Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Dec 2.
Published in final edited form as: Curr Gene Ther. 2015;15(6):531–540. doi: 10.2174/1566523215666150929110424

Contemporary Animal Models For Human Gene Therapy Applications

Chitra Gopinath 1, Trupti Job Nathar 1, Arkasubhra Ghosh 2, Dennis Durand Hickstein 3, Everette Jacob Remington Nelson 1,*
PMCID: PMC7709571  NIHMSID: NIHMS1646314  PMID: 26415576

Abstract

Over the past three decades, gene therapy has been making considerable progress as an alternative strategy in the treatment of many diseases. Since 2009, several studies have been reported in humans on the successful treatment of various diseases. Animal models mimicking human disease conditions are very essential at the preclinical stage before embarking on a clinical trial. In gene therapy, for instance, they are useful in the assessment of variables related to the use of viral vectors such as safety, efficacy, dosage and localization of transgene expression. However, choosing a suitable disease-specific model is of paramount importance for successful clinical translation. This review focuses on the animal models that are most commonly used in gene therapy studies, such as murine, canine, non-human primates, rabbits, porcine, and a more recently developed humanized mice. Though small and large animals both have their own pros and cons as disease-specific models, the choice is made largely based on the type and length of study performed. While small animals with a shorter life span could be well-suited for degenerative/aging studies, large animals with longer life span could suit longitudinal studies and also help with dosage adjustments to maximize therapeutic benefit. Recently, humanized mice or mouse-human chimaeras have gained interest in the study of human tissues or cells, thereby providing a more reliable understanding of therapeutic interventions. Thus, animal models are of great importance with regard to testing new vector technologies in vivo for assessing safety and efficacy prior to a gene therapy clinical trial.

Keywords: Animal models, Gene therapy, Genetic diseases, Viral vectors, Humanized mice, Clinical trials

INTRODUCTION

Gene therapy refers to the replacement of a defective disease-causing gene with a normal functional copy of the same gene to treat genetic diseases. Genetic mutations result in either loss or gain of function of genes leading to several genetic disorders including cancer. Therapeutic genes are delivered by either non-viral (physical or chemical) or viral methods into specific target cells [1, 2]. Viral vectors are highly efficient in gene transfer both in vitro and in vivo; however, major concerns such as immune response and insertional mutagenesis still remain [3]. Animal models are valuable tools in biomedical research to assess variables related to the use of viral vectors such as safety, efficacy, dosage and localization of transgene expression before moving onto an eventual human trial. A facility accommodating large animals like dogs, monkeys, and pigs is a cost-and-infrastructure intensive set-up as compared to a facility housing small animals like mice, rats and rabbits. Small animal models are a lot easier to handle and maintain, and since they have a short life span, they could be used to assess safety and efficacy within a short duration unlike large animal models. Though mouse genome is 99% similar to that of humans, [4] they do differ to some extent in the immunological make-up. On the other hand, using a large animal model could help with dosage adjustments, for instance, therapeutic interventions in dogs could very well correlate with small children [5]. Moreover, supportive studies on large models could be beneficial based on primary experimental conclusions made from small models to further improve therapeutics. A comparison between small and large animal models is summarized (Table 1). Here, we highlight some of the animal models of human diseases that are widely used in many gene therapy studies.

Table 1.

A comparative look at small and large animal models.

Parameters Small Models Large Models
Handling and maintenance Easy handling, inexpensive, minimal housing space, higher litter numbers Laborious, expensive, larger housing space, fewer litter numbers
Phenotypic expression Partially recapitulate complex human clinical features Closely mirror clinical presentations of genetic disorders with respect to genes, anatomy and physiology
Disease modelling Genetic disease models generated in less duration due to shorter gestation period (21 days) using techniques like ZNF, TALEN and CRISPR/Cas9 Takes longer duration to generate genetically modified animals due to longer gestation period (≥ 2 months)
Longitudinal studies Restricted to shorter experimental studies < 1.5 years Longer experimental studies of >2 years possible due to longer life span of the animal
Therapeutic interpretation Dosage adjustments cannot be correlated for clinical applications Dosage adjustments are similar to that of a small child

MURINE MODELS

Laboratory mice and rats are a favorite as they can be handled and maintained with ease. The litter size is more per cycle, thereby providing larger number of experimental animals. Mice are usually inbred and backcrossed resulting in genetically identical siblings in which case findings cannot be correlated with humans carrying genetic variations and signature gene polymorphisms. In the past, researchers have used spontaneous mutation models such as the severe combined immunodeficiency (SCID) mice [6], which were used in cancer gene therapy studies to model different types of cancer due to their naturally immune compromised state [7, 8]. Limitations of spontaneous mutation models include i) the intermittent rate at which these mutations occur [9] and ii) timely breeding based on visual identification of phenotypic differences.

With the advent of gene manipulation techniques, it is now possible to genetically engineer monogenic disease models within a short period of 6 to 8 months using methods such as zinc-finger nucleases (ZFN) [10, 11], transcription activator-like effector nucleases (TALEN) [12, 13] and a more recently developed clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 [14, 15]. These techniques are also recently reviewed elsewhere [16, 17]. Multiple gene knockout mouse can also be generated with ease using multiple CRISPR/Cas9 vector constructs [1822], which would be very useful to model complex genetic disorders involving multiple genes. A recent application of this new technology is generation of the Spata7 gene knockout mouse to model congenital eye disorders such as Leber’s congenital amaurosis (LCA) and juvenile retinitis pigmentosa (RP) [23, 24]. Genetically engineered mouse (GEM) models of metabolic diseases and cancers are of great interest in the study of loss of function of tumor suppressor genes or gain of function of proto-oncogenes. One recent example is the generation of Ace2 knockout mouse to mimic colitis leading to cancer [25]. Older cancerous xenograft models and chemically induced cancer models have limitations such as immunocompromised state and physiological side effects where the animals fail to resemble the disease and hence are unable to accurately respond to treatment. Taneja et al. gives an insight on few crucial transgenic and knockout mouse models of tumor suppressor genes [26].

Murine models are used in various gene therapy studies including those for cancer [27, 28], muscular dystrophies [29, 30] hematological [31, 32], neurological [33], respiratory [34], liver [35] and cardiovascular disorders [36]. In general, it is always a good practice to evaluate a novel therapeutic strategy in an animal model first before performing the same on human subjects to help assess safety and improve therapeutics. Sometimes, mouse models do not entirely mirror phenotypic features of human diseases in which case it becomes difficult to interpret and correlate crucial findings for human applications [37, 38]. In one study, lentiviral vectors were customized to improve specificity and potency for the treatment of chronic granulomatous disease (CGD) by gene therapy using tissue-specific and gene-specific promoter, gp91phox to drive therapeutic gene expression in phagocytes. The most promising vector giving therapeutic levels of expression was used to treat CGD mice [39]. Following in vitro testing of newly designed vectors, potential ones could be picked to determine their efficacy in vivo using small preclinical models.

A comparative preclinical study between adult mice and non-human primates (NHPs) showed that intravascular administration of adeno-associated viral vector serotype 9 (AAV9) was able to transduce and actively cross the blood-brain barrier comparatively better in mice than NHPs where poor transduction into brain and peripheral organs was observed. This could be due to the presence of low levels of pre-existing neutralizing antibodies (NAbs) in NHPs that could have blocked transduction into the brain and peripheral organs [40]. These results conclude that the presence of pre-existing NAbs, peripheral tropism and limited neuronal transduction are major concerns impeding human translation using AAV9 by intravascular delivery [41]. Similar differences in AAV9 transduction profile between dogs and mice have been previously reported [42, 43]. NAbs against AAV serotypes vary with respect to titers between species and serotypes, therefore prior screening for appropriate models for gene transfer experiments is crucial before conducting preclinical gene therapy studies [44].

Gene therapy was able to restore vision and delay retinal degeneration in a knockout mouse model of RP (CNGB1−/−) after therapeutic administration of recombinant AAVs (rAAVs) into the sub-retinal space of a two-week old CNGB1−/− mouse [45]. This study provided proof of concept that could be further tested in a large animal model (e.g. dog or non-human primate) before translating the findings to humans. AAV2-RPE65 gene therapy to treat LCA in humans has been a true success story in the last decade [4651].

In an anti-aging study, aged mice were induced to overexpress the enzyme telomerase by gene therapy. Telomerase has been shown to slow down the cells’ biological clock, thereby increasing the life span by 24% with a single treatment and without increasing the risk of cancer in the adult mice [52]. Another study showed reversal of loss of memory by gene therapy in mouse models during the initial stages of Alzheimer’s disease [53]. Preclinical testing for degenerative studies could be very time-consuming in a large animal model, whereas mice with their shorter life span could facilitate beneficial findings over a relatively short period of time.

It has been reported in a few cases that rat models when compared to mice were better in mimicking bioelectric phenotypes similar to those seen in human CF patients [34]. In addition, due to their smaller size and ease of handing, rats serve as better alternatives to large animals, yet recapitulating muscular lesions seen in human patients with Duchenne muscular dystrophy (DMD) [30].

CANINE MODELS

Dogs are excellent preclinical models used in many gene therapy studies. Over 58% of genetic diseases seen in the dogs closely depict the phenotype of human diseases caused by mutations in orthologous genes. The size of the animal is similar to that of a small child in which case dosage adjustments could be easily done. Immunological and clinical phenotypes in dogs are similar to those in humans most of the time [54].

Before using viral vectors to treat human diseases by gene therapy, the safety and efficacy of these vectors need to be first determined with the help of a reliable experimental model. For retinal gene therapies, consecutive local administration of rAAV vectors may be required for long-term expression of therapeutic gene in humans as the time window for administering the vector is unclear, although some preclinical reports claim that sustained expression could be achieved with one-time administration [55]. Immunosuppression for systemic bone marrow gene therapy requires non-myeloablative busulfan treatment in canine models to facilitate transplantation of gene-corrected hematopoietic stem cells [56]. In general, in order to boost levels and sustain expression of a therapeutic gene, immunosuppression in combination either with infusion of gene-corrected cells or administration of therapeutic gene-encoding viral vectors is necessary with systemic delivery [57]. Local administrations into the eyes have an immuneprivilege as they are compartmentalized and immune responses are limited to the injection site [58, 59]. The goal is to design a treatment modality involving minimal or complete elimination of prior immunosuppression that is commonly done in clinical practice.

Treatment of canine leukocyte adhesion deficiency (CLAD) using foamy virus vectors demonstrated amelioration of symptoms in four out of five CLAD dogs infused with vector-transduced autologous CD34+ hematopoietic stem cells (HSCs) following a non-myeloablative conditioning regime leading to complete reversal of the CLAD phenotype [60]. All treated dogs have been shown to sustain CD18 expression and essentially disease-free for more than 4 years post-treatment. There were no genotoxic effects reported and further the risk of integration near oncogenes was shown to be much lower compared to that of gammaretroviral vectors [61]. Disease characteristics in CLAD-affected dogs are similar to those in humans [62]. Therefore, dogs are considered as suitable large animal models for hematopoietic stem cell gene therapy [5]. In a few recent gene therapy studies, dogs have also been used as models for haemophilia [63, 64], ocular disorders [65, 66] and cardiovascular diseases [67, 68].

X-linked retinitis pigmentosa in humans and dogs is caused by defects in the retinitis pigmentosa GTPase regulator (RPGR) gene and results in early severe and progressive vision loss. It is one of the most common inherited forms of retinal degeneration in man. The defect was shown to be corrected with sub-retinal injections of AAV2/5 vectors carrying the human RPGR gene under the control of human interphotoreceptor retinoid-binding protein (IRBP) or G protein-coupled receptor kinase 1 (GRK1) promoters [69]. The similarities between humans and dogs in terms of eye anatomy, vision assessment [7073], disease characteristics and positive responses to gene therapy provide hope towards human translation in the near future.

Diabetes is caused due to prolonged impairment of glycemic control. A gene therapy study was conducted involving two genes, glucokinase and insulin that work synchronously and detect high blood sugar levels promoting the uptake of blood glucose into target cells. Long-term efficacy of this approach in a canine model of diabetes was also demonstrated. A one-time intramuscular administration of AAV1 encoding Gck and Ins was used to treat insulin deficient diabetic dogs. This study provided the first proof of concept in a large animal model for a gene transfer approach to treat diabetes in humans [74]. A recent AAV8-mediated gene therapy in a dog model to treat myotubular myopathy with a single-dose intravascular delivery significantly improved severe muscle weakness and respiratory impairment, and extended the life span to more than 1 year with no toxicity or immune response [75].

NON-HUMAN PRIMATE MODELS

Non-human primates (NHPs) such as African green monkeys, baboons, chimpanzees, cynomolgous monkeys, rhesus monkeys and owl monkeys are considered the most suited animal models for preclinical testing as they are evolutionarily and genetically very closely related to humans compared to any other mammals. Rhesus macaques, in particular, have contributed immensely to several preclinical studies, although selection of a suitable model remains crucial considering differences in immunological responses to viral vectors between NHPs and humans which could impede translation into human gene therapy trials [40, 7679]. Recently, successful generation of site-specific multiple gene targeted cynomolgus monkeys was demonstrated using the CRISPR/Cas9 system without any off-target effects paving way for generation of more genome engineered large animal models in the future [80].

NHP models accurately mirror the effectiveness of rAAV-mediated gene delivery to the airways in humans. Using an identical model system such as differentiated airway epithelial cells from the Indian Rhesus monkeys and from humans cultured at an air-liquid interface, the biology of rAAV-mediated gene transfer for three serotypes was assessed between the two species. While the airway epithelia from NHPs and humans have similar Na+ and Cl transport properties, rAAV transduction of airway epithelia of NHPs showed significant differences compared to those of humans in terms of the efficiency of transduction with all the three rAAV serotypes tested (AAV1, AAV2, AAV5) [81].

Another pilot study for cystic fibrosis (CF) gene therapy using a marmoset model showed that an intratracheal bolus dose of vesicular stomatitis virus G protein (VSV-G) pseudotyped HIV-1 based lentiviral (LV) vector carrying the LacZ reporter gene after airway treatment resulted in LacZ gene expression (X-gal). This was found primarily in the airway epithelia and alveolar regions. The LacZ gene expression was not seen in the liver or spleen. This study confirmed the transduction potential of CF-relevant airway cell types. The marmoset is an encouraging NHP model for testing and translating genetic treatments for CF ahead of clinical trials [82]. Marmosets are small and similar in body size to children and hence small volumes of viral vectors are sufficient for treatment which could be easily produced in the laboratory using the currently available methods of vector production.

NHPs have a vertebrate brain like that of humans in terms of neural circuitry, physiological and behavioral characteristics, which makes them a crucial and precise model for neurological diseases compared to other animals. Spontaneous atrophy of neurons occurs with aging in the brain of non-human primates similar to humans. Long-term expression of nerve growth factor (NGF) by LV-mediated intraparenchymal delivery has been shown to reverse the age-related cholinergic degeneration owing to sustained gene expression for one year in young monkeys [83]. These findings could pave way for potential testing in patients suffering from Alzheimer’s disease. Another study reported to be effective and first of its kind is the AAV2 delivery to the NHP hippocampus using frameless stereotactic methods [84], which is very similar to the stereotactic delivery methods used previously in clinical gene therapy trials [85].

Red-green colour blindness, which results from the absence of wavelength sensitive visual photopigments is a common single gene eye disorder. Gene therapy was performed on adult squirrel monkeys that were congenitally colour blind due to the absence of L-opsin. Addition of a third opsin to correct the defect in vision resulted in trichromatic colour vision in the treated monkeys [86]. This study provided a positive outlook on treating congenital vision defects in humans.

HUMANIZED MOUSE MODELS

Mouse-human chimeras or “humanized mice” are immunodeficient mice designed to express human transgenes or engrafted with human cells or tissue [87]. The use of humanized mice is comparatively beneficial as they are designed to mimic human biology and features of human disease conditions, the treatment response in which could provide a precise model to address various health problems such as the immunodeficiency disorders. Although bone marrow transplantation (BMT) or hematopoietic stem cell transplantation (HSCT) are currently available to treat various immune and genetic disorders, there are considerable limitations with each of these approaches [88]. Using some of the common immunosuppressed murine models such as the NOD/SCID (non-obese diabetic/severe combined immunodeficiency) and NSG (NOD/SCIDγc−/−) mice, human HSCs or patient-derived xenografts could be established for more precise clinical experimentation [89]. Due to differences in human and animal physiology, very often findings in any of the small preclinical models may not completely translate to humans. Hence, an in vivo model that closely mimics humans is better in order to translate findings from the laboratory bench to the clinical bedside.

Neutrophils in mice are different from that in humans in terms of their role, structure of surface molecules and certain signaling pathways. Recent research has made it possible to design immunodeficient mouse strains that would allow better engraftment of human hematopoietic cells by deleting the interleukin-2 (IL-2) receptor. Studies have demonstrated that functional human neutrophils tend to develop from human CD34+ cord blood stem cells in NOD-scid-γc−/− mice [90]. HIV-AIDS model and hu-HSC-transplanted NOD-scid-γc−/− mice have both been used in the treatment of HIV in various gene therapy studies [91]. Humanized models such as these could be used in the study of various human-specific autoimmune, metabolic, cancer and infectious diseases [9296].

Similar studies could be carried out for leukocyte adhesion deficiency type 1 (LAD1). Using a mouse model, it has been shown that the therapeutic human CD18 subunit was able to heterodimerize with the mouse CD11a subunit [97]. On the other hand, if a humanized mouse engrafted with human HSCs were to be used, heterodimerization of the human CD18 would occur with all of the four human CD11 subunits such as CD11a, CD11b, CD11c, CD11d, as it would in a human system. Humanized mice could serve as strong preclinical models to understand the blood biology in health and disease and therefore have an important role in biomedical research in the future.

Humanized murine liver models such as the immunosuppressed FRG mouse (Fah−/−/Rag 2−/−/Il2rg−/−) partially repopulated with human hepatocytes [98] were used to evaluate the efficiency of transduction of AAV variants into human hepatocyte xenografts. Out of 15 vectors tested in vitro and a few in vivo for various parameters such as the presence of neutralizing antibodies against the vectors [40], it was shown that the rAAV-LK03 serotype preferentially transduced human hepatocytes. This suggests that a human-mouse chimeric liver model could be used to accurately screen multiple gene therapy vectors for various factors such as virus entry based on serotype and species specificity and level of transgene expression post-infection, which could not have been precisely modeled in normal mice or NHP models otherwise [99].

PORCINE MODELS

Porcine models have served as desirable biomedical research models for a range of human diseases [100]. Preliminary data confirms the phylogenetic proximity of swine to humans as compared to the rodent species, which further closely relates them to humans in their physiology, histopathology, diet, metabolism and pharmacokinetics [101]. Many cases of porcine models with both spontaneous and targeted genetic mutations have been reported. The use of genetically engineered pigs is dramatically increasing in the field of biomedical disease modeling. This is due to their similarities to humans with regard to metabolism, physiology, genome organization, aging and pathology. However, spontaneous mutant models have their own disadvantages in terms of risks related to insertion of transgenes into undesired locations. Transgenic models of pigs for many diseases like Alzeihmer’s disease, diabetes, cancer, ophthalmological and cardiovascular disorders have been generated and found to be successful [102].

Swine is one of the most preferred models for cardiovascular diseases, as myocardial infarction could be produced in them in expected sizes and location with much ease. Also, its resting heart rates and left ventricular (LV) pressures are found to be analogous to humans. In one study, long-term expression of β-adrenergic receptor kinase carboxy terminus (β-ARKct) was evaluated to monitor cardiac function by AAV6-mediated myocardial gene delivery. β-ARKct is a peptide inhibitor of G protein-coupled receptor kinase 2 (GRK2), which is the key mechanism attributed to cardiac dysfunction. Therefore, AAV6 vectors encoding the β-ARKct gene was shown to result in long-term expression causing substantially significant amelioration of cardiac function and normalization of catecholaminergic axis and LV haemodynamics [103].

Several other studies stating the importance of porcine models for cardiac related disorders have been carried out where short-term over expression of inducible nitric oxide synthase (iNOS) gene was found to be effective in locally increasing nitric oxide (NO) levels thereby preventing intimal hyperplasia [104]. Another study showed that an alternatively spliced form of the vascular endothelial growth factor, VEGF165 also correspondingly acts upon adult cardiomyocyte cell cycle, consequently increasing both mitotic index and nuclear hyperplasia eventually leading to cardiomyocyte regeneration in pigs [105]. The Na+-Ca2+ exchanger/sarcoplasmic/endoplasmic reticulum Ca2+ ATPase (NCX/SERCA2a) ratio targeting cardiomyocyte Ca2+ cycling required to reduce diastolic SR Ca2+ leak and increase systolic contractility of the cardiomyocyte is higher in large animal models like pigs and humans, thus forming a critical link in translating basic concepts into clinical therapies [106]. Another study showed that dual AAV trans-splicing and hybrid vectors could transduce pig photoreceptors, the major cell targets for the treatment of inherited retinal dystrophies (IRDs), to levels which were about two to threefold lower than those attained with a single AAV vector of normal size. This efficiency of dual AAV vectors is significantly higher than in mice, which is potentially due to higher levels of dual AAV co-transduction being observed in pigs [107].

Cystic fibrosis is one of the diseases where successful gene therapy could be a major boon. Ion transport defect i.e. lack of cAMP-stimulated anion transport in sinus epithelia were corrected in a porcine model using an adenoviral (AdV) vector. Porcine sinus epithelia were transduced using an AV vector expressing CFTR, which lead to restoration of cAMP-mediated anion transport indicating that CFTR expression by a comparatively small number of cells could restore anion transport, a successful step in disease correction which was first demonstrated in a porcine model of cystic fibrosis. However, this still leaves behind a question of alteration in CF phenotype in sinus hypoplasia through CFTR correction [108]. The long life span of swine makes it amenable to the study of a disease like CF where lung deterioration progresses over a comparatively long period of time paving way for potential novel therapeutics to be tested over time.

In haemophilia A, a bleeding disorder caused by abnormalities in the coagulation factor VIII, gene therapy enabled sustained elevation of factor VIII levels. Currently available mouse models failed to extrapolate the human condition as they could not show similar coagulation ability. Hence, a porcine model was generated from F8 targeted fibroblasts using nuclear transfer. The infusion of human factor VIII using F8 targeting vector, constructed by insertion of two genomic DNA into the plasmid vector pHSv-TK/PGK-Neo resulted in reduced bleeding [100]. This would make way for porcine models as efficient animal models for gene therapy of haemophilia A.

The use of a porcine model is not just limited to the above mentioned disorders but is also widely used for various muscular genetic disorders like DMD and spinal muscular atrophy (SMA). SMA is an autosomal recessive disorder resulting either from the loss or mutation of two independent genes, survival motor neuron 1 and 2 (SMN1 and SMN2), both of which code for the same protein, SMN [109, 110]. Most of the functional SMN protein is from the SMN1 gene, while a small part of the protein is produced from the SMN2 gene. Therefore, mutations and/or absence of both the genes could lead to a more severe phenotype. As SMN2 gene is seen only in humans, naturally occurring mutations in this gene are rare in other species. Like other animals, mice possess only SMN1, the loss of which is embryonically lethal [111]. Hence, SMA mouse model with SMN1 gene knockdown was developed which showed remarkable survival with the use of scAAV9-SMN vector encoding the normal functional copy of the SMN1 gene [112114]. However, its relative permeability to blood-brain barrier and discrete symptoms like tail necrosis and cardiac defects that are unlikely seen in humans make the SMA mouse model unsuitable to study the human disease [115117]. Hence, a large animal model like the porcine model, the very first for SMA, was developed by post-natal reduction of SMN protein in motor neurons for proper understanding of the disease phenomenon. A scAAV9-hSMN vector that was delivered at 4 weeks post-knockdown rescued the gene expression and also improved the neuropathology [118]. This indicates that the degeneration process in motor neurons due to low SMN levels could be reversed even after the appearance of symptoms. Thus, a large animal model like porcine could be beneficial in mimicking the human pathology, especially for gradual onset diseases.

RABBIT MODELS

Rabbits are more often considered for the production of monoclonal and polyclonal antibodies. However, they are increasingly being used as experimental models recently. Oryctolagus cuniculus, white New Zealand rabbit is extensively used for clinical studies. Transgenic rabbits have previously been chosen as animal models for diverse studies including lipoprotein metabolism, atherosclerosis, cardiovascular research and hypertrophic cardiomyopathy [119].

One study showed prevention of arterial thrombus formation i.e. promoting thromboresistance by local overexpression of tissue plasminogen activator in a rabbit model [120]. Tissue plasminogen activator (tPA) catalyzes the rate-limiting step, which involves the conversion of proenzyme plasminogen to plasmin in the presence of fibrin [121]. Recombinant adenovirus vector expressing human tPA under the control of Rous sarcoma virus (RSV) promoter (Adv/RSV-tPA) was used to determine the impact of local tPA overexpression by delivering into the rabbit common femoral artery. Adv/RSV-tPA vector construct not only barred occluding intra-arterial thrombus formation, but also limited the complications by a decline in fibrin split product levels constantly seen with systemic haemorrhage [120]. Though transgenic mice are most extensively used for human diseases, it is found that in certain mutation cases they fail to display expected pathological symptoms. In terms of molecular composition of cardiac sarcomeric proteins, the mutant phenotypes were found to reflect more similarity with rabbits than rodents [119].

Another study where pathogenesis of transplant-related arteriosclerosis in rabbits due to smooth muscle cell replication was overcome by elimination of dividing cells for expression of herpes virus thymidine kinase (hsv-tk), which produces a toxic compound killing cells by phosphorylation of gancyclovir. Smooth muscle cells allow gene transfer only under de-endothelialization of arteries. Due to the resistance exhibited by these cells in gene transfer, certain manipulations to increase the porosity of arteries were made. These manipulations were successful in enhancing the efficiency of gene transfer in smooth muscle cells [122].

Limb ischemia is one of the prevailing problems due to lack of pharmacological interventions. The need for alternative treatments has led to the development of a novel strategy where angiogenic growth factors, such as hepatocyte growth factor (HGF) were shown to augment collateral artery development [123]. Intramuscular injection of HGF plasmid into rabbit ischemic hind limb not only showed significant augmentation of collateral vessel development, but also their extension from stem artery source to the end-point of reconstituted parent vessel. This study on rabbit ischemic hind limb models proved to be a potential therapy for peripheral arterial diseases thus making rabbits as efficient models for gene therapy.

For the very first time, a serious pathological condition like corneal neovascularization in the rabbit cornea has been significantly inhibited using decorin (proteoglycan). After examining multiple AAV serotypes (2, 5, 7, 8, and 9), AAV5, AAV8 and AAV9 were shown to lead to efficient transduction of the rabbit cornea. Superior transduction efficiency of AAV5 showed about 67% reduction in corneal neovascularization. Also, it was shown that decorin gene transfer into the cornea remarkably reduced VEGF induced corneal angiogenesis. Low inflammatory response with minimal confounding effects of wound healing enables a better understanding of molecular mechanisms with regard to corneal angiogenesis, eventually making rabbit the most frequently used animal model for angiogenesis research [124].

Safety of gene therapy approaches depends on various factors including vector dose, biodistribution and route of administration. Rabbits serve as excellent models to determine safety based on the above mentioned factors. In one such study, a rabbit model was used to characterize the safety of AAV serotypes i.e. AAV5 and AAV6 for liver-mediated human factor IX (hFIX) expression at different doses of 1×1012 or 1×1013 viral genomes/kg [125]. Both cohorts of AAV6 showed remarkable circulating therapeutic levels of hFIX, while slightly high dose of AAV5 was found to be effective. Also, no inhibitory antibodies to hFIX were found with AAV5 injected rabbits. While comparing the serotypes based on biodistribution, it was shown that AAV6 was found outside the liver and spleen in very low concentrations, whereas AAV5 was found in many tissues and organs. Rabbit strains have shown varied genetic background compared to both inbred and outbred strains. This can be an advantage for studying intricate disease models in view of the fact that they mimic human genetic diversity with high accuracy.

OTHER ANIMAL MODELS

Feline Models

Cats are of great interest for neurological disorders [126] since their brain is >50 times larger than the mouse brain and also its anatomy is more similar to that of a human than a rat or a mouse [127, 128]. Sandhoff disease (SD) is a neurodegenerative lysosomal storage disease caused by catalytic deficiency of β-N-acetylhexosaminidase (Hexβ) responsible for step-wise catabolism of GM2-ganglioside by removal of its terminal N-acetylgalactosamine residue. Hexβ results in Sandhoff disease, whereas the defect in the α-subunit of Hex results in Tay-Sachs disease. Since optimal production of Hex α and β is required for co-expression of both the subunits [129131], feline cDNAs of both the subunits were cloned in rhAAV8 vectors. Single intracranial injection of these vectors increased the life span of cats from 5 to 8 months. This increases the enzymatic activity to about 75-fold as compared to the normal with mild evidence of an immune infiltrate. Hence, feline model is preferred as an intermediate model between mouse and human studies for neurological disorders [132].

Bovine Models

The use of cattle for gene therapy is very rare, probably due to its size and also the high cost accrued due to large scale production of recombinant proteins and viral vectors [126]. However, using calves could be an alternative as they weigh around 30 kg, which is similar to that of an older child. Bovine model is the only available model for citrullinemia, an inborn error of urea cycle metabolism caused by deficiency of arginosuccinate synthatase leading to hyperammonemia. Inefficient pharmacological treatments where the oral administration of 15N4 -labelled urea was used to measure the nitrogen flux led to the use of viral vectors. Systemic administration of a first-generation E1-deleted AdV vector expressing human arginosuccinate synthetase successfully resulted in the transduction of hepatocytes and partial correction of the enzyme defect leading to restoration of urea synthesis [133].

Equine Models

Horses are one of the most commonly used models for osteoarthritis as this condition occurs in them naturally [134]. Osteoarthritis is a chronic, debilitating and expensive disease involving the joints [135]. A recent study on the therapeutic effects resulting from the intra-articular overexpression of the equine interleukin 1 receptor antagonist (IL-1Ra) gene by AdV-mediated gene transfer was established. Elevated levels of intra-articular expression of IL-1Ra after the in vivo delivery of equine IL-1Ra gene were observed up to 28 days. This resulted in a significant improvement in various clinical parameters such as pain, disease activity and prevention of articular cartilage, also with some beneficial effects on histologic parameters of articular cartilage and synovial membrane [134]. Surprisingly, most of the gene therapy experiments are performed in transgenic models instead of spontaneous mutant models such as the equine model.

CONCLUSION

Congenital and metabolic diseases caused by single gene defects leading to loss of function are viewed as potential candidates for gene therapy. Gene targeted knockout mouse model systems generated with the currently available gene manipulation techniques have their limitations as disease models for preclinical experiments which would make it difficult to develop novel therapies in an impending clinical trail. Therefore, it becomes a pertinent need to test large animal models like dogs and NHPs which are preferred based on various factors in close resemblance to humans such as similar size, anatomy, genetic background and disease pathology. Following an experimental trend from mice-to-dogs-to-humans could better validate various treatment modalities for diseases like cancer and other life-threatening human genetic disorders, thereby ensuring improved patient care in the future. Animals used to model human diseases affecting various organ systems are summarized (Table 2).

Table 2.

Animal models used in gene therapy for diseases related to various organ systems.

Animal Models Organ Systems References

Murine Muscular system [29, 30, 111, 114]
Circulatory system [31, 32, 36, 39]
Cancer [27, 28]
Ocular system [24, 45, 48]
Nervous system [33, 53]
Aging [52]

Canine Endocrine system [74]
Circulatory system [60, 61, 63, 64, 67, 68]
Ocular system [46, 47, 65, 66, 69]
Muscular system [42, 57, 75]

Non-human primates Ocular system [86]
Nervous system [83, 84]
Respiratory system [81, 82]

Humanized mice Cancer [95]
Circulatory system [88, 89, 92, 94]
Immune system [91, 96]
Hepatobiliary system [99]

Porcine Circulatory system [100, 103, 105]
Respiratory system [108]
Ocular system [107]
Muscular system [118]

Rabbit Circulatory system [120, 122]
Ocular system [124]

Other models (Feline, Bovine, Equine) Nervous system [132]
Hepatobiliary system [133]
Skeletal system [134]

ACKNOWLEDGEMENTS

Award of Ramalingaswami Fellowship by the Department of Biotechnology, Ministry of Science and Technology, Government of India to the senior author (E.J.R.N) is gratefully acknowledged.

Biography

graphic file with name nihms-1646314-b0001.gif

Footnotes

CONFLICT OF INTEREST

The author(s) confirm that this article content has no conflict of interest.

REFERENCES

  • [1].Felgner PL, Gadek TR, Holm M, et al. Lipofection: A highly efficient, lipid-mediated DNA-transfection procedure. Proc Natl Acad Sci USA 1987; 84(21): 7413–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [2].Miller DG, Adam MA, Miller AD. Gene transfer by retrovirus vector occurs only in cells that are actively replicating at the time of infection. Mol Cell Biol 1990; 10(8): 4239–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [3].Rosenberg SA, Aebersold P, Cornetta K, et al. Gene transfer into humans: immunotherapy of patients with advanced melanoma, using tumour infiltrating lymphocytes modified by retroviral gene transduction. N Engl J Med 1990; 323(9): 570–8. [DOI] [PubMed] [Google Scholar]
  • [4].Mouse Genome Sequencing Consortium, Waterston RH, Lindblad-Toh K, Birney E, et al. Initial sequencing and comparative analysis of the mouse genome. Nature 2002; 420(6915): 520–62. [DOI] [PubMed] [Google Scholar]
  • [5].Bauer TR Jr., Adler RL, Hickstein DD. Potential large animal models for gene therapy of human genetic diseases of immune and blood cell systems. ILAR J 2009; 50(2): 168–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [6].Bosma GC, Custer RP, Bosma MJ. A severe combined immunodeficiency mutation in the mouse. Nature 1983; 301(5900): 527–30. [DOI] [PubMed] [Google Scholar]
  • [7].Hu WX, Zeng ZJ, Luo SQ, Chen Q. Suicide gene therapy of human breast cancer in SCID mice model by the regulation of Tet-On. Chin Med J (Engl) 2004; 117(3): 434–9. [PubMed] [Google Scholar]
  • [8].Cook DR, Maxwell IH, Glode LM, et al. Gene therapy for B-cell lymphoma in a SCID mouse model using an immunoglobulin-regulated diphtheria toxin gene delivered by a novel adenovirus-polylysine conjugate. Cancer Biother 1994; 9(2): 131–41. [DOI] [PubMed] [Google Scholar]
  • [9].Stanford WL, Cohn JB, Cordes SP. Gene-trap mutagenesis: Past, present and beyond. Nat Rev Genet 2001; 2: 756–768. [DOI] [PubMed] [Google Scholar]
  • [10].Geurts AM, Cost GJ, Freyvert Y. Knockout rats via embryo microinjection of zinc-finger nucleases. Science 2009; 325(5939): 433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [11].Carroll Dana. Genome Engineering with zinc-finger nucleases. Genetics 2011; 188(4): 773–782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Miller JC, Tan S, Qiao G, et al. A TALE nuclease architecture for efficient genome editing. Nat Biotechnol 2011; 29(2): 143–8. [DOI] [PubMed] [Google Scholar]
  • [13].Tesson L, Usal C, Ménoret S, et al. Knockout rats generated by embryo microinjection of TALENs. Nat Biotechnol 2011; 29(8): 695–6. [DOI] [PubMed] [Google Scholar]
  • [14].Doudna JA, Charpentier E. Genome editing. The new frontier of genome engineering with CRISPR-Cas9. Science 2014; 346 (6213): 1258096. [DOI] [PubMed] [Google Scholar]
  • [15].Li D, Qiu Z, Shao Y, et al. Heritable gene targeting in the mouse and rat using a CRISPR-Cas system. Nat Biotechnol 2013; 31(8): 681–3. [DOI] [PubMed] [Google Scholar]
  • [16].Cai M, Yang Y. Targeted genome editing tools for disease modeling and gene therapy. Curr Gene Ther 2014; 14(1): 2–9. [DOI] [PubMed] [Google Scholar]
  • [17].Scharenberg AM, Duchateau P, Smith J. Genome engineering with TAL-effector nucleases and alternative modular nuclease technologies. Curr Gene Ther 2013; 13(4): 291–303. [DOI] [PubMed] [Google Scholar]
  • [18].Nakagawa Y, Sakuma T, Sakamoto T, et al. Production of knockout mice by DNA microinjection of various CRISPR/Cas9 vectors into freeze-thawed fertilized oocytes. BMC Biotechnol 2015; 15: 33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [19].Zhou J, Shen B, Zhang W, et al. One-step generation of different immunodeficient mice with multiple gene modifications by CRISPR/Cas9 mediated genome engineering. Int J Biochem Cell Biol 2014; 46: 49–55. [DOI] [PubMed] [Google Scholar]
  • [20].Fujii W, Onuma A, Sugiura K, Naito K. One-step generation of phenotype-expressing triple-knockout mice with heritable mutated alleles by the CRISPR/Cas9 system. J Reprod Dev 2014; 60(4): 324–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [21].Wang H, Yang H, Shivalila CS, et al. One-step generation of mice carrying mutations in multiple genes by CRISPR/Cas-mediated genome engineering. Cell 2013; 153(4): 910–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [22].Li W, Teng F, Li T, Zhou Q. Simultaneous generation and germ-line transmission of multiple gene mutations in rat using CRISPR-Cas systems. Nat Biotechnol 2013; 31(8): 684–6. [DOI] [PubMed] [Google Scholar]
  • [23].Zhong H, Eblimit A, Moayedi Y, et al. AAV8(Y733F)-mediated gene therapy in a Spata7 knockout mouse model of Leber congenital amaurosis and retinitis pigmentosa. Gene Ther 2015; 22(8): 619–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [24].Boye SL, Peshenko IV, Huang WC, et al. AAV-mediated gene therapy in the guanylate cyclase (RetGC1/RetGC2) double knockout mouse model of Leber congenital amaurosis. Hum Gene Ther 2013; 24(2): 189–202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [25].Liu C, Xiao L, Li F, et al. Generation of outbred Ace2 knockout mice by RNA transfection of TALENs displaying colitis reminiscent pathophysiology and inflammation. Transgenic Res 2015; 24(3): 433–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [26].Taneja P, Zhu S, Maglic D, et al. Transgenic and knockout mice models to reveal the functions of tumor suppressor genes. Clin Med Insights Oncol 2011; 5: 235–257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].Zhang L, Hedjran F, Larson C, et al. A novel immunocompetent murine model for replicating oncolytic adenoviral therapy. Cancer Gene Ther 2015; 22: 17–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [28].Morrison BJ, Tagaya Y, Steel JC, et al. Adenoviral-mediated interleukin-15 receptor-alpha gene therapy of murine breast cancer. Mol Ther 2006; 13: S165. [Google Scholar]
  • [29].DelloRusso C, Scott JM, Hartigan-O’Connor D, et al. Functional correction of adult mdx mouse muscle using gutted adenoviral vectors expressing full-length dystrophin. Proc Natl Acad Sci USA 2002; 99(20): 12979–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [30].Larcher T, Lafoux A, Tesson L, et al. Characterization of dystrophin deficient rats: a new model for Duchenne muscular dystrophy. PLoS One 2014; 9(10): e110371. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [31].Kuether EL, Schroeder JA, Fahs SA, et al. Lentivirus-mediated platelet gene therapy of murine hemophilia A with pre-existing anti-factor VIII immunity. J Thromb Haemost 2012; 10(8): 1570–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [32].Negre O, Bartholomae C, Beuzard Y, et al. Preclinical evaluation of efficacy and safety of an improved lentiviral vector for the treatment of β-thalassemia and sickle cell disease. Curr Gene Ther 2015; 15(1): 64–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [33].Haase G, Kennel P, Pettmann B, et al. Gene therapy of murine motor neuron disease using adenoviral vectors for neurotrophic factors. Nat Med 1997; 3(4): 429–36. [DOI] [PubMed] [Google Scholar]
  • [34].Tuggle KL, Birket SE, Cui X, et al. Characterization of defects in ion transport and tissue development in cystic fibrosis transmembrane conductance regulator (CFTR)-knockout rats. PLoS One 2014; 9(3): e91253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [35].Domvri K, Zarogoulidis P, Porpodis K, et al. Gene therapy in liver diseases: state-of-the-art and future perspectives. Curr Gene Ther 2012; 12(6): 463–83. [DOI] [PubMed] [Google Scholar]
  • [36].Mearini G, Stimpel D, Geertz B, et al. Mybpc3 gene therapy for neonatal cardiomyopathy enables long term disease prevention in mice. Nat Commun 2014; 5: 5515. [DOI] [PubMed] [Google Scholar]
  • [37].Shanks N, Greek R, Greek J. Are animal models predictive for humans? Philos Ethics Humanit Med 2009; 4: 2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [38].Vasireddy V, Mills JA, Gaddameedi R, et al. AAV-mediated gene therapy for choroideremia: preclinical studies in personalized models. PLoS One 2013; 8(5): e61396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [39].Barde I, Laurenti E, Verp S, et al. Lineage- and stage-restricted lentiviral vectors for the gene therapy of chronic granulomatous disease. Gene Ther 2011; 18(11): 1087–97. [DOI] [PubMed] [Google Scholar]
  • [40].Boutin S, Monteilhet V, Veron P, et al. Prevalence of serum IgG and neutralizing factors against adeno-associated virus (AAV) types 1, 2, 5, 6, 8, and 9 in the healthy population: implications for gene therapy using AAV vectors. Hum Gene Ther 2010; 21(6): 704–12. [DOI] [PubMed] [Google Scholar]
  • [41].Gray SJ, Matagne V, Bachaboina L, et al. Preclinical differences of intravascular AAV9 delivery to neurons and glia: a comparative study of adult mice and nonhuman primates. Mol Ther 2011; 19(6): 1058–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].Yue Y, Ghosh A, Long C, et al. A single intravenous injection of adeno-associated virus serotype-9 leads to whole body skeletal muscle transduction in dogs. Mol Ther 2008; 16(12): 1944–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [43].Ghosh A, Yue Y, Long C, et al. Efficient whole-body transduction with trans-splicing adeno-associated viral vectors. Mol Ther 2007; 15(4): 750–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [44].Rapti K, Louis-Jeune V, Kohlbrenner E, et al. Neutralizing antibodies against AAV serotypes 1, 2, 6, and 9 in sera of commonly used animal models. Mol Ther 2012; 20(1): 73–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [45].Koch S, Sothilingam V, Garrido GM, et al. Gene therapy restores vision and delays degeneration in the CNGB1 (−/−) mouse model of retinitis pigmentosa. Hum Mol Genet 2012; 21(20): 4486–96. [DOI] [PubMed] [Google Scholar]
  • [46].Acland GM, Aguirre GD, Ray J, et al. Gene therapy restores vision in a canine model of childhood blindness. Nat Genet 2001; 28(1): 92–5. [DOI] [PubMed] [Google Scholar]
  • [47].Acland GM, Aguirre GD, Bennett J, et al. Long-term restoration of rod and cone vision by single dose rAAV-mediated gene transfer to the retina in a canine model of childhood blindness. Mol Ther 2005; 12(6): 1072–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [48].Pang JJ, Chang B, Kumar A, et al. Gene therapy restores vision-dependent behavior as well as retinal structure and function in a mouse model of RPE65 Leber congenital amaurosis. Mol Ther 2006; 13(3): 565–72. [DOI] [PubMed] [Google Scholar]
  • [49].Maguire AM, Simonelli F, Pierce EA, et al. Safety and efficacy of gene transfer for Leber’s congenital amaurosis. N Engl J Med 2008; 358(21): 2240–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [50].Bainbridge JW, Smith AJ, Barker SS, et al. Effect of gene therapy on visual function in Leber’s congenital amaurosis. N Engl J Med 2008; 358(21): 2231–9. [DOI] [PubMed] [Google Scholar]
  • [51].Hauswirth WW, Aleman TS, Kaushal S, et al. Treatment of leber congenital amaurosis due to RPE65 mutations by ocular subretinal injection of adeno-associated virus gene vector: short-term results of a phase I trial. Hum Gene Ther 2008; 19(10): 979–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [52].Jesus BBD, Vera E, Schneeberger K, et al. Telomerase gene therapy in adult and old mice delays aging and increases longevity without increasing cancer. EMBO Mol Med 2012; 4(8): 691–704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [53].Damas AP, Valero J, Chen M, et al. Crtc1 activates a transcriptional program deregulated at early Alzheimer’s disease-related stages. J Neurosci 2014; 34(17): 5776–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [54].Felsburg PJ, Somberg RL, Hartnett BJ, et al. Canine X-linked severe combined immunodeficiency. A model for investigating the requirement for the common gamma chain (gamma c) in human lymphocyte development and function. Immunol Res 1998; 17(1–2): 63–73. [DOI] [PubMed] [Google Scholar]
  • [55].Weber M, Rabinowitz J, Provost N, et al. Recombinant adeno-associated virus serotype 4 mediates unique and exclusive long-term transduction of retinal pigmented epithelium in rat, dog, and nonhuman primate after subretinal delivery. Mol Ther 2003; 7(6): 774–81. [DOI] [PubMed] [Google Scholar]
  • [56].Sokolic RA, Bauer TR, Gu YC, et al. Nonmyeloablative conditioning with busulfan before matched littermate bone marrow transplantation results in reversal of the disease phenotype in canine leukocyte adhesion deficiency. Biol Blood Marrow Transplant 2005; 11(10): 755–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [57].Wang Z, Kuhr CS, Allen JM, et al. Sustained AAV-mediated dystrophin expression in a canine model of Duchenne muscular dystrophy with a brief course of immunosuppression. Mol Ther 2007; 15(6): 1160–6. [DOI] [PubMed] [Google Scholar]
  • [58].Willett K, Bennett J. Immunology of AAV-mediated gene transfer in the eye. Front Immunol 2013; 4: 261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [59].Zhou R, Caspi RR. Ocular immune privilege. F1000 Biol Rep 2010; 2 pii: 3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [60].Bauer TR Jr, Allen JM, Hai M, et al. Successful treatment of canine leukocyte adhesion deficiency by foamy virus vectors. Nat Med 2008; 14(1): 93–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [61].Bauer TR Jr, Tuschong LM, Calvo KR, et al. Long-term follow-up of foamy viral vector-mediated gene therapy for canine leukocyte adhesion deficiency. Mol Ther 2013; 21(5): 964–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [62].Renshaw HW, Chatburn C, Bryan GM, et al. Canine granulocytopathy syndrome: neutrophil dysfunction in a dog with recurrent infections. J Am Vet Med Assoc 1975; 166(5): 443–7. [PubMed] [Google Scholar]
  • [63].Du LM, Nurden P, Nurden AT, et al. Platelet-targeted gene therapy with human factor VIII establishes haemostasis in dogs with haemophilia A. Nat Commun 2013; 4: 2773. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [64].Cantore A, Ranzani M, Bartholomae CC, et al. Liver-directed lentiviral gene therapy in a dog model of hemophilia B. Sci Transl Med 2015; 7(277): 277ra28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [65].Mowat FM, Breuwer AR, Bartoe JT, et al. RPE65 gene therapy slows cone loss in RPE65 deficient dogs. Gene Ther 2013; 20(5): 545–55. [DOI] [PubMed] [Google Scholar]
  • [66].Wu Z, Hiriyanna S, Qian H, et al. A long-term efficacy study of gene replacement therapy for RPGR-associated retinal degeneration. Hum Mol Genet 2015; 24(14): 3956–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [67].Moulay G, Ohtani T, Ogut O, et al. Cardiac AAV9 gene delivery strategies in adult canines: assessment by long-term serial SPECT imaging of sodium iodide symporter expression. Mol Ther 2015; 23(7): 1211–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [68].Pan X, Yue Y, Zhang K, et al. AAV-8 Is More efficient than AAV-9 in transducing neonatal dog heart. Hum Gene Ther Methods 2015; 26(2): 54–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [69].Beltran WA, Cideciyan AV, Lewin AS, et al. Gene therapy rescues photoreceptor blindness in dogs and paves the way for treating human X-linked retinitis pigmentosa. Proc Natl Acad Sci USA 2012; 109(6): 2132–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [70].Narfström K, Wrigstad A, Nilsson SE. The Briard dog: a new animal model of congenital stationary night blindness. Br J Ophthalmol 1989; 73(9): 750–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [71].Komáromy AM, Varner SE, de Juan E, Acland GM, Aguirre GD. Application of a new subretinal injection device in the dog. Cell Transplant 2006; 15(6): 511–9. [DOI] [PubMed] [Google Scholar]
  • [72].Gearhart PM, Gearhart CC, Petersen-Jones SM. A novel method for objective vision testing in canine models of inherited retinal disease. Invest Ophthalmol Vis Sci 2008; 49(8): 3568–76. [DOI] [PubMed] [Google Scholar]
  • [73].Narfström K, Vaegan, Katz M, Bragadottir R, Rakoczy EP, Seeliger M. Assessment of structure and function over a 3-year period after gene transfer in RPE65−/− dogs. Doc Ophthalmol 2005; 111(1): 39–48. [DOI] [PubMed] [Google Scholar]
  • [74].Callejas D, Mann CJ, Ayuso E, et al. Treatment of diabetes and long-term survival after insulin and glucokinase gene therapy. Diabetes 2013; 62(5): 1718–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [75].Childers MK, Joubert R, Poulard K, et al. Gene therapy prolongs survival and restores function in murine and canine models of myotubular myopathy. Sci Transl Med 2014; 6(220): 220ra10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [76].Magalhaes I, Vudattu NK, Ahmed RK, et al. High content cellular immune profiling reveals differences between rhesus monkeys and men. Immunology 2010; 131(1): 128–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [77].Shen S, Pyo CW, Vu Q, Wang R, Geraghty DE. The essential detail: the genetics and genomics of the primate immune response. ILAR J 2013; 54(2): 181–95. [DOI] [PubMed] [Google Scholar]
  • [78].Mingozzi F and High KA. Immune responses to AAV vectors: overcoming barriers to successful gene therapy. Blood 2013; 122(1): 23–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [79].Poirier N, Mary C, Le Bas-Bernardet S, et al. Advantages of Papio anubis for preclinical testing of immunotoxicity of candidate therapeutic antagonistantibodies targeting CD28. MAbs 2014; 6(3): 697–707. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [80].Niu Y, Shen B, Cui Y, et al. Generation of gene-modified cynomolgus monkey via Cas9/RNA-mediated gene targeting in one-cell embryos. Cell 2014; 156(4): 836–43. [DOI] [PubMed] [Google Scholar]
  • [81].Liu X, Luo M, Trygg C, et al. Biological differences in rAAV transduction of airway epithelia in humans and in old world non-human primates. Mol Ther 2007; 15(12): 2114–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [82].Farrow N, Miller D, Cmielewski P, Donnelley M, Bright R, Parsons DW. Airway gene transfer in a non-human primate: Lentiviral gene expression in marmoset lungs. Sci Rep 2013; 3: 1287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [83].Nagahara AH, Bernot T, Moseanko R, et al. Long-term reversal of cholinergic neuronal decline in aged non-human primates by lentiviral NGF gene delivery. Exp Neurol 2009; 215(1): 153–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [84].Leung CH, Kliem MA, Heeke BL, et al. Assessment of hippocampal adeno-associated viral vector gene delivery via frameless stereotaxis in a nonhuman primate. Stereotact Funct Neurosurg 2011; 89(5): 275–85. [DOI] [PubMed] [Google Scholar]
  • [85].Marks WJ Jr, Ostrem JL, Verhagen L, et al. Safety and tolerability of intraputaminal delivery of CERE-120 (adeno-associated virus serotype 2-neurturin) to patients with idiopathic Parkinson’s disease: an open-label, phase I trial. Lancet Neurol 2008; 7(5): 400–8. [DOI] [PubMed] [Google Scholar]
  • [86].Mancuso K, Hauswirth WW, Li Q, et al. Gene therapy for redgreen colour blindness in adult primates. Nature 2009; 461(7265): 784–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [87].Shultz LD, Ishikawa F, Greiner DL. Humanized mice in translational biomedical research. Nat Rev Immunol 2007; 7(2): 118–30. [DOI] [PubMed] [Google Scholar]
  • [88].Mohty B and Mohty M. Long-term complications and side effects after allogeneic hematopoietic stem cell transplantation: an update. Blood Cancer J 2011; 1(4): e16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [89].Tanner A, Taylor SE, Decottignies W, Berges BK. Humanized mice as a model to study human hematopoietic stem cell transplantation. Stem Cells Dev 2014; 23(1): 76–82. [DOI] [PubMed] [Google Scholar]
  • [90].Coughlan AM, Freeley SJ, Robson MG. Humanised mice have functional human neutrophils. J Immunol Methods 2012; 385(1–2): 96–104. [DOI] [PubMed] [Google Scholar]
  • [91].Joseph A, Zheng JH, Chen K, et al. Inhibition of in vivo HIV infection in humanized mice by gene therapy of human hematopoietic stem cells with a lentiviral vector encoding a broadly neutralizing anti-HIV antibody. J Virol 2010; 84(13): 6645–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [92].Perumbeti A, Higashimoto T, Urbinati F, et al. A novel human gamma-globin gene vector for genetic correction of sickle cell anemia in a humanized sickle mouse model: critical determinants for successful correction. Blood 2009; 114(6): 1174–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [93].Frecha C, Costa C, Nègre D, et al. A novel lentiviral vector targets gene transfer into human hematopoietic stem cells in marrow from patients with bone marrow failure syndrome and in vivo in humanized mice. Blood 2012; 119(5): 1139–50. [DOI] [PubMed] [Google Scholar]
  • [94].Kassim SH, Li H, Vandenberghe LH, et al. Gene therapy in a humanized mouse model of familial hypercholesterolemia leads to marked regression of atherosclerosis. PLoS One 2010; 5(10): e13424. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [95].Vatakis DN, Bristol GC, Kim SG, et al. Using the BLT humanized mouse as a stem cell based gene therapy tumor model. J Vis Exp 2012; (70): e4181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [96].Anderson J, Li MJ, Palmer B, et al. Safety and efficacy of a lentiviral vector containing three anti-HIV genes--CCR5 ribozyme, tatrev siRNA, and TAR decoy--in SCID-hu mouse-derived T cells. Mol Ther 2007; 15(6): 1182–8. [DOI] [PubMed] [Google Scholar]
  • [97].Krauss JC, Bond LM, Todd RF, Willson JM. Expression of retroviral transduced human CD18 in murine cells: An in vitro model of gene therapy for leukocyte adhesion deficiency. Hum Gene Ther 1991; 2(3): 221–8. [DOI] [PubMed] [Google Scholar]
  • [98].Azuma H, Paulk N, Ranade A, et al. Robust expansion of human hepatocytes in Fah−/−/Rag2−/−/Il2rg−/− mice. Nat Biotechnol 2007; 25(8): 903–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [99].Lisowski L, Dane AP, Chu K, et al. Selection and evaluation of clinically relevant AAV variants in a xenograft liver model. Nature 2014; 506(7488): 382–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [100].Kashiwakura Y, Mimuro J, Onishi A. Porcine model of hemophilia A. PLoS ONE 2012; 7(11). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [101].Swindle MM, Makin A, Herron AJ, Clubb FJ, Frazier KS Jr Swine as models in biomedical research and toxicology testing. Vet Pathol 2011; 49(2): 344–56. [DOI] [PubMed] [Google Scholar]
  • [102].Gu G and Kues WA. Current progress of genetically engineered pig models for biomedical research. Bio Res Open Access 2014; 3(6): 255–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [103].Raake PWJ, Schlegel P, Ksienzyk J, et al. AAV6.bARKct cardiac gene therapy ameliorates cardiac function and normalizes the catecholaminergic axis in a clinically relevant large animal heart failure model. Eur Heart J 2011; 34, 1437–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [104].Shears LL II, Kibbe MR, Murdock AD, et al. Efficient inhibition of intimal hyperplasia by adenovirus-mediated inducible nitric oxide synthase gene transfer to rats and pigs in vivo. J Am Coll Surg 1998; 187(3): 295–306. [DOI] [PubMed] [Google Scholar]
  • [105].Laguens R, Meckert PC, Janavel GV, et al. Entrance in mitosis of adult cardiomyocytes in ischemic pig hearts after plasmid-mediated rhVEGF165 gene transfer. Gene Ther 2002; 9: 1676–81. [DOI] [PubMed] [Google Scholar]
  • [106].Pleger ST, Shan C, Ksienzyk J, et al. Cardiac AAV9-S100A1 gene therapy rescues post-ischemic heart failure in a preclinical large animal model. Sci Transl Med 2011; 3(92): 92ra64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [107].Colell P, Trapani I, Cesi G, et al. Efficient gene delivery to the cone-enriched pig retina by dual AAV vectors. Gene Ther 2014; 21: 450–6. [DOI] [PubMed] [Google Scholar]
  • [108].Potash AE, Wallen TJ, Karp PH, et al. Adenoviral gene transfer corrects the ion transport defect in the sinus epithelia of a porcine CF model. Mol Ther 2013; 21(5): 947–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [109].Arnold WD, Burghes AH. Spinal muscular atrophy: development and implementation of potential treatments. Ann Neurol 2013; 743: 348–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [110].Lorson CL, Hahnen E, Androphy EJ, et al. A single nucleotide in the SMN gene regulates splicing and is responsible for spinal muscular atrophy. Proc Natl Acad Sci U S A 1999; 9611: 6307–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [111].Monani UR, Sendtner M, Coovert DD et al. The human centromeric survival motor neuron gene (SMN2) rescues embryonic lethality in Smn−/− mice and results in a mouse with spinal muscular atrophy. Hum Mol Gent 2000; 9(3): 333–9. [DOI] [PubMed] [Google Scholar]
  • [112].Valori CF, Ning K, Wyles M, et al. Systemic delivery of scAAV9 expressing SMN prolongs survival in a model of spinal muscular atrophy. Sci Transl Med 2010; 235: 35ra42. [DOI] [PubMed] [Google Scholar]
  • [113].Foust KD, Wang X, McGovern VL, et al. Rescue of the spinal muscular atrophy phenotype in a mouse model by early postnatal delivery of SMN. Nat Biotechnol 2010; 283: 271–4. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • [114].Passini MA, Bu J, Roskelley EM, et al. CNS-targeted gene therapy improves survival and motor function in a mouse model of spinal muscular atrophy. J Clin Invest 2010; 1204: 1253–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [115].Hsieh-Li HM, Chang JG, Jong YJ, et al. A mouse model for spinal muscular atrophy. Nat Genet 2000; 241: 66–70. [DOI] [PubMed] [Google Scholar]
  • [116].Palladino A, Passamano L, Taglia A, et al. Cardiac involvement in patients with spinal muscular atrophies. Acta Myol 2011; 303: 175–8. [PMC free article] [PubMed] [Google Scholar]
  • [117].Bevan AK, Hutchinson KR, Foust KD, et al. Early heart failure in the SMNDelta7 model of spinal muscular atrophy and correction by postnatal scAAV9-SMN delivery. Hum Mol Genet 2010; 1920: 3895–905. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [118].Duque SI, Arnold WD, Odermatt P, et al. A Large Animal Model of Spinal Muscular Atrophy and Correction of Phenotype. ANN NEUROL 2015; 77: 399–414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [119].Bosze Z, Houdebine LM. Application of rabbits in biomedical research: a review. World Rabbit Sci. 2006; 14: 1–14. [Google Scholar]
  • [120].Waugh JM, Kattash M, Li J, et al. Gene therapy to promote thromboresistance: Local overexpression of tissue plasminogen activator to prevent arterial thrombosis in an in vivo rabbit model. Proc Natl Acad Sci U S A 1999; 96(3): 1065–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [121].Simon DI, Ezratty AM, Francis SA, Rennke H, Loscalzo. Fibrinogen is internalized and degraded by activated human monocytoid cells via Mac-1 (CD11 b/CD18): A nonplasmin fibrinolytic pathway. Blood 1993; 82: 2414–22. [PubMed] [Google Scholar]
  • [122].Rekhter MD, Shah N, Simari RD, et al. Graft permeabilization Facilitates Gene Therapy of Transplant Arteriosclerosis in a Rabbit Model. Circulation 1998; 98: 1335–41. [DOI] [PubMed] [Google Scholar]
  • [123].Taniyama Y, Morishita R, Aoki M, et al. Therapeutic angiogenesis induced by human hepatocyte growth factor gene in rat and rabbit hind limb ischemia models: preclinical study for treatment of peripheral arterial disease. Gene Ther 2001; 8: 181–9. [DOI] [PubMed] [Google Scholar]
  • [124].Mohan RR, Tovey JCK, Sharma A, Schultz GS, Cowden JW, Tandon A. Targeted Decorin gene therapy delivered with adeno-associated virus effectively retards corneal neovascularization in vivo. PLoS ONE 2011; 6(10): e26432. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [125].Favaro P, Finn JD, Siner JI, Wright JF, High KA, Arruda VR. Safety of liver gene transfer following peripheral intravascular delivery of adeno-associated virus (AAV)-5 and AAV-6 in a large animal model. Hum Gene Ther 2011; 22(7): 843–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [126].Casal M, Haskins M. Large animal models and gene therapy. Eur J Hum Genet 2006; 14: 266–72. [DOI] [PubMed] [Google Scholar]
  • [127].Berman AL: The Brainstem of the Cat: A Cytoarchitechtonic Atlas with Sterotactic Coordinates. Madison, WI: University of Wisconsin Press; 1968. [Google Scholar]
  • [128].Berman AL: The Thalamus and Basal Telencephalon of the Cat: A Cytoarchitechtonic Atlas with Sterotactic Coordinates. Madison, WI: University of Wisconsin Press; 1982. [Google Scholar]
  • [129].Arfi A, Bourgoin C, Basso L, Emiliani C, Tancini B, Chigorno V. Bicistronic lentiviral vector corrects beta-hexosaminidase deficiency in transduced and cross-corrected human Sandhoff fibroblasts. Neurobiol Dis 2005; 20: 583–93. [DOI] [PubMed] [Google Scholar]
  • [130].Guidotti JE, Mignon A, Haase G, Caillaud C, McDonell N, Kahn A. Adenoviral gene therapy of the Tay-Sachs disease in hexosaminidase A-deficient knock-out mice. Hum Mol Genet 1999; 8: 831–8. [DOI] [PubMed] [Google Scholar]
  • [131].Itakura T, Kuroki A, Ishibashi Y, Tsuji D, Kawashita E, Higashine Y. Inefficiency in GM2 ganglioside elimination by human lysosomal beta-hexosaminidase beta-subunit gene transfer to fibroblastic cell line derived from Sandhoff disease model mice. Biol Pharm Bull 2006; 29: 1564–9. [DOI] [PubMed] [Google Scholar]
  • [132].Bradbury AM, Cochran JN, Mccurdy VJ, et al. Therapeutic response in feline Sandhoff disease despite immunity to intracranial gene therapy. Mol Ther 2013; 21(7): 1306–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [133].Lee B, Dennis JA, Healy PJ, et al. Hepatocyte gene therapy in a large animal: A neonatal bovine model of citrullinemia. Proc Natl Acad Sci USA 1999; 96: 3981–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [134].Frisbie DD, Ghivizzani SC, Robbins PD, Evans CH, McIlwraith CW. Treatment of experimental equine osteoarthritis by in vivo delivery of the equine interleukin-1 receptor antagonist gene. Gene Ther 2002; 9: 12–20. [DOI] [PubMed] [Google Scholar]
  • [135].Evans CH, Gouze JH, Gouze E, Robbins PD, Ghivizzani SC. Osteoarthritis gene therapy. Gene Ther 2004; 11: 379–89. [DOI] [PubMed] [Google Scholar]

RESOURCES