. DiFiglia,et al.  A transgenic minipig model of Huntington's Disease. , 2013, Journal of Huntington's disease.

[76]  I. Bièche,et al.  Efficient intracerebral delivery of AAV5 vector encoding human ARSA in non-human primate. , 2010, Human molecular genetics.

[77]  C. Johnson Progress and Prospects , 1991 .