Preprint
Review

This version is not peer-reviewed.

Gammaretrovirus Infections in Humans in the Past, Present and Future: Have We Defeated the Pathogen?

Submitted:

01 December 2025

Posted:

03 December 2025

You are already at the latest version

Abstract
Gammaretroviruses are ubiquitous pathogens, often associated with the induction of neoplasia, especially leukemia, lymphoma, and sarcoma, and with a propensity to target the germline. The latter trait has left extensive evidence of their infectious competence in vertebrate genomes, the human genome being no exception. Despite the continuing activity of gammaretroviruses in mammals, including Old World monkeys, apes and gibbons, humans have apparently evaded novel infections by the virus class for the past 30 million years or so. Nevertheless, from the 1970’s onward, cell culture studies repeatedly discovered gammaretroviral components and/or virus replication in human samples. The last novel ‘human’ gammaretrovirus, identified in prostate cancer tissue, culminated in the XMRV frenzy of the 2000’s. In the end, that discovery was shown to be due to lab contamination with a murine gammaretrovirus. Contamination is also the likely source of the earlier findings. Complementation between genes of partially defect endogenous proviruses could have been another source of the virions observed. However, the capacity of many gammaretroviruses to replicate in human cell lines, as well as the presence of diverse infectious gammaretroviral species in our animal companions, for instance in mice, cats, pigs, monkeys, chickens, and bats, does not make a transmission to humans an improbable scenario. This review will summarize evidence for, or the lack of, gammaretrovirus infections in humans in the past, the present and the near future. Aspects linked to the probabilities of novel gammaretrovirus infections in humans, regarding exposure risk in connection to –modern– lifestyle, geography, diet, and habitat together with genetic and immune factors, will also be part of the review, as will be the estimated consequences of such novel infections.
Keywords: 
;  ;  ;  ;  ;  ;  

1. Introduction

In the family Retroviridae, subfamily Orthoretrovirinae, gammaretroviruses constitute one of the six genera recognized, which are the alpha-, beta-, gamma-, delta-, epsilon- and lenti- (retro)viruses. Spumaretroviruses belong to the subfamily Spumaretrovirinae. Alpharetroviruses are mainly found in birds, epsilonretroviruses in fish [1]. Phylogenetic analysis of reverse-transcriptase (RT) domains and of Env genes are the usual methods to define retrovirus genera [2]. An earlier classification scheme, which has now been abandoned, was based on the –budding– virion structure observed with electron microscopy (EM). Gammaretrovirus particles, for instance, with a centrally located, dense core and hardly any visible spike proteins, exhibit a “C type” morphology, like alpha-, delta, and epsilonretroviruses do [1]. Older studies, published around 1960-1980, reporting retrovirus discoveries based on virion type without further sequence analysis therefore need to be approached with some caution. For instance, the first description of the deltaretrovirus human T-lymphotropic virus (HTLV) was centered on its C-type morphology, and not on the genome structure [3].
At present, only two retroviruses circulate in humans, e.g. the first being the deltavirus HTLV, and the other the lentivirus human immunodeficiency virus (HIV). There is evidence that a betaretrovirus, a human variant of mouse mammary tumor virus (h-MMTV), also referred to as human betaretrovirus (HBRV), exists in humans, but no consensus has been reached yet [4,5,6,7,8,9]. The same is true for the presumed circulation of a second deltaretrovirus, bovine leukemia virus (BLV) in humans [9,10].
The current lack of infectious gamma- and, perhaps, betaretroviruses in humans is somewhat surprising. Since these two genera are ubiquitous in vertebrates, and have a propensity to target the germline, they abound as heritable endogenous retrovirus (ERV) sequences. An extensive genomic record documents their presence over millions of years of evolution [11,12,13]. The human genome likewise contains many gamma-like ERVs, showing that our ancestors were vulnerable to infection [14]. Germline gamma-related proviruses, or what remains of them, together with epsilon-related viral sequences, are known as Class I ERVs [15]. Class II ERVs are alpha-, beta-, delta- or lenti-like, and class III are spuma-like ERVs. In primate genomes, gamma-like ERVs exceed the number of beta-like ERVs; together they constitute the most abundant ERV group in vertebrates [11]. Recombinant beta-gamma retroviruses also exist, for instance the Old World monkey (OWM) simian retroviruses (SRVs), and simian endogenous retrovirus (SERV), which have beta-like gag-pol genes and a gamma-like env [16,17,18]. In primate genomes, lenti-like ERVS are present in some prosimians; spuma-like ERVs are scarce, and delta-like ERVS have not been found [19,20,21,22,23].
Gammaretroviral cross-species transmissions are relatively common, especially between closely related species; class switching is rare [11,24,25]. An example of a class switch are the avian reticuloendotheliosis viruses (REVs), which have recently been transmitted from mammals to birds [26,27]. The ERV burden in a contemporary genome can inform us on the history of retrovirus infections, evolution, and cross-species transmissions in the –distant– past, since ERVs are demonstrably suitable for conveying this information [28,29,30,31,32,33,34,35]. However, such evidence will never be complete, since absence in a genome is not proof of absence of infection; germline infection and fixation of a trait are equally important for later detection.
To discover current retrovirus infections, diagnostic methods such as serology, Western blotting, PCR, and sequencing can be utilized. Large population surveys for novel retroviruses have not been done yet, although some early efforts have been published in the past [36,37,38,39,40,41,42,43,44].
Since two retrovirus genera, delta- and lentiviruses, already infect humans, and there is some evidence of human infection with a betaretrovirus, this review will summarize evidence for, or the absence of, another major group of retroviruses, namely the gammaretroviruses. Infection with gammaretroviruses, or the lack thereoff, in the past, the present and the future will be reviewed in the light of the omnipresence of infectious variants in domestic animals, such as poultry, mice, cats, pigs, and in many primate species. The review will also discuss factors influencing the infection risk, including lifestyle, medical interventions, geography, diet, and habitat together with genetic and immune factors together with the anticipated consequences of such infections.

2. Materials and Methods

Selection of ICTV-Listed Gammaretroviruses to be Included in the Review

The International Committee on Taxonomy of Viruses (ICTV) 2024 list contains 15 examplar isolates of gammaretroviruses [1,45]. These are REV, Trager duck spleen necrosis virus (SNV), chick syncytial virus, Finkel-Biskis-Jinkins murine sarcoma virus, Harvey murine sarcoma virus, Kirsten murine sarcoma virus, Moloney murine sarcoma virus, murine leukemia virus (MLV), Hardy-Zuckerman feline sarcoma virus, Snyder-Theilen feline sarcoma virus, feline leukemia virus (FeLV), woolly monkey sarcoma virus (WSMV), gibbon ape leukemia virus (GALV), koala retrovirus (KoRV), and porcine type-C oncovirus (PCOV) (Table 1). Hosts of the 15 virus species are then birds (3×), mice (5×), cats (3×), primates (2×), pig (1×) and koala (1×). Unfortunately, the ICTV list is not very practical when looking for retroviruses that could threaten humans as it contains seven sarcoma viruses, which are replication defective, and originated through recombination of retroviral sequences with cellular oncogenes [46,47,48]. WMSV likely developed as a defective, oncogene-carrying GALV variant in a pet monkey after co-housing with a GALV-infected gibbon [49,50]. The WMSV helper virus, however, is closely related to GALV [51]. The porcine PCOV designation probably encompasses all pig gammaretroviruses, although the sequence provided by ICTV is one of a defective ERV with limited homology to the replication-competent porcine endogenous retroviruses PERV-A, -B, and -C. PERV-A and -B, as well as naturally occuring A/C recombinant viruses can infect humans cells, with especially some of the A/C recombinants replicating at high titers [52]. Thus, regarding putative infectivity for humans, we are left with eight ICTV-recognized gammaretroviruses: the three avian species, and the mammalian MLV, FeLV, GALV, KoRV, and PCOV/PERV (Table 1).

Selection of ICTV-Listed Betaretroviruses with a Gamma-Type Env Gene

The env genes of the alpha/gamma/deltaretroviruses (‘gamma-type’ env) differ significantly from those of the beta-/lentiretrovirus genus (‘beta-type’ env). In contrast to the beta-type Env, gamma-type Env subunits are covalently linked [17]. Furthermore, gamma-type env genes encode a highly conserved ‘immunosuppressive domain’ in the transmembrane subunit, and require the viral protease for cleavage of the cytoplasmic tail during virion maturation [18]. Retroviruses with a gamma-type Env use multi-membrane spanning transporter proteins as entry receptors, the majority belonging to the solute carrier (SLC) superfamily of cellular transporters [18]. Recombination involving the env gene is common in beta- and gammaretroviruses [17]. Recombinant viruses, with betaretroviral gag-pol genes and a gamma-type env gene, have been designated type D retroviruses in the past based on virion maturation and morphology. ICTV classifies type D recombinants as betaretrovirus, albeit with the remark that they carry unique Env proteins. The ICTV member species list contains, out of the five betavirus members, three examplar isolates of such type D viruses: the langur virus Po-1-Lu, Mason-Pfizer monkey virus (MPMV), also known as SRV-3, and NWM squirrel monkey retrovirus (SMRV). Other examples of type D retroviruses are the replication-competent ERVs OWM SERV, and feline RD-114.

Replication-Compentent ERVs not Listed by ICTV

Targeting the germline, putatively through infection of the early embryo, is a hallmark of the beta- and gammaretrovirus replication cycle. The ICTV list contains many examplar isolates which have integrated counterparts in the host germline. In fact, except for the sarcomaviruses and GALV, all have. Although host defenses suppress expression, many proviruses retain some capacity to replicate and produce infectious virions for a long time after integration. The above list of potential retroviral threats should thus be extended with additional replication-competent ERVs that have shown infectious potential, or for which virions have been detected in cell culture, which are the type D ERVs mentioned in section 2.1.2., but also the primate ERVs baboon endogenous virus (BaEV), MAC-1 isolated from Macaca arctoides and CPC-1 from the King colobus, Colobus polykomos [53,54,55,56]. Table 1 gives an overview of gammaretroviruses and betaretroviruses with a gamma-type env gene discussed in the review.

Search Strategy

The PubMed database [57] and Google Scholar [58] were manually searched for peer-reviewed publications on the topic of human retrovirus infections and endogenous retroviruses, using appropriate terms to the topic, and combinations thereof, such as specific virus names from section 2.1, “gammaretrovirus”, “class I ERV”, “type C retrovirus”, “HERV”, “detection”, “human”, etc. Abstracts were inspected for relevance; from the selected abstracts, only those for which a full-length publication could be retrieved were read and included. In addition, references in the retrieved papers were inspected and used when appropriate. Year of publication was not a limiting factor.

3. Results

3.1. Germline Gammaretrovirus Infections in Humans in the Past

Retroviruses with homology to gammaretroviruses integrated in the vertebrate germline already long before the rise of the order of primates in the late Cretaceous, 70-80 million years ago (mya) [11,59,60]. From then on, gammaretroviruses continued infecting primates [14,61,62]. Human endogenous retrovirus type W, HERV-W, entered the ancestral primate germline approximately 63 mya, thus around the time of the prosimian/simian split, which is estimated to have occurred around 55.8-70 mya [63]. HERV-Pb (27-36 mya), HERV-E (10.7- 41.3 mya), HERV-V (̴ 43 mya), HERV-T (̴ 32 mya) as well as HERV-H, HERV-FRD and HERV-IP-T47D entered the primate germline just before, or around the time when the NWM branched off from the OWM around 34-56 mya [64,65,66,67,68,69,70]. HERV-R and HERV-I ended up in the ancestral human genome around 28 mya, thus around the split of the ape lineage from that of the OWM [71,72].
After integration, ERVs commonly proliferate for substantial periods of time, but humans did not acquire any new ERV lineage after 30 mya [73]. The only lineage expanding after that time was the betaretrovirus HERV-K, where polymorphic integrations still exist in the human population [73,74,75]. Interestingly, in the genus Homo, mainly gammaretroviral env genes have been domesticated, either as syncytins involved in placentation, HERV-W, HERV-FRD, and HERV-V, or as antiviral proteins, HERV-T and HERV-V [66,70,76,77,78,79].

3.2. Ancient Gammaretroviruses in OW Primates with no Evidence of Human Infection

Focussing on the OW primates, the African and Asian primate lineage from which humans evolved, it is clear from cell culture, DNA hybridization studies and genome analyses that, especially the Cercopithecinae and Colobinae repeatedly suffered from gammaretrovirus infections during their evolutionary time, for which the proviruses did not end up in the ape/hominoid lineage, and thus presumably did not infect ancestral apes and hominoids [34]. Examples here are, for instance BaEV, Papio cynocephalus endogenous virus (PcEV) and MAC-1 [54,55,80]. A few of these, including BaEV and MAC-1, can be induced from monkey tissues to produce infectious virions, suggesting that their germline acquisition happened relatively recently [53,81,82,83,84]. Productive infection of a human fibroblast cell line with syncytium formation was seen for BaEV, showing that the absence of BaEV sequences in humans is not due to an infection block at the cellular level [85]. Nowadays, lentiviral vectors pseudotyped with BaEV Env are routinely used to transduce human cells of the myeloid and lymphoid lineages, confirming that BaEV can interact efficiently with human primary cells [86]. It must be noted, however, that the BaEV env gene was modified in various ways to increase performance. For instance, in one construct, the cytoplasmic tail of the wild type gene was replaced by that of MLV [86,87]. MAC-1 was originally isolated by coculture of a stump-tailed macaque spleen cell line with the human A549 carcinoma cell line [54]. Cell-free supernatant from the culture was then used to transmit the virus to non-infected A549 cells, showing that MAC-1 can efficiently replicate in human cells [54].
A type D retrovirus, SERV, is also present in OWM, but is again not found in ape or human genomes [16,88]. SERV is closely related to the exogenous type D simian retroviruses, SRVs, which are the causative agents of immunodeficiency and neoplasms in captive macaques [89,90,91]. The African green monkey kidney tissue-derived Vero cell line, widely used for the production of viral vectors and vaccines, can produce SERV and BaEV virions upon chemical induction [84,92,93].
Another interesting OWM gammaretrovirus is Pan troglodytes endogenous retrovirus (PtERV1), also known as PtG1a or CERV1, which is, as its name suggests, found in chimpanzee and in gorilla genomes, but not in Asian –lesser– apes and human DNA [29,83,94,95]. The high copy numbers of PtERV1 proviruses in the two African ape genomes are not due to common descent, but arose from independent transmissions [94,96]. Human cell lines naturally expressing the PtERV1 receptor SLC52a2, a riboflavin transporter, are susceptible to infection with PtERV1. Therefore, the absence of the virus in humans has been attributed to either resistance, possibly through the expression of antiviral factors such as APOBEC3 or TRIM5alpha, or to a combination of time and place, with human ancestors not being present in Africa during the time the virus was circulating there [94,97]. Human and chimpanzee TRIM5alpha, but not the gorilla variant, have been shown to be capable of restricting a resurrected PtERV1 core protein [98]. Since the restriction capacity depended on a single amino acid change, it could be that chimpanzee TRIM5alpha mutated, or that a variant was strongly selected for, after PtERV1 infection [98]. Alternatively, TRIM5alpha activity may not be the only factor protecting against PtERV1 invasion [98].
In summary, although gammaretroviruses have been able to infect the germline from the very beginning of the emergence of mammals, and gammaretrovirus species flourished in OWMs from the late Miocene till present times, humans have so far avoided getting infected. At least, germline evidence for infection is lacking. Similarly, the Asian orangutan shows no evidence of having contracted any gammaretrovirus infection after speciation, but African apes have been susceptible to at least one OWM gammaretrovirus, namely PtERV1.

3.3. Gammaretrovirus Detection in Human Samples in the Recent Past

Inspired by the finding of viruses in transmissable chicken and mouse malignancies in the first half of the last century, which provide an appealing explanation for cancer development, screening of human samples for oncogenic –retro– viruses became a favorite topic with many researchers [99,100,101,102]. In the 1960s and -70s, methods employed were mainly cell culture, inoculation, electron microscopy and RNA-dependent-DNA polymerase assays, which resulted in the detection of type C retroviral particles in, or from, human malignancies, or an indication of retroviral replication. As type C virions are characteristic for both gamma- and deltaretroviruses, hybridization studies or sequence analysis is required for further identification. In addition, using either Mg2+ or Mn2+ in reverse transcriptase (RT) assays may help to differentiate, as gammaretroviral RT prefers Mn2+ over Mg2+, with the reverse being true for deltaretroviruses [3]. Type C particles obtained from a cutaneous T-cell lymphoma of which the RT showed a preference for Mg2+, were later indeed shown to belong to a different retroviral lineage, the deltaretroviruses [3]. Studies on detection of type C virions in human samples withour further analysis will therefore not be considered here.

3.3.1. Cell Culture-Associated Gammaretrovirus Detection in the 1970s

McAllister et al. inoculated human rhabdomyosarcoma (RD) cells into kittens, and recovered the RD-114 cell line from an ensuing brain tumor [103]. The cell line expressed a gammaretrovirus which they believed to be of human origin as it lacked homology to FeLV, or any other ‘well-studied’ type C virus [103]. RD-114 was soon shown to be an inducible endogenous virus of cats, passaging of the human tissue in kittens being responsible for its ‘human’ infection [104,105,106]. Another type C virus, or possibly two, were isolated from a patient with acute myelogenous leukemia, and showed homology to WMSV and GALV [107,108,109,110]. The virus, named HL23V, was likely the result of a laboratory contamination [111,112,113]. The same was true for a BaEV-variant with partial homology to HL23V, isolated from tissues of six leukemic patients, and a distinct type C virus with homology to Rauscher murine leukemia virus, in six patients with either leukemia, Hodgkin’s disease or multiple myeloma [114,115]. The findings led the authors to speculate that “the results suggest acquisition of at least three types of type-C viral sequences in the human population” [115]. As all the above experiments used either passage of human tissue in animals, or coculture with animal cell lines, and animal material was commonly present and propagated in the labs, contamination from animal sources is the most plausible explanation for the findings. In one of the publications this option was shortly considered: “although obviously a very remote possibility, it was conceivable that the results described here were due to the selective, accidental contamination of the leukaemic tissues with primate tissues contamination”, since “this laboratory uses DNA from many non-human primates” [114]. But even after detecting 3-8% hybridization of a labelled baboon DNA probe to 3 out of 6 human leukemic samples, the authors surprisingly stated that “the human leukaemic DNA samples lacked baboon-specific sequences” [114].
Since all the above results were obtained in the pre-PCR era, and were based on virus propagation, virion detection and DNA hybridization, it is clear that animal gammaretroviruses can readily infect, and spread in, human cell lines under laboratory conditions.

3.3.2. MSRV is an Endogenous Retrovirus, HERV-W

When a transmissible retrovirus, multiple sclerosis associated retrovirus (MSRV), was isolated from multiple sclerosis (MS) patients, it was immediately suspected to be of endogenous origin [116,117,118,119]. Indeed, further analysis showed that MSRV was derived from endogenous retroviral sequences of the HERV-W family of gammaretroviruses [120]. The HERV-W env gene product was later identified as syncytin 1, a protein involved in human placental morphogenesis [76]. A systematic review showed that HERV-W sequences are overexpressed in MS, which accounts for the findings in patients [121]. Complementation between fragmented genomes likely accounted for the RT activity measured and the virions observed.

3.3.3. The XMRV Case, 2006-2012

A next gammaretrovirus with homology to murine gammaretroviruses, termed xenotropic murine leukemia virus-related virus (XMRV), was detected in human prostate cancer tissue in 2006 [122]. Subsequent studies associated the virus with myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS), a finding strongly emphasized by the senior author, which caused quite a stir among the scientific community and the despairing patients [122]. In 2011, after many worldwide follow-up studies, which mostly failed to find the virus whatever the patient group, XMRV was definitely shown to be a murine contaminant of the orginal human prostate tumor xenograft that had earlier been passaged in mice [123]. The first reports have now been retracted by the editors of the respective journals [124,125]. In 2005, screening of eight human melanoma cell lines had already shown that half of them were productively infected by MLV [126]. The authors therefore strongly recommended “mandatory testing of melanoma and other human cell lines for contamination with infectious MLV or other animal retroviruses, ……, in order to avoid artificial experimental data”, but clearly their advice was ignored. In 1993, HIV-infected HUT78 cells were found to be co-infected with GALV [127,128]. In 2008, contamination of three human cell lines by MLV or by GALV was reported [129]. In 2009, a 293T cell batch was found to be infected with both a MLV variant and a type D retrovirus [130]. Six further human cell lines were discovered to be contaminated with SMRV after its initial detection in a virus-producing human lymphoblastoid cell line [131,132]. A 2015 screen of 577 human cell lines for MLV found that 19 of them contained MLV sequences, with 17 being productively infected as evidenced by PCR analysis, sequencing, and reverse transcriptase activity [133]. Passaging of human tumor cells in immunodeficient mice, and the use of murine feeder cells in cell cultures were proposed as the most likely explanations for the findings [126,133]. Once a cell line is infected with MLV, further contamination of other cell lines exacerbates the problem. Sometimes a subclone, but not the original cell line, was found to be MLV-positive, as was the case for the HeLa subclone BT-B [133]. In the same study, blood samples from 30 healthy volunteers were all negative for MLV [133]. Thus, the widespread laboratory contamination of human cell lines with mouse-derived gammaretroviruses again underlines the ease with which human-derived cells can be infected, and demonstrates their ability to support the subsequent replication steps.

3.3.4. Human Retrovirus-5 Turned out to be a Rabbit Endogenous Retrovirus

In addition to contamination by gammaretroviruses, a betaretrovirus, tentatively named human retrovirus 5 (HRV-5) was detected by PCR analysis in salivary gland tissue from a patient with Sjögren’s syndrome [134]. Soon, HRV-5 was shown to result from a laboratory contamination with a rabbit endogenous retrovirus, RERV-H [134,135]. An earlier search for a virus in the condition through exposure of a human lymphoblastoid cell line to salivary tissue from Sjögren’s syndrome patients, and analysis of tissue without coculturing, had led to the detection of, not further characterized, retroviral activity associated with intracisternal type A retroviral particles [136,137]. Later, researchers suggested a causal role for other retroviruses, namely HTLV or HIV, in the disease [138]. At the moment, Sjögren’s syndrome is considered an auto-immune disease, associated with Epstein-Barr virus infection [139].
A summary of human “rumor’ viruses up till 2008, many of them being HERVs, is provided by Voisset et al. [140].

3.4. Retroviruses Currently Infecting Humans

To date, human-specific lineages of delta- and lentiretroviruses, HTLV and HIV, respectively, have firmly established themselves in the human population, while zoonotic transmissions of spumaretroviruses are ongoing [34,141,142].

3.4.1. The Human Deltaretroviruses

The circulating human lineages of deltaretroviruses are known as HTLV 1-3. They have been transmitted from OW primates starting around 30,000 years ago [143]. More recent cross-species transmissions are rare, but do occur [144,145,146,147]. HTLV-1, initially described as a type C retrovirus detected in PBMC from a patient with a cutaneous T-cell lymphoma and from two T-cell lymphoblastoid cell lines, is the causative agent of adult T-cell leukemia (ATLL) and HTLV-1-associated myelopathy/tropical spastic paraparesis [3,148]. Until now, no pathology has been associated with HTLV-2 and -3 infections [147,148].

3.4.2. The Human Lentiviruses, HIV-1 and HIV-2

The history of the, relatively recent zoonotic transmissions of simian lentiviruses from chimpanzees and mangabeys to humans around the turn of the 20th century is all too familiar [149,150,151,152]. In short, two simian immunodeficiency virus (SIV) species, from African apes and sooty mangabeys, respectively, were transmitted, likely through hunting and bushmeat preparation, from non-human primates to humans, resulting in human-adapted lentivirus genera HIV-1 and HIV-2, respectively [149,150,151,152]. At least four transmissions of ape SIV strains, named HIV-1 group M, N, O and P, have been recorded [149,150,153]. Of these, group M viruses are responsible for the HIV-1 pandemic, while the other three are uncommon. HIV-2 from Western Africa can, similar to HIV-1, cause acquired immunodeficiency syndrome, AIDS, but appears to decrease in prevalence [154,155,156,157]. Undoubtedly, there have been more transmissions from SIV to humans, but not all of those were probably successful, as viral adaptations are needed when changing host species [158,159,160]. However, even when the virus was able to replicate after the initial transmission, keeping in mind the high mortality seen after HIV infection, together with the slow dissemination of a retrovirus in a confined population, namely spreading only by intimate contact, it can be assumed that any prior SIV transmission has been invisible to the world.

3.4.3. Spumaviruses in Humans

Spumaviruses, also known as foamy viruses due to the frothy appearance of infected cell cultures, are distinct from the Orthoretroviridae, and reside in their own family of Spumaretroviridae [1]. Spumaviruses are found in many mammalian species, including cattle, horses, cats, bats and primates [161]. Phylogenetic analysis shows strong virus-host co-evolution, with cross-species transmission predominantly seen within the host clade [161,162]. A survey under 204 individuals with long-term, high-risk exposure to domestic cats indeed provided no evidence of feline spumavirus infection [163,164]. However, seroreactivity to bovine spumavirus antigens was seen in 7% of individuals with professional exposure to cattle [162]. Surprisingly, despite the high prevalence of spumaviruses in primate species worldwide, including the three ape species, there is no genuine human foamy virus strain [161]. All spumaviruses detected in humans are the result of cross-species transmissions, including the ‘human’ foamy virus isolated in 1971, that turned out to be of chimpanzee origin [161]. The main mode of simian foamy virus (SFV) transmission from non-human primates to humans is through biting or scratching [141,142]. SFV infections are prevalent in African hunters and in laboratory personnel, and are mainly associated with biting accidents [141,142,165]. As these infections are persistent, there is an increasing possibility of a human-specific strain emerging [141,142]. Yet, human-to-human transmission of SFV infections has not –yet– been documented [164,165]. Interestingly, SFVs transmitted from NWM do not persist in humans, even though antibody prevalence in primate workers can be as high as 12-19% [166,167].

3.4.4. Unresolved Human Retrovirus Infections

Apart from the established human deltaretrovirus and lentivirus lineages recognized together with the frequently documented cross-species spumavirus transmissions, a few other retroviruses have been postulated to circulate in humans. Since the discovery of a retrovirus in mammary tumors of mice, the betaretrovirus mouse mammary tumor virus (MMTV), scientists have been searching for a human variant causing similar pathology. Early EM analysis discovered viral particles, including B type particles, in human breast cancer tissue and in human breast milk [168,169,170]. Subsequently, RT activity and nucleic acid homologous to MMTV were found in human milk from women with a history of familial breast cancer and in breast tumors themselves [171,172]. Although evidence is mounting for a causative role of an MMTV-like human betaretrovirus, HBRV, in certain types of breast cancer, in lymphomas, and, possibly, also in primary biliary cholangitis, not everyone is convinced yet [5,6,7,8,9,173,174]. BLV, a bovine deltaretrovirus that can infect human cells, has likewise been associated with breast cancer [175], see also [176,177].
Summarizing contemporary human retrovirus infections, it is clear that human-specific virus lineages exist for delta- and lenti-retroviruses, that humans are repeatedly infected by simian spumaviruses, and that there is mounting evidence for a rodent-derived betaretrovirus circulating in the human population. Since alpha- and epsilonretroviruses are not known to infect mammals, the only ‘missing’ infection in humans is then one by a member of the gammaretrovirus genus.

3.5. Current Infectious Gammaretrovirus Species

3.5.1. Infectious Avian Gammaretroviruses

The ICTV-listed avian gammaretroviruses, three strains of REV, constitute an interclass transmission of a mammalian gammaretrovirus in the recent past [26,27,178,179]. Wildtype REV is not infectious for human cell lines [180,181]. An earlier study, however, reported that SNV can infect HeLa cells with establishment of an integrated provirus, but that a posttranscriptional block prevents viral replication [182]. REV infection in poultry can be diminished by testing and selection but flocks can still become infected through infection, or vaccination with fowlpox virus or Marek’s disease virus, a herpesvirus, vaccines due to REV’s ability to integrate into the genome of large DNA viruses [181,183,184,185]. Fortunately for humans, avipoxviruses, of which fowlpox is the prototype, have a very limited host range, infecting only avian species, in contrast to other poxviruses [186].

3.5.2. Infectious Mammalian Gammaretroviruses

Research summarized in section 3.3.1 on contaminating mammalian gammaretroviruses in human cell lines showed that many non-avian members of the genus, especially the MLV-like viruses, are capable of infecting humans at the cellular level [187]. Certain porcine PERV variants also have the ability to do so, with integration of a provirus, although they have not been observed to replicate or spread in humans after xenotransplantation of pig tissue [188,189,190,191]. GALV replicates efficiently in a human T cell line, but very poorly in human primary T cells [129,192]. KoRV type A, KoRV-A, replicates very well in the human HEK293T cell line, while lentiviral vectors pseudotyped with the wt KoRV-A env gene efficiently transduce human primary monocytes as well as B- and NK-cells [193,194,195]. An Australian bat virus related to KoRV and GALV, Hervey pteropid gammaretrovirus (HPG), is also able to infect human, though not mouse, cells [196]. Since GALV and KoRV are closely related, it has been suggested that they originate from a common source, such as a rodent or possibly a bat species [31,49,50,197,198,199]. GALV is has first been reported in captive gibbons from research colonies, while it could not be isolated from 76 gibbons kept in North-American zoos, nor in 23 gibbons from seven species residing in the European Union [200,201]. Still, anti-GALV capsid p30 antibody prevalence was 28% in the American zoo gibbons, whereby Nomascus gibbons showed a higher prevalence than Hylobates and Symphalangus gibbons [200]. It was not investigated whether the seroprevalence could be due to antibody cross-reactivity to, possibly, ERV epitopes [201]. Earlier, Kawakami et al. reported shedding of an infectious C-type virus by four healthy Hylobates gibbons kept as pets or in zoos, and by two laboratory animals, though it is not clear whether or not the virus was GALV [202]. GALV can be transmitted between animals, but there is no evidence for seroconversion or virus replication in human gibbon handlers, nor were any of the 338 pre-treatment samples in a clinical trial with GALVenv-carrying retroviral vectors positive for such sequences [203,204]. Contrarywise, it has been speculated that infected humans were the source of GALV in gibbons [205]. Surprisingly, GALV infection in gibbons has not been reported beyond the 1970s, strenghtening the hypothesis that iatrogenic transmission in US research facilities and not natural infection in the wild, has been the source of the reported cases [201].Another monkey virus, SRV4, isolated from Japanses macaques with an infectious hemorrhagic disease, has been reported to replicate in human cell lines and in humanized mice [206].
A post-transcriptional block was observed for FeLV type B replication in human PBMC, although most human cancer-derived cell lines but also human non-transformed keratinocytes and lung fibroblasts were fully permissive [207]. FeLV type A has a low affinity for the human variant of its receptor, thiamine transport protein 1 (THTR1), and is thus an unlikely candidate for a zoonotic transmission [208]. Natural isolates of FeLV commonly consist of mixtures of FeLV-A and -B, and can be shed in cat saliva and other bodily fluids [207]. A survey of veterinary conference attendees for evidence of zoonotic infection by feline retroviruses detected no anti-FeLV antibodies nor viral DNA in the highly cat-exposed participants [163]. However, virus transmission from local domestic cats to other felines, namely to Florida panthers, to Iberian lynx and to North American pumas, has been recorded [209,210,211].

3.5.3. Replication-Competent ERVs and the Type D Retroviruses

Both endogenous SERV, as well as exogenous SRVs, have the capacity to generate virions, while SRVs can spread within primate colonies [84,89,90,91,92]. It can be speculated that, given their high sequence homology to SERV, exogenous SRVs, including MPMV/SRV3 and Po-1-Lu, at least partly derive from monkey ERVs [91,212].
Simian D type viruses and the feline ERV RD-114 do replicate in human cell lines, with the induction of RT activity and syncytium formation [85,103,213,214]. The same is true for other replication-competent ERVs, such as BaEV, MAC-1, and CPC-1 [53,54,215]. However, being able to replicate in a cell line is not a reliable predictor of the ability to replicate in primary cells or to productively infect an organism. Evidence for a simian D type retrovirus infection, with virus isolation, serum reactivity and PCR amplification of viral DNA was found in Burkitt’s-type B-cell lymphoma biopsies obtained from a patient with AIDS [216]. As the patient had had no exposure to monkeys, and an autopsy was denied, the authors could not completely rule out contamination since type D viruses have also been discovered as cell line contaminants [131,216,217,218]. Lerche et al. later reported serological evidence for a type D virus infection in two healthy individuals occupationally exposed to monkeys [219]. However, the virus could not be isolated, PCR-amplified or transmitted, suggestive of very limited replication, if at all.
Since gammaretroviruses are frequent laboratory contaminants but have not yet been isolated from humans, it is likely that they need to overcome significant obstacles to accomplish successful cross-species transmission. That observation corresponds with the lack of any gammaretrovirus on the 2024 WHO priority pathogens list, where HIV (Lentivirus humimdef1) is the only retrovirus assigned a Public Health Emergencies of International Concern medium risk level for pandemic potential [220]. Still, the relatively common cross-species transmissions of gammaretroviruses observed, the ease of human cell culture infections, and the many animal species carrying infectious variants that surround us, should keep us alert.

3.6. Genetic and Immune Factors Associated with –Gamma–Retrovirus Resistance in Humans

Already some time ago, it was seen that the so-called animal RNA tumor viruses were effectively lysed by human serum through complement activation [221]. No such effect was found for HTLV and HIV [222,223]. Later, it was shown that inactivation through the complement pathway depends both on the producer cell line, thus the host species, and the viral Env protein, and is therefore an unpredictable mechanism to prevent virus infections [224]. Over the evolutionary timespan, humans have lost the ability to synthesize various carbohydrate antigens, resulting in a loss of certain membrane-associated glycans, such as the complex alpha-gal epitope and N-glycolyl neuraminic acid (Neu-5Gc) [225]. The alpha-gal epitope was lost early in evolution of the OW primate lineage, but is still present as a major carbohydrate antigen in non-primate mammals, prosimians and NWM [225]. Naturally occurring human anti-alpha-galactosyl antibodies recruit complement factors to inactivate retroviruses expressing the target carbohydrate on their envelope, offering a defense against zoonotic infections [225,226]. However, depleting anti-alpha-gal antibodies did not enhance the risk of PERV infection in experimental pig-to-baboon organ transplantations [227].
Should an incoming virus have survived the first innate immune barriers, receptor incompatibility would be a next hurdle. Retroviruses with a gamma-type Env use a member of the SLC superfamily as a cellular receptor [18]. Viruses of the so-called RDR superinfection interference group, comprising, amongst others, BaEV, RD-114, the primate D-type viruses, and the avian gammaretroviruses, all use the neutral amino acid transporters SLC1a4/ASCT1 and/or SLCa5/ASCT2 for entry [18,32]. Human syncytin 1, a repurposed HERV-W Env, also binds to SLC1a4/5 [228]. SLC20a1/PiT1 is used by e.g. KoRV-A, FeLV-B, and GALV [18]. Competition for entry by another Env using a same receptor can lead to superinfection resistance, whereby an incoming virus cannot enter a cell as its receptor is already occupied or downregulated by another Env [229]. ERV Envs can be domesticated to become part of the innate immune system. In humans, a HERV-T Env, named suppressyn, acts against SLC1a5/ASCT2-using retroviruses, while HERV-W Env is capable of restricting avian SNV infectivity through SLC1a4 [78,79,230]. SLC1a4/5 proteins are well conserved among vertebrates, although chickens have lost the SLC1a5 gene [231]. Interferon-stimulated genes (ISGs) are genes activated after interferon release following virus infection [232,233]. Such restriction factors are, for example, the APOBEC3 family, TRIM5α, tetherin, SAMHD1, the HUSH complex, ZAP, and SERINC5; they inhibit the viral replication cycle at specific points [234,235].
Thus, viral proteins themselves have been recruited by the innate immune system to combat further virus infections showing that host defenses are surprisingly adaptable. Still, the immune system can never be 100% efficient. In immunocompetent individuals, the majority of infections will probably be prevented, but this may be different in those with a genetic flaw or an acquired immunodeficiency. However, the combined action of multiple antiviral mechanisms may explain why, despite regular exposure to animal gammaretroviruses, humans appear to be quite resistant to infection.

3.7. Exposure Risk and Gammaretrovirus Infections in Humans

3.7.1. Natural Exposure to Gammaretroviruses

The most effective way of not getting infected is of course not being around when and where a virus is circulating. It has been speculated that humans did just that when PtERV1 was spreading to ancestral chimpanzees and gorillas in Africa some 3-4 mya [94]. Possibly, ancient hominids were in Eurasia at that time, or alternatively, they could have lived in a different habitat, such as a savannah environment instead of tropical forest [94]. Another explanation, although less appealing to the imagination, would be incomplete lineage sorting, with a prehistoric loss of provirus-carrying chromosomes due to the relatively small effective population size of human ancestors [94].
It has been shown that shared habitats are, and have been, important for natural cross-species transmission, although for instance bats may carry retroviruses over long distances within their territory [28,30,49,236,237,238]. Retrovirus particles are unstable in the environment, and therefore need close contact between individuals for transmission, that is, a direct exchange of body fluids such as saliva, breast milk or blood. Thus, exposure to a virus-carrying species, both in geography and in ecology, as well as through behavior and diet, e.g. hunting for meat, with the danger of being scratched or bitten, are important for a zoonotic transfer [239]. A systematic analysis suggested primates to be the most likely source of future human retrovirus infections, both through bushmeat hunting and via laboratories [240]. Gammaretroviruses carried by extant primates are mainly found in OWM, such as the exogenous viruses SRV and GALV, and a variety of replication-competent ERVs. Their primate hosts can be encountered in the wild in Africa and Asia, but are also sold as pets, kept in zoos, maintained in temples, and held in primate research centers, facilitating ample human contact opportunities. Non-primate animal species could be another reservoir for zoonotic viruses, with rodents and bats being prime suspects. However, domestic animals are probably a much bigger virus source due to their large numbers and their proximity to humans. The combined biomass of humans and domestic mammals increased around five-fold since 1850, while the biomass of wild mammals decreased by 50%, so that the latter now comprise only around 10% of the total mammalian biomass on earth [241]. Exposure risks will be further discussed in the next sections.

3.7.2. Man-Made Exposure to Gammaretroviruses

In Eurasia, the domestication of livestock, beginning around 6,500 years ago, enabled the transmission of zoonotic pathogens that were not found in humans before that time [242]. The ongoing surge of the human population accompanied by growing numbers of livestock and pet animals, as well as human-enforced declines in the natural habitats of wild animals makes the human species progressively vulnerable to pathogens carried by animals. Interestingly, (in)breeding and hybridization associated with domestication have been reported to increase the number of ERV loci in both pigs and mice [13,187,243]. So, people have been intensifying their contact with animals on a global and industrial scale for quite some time. But, this practice has not –yet– resulted in a detectable gammaretrovirus infection.
Modern biomedical practices and clinical inventions would be another way of confronting the human population with a gammaretrovirus. As summarized in section 3.3, various gammaretroviruses have been found to contaminate human cell lines in laboratory settings. In a worst case scenario, such a virus could continue to adapt and maybe escape from the lab. However, cell culture contaminations have been reported for decades, with, again, no discernable human gammaretrovirus infection. Inoculation of cell culture products into a large number of humans, as is done with live-attenuated vaccines, could enhance the chances of a productive infection. RD-114 virions have been detected in live attenuated vaccines for dogs made in a cat-derived cell line [244]. The infectious RD-114 present in the Crandell-Rees feline kidney cell line used here originated from recombination between different proviruses [245]. Although RD-114 can use canine SLC1a4/5 for entry, no productive infection was detected in dogs subcutaneously inoculated with a virus stock [246,247]. From some human live-attenuated vaccines, especially Vero cell line-produced Rotateq and Rotarix, both oral anti-rotavirus vaccines, SRV DNA could be PCR-amplified. As DNase treatment eliminated the signal, contaminating African green monkey DNA, but not viral RNA had probably been present in the vaccine batches [248]. Subsequently, it was shown that ‘SRV’ sequences from the Vero cell line were in fact endogenous SERV DNA [84]. SERVagm-Vero did not replicate in human A-204 and Raji cell lines [84]. A study in Japan, analyzing five Vero-based vaccines, plus 783 clinical samples obtained from patients with various diseases and 1000 samples from healthy controls, found no evidence of SERV infection, except that eight healthy controls >30 years showed seropositivity for a SERV Pol antigen, but not for Gag or Env [249]. The vaccines analyzed, all produced in Vero cells since 1980, were three inactivated and two oral live-attenuated vaccines [249]. Live-attenuated mumps, measles & rubella and yellow fever vaccines manufactured in chicken embryos or in a chicken embryonic fibroblast cell line were shown to be contaminated with the alpharetrovirus avian leukosis virus, but human infection in recipients was not observed [36,248,250,251,252]. Of course, biotechnology products made in mammalian cell lines, such as monoclonal antibodies and vaccines, are nowadays routinely screened for contaminating viruses, but no procedure will be 100% reliable [253,254].
Another man-made medical intervention, transplantation of animal tissue or complete organs in humans, which requires lifetime immune suppression, has sofar not resulted in BaEV or PERV infection in recipients of baboon bone marrow, baboon liver or various pig tissues [255,256,257]. Currently, the first xenotransplantation of a kidney originating from a pig carrying 69 genomic edits, including the inactivation of 59 PERV loci, has been reported [258,259]. Other companies aim to reduce the PERV content in pigs destined for xenotransplantation procedures by selective breeding [260]. A clinical trial using kidneys from pigs with 10 genomic edits, none of them PERV elements, has started recruiting in August 2025 [261]. As neutralizing antibodies against PERV can be elicited in rats, development of an anti-PERV vaccine to protect recipients of pig organs has been suggested as an alternative approach [262].
Introducing a gene of interest into humans has been done using MLV-based gammaretroviral vectors [263]. Since safety issues arose, the use of so-called self-inactivating (SIN) vectors effectively reduces the chances of creating a gammaretrovirus in a patient [263,264]. Here, three plasmids, one coding for gag-pol, a second for env, and a third carrying the gene of interest flanked by partially defective LTRs, are used to produce viral particles in vitro. Another way of protecting laboratory personel against MLV when working with lentiviral pseudotyping systems, is the use of an ecotropic, thus mouse-only, MLV env gene, together with transient expression of its receptor, SLC7a1, in the human producer cell line [265]. Besides, alternative delivery methods for nucleic acids, which eliminate the need for retroviral vectors, have now also been developed [266]. However, not everyone has abandoned the clinical use of replicating viral vectors. A worrisome development is the use of non-human viruses as oncolytic or, despite the above-mentioned safety concerns, as gene-delivery agents in cancer treatment. Koppers Lalic and Hoeben estimated the ‘relative environmental risk’ of these replicating retroviral vectors (RRVs) as ‘high’ [267]. RRVs, based on either MLV or GALV equipped with a transgene cassette, have been tried in mouse models first; MLV RRVs subsequently entered clinical trials [268,269,270]. One study analyzed patient samples from a trial on glioblastoma treatment with a MLV RRV, Toca 511, expressing a modified yeast cytosine deaminase to convert the prodrug 5-fluorocytosine into cytotoxic 5-fluorouracil [271]. In patients with detectable viremia, the development of neutralizing antibodies coincided with viral sequences disappearing from the blood [271]. Patient-derived viral sequences suggested APOBEC3-activity during reverse transcription [271]. In 13 patient samples sequenced, MLV did not revert to wildtype, in line with the insert having been reported to be quite stable when directly cloned after the stop codon of the env gene [270,271]. A 2023 study reported no evidence of the presence of replication competent retrovirus (RCR) in 338 pre-treatment and 1,595 post-treatment peripheral blood samples obtained from 60 clinical trials [204]. The authors focused here on RCR during the production process of retroviral vectors to be used in clinical trials, exposing the participants to a replicating virus, not so much on the generation of RCR in treated individuals. In the 1990s, RCR MLV recombinants originating from a producer cell line were found to be the cause of lymphoma in three rhesus macaques receiving experimentally transduced bone marrow cells [272,273,274]. The majority of the studies analyzed in 2023 had used vectors with intact long terminal repeats (LTRs), pseudotyped with a GALV Env [204]. In line with the above results, Farley et al. recommend to unify and reduce testing of third-generation retroviral vector systems, except that they “note that there are conditionally replicating RVs (crRVs) being used for cancer therapy, where generation of RCV (replication-competent virus) risk must be considered in a different light” [275].
A list of gammaretrovirus threats to humans, as well as the their most likely transmission sources with recommendations to decrease the likelihood of transmission, is provided in Table 2. It must be remarked that almost all listed gammaretroviruses exist in an endogenized form in their natural hosts, whereby the probability of provirus activation decreases with time since endogenization. For instance, SERV activation in Vero cells requires chemical induction, while PERV needs mitogenic stimulation to be released from pig primary cells [84,276].
In summary, animal domestication combined with a human-induced decline in the habitat of wild animals, as well as current healthcare inventions, such as vaccination, xenotransplantation, and retroviral vector therapy, facilitate exposure of the human population to animal gammaretroviruses. But, to date, no replication competent virus variant let alone a circulating virus is known to have emerged from these activities.

3.8. Consequences of Novel Human Gammaretrovirus Infections

Assuming a productive novel gammaretrovirus infection, consequences for humankind can be envisaged on both the short term, related to morbidity and mortality from the infection, and on the long term, by influencing evolution through provirus integration in the germline. In order to have a significant effect, the novel virus should be transmittable, and not be limited to a single individual, which may be the case in clinical trial participants, or in immunosuppressed individuals. As retroviruses spread through intimate contact, e.g. between sexual partners, from mother to child in utero or through breastfeeding, sufficient opportunities to do so would be essential. Between humans, transmission through biting and scratching is less likely. Blood transfusion, surgical procedures, or the use of dirty needles could, however, lead to health-care associated transmission. In addition, unlike what is known for HIV and HTLV, gibbons and cats can also shed infectious GALV and FeLV, respectively, in urine and feces, while infectious FeLV is consistently detectable in saliva of viremic cats [202,280,281,282,283]. GALV production was found to be very high in the post-mortem material from the oral cavity of a gibbon with lymphocytic leukemia, likewise implicating the possibility of oral transmission [284].
Regarding pathology, the frequent use of the term ‘leukemia’ in the names of gammaretroviruses already hints at the most common condition associated with infection. Indeed, malignancies, such as leukemia, lymphoma, and sarcoma prevail in animals, but immunosuppression and neurological conditions are also seen [1]. The most recent novel primate gammaretrovirus is GALV, whereby infection is either asymptomatic or is associated with acute leukemia and lymphoma [200,284,285]. Recency of the virus is suggested by the lack of endogenous GALV counterparts in the gibbon genome [284,286]. For related KORV, which is a relatively young pathogen of koalas, germline proviruses are already present [287,288]. Which brings us to the long-term consequences of retrovirus infections. The tendency to target the early embryo leads to germline integrations that can affect evolution at the species level. Random integrations can activate or inactivate genes, while chromosomal rearrangements can be induced by recombination between different proviruses. Subsequent activation of a provirus can result in changes in protein expression and thus in cellular biology. For the most recently active human ERV family, HERV-K (HML-2), insertional polymorphisms were indeed found to create genetic variation, while specific HML-2 expression profiles are associated with various diseases [289,290,291]. In addition, novel retroviruses might develop through recombination with ERV sequences. Throughout retroviral evolution, env-snatching is common in beta- and gamma-retroviruses, whereby preferentially gamma-type envs are targeted, resulting in type D retroviruses, but also in gamma-gamma recombinants [32,80,292,293,294]. In humans, a gamma-env could potentially recombine with HERV-K sequences, of which intact LTRs and genes are encoded in our genome [295,296]. The sarcoma viruses listed by ICTV indicate that gammaretroviruses do also recombine with non-ERV host genes. Although such recombinants will need a helper virus for replication, the capacity to shuttle host genes between cells would increase their oncogenic competence.
In the short term, a productive gammaretrovirus infection could lead to significant morbidity and mortality with the possibility of a new virus emerging, which could lead to an epidemic. Should the retrovirus reach the human germline, it is likely to influence evolution of the species.

4. Discussion

There is no evidence that ancestral humans were ever infected by a gammaretrovirus in the past 30 million years, nor that such a virus is present in the population today. Our cousins, the apes, were apparently also spared, except for an encounter with PtERV1 around 3-4 mya [94]. Gammaretrovirus infections, however, were common in the OWM lineage, with multiple inducible ERVs testifying to the significant virus burden in the recent past. Other retroviruses have had less trouble spreading between OW primates and us, as exemplified by the recent HIV pandemic, and the past and ongoing infections with primate foamy- and deltaretroviruses. For HIV and HTLV, hunting and butchering non-human primates has been proposed as the most likely transmission route, linking diet to infection risk [145,146,150,297]. For SFV, any exposure to non-human primates will do [113,297,298]. The latest primate gammaretrovirus victims are apparently gibbons kept in research facilities. The failure to detect GALV in zoo gibbons together with a lack of germline integrations suggests a recent acquisition. Human activity was suspected of being causal here, as blood and tissue from malaria, kuru and dengue patients as well as rodent material has been inoculated in the 1960s into gibbons in US medical research facilities in Thailand and in the USA [201,205]. A cross-species transmission from a wild animal could, however, likewise be responsible, especially since the Thai institute kept a free-ranging colony of indigenous gibbons. GALV-like replication competent proviruses can be found in the rodent species Melomys and Mastomys, although virus isolation from six Australian Melomys burtoni blood samples and tissues was unsuccessful [50,197,299]. Since GALV infection has not been reported in gibbons since 1978, and the number of cases was limited anyway, the risk of human infection is estimated to be low [201]. Co-housing of monkeys in research facilities or elsewhere is an effective way of spreading primate retroviruses, as evidenced by SIV transmission from sooty mangabeys to macaques, SRV transmission between macaques, and GALV transmission to a NWM [49,50,51,300,301,302]. Despite the vulnerability of captive non-human primates to retroviruses, human caretakers have not yet found to be infected, except with SFV [165,203,303]. Type D SRVs have historically circulated in captive macaques, but improved diagnostics, selective breeding and vaccination have nowadays decreased their prevalence significantly [277,304,305]. Research groups have recently reported that, for unexplained reasons, they were unable to infect macaques with SRV, and also that the virus is no longer capable of transmitting between cage mates [306,307]. Should macaques have become less susceptible to SRV infection, then the risk of SRV spreading to humans would decrease concomitantly, although monkeys in the wild may still carry the virus. Further retroviruses with a gamma-type env mainly exist in primate DNA as latent, but inducible, proviruses. Nevertheless, expression in cell culture of, for instance, BaEV, MAC-1, or SERV, requires chemical activation, cocultivation and/or long-term culture, suggesting that casual contact with monkeys will not be sufficient for transmission.
Contact with live animals could, theoretically, lead to FeLV transmission from domestic cats, as around 600 million cats around the globe live near humans [308]. FeLV is limited to domestic cats and closely related Felis species, suggestive of a relatively recent origin [309]. Prevalence of progressive infections, whereby the virus is not contained by the immune system, but is shed in, for instance, saliva, varies. It ranges between 0-21% in domestic cats, and rises up to 28.4% in European wildcats (Felis sylvestris) [278,310,311]. Nonetheless, despite the high prevalence in cats and the regular exposure of humans to them, no human FeLV infection has been recorded so far [163,207]. Similar to the situation with monkey ERVs, the inducible RD-114 provirus is hardly expressed in healthy cats, and would therefore not be easily transmitted through direct contact [213]. Exposure to koalas constitutes a risk of contracting KORV, especially since KORV-A has a prevalence of 100% in koala populations, while non-A strain prevalence ranges between 10.3-93.9% [312,313,314]. Yet, human infections have, again, not been reported. As for the avian gammaretrovirus group, it has been suggested that the REVs were introduced into experimental birds through inoculation of contaminated Plasmodium lophurae stocks in the 1930s [27]. Nowadays, REV spreads in wild and domestic birds as a hitchhiker in the fowlpox genome and in in live attenuated fowlpox vaccines, respectively. As avipoxviruses do not infect mammals, human infection through bird exposure is unlikely. However, it would be interesting to analyze human herpesvirus genomes for retroviral integrations, as for instance, virulence of Epstein-Barr virus varies globally, which has been attributed to strain variation [315].
Modern lifestyle, in the form of medical developments, such as xenotransplantation and vector therapy, is a novel risk factor to contract murine or porcine gammaretroviruses, as millennia of living together with mice and pigs did not result in a human MLV or PERV infection. Fortunately, PERV does not appear to be expressed nor to be transmitted from transplanted pig organs, and PERV-free pigs are available now. Treatment with RRVs is limited at present, not only because people are aware of the risks, but also because the therapy failed to meet expectations [316,317]. Still, an analysis of blood samples of clinical trial participants administered a gammaretroviral RRV gives us a glimpse of what could be the early consequences should humans get infected. Transient quantifiable viral DNA levels were seen in blood from a subset of patients, indicative of viral spread but also of systemic control [271]. An autopsy performed on one of the recipients showed low viral DNA levels in spleen, lymph node, liver and bone marrow as well, further evidence of virus replication and spread [271].
Summarizing all of the above studies, what can we humans expect from the gammaretrovirus family in the future? Despite all possible threats from wild and domestic animals, the invention of medical treatments such as vaccination, xenotransplantation and RRV therapy, and despite the apparently effortless in vitro infection of human cells by most retroviruses with gamma-type env genes, a human infection has not been documented, yet. Undoubtedly, the past issues with contaminating ‘rumor’ viruses and with HERVs, discouraged further human testing. In addition, diagnostic assays may be hindered by low DNA or antibody levels, either due to suboptimal replication, fast immune control or the absence of antibodies when infected early in life. In cats, four types of FeLV infection status are recognized, depending on the effectivity of the immune response, namely, progressively infected, regressively infected, focally infected, and abortively infected [278]. For the latter two types, which are more common than progressive infection, detecting viral DNA can be challenging, as virus DNA is only locally present, or was no longer detectable, indicative of virus eradication [278]. Gibbons infected by GALV either develop a persistent antibody response with very low or no measurable viral DNA levels, or, due to the absence of an antibody response, become highly viremic [200,318]. Relying on a single diagnostic test is therefore not enough. To define a human infection a virus should be characterized, the presence of only antibodies will not suffice. However, antibodies could be all that remains of an infection. Until now, human innate, and probably adaptive, immune responses are important factors in combating gammaretrovirus infections. Individuals treated with an MLV RRV suppressed viral replication within weeks, presumably through the generation of neutralizing antibodies [271]. Overall, adequate immune responses may the most important factor in avoiding gammaretrovirus infection, since humans have certainly met the viruses when hunting monkeys, conducting research on animals, caring for pets, livestock and poultry, and, in the last century, through medical procedures involving non-human material, or replicating virus. Alternatively, immune responses may have only inhibited large-scale replication, disabling further adaptation of the virus, thus reducing it to a dead-end infection.
Since gammaretrovirus infection is commonly associated with leukemia or lymphoma, cancer statistics could be used to investigate potential outbreaks. Current data do not show any remarkable increases of leukemia and lymphoma cases, although, for instance, incidence rates for lymphoma subtypes vary widely between countries and over the years [319,320,321]. So, given that all known gammaretroviruses, including betaretroviruses with gamma-type env genes have apparently not infected humans in the past, and are unlikely to do so in the near future, what would a viral threat from that direction look like? Appreciating the molecular divergence of the viruses listed above, with the fast generation of distinct strains and the recombinant viruses often observed, a novel recombinant virus seems the most plausible prediction. Recombination with ERV sequences is not uncommon, and can lead to a virus with novel characteristics. An alternative could be a surprise virus from an unexpected source. For instance, dogs, our long time companions, harbor replication competent gamma-ERVs designated CfERV-Fc1(a), which are highly expressed in dogs with leukemia and lymphoma [322]. Should such a virus end up in a person with some genetic flaw, leading to an inadequate immune response, but being capable of living a long and fruitful life, a new human pathogen may be born. But for now, the conclusion should be that we humans have indeed evaded gammaretrovirus infections for a very long time, but the situation may change in the future, whereby not an ICTV-listed virus, but a novel ERV-recombinant, or a virus from an unexpected source would be the most likely candidate, unless we keep treating humans with RRVs.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The author declares no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
APOBEC3 Apolipoprotein B mRNA editing enzyme, catalytic polypeptide 3
DNA Deoxyribonucleic acid
HUSH Human silencing hub
PBMC Peripheral blood mononuclear cells
PCR Polymerase Chain Reaction
RNA Ribonucleic acid
SAMHD1 SAM domain and HD domain-containing protein 1
SERINC5 Serine incorporator 5
TRIM5α Tripartite motif-containing protein 5α
ZAP Zinc finger antiviral protein

References

  1. Coffin, J.; Blomberg, J.; Fan, H.; Gifford, R.; Hatziioannou, T.; Lindemann, D.; Mayer, J.; Stoye, J.; Tristem, M.; Johnson, W.; et al. ICTV Virus Taxonomy Profile: Retroviridae 2021. The Journal of general virology 2021, 102. [Google Scholar] [CrossRef] [PubMed]
  2. International Committee on Taxonomy of Viruses: Retroviridae. Available online: https://ictv.global/report/chapter/retroviridae/retroviridae (accessed on 24 February 2025).
  3. Poiesz, B.J.; Ruscetti, F.W.; Gazdar, A.F.; Bunn, P.A.; Minna, J.D.; Gallo, R.C. Detection and isolation of type C retrovirus particles from fresh and cultured lymphocytes of a patient with cutaneous T-cell lymphoma. Proceedings of the National Academy of Sciences of the United States of America 1980, 77, 7415–7419. [Google Scholar] [CrossRef]
  4. Lasfargues, E.Y.; Moore, D.H. Proceedings: Search for a viral etiology of human breast cancer. J Invest Dermatol 1974, 63, 125–132. [Google Scholar] [CrossRef]
  5. Lessi, F.; Grandi, N.; Mazzanti, C.M.; Civita, P.; Scatena, C.; Aretini, P.; Bandiera, P.; Fornaciari, A.; Giuffra, V.; Fornaciari, G.; et al. A human MMTV-like betaretrovirus linked to breast cancer has been present in humans at least since the copper age. Aging (Albany NY) 2020, 12, 15978–15994. [Google Scholar] [CrossRef]
  6. Goubran, M.; Wang, W.; Indik, S.; Faschinger, A.; Wasilenko, S.T.; Bintner, J.; Carpenter, E.J.; Zhang, G.; Nuin, P.; Macintyre, G.; et al. Isolation of a Human Betaretrovirus from Patients with Primary Biliary Cholangitis. Viruses 2022, 14. [Google Scholar] [CrossRef]
  7. Bevilacqua, G. The Viral Origin of Human Breast Cancer: From the Mouse Mammary Tumor Virus (MMTV) to the Human Betaretrovirus (HBRV). Viruses 2022, 14. [Google Scholar] [CrossRef] [PubMed]
  8. Lawson, J.S.; Glenn, W.K. Mouse Mammary Tumour Virus (MMTV) in Human Breast Cancer-The Value of Bradford Hill Criteria. Viruses 2022, 14. [Google Scholar] [CrossRef]
  9. Brantley, K.D.; Tamimi, R.M. The association between infectious agents and breast cancer: a review of the epidemiologic evidence. Breast Cancer Res Treat 2024, 207, 235–252. [Google Scholar] [CrossRef] [PubMed]
  10. Martinez Cuesta, L.; Lendez, P.A.; Nieto Farias, M.V.; Dolcini, G.L.; Ceriani, M.C. Can Bovine Leukemia Virus Be Related to Human Breast Cancer? A Review of the Evidence. J Mammary Gland Biol Neoplasia 2018, 23, 101–107. [Google Scholar] [CrossRef]
  11. Hayward, A.; Cornwallis, C.K.; Jern, P. Pan-vertebrate comparative genomics unmasks retrovirus macroevolution. Proceedings of the National Academy of Sciences of the United States of America 2015, 112, 464–469. [Google Scholar] [CrossRef]
  12. van der Kuyl, A.C.; Berkhout, B. Viruses in the reproductive tract: On their way to the germ line? Virus research 2020, 286, 198101. [Google Scholar] [CrossRef]
  13. Wang, J.; Han, G.Z. Genome mining shows that retroviruses are pervasively invading vertebrate genomes. Nature communications 2023, 14, 4968. [Google Scholar] [CrossRef]
  14. Li, Y.; Zhang, G.; Cui, J. Origin and Deep Evolution of Human Endogenous Retroviruses in Pan-Primates. Viruses 2022, 14. [Google Scholar] [CrossRef]
  15. Gifford, R.; Kabat, P.; Martin, J.; Lynch, C.; Tristem, M. Evolution and distribution of class II-related endogenous retroviruses. Journal of virology 2005, 79, 6478–6486. [Google Scholar] [CrossRef]
  16. van der Kuyl, A.C.; Mang, R.; Dekker, J.T.; Goudsmit, J. Complete nucleotide sequence of simian endogenous type D retrovirus with intact genome organization: evidence for ancestry to simian retrovirus and baboon endogenous virus. Journal of virology 1997, 71, 3666–3676. [Google Scholar] [CrossRef]
  17. Henzy, J.E.; Johnson, W.E. Pushing the endogenous envelope. Philosophical transactions of the Royal Society of London. Series B, Biological sciences 2013, 368, 20120506. [Google Scholar] [CrossRef] [PubMed]
  18. Hogan, V.; Johnson, W.E. Unique Structure and Distinctive Properties of the Ancient and Ubiquitous Gamma-Type Envelope Glycoprotein. Viruses 2023, 15. [Google Scholar] [CrossRef] [PubMed]
  19. Cordonnier, A.; Casella, J.F.; Heidmann, T. Isolation of novel human endogenous retrovirus-like elements with foamy virus-related pol sequence. Journal of virology 1995, 69, 5890–5897. [Google Scholar] [CrossRef] [PubMed]
  20. Gifford, R.J.; Katzourakis, A.; Tristem, M.; Pybus, O.G.; Winters, M.; Shafer, R.W. A transitional endogenous lentivirus from the genome of a basal primate and implications for lentivirus evolution. Proceedings of the National Academy of Sciences of the United States of America 2008, 105, 20362–20367. [Google Scholar] [CrossRef]
  21. Gilbert, C.; Maxfield, D.G.; Goodman, S.M.; Feschotte, C. Parallel germline infiltration of a lentivirus in two Malagasy lemurs. PLoS genetics 2009, 5, e1000425. [Google Scholar] [CrossRef]
  22. Katzourakis, A.; Aiewsakun, P.; Jia, H.; Wolfe, N.D.; LeBreton, M.; Yoder, A.D.; Switzer, W.M. Discovery of prosimian and afrotherian foamy viruses and potential cross species transmissions amidst stable and ancient mammalian co-evolution. Retrovirology 2014, 11, 61. [Google Scholar] [CrossRef]
  23. Hron, T.; Elleder, D.; Gifford, R.J. Deltaretroviruses have circulated since at least the Paleogene and infected a broad range of mammalian species. Retrovirology 2019, 16, 33. [Google Scholar] [CrossRef]
  24. Herniou, E.; Martin, J.; Miller, K.; Cook, J.; Wilkinson, M.; Tristem, M. Retroviral diversity and distribution in vertebrates. Journal of virology 1998, 72, 5955–5966. [Google Scholar] [CrossRef]
  25. Martin, J.; Herniou, E.; Cook, J.; O’Neill, R.W.; Tristem, M. Interclass transmission and phyletic host tracking in murine leukemia virus-related retroviruses. Journal of virology 1999, 73, 2442–2449. [Google Scholar] [CrossRef]
  26. Barbacid, M.; Hunter, E.; Aaronson, S.A. Avian reticuloendotheliosis viruses: evolutionary linkage with mammalian type C retroviruses. Journal of virology 1979, 30, 508–514. [Google Scholar] [CrossRef] [PubMed]
  27. Niewiadomska, A.M.; Gifford, R.J. The extraordinary evolutionary history of the reticuloendotheliosis viruses. PLoS biology 2013, 11, e1001642. [Google Scholar] [CrossRef]
  28. van der Kuyl, A.C.; Dekker, J.T.; Goudsmit, J. Distribution of baboon endogenous virus among species of African monkeys suggests multiple ancient cross-species transmissions in shared habitats. Journal of virology 1995, 69, 7877–7887. [Google Scholar] [CrossRef] [PubMed]
  29. Jern, P.; Sperber, G.O.; Blomberg, J. Divergent patterns of recent retroviral integrations in the human and chimpanzee genomes: probable transmissions between other primates and chimpanzees. Journal of virology 2006, 80, 1367–1375. [Google Scholar] [CrossRef] [PubMed]
  30. Zhuo, X.; Feschotte, C. Cross-species transmission and differential fate of an endogenous retrovirus in three mammal lineages. PLoS pathogens 2015, 11, e1005279. [Google Scholar] [CrossRef]
  31. Alfano, N.; Michaux, J.; Morand, S.; Aplin, K.; Tsangaras, K.; Löber, U.; Fabre, P.H.; Fitriana, Y.; Semiadi, G.; Ishida, Y.; et al. Endogenous Gibbon Ape Leukemia Virus Identified in a Rodent (Melomys burtoni subsp.) from Wallacea (Indonesia). Journal of virology 2016, 90, 8169–8180. [Google Scholar] [CrossRef]
  32. Sinha, A.; Johnson, W.E. Retroviruses of the RDR superinfection interference group: ancient origins and broad host distribution of a promiscuous Env gene. Current opinion in virology 2017, 25, 105–112. [Google Scholar] [CrossRef]
  33. Greenwood, A.D.; Ishida, Y.; O’Brien, S.P.; Roca, A.L.; Eiden, M.V. Transmission, evolution, and endogenization: lessons learned from recent retroviral invasions. Microbiology and molecular biology reviews : MMBR 2018, 82, e00044–00017. [Google Scholar] [CrossRef]
  34. van der Kuyl, A.C. Contemporary distribution, estimated age, and prehistoric migrations of Old World monkey retroviruses. Epidemiologia 2021, 2, 46–67. [Google Scholar] [CrossRef]
  35. Williams, Z.H.; Imedio, A.D.; Gaucherand, L.; Lee, D.C.; Mostafa, S.M.; Phelan, J.P.; Coffin, J.M.; Johnson, W.E. Recombinant origin and interspecies transmission of a HERV-K(HML-2)-related primate retrovirus with a novel RNA transport element. eLife 2024, 13. [Google Scholar] [CrossRef] [PubMed]
  36. Piraino, F.; Krumbiegel, E.R.; Wisniewski, H.J. Serologic survey of man for avian leukosis virus infection. Journal of immunology (Baltimore, Md. : 1950) 1967, 98, 702–706. [Google Scholar] [CrossRef] [PubMed]
  37. Aaronson, S.A.; Schlom, J. The search for RNA tumor viruses in human cancer. Prog Clin Cancer 1975, 6, 51–63. [Google Scholar]
  38. Szakacs, J.E.; Szakacs, M.R. Search for C-type particles in human neoplasia. Ann Clin Lab Sci 1975, 5, 14–22. [Google Scholar]
  39. Aoki, T.; Walling, M.J.; Bushar, G.S.; Liu, M.; Hsu, K.C. Natural antibodies in sera from healthy humans to antigens on surfaces of type C RNA viruses and cells from primates. Proceedings of the National Academy of Sciences of the United States of America 1976, 73, 2491–2495. [Google Scholar] [CrossRef]
  40. Ohtsuki, Y.; Seman, G.; Maruyama, K.; Bowen, J.M.; Johnson, D.E.; Dmochowski, L. Ultrastructural studies of human prostatic neoplasia. Cancer 1976, 37, 2295–2305. [Google Scholar] [CrossRef]
  41. Stephenson, J.R.; Aaronson, S.A. Search for antigens and antibodies crossreactive with type C viruses of the woolly monkeys and gibbon ape in animal models and in humans. Proceedings of the National Academy of Sciences of the United States of America 1976, 73, 1725–1729. [Google Scholar] [CrossRef] [PubMed]
  42. Charman, H.P.; Rahman, R.; White, M.H.; Kim, N.; Gilden, R.V. Radioimmunoassay for the major structural protein of Mason-Pfizer monkey virus: Attempts to detect the presence of antigen or antibody in humans. Int J Cancer 1977, 19, 498–504. [Google Scholar] [CrossRef]
  43. Lerche, N.W.; Heneine, W.; Kaplan, J.E.; Spira, T.; Yee, J.L.; Khabbaz, R.F. An expanded search for human infection with simian type D retrovirus. AIDS research and human retroviruses 1995, 11, 527–529. [Google Scholar] [CrossRef]
  44. Morozov, V.A.; Lagaye, S.; Lyakh, L.; ter Meulen, J. Type D retrovirus markers in healthy Africans from Guinea. Research in virology 1996, 147, 341–351. [Google Scholar] [CrossRef]
  45. International Committee on Taxonomy of Viruses: ICTV gammaretroviruses. Available online: https://ictv.global/report/chapter/retroviridae/retroviridae/gammaretrovirus (accessed on 11 January 2025).
  46. van Zaane, D.; Bloemers, H.P. The genome of the mammalian sarcoma viruses. Biochim Biophys Acta 1978, 516, 249–268. [Google Scholar] [CrossRef]
  47. Robbins, K.C.; Hill, R.L.; Aaronson, S.A. Primate origin of the cell-derived sequences of simian sarcoma virus. Journal of virology 1982, 41, 721–725. [Google Scholar] [CrossRef] [PubMed]
  48. Vogt, P.K. Retroviral oncogenes: a historical primer. Nat Rev Cancer 2012, 12, 639–648. [Google Scholar] [CrossRef] [PubMed]
  49. Denner, J. Transspecies Transmission of Gammaretroviruses and the Origin of the Gibbon Ape Leukaemia Virus (GaLV) and the Koala Retrovirus (KoRV). Viruses 2016, 8. [Google Scholar] [CrossRef] [PubMed]
  50. Hayward, J.A.; Tian, S.; Tachedjian, G. GALV-KoRV-related retroviruses in diverse Australian and African rodent species. Virus Evol 2024, 10, veae061. [Google Scholar] [CrossRef]
  51. Alfano, N.; Kolokotronis, S.O.; Tsangaras, K.; Roca, A.L.; Xu, W.; Eiden, M.V.; Greenwood, A.D. Episodic Diversifying Selection Shaped the Genomes of Gibbon Ape Leukemia Virus and Related Gammaretroviruses. Journal of virology 2016, 90, 1757–1772. [Google Scholar] [CrossRef]
  52. Denner, J. Co-Cultivation Assays for Detecting Infectious Human-Tropic Porcine Endogenous Retroviruses (PERVs). International journal of molecular sciences 2025, 26. [Google Scholar] [CrossRef]
  53. Sherwin, S.A.; Todaro, G.J. A new endogenous primate type C virus isolated from the Old World monkey Colobus polykomos. Proceedings of the National Academy of Sciences of the United States of America 1979, 76, 5041–5045. [Google Scholar] [CrossRef]
  54. Todaro, G.J.; Benveniste, R.E.; Sherwin, S.A.; Sherr, C.J. MAC-1, a new genetically transmitted type C virus of primates: “low frequency” activation from stumptail monkey cell cultures. Cell 1978, 13, 775–782. [Google Scholar] [CrossRef] [PubMed]
  55. Cohen, M.; Rein, A.; Stephens, R.M.; O’Connell, C.; Gilden, R.V.; Shure, M.; Nicolson, M.O.; McAllister, R.M.; Davidson, N. Baboon endogenous virus genome: molecular cloning and structural characterization of nondefective viral genomes from DNA of a baboon cell strain. Proceedings of the National Academy of Sciences of the United States of America 1981, 78, 5207–5211. [Google Scholar] [CrossRef] [PubMed]
  56. Rice, N.R.; Bonner, T.I.; Gilden, R.V. Nucleic acid homology between avian and mammalian type C viruses: relatedness of reticuloendotheliosis virus cdna to cloned proviral DNA of the endogenous Colobus virus CPC-1. Virology 1981, 114, 286–290. [Google Scholar] [CrossRef]
  57. PubMed database. Available online: https://pubmed.ncbi.nlm.nih.gov/ (accessed on 19 April 2025).
  58. Google Scholar. Available online: https://scholar.google.nl (accessed on January 22 2025).
  59. Tavaré, S.; Marshall, C.R.; Will, O.; Soligo, C.; Martin, R.D. Using the fossil record to estimate the age of the last common ancestor of extant primates. Nature 2002, 416, 726–729. [Google Scholar] [CrossRef] [PubMed]
  60. Xu, X.; Zhao, H.; Gong, Z.; Han, G.Z. Endogenous retroviruses of non-avian/mammalian vertebrates illuminate diversity and deep history of retroviruses. PLoS pathogens 2018, 14, e1007072. [Google Scholar] [CrossRef]
  61. Sverdlov, E.D. Retroviruses and primate evolution. BioEssays : news and reviews in molecular, cellular and developmental biology 2000, 22, 161–171. [Google Scholar] [CrossRef]
  62. Diehl, W.E.; Patel, N.; Halm, K.; Johnson, W.E. Tracking interspecies transmission and long-term evolution of an ancient retrovirus using the genomes of modern mammals. eLife 2016, 5, e12704. [Google Scholar] [CrossRef]
  63. Kim, H.S.; Kim, D.S.; Huh, J.W.; Ahn, K.; Yi, J.M.; Lee, J.R.; Hirai, H. Molecular characterization of the HERV-W env gene in humans and primates: expression, FISH, phylogeny, and evolution. Mol Cells 2008, 26, 53–60. [Google Scholar] [CrossRef]
  64. Mager, D.L.; Freeman, J.D. HERV-H endogenous retroviruses: presence in the New World branch but amplification in the Old World primate lineage. Virology 1995, 213, 395–404. [Google Scholar] [CrossRef]
  65. Seifarth, W.; Baust, C.; Schön, U.; Reichert, A.; Hehlmann, R.; Leib-Mösch, C. HERV-IP-T47D, a novel type C-related human endogenous retroviral sequence derived from T47D particles. AIDS research and human retroviruses 2000, 16, 471–480. [Google Scholar] [CrossRef] [PubMed]
  66. Blaise, S.; de Parseval, N.; Bénit, L.; Heidmann, T. Genomewide screening for fusogenic human endogenous retrovirus envelopes identifies syncytin 2, a gene conserved on primate evolution. Proceedings of the National Academy of Sciences of the United States of America 2003, 100, 13013–13018. [Google Scholar] [CrossRef]
  67. Aagaard, L.; Villesen, P.; Kjeldbjerg, A.L.; Pedersen, F.S. The approximately 30-million-year-old ERVPb1 envelope gene is evolutionarily conserved among hominoids and Old World monkeys. Genomics 2005, 86, 685–691. [Google Scholar] [CrossRef]
  68. Yi, J.M.; Kim, H.S. Molecular evolution of the HERV-E family in primates. Archives of virology 2006, 151, 1107–1116. [Google Scholar] [CrossRef]
  69. Kjeldbjerg, A.L.; Villesen, P.; Aagaard, L.; Pedersen, F.S. Gene conversion and purifying selection of a placenta-specific ERV-V envelope gene during simian evolution. BMC evolutionary biology 2008, 8, 266. [Google Scholar] [CrossRef]
  70. Blanco-Melo, D.; Gifford, R.J.; Bieniasz, P.D. Co-option of an endogenous retrovirus envelope for host defense in hominid ancestors. eLife 2017, 6. [Google Scholar] [CrossRef]
  71. Kim, H.S.; Yi, J.M.; Hirai, H.; Huh, J.W.; Jeong, M.S.; Jang, S.B.; Kim, C.G.; Saitou, N.; Hyun, B.H.; Lee, W.H. Human Endogenous Retrovirus (HERV)-R family in primates: chromosomal location, gene expression, and evolution. Gene 2006, 370, 34–42. [Google Scholar] [CrossRef]
  72. Lee, J.W.; Kim, H.S. Endogenous retrovirus HERV-I LTR family in primates: sequences, phylogeny, and evolution. Archives of virology 2006, 151, 1651–1658. [Google Scholar] [CrossRef]
  73. Magiorkinis, G.; Blanco-Melo, D.; Belshaw, R. The decline of human endogenous retroviruses: extinction and survival. Retrovirology 2015, 12, 8. [Google Scholar] [CrossRef] [PubMed]
  74. Marchi, E.; Kanapin, A.; Magiorkinis, G.; Belshaw, R. Unfixed endogenous retroviral insertions in the human population. Journal of virology 2014, 88, 9529–9537. [Google Scholar] [CrossRef] [PubMed]
  75. Macfarlane, C.M.; Badge, R.M. Genome-wide amplification of proviral sequences reveals new polymorphic HERV-K(HML-2) proviruses in humans and chimpanzees that are absent from genome assemblies. Retrovirology 2015, 12, 35. [Google Scholar] [CrossRef]
  76. Mi, S.; Lee, X.; Li, X.; Veldman, G.M.; Finnerty, H.; Racie, L.; LaVallie, E.; Tang, X.Y.; Edouard, P.; Howes, S.; et al. Syncytin is a captive retroviral envelope protein involved in human placental morphogenesis. Nature 2000, 403, 785–789. [Google Scholar] [CrossRef]
  77. Esnault, C.; Cornelis, G.; Heidmann, O.; Heidmann, T. Differential evolutionary fate of an ancestral primate endogenous retrovirus envelope gene, the EnvV syncytin, captured for a function in placentation. PLoS genetics 2013, 9, e1003400. [Google Scholar] [CrossRef] [PubMed]
  78. Sugimoto, J.; Sugimoto, M.; Bernstein, H.; Jinno, Y.; Schust, D. A novel human endogenous retroviral protein inhibits cell-cell fusion. Sci Rep 2013, 3, 1462. [Google Scholar] [CrossRef] [PubMed]
  79. Frank, J.A.; Singh, M.; Cullen, H.B.; Kirou, R.A.; Benkaddour-Boumzaouad, M.; Cortes, J.L.; Garcia Pérez, J.; Coyne, C.B.; Feschotte, C. Evolution and antiviral activity of a human protein of retroviral origin. Science (New York, N.Y.) 2022, 378, 422–428. [Google Scholar] [CrossRef] [PubMed]
  80. Mang, R.; Goudsmit, J.; van der Kuyl, A.C. Novel endogenous type C retrovirus in baboons: complete sequence, providing evidence for baboon endogenous virus gag-pol ancestry. Journal of virology 1999, 73, 7021–7026. [Google Scholar] [CrossRef]
  81. Todaro, G.J.; Sherr, C.J.; Benveniste, R.E.; Lieber, M.M.; Melnick, J.L. Type C viruses of baboons: isolation from normal cell cultures. Cell 1974, 2, 55–61. [Google Scholar] [CrossRef]
  82. Benveniste, R.E.; Lieber, M.M.; Livingston, D.M.; Sherr, C.J.; Todaro, G.J.; Kalter, S.S. Infectious C-type virus isolated from a baboon placenta. Nature 1974, 248, 17–20. [Google Scholar] [CrossRef]
  83. Bonner, T.I.; Birkenmeier, E.H.; Gonda, M.A.; Mark, G.E.; Searfoss, G.H.; Todaro, G.J. Molecular cloning of a family of retroviral sequences found in chimpanzee but not human DNA. Journal of virology 1982, 43, 914–924. [Google Scholar] [CrossRef]
  84. Ma, H.; Ma, Y.; Ma, W.; Williams, D.K.; Galvin, T.A.; Khan, A.S. Chemical induction of endogenous retrovirus particles from the vero cell line of African green monkeys. Journal of virology 2011, 85, 6579–6588. [Google Scholar] [CrossRef]
  85. Sommerfelt, M.A.; Weiss, R.A. Receptor interference groups of 20 retroviruses plating on human cells. Virology 1990, 176, 58–69. [Google Scholar] [CrossRef]
  86. Girard-Gagnepain, A.; Amirache, F.; Costa, C.; Lévy, C.; Frecha, C.; Fusil, F.; Nègre, D.; Lavillette, D.; Cosset, F.L.; Verhoeyen, E. Baboon envelope pseudotyped LVs outperform VSV-G-LVs for gene transfer into early-cytokine-stimulated and resting HSCs. Blood 2014, 124, 1221–1231. [Google Scholar] [CrossRef] [PubMed]
  87. Bernadin, O.; Amirache, F.; Girard-Gagnepain, A.; Moirangthem, R.D.; Lévy, C.; Ma, K.; Costa, C.; Nègre, D.; Reimann, C.; Fenard, D.; et al. Baboon envelope LVs efficiently transduced human adult, fetal, and progenitor T cells and corrected SCID-X1 T-cell deficiency. Blood Adv 2019, 3, 461–475. [Google Scholar] [CrossRef]
  88. Ikeda, M.; Satomura, K.; Sekizuka, T.; Hanada, K.; Endo, T.; Osada, N. Comprehensive phylogenomic analysis reveals a novel cluster of simian endogenous retroviral sequences in Colobinae monkeys. American journal of primatology 2018, 80, e22882. [Google Scholar] [CrossRef] [PubMed]
  89. Power, M.D.; Marx, P.A.; Bryant, M.L.; Gardner, M.B.; Barr, P.J.; Luciw, P.A. Nucleotide sequence of SRV-1, a type D simian acquired immune deficiency syndrome retrovirus. Science (New York, N.Y.) 1986, 231, 1567–1572. [Google Scholar] [CrossRef]
  90. Heidecker, G.; Lerche, N.W.; Lowenstine, L.J.; Lackner, A.A.; Osborn, K.G.; Gardner, M.B.; Marx, P.A. Induction of simian acquired immune deficiency syndrome (SAIDS) with a molecular clone of a type D SAIDS retrovirus. Journal of virology 1987, 61, 3066–3071. [Google Scholar] [CrossRef] [PubMed]
  91. Grant, R.; Keele, B.; Kuller, L.; Watanabe, R.; Perret, A.; Smedley, J. Identification of novel simian endogenous retroviruses that are indistinguishable from simian retrovirus (SRV) on current SRV diagnostic assays. Journal of medical primatology 2017, 46, 158–161. [Google Scholar] [CrossRef]
  92. Sakuma, C.; Sekizuka, T.; Kuroda, M.; Kasai, F.; Saito, K.; Ikeda, M.; Yamaji, T.; Osada, N.; Hanada, K. Novel endogenous simian retroviral integrations in Vero cells: implications for quality control of a human vaccine cell substrate. Sci Rep 2018, 8, 644. [Google Scholar] [CrossRef]
  93. Kiesslich, S.; Kamen, A.A. Vero cell upstream bioprocess development for the production of viral vectors and vaccines. Biotechnol Adv 2020, 44, 107608. [Google Scholar] [CrossRef]
  94. Yohn, C.T.; Jiang, Z.; McGrath, S.D.; Hayden, K.E.; Khaitovich, P.; Johnson, M.E.; Eichler, M.Y.; McPherson, J.D.; Zhao, S.; Pääbo, S.; et al. Lineage-specific expansions of retroviral insertions within the genomes of African great apes but not humans and orangutans. PLoS biology 2005, 3, e110. [Google Scholar] [CrossRef]
  95. Polavarapu, N.; Bowen, N.J.; McDonald, J.F. Identification, characterization and comparative genomics of chimpanzee endogenous retroviruses. Genome biology 2006, 7, R51. [Google Scholar] [CrossRef]
  96. Kim, Y.J.; Han, K. Endogenous retrovirus-mediated genomic variations in chimpanzees. Mob Genet Elements 2014, 4, 1–4. [Google Scholar] [CrossRef]
  97. AbuEed, L.; Miyake, A.; Wanjala, N.; Pramono, D.; Abdillah, D.; Imamura, M.; Shimojima, M.; Denner, J.; Kawasaki, J.; Nishigaki, K. Riboflavin transporter: evidence of a role as entry receptor for chimpanzee endogenous retrovirus. Virus Evol 2025, 11, veaf031. [Google Scholar] [CrossRef] [PubMed]
  98. Kaiser, S.M.; Malik, H.S.; Emerman, M. Restriction of an extinct retrovirus by the human TRIM5alpha antiviral protein. Science (New York, N.Y.) 2007, 316, 1756–1758. [Google Scholar] [CrossRef] [PubMed]
  99. Rous, P. A sarcoma of the fowl transmissable by an agent separable from the tumor cells. The Journal of experimental medicine 1911, 13, 397–411. [Google Scholar] [CrossRef]
  100. Bittner, J.J. The preservation by freezing and drying in vacuo of the milk-influence for the development of breast cancer in mice. Science (New York, N.Y.) 1941, 93, 527–528. [Google Scholar] [CrossRef] [PubMed]
  101. Duran-Reynals, F.; Shrigley, E.W. A study of five transplantable chicken sarcomas induced by viruses. Cancer Res 1946, 6, 535–544. [Google Scholar]
  102. Kunii, A.; Furth, J. Mammary carcinoma in mice bearing a transplantable mammotropic tumor, carrying the Bittner virus. Proc Soc Exp Biol Med 1963, 114, 709–714. [Google Scholar] [CrossRef]
  103. McAllister, R.M.; Nicolson, M.; Gardner, M.B.; Rongey, R.W.; Rasheed, S.; Sarma, P.S.; Huebner, R.J.; Hatanaka, M.; Oroszlan, S.; Gilden, R.V.; et al. C-type virus released from cultured human rhabdomyosarcoma cells. Nat New Biol 1972, 235, 3–6. [Google Scholar] [CrossRef]
  104. Fischinger, P.J.; Peebles, P.T.; Nomura, S.; Haapala, D.K. Isolation of RD-114-like oncornavirus from a cat cell line. Journal of virology 1973, 11, 978–985. [Google Scholar] [CrossRef]
  105. Okabe, H.; Gilden, R.V.; Hatanaka, M. RD 114 virus-specific sequences in feline cellular RNA: detection and characterization. Journal of virology 1973, 12, 984–994. [Google Scholar] [CrossRef]
  106. Ruprecht, R.M.; Goodman, N.C.; Spiegelman, S. Determination of natural host taxonomy of RNA tumor viruses by molecular hybridization: application to RD-114, a candidate human virus. Proceedings of the National Academy of Sciences of the United States of America 1973, 70, 1437–1441. [Google Scholar] [CrossRef] [PubMed]
  107. Gallagher, R.E.; Gallo, R.C. Type C RNA tumor virus isolated from cultured human acute myelogenous leukemia cells. Science (New York, N.Y.) 1975, 187, 350–353. [Google Scholar] [CrossRef] [PubMed]
  108. Teich, N.M.; Weiss, R.A.; Salahuddin, S.Z.; Gallagher, R.E.; Gillespie, D.H.; Gallo, R.C. Infective transmission and characterisation of a C-type virus released by cultured human myeloid leukaemia cells. Nature 1975, 256, 551–555. [Google Scholar] [CrossRef] [PubMed]
  109. Reitz, M.S.; Miller, N.R.; Wong-Staal, F.; Gallagher, R.E.; Gallo, R.C.; Gillespie, D.H. Primate type-C virus nucleic acid sequences (woolly monkey and baboon types) in tissues from a patient with acute myelogenous leukemia and in viruses isolated from cultured cells of the same patient. Proceedings of the National Academy of Sciences of the United States of America 1976, 73, 2113–2117. [Google Scholar] [CrossRef]
  110. Sahagan, B.G.; Haseltine, W.A. Relationship of retroviruses isolated from human leukemia tissues to the woolly monkey-gibbon ape leukemia viruses. Journal of virology 1980, 34, 390–401. [Google Scholar] [CrossRef]
  111. Okabe, H.; Gilden, R.V.; Hatanaka, M.; Stephenson, J.R.; Gallagher, R.E.; Aaronson, S.A.; Gallo, R.C.; Tronick, S.R. Immunological and biochemical characterisation of type C viruses isolated from cultured human AML cells. Nature 1976, 260, 264–266. [Google Scholar] [CrossRef]
  112. Mishra, L.; Hong, D.; Baluda, M.A. Homology between HL-23V and primate viruses and search for proviral DNA sequences of simian sarcoma associated virus and baboon endogenous virus in DNA from human leukemic cells. Leuk Res 1979, 3, 285–296. [Google Scholar] [CrossRef]
  113. Bergholz, C.M.; Wolfe, L.G.; Schulz, G.A.; Deinhardt, F.; Miller, N.R.; Reitz, M.S. Isolation of a virus closely related to gibbon ape leukaemia virus from cells infected with virus (HL-23V) released by human leukaemic cells. The Journal of general virology 1980, 48, 111–121. [Google Scholar] [CrossRef]
  114. Wong-Staal, F.; Gillespie, D.; Gallo, R.C. Proviral sequences of baboon endogenous type C RNA virus in DNA of human leukaemic tissues. Nature 1976, 262, 190–195. [Google Scholar] [CrossRef]
  115. Aulakh, G.S.; Gallo, R.C. Rauscher-leukemia-virus-related sequences in human DNA: presence in some tissues of some patients with hemotopoietic neoplasias and absence in DNA from other tissues. Proceedings of the National Academy of Sciences of the United States of America 1977, 74, 353–357. [Google Scholar] [CrossRef]
  116. Perron, H.; Lalande, B.; Gratacap, B.; Laurent, A.; Genoulaz, O.; Geny, C.; Mallaret, M.; Schuller, E.; Stoebner, P.; Seigneurin, J.M. Isolation of retrovirus from patients with multiple sclerosis. Lancet (London, England) 1991, 337, 862–863. [Google Scholar] [CrossRef]
  117. Perron, H.; Gratacap, B.; Lalande, B.; Genoulaz, O.; Laurent, A.; Geny, C.; Mallaret, M.; Innocenti, P.; Schuller, E.; Stoebner, P.; et al. In vitro transmission and antigenicity of a retrovirus isolated from a multiple sclerosis patient. Research in virology 1992, 143, 337–350. [Google Scholar] [CrossRef] [PubMed]
  118. Perron, H.; Garson, J.A.; Bedin, F.; Beseme, F.; Paranhos-Baccala, G.; Komurian-Pradel, F.; Mallet, F.; Tuke, P.W.; Voisset, C.; Blond, J.L.; et al. Molecular identification of a novel retrovirus repeatedly isolated from patients with multiple sclerosis. The Collaborative Research Group on Multiple Sclerosis. Proceedings of the National Academy of Sciences of the United States of America 1997, 94, 7583–7588. [Google Scholar] [CrossRef] [PubMed]
  119. Garson, J.A.; Tuke, P.W.; Giraud, P.; Paranhos-Baccala, G.; Perron, H. Detection of virion-associated MSRV-RNA in serum of patients with multiple sclerosis. Lancet (London, England) 1998, 351, 33. [Google Scholar] [CrossRef]
  120. Komurian-Pradel, F.; Paranhos-Baccala, G.; Bedin, F.; Ounanian-Paraz, A.; Sodoyer, M.; Ott, C.; Rajoharison, A.; Garcia, E.; Mallet, F.; Mandrand, B.; et al. Molecular cloning and characterization of MSRV-related sequences associated with retrovirus-like particles. Virology 1999, 260, 1–9. [Google Scholar] [CrossRef] [PubMed]
  121. Morandi, E.; Tanasescu, R.; Tarlinton, R.E.; Constantinescu, C.S.; Zhang, W.; Tench, C.; Gran, B. The association between human endogenous retroviruses and multiple sclerosis: A systematic review and meta-analysis. PloS one 2017, 12, e0172415. [Google Scholar] [CrossRef]
  122. Coffin, J.M.; Kearney, M.F. False Alarm: XMRV, Cancer, and Chronic Fatigue Syndrome. Annual review of virology 2024, 11, 261–281. [Google Scholar] [CrossRef]
  123. Paprotka, T.; Delviks-Frankenberry, K.A.; Cingöz, O.; Martinez, A.; Kung, H.J.; Tepper, C.G.; Hu, W.S.; Fivash, M.J., Jr.; Coffin, J.M.; Pathak, V.K. Recombinant origin of the retrovirus XMRV. Science (New York, N.Y.) 2011, 333, 97–101. [Google Scholar] [CrossRef]
  124. Alberts, B. Retraction. Science (New York, N.Y.) 2011, 334, 1636. [Google Scholar] [CrossRef]
  125. Urisman, A.; Molinaro, R.J.; Fischer, N.; Plummer, S.J.; Casey, G.; Klein, E.A.; Malathi, K.; Magi-Galluzzi, C.; Tubbs, R.R.; Ganem, D.; et al. Retraction. Identification of a novel gammaretrovirus in prostate tumors of patients homozygous for R462Q RNASEL variant. PLoS pathogens 2012, 8. [Google Scholar] [CrossRef]
  126. Deichmann, M.; Huder, J.B.; Kleist, C.; Näher, H.; Schüpbach, J.; Böni, J. Detection of reverse transcriptase activity in human melanoma cell lines and identification of a murine leukemia virus contaminant. Arch Dermatol Res 2005, 296, 345–352. [Google Scholar] [CrossRef] [PubMed]
  127. Burtonboy, G.; Delferriere, N.; Mousset, B.; Heusterspreute, M. Isolation of a C-type retrovirus from an HIV infected cell line. Archives of virology 1993, 130, 289–300. [Google Scholar] [CrossRef]
  128. Parent, I.; Qin, Y.; Vandenbroucke, A.T.; Walon, C.; Delferrière, N.; Godfroid, E.; Burtonboy, G. Characterization of a C-type retrovirus isolated from an HIV infected cell line: complete nucleotide sequence. Archives of virology 1998, 143, 1077–1092. [Google Scholar] [CrossRef]
  129. Takeuchi, Y.; McClure, M.O.; Pizzato, M. Identification of gammaretroviruses constitutively released from cell lines used for human immunodeficiency virus research. Journal of virology 2008, 82, 12585–12588. [Google Scholar] [CrossRef]
  130. Stang, A.; Petrasch-Parwez, E.; Brandt, S.; Dermietzel, R.; Meyer, H.E.; Stühler, K.; Liffers, S.T.; Uberla, K.; Grunwald, T. Unintended spread of a biosafety level 2 recombinant retrovirus. Retrovirology 2009, 6, 86. [Google Scholar] [CrossRef] [PubMed]
  131. Oda, T.; Ikeda, S.; Watanabe, S.; Hatsushika, M.; Akiyama, K.; Mitsunobu, F. Molecular cloning, complete nucleotide sequence, and gene structure of the provirus genome of a retrovirus produced in a human lymphoblastoid cell line. Virology 1988, 167, 468–476. [Google Scholar] [CrossRef]
  132. Uphoff, C.C.; Denkmann, S.A.; Steube, K.G.; Drexler, H.G. Detection of EBV, HBV, HCV, HIV-1, HTLV-I and -II, and SMRV in human and other primate cell lines. J Biomed Biotechnol 2010, 2010, 904767. [Google Scholar] [CrossRef]
  133. Uphoff, C.C.; Lange, S.; Denkmann, S.A.; Garritsen, H.S.; Drexler, H.G. Prevalence and characterization of murine leukemia virus contamination in human cell lines. PloS one 2015, 10, e0125622. [Google Scholar] [CrossRef]
  134. Griffiths, D.J.; Voisset, C.; Venables, P.J.; Weiss, R.A. Novel endogenous retrovirus in rabbits previously reported as human retrovirus 5. Journal of virology 2002, 76, 7094–7102. [Google Scholar] [CrossRef] [PubMed]
  135. Forsman, A.; Uzameckis, D.; Rönnblom, L.; Baecklund, E.; Aleskog, A.; Bindra, A.; Pipkorn, R.; Lejniece, S.; Kozireva, S.; Murovska, M.; et al. Single-tube nested quantitative PCR: a rational and sensitive technique for detection of retroviral DNA. Application to RERV-H/HRV-5 and confirmation of its rabbit origin. Journal of virological methods 2003, 111, 1–11. [Google Scholar] [CrossRef]
  136. Garry, R.F.; Fermin, C.D.; Hart, D.J.; Alexander, S.S.; Donehower, L.A.; Luo-Zhang, H. Detection of a human intracisternal A-type retroviral particle antigenically related to HIV. Science (New York, N.Y.) 1990, 250, 1127–1129. [Google Scholar] [CrossRef]
  137. Yamano, S.; Renard, J.N.; Mizuno, F.; Narita, Y.; Uchida, Y.; Higashiyama, H.; Sakurai, H.; Saito, I. Retrovirus in salivary glands from patients with Sjögren’s syndrome. J Clin Pathol 1997, 50, 223–230. [Google Scholar] [CrossRef] [PubMed]
  138. Sipsas, N.V.; Gamaletsou, M.N.; Moutsopoulos, H.M. Is Sjögren’s syndrome a retroviral disease? Arthritis Res Ther 2011, 13, 212. [Google Scholar] [CrossRef]
  139. Borghol, A.H.; Bitar, E.R.; Hanna, A.; Naim, G.; Rahal, E.A. The role of Epstein-Barr virus in autoimmune and autoinflammatory diseases. Crit Rev Microbiol 2025, 51, 296–316. [Google Scholar] [CrossRef]
  140. Voisset, C.; Weiss, R.A.; Griffiths, D.J. Human RNA “rumor” viruses: the search for novel human retroviruses in chronic disease. Microbiology and molecular biology reviews : MMBR 2008, 72, 157–196. [Google Scholar] [CrossRef]
  141. Betsem, E.; Rua, R.; Tortevoye, P.; Froment, A.; Gessain, A. Frequent and recent human acquisition of simian foamy viruses through apes’ bites in central Africa. PLoS pathogens 2011, 7, e1002306. [Google Scholar] [CrossRef]
  142. Mouinga-Ondémé, A.; Kazanji, M. Simian foamy virus in non-human primates and cross-species transmission to humans in Gabon: an emerging zoonotic disease in central Africa? Viruses 2013, 5, 1536–1552. [Google Scholar] [CrossRef]
  143. Van Dooren, S.; Salemi, M.; Vandamme, A.M. Dating the origin of the African human T-cell lymphotropic virus type-i (HTLV-I) subtypes. Molecular biology and evolution 2001, 18, 661–671. [Google Scholar] [CrossRef] [PubMed]
  144. Calattini, S.; Chevalier, S.A.; Duprez, R.; Bassot, S.; Froment, A.; Mahieux, R.; Gessain, A. Discovery of a new human T-cell lymphotropic virus (HTLV-3) in Central Africa. Retrovirology 2005, 2, 30. [Google Scholar] [CrossRef] [PubMed]
  145. Wolfe, N.D.; Heneine, W.; Carr, J.K.; Garcia, A.D.; Shanmugam, V.; Tamoufe, U.; Torimiro, J.N.; Prosser, A.T.; Lebreton, M.; Mpoudi-Ngole, E.; et al. Emergence of unique primate T-lymphotropic viruses among central African bushmeat hunters. Proceedings of the National Academy of Sciences of the United States of America 2005, 102, 7994–7999. [Google Scholar] [CrossRef] [PubMed]
  146. Zheng, H.; Wolfe, N.D.; Sintasath, D.M.; Tamoufe, U.; Lebreton, M.; Djoko, C.F.; Diffo Jle, D.; Pike, B.L.; Heneine, W.; Switzer, W.M. Emergence of a novel and highly divergent HTLV-3 in a primate hunter in Cameroon. Virology 2010, 401, 137–145. [Google Scholar] [CrossRef] [PubMed]
  147. Mahieux, R.; Gessain, A. HTLV-3/STLV-3 and HTLV-4 viruses: discovery, epidemiology, serology and molecular aspects. Viruses 2011, 3, 1074–1090. [Google Scholar] [CrossRef]
  148. Ferreira, O.C., Jr.; Planelles, V.; Rosenblatt, J.D. Human T-cell leukemia viruses: epidemiology, biology, and pathogenesis. Blood Rev 1997, 11, 91–104. [Google Scholar] [CrossRef] [PubMed]
  149. Zhu, T.; Korber, B.T.; Nahmias, A.J.; Hooper, E.; Sharp, P.M.; Ho, D.D. An African HIV-1 sequence from 1959 and implications for the origin of the epidemic. Nature 1998, 391, 594–597. [Google Scholar] [CrossRef]
  150. Gao, F.; Bailes, E.; Robertson, D.L.; Chen, Y.; Rodenburg, C.M.; Michael, S.F.; Cummins, L.B.; Arthur, L.O.; Peeters, M.; Shaw, G.M.; et al. Origin of HIV-1 in the chimpanzee Pan troglodytes troglodytes. Nature 1999, 397, 436–441. [Google Scholar] [CrossRef]
  151. Worobey, M.; Gemmel, M.; Teuwen, D.E.; Haselkorn, T.; Kunstman, K.; Bunce, M.; Muyembe, J.J.; Kabongo, J.M.; Kalengayi, R.M.; Van Marck, E.; et al. Direct evidence of extensive diversity of HIV-1 in Kinshasa by 1960. Nature 2008, 455, 661–664. [Google Scholar] [CrossRef]
  152. Faria, N.R.; Rambaut, A.; Suchard, M.A.; Baele, G.; Bedford, T.; Ward, M.J.; Tatem, A.J.; Sousa, J.D.; Arinaminpathy, N.; Pépin, J.; et al. HIV epidemiology. The early spread and epidemic ignition of HIV-1 in human populations. Science (New York, N.Y.) 2014, 346, 56–61. [Google Scholar] [CrossRef]
  153. Sharp, P.M.; Hahn, B.H. Origins of HIV and the AIDS pandemic. Cold Spring Harb Perspect Med 2011, 1, a006841. [Google Scholar] [CrossRef]
  154. Peeters, M.; Koumare, B.; Mulanga, C.; Brengues, C.; Mounirou, B.; Bougoudogo, F.; Ravel, S.; Bibollet-Ruche, F.; Delaporte, E. Genetic subtypes of HIV type 1 and HIV type 2 strains in commercial sex workers from Bamako, Mali. AIDS research and human retroviruses 1998, 14, 51–58. [Google Scholar] [CrossRef]
  155. van der Loeff, M.F.; Awasana, A.A.; Sarge-Njie, R.; van der Sande, M.; Jaye, A.; Sabally, S.; Corrah, T.; McConkey, S.J.; Whittle, H.C. Sixteen years of HIV surveillance in a West African research clinic reveals divergent epidemic trends of HIV-1 and HIV-2. Int J Epidemiol 2006, 35, 1322–1328. [Google Scholar] [CrossRef] [PubMed]
  156. da Silva, Z.J.; Oliveira, I.; Andersen, A.; Dias, F.; Rodrigues, A.; Holmgren, B.; Andersson, S.; Aaby, P. Changes in prevalence and incidence of HIV-1, HIV-2 and dual infections in urban areas of Bissau, Guinea-Bissau: is HIV-2 disappearing? AIDS (London, England) 2008, 22, 1195–1202. [Google Scholar] [CrossRef] [PubMed]
  157. Heitzinger, K.; Sow, P.S.; Dia Badiane, N.M.; Gottlieb, G.S.; N’Doye, I.; Toure, M.; Kiviat, N.B.; Hawes, S.E. Trends of HIV-1, HIV-2 and dual infection in women attending outpatient clinics in Senegal, 1990-2009. Int J STD AIDS 2012, 23, 710–716. [Google Scholar] [CrossRef]
  158. Yuan, Z.; Kang, G.; Ma, F.; Lu, W.; Fan, W.; Fennessey, C.M.; Keele, B.F.; Li, Q. Recapitulating Cross-Species Transmission of Simian Immunodeficiency Virus SIVcpz to Humans by Using Humanized BLT Mice. Journal of virology 2016, 90, 7728–7739. [Google Scholar] [CrossRef]
  159. Sato, K.; Misawa, N.; Takeuchi, J.S.; Kobayashi, T.; Izumi, T.; Aso, H.; Nagaoka, S.; Yamamoto, K.; Kimura, I.; Konno, Y.; et al. Experimental Adaptive Evolution of Simian Immunodeficiency Virus SIVcpz to Pandemic Human Immunodeficiency Virus Type 1 by Using a Humanized Mouse Model. Journal of virology 2018, 92. [Google Scholar] [CrossRef]
  160. Sauter, D.; Kirchhoff, F. Key Viral Adaptations Preceding the AIDS Pandemic. Cell host & microbe 2019, 25, 27–38. [Google Scholar] [CrossRef]
  161. Khan, A.S.; Bodem, J.; Buseyne, F.; Gessain, A.; Johnson, W.; Kuhn, J.H.; Kuzmak, J.; Lindemann, D.; Linial, M.L.; Löchelt, M.; et al. Spumaretroviruses: Updated taxonomy and nomenclature. Virology 2018, 516, 158–164. [Google Scholar] [CrossRef]
  162. Kehl, T.; Tan, J.; Materniak, M. Non-simian foamy viruses: molecular virology, tropism and prevalence and zoonotic/interspecies transmission. Viruses 2013, 5, 2169–2209. [Google Scholar] [CrossRef]
  163. Butera, S.T.; Brown, J.; Callahan, M.E.; Owen, S.M.; Matthews, A.L.; Weigner, D.D.; Chapman, L.E.; Sandstrom, P.A. Survey of veterinary conference attendees for evidence of zoonotic infection by feline retroviruses. J Am Vet Med Assoc 2000, 217, 1475–1479. [Google Scholar] [CrossRef]
  164. Pinto-Santini, D.M.; Stenbak, C.R.; Linial, M.L. Foamy virus zoonotic infections. Retrovirology 2017, 14, 55. [Google Scholar] [CrossRef]
  165. Heneine, W.; Switzer, W.M.; Sandstrom, P.; Brown, J.; Vedapuri, S.; Schable, C.A.; Khan, A.S.; Lerche, N.W.; Schweizer, M.; Neumann-Haefelin, D.; et al. Identification of a human population infected with simian foamy viruses. Nat Med 1998, 4, 403–407. [Google Scholar] [CrossRef]
  166. Stenbak, C.R.; Craig, K.L.; Ivanov, S.B.; Wang, X.; Soliven, K.C.; Jackson, D.L.; Gutierrez, G.A.; Engel, G.; Jones-Engel, L.; Linial, M.L. New World simian foamy virus infections in vivo and in vitro. Journal of virology 2014, 88, 982–991. [Google Scholar] [CrossRef]
  167. Santos, A.F.; Cavalcante, L.T.F.; Muniz, C.P.; Switzer, W.M.; Soares, M.A. Simian foamy viruses in Central and South America: A new world of discovery. Viruses 2019, 11. [Google Scholar] [CrossRef]
  168. Di Virgilio, G.; Lavenda, N.; Siegel, D. Viral particles in human breast cancer. Oncologia 1965, 19, 341–348. [Google Scholar] [CrossRef]
  169. Dmochowski, L.; Seman, G.; Gallager, H.S. Viruses as possible etiologic factors in human breast cancer. Cancer 1969, 24, 1241–1249. [Google Scholar] [CrossRef]
  170. Schlom, J.; Spiegelman, S.; Moore, D. RNA-dependent DNA polymerase activity in virus-like particles isolated from human milk. Nature 1971, 231, 97–100. [Google Scholar] [CrossRef] [PubMed]
  171. Dion, A.S.; Sarkar, N.H.; Moore, D.H. An attempt to correlate RNA-templated DNA polymerase activity with virus-like particles in human milk: murine mammary tumor virus (MuMTV) as a model system. Johns Hopkins Med J Suppl 1973, 2, 267–281. [Google Scholar] [PubMed]
  172. Das, M.R.; Mink, M.M. Sequence homology of nucleic acids from human breast cancer cells and complementary DNA’s from murine mammary tumor virus and Mason-Pfizer monkey virus. Cancer Res 1979, 39, 5106–5113. [Google Scholar]
  173. Xu, L.; Shen, Z.; Guo, L.; Fodera, B.; Keogh, A.; Joplin, R.; O’Donnell, B.; Aitken, J.; Carman, W.; Neuberger, J.; et al. Does a betaretrovirus infection trigger primary biliary cirrhosis? Proceedings of the National Academy of Sciences of the United States of America 2003, 100, 8454–8459. [Google Scholar] [CrossRef]
  174. Amarante, M.K.; de Sousa Pereira, N.; Vitiello, G.A.F.; Watanabe, M.A.E. Involvement of a mouse mammary tumor virus (MMTV) homologue in human breast cancer: Evidence for, against and possible causes of controversies. Microb Pathog 2019, 130, 283–294. [Google Scholar] [CrossRef] [PubMed]
  175. Graves, D.C.; Ferrer, J.F. In vitro transmission and propagation of the bovine leukemia virus in monolayer cell cultures. Cancer Res 1976, 36, 4152–4159. [Google Scholar]
  176. Saeedi-Moghaddam, F.; Mohammaditabar, M.; Mozhgani, S.H. Bovine leukemia virus (BLV) and risk of breast cancer; a systematic review and meta-analysis. Retrovirology 2024, 21, 20. [Google Scholar] [CrossRef]
  177. Blanco, R.; Quezada-Romegialli, C.; Muñoz, J.P. Bovine Leukemia Virus and Human Breast Cancer: A Review of Clinical and Molecular Evidence. Viruses 2025, 17. [Google Scholar] [CrossRef] [PubMed]
  178. Purchase, H.G.; Ludford, C.; Nazerian, K.; Cox, H.W. A new group of oncogenic viruses: reticuloendotheliosis, chick syncytial, duck infectious anemia, and spleen necrosis viruses. J Natl Cancer Inst 1973, 51, 489–499. [Google Scholar] [PubMed]
  179. Purchase, H.G.; Witter, R.L. The reticuloendotheliosis viruses. Curr Top Microbiol Immunol 1975, 71, 103–124. [Google Scholar] [CrossRef] [PubMed]
  180. Gautier, R.; Jiang, A.; Rousseau, V.; Dornburg, R.; Jaffredo, T. Avian reticuloendotheliosis virus strain A and spleen necrosis virus do not infect human cells. Journal of virology 2000, 74, 518–522. [Google Scholar] [CrossRef]
  181. Schat, K.A.; Erb, H.N. Lack of evidence that avian oncogenic viruses are infectious for humans: a review. Avian diseases 2014, 58, 345–358. [Google Scholar] [CrossRef]
  182. Koo, H.M.; Brown, A.M.; Ron, Y.; Dougherty, J.P. Spleen necrosis virus, an avian retrovirus, can infect primate cells. Journal of virology 1991, 65, 4769–4776. [Google Scholar] [CrossRef]
  183. Awad, A.M.; Abd El-Hamid, H.S.; Abou Rawash, A.A.; Ibrahim, H.H. Detection of reticuloendotheliosis virus as a contaminant of fowl pox vaccines. Poult Sci 2010, 89, 2389–2395. [Google Scholar] [CrossRef]
  184. Fadly, A.; Garcia, M.C. Detection of reticuloendotheliosis virus in live virus vaccines of poultry. Dev Biol (Basel) 2006, 126, 301–305; discussion 327. [Google Scholar]
  185. Chacón, R.D.; Astolfi-Ferreira, C.S.; Valdeiglesias Ichillumpa, S.; Lage Hagemann, H.; Furlan Rocha, M.; Fernandes Magalhães, L.; Freitas Raso, T.; Ferreira, A.J.P. First Complete Genome of Reticuloendotheliosis Virus in a Mallard Duck from Brazil: Phylogenetic Insights and Evolutionary Analysis. Pathogens 2025, 14. [Google Scholar] [CrossRef]
  186. Baxby, D.; Paoletti, E. Potential use of non-replicating vectors as recombinant vaccines. Vaccine 1992, 10, 8–9. [Google Scholar] [CrossRef]
  187. Kozak, C.A. Origins of the endogenous and infectious laboratory mouse gammaretroviruses. Viruses 2014, 7, 1–26. [Google Scholar] [CrossRef]
  188. Specke, V.; Rubant, S.; Denner, J. Productive infection of human primary cells and cell lines with porcine endogenous retroviruses. Virology 2001, 285, 177–180. [Google Scholar] [CrossRef] [PubMed]
  189. Moalic, Y.; Blanchard, Y.; Félix, H.; Jestin, A. Porcine endogenous retrovirus integration sites in the human genome: features in common with those of murine leukemia virus. Journal of virology 2006, 80, 10980–10988. [Google Scholar] [CrossRef] [PubMed]
  190. Lee, D.; Kim, N.Y.; Bae, G.E.; Lee, H.J.; Kwon, M.; Kim, S.S.; Lee, H.T.; Yang, J.M.; Kim, Y.B. Transmissible infection of human 293T cells with porcine endogenous retroviruses subgroup a from NIH-miniature pig. Transplant Proc 2008, 40, 3742–3745. [Google Scholar] [CrossRef]
  191. Denner, J. Monitoring for PERV Following Xenotransplantation. Transpl Int 2024, 37, 13491. [Google Scholar] [CrossRef]
  192. Lamers, C.H.; Willemsen, R.A.; van Elzakker, P.M.; Gratama, J.W.; Debets, R. Gibbon ape leukemia virus poorly replicates in primary human T lymphocytes: implications for safety testing of primary human T lymphocytes transduced with GALV-pseudotyped vectors. J Immunother 2009, 32, 272–279. [Google Scholar] [CrossRef]
  193. Fiebig, U.; Hartmann, M.G.; Bannert, N.; Kurth, R.; Denner, J. Transspecies transmission of the endogenous koala retrovirus. Journal of virology 2006, 80, 5651–5654. [Google Scholar] [CrossRef]
  194. Shojima, T.; Hoshino, S.; Abe, M.; Yasuda, J.; Shogen, H.; Kobayashi, T.; Miyazawa, T. Construction and characterization of an infectious molecular clone of Koala retrovirus. Journal of virology 2013, 87, 5081–5088. [Google Scholar] [CrossRef] [PubMed]
  195. Renner, A.; Stahringer, A.; Ruppel, K.E.; Fricke, S.; Koehl, U.; Schmiedel, D. Development of KoRV-pseudotyped lentiviral vectors for efficient gene transfer into freshly isolated immune cells. Gene therapy 2024, 31, 378–390. [Google Scholar] [CrossRef]
  196. Hayward, J.A.; Tachedjian, M.; Kohl, C.; Johnson, A.; Dearnley, M.; Jesaveluk, B.; Langer, C.; Solymosi, P.D.; Hille, G.; Nitsche, A.; et al. Infectious KoRV-related retroviruses circulating in Australian bats. Proceedings of the National Academy of Sciences of the United States of America 2020, 117, 9529–9536. [Google Scholar] [CrossRef]
  197. Simmons, G.; Clarke, D.; McKee, J.; Young, P.; Meers, J. Discovery of a novel retrovirus sequence in an Australian native rodent (Melomys burtoni): a putative link between gibbon ape leukemia virus and koala retrovirus. PloS one 2014, 9, e106954. [Google Scholar] [CrossRef]
  198. Gregory Stewart, S.; Gervais, H. The Origins of Gibbon Ape Leukaemia Virus. In Primates, Mark, B., Maurice, P., Eds.; IntechOpen: London, 2017. [Google Scholar]
  199. McMichael, L.; Smith, C.; Gordon, A.; Agnihotri, K.; Meers, J.; Oakey, J. A novel Australian flying-fox retrovirus shares an evolutionary ancestor with Koala, Gibbon and Melomys gamma-retroviruses. Virus genes 2019, 55, 421–424. [Google Scholar] [CrossRef] [PubMed]
  200. Siegal-Willott, J.L.; Jensen, N.; Kimi, D.; Taliaferro, D.; Blankenship, T.; Malinsky, B.; Murray, S.; Eiden, M.V.; Xu, W. Evaluation of captive gibbons (Hylobates spp., Nomascus spp., Symphalangus spp.) in North American Zoological Institutions for Gibbon Ape Leukemia Virus (GALV). J Zoo Wildl Med 2015, 46, 27–33. [Google Scholar] [CrossRef] [PubMed]
  201. Brown, K.; Tarlinton, R.E. Is gibbon ape leukaemia virus still a threat? Mammal Review 2017, 47, 53–61. [Google Scholar] [CrossRef]
  202. Kawakami, T.G.; Sun, L.; McDowell, T.S. Infectious primate type-C virus shed by healthy gibbons. Nature 1977, 268, 448–450. [Google Scholar] [CrossRef]
  203. Krakower, J.M.; Tronick, S.R.; Gallagher, R.E.; Gallo, R.C.; Aaronson, S.A. Antigenic characterization of a new gibbon ape leukemia virus isolate: seroepidemiologic assessment of an outbreak of gibbon leukemia. Int J Cancer 1978, 22, 715–720. [Google Scholar] [CrossRef]
  204. Cornetta, K.; Yao, J.; House, K.; Duffy, L.; Adusumilli, P.S.; Beyer, R.; Booth, C.; Brenner, M.; Curran, K.; Grilley, B.; et al. Replication competent retrovirus testing (RCR) in the National Gene Vector Biorepository: No evidence of RCR in 1,595 post-treatment peripheral blood samples obtained from 60 clinical trials. Molecular therapy : the journal of the American Society of Gene Therapy 2023, 31, 801–809. [Google Scholar] [CrossRef]
  205. McKee, J.; Clark, N.; Shapter, F.; Simmons, G. A new look at the origins of gibbon ape leukemia virus. Virus genes 2017, 53, 165–172. [Google Scholar] [CrossRef]
  206. Sato, K.; Kobayashi, T.; Misawa, N.; Yoshikawa, R.; Takeuchi, J.S.; Miura, T.; Okamoto, M.; Yasunaga, J.; Matsuoka, M.; Ito, M.; et al. Experimental evaluation of the zoonotic infection potency of simian retrovirus type 4 using humanized mouse model. Sci Rep 2015, 5, 14040. [Google Scholar] [CrossRef]
  207. Terry, A.; Kilbey, A.; Naseer, A.; Levy, L.S.; Ahmad, S.; Watts, C.; Mackay, N.; Cameron, E.; Wilson, S.; Neil, J.C. Barriers to Infection of Human Cells by Feline Leukemia Virus: Insights into Resistance to Zoonosis. Journal of virology 2017, 91. [Google Scholar] [CrossRef]
  208. Mendoza, R.; Anderson, M.M.; Overbaugh, J. A putative thiamine transport protein is a receptor for feline leukemia virus subgroup A. Journal of virology 2006, 80, 3378–3385. [Google Scholar] [CrossRef]
  209. Brown, M.A.; Cunningham, M.W.; Roca, A.L.; Troyer, J.L.; Johnson, W.E.; O’Brien, S.J. Genetic characterization of feline leukemia virus from Florida panthers. Emerging infectious diseases 2008, 14, 252–259. [Google Scholar] [CrossRef]
  210. Meli, M.L.; Cattori, V.; Martínez, F.; López, G.; Vargas, A.; Simón, M.A.; Zorrilla, I.; Muñoz, A.; Palomares, F.; López-Bao, J.V.; et al. Feline leukemia virus and other pathogens as important threats to the survival of the critically endangered Iberian lynx (Lynx pardinus). PloS one 2009, 4, e4744. [Google Scholar] [CrossRef] [PubMed]
  211. Petch, R.J.; Gagne, R.B.; Chiu, E.; Mankowski, C.; Rudd, J.; Roelke-Parker, M.; Vickers, T.W.; Logan, K.A.; Alldredge, M.; Clifford, D.; et al. Feline Leukemia Virus Frequently Spills Over from Domestic Cats to North American Pumas. Journal of virology 2022, 96, e0120122. [Google Scholar] [CrossRef] [PubMed]
  212. Sommerfelt, M.A.; Harkestad, N.; Hunter, E. The endogenous langur type D retrovirus PO-1-Lu and its exogenous counterparts in macaque and langur monkeys. Virology 2003, 315, 275–282. [Google Scholar] [CrossRef]
  213. Todaro, G.J.; Benveniste, R.E.; Lieber, M.M.; Livingston, D.M. Infectious type C viruses released by normal cat embryo cells. Virology 1973, 55, 506–515. [Google Scholar] [CrossRef] [PubMed]
  214. Filbert, J.E.; McAllister, R.M.; Nicolson, M.O.; Gilden, R.V.; Lennette, E.H. RD-114 virus infectivity assay by measurements of DNA polymerase activity and virus group specific antigen. Proc Soc Exp Biol Med 1974, 145, 366–370. [Google Scholar] [CrossRef]
  215. Birkenmeier, E.H.; Bonner, T.I.; Reynolds, K.; Searfoss, G.H.; Todaro, G.J. Colobus type C virus: molecular cloning of unintegrated viral DNA and characterization of the endogenous viral genomes of Colobus. Journal of virology 1982, 41, 842–854. [Google Scholar] [CrossRef]
  216. Bohannon, R.C.; Donehower, L.A.; Ford, R.J. Isolation of a type D retrovirus from B-cell lymphomas of a patient with AIDS. Journal of virology 1991, 65, 5663–5672. [Google Scholar] [CrossRef] [PubMed]
  217. Krause, H.; Wunderlich, V.; Uckert, W. Molecular cloning of a type D retrovirus from human cells (PMFV) and its homology to simian acquired immunodeficiency type D retroviruses. Virology 1989, 173, 214–222. [Google Scholar] [CrossRef]
  218. Asikainen, K.; Vesanen, M.; Kuittinen, T.; Vaheri, A. Identification of human type D retrovirus as a contaminant in a neuroblastoma cell line. Archives of virology 1993, 129, 357–361. [Google Scholar] [CrossRef]
  219. Lerche, N.W.; Switzer, W.M.; Yee, J.L.; Shanmugam, V.; Rosenthal, A.N.; Chapman, L.E.; Folks, T.M.; Heneine, W. Evidence of infection with simian type D retrovirus in persons occupationally exposed to nonhuman primates. Journal of virology 2001, 75, 1783–1789. [Google Scholar] [CrossRef] [PubMed]
  220. WHO. Public Health Emergencies of International Concern. Available online: www.who.int/publications/m/item/pathogens-prioritization-a-scientific-framework-for-epidemic-and-pandemic-research-preparedness (accessed on 22 May 2025).
  221. Welsh, R.M., Jr.; Cooper, N.R.; Jensen, F.C.; Oldstone, M.B. Human serum lyses RNA tumour viruses. Nature 1975, 257, 612–614. [Google Scholar] [CrossRef] [PubMed]
  222. Hoshino, H.; Tanaka, H.; Miwa, M.; Okada, H. Human T-cell leukaemia virus is not lysed by human serum. Nature 1984, 310, 324–325. [Google Scholar] [CrossRef]
  223. Banapour, B.; Sernatinger, J.; Levy, J.A. The AIDS-associated retrovirus is not sensitive to lysis or inactivation by human serum. Virology 1986, 152, 268–271. [Google Scholar] [CrossRef]
  224. Takeuchi, Y.; Cosset, F.L.; Lachmann, P.J.; Okada, H.; Weiss, R.A.; Collins, M.K. Type C retrovirus inactivation by human complement is determined by both the viral genome and the producer cell. Journal of virology 1994, 68, 8001–8007. [Google Scholar] [CrossRef]
  225. Galili, U. Mutations Inactivating Biosynthesis of Dispensable Carbohydrate-Antigens Prevented Extinctions in Primate/Human Lineage Evolution. Journal of molecular evolution 2025. [Google Scholar] [CrossRef]
  226. Rother, R.P.; Fodor, W.L.; Springhorn, J.P.; Birks, C.W.; Setter, E.; Sandrin, M.S.; Squinto, S.P.; Rollins, S.A. A novel mechanism of retrovirus inactivation in human serum mediated by anti-alpha-galactosyl natural antibody. The Journal of experimental medicine 1995, 182, 1345–1355. [Google Scholar] [CrossRef]
  227. Moscoso, I.; Hermida-Prieto, M.; Mañez, R.; Lopez-Pelaez, E.; Centeno, A.; Diaz, T.M.; Domenech, N. Lack of cross-species transmission of porcine endogenous retrovirus in pig-to-baboon xenotransplantation with sustained depletion of anti-alphagal antibodies. Transplantation 2005, 79, 777–782. [Google Scholar] [CrossRef] [PubMed]
  228. Lavillette, D.; Marin, M.; Ruggieri, A.; Mallet, F.; Cosset, F.L.; Kabat, D. The envelope glycoprotein of human endogenous retrovirus type W uses a divergent family of amino acid transporters/cell surface receptors. Journal of virology 2002, 76, 6442–6452. [Google Scholar] [CrossRef]
  229. Nethe, M.; Berkhout, B.; van der Kuyl, A.C. Retroviral superinfection resistance. Retrovirology 2005, 2, 52. [Google Scholar] [CrossRef]
  230. Ponferrada, V.G.; Mauck, B.S.; Wooley, D.P. The envelope glycoprotein of human endogenous retrovirus HERV-W induces cellular resistance to spleen necrosis virus. Archives of virology 2003, 148, 659–675. [Google Scholar] [CrossRef]
  231. Gesemann, M.; Lesslauer, A.; Maurer, C.M.; Schönthaler, H.B.; Neuhauss, S.C. Phylogenetic analysis of the vertebrate excitatory/neutral amino acid transporter (SLC1/EAAT) family reveals lineage specific subfamilies. BMC evolutionary biology 2010, 10, 117. [Google Scholar] [CrossRef]
  232. Green, R.; Ireton, R.C.; Gale, M., Jr. Interferon-stimulated genes: new platforms and computational approaches. Mamm Genome 2018, 29, 593–602. [Google Scholar] [CrossRef] [PubMed]
  233. Schoggins, J.W. Interferon-Stimulated Genes: What Do They All Do? Annual review of virology 2019, 6, 567–584. [Google Scholar] [CrossRef] [PubMed]
  234. Kassiotis, G.; Stoye, J.P. Evolution of antiviral host defenses against a backdrop of endogenous retroelements. Science (New York, N.Y.) 2025, 389, 588–593. [Google Scholar] [CrossRef]
  235. Nchioua, R.; Kmiec, D.; Krchlikova, V.; Mattes, S.; Noettger, S.; Bibollet-Ruche, F.; Russell, R.M.; Sparrer, K.M.J.; Charpentier, T.; Tardy, F.; et al. Host ZAP activity correlates with the levels of CpG suppression in primate lentiviruses. Proceedings of the National Academy of Sciences of the United States of America 2025, 122, e2419489122. [Google Scholar] [CrossRef]
  236. Jin, M.J.; Rogers, J.; Phillips-Conroy, J.E.; Allan, J.S.; Desrosiers, R.C.; Shaw, G.M.; Sharp, P.M.; Hahn, B.H. Infection of a yellow baboon with simian immunodeficiency virus from African green monkeys: evidence for cross-species transmission in the wild. Journal of virology 1994, 68, 8454–8460. [Google Scholar] [CrossRef]
  237. Mang, R.; Maas, J.; van Der Kuyl, A.C.; Goudsmit, J. Papio cynocephalus endogenous retrovirus among old world monkeys: evidence for coevolution and ancient cross-species transmissions. Journal of virology 2000, 74, 1578–1586. [Google Scholar] [CrossRef] [PubMed]
  238. Nyamota, R.; Owino, V.; Murungi, E.K.; Villinger, J.; Otiende, M.; Masiga, D.; Thuita, J.; Lekolool, I.; Jeneby, M. Broad diversity of simian immunodeficiency virus infecting Chlorocebus species (African green monkey) and evidence of cross-species infection in Papio anubis (olive baboon) in Kenya. Journal of medical primatology 2020, 49, 165–178. [Google Scholar] [CrossRef]
  239. Parrish, C.R.; Holmes, E.C.; Morens, D.M.; Park, E.C.; Burke, D.S.; Calisher, C.H.; Laughlin, C.A.; Saif, L.J.; Daszak, P. Cross-species virus transmission and the emergence of new epidemic diseases. Microbiology and molecular biology reviews : MMBR 2008, 72, 457–470. [Google Scholar] [CrossRef]
  240. Kreuder Johnson, C.; Hitchens, P.L.; Smiley Evans, T.; Goldstein, T.; Thomas, K.; Clements, A.; Joly, D.O.; Wolfe, N.D.; Daszak, P.; Karesh, W.B.; et al. Spillover and pandemic properties of zoonotic viruses with high host plasticity. Sci Rep 2015, 5, 14830. [Google Scholar] [CrossRef]
  241. Greenspoon, L.; Ramot, N.; Moran, U.; Roll, U.; Phillips, R.; Noor, E.; Milo, R. The global biomass of mammals since 1850. Nature communications 2025, 16, 8338. [Google Scholar] [CrossRef]
  242. Sikora, M.; Canteri, E.; Fernandez-Guerra, A.; Oskolkov, N.; Ågren, R.; Hansson, L.; Irving-Pease, E.K.; Mühlemann, B.; Holtsmark Nielsen, S.; Scorrano, G.; et al. The spatiotemporal distribution of human pathogens in ancient Eurasia. Nature 2025, 643, 1011–1019. [Google Scholar] [CrossRef]
  243. Mang, R.; Maas, J.; Chen, X.; Goudsmit, J.; van Der Kuyl, A.C. Identification of a novel type C porcine endogenous retrovirus: evidence that copy number of endogenous retroviruses increases during host inbreeding. The Journal of general virology 2001, 82, 1829–1834. [Google Scholar] [CrossRef]
  244. Miyazawa, T.; Yoshikawa, R.; Golder, M.; Okada, M.; Stewart, H.; Palmarini, M. Isolation of an infectious endogenous retrovirus in a proportion of live attenuated vaccines for pets. Journal of virology 2010, 84, 3690–3694. [Google Scholar] [CrossRef]
  245. Shimode, S.; Sakuma, T.; Yamamoto, T.; Miyazawa, T. Establishment of CRFK cells for vaccine production by inactivating endogenous retrovirus with TALEN technology. Sci Rep 2022, 12, 6641. [Google Scholar] [CrossRef] [PubMed]
  246. Narushima, R.; Horiuchi, N.; Usui, T.; Ogawa, T.; Takahashi, T.; Shimazaki, T. Experimental infection of dogs with a feline endogenous retrovirus RD-114. Acta veterinaria Scandinavica 2011, 53, 3. [Google Scholar] [CrossRef] [PubMed]
  247. Yoshikawa, R.; Yasuda, J.; Kobayashi, T.; Miyazawa, T. Canine ASCT1 and ASCT2 are functional receptors for RD-114 virus in dogs. The Journal of general virology 2012, 93, 603–607. [Google Scholar] [CrossRef]
  248. Victoria, J.G.; Wang, C.; Jones, M.S.; Jaing, C.; McLoughlin, K.; Gardner, S.; Delwart, E.L. Viral nucleic acids in live-attenuated vaccines: detection of minority variants and an adventitious virus. Journal of virology 2010, 84, 6033–6040. [Google Scholar] [CrossRef]
  249. Fukumoto, H.; Hishima, T.; Hasegawa, H.; Saeki, H.; Kuroda, M.; Katano, H. Evaluation of Vero-cell-derived simian endogenous retrovirus infection in humans by detection of viral genome in clinicopathological samples and commercialized vaccines and by serology of Japanese general population. Vaccine 2016, 34, 2700–2706. [Google Scholar] [CrossRef]
  250. Tsang, S.X.; Switzer, W.M.; Shanmugam, V.; Johnson, J.A.; Goldsmith, C.; Wright, A.; Fadly, A.; Thea, D.; Jaffe, H.; Folks, T.M.; et al. Evidence of avian leukosis virus subgroup E and endogenous avian virus in measles and mumps vaccines derived from chicken cells: investigation of transmission to vaccine recipients. Journal of virology 1999, 73, 5843–5851. [Google Scholar] [CrossRef]
  251. Hussain, A.I.; Shanmugam, V.; Switzer, W.M.; Tsang, S.X.; Fadly, A.; Thea, D.; Helfand, R.; Bellini, W.J.; Folks, T.M.; Heneine, W. Lack of evidence of endogenous avian leukosis virus and endogenous avian retrovirus transmission to measles, mumps, and rubella vaccine recipients. Emerging infectious diseases 2001, 7, 66–72. [Google Scholar] [CrossRef]
  252. Hussain, A.I.; Johnson, J.A.; Da Silva Freire, M.; Heneine, W. Identification and characterization of avian retroviruses in chicken embryo-derived yellow fever vaccines: investigation of transmission to vaccine recipients. Journal of virology 2003, 77, 1105–1111. [Google Scholar] [CrossRef] [PubMed]
  253. Ajayi, O.O.; Cullinan, J.L.; Basria, I.; Fuentes-Arias, M.; Osuna-Najarro, A.; Johnson, S.; Faison, T.; Lute, S. Analysis of Virus Clearance for Biotechnology Manufacturing Processes from Early to Late Phase Development. PDA J Pharm Sci Technol 2025, 79, 252–273. [Google Scholar] [CrossRef]
  254. Chin, P.J.; Lambert, C.; Beurdelay, P.; Charlebois, R.L.; Colinet, A.S.; Eloit, M.; Gilchrist, S.; Gilleece, M.; Hess, M.; Leimbach, A.; et al. Virus detection by short read high throughput sequencing in a high virus low cellular background. NPJ Vaccines 2025, 10, 61. [Google Scholar] [CrossRef] [PubMed]
  255. Allan, J.S.; Broussard, S.R.; Michaels, M.G.; Starzl, T.E.; Leighton, K.L.; Whitehead, E.M.; Comuzzie, A.G.; Lanford, R.E.; Leland, M.M.; Switzer, W.M.; et al. Amplification of simian retroviral sequences from human recipients of baboon liver transplants. AIDS research and human retroviruses 1998, 14, 821–824. [Google Scholar] [CrossRef]
  256. Michaels, M.G.; Kaufman, C.; Volberding, P.A.; Gupta, P.; Switzer, W.M.; Heneine, W.; Sandstrom, P.; Kaplan, L.; Swift, P.; Damon, L.; et al. Baboon bone-marrow xenotransplant in a patient with advanced HIV disease: case report and 8-year follow-up. Transplantation 2004, 78, 1582–1589. [Google Scholar] [CrossRef] [PubMed]
  257. Denner, J.; Jhelum, H.; Ban, J.; Krabben, L.; Kaufer, B.B. How to Detect Porcine Endogenous Retrovirus (PERV) Infections in Patients After Transplantation of Pig Organs. Xenotransplantation 2025, 32, e70028. [Google Scholar] [CrossRef] [PubMed]
  258. Anand, R.P.; Layer, J.V.; Heja, D.; Hirose, T.; Lassiter, G.; Firl, D.J.; Paragas, V.B.; Akkad, A.; Chhangawala, S.; Colvin, R.B.; et al. Design and testing of a humanized porcine donor for xenotransplantation. Nature 2023, 622, 393–401. [Google Scholar] [CrossRef]
  259. Kawai, T.; Williams, W.W.; Elias, N.; Fishman, J.A.; Crisalli, K.; Longchamp, A.; Rosales, I.A.; Duggan, M.; Kimura, S.; Morena, L.; et al. Xenotransplantation of a Porcine Kidney for End-Stage Kidney Disease. The New England journal of medicine 2025, 392, 1933–1940. [Google Scholar] [CrossRef]
  260. The organ farm. Available online: www.science.org/content/article/can-gene-edited-pigs-solve-organ-transplant-shortage (accessed on 7 november 2025).
  261. Study to Evaluate the Safety and Efficacy of the 10 GE Xenokidney in Patients With ESRD (EXPAND). Available online: https://clinicaltrials.gov/study/NCT06878560 (accessed on October 27 2025).
  262. Ban, J.; Krabben, L.; Kaufer, B.B.; Denner, J. Neutralizing Antibodies Against the Porcine Endogenous Retroviruses (PERVs). Viruses 2025, 17. [Google Scholar] [CrossRef]
  263. Maetzig, T.; Galla, M.; Baum, C.; Schambach, A. Gammaretroviral vectors: biology, technology and application. Viruses 2011, 3, 677–713. [Google Scholar] [CrossRef] [PubMed]
  264. Watanabe, N.; McKenna, M.K. Generation of CAR T-cells using γ-retroviral vector. Methods Cell Biol 2022, 167, 171–183. [Google Scholar] [CrossRef] [PubMed]
  265. Weber, S.L.K.; Heinzel, T.; Iyer-Bierhoff, A. Protocol for ecotropic pseudotyped lentiviral transduction of “murinized” human cells while decreasing risk of infection for laboratory personnel. STAR Protoc 2025, 6, 103708. [Google Scholar] [CrossRef]
  266. Taghdiri, M.; Mussolino, C. Viral and Non-Viral Systems to Deliver Gene Therapeutics to Clinical Targets. International journal of molecular sciences 2024, 25. [Google Scholar] [CrossRef]
  267. Koppers-Lalic, D.; Hoeben, R.C. Non-human viruses developed as therapeutic agent for use in humans. Rev Med Virol 2011, 21, 227–239. [Google Scholar] [CrossRef]
  268. Lu, Y.C.; Luo, Y.P.; Wang, Y.W.; Tai, C.K. Highly efficient gene transfer to solid tumors in vivo by tumor-selective replicating retrovirus vectors. Int J Mol Med 2010, 25, 769–775. [Google Scholar] [CrossRef]
  269. Lu, Y.C.; Chen, Y.J.; Yu, Y.R.; Lai, Y.H.; Cheng, J.C.; Li, Y.F.; Shen, C.H.; Tai, C.K. Replicating retroviral vectors for oncolytic virotherapy of experimental hepatocellular carcinoma. Oncol Rep 2012, 28, 21–26. [Google Scholar] [CrossRef]
  270. Collins, S.A.; Shah, A.H.; Ostertag, D.; Kasahara, N.; Jolly, D.J. Clinical development of retroviral replicating vector Toca 511 for gene therapy of cancer. Expert Opin Biol Ther 2021, 21, 1199–1214. [Google Scholar] [CrossRef] [PubMed]
  271. Hogan, D.J.; Zhu, J.J.; Diago, O.R.; Gammon, D.; Haghighi, A.; Lu, G.; Das, A.; Gruber, H.E.; Jolly, D.J.; Ostertag, D. Molecular Analyses Support the Safety and Activity of Retroviral Replicating Vector Toca 511 in Patients. Clin Cancer Res 2018, 24, 4680–4693. [Google Scholar] [CrossRef]
  272. Donahue, R.E.; Kessler, S.W.; Bodine, D.; McDonagh, K.; Dunbar, C.; Goodman, S.; Agricola, B.; Byrne, E.; Raffeld, M.; Moen, R.; et al. Helper virus induced T cell lymphoma in nonhuman primates after retroviral mediated gene transfer. The Journal of experimental medicine 1992, 176, 1125–1135. [Google Scholar] [CrossRef]
  273. Vanin, E.F.; Kaloss, M.; Broscius, C.; Nienhuis, A.W. Characterization of replication-competent retroviruses from nonhuman primates with virus-induced T-cell lymphomas and observations regarding the mechanism of oncogenesis. Journal of virology 1994, 68, 4241–4250. [Google Scholar] [CrossRef] [PubMed]
  274. Purcell, D.F.; Broscius, C.M.; Vanin, E.F.; Buckler, C.E.; Nienhuis, A.W.; Martin, M.A. An array of murine leukemia virus-related elements is transmitted and expressed in a primate recipient of retroviral gene transfer. Journal of virology 1996, 70, 887–897. [Google Scholar] [CrossRef]
  275. Farley, D.; Stockdale, S.; Moore-Kelly, C.; Miskin, J.; Reiser, J.; Mitrophanous, K. Risks of replication-competent retro/lentivirus from associated vector systems: Is it time for a roadmap toward reduced testing? Molecular Therapy Methods & Clinical Development 2025, 33, 101601. [Google Scholar] [CrossRef]
  276. Wilson, C.A.; Wong, S.; Muller, J.; Davidson, C.E.; Rose, T.M.; Burd, P. Type C retrovirus released from porcine primary peripheral blood mononuclear cells infects human cells. Journal of virology 1998, 72, 3082–3087. [Google Scholar] [CrossRef]
  277. Lerche, N.W.; Marx, P.A.; Gardner, M.B. Elimination of type D retrovirus infection from group-housed rhesus monkeys using serial testing and removal. Lab Anim Sci 1991, 41, 123–127. [Google Scholar]
  278. Westman, M.E.; Coggins, S.J.; van Dorsselaer, M.; Norris, J.M.; Squires, R.A.; Thompson, M.; Malik, R. Feline leukaemia virus (FeLV) infection in domestic pet cats in Australia and New Zealand: Guidelines for diagnosis, prevention and management. Aust Vet J 2025, 103, 617–635. [Google Scholar] [CrossRef]
  279. Olagoke, O.; Miller, D.; Hemmatzadeh, F.; Stephenson, T.; Fabijan, J.; Hutt, P.; Finch, S.; Speight, N.; Timms, P. Induction of neutralizing antibody response against koala retrovirus (KoRV) and reduction in viral load in koalas following vaccination with recombinant KoRV envelope protein. NPJ Vaccines 2018, 3, 30. [Google Scholar] [CrossRef]
  280. Francis, D.P.; Essex, M.; Hardy, W.D., Jr. Excretion of feline leukaemia virus by naturally infected pet cats. Nature 1977, 269, 252–254. [Google Scholar] [CrossRef]
  281. Kawakami, T.G.; Sun, L.; McDowell, T.S. Natural transmission of gibbon leukemia virus. J Natl Cancer Inst 1978, 61, 1113–1115. [Google Scholar]
  282. Gomes-Keller, M.A.; Tandon, R.; Gönczi, E.; Meli, M.L.; Hofmann-Lehmann, R.; Lutz, H. Shedding of feline leukemia virus RNA in saliva is a consistent feature in viremic cats. Veterinary microbiology 2006, 112, 11–21. [Google Scholar] [CrossRef] [PubMed]
  283. Cattori, V.; Tandon, R.; Riond, B.; Pepin, A.C.; Lutz, H.; Hofmann-Lehmann, R. The kinetics of feline leukaemia virus shedding in experimentally infected cats are associated with infection outcome. Veterinary microbiology 2009, 133, 292–296. [Google Scholar] [CrossRef] [PubMed]
  284. Gallo, R.C.; Gallagher, R.E.; Wong-Staal, F.; Aoki, T.; Markham, P.D.; Schetters, H.; Ruscetti, F.; Valerio, M.; Walling, M.J.; O’Keeffe, R.T.; et al. Isolation and tissue distribution of type-C virus and viral components from a gibbon ape (Hylobates lar) with lymphocytic leukemia. Virology 1978, 84, 359–373. [Google Scholar] [CrossRef]
  285. Kawakami, T.G.; Huff, S.D.; Buckley, P.M.; Dungworth, D.L.; Synder, S.P.; Gilden, R.V. C-type virus associated with gibbon lymphosarcoma. Nat New Biol 1972, 235, 170–171. [Google Scholar] [CrossRef]
  286. Oliveira, N.M.; Farrell, K.B.; Eiden, M.V. In vitro characterization of a koala retrovirus. Journal of virology 2006, 80, 3104–3107. [Google Scholar] [CrossRef]
  287. Hanger, J.J.; Bromham, L.D.; McKee, J.J.; O’Brien, T.M.; Robinson, W.F. The nucleotide sequence of koala (Phascolarctos cinereus) retrovirus: a novel type C endogenous virus related to Gibbon ape leukemia virus. Journal of virology 2000, 74, 4264–4272. [Google Scholar] [CrossRef]
  288. Blyton, M.D.J.; Young, P.R.; Moore, B.D.; Chappell, K.J. Geographic patterns of koala retrovirus genetic diversity, endogenization, and subtype distributions. Proceedings of the National Academy of Sciences of the United States of America 2022, 119, e2122680119. [Google Scholar] [CrossRef] [PubMed]
  289. Garcia-Montojo, M.; Doucet-O’Hare, T.; Henderson, L.; Nath, A. Human endogenous retrovirus-K (HML-2): a comprehensive review. Crit Rev Microbiol 2018, 44, 715–738. [Google Scholar] [CrossRef]
  290. Karamitros, T.; Hurst, T.; Marchi, E.; Karamichali, E.; Georgopoulou, U.; Mentis, A.; Riepsaame, J.; Lin, A.; Paraskevis, D.; Hatzakis, A.; et al. Human Endogenous Retrovirus-K HML-2 integration within RASGRF2 is associated with intravenous drug abuse and modulates transcription in a cell-line model. Proceedings of the National Academy of Sciences of the United States of America 2018, 115, 10434–10439. [Google Scholar] [CrossRef] [PubMed]
  291. Xue, B.; Sechi, L.A.; Kelvin, D.J. Human Endogenous Retrovirus K (HML-2) in Health and Disease. Frontiers in microbiology 2020, 11, 1690. [Google Scholar] [CrossRef] [PubMed]
  292. van der Kuyl, A.C.; Dekker, J.T.; Goudsmit, J. Discovery of a new endogenous type C retrovirus (FcEV) in cats: evidence for RD-114 being an FcEV(Gag-Pol)/baboon endogenous virus BaEV(Env) recombinant. Journal of virology 1999, 73, 7994–8002. [Google Scholar] [CrossRef]
  293. Henzy, J.E.; Gifford, R.J.; Johnson, W.E.; Coffin, J.M. A novel recombinant retrovirus in the genomes of modern birds combines features of avian and mammalian retroviruses. Journal of virology 2014, 88, 2398–2405. [Google Scholar] [CrossRef]
  294. Vargiu, L.; Rodriguez-Tomé, P.; Sperber, G.O.; Cadeddu, M.; Grandi, N.; Blikstad, V.; Tramontano, E.; Blomberg, J. Classification and characterization of human endogenous retroviruses; mosaic forms are common. Retrovirology 2016, 13, 7. [Google Scholar] [CrossRef]
  295. Tönjes, R.R.; Löwer, R.; Boller, K.; Denner, J.; Hasenmaier, B.; Kirsch, H.; König, H.; Korbmacher, C.; Limbach, C.; Lugert, R.; et al. HERV-K: the biologically most active human endogenous retrovirus family. J Acquir Immune Defic Syndr Hum Retrovirol 1996, 13 Suppl 1, S261–267. [Google Scholar] [CrossRef] [PubMed]
  296. Berkhout, B.; Jebbink, M.; Zsiros, J. Identification of an active reverse transcriptase enzyme encoded by a human endogenous HERV-K retrovirus. Journal of virology 1999, 73, 2365–2375. [Google Scholar] [CrossRef]
  297. Wolfe, N.D.; Switzer, W.M.; Carr, J.K.; Bhullar, V.B.; Shanmugam, V.; Tamoufe, U.; Prosser, A.T.; Torimiro, J.N.; Wright, A.; Mpoudi-Ngole, E.; et al. Naturally acquired simian retrovirus infections in central African hunters. Lancet (London, England) 2004, 363, 932–937. [Google Scholar] [CrossRef]
  298. Switzer, W.M.; Bhullar, V.; Shanmugam, V.; Cong, M.E.; Parekh, B.; Lerche, N.W.; Yee, J.L.; Ely, J.J.; Boneva, R.; Chapman, L.E.; et al. Frequent simian foamy virus infection in persons occupationally exposed to nonhuman primates. Journal of virology 2004, 78, 2780–2789. [Google Scholar] [CrossRef]
  299. Mottaghinia, S.; Stenzel, S.; Tsangaras, K.; Nikolaidis, N.; Laue, M.; Müller, K.; Hölscher, H.; Löber, U.; McEwen, G.K.; Donnellan, S.C.; et al. A recent gibbon ape leukemia virus germline integration in a rodent from New Guinea. Proceedings of the National Academy of Sciences of the United States of America 2024, 121, e2220392121. [Google Scholar] [CrossRef]
  300. Lerche, N.W.; Henrickson, R.V.; Maul, D.H.; Gardner, M.B. Epidemiologic aspects of an outbreak of acquired immunodeficiency in rhesus monkeys (Macaca mulatta). Lab Anim Sci 1984, 34, 146–150. [Google Scholar]
  301. Gardner, M.B. The history of simian AIDS. Journal of medical primatology 1996, 25, 148–157. [Google Scholar] [CrossRef] [PubMed]
  302. Yoshikawa, R.; Okamoto, M.; Sakaguchi, S.; Nakagawa, S.; Miura, T.; Hirai, H.; Miyazawa, T. Simian retrovirus 4 induces lethal acute thrombocytopenia in Japanese macaques. Journal of virology 2015, 89, 3965–3975. [Google Scholar] [CrossRef] [PubMed]
  303. Murphy, H.W.; Miller, M.; Ramer, J.; Travis, D.; Barbiers, R.; Wolfe, N.D.; Switzer, W.M. Implications of simian retroviruses for captive primate population management and the occupational safety of primate handlers. J Zoo Wildl Med 2006, 37, 219–233. [Google Scholar] [CrossRef]
  304. Schroder, M.A.; Fisk, S.K.; Lerche, N.W. Eradication of simian retrovirus type D from a colony of cynomolgus, rhesus, and stump-tailed macaques by using serial testing and removal. Contemp Top Lab Anim Sci 2000, 39, 16–23. [Google Scholar]
  305. Montiel, N.A. An updated review of simian betaretrovirus (SRV) in macaque hosts. Journal of medical primatology 2010, 39, 303–314. [Google Scholar] [CrossRef]
  306. Yee, J.L.; Grant, R.; Van Rompay, K.K.; Kuller, L.; Carpenter, A.; Watanabe, R.; Huebner, R.; Agricola, B.; Smedley, J.; Roberts, J.A. Emerging diagnostic challenges and characteristics of simian betaretrovirus infections in captive macaque colonies. Journal of medical primatology 2017, 46, 149–153. [Google Scholar] [CrossRef]
  307. Nham, P.; Halley, B.; Van Rompay, K.K.A.; Roberts, J.; Yee, J. Simian retrovirus transmission in rhesus macaques. Journal of medical primatology 2024, 53, e12726. [Google Scholar] [CrossRef]
  308. Driscoll, C.A.; Clutton-Brock, J.; Kitchener, A.C.; O’Brien, S.J. The Taming of the cat. Genetic and archaeological findings hint that wildcats became housecats earlier--and in a different place--than previously thought. Sci Am 2009, 300, 68–75. [Google Scholar] [CrossRef] [PubMed]
  309. Benveniste, R.E.; Sherr, C.J.; Todaro, G.J. Evolution of type C viral genes: origin of feline leukemia virus. Science (New York, N.Y.) 1975, 190, 886–888. [Google Scholar] [CrossRef]
  310. Giselbrecht, J.; Jähne, S.; Bergmann, M.; Meli, M.L.; Pineroli, B.; Boenzli, E.; Teichmann-Knorrn, S.; Zablotski, Y.; Pennisi, M.G.; Layachi, N.; et al. Prevalence of Different Courses of Feline Leukaemia Virus Infection in Four European Countries. Viruses 2023, 15. [Google Scholar] [CrossRef]
  311. Stubbe, S.P.; Lang, J.; Nagler, N.; Müller, S.F.; Lierz, M. High Prevalence of Antigens of and Specific Antibodies Against Various Viral Pathogens in European Wildcats (Felis silvestris) from Southwest Germany, 2020-22. J Wildl Dis 2025, 61, 663–673. [Google Scholar] [CrossRef]
  312. Hashem, M.A.; Kayesh, M.E.H.; Yamato, O.; Maetani, F.; Eiei, T.; Mochizuki, K.; Sakurai, H.; Ito, A.; Kannno, H.; Kasahara, T.; et al. Coinfection with koala retrovirus subtypes A and B and its impact on captive koalas in Japanese zoos. Archives of virology 2019, 164, 2735–2745. [Google Scholar] [CrossRef]
  313. Zheng, H.; Pan, Y.; Tang, S.; Pye, G.W.; Stadler, C.K.; Vogelnest, L.; Herrin, K.V.; Rideout, B.A.; Switzer, W.M. Koala retrovirus diversity, transmissibility, and disease associations. Retrovirology 2020, 17, 34. [Google Scholar] [CrossRef]
  314. Joyce, B.A.; Blyton, M.D.J.; Johnston, S.D.; Meikle, W.D.; Vinette Herrin, K.; Madden, C.; Young, P.R.; Chappell, K.J. Diversity and transmission of koala retrovirus: a case study in three captive koala populations. Sci Rep 2022, 12, 15787. [Google Scholar] [CrossRef] [PubMed]
  315. Kanda, T.; Yajima, M.; Ikuta, K. Epstein-Barr virus strain variation and cancer. Cancer Sci 2019, 110, 1132–1139. [Google Scholar] [CrossRef] [PubMed]
  316. Tocagen’s cancer drug fails to improve survival in glioma trial. Available online: www.clinicaltrialsarena.com/news/tocagen-glioma-phaseiii-fails/ (accessed on November 5 2025).
  317. Cloughesy, T.F.; Petrecca, K.; Walbert, T.; Butowski, N.; Salacz, M.; Perry, J.; Damek, D.; Bota, D.; Bettegowda, C.; Zhu, J.J.; et al. Effect of Vocimagene Amiretrorepvec in Combination With Flucytosine vs Standard of Care on Survival Following Tumor Resection in Patients With Recurrent High-Grade Glioma: A Randomized Clinical Trial. JAMA Oncol 2020, 6, 1939–1946. [Google Scholar] [CrossRef]
  318. Reitz, M.S., Jr.; Voltin, M.; Gallo, R.C. Characterization of a partial provirus from a gibbon ape naturally infected with gibbon ape leukemia virus. Virology 1980, 104, 474–481. [Google Scholar] [CrossRef] [PubMed]
  319. Cohen, J.I.; Iwatsuki, K.; Ko, Y.H.; Kimura, H.; Manoli, I.; Ohshima, K.; Pittaluga, S.; Quintanilla-Martinez, L.; Jaffe, E.S. Epstein-Barr virus NK and T cell lymphoproliferative disease: report of a 2018 international meeting. Leuk Lymphoma 2020, 61, 808–819. [Google Scholar] [CrossRef]
  320. Cancer Today. Available online: https://gco.iarc.fr/today/en (accessed on 11 November 2025).
  321. Liang, Y.; Tang, Y.; WanYan, Y.; Li, E. Comparative global burden analysis of lymphoma subtypes: a statistical evaluation of severity across global regions. Front Public Health 2025, 13, 1590093. [Google Scholar] [CrossRef] [PubMed]
  322. Jarosz, A.S.; Pendleton, A.L.; Lashbrook, M.J.; Cech, E.; Altieri, M.; Kunch, A.; Modiano, J.F.; Halo, J.V. Expression and high levels of insertional polymorphism of an endogenous gammaretrovirus lineage in dogs. PLoS genetics 2023, 19, e1011083. [Google Scholar] [CrossRef] [PubMed]
Table 1. Gamma- and betaretroviruses included in the review.
Table 1. Gamma- and betaretroviruses included in the review.
Retrovirus group Virus name* Abbreviation* Host species
ICTV examplar gammaretroviruses reticuloendotheliosis virus REV bird
Trager duck spleen necrosis virus SNV bird
chick syncytial virus CSV bird
murine leukemia virus MLV mouse
feline leukemia virus FeLV cat
gibbon ape leukemia virus GALV gibbon
koala retrovirus KoRV koala
porcine type-C oncovirus PCOV/PERV pig
ICTV examplar gammaretroviruses needing a helper virus Finkel-Biskis-Jinkins murine sarcoma virus - mouse
Harvey murine sarcoma virus - mouse
Kirsten murine sarcoma virus - mouse
Moloney murine sarcoma virus - mouse
Hardy-Zuckerman feline sarcoma virus - cat
Snyder-Theilen feline sarcoma virus - cat
woolly monkey sarcoma virus WMSV woolly monkey
ICTV examplar betaretroviruses with a gamma-type env gene Po-1-Lu Po-1-Lu langur monkey
Mason-Pfizer monkey virus MPMV/SRV3 rhesus monkey
squirrel monkey retrovirus SMRV squirrel monkey
Non-ICTV gammaretroviruses baboon endogenous virus BaEV baboon
MAC-1 MAC-1 rhesus monkey
CPC-1 CPC-1 colobus monkey
Non-ICTV betaretroviruses with a gamma-type env gene simian endogenous retrovirus SERV African green monkey
RD-114 RD-114 cat
* Common name/abbreviation, which is not the offical ICTV name/abbreviation since 2023.
Table 2. Possible sources of gammaretrovirus transmission to humans with recommendations to decrease risks.
Table 2. Possible sources of gammaretrovirus transmission to humans with recommendations to decrease risks.
Gammaretrovirus1 Possible infection source Recommendation
Avian gammaretroviruses
(REV, SNV, CSV)
Poultry products Screen flocks
GALV Contact with gibbons
Clinical use of RRVs
Protective measures2
Limit use
MLV Clinical use of RRVs Limit use
FeLV Contact with pet cats Vaccinate cats3
KORV Exposure to koalas Protective measures
Vaccinate koalas4
PERV/PCOV Xenotransplantation Edit pig genome
SRV1-3 Exposure to primates Protective measures2
Primate ERVs5
(Po-1-Lu, SMRV, BaEV,
MAC-1, CPC-1, SERV)
Exposure to primates
Cell line products
Protective measures
Perform quality checks
Cat ERV RD-114 Cell line products Edit cell line genome
‘Sarcoma’ retroviruses None None
1 The color of the row indicates the level of endogenization, with white implying no endogenization in the original animal source, light gray depicts viruses with both endogenous and exogenous variants, and dark gray represents endogenous proviruses that are activated only under specific conditions. 2 GALV infection in gibbons has not been reported since the 1970s. SRV prevalence in captive monkeys has declined significantly after 1990 [277]. 3 Cat vaccines are available in most countries, and are strongly advised [278]. 4 No vaccine has been marketed yet, but a therapeutic vaccine proved to be effective [279]. 5 Xenotransplantation of baboon tissue has been discontinued.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.
Copyright: This open access article is published under a Creative Commons CC BY 4.0 license, which permit the free download, distribution, and reuse, provided that the author and preprint are cited in any reuse.
Prerpints.org logo

Preprints.org is a free preprint server supported by MDPI in Basel, Switzerland.

Subscribe

Disclaimer

Terms of Use

Privacy Policy

Privacy Settings

© 2025 MDPI (Basel, Switzerland) unless otherwise stated