Tailored Pig Models for Preclinical Efficacy and Safety Testing of Targeted Therapies

Despite enormous advances in translational biomedical research, there remains a growing demand for improved animal models of human disease. This is particularly true for diseases where rodent models do not reflect the human disease phenotype. Compared to rodents, pig anatomy and physiology are more similar to humans in cardiovascular, immune, respiratory, skeletal muscle, and metabolic systems. Importantly, efficient and precise techniques for genetic engineering of pigs are now available, facilitating the creation of tailored large animal models that mimic human disease mechanisms at the molecular level. In this article, the benefits of genetically engineered pigs for basic and translational research are exemplified by a novel pig model of Duchenne muscular dystrophy and by porcine models of cystic fibrosis. Particular emphasis is given to potential advantages of using these models for efficacy and safety testing of targeted therapies, such as exon skipping and gene editing, for example, using the clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated system. In general, genetically tailored pig models have the potential to bridge the gap between proof-of-concept studies in rodents and clinical trials in patients, thus supporting translational medicine.

[1]  K. Boycott,et al.  Rare-disease genetics in the era of next-generation sequencing: discovery to translation , 2013, Nature Reviews Genetics.

[2]  Eric P. Hoffman,et al.  Dystrophin: The protein product of the duchenne muscular dystrophy locus , 1987, Cell.

[3]  R. Prather,et al.  Meganucleases Revolutionize the Production of Genetically Engineered Pigs for the Study of Human Diseases , 2016, Toxicologic pathology.

[4]  Yifan Dai,et al.  Targeted disruption of the α1,3-galactosyltransferase gene in cloned pigs , 2002, Nature Biotechnology.

[5]  Jeong-Sun Seo,et al.  Targeting efficiency of a-1,3-galactosyl transferase gene in pig fetal fibroblast cells , 2003, Experimental & Molecular Medicine.

[6]  S. Richter,et al.  The ΔF508 Mutation Causes CFTR Misprocessing and Cystic Fibrosis–Like Disease in Pigs , 2011, Science Translational Medicine.

[7]  B. Davidson,et al.  A NOVEL GENE DELIVERY METHOD TRANSDUCES PORCINE PANCREATIC DUCT EPITHELIAL CELLS , 2013, Gene Therapy.

[8]  L. Touqui,et al.  Mouse models of cystic fibrosis: phenotypic analysis and research applications. , 2011, Journal of cystic fibrosis : official journal of the European Cystic Fibrosis Society.

[9]  H. Kim,et al.  A guide to genome engineering with programmable nucleases , 2014, Nature Reviews Genetics.

[10]  Martin J. Aryee,et al.  GUIDE-Seq enables genome-wide profiling of off-target cleavage by CRISPR-Cas nucleases , 2014, Nature Biotechnology.

[11]  Daniel F. Voytas,et al.  Efficient TALEN-mediated gene knockout in livestock , 2012, Proceedings of the National Academy of Sciences.

[12]  David K. Meyerholz,et al.  Disruption of the CFTR Gene Produces a Model of Cystic Fibrosis in Newborn Pigs , 2008, Science.

[13]  Wei Li,et al.  One-step generation of knockout pigs by zygote injection of CRISPR/Cas system , 2014, Cell Research.

[14]  Hong Wang,et al.  Functional disruption of the dystrophin gene in rhesus monkey using CRISPR/Cas9. , 2015, Human molecular genetics.

[15]  D. Sen,et al.  Adeno‐associated virus (AAV) vectors in gene therapy: immune challenges and strategies to circumvent them , 2013, Reviews in medical virology.

[16]  G. Comi,et al.  Ataluren treatment of patients with nonsense mutation dystrophinopathy , 2014, Muscle & nerve.

[17]  Bronwen L. Aken,et al.  Analyses of pig genomes provide insight into porcine demography and evolution , 2012, Nature.

[18]  S. Richter,et al.  Cystic Fibrosis Pigs Develop Lung Disease and Exhibit Defective Bacterial Eradication at Birth , 2010, Science Translational Medicine.

[19]  Kevin A. Rocco,et al.  Efficient intratracheal delivery of airway epithelial cells in mice and pigs. , 2015, American journal of physiology. Lung cellular and molecular physiology.

[20]  Steve D. M. Brown,et al.  The mammalian gene function resource: the international knockout mouse consortium , 2012, Mammalian Genome.

[21]  W. Berdon,et al.  Gastrointestinal manifestations of cystic fibrosis. , 1987, Seminars in roentgenology.

[22]  Mark A. Kay,et al.  Progress and problems with the use of viral vectors for gene therapy , 2003, Nature Reviews Genetics.

[23]  R. Prather,et al.  Gene targeting with zinc finger nucleases to produce cloned eGFP knockout pigs , 2011, Molecular reproduction and development.

[24]  D. Geddes,et al.  Inflammation in cystic fibrosis--when and why? Friend or foe? , 2007, Seminars in Respiratory and Critical Care Medicine.

[25]  A. Nakamura,et al.  Mammalian Models of Duchenne Muscular Dystrophy: Pathological Characteristics and Therapeutic Applications , 2011, Journal of Biomedicine and Biotechnology.

[26]  E. Wolf,et al.  Sequential targeting of CFTR by BAC vectors generates a novel pig model of cystic fibrosis , 2012, Journal of Molecular Medicine.

[27]  M. Nishihara,et al.  Generation of muscular dystrophy model rats with a CRISPR/Cas system , 2014, Scientific Reports.

[28]  George P Patrinos,et al.  Defining the disease liability of variants in the cystic fibrosis transmembrane conductance regulator gene , 2013, Nature Genetics.

[29]  R. Wanke,et al.  Tissue Sampling Guides for Porcine Biomedical Models , 2016, Toxicologic pathology.

[30]  Kenji Nakamura,et al.  Targeted disruption of exon 52 in the mouse dystrophin gene induced muscle degeneration similar to that observed in Duchenne muscular dystrophy. , 1997, Biochemical and biophysical research communications.

[31]  M. Welsh,et al.  Loss of Anion Transport without Increased Sodium Absorption Characterizes Newborn Porcine Cystic Fibrosis Airway Epithelia , 2010, Cell.

[32]  M. Welsh,et al.  Reduced Airway Surface pH Impairs Bacterial Killing in the Porcine Cystic Fibrosis Lung , 2012, Nature.

[33]  M. Welsh,et al.  Production of CFTR-null and CFTR-DeltaF508 heterozygous pigs by adeno-associated virus-mediated gene targeting and somatic cell nuclear transfer. , 2008, The Journal of clinical investigation.

[34]  C. Sheridan,et al.  Gene therapy finds its niche , 2011, Nature Biotechnology.

[35]  J. Vogel,et al.  CRISPR RNA maturation by trans-encoded small RNA and host factor RNase III , 2011, Nature.

[36]  G. McLennan,et al.  Loss of cystic fibrosis transmembrane conductance regulator function produces abnormalities in tracheal development in neonatal pigs and young children. , 2010, American journal of respiratory and critical care medicine.

[37]  D. Meyerholz,et al.  Sonographic evidence of abnormal tracheal cartilage ring structure in cystic fibrosis , 2015, The Laryngoscope.

[38]  E. Lander,et al.  Development and Applications of CRISPR-Cas9 for Genome Engineering , 2014, Cell.

[39]  J. Renaud,et al.  Characterization of Dystrophin Deficient Rats: A New Model for Duchenne Muscular Dystrophy , 2014, PloS one.

[40]  Marie-Pierre Dubé,et al.  Human monogenic disorders — a source of novel drug targets , 2006, Nature Reviews Genetics.

[41]  Chady H Hakim,et al.  Animal models of Duchenne muscular dystrophy: from basic mechanisms to gene therapy , 2015, Disease Models & Mechanisms.

[42]  David J. Chen,et al.  DNA double strand break repair via non-homologous end-joining. , 2013, Translational cancer research.

[43]  A. Muotri,et al.  Pig models of neurodegenerative disorders: Utilization in cell replacement‐based preclinical safety and efficacy studies , 2014, The Journal of comparative neurology.

[44]  Steve Cunningham,et al.  Repeated nebulisation of non-viral CFTR gene therapy in patients with cystic fibrosis: a randomised, double-blind, placebo-controlled, phase 2b trial , 2015 .

[45]  E. Wolf,et al.  Transgenic pigs as models for translational biomedical research , 2010, Journal of Molecular Medicine.

[46]  Ronald G. Crystal,et al.  Genetic medicines: treatment strategies for hereditary disorders , 2006, Nature Reviews Genetics.

[47]  R. Finkel Read-Through Strategies for Suppression of Nonsense Mutations in Duchenne/ Becker Muscular Dystrophy: Aminoglycosides and Ataluren (PTC124) , 2010, Journal of child neurology.

[48]  F. Gleason,et al.  Prevention of muscular dystrophy in mice by CRISPR/Cas9-mediated editing of germline DNA , 2014 .

[49]  K. Chu,et al.  Characterization of Defects in Ion Transport and Tissue Development in Cystic Fibrosis Transmembrane Conductance Regulator (CFTR)-Knockout Rats , 2014, PloS one.

[50]  E. Hoffman,et al.  Impaired mucus detachment disrupts mucociliary transport in a piglet model of cystic fibrosis , 2014, Science.

[51]  R. Wanke,et al.  Dystrophin-deficient pigs provide new insights into the hierarchy of physiological derangements of dystrophic muscle. , 2013, Human molecular genetics.

[52]  E. Hoffman,et al.  Intestinal CFTR expression alleviates meconium ileus in cystic fibrosis pigs. , 2013, The Journal of clinical investigation.

[53]  C. Spurney Cardiomyopathy of duchenne muscular dystrophy: Current understanding and future directions , 2011, Muscle & nerve.

[54]  Jacqueline Corrigan-Curay,et al.  Genome Editing Technologies: Defining a Path to Clinic: Genomic Editing: Establishing Preclinical Toxicology Standards, Bethesda, Maryland 10 June 2014. , 2015, Molecular therapy : the journal of the American Society of Gene Therapy.

[55]  Michelle H. Prickett,et al.  Gene therapy in cystic fibrosis. , 2013, Translational research : the journal of laboratory and clinical medicine.

[56]  Florian Schmidt,et al.  CRISPR genome engineering and viral gene delivery: A case of mutual attraction , 2015, Biotechnology journal.

[57]  S. Matalon,et al.  The CFTR and ENaC debate: how important is ENaC in CF lung disease? , 2012, American journal of physiology. Lung cellular and molecular physiology.

[58]  F. Mingozzi,et al.  Humoral immunity to AAV vectors in gene therapy: challenges and potential solutions. , 2013, Discovery medicine.

[59]  R. Wanke,et al.  First inducible transgene expression in porcine large animal models , 2012, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[60]  E. Wolf,et al.  Genetically engineered pig models for diabetes research , 2013, Transgenic Research.

[61]  INFRAFRONTIER Consortium INFRAFRONTIER – – providing mutant mouse resources as research tools for the international scientific community , 2015 .

[62]  K. Davies,et al.  Therapy for Duchenne muscular dystrophy: renewed optimism from genetic approaches , 2013, Nature Reviews Genetics.

[63]  Yolanda Santiago,et al.  Efficient generation of a biallelic knockout in pigs using zinc-finger nucleases , 2011, Proceedings of the National Academy of Sciences.

[64]  K. Davies,et al.  Functional correction in mouse models of muscular dystrophy using exon-skipping tricyclo-DNA oligomers , 2015, Nature Medicine.

[65]  Garry R. Cutting,et al.  Cystic fibrosis genetics: from molecular understanding to clinical application , 2014, Nature Reviews Genetics.

[66]  David V Schaffer,et al.  The AAV Vector Toolkit: Poised at the Clinical Crossroads. , 2012, Molecular therapy : the journal of the American Society of Gene Therapy.

[67]  K. Davies The era of genomic medicine. , 2013, Clinical medicine.

[68]  Yifan Dai,et al.  Targeted disruption of the alpha1,3-galactosyltransferase gene in cloned pigs. , 2002, Nature biotechnology.

[69]  A. Coates,et al.  Efficient Gene Delivery to Pig Airway Epithelia and Submucosal Glands Using Helper-Dependent Adenoviral Vectors , 2013, Molecular therapy. Nucleic acids.

[70]  David K Meyerholz,et al.  Disease phenotype of a ferret CFTR-knockout model of cystic fibrosis. , 2010, The Journal of clinical investigation.

[71]  J. Olsen,et al.  Ferret and pig models of cystic fibrosis: prospects and promise for gene therapy. , 2014, Human gene therapy. Clinical development.

[72]  Kwang-Wook Park,et al.  Production of α-1,3-Galactosyltransferase Knockout Pigs by Nuclear Transfer Cloning , 2002, Science.

[73]  R. Finkel,et al.  Phase 2a Study of Ataluren-Mediated Dystrophin Production in Patients with Nonsense Mutation Duchenne Muscular Dystrophy , 2013, PloS one.

[74]  Hans Clevers,et al.  Functional repair of CFTR by CRISPR/Cas9 in intestinal stem cell organoids of cystic fibrosis patients. , 2013, Cell stem cell.

[75]  D. Meyerholz,et al.  Sinus Hypoplasia Precedes Sinus Infection in a Porcine Model of Cystic Fibrosis , 2012, The Laryngoscope.

[76]  M. Welsh,et al.  Human cystic fibrosis airway epithelia have reduced Cl− conductance but not increased Na+ conductance , 2011, Proceedings of the National Academy of Sciences.

[77]  T. Partridge The mdx mouse model as a surrogate for Duchenne muscular dystrophy , 2013, The FEBS journal.

[78]  Roger M. Stein,et al.  Financing drug discovery for orphan diseases. , 2014, Drug discovery today.

[79]  Jeffry D. Sander,et al.  CRISPR-Cas systems for editing, regulating and targeting genomes , 2014, Nature Biotechnology.

[80]  A. Schnieke,et al.  The new pig on the block: modelling cancer in pigs , 2013, Transgenic Research.

[81]  E. Alton,et al.  Cystic fibrosis gene therapy: successes, failures and hopes for the future , 2009, Expert review of respiratory medicine.

[82]  Daniel G. Anderson,et al.  Non-viral vectors for gene-based therapy , 2014, Nature Reviews Genetics.

[83]  William H. Majoros,et al.  Multiplex CRISPR/Cas9-Based Genome Editing for Correction of Dystrophin Mutations that Cause Duchenne Muscular Dystrophy , 2015, Nature Communications.

[84]  B. N. Day,et al.  Production of α-1,3-Galactosyltransferase Knockout Pigs by Nuclear Transfer Cloning , 2002, Science.