key: cord-0809699-o66dosgp authors: Stingi, Aureliano; Cirillo, Luca title: SARS‐CoV‐2 infection and cancer: Evidence for and against a role of SARS‐CoV‐2 in cancer onset date: 2021-06-03 journal: Bioessays DOI: 10.1002/bies.202000289 sha: 81ad1a44e191cabc6146c8607e2077ba5cd0687b doc_id: 809699 cord_uid: o66dosgp Despite huge efforts towards understanding the severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) pathogenesis, little is known about the long‐term consequences of the disease. Here, we critically review existing literature about oncogenesis as a potential long‐term effect of SARS‐CoV‐2 infection. Like other viral infections, SARS‐CoV‐2 may promote cancer onset by inhibiting tumor suppressor genes. We conclude that, although unlikely, such hypothesis cannot be excluded a priori and we delineate an experimental approach to address it. To date, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and the ongoing outbreak of the novel coronavirus disease 2019 (known as has rapidly become one of the most important global health problems-infecting more than 150 million people worldwide and causing over 3 million deaths. Besides acute symptoms, there are rising concerns about long-term effects of the viral infection. Convalescent patients, with no detectable viral load, report a variety of long-lasting symptoms that puzzle scientists and clinicians. [1, 2] These long-lasting symptoms are worrisome from a public health perspective, as people experiencing them may need medical care for months or years. A recent report proposed that among long-term effects linked to SARS-CoV-2 infection there may be an oncogenic potential. [3] Here, we further discuss this hypothesis by comparing SARS-CoV-2 with other oncogenic viruses and evaluate potential molecular mechanisms a causal link between viral infections and some forms of cancer [5, 6] and paved the way to understanding the oncogenic potential of viruses. Such oncogenic potential was later described in other animal viruses, including human viruses. [7, 8] Today, the link between viral infection and cancer is well established and recognized as one of the most pressing public health problems. The mechanism through which a viral infection degenerates into cancer can vary from a sustained inflammatory reaction to a suppression of the immune system to an active reprogramming of the host cell. Broadly speaking, a virus can directly trigger cell transformation in the following ways: by providing an external oncogene, by over-activating human oncogenes, and/or by inhibiting tumor suppressors. [9] A textbook example of virus-mediated inhibition of oncosuppressor is the human papilloma virus (HPV). HPV infected cells express two viral proteins named E6 and E7 that bind and inhibit the two tumor suppressors p53 and pRB. HPV E7 protein contains a LXCXE motif and mutation or deletion of this short sequence abolishes the interaction between E7 and pRB. [10] In addition, the oncogenic potential of different HPV strains seems to correlate with the amount of E6 and E7 expression and with their affinity to their targets. [11, 12] Overall, it is not uncommon for a virus to spark cell transformation and cancer. Since Sars-CoV-2 is a novel virus with uncertainty about its long-term effects, we asked whether Sars-CoV-2 infection could promote cancer onset. Multiple mechanisms could link Sars-CoV-2 infection with cancer onset. The most important one being a potential inhibition of oncosuppressors in Sars-CoV-2-infected cells. SARS-CoV, the virus responsible for the 2002 pandemic of the severe acute respiratory syndrome (SARS), expresses the endoribonuclease non-structural protein 15 (Nsp15) that interacts with pRB tumor suppressor through a LXCXE motif. [13] The interaction between Nsp15 and pRB induces its nuclear export and ubiquitination, targeting pRB for proteasomal degradation. NIH-3T cells expressing Nsp15 lose contact inhibition and display an increased proliferative potential compared to wild-type cells-indicating a role of Nsp15 in cell transformation. [13] Altogether, Nsp15 expression has the potential to inactivate pRB, to induce cell transformation and eventually cancer. Nsp15 nuclease is present and conserved in the SARS-CoV-2 genome, sharing 88.7% sequence similarity with its orthologue in SARS-CoV. In addition, the pRB interacting motif LXCXE is conserved both in terms of sequence and primary structure location. Although no experimental evidence exists to date, an interaction between Nsp15 and the tumor suppressor pRB seems highly likely. In this instance, a cell infected with SARS-CoV-2 would experience Nsp15-mediated pRB inhibition. In addition to pRB inhibition, Sars-CoV-1 could also increase p53 degradation via non-structural protein 3 (Nsp3) protein. The papainlike protease of Nsp3 interacts and stabilizes the E3 ubiquitin ligase RCHY1 (ring-finger and CHY zinc-finger domain-containing). [14] One of the main targets of RCHY1 is the tumor suppressor p53. [15, 16] Stabilization of RCHY1 results in decreased levels of p53 and a weaker G1 checkpoint. Similar to Nsp15, Nsp3 is highly conserved between SARS-CoV-1 and SARS-CoV-2 (76% sequence similarity) suggesting that Nsp3 from SARS-CoV-2 has the potential to reduce p53 levels increasing the likelihood of cell transformation. Both p53 and pRB are widely recognized as important tumor suppressor genes. p53 is the most frequently mutated gene in human cancer [17] and pRB is often lost in human malignancies (reviewed in [18, 19] ). In addition, germ-line mutation of either gene results in genetic diseases associated with increased cancer risk. [20, 21] The ability of SARS-CoV-2′s proteins to inhibit both p53 and pRB suggests that SARS-CoV-2 may have an oncogenic potential through a mechanism similar to HPV (see above). In the previous section, we described how some proteins of SARS-CoV-2 may exert oncogenic potential by inhibiting tumor suppressor genes. However, the relationship between a virus and its host is a complex phenomenon, involving several regulators from both the virus and the host. Although the expression of certain proteins may have an oncogenic effect, this does not mean that the viral infection necessarily degenerates into cancer. Well-known oncogenic viruses typically establish long-term relationships with their host through some forms of latency or persistence. Two infamous examples are HPV, which follows the differentiation of keratinocytes as well as herpes simplex virus-which remains latent into neural ganglia. Unlike these oncogenic viruses, most Sars-CoV-2 infections resolve in a limited amount of time. A recent work speculated about the possibility of viral latency in the testis [22] ; however, Pan and colleagues found no evidence of Sars-CoV-2 genome in the semen of convalescent males. [23] While the persistence of Sars-CoV-2 RNA in fecal samples may indicate a potential gastrointestinal reservoir, no direct evidence has been reported. Furthermore, RNA persistence in fecal samples is limited to a period of months which is much shorter than the infections caused by other oncogenic viruses. [24] [25] [26] Other reports of "chronic" SARS-CoV-2 infections or "reactivation" of the virus can be more easily explained by the worsening of low-grade infections rather than by the existence of a viral reservoir. The fact that SARS-CoV-2 does not seem to establish a long-lasting infection argues against its role in cancer onset. If Sars-CoV-2 does not establish a stable relationship with its host, the only possibility for cell transformation relies on the ability of infected cells to escape immune surveillance and the cytopathic effect of the virus. This possibility is particularly attractive considering the link between pRB haploinsufficiency and chromosomal instability. The loss of a single pRB copy induces chromosomal instability [27] and creates an optimal environment for cell transformation, even in the case of an acute and transient event. [28] Although Nsp15-mediated pRB inhibition and genomic instability are likely to self-resolve with virus clearance, the mutations that originated through genomic instability are irreversible. Some of these mutations may have long-term effects and increase susceptibility to cancer even years after the primary infection. However, SARS-CoV-2 infection results in extensive tissue damage and cell death in cultured cells and organoids [29] [30] [31] ; this indicates that the virus tends to kill its host and severely reduces the likelihood for cell transformation. Some lines of evidence suggest that alternative outcomes of viral infection exist. Recent results indicate that the egression strategy of β-coronaviruses does not cause major alterations of the plasma membrane or increases in cell death 18 h post-infection. [32, 33] In addition, SARS-CoV-2 infects but does not replicate in at least some cell types such as monocytes and monocytederived macrophages. [34] The reduced cytotoxicity and the existence of "abortive" viral cycle indicate that cell death may not be the only pos- [35] ). For example, SARS-CoV-1 inhibits pRB phosphorylation through 3a and 7b proteins and inhibits cyclin-cyclindependent kinases activity through the N-protein, inducing a cell cycle arrest. [36] [37] [38] Similarly, the coronavirus infectious bronchitis virus induces a G2/M arrest in cells lacking p53 [39] and SARS-CoV-2 infected cells also arrest in G2/M. [40] Moreover, cells infected by SARS-CoV-2 activate the pro-apoptotic regulator caspase 8 and undergo programmed cell death. [41] Overall, even postulating the cell survival fol- A final piece of evidence arguing against oncogenic potential of SARS-CoV-2 comes from epidemiological data on SARS-CoV-1. Extensive follow-up studies on the long-term symptoms of SARS-CoV-1 have not reported increased cancer incidence thus far [42] [43] [44] arguing against an increased susceptibility of cancer in SARS-CoV-1-infected individuals. It must be noted that only a subset of the 8096 total reported cases of SARS-CoV-1 were analyzed to epidemiologically investigate the long-term effects of the syndrome. In addition, it is plausible that increased incidence of cancer has not yet arisen in the relatively short In previous sections, we speculated about the mechanisms that could link SARS-CoV-2 with cancer and reviewed the evidence for and against such a hypothesis. In this section, we consider how to address any outstanding questions experimentally. Histology is a classic technique to detect early signs of cell transformation in biopsies and autopsies. Thus far, post-mortem histological analysis of SARS-CoV-2 focused on patients that have died from the infection without specifically searching for cancer markers. [45, 46] In the future, it would be interesting to study individuals that have recovered from the disease and died of independent causes at different time after the infection. This analysis should focus primarily on tissues with high trophism for SARS-CoV-2, such as the lungs, as these are the prime suspect location for cancer to potentially develop. In parallel with the histological analysis, important evidence on the oncogenic potential of SARS-CoV-2 could come from epidemiological studies on patients that have recovered from COVID-19 syndrome. The unprecedented detail and quality of epidemiological data collected during the COVID-19 pandemic, along with recent advancement in data analysis, will be invaluable tools for evaluating the long-term effects of the infection. Both histological and epidemiological data could show a correlation between COVID-19 and cancer, but a more direct approach is needed to demonstrate causation. In this context, the cellular effects of the virus on cell physiology remain only partially understood. One should assess the long-term effects of SARS-CoV-2 on cell cultures and organoids using functional assays such as proliferation curves, soft agar assay, and cell cycle profile analysis. A replication-defective or attenuated version of the virus is an important tool to differentiate the viral effects on cell physiology from its cytotoxicity. Although cells and organoids enable investigations of biological phenomena at high resolution, they often fall short in capturing the complexity of an organism. For this reason, it is crucial to assess the long-term effects of mild SARS-CoV-2 infection in laboratory animals. The effects of SARS-CoV-2 infection can be modeled in several animals. [47, 48] The development of mice expressing human angiotensin-converting enzyme 2 (hACE2 -K18-hACE2 mice) receptors [49, 50, 51] and the availability of cancer mouse models [52] makes Mus musculus a promising model to study the effect of SARS-CoV-2 infection and cancer development. It would be interesting to cross p53−/+ [53, 54] with K18-hACE2 mice to investigate the effect of tumor development in SARS-CoV-2-infected mice compared to noninfected mice. The novel coronavirus, SARS-CoV-2, emerged for the first time in China in December 2019. It is characterized by modest mortality (Infection Fatality Rate, circa 0.6) and high infectiousness that can cause a severe respiratory syndrome known as COVID-19. [55] There is a growing body of evidence that indicates long-term effects well after primary SARS-CoV-2 infections have resolved. Among these long-term effects, we have speculated about the oncogenic potential of SARS-CoV-2. pneumonia. [56] [57] [58] [59] Nevertheless, the effects of SARS-CoV-2 infection on a single cell remain unclear and may be dramatically different depending on the cell type and the initial viral load. In conclusion, the role of SARS-CoV-2 in cell transformation is unlikely but cannot be excluded in specific contexts. Additional studies on the long-term effects of the virus are needed to shed light on such possibility. We would like to thank Prof. Patrick Meraldi (University of Geneva, Geneva) and Prof. Jonathon Pines (The Institute for Cancer Research, London) for their support. We are grateful to Matteo Bosso (Institute of Molecular Virology, Ulm University Hospital) for critically reading the manuscript. A.S. is supported by IGE3 grant and L.C. is supported by the SNF fellowship P2GEP3-187775. The authors declare no conflicts of interest. Data sharing not applicable -no new data generated. Short-and potential long-term adverse health outcomes of COVID-19: A rapid review Long-term consequences of COVID-19: Research needs. The Lancet Infectious Diseases Persistent SARS-CoV-2 infection and the risk for cancer Sun exposure and melanoma, certainties and weaknesses of the present knowledge Experimentelle Leukämie bei Hühnern. II. Zeitschrift für Hygiene und Infektionskrankheiten A transmissible avian neoplasm. (Sarcoma of the common fowl A sarcoma involving the jaws in African children Morphological and biological studies on a virus in cultured lymphoblasts from Burkitt's lymphoma Human viral oncogenesis: A cancer hallmarks analysis Structural basis for the inactivation of retinoblastoma tumor suppressor by SV40 large T antigen The human papillomavirus (HPV)-related cancer biology: An overview Review of HPVrelated diseases and cancers The coronavirus endoribonuclease Nsp15 interacts with retinoblastoma tumor suppressor protein P53 down-regulates SARS coronavirus replication and is targeted by the SARS-unique domain and PLpro via E3 ubiquitin ligase RCHY1 Pirh2, a p53-induced ubiquitin-protein ligase, promotes p53 degradation Molecular basis of Pirh2-mediated p53 ubiquitylation Uncovering the role of p53 splice variants in human malignancy: A clinical perspective Cellular mechanisms of tumour suppression by the retinoblastoma gene Hallmarks of cancer: The next generation Li-Fraumeni syndrome: Cancer risk assessment and clinical management The genomic landscape of retinoblastoma: A review The big challenge of SARS-CoV-2 latency: Testes as reservoir No evidence of severe acute respiratory syndrome-coronavirus 2 in semen of males recovering from coronavirus disease 2019 Potential intestinal infection and faecal-oral transmission of SARS-CoV-2 SARS-CoV-2 shedding from asymptomatic patients: Contribution of potential extrapulmonary tissue reservoirs Prolonged viral shedding in feces of pediatric patients with coronavirus disease 2019 Haploinsufficiency of an RB -E2F1 -Condensin II complex leads to aberrant replication and aneuploidy Loss of pRB causes centromere dysfunction and chromosomal instability Pathogenesis of COVID-19 from a cell biology perspective Infection of bat and human intestinal organoids by SARS-CoV-2 Morphogenesis and cytopathic effect of SARS-CoV-2 infection in human airway epithelial cells β-Coronaviruses use lysosomes for egress instead of the biosynthetic secretory pathway Coronavirus biology and replication: Implications for SARS-CoV-2 Monocytes and macrophages, targets of SARS-CoV-2: The clue for Covid-19 immunoparalysis A mini-review on cell cycle regulation of coronavirus infection The nucleocapsid protein of severe acute respiratory syndrome-coronavirus inhibits the activity of cyclin-cyclin-dependent kinase complex and blocks S phase progression in mammalian cells G1 phase cell cycle arrest induced by SARS-CoV 3a protein via the cyclin D3/pRb pathway SARS coronavirus 7a protein blocks cell cycle progression at G0/G1 phase via the cyclin D3/pRb pathway Cell cycle arrest and apoptosis induced by the coronavirus infectious bronchitis virus in the absence of p53 The global phosphorylation landscape of SARS-CoV-2 infection SARS-CoV-2 triggers inflammatory responses and cell death through caspase-8 activation Mental morbidities and chronic fatigue in severe acute respiratory syndrome survivors: Long-term follow-up The long-term impact of severe acute respiratory syndrome on pulmonary function, exercise capacity and health status Long-term bone and lung consequences associated with hospital-acquired severe acute respiratory syndrome: A 15-year follow-up from a prospective cohort study Histopathological findings and viral tropism in UK patients with severe fatal COVID-19: A post-mortem study Early histologic findings of pulmonary SARS-CoV-2 infection detected in a surgical specimen Animal and translational models of SARS-CoV-2 infection and COVID-19 Animal models for COVID-19 A SARS-CoV-2 infection model in mice demonstrates protection by neutralizing antibodies Comparison of transgenic and adenovirus hACE2 mouse models for SARS-CoV-2 infection Rapid generation of a mouse model for Middle East respiratory syndrome Transgenic mouse models in cancer research Spontaneous and carcinogen-induced tumorigenesis in p53-deficient mice A systematic review and meta-analysis of published research data on COVID-19 infectionfatality rates Pulmonary fibrosis and COVID-19: The potential role for antifibrotic therapy COVID-19: The potential treatment of pulmonary fibrosis associated with SARS-CoV-2 infection Pulmonary fibrosis secondary to COVID-19: A call to arms? The Lancet Respiratory Medicine Is the presence of lung injury in COVID-19 an independent risk factor for secondary lung cancer? Medical Hypotheses SARS-CoV-2 infection and cancer