Next Article in Journal
A Novel Vitronectin Peptide Facilitates Differentiation of Oligodendrocytes from Human Pluripotent Stem Cells (Synthetic ECM for Oligodendrocyte Differentiation)
Previous Article in Journal
Genome-Wide Identification and Expression Analysis of Potential Antiviral Tripartite Motif Proteins (TRIMs) in Grass Carp (Ctenopharyngodon idella)
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Animal Models in Human Adenovirus Research

Leibniz Institute for Experimental Virology (HPI), Martinistr. 52, 20251 Hamburg, Germany
*
Authors to whom correspondence should be addressed.
Biology 2021, 10(12), 1253; https://doi.org/10.3390/biology10121253
Submission received: 11 November 2021 / Revised: 24 November 2021 / Accepted: 30 November 2021 / Published: 1 December 2021
(This article belongs to the Special Issue Mechanisms of Adenovirus Pathogenesis)

Abstract

:

Simple Summary

Animal models are widely used to study various aspects of human diseases and disorders. Likewise, they are indispensable for preclinical testing of medicals and vaccines. Human adenovirus infections are usually self-limiting, and can cause mild respiratory symptoms with fever, eye infection or gastrointestinal symptoms, but occasional local outbreaks with severe disease courses have been reported. In addition, adenovirus infections pose a serious risk for children and patients with a weakened immune system. Human adenovirus research in animal models to study adenovirus-induced disease and tumor development started in the 1950s. Various animal species have been tested for their susceptibility to human adenovirus infection since then, and some have been shown to mimic key characteristics of the infection in humans, including persistent infection. Furthermore, some rodent species have been found to develop tumors upon human adenovirus infection. Our review summarizes the current knowledge on animal models in human adenovirus research, describing the pros and cons along with important findings and future perspectives.

Abstract

Human adenovirus (HAdV) infections cause a wide variety of clinical symptoms, ranging from mild upper respiratory tract disease to lethal outcomes, particularly in immunocompromised individuals. To date, neither widely available vaccines nor approved antiadenoviral compounds are available to efficiently deal with HAdV infections. Thus, there is a need to thoroughly understand HAdV-induced disease, and for the development and preclinical evaluation of HAdV therapeutics and/or vaccines, and consequently for suitable standardizable in vitro systems and animal models. Current animal models to study HAdV pathogenesis, persistence, and tumorigenesis include rodents such as Syrian hamsters, mice, and cotton rats, as well as rabbits. In addition, a few recent studies on other species, such as pigs and tree shrews, reported promising data. These models mimic (aspects of) HAdV-induced pathological changes in humans and, although they are relevant, an ideal HAdV animal model has yet to be developed. This review summarizes the available animal models of HAdV infection with comprehensive descriptions of virus-induced pathogenesis in different animal species. We also elaborate on rodent HAdV animal models and how they contributed to insights into adenovirus-induced cell transformation and cancer.

1. Introduction

Animal models are of the utmost importance for basic and applied research to not only expand and verify in vitro findings, but also to study diseases and therapies in physiological settings. In infectious diseases research, they contributed to fundamental insights into disease pathogenesis, and are essential to study mode-of-action and efficacies of anti-infectives and prophylactic treatments [1]. An ideal animal model recapitulates a particular disease or condition in a non-human organism in terms of key phenotypic, pathophysiologic, and histopathological characteristics, as well as response to treatment [2,3]. Animal models have greatly improved our knowledge on the course of viral infections including disease, associated pathology, persistence, and viral transformation in human adenovirus (HAdV) infections [4,5]. Moreover, preclinical evaluation of antiadenoviral treatment options, adenovirus-based therapeutics, and vaccines rely on data from experiments in relevant in vivo settings, as is happening currently with COVID-19 vaccines [6]. Here, different HAdV types and simian adenoviruses serve as excellent vaccine vectors, and are used to potentially fight off zoonotic diseases such as AIDS, malaria, and Ebola, besides COVID-19 [7]. In vivo studies on adenoviral vectors in general and their use in vaccine- and gene therapy approaches in particular have been extensively reviewed. The same applies to oncolytic adenoviruses in animal models that are not part of this work, but were reviewed by others recently [8,9,10,11,12]. This review outlines the current knowledge on HAdV susceptibility of different animal models, highlighting key features, strengths, and limitations. We set the focus on studies that predominantly report adenovirus pathogenesis and adenovirus-induced cell transformation.

2. Main Text

2.1. Adenovirus Disease in Humans and the Importance of Animal Models in Adenovirus Research

HAdVs belong to the genus Mastadenovirus, in the virus family Adenoviridae. They are grouped into different species (A to G), and subsequently classified in more than 100 different types, based on viral sequences and serological data [13]. HAdV infections of humans are common, with high seroprevalences worldwide [14]. These non-enveloped, double-stranded DNA viruses have genome sizes of approximately 24–48 kb, depending on the type [15].
Clinical symptoms in infected individuals vary, and there are currently no widely available vaccines nor approved specific antiadenoviral compounds available to prevent and treat HAdV infections [16]. While immunocompetent individuals develop rather mild symptoms, infections of pediatric or immunocompromised patients can cause severe and sometimes lethal disease [17]. Furthermore, different HAdV types have different tissue tropisms, and infections of the respiratory, gastrointestinal, and urinary tracts, as well as the eyes, have been reported [17]. The tissue tropism largely correlates with clinical signs in humans that range from respiratory disease, conjunctivitis, and gastroenteritis of different severities. Moreover, some types are discussed to be risk factors for obesity, as extensively reviewed elsewhere [18,19]. To understand HAdV pathogenesis and infection-related consequences, researchers have been on a ~70 year-long quest for suitable animal models that phenocopy HAdV infection of humans. These models included various rodents including xenotransplanted and genetically engineered transgenic mice and hamsters, pigs, non-human primates, and other species. All these models facilitate HAdV replication and induction of HAdV disease signs to different extents, and are employed to recapitulate aspects of the human disease. However, an ideal model that mimics the human disease has not been established to date, partly due to the species-specific nature of adenoviruses. The search remains ongoing as key findings from carefully designed and well-controlled studies in animal models continue to be of great importance to biomedical research [20].

2.2. First Experimental HAdV Infections of Animals

The first experimental infections of animals with HAdVs date back to the 1950s and 1960s [21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36]. All of these studies confirmed findings by Rowe and colleagues, who did not observe clinical disease symptoms in experimentally infected rabbits, mice, hamsters, Guinea pigs, cats, ferrets, rats, and even non-human primates (Figure 1) [21]. Another follow-up study also reported asymptomatic persistent infection of rabbits [22]. Interestingly, they detected persistent HAdV-C5 in experimentally infected adult rabbits at 8 weeks p.i. by virus re-isolation from spleen homogenates. Subsequent approaches evaluated HAdV-induced tumors in rodents as described below [26,28,29,30,31,32,33,34,35], along with the susceptibility of dogs and even pigs to different HAdV types using different infectious doses, but none of these animals proved to be suitable to study HAdV infection [23,24,25].

2.3. Syrian Hamsters

(Golden) Syrian hamsters (Mesocricetus auratus) are medium-sized rodents that are frequently used as animal model in various fields of research. Moreover, a decent number of molecular tools and reagents are available to study host response to infection in this hamster species [42]. They are susceptible to a wide variety of DNA and RNA viruses [42], and are used as an adenovirus animal model since 1962 (Table 1) [26,27,28,29]. The Syrian hamster model is the most commonly used HAdV animal model that has successfully been applied to studies on basic HAdV pathogenesis, and especially tumorigenesis, countermeasure and (oncolytic) vector development [43]. The first records on the HAdV-susceptibility of Syrian hamsters originate from the a study performed by Trentin and colleagues, who showed that HAdV-A12 induced sarcomas over a course of 33–90 days post-infection (Figure 1) [28]. These results were confirmed by three more publications in the same year that additionally demonstrated tumor induction by HAdV-A18, as well as fatal infection of newborn hamsters with high infectious doses of HAdV-C5 [26,27,29]. Subsequent studies further corroborated these data, and also reported oncogenic properties of HAdV-B3 and B7 [30,34,44,45,46]. Much is known about the mechanisms of adenoviral tumor induction and cell transformation in vitro, but the subjects still are a matter of investigation. It is discussed that some HAdVs (such as A12, A18, B3, and B7) lead to abortive infection, and subsequently to cell transformation in hamsters, while other HAdVs replicate and cause cell pathology in these animals. Nevertheless, adenoviral cell transformation in general and HAdV-induced tumors in Syrian hamsters in particular represent an excellent system for DNA virus tumorigenesis, and built the cornerstone for various following reports on the development of adenoviral vectors that have been reviewed extensively [47,48,49,50]. Intriguingly, other hamster species have also been shown to be susceptible to HAdV-induced tumors. A single 1974 study describes HAdV-A12-induced tumor development in newborn Armenian (Cricetulus migratorius) and Chinese hamsters (Cricetulus griseus) [51].
Syrian hamsters that were used in HAdV-C5 and C6 pathogenesis studies were found to have viral titers in blood and organ samples, and serologic as well as histologic evidence of infection [52,53,54,55,56,57]. Besides HAdV-C5 and C6 replication in the lungs and other organs, the virus especially targets the liver (hepatocytes, Kupffer cells) to cause inflammation and hepatocellular necrosis, accompanied by elevated levels of liver enzymes. Weight loss as a robust clinical sign of infection has only been observed in chemically immunosuppressed Syrian hamsters that showed transiently reduced body weights upon HAdV-C5 infection [55,56]. Interestingly, male Syrian hamsters seem to be more susceptible than females, which has also been observed for other viral infections like COVID19 lately [57,58,59].
In recent studies, Radke and colleagues reported that HAdV-B14 and an emerging variant, B14p1, cause severe lung pathogenesis in intratracheally infected in Syrian hamsters characterized by local infiltrations of inflammatory cells developing into bronchopneumonia. However, although the authors did not report clinically apparent disease signs, these studies in conjunction with the studies that Tollefson and colleagues performed in Syrian hamsters allowed for comparisons of the pathogenicity of the different HAdVs and assessment of respective immune responses [55,56,60,61].
Toth and colleagues introduced the genetically modified Syrian hamster model in 2015 (Figure 1) [39]. These STAT2 knockout animals [62] show decreased interferon signaling, which facilitates higher HAdV-C5 replication, more severe liver pathology, and increased mortality when compared to wild type Syrian hamsters upon intravenous infection, and thereby resemble HAdV infection of immunosuppressed humans. Moreover, the interferon immune response was deregulated in infected STAT2 knockout hamsters [39]. Both wild type and genetically modified hamsters have been proven useful for efficacy studies on HAdV therapeutics and vaccines, as well as on vector development [43,53,54,55,63,64,65,66,67,68,69,70,71,72,73].

2.4. Cotton Rats

Cotton rats are small rodents that are susceptible to various human pathogens, and especially to upper respiratory tract infections where mice and rats are typically rather resistant [74,75]. The first report on hispid cotton rats (Sigmodon hispidus) as an HAdV animal model dates back to 1984 when Pacini and colleagues used moderate HAdV-C5 titers to infect one-month-old animals, and detected viral titers in the lungs and nasal mucosa and seroconversion as soon as six days post-infection (Figure 1, Table 1) [37]. Histopathological examinations of lung samples from infected cotton rats revealed transient peribronchial immune cell infiltration and other subtle signs of pneumonia [37]. No clinical symptoms were observed by Pacini and colleagues, but dose-dependent disease has been reported in a follow-up study, in which high HAdV-C5 doses led to more severe lung damage, and high-dose-infected animals died within the first week post-infection [76].
Cotton rats are also employed as animal models for the adenovirus-induced eye disease adenoviral epidemic keratoconjunctivitis (EKC) [78,79,80]. EKC is highly contagious, and characterized by eye inflammation and visual disturbances caused by corneal opacities; EKC outbreaks occur regularly [81,82]. Ocular HAdV-C5 and D8-infection of cotton rats show hallmarks of the human disease, including virus replication and development of the aforementioned subepithelial corneal opacities [78,79,80].
This HAdV-animal model is susceptible to HAdV infection, and even resembles human EKC. Consequently, cotton rats have been used in various approaches to test therapeutic interventions and to study oncolytic adenoviruses [65,79,80,83,84,85,86].

2.5. New Zealand Rabbits

Rabbits have served as models of human infectious diseases and ophthalmology for decades [87,88]. New Zealand (NZ) rabbits (Oryctolagus cuniculus) have been used as an animal model species for investigating persistent [22,36] and ocular HAdV infection (Table 1) [38,89,90,91,92,93,94,95]. Both papers reporting prolonged HAdV-C5 infections observed persistence of the virus in spleens for months [22,36], as well as presence of anti-HAdV-C5 antibodies in rabbit sera for up to a year post-infection [36]. The initial study on HAdV-C5-infected rabbits as a model for basic and preclinical research on EKC was published by Gordon and colleagues in 1992 (Figure 1) [38]. Through intraocular infection of female NZ rabbits, they demonstrated that several features of human adenoviral eye infection can be reproduced in this animal model, such as acute conjunctivitis, iritis, and corneal edema, followed by infiltrating immune cells at around 14 days post-infection. In addition, infected rabbits mounted a neutralizing antibody response, and confirmed previous findings that rabbits respond to infection via the humoral immune response. This ocular model of HAdV-C5 infection has been continuously used since then to assess efficacies topical use of the broad spectrum antiviral cidofovir and other treatment options against adenoviral eye infections [89,90,91,92,93,94,95]. It is very likely that NZ rabbits will serve in future studies that investigate the pathology and countermeasures against adenovirus-induced human EKC.

2.6. Rats

Rats (Rattus norvegicus) are commonly used as laboratory animal models that mimic aspects of human diseases or the human organism, especially in oncology, pharmacology, and toxicology research [96,97]. As described for other rodents above, rats also develop tumors upon HAdV infection. That was first described by Huebner and colleagues in 1963, who showed that HAdV-A12 infections induce peritoneal tumors in rats with 30% efficiency [98]. Interestingly, HAdV-A12-induced sarcomas and even retinoblastomas have been observed by other groups [32,99,100,101]. In contrast, HAdV-D9 has been shown to cause tumors in rats more efficiently, as investigated in a series of papers by Javier and colleagues. First, they showed that subcutaneous HAdV-D9 inoculation of newborn female rats led to mammary tumors during weeks to several months post-infection that exhibited three distinct phenotypes: fibroadenoma, phylloide-like tumors, and solid sarcomas. HAdV-D9-induced tumors developed, dependent on estrogen, as ovariectomized rats did not develop tumors upon infection whereas treatment with a synthetic estrogen promoted tumor formation [102]. With that, they confirmed earlier studies on HAdV-D9-related tumorigenesis [103,104]. Further analyses revealed that HAdV-D9 persisted via genomic integration in tumor cells [102]. In subsequent studies, Javier and colleagues could pinpoint the HAdV-D9 E4 ORF1 gene as the major oncogene that drives mammary tumor formation in infected rats [105,106,107]. The rat model for HAdV-infection has been used in one more study that aimed to assess the effects of HAdV-C5 major capsid protein modifications on tissue distribution within infected animals [108]. Male animals that were infected with the HAdV-C5 wild type control exhibited a hepatotropic virus dissemination with no clinical signs of an HAdV-C5-induced disease over a course of five days post-infection. Interestingly though, the study demonstrated certain penton and fiber mutations lead to viruses that are detargeted from rat livers [108].
These data indicate that rats are a good model to study HAdV-induced tumors, and possibly also adenoviral vector development, but are not suitable for pathogenesis assessment and preclinical evaluation of HAdV-vaccines and antivirals.

2.7. Mice

Non-transgenic immunocompetent laboratory mice (Mus musculus) were introduced to HAdV research in 1964, when two American laboratories concurrently showed that HAdV-A12 can induce tumors in newborn mice, comparable to those in Syrian hamsters (Figure 1) [32,33]. However, HAdV tumor induction has not been followed up upon in mice.
Comparable to the infection of Syrian hamsters, the primary target organ for HAdV replication is the mouse liver, leading to hepatocellular necrosis, and even fatal hepatitis, in HAdV-C5-infected mice, depending on the infectious dose [53,77,109,110]
Other studies reported no or only subtle viral replication, which is partly explained by the absence of the entry receptor CD46 that is used by some HAdVs [14,111,112,113]. This poses a major drawback, and prompted HAdV researchers to assess alternative models such as HAdV infection of transgenic mice [54,114,115,116] or ocular HAdV infection [117,118].
Mice are the most used animal model, with a vast variety of tools and reagents available to study nearly all aspects of host responses to infection. Thus, several studies have investigated antiviral therapies and oncolytic adenoviruses in mice [53,54,119,120,121,122,123,124,125,126], and this model will likely remain important for HAdV research.
As additional mouse models, humanized mice are routinely used to study human infectious diseases [127,128,129]. These models are generated e.g., by engraftment with functional human cells or tissues. To establish an HAdV small animal model that reproduces acute and chronic infection, Rodriguez and colleagues recently presented a humanized mouse model (Figure 1) [40]. These JAX NSG-A2 mice received HLA-A2-matching CD34+ human hematopoietic stem and progenitor cells (HSPCs), and were HAdV-C2-infected intravenously nine weeks post-transplantation. Successful infection was confirmed by the detection of adenoviral RNA in blood samples, and systemic infection was excluded for most animals, although low levels of viral mRNA could be detected in bone marrow and inguinal lymph node samples. Aside from asymptomatic infections, which are most probably due to the differing degrees of “humanization”, they observed acute infection presented as lethargy, weight loss, and even death in roughly one third of all infected NSG-A2 mice. Recorded histopathological lesions in livers of infected mice included intracytoplasmic vacuoles in hepatocytes, an increase in cell proliferation, an influx of monocytes and macrophages, and signs of fibrosis [40]. HAdV-C2 persisted in asymptomatic mice, and these mice mounted an HAdV-specific adaptive immune response.
The humanized mouse model of HAdV infection requires further evaluation, and could be a valuable alternative animal model to study HAdV persistence and reactivation.

2.8. Non-Human Primates

Due to a close phylogenetic relationship to humans, non-human primates are often used as models to understand infectious diseases of humans, as well as zoonotic and anthroponotic infections [130]. HAdV infection has been evaluated in non-human primate species with differing susceptibilities. First, experimental studies of various HAdV types from different species in intranasally, intracerebrally, and subcutaneously infected non-human primates were performed by Rowe et al., and failed to detect any symptoms of an HAdV-induced disease [21]. However, efficacy studies of adenovirus-based vaccines and gene delivery vectors provide evidence that non-human primates are susceptible to HAdV infection with no clinical signs of HAdV-induced respiratory disease reported to date [131,132,133,134,135,136]. Moreover, a single study described HAdV-A12-induced eye tumors in 3 out of 21 baboons (Papio spp.) 1 to 3 years post-infection when the virus was injected intraocularly [137].
Most recent data come from HAdV-B55 infection in tree shrews (Tupaia belangeri chinensis), squirrel-sized Asian non-human primates (Figure 1) [138]. Li and colleagues infected Chinese tree shrews intranasally, and showed that they were permissive to infection as evidenced by viral replication in the upper and lower airways and the lungs, resulting in severe pneumonia (Table 1). They additionally demonstrated that tree shrews reacted to HAdV-B55 infection with rapid seroconversion, elevated body temperatures, and upregulation of pro-inflammatory cytokines in PBMCs [41]. The tree shrew therefore poses a promising model for HAdV-induced clinical disease and research on the zoonotic potential of HAdVs that should be followed up upon, especially using other HAdVs for infection.

2.9. Pigs

Pigs (Sus scrofa) have long been excellent models for research on diseases affecting humans [139]. Many characteristics of pig anatomy and physiology resemble the situation in humans, which is why there are several advantages of the porcine model over rodents and other small animal models [140]. Their application as an animal model in HAdV research, however, has been infrequent, and can be attributed to practical reasons such as required space, costs, expense, and handling. The 1962 Betts and Jennings papers were the first to report intratracheal HAdV infection of pigs causing bronchopneumonia, accompanied by focal alveolar necrosis and lymphoid hyperplasia [24,25]. Conversely, no clinical signs were observed (Table 1) [24,25]. These initial investigations were followed up by two more recent publications, confirming the HAdV susceptibility of pigs. Intravenous HAdV-C5 injection of pigs resulted in moderate lung pathology, and viral DNA could be detected in lung, liver, kidney, and blood samples at early time points post-infection, suggesting virus replication [110]. No obvious abnormalities were detected upon histopathological examinations of liver and kidneys tissues, and clinical disease signs were absent [110]. Work by Koodie and colleagues demonstrated active replication of an HAdV-C5/B3 chimera in the lungs and spleen of intravenously infected immunocompetent pigs [141].
Collectively, these results suggest that pigs could be used as an HAdV animal model to study the disease and for countermeasure development. Future directions should include more thorough characterization of HAdV-induced pathology and immune responses in pigs.

2.10. Guinea Pigs

Guinea pigs (Cavia porcellus) are important models in human bacterial infections [142]. In addition, they are susceptible to human pathogenic viruses, such as influenza A and Zika [143,144]. An early study from 1974 describes persistent HAdV-C5 infection of guinea pigs (Table 1) [145]. After intracardial HAdV-C5 infection of male guinea pigs, Faucon and colleagues could re-isolate virus from different tissues, from blood and from spleens for long periods of times post-infection, indicating persistent infection [145]. No clinical signs or seroconversion of animals were reported in that study. A follow-up study that aimed at establishing guinea pigs as an HAdV infection model used the same virus (HAdV-C5), but at a lower infectious dose, and female guinea pigs that were infected intranasally. These animals had high viral titers and detectable viral gene expression in lung tissues early in infection, and exhibited considerable lung damage as observed by immunopathologic examination of the lungs at different time points post-infection. Moreover, the animals seroconverted to HAdV-C5 from three weeks post-infection on and persistent infections as shown by Faucon and colleagues could not be detected [146]. Notably, HAdV-induced tumor development has either not been investigated or was not reported in guinea pigs, even though they belong to the order Rodentia. These unresolved and even contradicting results combined with a lack of reagent availability explain why more research is needed to establish guinea pigs as an HAdV animal model. Nevertheless, guinea pigs have served in preclinical trials of HAdV-C5-based vector vaccines [147].

3. Conclusions and Perspectives

Basic and applied research on the various facets of HAdV infection including work in preclinical models will remain important for the next decades. Basic research will further reveal mechanisms and nuances of viral pathogenesis, persistence, and cell transformation. Applied research includes important research areas such as the testing of therapeutics and the evaluation of vector attenuation and immunogenicity, as well as (protective) efficacy of adenoviral vaccine and gene therapy vectors. Thus, the quest for an ideal small animal model that resembles key characteristics of HAdV infections will continue as, thus far, no HAdV animal model can replicate the most important aspects of HAdV infection of humans, and most available models are far from ideal (Table 2).
The Syrian hamster and cotton rat models are undoubtedly the most relevant and most utilized models to date, although they have their limitations as outlined above and summarized in Table 2. Novel approaches like the tree shrew model or infection of pigs must prove their worth.
The availability of various tools including suitable animal models to study HAdVs is particularly important as the adenoviruses are diversifying, and more and more cases of interspecies transmission pose the risk of zoonotic and anthroponotic spillover events [148,149,150,151,152,153,154,155,156]. Interesting and germane research questions that could be addressed using wild type and chimeric viruses in in vitro and in vivo studies include, but are certainly not limited to: What defines the host range of adenoviruses? How do recombination events facilitate spillover infections? How can we improve outbreak preparedness? How likely are cross-species transmission events from non-human primates to humans (or vice versa)? Notably, the non-human primate isolates belong to the genus Mastadenovirus in the same way as all HAdVs, and the adenovirus-induced disease in non-human primates is comparable to that in humans [157]. Similar to in humans, disease signs include mild to moderate respiratory and/or enteric symptoms, and even eye infections have been reported [157]. The field is beginning to understand aspects of host-range determinants, and both host and particularly viral factors seem to play important roles in this process [156,158]. The adenoviral DNA binding protein (DBP) is such a host range determinant, and future work will further unravel its role in HAdV cross-species transmission and adaptation of the virus to new hosts [158,159]. For all that, well-coordinated interdisciplinary research and a toolbox that includes appropriate HAdV animal models are crucial to thoroughly understand the HAdV-induced disease and its zoonotic potential.

Author Contributions

All authors contributed significantly to the manuscript as follows. Conceptualization, L.D.B., W.-H.I. and T.D.; writing—original draft preparation, L.D.B.; writing—review and editing, L.D.B., W.-H.I. and T.D.; visualization, L.D.B.; funding acquisition, T.D. All authors have read and agreed to the published version of the manuscript.

Funding

The Leibniz Institute for Experimental Virology (HPI) is supported by the German Federal Ministry of Health and the Freie und Hansestadt Hamburg.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Colby, L.A.; Quenee, L.E.; Zitzow, L.A. Considerations for Infectious Disease Research Studies Using Animals. Comp. Med. 2017, 67, 222–231. [Google Scholar] [PubMed]
  2. Prabhakar, S. Translational research challenges: Finding the right animal models. J. Investig. Med. 2012, 60, 1141–1146. [Google Scholar] [CrossRef] [PubMed]
  3. Mergenthaler, P.; Meisel, A. Animal Models: Value and Translational Potency. In Principles of Translational Science in Medicine, 2nd ed.; Wehling, M., Ed.; Academic Press: Cambridge, MA, USA, 2015; pp. 83–90. [Google Scholar] [CrossRef]
  4. Ruiz, S.I.; Zumbrun, E.E.; Nalca, A. Animal Models of Human Viral Diseases. In Animal Models for the Study of Human Disease, 2nd ed.; Conn, P.M., Ed.; Academic Press: Cambridge, MA, USA, 2017; pp. 853–901. [Google Scholar] [CrossRef]
  5. Tessier, T.M.; Dodge, M.J.; MacNeil, K.M.; Evans, A.M.; Prusinkiewicz, M.A.; Mymryk, J.S. Almost famous: Human adenoviruses (and what they have taught us about cancer). Tumour Virus Res. 2021, 12, 200225. [Google Scholar] [CrossRef] [PubMed]
  6. Mendonca, S.A.; Lorincz, R.; Boucher, P.; Curiel, D.T. Adenoviral vector vaccine platforms in the SARS-CoV-2 pandemic. NPJ Vaccines 2021, 6, 97. [Google Scholar] [CrossRef] [PubMed]
  7. Vrba, S.M.; Kirk, N.M.; Brisse, M.E.; Liang, Y.; Ly, H. Development and Applications of Viral Vectored Vaccines to Combat Zoonotic and Emerging Public Health Threats. Vaccines 2020, 8, 680. [Google Scholar] [CrossRef] [PubMed]
  8. Wold, W.S.; Toth, K. Adenovirus vectors for gene therapy, vaccination and cancer gene therapy. Curr. Gene Ther. 2013, 13, 421–433. [Google Scholar] [CrossRef] [PubMed]
  9. Crystal, R.G. Adenovirus: The first effective in vivo gene delivery vector. Hum. Gene Ther. 2014, 25, 3–11. [Google Scholar] [CrossRef] [Green Version]
  10. Lee, C.S.; Bishop, E.S.; Zhang, R.; Yu, X.; Farina, E.M.; Yan, S.; Zhao, C.; Zheng, Z.; Shu, Y.; Wu, X.; et al. Adenovirus-Mediated Gene Delivery: Potential Applications for Gene and Cell-Based Therapies in the New Era of Personalized Medicine. Genes Dis. 2017, 4, 43–63. [Google Scholar] [CrossRef]
  11. Goswami, R.; Subramanian, G.; Silayeva, L.; Newkirk, I.; Doctor, D.; Chawla, K.; Chattopadhyay, S.; Chandra, D.; Chilukuri, N.; Betapudi, V. Gene Therapy Leaves a Vicious Cycle. Front. Oncol. 2019, 9, 297. [Google Scholar] [CrossRef] [PubMed]
  12. McKenna, M.K.; Rosewell-Shaw, A.; Suzuki, M. Modeling the Efficacy of Oncolytic Adenoviruses In Vitro and In Vivo: Current and Future Perspectives. Cancers 2020, 12, 619. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Seto, D.; Chodosh, J.; Brister, J.R.; Jones, M.S. Members of the Adenovirus Research Community, Using the whole-genome sequence to characterize and name human adenoviruses. J. Virol. 2011, 85, 5701–5702. [Google Scholar] [CrossRef] [Green Version]
  14. Mennechet, F.J.D.; Paris, O.; Ouoba, A.R.; Salazar Arenas, S.; Sirima, S.B.; Takoudjou Dzomo, G.R.; Diarra, A.; Traore, I.T.; Kania, D.; Eichholz, K.; et al. A review of 65 years of human adenovirus seroprevalence. Expert Rev. Vaccines 2019, 18, 597–613. [Google Scholar] [CrossRef] [PubMed]
  15. Harrach, B.; Benkő, M. Adenoviruses (Adenoviridae). In Encyclopedia of Virology, 4th ed.; Bamford, D., Zuckerman, M., Eds.; Academic Press: Amsterdam, The Netherlands, 2021; Volume 2, pp. 3–16. [Google Scholar] [CrossRef]
  16. Dodge, M.J.; MacNeil, K.M.; Tessier, T.M.; Weinberg, J.B.; Mymryk, J.S. Emerging antiviral therapeutics for human adenovirus infection: Recent developments and novel strategies. Antivir. Res. 2021, 188, 105034. [Google Scholar] [CrossRef] [PubMed]
  17. Lion, T. Adenovirus infections in immunocompetent and immunocompromised patients. Clin. Microbiol. Rev. 2014, 27, 441–462. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Pasarica, M.; Dhurandhar, N.V. Infectobesity: Obesity of Infectious Origin. In Advances in Food and Nutrition Research; Taylor, S.L., Ed.; Academic Press: Cambridge, MA, USA, 2007; Volume 52, pp. 61–102. [Google Scholar] [CrossRef]
  19. Ponterio, E.; Gnessi, L. Adenovirus 36 and Obesity: An Overview. Viruses 2015, 7, 3719–3740. [Google Scholar] [CrossRef] [Green Version]
  20. Barre-Sinoussi, F.; Montagutelli, X. Animal models are essential to biological research: Issues and perspectives. Future Sci. OA 2015, 1, FSO63. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Rowe, W.P.; Huebner, R.J.; Hartley, J.W.; Ward, T.G.; Parrott, R.H. Studies of the Adenoidal-Pharyngeal-Conjunctival (APC) Group of Viruses. Am. J. Epidemiol. 1955, 61, 197–218. [Google Scholar] [CrossRef]
  22. Pereira, H.G.; Kelly, B. Latent infection of rabbits by adenovirus type 5. Nature 1957, 180, 615–616. [Google Scholar] [CrossRef] [PubMed]
  23. Sinha, S.K.; Fleming, L.W.; Scholes, S. Current considerations in public health of the role of animals in relation to human viral diseases. J. Am. Vet. Med. Assoc. 1960, 136, 481–485. [Google Scholar]
  24. Betts, A.O.; Jennings, A.R.; Lamont, P.H.; Page, Z. Inoculation of pigs with adenoviruses of man. Nature 1962, 193, 45–46. [Google Scholar] [CrossRef] [PubMed]
  25. Jennings, A.R.; Betts, A.O. Human adenoviruses in pigs. Ann. N.Y. Acad. Sci. 1962, 101, 485–492. [Google Scholar] [CrossRef] [PubMed]
  26. Huebner, R.J.; Rowe, W.P.; Lane, W.T. Oncogenic effects in hamsters of human adenovirus types 12 and 18. Proc. Natl. Acad. Sci. USA 1962, 48, 2051–2058. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Pereira, H.G.; Allison, A.C.; Niven, J.S.F. Fatal Infection of New-Born Hamsters by an Adenovirus of Human Origin. Nature 1962, 196, 244–245. [Google Scholar] [CrossRef]
  28. Trentin, J.J.; Yabe, Y.; Taylor, G. The quest for human cancer viruses: A new approach to an old problem reveals cancer induction in hamsters by human adenovirus. Science 1962, 137, 835–841. [Google Scholar] [CrossRef] [PubMed]
  29. Yabe, Y.; Trentin, J.J.; Taylor, G. Cancer induction in hamsters by human type 12 adenovirus. Effect of age and of virus dose. Proc. Soc. Exp. Biol. Med. 1962, 111, 343–344. [Google Scholar] [CrossRef]
  30. Girardi, A.J.; Hilleman, M.R.; Zwickey, R.E. Tests in Hamsters for Oncogenic Quality of Ordinary Viruses Including Adenovirus Type 7. Proc. Soc. Exp. Biol. Med. 1964, 115, 1141–1150. [Google Scholar] [CrossRef] [PubMed]
  31. Pope, J.H.; Rowe, W.P. Immunofluorescent Studies of Adenovirus 12 Tumors and of Cells Transformed or Infected by Adenoviruses. J. Exp. Med. 1964, 120, 577–588. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Rabson, A.S.; Kirschstein, R.L.; Paul, F.J. Tumors Produced by Adenovirus 12 in Mastomys and Mice. JNCI J. National Cancer Inst. 1964, 32, 77–87. [Google Scholar] [CrossRef]
  33. Yabe, Y.; Samper, L.; Bryan, E.; Taylor, G.; Trentin, J.J. Oncogenic Effect of Human Adenovirus Type 12, in Mice. Science 1964, 143, 46–47. [Google Scholar] [CrossRef] [PubMed]
  34. Huebner, R.J.; Casey, M.J.; Chanock, R.M.; Schell, K. Tumors induced in hamsters by a strain of adenovirus type 3: Sharing of tumor antigens and “neoantigens” with those produced by adenovirus type 7 tumors. Proc. Natl. Acad. Sci. USA 1965, 54, 381–388. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Pereira, M.S.; Pereira, H.G.; Clarke, S.K.R. Human Adenovirus Type 31 a New Serotype with Oncogenic Properties. Lancet 1965, 285, 21–23. [Google Scholar] [CrossRef]
  36. Reddick, R.A.; Lefkowitz, S.S. In vitro immune responses of rabbits with persistent adenovirus type 5 infection. J. Immunol. 1969, 103, 687–694. [Google Scholar]
  37. Pacini, D.L.; Dubovi, E.J.; Clyde, W.A., Jr. A new animal model for human respiratory tract disease due to adenovirus. J. Infect. Dis. 1984, 150, 92–97. [Google Scholar] [CrossRef] [PubMed]
  38. Gordon, Y.J.; Romanowski, E.; Araullo-Cruz, T. An ocular model of adenovirus type 5 infection in the NZ rabbit. Investig. Ophthalmol. Vis. Sci. 1992, 33, 574–580. [Google Scholar]
  39. Toth, K.; Lee, S.R.; Ying, B.; Spencer, J.F.; Tollefson, A.E.; Sagartz, J.E.; Kong, I.K.; Wang, Z.; Wold, W.S. STAT2 Knockout Syrian Hamsters Support Enhanced Replication and Pathogenicity of Human Adenovirus, Revealing an Important Role of Type I Interferon Response in Viral Control. PLoS Pathog. 2015, 11, e1005084. [Google Scholar] [CrossRef] [Green Version]
  40. Rodriguez, E.; Ip, W.H.; Kolbe, V.; Hartmann, K.; Pilnitz-Stolze, G.; Tekin, N.; Gomez-Medina, S.; Munoz-Fontela, C.; Krasemann, S.; Dobner, T. Humanized Mice Reproduce Acute and Persistent Human Adenovirus Infection. J. Infect. Dis. 2017, 215, 70–79. [Google Scholar] [CrossRef] [PubMed]
  41. Li, X.; Zhou, Z.; Liu, W.; Fan, Y.; Luo, Y.; Li, K.; Zheng, Z.; Tian, X.; Zhou, R. Chinese tree shrew: A permissive model for in vitro and in vivo replication of human adenovirus species B. Emerg. Microbes Infect. 2021, 10, 424–438. [Google Scholar] [CrossRef] [PubMed]
  42. Miao, J.; Chard, L.S.; Wang, Z.; Wang, Y. Syrian Hamster as an Animal Model for the Study on Infectious Diseases. Front. Immunol. 2019, 10, 2329. [Google Scholar] [CrossRef] [Green Version]
  43. Wold, W.S.; Toth, K. Syrian Hamster as an Animal Model to Study Oncolytic Adenoviruses and to Evaluate the Efficacy of Antiviral Compounds. In Advances in Cancer Research; Curiel, D.T., Fisher, P.B., Eds.; Academic Press: Cambridge, MA, USA, 2012; Volume 115, pp. 69–92. [Google Scholar] [CrossRef]
  44. Huebner, R.J.; Chanock, R.M.; Rubin, B.A.; Casey, M.J. Induction by Adenovirus Type 7 of Tumors in Hamsters Having the Antigenic Characteristics of Sv40 Virus. Proc. Natl. Acad. Sci. USA 1964, 52, 1333–1340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. McAllister, R.M.; Goodheart, C.R.; Mirabal, V.Q.; Huebner, R.J. Human adenoviruses: Tumor production in hamsters by type 12 and 18 grown from single plaques. Proc. Soc. Exp. Biol. Med. 1966, 122, 455–458. [Google Scholar] [CrossRef] [PubMed]
  46. Nakajima, T.; Mukai, N. Cell origin of human adenovirus type 12-induced subcutaneous tumor in Syrian hamsters. Acta Neuropathol. 1979, 45, 187–194. [Google Scholar] [CrossRef] [PubMed]
  47. Doerfler, W. Abortive infection and malignant transformation by adenoviruses: Integration of viral DNA and control of viral gene expression by specific patterns of DNA methylation. Adv. Virus Res. 1991, 39, 89–128. [Google Scholar] [CrossRef]
  48. Doerfler, W. A new concept in (adenoviral) oncogenesis: Integration of foreign DNA and its consequences. Biochim. Biophys. Acta (BBA)-Rev. Cancer 1996, 1288, F79–F99. [Google Scholar] [CrossRef]
  49. Hearing, P. Adenovirus Transformation. In DNA Tumor Viruses; Springer: New York, NY, USA, 2009; pp. 145–162. [Google Scholar] [CrossRef]
  50. Doerfler, W. Human Adenovirus Type 12. In Adenovirus Methods and Protocols, 2nd ed.; Wold, W.S., Tollefson, A.E., Eds.; Humana Press: Totowa, NJ, USA, 2007; pp. 197–211. [Google Scholar] [CrossRef]
  51. Kang, Y.S.; Hahn, S. Spontaneous Morphological Transformation in Adenovirus Type 12 Induced Tumor Cells of Armenian and Chinese Hamsters. Korean J. Zool. 1974, 17, 51–56. [Google Scholar]
  52. Hjorth, R.N.; Bonde, G.M.; Pierzchala, W.A.; Vernon, S.K.; Wiener, F.P.; Levner, M.H.; Lubeck, M.D.; Hung, P.P. A new hamster model for adenoviral vaccination. Arch. Virol. 1988, 100, 279–283. [Google Scholar] [CrossRef] [PubMed]
  53. Lichtenstein, D.L.; Spencer, J.F.; Doronin, K.; Patra, D.; Meyer, J.M.; Shashkova, E.V.; Kuppuswamy, M.; Dhar, D.; Thomas, M.A.; Tollefson, A.E.; et al. An acute toxicology study with INGN 007, an oncolytic adenovirus vector, in mice and permissive Syrian hamsters; comparisons with wild-type Ad5 and a replication-defective adenovirus vector. Cancer Gene Ther. 2009, 16, 644–654. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Ying, B.; Toth, K.; Spencer, J.F.; Meyer, J.; Tollefson, A.E.; Patra, D.; Dhar, D.; Shashkova, E.V.; Kuppuswamy, M.; Doronin, K.; et al. INGN 007, an oncolytic adenovirus vector, replicates in Syrian hamsters but not mice: Comparison of biodistribution studies. Cancer Gene Ther. 2009, 16, 625–637. [Google Scholar] [CrossRef] [PubMed]
  55. Tollefson, A.E.; Spencer, J.F.; Ying, B.; Buller, R.M.; Wold, W.S.; Toth, K. Cidofovir and brincidofovir reduce the pathology caused by systemic infection with human type 5 adenovirus in immunosuppressed Syrian hamsters, while ribavirin is largely ineffective in this model. Antivir. Res. 2014, 112, 38–46. [Google Scholar] [CrossRef] [PubMed]
  56. Tollefson, A.E.; Ying, B.; Spencer, J.F.; Sagartz, J.E.; Wold, W.S.M.; Toth, K. Pathology in Permissive Syrian Hamsters after Infection with Species C Human Adenovirus (HAdV-C) Is the Result of Virus Replication: HAdV-C6 Replicates More and Causes More Pathology than HAdV-C5. J. Virol. 2017, 91, e00284-17. [Google Scholar] [CrossRef] [Green Version]
  57. Ying, B.; Spencer, J.F.; Tollefson, A.E.; Wold, W.S.M.; Toth, K. Male Syrian hamsters are more susceptible to intravenous infection with species C human adenoviruses than are females. Virology 2018, 514, 66–78. [Google Scholar] [CrossRef]
  58. Dhakal, S.; Ruiz-Bedoya, C.A.; Zhou, R.; Creisher, P.S.; Villano, J.S.; Littlefield, K.; Ruelas Castillo, J.; Marinho, P.; Jedlicka, A.E.; Ordonez, A.A.; et al. Sex Differences in Lung Imaging and SARS-CoV-2 Antibody Responses in a COVID-19 Golden Syrian Hamster Model. mBio 2021, 12, e0097421. [Google Scholar] [CrossRef] [PubMed]
  59. Yuan, L.; Zhu, H.; Zhou, M.; Ma, J.; Chen, R.; Chen, Y.; Chen, L.; Wu, K.; Cai, M.; Hong, J.; et al. Gender associates with both susceptibility to infection and pathogenesis of SARS-CoV-2 in Syrian hamster. Signal Transduct. Target. Ther. 2021, 6, 136. [Google Scholar] [CrossRef] [PubMed]
  60. Radke, J.R.; Yong, S.L.; Cook, J.L. Low-Level Expression of the E1B 20-Kilodalton Protein by Adenovirus 14p1 Enhances Viral Immunopathogenesis. J. Virol. 2016, 90, 497–505. [Google Scholar] [CrossRef] [Green Version]
  61. Radke, J.R.; Covert, H.J.; Bauer, F.; Ananthanarayanan, V.; Cook, J.L. Adenovirus 14p1 Immunopathogenesis during Lung Infection in the Syrian Hamster. Viruses 2020, 12, 595. [Google Scholar] [CrossRef] [PubMed]
  62. Fan, Z.; Li, W.; Lee, S.R.; Meng, Q.; Shi, B.; Bunch, T.D.; White, K.L.; Kong, I.K.; Wang, Z. Efficient gene targeting in golden Syrian hamsters by the CRISPR/Cas9 system. PLoS ONE 2014, 9, e109755. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Thomas, M.A.; Spencer, J.F.; Toth, K.; Sagartz, J.E.; Phillips, N.J.; Wold, W.S. Immunosuppression enhances oncolytic adenovirus replication and antitumor efficacy in the Syrian hamster model. Mol. Ther. 2008, 16, 1665–1673. [Google Scholar] [CrossRef] [PubMed]
  64. Dhar, D.; Spencer, J.F.; Toth, K.; Wold, W.S. Effect of preexisting immunity on oncolytic adenovirus vector INGN 007 antitumor efficacy in immunocompetent and immunosuppressed Syrian hamsters. J. Virol. 2009, 83, 2130–2139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Sonabend, A.M.; Ulasov, I.V.; Han, Y.; Rolle, C.E.; Nandi, S.; Cao, D.; Tyler, M.A.; Lesniak, M.S. Biodistribution of an oncolytic adenovirus after intracranial injection in permissive animals: A comparative study of Syrian hamsters and cotton rats. Cancer Gene Ther. 2009, 16, 362–372. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Diaconu, I.; Cerullo, V.; Escutenaire, S.; Kanerva, A.; Bauerschmitz, G.J.; Hernandez-Alcoceba, R.; Pesonen, S.; Hemminki, A. Human adenovirus replication in immunocompetent Syrian hamsters can be attenuated with chlorpromazine or cidofovir. J. Gene Med. 2010, 12, 435–445. [Google Scholar] [CrossRef]
  67. Dhar, D.; Toth, K.; Wold, W.S. Syrian hamster tumor model to study oncolytic Ad5-based vectors. Methods Mol. Biol. 2012, 797, 53–63. [Google Scholar] [CrossRef] [PubMed]
  68. Ying, B.; Tollefson, A.E.; Spencer, J.F.; Balakrishnan, L.; Dewhurst, S.; Capella, C.; Buller, R.M.; Toth, K.; Wold, W.S. Ganciclovir inhibits human adenovirus replication and pathogenicity in permissive immunosuppressed Syrian hamsters. Antimicrob. Agents Chemother. 2014, 58, 7171–7181. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Schaar, K.; Geisler, A.; Kraus, M.; Pinkert, S.; Pryshliak, M.; Spencer, J.F.; Tollefson, A.E.; Ying, B.; Kurreck, J.; Wold, W.S.; et al. Anti-adenoviral Artificial MicroRNAs Expressed from AAV9 Vectors Inhibit Human Adenovirus Infection in Immunosuppressed Syrian Hamsters. Mol. Ther. Nucleic Acids 2017, 8, 300–316. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Toth, K.; Spencer, J.F.; Ying, B.; Tollefson, A.E.; Wold, W.S.M. HAdV-C6 Is a More Relevant Challenge Virus than HAdV-C5 for Testing Antiviral Drugs with the Immunosuppressed Syrian Hamster Model. Viruses 2017, 9, 147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Toth, K.; Tollefson, A.E.; Spencer, J.F.; Ying, B.; Wold, W.S.M. Combination therapy with brincidofovir and valganciclovir against species C adenovirus infection in the immunosuppressed Syrian hamster model allows for substantial reduction of dose for both compounds. Antivir. Res. 2017, 146, 121–129. [Google Scholar] [CrossRef] [PubMed]
  72. Toth, K.; Spencer, J.F.; Ying, B.; Tollefson, A.E.; Hartline, C.B.; Richard, E.T.; Fan, J.; Lyu, J.; Kashemirov, B.A.; Harteg, C.; et al. USC-087 protects Syrian hamsters against lethal challenge with human species C adenoviruses. Antivir. Res. 2018, 153, 1–9. [Google Scholar] [CrossRef] [PubMed]
  73. Wold, W.S.M.; Tollefson, A.E.; Ying, B.; Spencer, J.F.; Toth, K. Drug development against human adenoviruses and its advancement by Syrian hamster models. FEMS Microbiol. Rev. 2019, 43, 380–388. [Google Scholar] [CrossRef] [PubMed]
  74. Blanco, J.C.; Pletneva, L.; Boukhvalova, M.; Richardson, J.Y.; Harris, K.A.; Prince, G.A. The cotton rat: An underutilized animal model for human infectious diseases can now be exploited using specific reagents to cytokines, chemokines, and interferons. J. Interferon Cytokine Res. 2004, 24, 21–28. [Google Scholar] [CrossRef] [PubMed]
  75. Green, M.G.; Huey, D.; Niewiesk, S. The cotton rat (Sigmodon hispidus) as an animal model for respiratory tract infections with human pathogens. Lab. Anim. 2013, 42, 170–176. [Google Scholar] [CrossRef]
  76. Prince, G.A.; Porter, D.D.; Jenson, A.B.; Horswood, R.L.; Chanock, R.M.; Ginsberg, H.S. Pathogenesis of adenovirus type 5 pneumonia in cotton rats (Sigmodon hispidus). J. Virol. 1993, 67, 101–111. [Google Scholar] [CrossRef] [Green Version]
  77. Duncan, S.J.; Gordon, F.C.; Gregory, D.W.; McPhie, J.L.; Postlethwaite, R.; White, R.; Willcox, H.N. Infection of mouse liver by human adenovirus type 5. J. Gen. Virol. 1978, 40, 45–61. [Google Scholar] [CrossRef] [PubMed]
  78. Tsai, J.C.; Garlinghouse, G.; McDonnell, P.J.; Trousdale, M.D. An experimental animal model of adenovirus-induced ocular disease. The cotton rat. Arch. Ophthalmol. 1992, 110, 1167–1170. [Google Scholar] [CrossRef] [PubMed]
  79. Trousdale, M.D.; Goldschmidt, P.L.; Nobrega, R. Activity of ganciclovir against human adenovirus type-5 infection in cell culture and cotton rat eyes. Cornea 1994, 13, 435–439. [Google Scholar] [CrossRef] [PubMed]
  80. Kaneko, H.; Mori, S.; Suzuki, O.; Iida, T.; Shigeta, S.; Abe, M.; Ohno, S.; Aoki, K.; Suzutani, T. The cotton rat model for adenovirus ocular infection: Antiviral activity of cidofovir. Antivir. Res. 2004, 61, 63–66. [Google Scholar] [CrossRef]
  81. Lund, O.E.; Stefani, F.H. Corneal histology after epidemic keratoconjunctivitis. Arch. Ophthalmol. 1978, 96, 2085–2088. [Google Scholar] [CrossRef] [PubMed]
  82. Gonzalez, G.; Yawata, N.; Aoki, K.; Kitaichi, N. Challenges in management of epidemic keratoconjunctivitis with emerging recombinant human adenoviruses. J. Clin. Virol. 2019, 112, 1–9. [Google Scholar] [CrossRef] [PubMed]
  83. Toth, K.; Spencer, J.F.; Tollefson, A.E.; Kuppuswamy, M.; Doronin, K.; Lichtenstein, D.L.; Wold, W.S.M. Immune-Competent Cotton Rat Animal Model for Evaluation of Oncolytic Adenoviruses. Mol. Ther. 2004, 9, S390. [Google Scholar] [CrossRef]
  84. Toth, K.; Spencer, J.F.; Tollefson, A.E.; Kuppuswamy, M.; Doronin, K.; Lichtenstein, D.L.; La Regina, M.C.; Prince, G.A.; Wold, W.S. Cotton rat tumor model for the evaluation of oncolytic adenoviruses. Hum. Gene Ther. 2005, 16, 139–146. [Google Scholar] [CrossRef] [PubMed]
  85. Steel, J.C.; Morrison, B.J.; Mannan, P.; Abu-Asab, M.S.; Wildner, O.; Miles, B.K.; Yim, K.C.; Ramanan, V.; Prince, G.A.; Morris, J.C. Immunocompetent syngeneic cotton rat tumor models for the assessment of replication-competent oncolytic adenovirus. Virology 2007, 369, 131–142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Knowles, M.K.; Roberts, D.; Craig, S.; Sheen, M.; Nadin-Davis, S.A.; Wandeler, A.I. In vitro and in vivo genetic stability studies of a human adenovirus type 5 recombinant rabies glycoprotein vaccine (ONRAB). Vaccine 2009, 27, 2662–2668. [Google Scholar] [CrossRef] [PubMed]
  87. Peng, X.; Knouse, J.A.; Hernon, K.M. Rabbit Models for Studying Human Infectious Diseases. Comp. Med. 2015, 65, 499–507. [Google Scholar]
  88. Esteves, P.J.; Abrantes, J.; Baldauf, H.M.; BenMohamed, L.; Chen, Y.; Christensen, N.; Gonzalez-Gallego, J.; Giacani, L.; Hu, J.; Kaplan, G.; et al. The wide utility of rabbits as models of human diseases. Exp. Mol. Med. 2018, 50, 1–10. [Google Scholar] [CrossRef] [PubMed]
  89. Gordon, Y.J.; Romanowski, E.G.; Araullo-Cruz, T. Topical HPMPC inhibits adenovirus type 5 in the New Zealand rabbit ocular replication model. Investig. Ophthalmol. Vis. Sci. 1994, 35, 4135–4143. [Google Scholar]
  90. De Oliveira, C.B.; Stevenson, D.; LaBree, L.; McDonnell, P.J.; Trousdale, M.D. Evaluation of Cidofovir (HPMPC, GS-504) against adenovirus type 5 infection in vitro and in a New Zealand rabbit ocular model. Antivir. Res. 1996, 31, 165–172. [Google Scholar] [CrossRef]
  91. Romanowski, E.G.; Gordon, Y.J.; Araullo-Cruz, T.; Yates, K.A.; Kinchington, P.R. The antiviral resistance and replication of cidofovir-resistant adenovirus variants in the New Zealand White rabbit ocular model. Investig. Ophthalmol. Vis. Sci. 2001, 42, 1812–1815. [Google Scholar]
  92. Romanowski, E.G.; Yates, K.A.; Gordon, Y.J. Antiviral prophylaxis with twice daily topical cidofovir protects against challenge in the adenovirus type 5/New Zealand rabbit ocular model. Antivir. Res. 2001, 52, 275–280. [Google Scholar] [CrossRef]
  93. Epstein, S.P.; Pashinsky, Y.Y.; Gershon, D.; Winicov, I.; Srivilasa, C.; Kristic, K.J.; Asbell, P.A. Efficacy of topical cobalt chelate CTC-96 against adenovirus in a cell culture model and against adenovirus keratoconjunctivitis in a rabbit model. BMC Ophthalmol. 2006, 6, 22. [Google Scholar] [CrossRef] [Green Version]
  94. Romanowski, E.G.; Yates, K.A.; Teuchner, B.; Nagl, M.; Irschick, E.U.; Gordon, Y.J. N-chlorotaurine is an effective antiviral agent against adenovirus in vitro and in the Ad5/NZW rabbit ocular model. Investig. Ophthalmol. Vis. Sci. 2006, 47, 2021–2026. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Clement, C.; Capriotti, J.A.; Kumar, M.; Hobden, J.A.; Foster, T.P.; Bhattacharjee, P.S.; Thompson, H.W.; Mahmud, R.; Liang, B.; Hill, J.M. Clinical and antiviral efficacy of an ophthalmic formulation of dexamethasone povidone-iodine in a rabbit model of adenoviral keratoconjunctivitis. Investig. Ophthalmol. Vis. Sci. 2011, 52, 339–344. [Google Scholar] [CrossRef] [PubMed]
  96. Szpirer, C. Rat models of human diseases and related phenotypes: A systematic inventory of the causative genes. J. Biomed. Sci. 2020, 27, 84. [Google Scholar] [CrossRef]
  97. Wilson, J.M.; Makidon, P.E.; Bergin, I.L. Rat Models of Infectious Disease. In The Laboratory Rat, 3rd ed.; Suckow, M.A., Hankenson, F.C., Wilson, R.P., Foley, P.L., Eds.; Academic Press: Cambridge, MA, USA, 2020; pp. 1107–1134. [Google Scholar] [CrossRef]
  98. Huebner, R.J.; Rowe, W.P.; Turner, H.C.; Lane, W.T. Specific Adenovirus Complement-Fixing Antigens in Virus-Free Hamster and Rat Tumors. Proc. Natl. Acad. Sci. USA 1963, 50, 379–389. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Kobayashi, S.; Mukai, N. Retinoblastoma-like tumors induced by human adenovirus type 12 in rats. Cancer Res. 1974, 34, 1646–1651. [Google Scholar] [PubMed]
  100. Mukai, N.; Murao, T. Retinal tumor induction by ocular inoculation of human adenovirus in 3-day-old rats. J. Neuropathol. Exp. Neurol. 1975, 34, 28–35. [Google Scholar] [CrossRef]
  101. Jonsson, N.; Ankerst, J. Studies on adenovirus type 9-induced mammary fibroadenomas in rats and their malignant transformation. Cancer 1977, 39, 2513–2519. [Google Scholar] [CrossRef]
  102. Javier, R.; Raska, K., Jr.; Macdonald, G.J.; Shenk, T. Human adenovirus type 9-induced rat mammary tumors. J. Virol. 1991, 65, 3192–3202. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Ankerst, J.; Jonsson, N.; Kjellen, L.; Norrby, E.; Sjogren, H.O. Induction of mammary fibroadenomas in rats by adenovirus type 9. Int. J. Cancer 1974, 13, 286–290. [Google Scholar] [CrossRef] [PubMed]
  104. Ankerst, J.; Jonsson, N. Adenovirus type 9-induced tumorigenesis in the rat mammary gland related to sex hormonal state. J. Natl. Cancer Inst. 1989, 81, 294–298. [Google Scholar] [CrossRef] [PubMed]
  105. Javier, R.; Raska, K., Jr.; Shenk, T. Requirement for the adenovirus type 9 E4 region in production of mammary tumors. Science 1992, 257, 1267–1271. [Google Scholar] [CrossRef] [PubMed]
  106. Javier, R.T. Adenovirus type 9 E4 open reading frame 1 encodes a transforming protein required for the production of mammary tumors in rats. J. Virol. 1994, 68, 3917–3924. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Thomas, D.L.; Shin, S.; Jiang, B.H.; Vogel, H.; Ross, M.A.; Kaplitt, M.; Shenk, T.E.; Javier, R.T. Early region 1 transforming functions are dispensable for mammary tumorigenesis by human adenovirus type 9. J. Virol. 1999, 73, 3071–3079. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Nicol, C.G.; Graham, D.; Miller, W.H.; White, S.J.; Smith, T.A.; Nicklin, S.A.; Stevenson, S.C.; Baker, A.H. Effect of adenovirus serotype 5 fiber and penton modifications on in vivo tropism in rats. Mol. Ther. 2004, 10, 344–354. [Google Scholar] [CrossRef] [PubMed]
  109. Postlethwaite, R. Liver Damage Induced in Mice by Human Adenovirus Type 5. Scott. Med. J. 1973, 18, 131. [Google Scholar] [CrossRef]
  110. Jogler, C.; Hoffmann, D.; Theegarten, D.; Grunwald, T.; Uberla, K.; Wildner, O. Replication properties of human adenovirus in vivo and in cultures of primary cells from different animal species. J. Virol. 2006, 80, 3549–3558. [Google Scholar] [CrossRef] [Green Version]
  111. Gaggar, A.; Shayakhmetov, D.M.; Lieber, A. CD46 is a cellular receptor for group B adenoviruses. Nat. Med. 2003, 9, 1408–1412. [Google Scholar] [CrossRef] [PubMed]
  112. Persson, B.D.; John, L.; Rafie, K.; Strebl, M.; Frangsmyr, L.; Ballmann, M.Z.; Mindler, K.; Havenga, M.; Lemckert, A.; Stehle, T.; et al. Human species D adenovirus hexon capsid protein mediates cell entry through a direct interaction with CD46. Proc. Natl. Acad. Sci. USA 2021, 118, e2020732118. [Google Scholar] [CrossRef] [PubMed]
  113. Hemsath, J.R.; Liaci, A.M.; Rubin, J.D.; Parrett, B.J.; Lu, S.-C.; Nguyen, T.V.; Turner, M.A.; Chen, C.Y.; Cupelli, K.; Reddy, V.S.; et al. Ex Vivo and In Vivo CD46 Receptor Utilization by Species D Human Adenovirus Serotype 26 (HAdV26). J. Virol. 2021. [Google Scholar] [CrossRef] [PubMed]
  114. Ginsberg, H.S.; Moldawer, L.L.; Sehgal, P.B.; Redington, M.; Kilian, P.L.; Chanock, R.M.; Prince, G.A. A mouse model for investigating the molecular pathogenesis of adenovirus pneumonia. Proc. Natl. Acad. Sci. USA 1991, 88, 1651–1655. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Oualikene, W.; Gonin, P.; Eloit, M. Short and long term dissemination of deletion mutants of adenovirus in permissive (cotton rat) and non-permissive (mouse) species. J. Gen. Virol. 1994, 75 Pt 10, 2765–2768. [Google Scholar] [CrossRef]
  116. Billerbeck, E.; Horwitz, J.A.; Labitt, R.N.; Donovan, B.M.; Vega, K.; Budell, W.C.; Koo, G.C.; Rice, C.M.; Ploss, A. Characterization of human antiviral adaptive immune responses during hepatotropic virus infection in HLA-transgenic human immune system mice. J. Immunol. 2013, 191, 1753–1764. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Zhou, X.; Robinson, C.M.; Rajaiya, J.; Dehghan, S.; Seto, D.; Jones, M.S.; Dyer, D.W.; Chodosh, J. Analysis of human adenovirus type 19 associated with epidemic keratoconjunctivitis and its reclassification as adenovirus type 64. Investig. Ophthalmol. Vis. Sci. 2012, 53, 2804–2811. [Google Scholar] [CrossRef] [PubMed]
  118. Mukherjee, S.; Zhou, X.; Rajaiya, J.; Chodosh, J. Ultrastructure of adenovirus keratitis. Investig. Ophthalmol. Vis. Sci. 2015, 56, 472–477. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Mentel, R.; Wegner, U. Evaluation of the efficacy of 2′,3′-dideoxycytidine against adenovirus infection in a mouse pneumonia model. Antivir. Res. 2000, 47, 79–87. [Google Scholar] [CrossRef]
  120. Paielli, D.L.; Wing, M.S.; Rogulski, K.R.; Gilbert, J.D.; Kolozsvary, A.; Kim, J.H.; Hughes, J.; Schnell, M.; Thompson, T.; Freytag, S.O. Evaluation of the biodistribution, persistence, toxicity, and potential of germ-line transmission of a replication-competent human adenovirus following intraprostatic administration in the mouse. Mol. Ther. 2000, 1, 263–274. [Google Scholar] [CrossRef] [PubMed]
  121. Fueyo, J.; Gomez-Manzano, C.; Alemany, R.; Lee, P.S.; McDonnell, T.J.; Mitlianga, P.; Shi, Y.X.; Levin, V.A.; Yung, W.K.; Kyritsis, A.P. A mutant oncolytic adenovirus targeting the Rb pathway produces anti-glioma effect in vivo. Oncogene 2000, 19, 2–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Tallone, T.; Malin, S.; Samuelsson, A.; Wilbertz, J.; Miyahara, M.; Okamoto, K.; Poellinger, L.; Philipson, L.; Pettersson, S. A mouse model for adenovirus gene delivery. Proc. Natl. Acad. Sci. USA 2001, 98, 7910–7915. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Young, A.M.; Archibald, K.M.; Tookman, L.A.; Pool, A.; Dudek, K.; Jones, C.; Williams, S.L.; Pirlo, K.J.; Willis, A.E.; Lockley, M.; et al. Failure of translation of human adenovirus mRNA in murine cancer cells can be partially overcome by L4-100K expression in vitro and in vivo. Mol. Ther. 2012, 20, 1676–1688. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Hassan, F.; Lossie, S.L.; Kasik, E.P.; Channon, A.M.; Ni, S.; Kennedy, M.A. A mouse model study of toxicity and biodistribution of a replication defective adenovirus serotype 5 virus with its genome engineered to contain a decoy hyper binding site to sequester and suppress oncogenic HMGA1 as a new cancer treatment therapy. PLoS ONE 2018, 13, e0192882. [Google Scholar] [CrossRef] [PubMed]
  125. Doszpoly, A.; de la Cuesta, F.; Lopez-Gordo, E.; Benezech, C.; Nicklin, S.A.; Baker, A.H. Human Adenovirus Serotype 5 Is Sensitive to IgM-Independent Neutralization In Vitro and In Vivo. Viruses 2019, 11, 616. [Google Scholar] [CrossRef] [Green Version]
  126. Bates, E.A.; Counsell, J.R.; Alizert, S.; Baker, A.T.; Suff, N.; Boyle, A.; Bradshaw, A.C.; Waddington, S.N.; Nicklin, S.A.; Baker, A.H.; et al. In Vitro and In Vivo Evaluation of Human Adenovirus Type 49 as a Vector for Therapeutic Applications. Viruses 2021, 13, 1483. [Google Scholar] [CrossRef]
  127. Shultz, L.D.; Ishikawa, F.; Greiner, D.L. Humanized mice in translational biomedical research. Nat. Rev. Immunol. 2007, 7, 118–130. [Google Scholar] [CrossRef] [PubMed]
  128. Legrand, N.; Ploss, A.; Balling, R.; Becker, P.D.; Borsotti, C.; Brezillon, N.; Debarry, J.; de Jong, Y.; Deng, H.; Di Santo, J.P.; et al. Humanized mice for modeling human infectious disease: Challenges, progress, and outlook. Cell Host Microbe 2009, 6, 5–9. [Google Scholar] [CrossRef] [Green Version]
  129. Dash, P.K.; Gorantla, S.; Poluektova, L.; Hasan, M.; Waight, E.; Zhang, C.; Markovic, M.; Edagwa, B.; Machhi, J.; Olson, K.E.; et al. Humanized Mice for Infectious and Neurodegenerative disorders. Retrovirology 2021, 18, 13. [Google Scholar] [CrossRef] [PubMed]
  130. Estes, J.D.; Wong, S.W.; Brenchley, J.M. Nonhuman primate models of human viral infections. Nat. Rev. Immunol. 2018, 18, 390–404. [Google Scholar] [CrossRef] [PubMed]
  131. Lubeck, M.D.; Davis, A.R.; Chengalvala, M.; Natuk, R.J.; Morin, J.E.; Molnar-Kimber, K.; Mason, B.B.; Bhat, B.M.; Mizutani, S.; Hung, P.P. Immunogenicity and efficacy testing in chimpanzees of an oral hepatitis B vaccine based on live recombinant adenovirus. Proc. Natl. Acad. Sci. USA 1989, 86, 6763–6767. [Google Scholar] [CrossRef] [Green Version]
  132. Lubeck, M.D.; Natuk, R.; Myagkikh, M.; Kalyan, N.; Aldrich, K.; Sinangil, F.; Alipanah, S.; Murthy, S.C.; Chanda, P.K.; Nigida, S.M., Jr.; et al. Long-term protection of chimpanzees against high-dose HIV-1 challenge induced by immunization. Nat. Med. 1997, 3, 651–658. [Google Scholar] [CrossRef] [PubMed]
  133. Lubeck, M.D.; Natuk, R.J.; Chengalvala, M.; Chanda, P.K.; Murthy, K.K.; Murthy, S.; Mizutani, S.; Lee, S.G.; Wade, M.S.; Bhat, B.M.; et al. Immunogenicity of recombinant adenovirus-human immunodeficiency virus vaccines in chimpanzees following intranasal administration. AIDS Res. Hum. Retrovir. 1994, 10, 1443–1449. [Google Scholar] [CrossRef] [PubMed]
  134. Chengalvala, M.V.; Bhat, B.M.; Bhat, R.A.; Dheer, S.K.; Lubeck, M.D.; Purcell, R.H.; Murthy, K.K. Replication and immunogenicity of Ad7-, Ad4-, and Ad5-hepatitis B virus surface antigen recombinants, with or without a portion of E3 region, in chimpanzees. Vaccine 1997, 15, 335–339. [Google Scholar] [CrossRef]
  135. Shiver, J.W.; Fu, T.M.; Chen, L.; Casimiro, D.R.; Davies, M.E.; Evans, R.K.; Zhang, Z.Q.; Simon, A.J.; Trigona, W.L.; Dubey, S.A.; et al. Replication-incompetent adenoviral vaccine vector elicits effective anti-immunodeficiency-virus immunity. Nature 2002, 415, 331–335. [Google Scholar] [CrossRef]
  136. Liu, J.; O’Brien, K.L.; Lynch, D.M.; Simmons, N.L.; La Porte, A.; Riggs, A.M.; Abbink, P.; Coffey, R.T.; Grandpre, L.E.; Seaman, M.S.; et al. Immune control of an SIV challenge by a T-cell-based vaccine in rhesus monkeys. Nature 2009, 457, 87–91. [Google Scholar] [CrossRef]
  137. Mukai, N.; Kalter, S.S.; Cummins, L.B.; Matthews, V.A.; Nishida, T.; Nakajima, T. Retinal tumor induced in the baboon by human adenovirus 12. Science 1980, 210, 1023–1025. [Google Scholar] [CrossRef]
  138. Li, R.; Zanin, M.; Xia, X.; Yang, Z. The tree shrew as a model for infectious diseases research. J. Thorac. Dis. 2018, 10 (Suppl. 19), S2272–S2279. [Google Scholar] [CrossRef] [PubMed]
  139. Meurens, F.; Summerfield, A.; Nauwynck, H.; Saif, L.; Gerdts, V. The pig: A model for human infectious diseases. Trends Microbiol. 2012, 20, 50–57. [Google Scholar] [CrossRef] [PubMed]
  140. Swindle, M.M.; Makin, A.; Herron, A.J.; Clubb, F.J., Jr.; Frazier, K.S. Swine as models in biomedical research and toxicology testing. Vet. Pathol. 2012, 49, 344–356. [Google Scholar] [CrossRef] [PubMed]
  141. Koodie, L.; Robertson, M.G.; Chandrashekar, M.; Ruth, G.; Dunning, M.; Bianco, R.W.; Davydova, J. Rodents Versus Pig Model for Assessing the Performance of Serotype Chimeric Ad5/3 Oncolytic Adenoviruses. Cancers 2019, 11, 298. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Padilla-Carlin, D.J.; McMurray, D.N.; Hickey, A.J. The Guinea Pig as a Model of Infectious Diseases. Comp. Med. 2008, 58, 324–340. [Google Scholar] [PubMed]
  143. Lowen, A.C.; Mubareka, S.; Tumpey, T.M.; Garcia-Sastre, A.; Palese, P. The guinea pig as a transmission model for human influenza viruses. Proc. Natl. Acad. Sci. USA 2006, 103, 9988–9992. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Kumar, M.; Krause, K.K.; Azouz, F.; Nakano, E.; Nerurkar, V.R. A guinea pig model of Zika virus infection. Virol. J. 2017, 14, 75. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Faucon, N.; Chardonnet, Y.; Sohier, R. Persistence of Adenovirus 5 in guinea pigs. Infect. Immun. 1974, 10, 11–15. [Google Scholar] [CrossRef] [Green Version]
  146. Vitalis, T.Z.; Keicho, N.; Itabashi, S.; Hayashi, S.; Hogg, J.C. A model of latent adenovirus 5 infection in the guinea pig (Cavia porcellus). Am. J. Respir. Cell. Mol. Biol. 1996, 14, 225–231. [Google Scholar] [CrossRef]
  147. Wu, S.; Kroeker, A.; Wong, G.; He, S.; Hou, L.; Audet, J.; Wei, H.; Zhang, Z.; Fernando, L.; Soule, G.; et al. An Adenovirus Vaccine Expressing Ebola Virus Variant Makona Glycoprotein Is Efficacious in Guinea Pigs and Nonhuman Primates. J. Infect. Dis. 2016, 214 (Suppl. 3), S326–S332. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Pauly, M.; Akoua-Koffi, C.; Buchwald, N.; Schubert, G.; Weiss, S.; Couacy-Hymann, E.; Anoh, A.E.; Mossoun, A.; Calvignac-Spencer, S.; Leendertz, S.A.; et al. Adenovirus in Rural Cote D’Ivoire: High Diversity and Cross-Species Detection. Ecohealth 2015, 12, 441–452. [Google Scholar] [CrossRef] [Green Version]
  149. Bailey, E.S.; Fieldhouse, J.K.; Choi, J.Y.; Gray, G.C. A Mini Review of the Zoonotic Threat Potential of Influenza Viruses, Coronaviruses, Adenoviruses, and Enteroviruses. Front. Public Health 2018, 6, 104. [Google Scholar] [CrossRef]
  150. Lange, C.E.; Niama, F.R.; Cameron, K.; Olson, S.H.; Aime Nina, R.; Ondzie, A.; Bounga, G.; Smith, B.R.; Pante, J.; Reed, P.; et al. First evidence of a new simian adenovirus clustering with Human mastadenovirus F viruses. Virol. J. 2019, 16, 147. [Google Scholar] [CrossRef]
  151. Borkenhagen, L.K.; Fieldhouse, J.K.; Seto, D.; Gray, G.C. Are adenoviruses zoonotic? A systematic review of the evidence. Emerg. Microbes Infect. 2019, 8, 1679–1687. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Medkour, H.; Amona, I.; Akiana, J.; Davoust, B.; Bitam, I.; Levasseur, A.; Tall, M.L.; Diatta, G.; Sokhna, C.; Hernandez-Aguilar, R.A.; et al. Adenovirus Infections in African Humans and Wild Non-Human Primates: Great Diversity and Cross-Species Transmission. Viruses 2020, 12, 657. [Google Scholar] [CrossRef] [PubMed]
  153. Mollentze, N.; Streicker, D.G. Viral zoonotic risk is homogenous among taxonomic orders of mammalian and avian reservoir hosts. Proc. Natl. Acad. Sci. USA 2020, 117, 9423–9430. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Mollentze, N.; Babayan, S.A.; Streicker, D.G. Identifying and prioritizing potential human-infecting viruses from their genome sequences. PLoS Biol. 2021, 19, e3001390. [Google Scholar] [CrossRef] [PubMed]
  155. Medkour, H.; Castaneda, S.; Amona, I.; Fenollar, F.; Andre, C.; Belais, R.; Mungongo, P.; Muyembe-Tamfum, J.J.; Levasseur, A.; Raoult, D.; et al. Potential zoonotic pathogens hosted by endangered bonobos. Sci. Rep. 2021, 11, 6331. [Google Scholar] [CrossRef] [PubMed]
  156. Kremer, E.J. What is the risk of a deadly adenovirus pandemic? PLoS Pathog. 2021, 17, e1009814. [Google Scholar] [CrossRef] [PubMed]
  157. Wachtman, L.; Mansfield, K. Viral Diseases of Nonhuman Primates. In Nonhuman Primates in Biomedical Research, 2nd ed.; Abee, C.R., Mansfield, K., Tardif, S., Morris, T., Eds.; Academic Press: Cambridge, MA, USA, 2012; Volume 2, pp. 1–104. [Google Scholar] [CrossRef]
  158. Lucher, L.A. Abortive Adenovirus Infection and Host Range Determinants. In The Molecular Repertoire of Adenoviruses; Doerfler, W., Böhm, B., Eds.; Springer: Berlin/Heidelberg, Germany, 1995; Volume 199. [Google Scholar] [CrossRef]
  159. Brough, D.E.; Rice, S.A.; Sell, S.; Klessig, D.F. Restricted changes in the adenovirus DNA-binding protein that lead to extended host range or temperature-sensitive phenotypes. J. Virol. 1985, 55, 206–212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Landmark publications on animal models in HAdV research. HAdV research in small animal models was pioneered by Rowe et al. in 1955 at the NIH in Bethesda, MD (USA) [21]. That work laid the foundation for various follow-up studies and novel approaches to establish a suitable animal model to study HAdV infection, including HAdV-induced pathogenesis and cancer development in vivo. The most relevant models are Syrian hamsters [28], mice [32,33], cotton rats [37], New Zealand (NZ) rabbits [38], STAT2 knockout (KO) Syrian hamsters [39], humanized mice [40], and Chinese tree shrews [41] (sorted by publication date of the first report on the respective animal model).
Figure 1. Landmark publications on animal models in HAdV research. HAdV research in small animal models was pioneered by Rowe et al. in 1955 at the NIH in Bethesda, MD (USA) [21]. That work laid the foundation for various follow-up studies and novel approaches to establish a suitable animal model to study HAdV infection, including HAdV-induced pathogenesis and cancer development in vivo. The most relevant models are Syrian hamsters [28], mice [32,33], cotton rats [37], New Zealand (NZ) rabbits [38], STAT2 knockout (KO) Syrian hamsters [39], humanized mice [40], and Chinese tree shrews [41] (sorted by publication date of the first report on the respective animal model).
Biology 10 01253 g001
Table 1. Wild type HAdV susceptibility and wild type HAdV-induced disease in different relevant animal models. The corresponding references can be found in the respective paragraph text.
Table 1. Wild type HAdV susceptibility and wild type HAdV-induced disease in different relevant animal models. The corresponding references can be found in the respective paragraph text.
Species HAdV Type(s) + HAdV Dose (Range) # Infection Route Clinical Signs Of A Systemic Disease Histopathological Lesions Induction Of Neutralizing Antibodies? HAdV-Induced Tumors?
Cotton rat (Sigmodon
hispidus)
C5, D8, D37 and E4 102–1010 i.m., i.n., i.o. No * Lung, airway and eye damage Yes No
Guinea pig (Cavia
porcellus)
C5 107–8 × 108 i.c., i.n. Not reported Lung and airway damage Yes No
Humanized mouse (Mus musculus) C2 1.4 × 104–1.4 × 108 i.v. Lethargy, weight loss Liver damage Yes No
Mouse (Mus musculus) A12, C5, D37 and D64 105–1.4 × 1011 i.m., i.n., i.pe., i.pu., i.v., i.o., s.c. No * Liver, eye and lung damage Yes Yes
New Zealand rabbit (Oryctolagus cuniculus) C5 5.7 × 105–1.6 × 109 i.v., i.o. Eye pathology Lung and eye damage Yes No
Pig (Sus scrofa) C5 1.6 × 103–1010 i.v., i.t. No Moderate lung damage Not reported No
Rat (rattus norvegicus) A12, C5 and D9 5 × 107–3 × 1011 i.o. i.pe., s.c. No Not reported Not reported Yes
Syrian hamster (Mesocricetus auratus) A12, A18, B3, B7, B14, C5 and C6 1.5 × 1010–2 × 1011 i.i., i.n., i.pu., i.t., s.c., i.v. Weight loss with some HAdV types § Liver, lung and airway damage Yes Yes
Tree shrew (Tupaia
belangeri chinensis)
B55 5 × 105 i.n. Weight loss and body temperature increase Lung damage Yes No
+ Without mutant HAdVs or engineered oncolytic HAdVs. # Not all references reported accurate TCID50 values; where appropriate, PFU were converted to TCID50 (pfu [mL]/TCID50 [mL] = 0.7). * A lethal dose of 3.6 × 109 TCID50 has been reported for cotton rats [76], and ~1.4 × 109 TCID50 for mice [77]. § Weight loss and lethal challenge have been reported in chemically immunosuppressed Syrian hamsters [55,56]. i.c., intracardial; i.i., intraintestinal; i.m., intramuscular; i.n., intranasal; i.o., intraocular, i.pe., intraperitoneal; i.pu., intrapulmonal; i.t., intratracheal; i.v., intravenous; s.c., subcutaneous.
Table 2. Available HAdV animal models—their strengths and limitations.
Table 2. Available HAdV animal models—their strengths and limitations.
Model Animal Strengths Limitations
Cotton rat
HAdV replicates in the upper respiratory tract and the lungs and causes pneumonia
resembles human EKC
used in many studies → good comparability between studies
difficult animal handling
subtle systemic disease signs,
dependent on the infectious dose
Guinea pig
HAdV replicates in the lungs and causes pneumonia
persistent HAdV infection (?)
few studies available → limited comparability between studies
Humanized mouse
persistent HAdV infection
robust clinical readouts like weight loss and lethargy
high expenses
laborious to generate
few studies available → limited comparability between studies
Immunosuppressed hamster
resembles HAdV infection of immunosuppressed
humans
weight loss as a robust clinical readout
chemical immunosuppression
required
Mouse
countless molecular and genetic tools available
HAdV-induced tumors
mostly non-permissive
subtle clinical disease signs
New Zealand rabbit
resembles human EKC, used in many studies → good comparability between studies
persistent HAdV infection
higher maintenance costs
compared to mice and rats
no systemic infection
Pig
HAdV replicates in the lungs and causes pneumonia
animal size and handling
no signs of a clinical disease
few studies available → limited comparability between studies
Rat
HAdV-induced tumors
non-permissive
no signs of a clinical disease
STAT2 KO
hamster
increased HAdV replication compared to wild type
hamsters
resembles aspects of HAdV infection of immunosuppressed humans
limited availability
animals are immunosuppressed
Syrian hamster
most established HAdV animal model
used in many efficacy studies on HAdV therapeutics → good comparability between studies
various molecular tools available
virus replication in various organs
HAdV-induced tumors
no signs of a clinical disease
Tree shrew
resembles human HAdV-induced pneumonia
robust clinical readouts light weight loss and fever
suitable for studies that assess zoonotic potential of HAdVs (?)
limited availability
few studies available → limited comparability between studies
EKC, (adenoviral) epidemic keratoconjunctivitis; KO, knockout.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Bertzbach, L.D.; Ip, W.-H.; Dobner, T. Animal Models in Human Adenovirus Research. Biology 2021, 10, 1253. https://doi.org/10.3390/biology10121253

AMA Style

Bertzbach LD, Ip W-H, Dobner T. Animal Models in Human Adenovirus Research. Biology. 2021; 10(12):1253. https://doi.org/10.3390/biology10121253

Chicago/Turabian Style

Bertzbach, Luca D., Wing-Hang Ip, and Thomas Dobner. 2021. "Animal Models in Human Adenovirus Research" Biology 10, no. 12: 1253. https://doi.org/10.3390/biology10121253

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop