Should We Expect an Increase in the Number of Cancer Cases in People with Long COVID?
Abstract
:1. Introduction
2. Senescence and Viral Infections
3. Chronic Inflammation and Viral Infections
4. Chronic Virus Infection, Residual Virus Proteins and Cancer Risk
5. The Oncogenic Potential of SARS-CoV-2
6. Immunosuppression
7. Autophagy
8. Is There Something We Have Overlooked?
9. Limitations and Perspectives
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Plummer, M.; de Martel, C.; Vignat, J.; Ferlay, J.; Bray, F.; Franceschi, S. Global burden of cancers attributable to infections in 2012: A synthetic analysis. Lancet Glob. Health 2016, 4, e609–e616. [Google Scholar] [CrossRef] [Green Version]
- Epstein, M.A.; Achong, B.G.; Barr, Y.M. Virus particles in cultured lymphoblasts from Burkitt’s lymphoma. Lancet 1964, 1, 702–703. [Google Scholar] [CrossRef] [PubMed]
- Kellogg, C.; Kouznetsova, V.L.; Tsigelny, I.F. Implications of viral infection in cancer development. Biochim. Biophys. Acta Rev. Cancer 2021, 1876, 188622. [Google Scholar] [CrossRef] [PubMed]
- Chen, C.J.; You, S.L.; Hsu, W.L.; Yang, H.I.; Lee, M.H.; Chen, H.C.; Chen, Y.Y.; Liu, J.; Hu, H.H.; Lin, Y.J.; et al. Epidemiology of virus Infection and human cancer. Recent Results Cancer Res. 2021, 217, 13–45. [Google Scholar] [CrossRef]
- Mesri, E.A.; Feitelson, M.A.; Munger, K. Human viral oncogenesis: A cancer hallmarks analysis. Cell Host Microbe 2014, 15, 266–282. [Google Scholar] [CrossRef] [Green Version]
- Johns Hopkins. Coronavirus Resourse Center. Available online: https://coronavirus.jhu.edu/map.html (accessed on 29 August 2022).
- Wang, D.; Hu, B.; Hu, C.; Zhu, F.; Liu, X.; Zhang, J.; Wang, B.; Xiang, H.; Cheng, Z.; Xiong, Y.; et al. Clinical characteristics of 138 hospitalized patients with 2019 novel coronavirus-infected pneumonia in Wuhan, China. JAMA 2020, 323, 1061–1069. [Google Scholar] [CrossRef]
- Long, B.; Brady, W.J.; Koyfman, A.; Gottlieb, M. Cardiovascular complications in COVID-19. Am. J. Emerg. Med. 2020, 38, 1504–1507. [Google Scholar] [CrossRef]
- Gupta, A.; Madhavan, M.V.; Sehgal, K.; Nair, N.; Mahajan, S.; Sehrawat, T.S.; Bikdeli, B.; Ahluwalia, N.; Ausiello, J.C.; Wan, E.Y.; et al. Extrapulmonary manifestations of COVID-19. Nat. Med. 2020, 26, 1017–1032. [Google Scholar] [CrossRef] [PubMed]
- Xie, Y.; Al-Aly, Z. Risks and burdens of incident diabetes in long COVID: A cohort study. Lancet Diabetes Endocrinol. 2022, 10, 311–321. [Google Scholar] [CrossRef] [PubMed]
- Montori, V.M. Patients surviving COVID-19 had increased risk for incident diabetes vs. persons without COVID-19. Ann. Intern. Med. 2022, 175, JC93. [Google Scholar] [CrossRef]
- Groff, D.; Sun, A.; Ssentongo, A.E.; Ba, D.M.; Parsons, N.; Poudel, G.R.; Lekoubou, A.; Oh, J.S.; Ericson, J.E.; Ssentongo, P.; et al. Short-term and long-term rates of postacute sequelae of SARS-CoV-2 infection: A systematic review. JAMA Netw. Open 2021, 4, e2128568. [Google Scholar] [CrossRef]
- Taquet, M.; Dercon, Q.; Luciano, S.; Geddes, J.R.; Husain, M.; Harrison, P.J. Incidence, co-occurrence, and evolution of long-COVID features: A 6-month retrospective cohort study of 273,618 survivors of COVID-19. PLoS Med. 2021, 18, e1003773. [Google Scholar] [CrossRef]
- Lopez-Leon, S.; Wegman-Ostrosky, T.; Perelman, C.; Sepulveda, R.; Rebolledo, P.A.; Cuapio, A.; Villapol, S. More than 50 long-term effects of COVID-19: A systematic review and meta-analysis. Sci. Rep. 2021, 11, 16144. [Google Scholar] [CrossRef]
- Merad, M.; Blish, C.A.; Sallusto, F.; Iwasaki, A. The immunology and immunopathology of COVID-19. Science 2022, 375, 1122–1127. [Google Scholar] [CrossRef]
- Jacobs, J.J.L. Persistent SARS-2 infections contribute to long COVID-19. Med. Hypotheses 2021, 149, 110538. [Google Scholar] [CrossRef]
- Acosta-Ampudia, Y.; Monsalve, D.M.; Rojas, M.; Rodriguez, Y.; Zapata, E.; Ramirez-Santana, C.; Anaya, J.M. Persistent autoimmune activation and proinflammatory state in Post-Coronavirus Disease 2019 Syndrome. J. Infect. Dis. 2022, 225, 2155–2162. [Google Scholar] [CrossRef]
- Galeotti, C.; Bayry, J. Autoimmune and inflammatory diseases following COVID-19. Nat. Rev. Rheumatol. 2020, 16, 413–414. [Google Scholar] [CrossRef]
- Zheng, K.I.; Feng, G.; Liu, W.Y.; Targher, G.; Byrne, C.D.; Zheng, M.H. Extrapulmonary complications of COVID-19: A multisystem disease? J. Med. Virol. 2021, 93, 323–335. [Google Scholar] [CrossRef]
- Thakur, V.; Ratho, R.K.; Kumar, P.; Bhatia, S.K.; Bora, I.; Mohi, G.K.; Saxena, S.K.; Devi, M.; Yadav, D.; Mehariya, S. Multi-Organ involvement in COVID-19: Beyond pulmonary manifestations. J. Clin. Med. 2021, 10, 446. [Google Scholar] [CrossRef]
- Letko, M.; Marzi, A.; Munster, V. Functional assessment of cell entry and receptor usage for SARS-CoV-2 and other lineage B betacoronaviruses. Nat. Microbiol. 2020, 5, 562–569. [Google Scholar] [CrossRef] [Green Version]
- Yan, R.; Zhang, Y.; Li, Y.; Xia, L.; Guo, Y.; Zhou, Q. Structural basis for the recognition of SARS-CoV-2 by full-length human ACE2. Science 2020, 367, 1444–1448. [Google Scholar] [CrossRef] [Green Version]
- Hikmet, F.; Mear, L.; Edvinsson, A.; Micke, P.; Uhlen, M.; Lindskog, C. The protein expression profile of ACE2 in human tissues. Mol. Syst. Biol. 2020, 16, e9610. [Google Scholar] [CrossRef]
- Dugue, P.A.; Bassett, J.K.; Wong, E.M.; Joo, J.E.; Li, S.; Yu, C.; Schmidt, D.F.; Makalic, E.; Doo, N.W.; Buchanan, D.D.; et al. Biological aging measures based on blood DNA methylation and risk of cancer: A prospective study. JNCI Cancer Spectr. 2021, 5, pkaa109. [Google Scholar] [CrossRef]
- Gorgoulis, V.; Adams, P.D.; Alimonti, A.; Bennett, D.C.; Bischof, O.; Bishop, C.; Campisi, J.; Collado, M.; Evangelou, K.; Ferbeyre, G.; et al. Cellular senescence: Defining a path forward. Cell 2019, 179, 813–827. [Google Scholar] [CrossRef]
- Campisi, J.; Andersen, J.K.; Kapahi, P.; Melov, S. Cellular senescence: A link between cancer and age-related degenerative disease? Semin. Cancer Biol. 2011, 21, 354–359. [Google Scholar] [CrossRef] [Green Version]
- Coppe, J.P.; Patil, C.K.; Rodier, F.; Sun, Y.; Munoz, D.P.; Goldstein, J.; Nelson, P.S.; Desprez, P.Y.; Campisi, J. Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor. PLoS Biol. 2008, 6, 2853–2868. [Google Scholar] [CrossRef] [PubMed]
- Takasugi, M.; Okada, R.; Takahashi, A.; Virya Chen, D.; Watanabe, S.; Hara, E. Small extracellular vesicles secreted from senescent cells promote cancer cell proliferation through EphA2. Nat. Commun. 2017, 8, 15729. [Google Scholar] [CrossRef] [Green Version]
- Fulop, T.; Dupuis, G.; Baehl, S.; Le Page, A.; Bourgade, K.; Frost, E.; Witkowski, J.M.; Pawelec, G.; Larbi, A.; Cunnane, S. From inflamm-aging to immune-paralysis: A slippery slope during aging for immune-adaptation. Biogerontology 2016, 17, 147–157. [Google Scholar] [CrossRef]
- Salminen, A. Immunosuppressive network promotes immunosenescence associated with aging and chronic inflammatory conditions. J. Mol. Med. 2021, 99, 1553–1569. [Google Scholar] [CrossRef]
- Baz-Martinez, M.; Da Silva-Alvarez, S.; Rodriguez, E.; Guerra, J.; El Motiam, A.; Vidal, A.; Garcia-Caballero, T.; Gonzalez-Barcia, M.; Sanchez, L.; Munoz-Fontela, C.; et al. Cell senescence is an antiviral defense mechanism. Sci. Rep. 2016, 6, 37007. [Google Scholar] [CrossRef] [Green Version]
- Seoane, R.; Vidal, S.; Bouzaher, Y.H.; El Motiam, A.; Rivas, C. The interaction of viruses with the cellular senescence response. Biology 2020, 9, 455. [Google Scholar] [CrossRef]
- Reddel, R.R. Senescence: An antiviral defense that is tumor suppressive? Carcinogenesis 2010, 31, 19–26. [Google Scholar] [CrossRef] [PubMed]
- Camell, C.D.; Yousefzadeh, M.J.; Zhu, Y.; Prata, L.; Huggins, M.A.; Pierson, M.; Zhang, L.; O’Kelly, R.D.; Pirtskhalava, T.; Xun, P.; et al. Senolytics reduce coronavirus-related mortality in old mice. Science 2021, 373. [Google Scholar] [CrossRef]
- Tripathi, U.; Nchioua, R.; Prata, L.; Zhu, Y.; Gerdes, E.O.W.; Giorgadze, N.; Pirtskhalava, T.; Parker, E.; Xue, A.; Espindola-Netto, J.M.; et al. SARS-CoV-2 causes senescence in human cells and exacerbates the senescence-associated secretory phenotype through TLR-3. Aging 2021, 13, 21838–21854. [Google Scholar] [CrossRef]
- Evangelou, K.; Veroutis, D.; Paschalaki, K.; Foukas, P.G.; Lagopati, N.; Dimitriou, M.; Papaspyropoulos, A.; Konda, B.; Hazapis, O.; Polyzou, A.; et al. Pulmonary infection by SARS-CoV-2 induces senescence accompanied by an inflammatory phenotype in severe COVID-19: Possible implications for viral mutagenesis. Eur. Respir. J. 2022, 60, 2102951. [Google Scholar] [CrossRef]
- Balnis, J.; Madrid, A.; Hogan, K.J.; Drake, L.A.; Adhikari, A.; Vancavage, R.; Singer, H.A.; Alisch, R.S.; Jaitovich, A. Persistent blood DNA methylation changes one year after SARS-CoV-2 infection. Clin. Epigenetics 2022, 14, 94. [Google Scholar] [CrossRef]
- Mongelli, A.; Barbi, V.; Gottardi Zamperla, M.; Atlante, S.; Forleo, L.; Nesta, M.; Massetti, M.; Pontecorvi, A.; Nanni, S.; Farsetti, A.; et al. Evidence for biological age acceleration and telomere shortening in COVID-19 survivors. Int. J. Mol. Sci. 2021, 22, 6151. [Google Scholar] [CrossRef]
- Cao, X.; Li, W.; Wang, T.; Ran, D.; Davalos, V.; Planas-Serra, L.; Pujol, A.; Esteller, M.; Wang, X.; Yu, H. Accelerated biological aging in COVID-19 patients. Nat. Commun. 2022, 13, 2135. [Google Scholar] [CrossRef]
- Victor, J.; Deutsch, J.; Whitaker, A.; Lamkin, E.N.; March, A.; Zhou, P.; Botten, J.W.; Chatterjee, N. SARS-CoV-2 triggers DNA damage response in Vero E6 cells. Biochem. Biophys. Res. Commun. 2021, 579, 141–145. [Google Scholar] [CrossRef]
- Franzen, J.; Nuchtern, S.; Tharmapalan, V.; Vieri, M.; Nikolic, M.; Han, Y.; Balfanz, P.; Marx, N.; Dreher, M.; Brummendorf, T.H.; et al. Epigenetic clocks are not accelerated in COVID-19 patients. Int. J. Mol. Sci. 2021, 22, 9306. [Google Scholar] [CrossRef]
- Karin, M. Nuclear factor-kappaB in cancer development and progression. Nature 2006, 441, 431–436. [Google Scholar] [CrossRef] [PubMed]
- Woller, N.; Kuhnel, F. Virus infection, inflammation and prevention of cancer. Recent Results Cancer Res. 2014, 193, 33–58. [Google Scholar] [CrossRef]
- Sato, Y.; Takahashi, S.; Kinouchi, Y.; Shiraki, M.; Endo, K.; Matsumura, Y.; Kakuta, Y.; Tosa, M.; Motida, A.; Abe, H.; et al. IL-10 deficiency leads to somatic mutations in a model of IBD. Carcinogenesis 2006, 27, 1068–1073. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Doykov, I.; Hallqvist, J.; Gilmour, K.C.; Grandjean, L.; Mills, K.; Heywood, W.E. ‘The long tail of Covid-19’—The detection of a prolonged inflammatory response after a SARS-CoV-2 infection in asymptomatic and mildly affected patients. F1000Research 2020, 9, 1349. [Google Scholar] [CrossRef]
- Sieminska, I.; Weglarczyk, K.; Surmiak, M.; Kurowska-Baran, D.; Sanak, M.; Siedlar, M.; Baran, J. Mild and asymptomatic COVID-19 convalescents present long-term endotype of immunosuppression associated with neutrophil subsets possessing regulatory functions. Front. Immunol. 2021, 12, 748097. [Google Scholar] [CrossRef]
- Queiroz, M.A.F.; Neves, P.; Lima, S.S.; Lopes, J.D.C.; Torres, M.; Vallinoto, I.; Bichara, C.D.A.; Dos Santos, E.F.; de Brito, M.; da Silva, A.L.S.; et al. Cytokine profiles associated with acute COVID-19 and long COVID-19 syndrome. Front. Cell. Infect. Microbiol. 2022, 12, 922422. [Google Scholar] [CrossRef]
- Johnson, D.E.; O’Keefe, R.A.; Grandis, J.R. Targeting the IL-6/JAK/STAT3 signalling axis in cancer. Nat. Rev. Clin. Oncol. 2018, 15, 234–248. [Google Scholar] [CrossRef]
- Rohrhofer, J.; Graninger, M.; Lettenmaier, L.; Schweighardt, J.; Gentile, S.A.; Koidl, L.; Ret, D.; Stingl, M.; Puchhammer-Stockl, E.; Untersmayr, E. Association between Epstein-Barr-Virus reactivation and development of Long-COVID fatigue. Allergy 2022, 78, 297–299. [Google Scholar] [CrossRef]
- Gatto, I.; Biagioni, E.; Coloretti, I.; Farinelli, C.; Avoni, C.; Caciagli, V.; Busani, S.; Sarti, M.; Pecorari, M.; Gennari, W.; et al. Cytomegalovirus blood reactivation in COVID-19 critically ill patients: Risk factors and impact on mortality. Intensive Care Med. 2022, 48, 706–713. [Google Scholar] [CrossRef]
- Akira, S.; Uematsu, S.; Takeuchi, O. Pathogen recognition and innate immunity. Cell 2006, 124, 783–801. [Google Scholar] [CrossRef] [Green Version]
- Nelemans, T.; Kikkert, M. Viral Innate Immune Evasion and the Pathogenesis of Emerging RNA Virus Infections. Viruses 2019, 11, 961. [Google Scholar] [CrossRef] [Green Version]
- Coutard, B.; Valle, C.; de Lamballerie, X.; Canard, B.; Seidah, N.G.; Decroly, E. The spike glycoprotein of the new coronavirus 2019-nCoV contains a furin-like cleavage site absent in CoV of the same clade. Antivir. Res. 2020, 176, 104742. [Google Scholar] [CrossRef]
- Swanson, K.V.; Deng, M.; Ting, J.P. The NLRP3 inflammasome: Molecular activation and regulation to therapeutics. Nat. Rev. Immunol. 2019, 19, 477–489. [Google Scholar] [CrossRef]
- Zhao, C.; Zhao, W. NLRP3 Inflammasome-A Key Player in Antiviral Responses. Front. Immunol. 2020, 11, 211. [Google Scholar] [CrossRef] [Green Version]
- Albornoz, E.A.; Amarilla, A.A.; Modhiran, N.; Parker, S.; Li, X.X.; Wijesundara, D.K.; Aguado, J.; Pliego Zamora, A.; McMillan, C.L.D.; Liang, B.; et al. SARS-CoV-2 drives NLRP3 inflammasome activation in human microglia through spike protein. Mol. Psychiatry 2022. [Google Scholar] [CrossRef]
- Franceschi, C.; Bonafe, M.; Valensin, S.; Olivieri, F.; De Luca, M.; Ottaviani, E.; De Benedictis, G. Inflamm-aging. An evolutionary perspective on immunosenescence. Ann. N. Y. Acad. Sci. 2000, 908, 244–254. [Google Scholar] [CrossRef]
- Ferrucci, L.; Fabbri, E. Inflammageing: Chronic inflammation in ageing, cardiovascular disease, and frailty. Nat. Rev. Cardiol. 2018, 15, 505–522. [Google Scholar] [CrossRef]
- Randall, R.E.; Griffin, D.E. Within host RNA virus persistence: Mechanisms and consequences. Curr. Opin. Virol. 2017, 23, 35–42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Farazi, P.A.; DePinho, R.A. Hepatocellular carcinoma pathogenesis: From genes to environment. Nat. Rev. Cancer 2006, 6, 674–687. [Google Scholar] [CrossRef]
- Isaguliants, M.; Bayurova, E.; Avdoshina, D.; Kondrashova, A.; Chiodi, F.; Palefsky, J.M. Oncogenic Effects of HIV-1 Proteins, Mechanisms Behind. Cancers 2021, 13, 305. [Google Scholar] [CrossRef]
- Cheung, C.C.L.; Goh, D.; Lim, X.; Tien, T.Z.; Lim, J.C.T.; Lee, J.N.; Tan, B.; Tay, Z.E.A.; Wan, W.Y.; Chen, E.X.; et al. Residual SARS-CoV-2 viral antigens detected in GI and hepatic tissues from five recovered patients with COVID-19. Gut 2022, 71, 226–229. [Google Scholar] [CrossRef]
- Natarajan, A.; Zlitni, S.; Brooks, E.F.; Vance, S.E.; Dahlen, A.; Hedlin, H.; Park, R.M.; Han, A.; Schmidtke, D.T.; Verma, R.; et al. Gastrointestinal symptoms and fecal shedding of SARS-CoV-2 RNA suggest prolonged gastrointestinal infection. Med 2022, 3, 371–387.e379. [Google Scholar] [CrossRef]
- Zuo, T.; Wu, X.; Wen, W.; Lan, P. Gut microbiome alterations in COVID-19. Genom. Proteom. Bioinform. 2021, 19, 679–688. [Google Scholar] [CrossRef]
- Verrienti, P.; Cito, G.; Di Maida, F.; Tellini, R.; Cocci, A.; Minervini, A.; Natali, A. The impact of COVID-19 on the male genital tract: A qualitative literature review of sexual transmission and fertility implications. Clin. Exp. Reprod. Med. 2022, 49, 9–15. [Google Scholar] [CrossRef]
- Kresch, E.; Achua, J.; Saltzman, R.; Khodamoradi, K.; Arora, H.; Ibrahim, E.; Kryvenko, O.N.; Almeida, V.W.; Firdaus, F.; Hare, J.M.; et al. COVID-19 endothelial dysfunction can cause erectile dysfunction: Histopathological, immunohistochemical, and ultrastructural study of the human penis. World J. Mens. Health 2021, 39, 466–469. [Google Scholar] [CrossRef]
- Pan, F.; Xiao, X.; Guo, J.; Song, Y.; Li, H.; Patel, D.P.; Spivak, A.M.; Alukal, J.P.; Zhang, X.; Xiong, C.; et al. No evidence of severe acute respiratory syndrome-coronavirus 2 in semen of males recovering from coronavirus disease 2019. Fertil. Steril. 2020, 113, 1135–1139. [Google Scholar] [CrossRef]
- Zollner, A.; Koch, R.; Jukic, A.; Pfister, A.; Meyer, M.; Rossler, A.; Kimpel, J.; Adolph, T.E.; Tilg, H. Postacute COVID-19 is Characterized by Gut Viral Antigen Persistence in Inflammatory Bowel Diseases. Gastroenterology 2022, 163, 495–506.e498. [Google Scholar] [CrossRef]
- Goh, D.; Lim, J.C.T.; Fernaindez, S.B.; Joseph, C.R.; Edwards, S.G.; Neo, Z.W.; Lee, J.N.; Caballero, S.G.; Lau, M.C.; Yeong, J.P.S. Case report: Persistence of residual antigen and RNA of the SARS-CoV-2 virus in tissues of two patients with long COVID. Front. Immunol. 2022, 13, 939989. [Google Scholar] [CrossRef]
- Bussani, R.; Zentilin, L.; Correa, R.; Colliva, A.; Silvestri, F.; Zacchigna, S.; Collesi, C.; Giacca, M. Persistent SARS-CoV-2 infection in patients seemingly recovered from COVID-19. J. Pathol. 2023, 259, 254–263. [Google Scholar] [CrossRef]
- Peluso, M.J.; Anglin, K.; Durstenfeld, M.S.; Martin, J.N.; Kelly, J.D.; Hsue, P.Y.; Henrich, T.J.; Deeks, S.G. Effect of oral nirmatrelvir on Long COVID symptoms: 4 cases and rationale for systematic studies. Pathog. Immun. 2022, 7, 95–103. [Google Scholar] [CrossRef]
- Owen, D.R.; Allerton, C.M.N.; Anderson, A.S.; Aschenbrenner, L.; Avery, M.; Berritt, S.; Boras, B.; Cardin, R.D.; Carlo, A.; Coffman, K.J.; et al. An oral SARS-CoV-2 M(pro) inhibitor clinical candidate for the treatment of COVID-19. Science 2021, 374, 1586–1593. [Google Scholar] [CrossRef]
- Briggs, E.; Ward, W.; Rey, S.; Law, D.; Nelson, K.; Bois, M.; Ostrov, N.; Lee, H.H.; Laurent, J.M.; Mita, P. Assessment of potential SARS-CoV-2 virus integration into human genome reveals no significant impact on RT-qPCR COVID-19 testing. Proc. Natl. Acad. Sci. USA 2021, 118, e2113065118. [Google Scholar] [CrossRef]
- Parry, R.; Gifford, R.J.; Lytras, S.; Ray, S.C.; Coin, L.J.M. No evidence of SARS-CoV-2 reverse transcription and integration as the origin of chimeric transcripts in patient tissues. Proc. Natl. Acad. Sci. USA 2021, 118, e2109066118. [Google Scholar] [CrossRef]
- Smits, N.; Rasmussen, J.; Bodea, G.O.; Amarilla, A.A.; Gerdes, P.; Sanchez-Luque, F.J.; Ajjikuttira, P.; Modhiran, N.; Liang, B.; Faivre, J.; et al. No evidence of human genome integration of SARS-CoV-2 found by long-read DNA sequencing. Cell Rep. 2021, 36, 109530. [Google Scholar] [CrossRef]
- Zhang, L.; Richards, A.; Barrasa, M.I.; Hughes, S.H.; Young, R.A.; Jaenisch, R. Reverse-transcribed SARS-CoV-2 RNA can integrate into the genome of cultured human cells and can be expressed in patient-derived tissues. Proc. Natl. Acad. Sci. USA 2021, 118, e2105968118. [Google Scholar] [CrossRef]
- Craddock, V.; Mahajan, A.; Krishnamachary, B.; Spikes, L.; Chalise, P.; Dhillon, N.K. Persistent presence of Spike protein and viral RNA in the circulation of individuals with post-acute sequelae of COVID-19. medRxiv 2022. [Google Scholar]
- Swank, Z.; Senussi, Y.; Manickas-Hill, Z.; Yu, X.G.; Li, J.Z.; Alter, G.; Walt, D.R. Persistent circulating SARS-CoV-2 spike is associated with post-acute COVID-19 sequelae. Clin. Infect. Dis. 2022, 76, e487–e490. [Google Scholar] [CrossRef]
- Kanduc, D. From anti-severe acute respiratory syndrome Coronavirus 2 immune response to cancer onset via molecular mimicry and cross-reactivity. Glob. Med. Genet. 2021, 8, 176–182. [Google Scholar] [CrossRef]
- Boulias, K.; Lieberman, J.; Greer, E.L. An epigenetic clock measures accelerated aging in treated HIV infection. Mol. Cell 2016, 62, 153–155. [Google Scholar] [CrossRef] [Green Version]
- Stingi, A.; Cirillo, L. SARS-CoV-2 infection and cancer: Evidence for and against a role of SARS-CoV-2 in cancer onset. Bioessays 2021, 43, e2000289. [Google Scholar] [CrossRef]
- Cardozo, C.M.; Hainaut, P. Viral strategies for circumventing p53: The case of severe acute respiratory syndrome coronavirus. Curr. Opin. Oncol. 2021, 33, 149–158. [Google Scholar] [CrossRef] [PubMed]
- Gomez-Carballa, A.; Martinon-Torres, F.; Salas, A. Is SARS-CoV-2 an oncogenic virus? J. Infect. 2022, 85, 573–607. [Google Scholar] [CrossRef] [PubMed]
- Burkhart, D.L.; Sage, J. Cellular mechanisms of tumour suppression by the retinoblastoma gene. Nat. Rev. Cancer 2008, 8, 671–682. [Google Scholar] [CrossRef]
- Saini, G.; Aneja, R. Cancer as a prospective sequela of long COVID-19. Bioessays 2021, 43, e2000331. [Google Scholar] [CrossRef]
- Bhardwaj, K.; Liu, P.; Leibowitz, J.L.; Kao, C.C. The coronavirus endoribonuclease Nsp15 interacts with retinoblastoma tumor suppressor protein. J. Virol. 2012, 86, 4294–4304. [Google Scholar] [CrossRef] [Green Version]
- Manning, A.L.; Longworth, M.S.; Dyson, N.J. Loss of pRB causes centromere dysfunction and chromosomal instability. Genes Dev. 2010, 24, 1364–1376. [Google Scholar] [CrossRef] [Green Version]
- Surget, S.; Khoury, M.P.; Bourdon, J.C. Uncovering the role of p53 splice variants in human malignancy: A clinical perspective. OncoTargets Ther. 2013, 7, 57–68. [Google Scholar] [CrossRef] [Green Version]
- Ebrahimi Sadrabadi, A.; Bereimipour, A.; Jalili, A.; Gholipurmalekabadi, M.; Farhadihosseinabadi, B.; Seifalian, A.M. The risk of pancreatic adenocarcinoma following SARS-CoV family infection. Sci. Rep. 2021, 11, 12948. [Google Scholar] [CrossRef]
- Zhao, X.; Wu, X.; Xiao, J.; Zhang, L.; Hao, Y.; Xiao, C.; Zhang, B.; Li, J.; Jiang, X. Are female-specific cancers long-term sequelae of COVID-19? Evidence from a large-scale genome-wide cross-trait analysis. medRxiv 2022. [Google Scholar] [CrossRef]
- Shen, Q.; Wang, J.; Zhao, L. To investigate the internal association between SARS-CoV-2 infections and cancer through bioinformatics. Math. Biosci. Eng. 2022, 19, 11172–11194. [Google Scholar] [CrossRef]
- Ahmadi, E.; Changaei, M.; Teymouri, A.; Alipour, B.; Izad, M. Cancer Related-Genes Enriched in Peripheral Blood Mononuclear Cells (PBMCs) of COVID-19 Patients. a Bioinformatics Study; Research Square: Durham, NC, USA, 2022. [Google Scholar] [CrossRef]
- Casey, S.C.; Li, Y.; Felsher, D.W. An essential role for the immune system in the mechanism of tumor regression following targeted oncogene inactivation. Immunol. Res. 2014, 58, 282–291. [Google Scholar] [CrossRef] [Green Version]
- Ghosh, S.; Dellibovi-Ragheb, T.A.; Kerviel, A.; Pak, E.; Qiu, Q.; Fisher, M.; Takvorian, P.M.; Bleck, C.; Hsu, V.W.; Fehr, A.R.; et al. β-Coronaviruses use lysosomes for egress Instead of the biosynthetic secretory pathway. Cell 2020, 183, 1520–1535.e14. [Google Scholar] [CrossRef]
- Boumaza, A.; Gay, L.; Mezouar, S.; Bestion, E.; Diallo, A.B.; Michel, M.; Desnues, B.; Raoult, D.; La Scola, B.; Halfon, P.; et al. Monocytes and macrophages, targets of severe acute respiratory syndrome Coronavirus 2: The clue for Coronavirus Disease 2019 immunoparalysis. J. Infect. Dis. 2021, 224, 395–406. [Google Scholar] [CrossRef]
- Phetsouphanh, C.; Darley, D.R.; Wilson, D.B.; Howe, A.; Munier, C.M.L.; Patel, S.K.; Juno, J.A.; Burrell, L.M.; Kent, S.J.; Dore, G.J.; et al. Immunological dysfunction persists for 8 months following initial mild-to-moderate SARS-CoV-2 infection. Nat. Immunol. 2022, 23, 210–216. [Google Scholar] [CrossRef]
- Lee, S.; Yu, Y.; Trimpert, J.; Benthani, F.; Mairhofer, M.; Richter-Pechanska, P.; Wyler, E.; Belenki, D.; Kaltenbrunner, S.; Pammer, M.; et al. Virus-induced senescence is a driver and therapeutic target in COVID-19. Nature 2021, 599, 283–289. [Google Scholar] [CrossRef]
- Blanco-Melo, D.; Nilsson-Payant, B.E.; Liu, W.C.; Uhl, S.; Hoagland, D.; Moller, R.; Jordan, T.X.; Oishi, K.; Panis, M.; Sachs, D.; et al. Imbalanced host response to SARS-CoV-2 drives development of COVID-19. Cell 2020, 181, 1036–1045.e1039. [Google Scholar] [CrossRef]
- Li, Y.; Renner, D.M.; Comar, C.E.; Whelan, J.N.; Reyes, H.M.; Cardenas-Diaz, F.L.; Truitt, R.; Tan, L.H.; Dong, B.; Alysandratos, K.D.; et al. SARS-CoV-2 induces double-stranded RNA-mediated innate immune responses in respiratory epithelial-derived cells and cardiomyocytes. Proc. Natl. Acad. Sci. USA 2021, 118, e2022643118. [Google Scholar] [CrossRef] [PubMed]
- Kee, J.; Thudium, S.; Renner, D.M.; Glastad, K.; Palozola, K.; Zhang, Z.; Li, Y.; Lan, Y.; Cesare, J.; Poleshko, A.; et al. SARS-CoV-2 disrupts host epigenetic regulation via histone mimicry. Nature 2022, 610, 381–388. [Google Scholar] [CrossRef]
- Tarakhovsky, A.; Prinjha, R.K. Drawing on disorder: How viruses use histone mimicry to their advantage. J. Exp. Med. 2018, 215, 1777–1787. [Google Scholar] [CrossRef] [Green Version]
- van der Donk, L.E.H.; Eder, J.; van Hamme, J.L.; Brouwer, P.J.M.; Brinkkemper, M.; van Nuenen, A.C.; van Gils, M.J.; Sanders, R.W.; Kootstra, N.A.; Bermejo-Jambrin, M.; et al. SARS-CoV-2 infection activates dendritic cells via cytosolic receptors rather than extracellular TLRs. Eur. J. Immunol. 2002, 52, 646–655. [Google Scholar] [CrossRef]
- van der Sluis, R.M.; Cham, L.B.; Gris-Oliver, A.; Gammelgaard, K.R.; Pedersen, J.G.; Idorn, M.; Ahmadov, U.; Hernandez, S.S.; Cémalovic, E.; Godsk, S.H.; et al. TLR2 and TLR7 mediate distinct immunopathological and antiviral plasmacytoid dendritic cell responses to SARS-CoV-2 infection. EMBO J. 2022, 41, e109622. [Google Scholar] [CrossRef]
- Bi, J. NK cell dysfunction in patients with COVID-19. Cell Mol. Immunol. 2022, 19, 127–129. [Google Scholar] [CrossRef]
- Bernal, E.; Gimeno, L.; Alcaraz, M.J.; Quadeer, A.A.; Moreno, M.; Martínez-Sánchez, M.V.; Campillo, J.A.; Gomez, J.M.; Pelaez, A.; García, E.; et al. Activating Killer-Cell Immunoglobulin-Like Receptors Are Associated With the Severity of Coronavirus Disease 2019. J. Infect. Dis. 2021, 224, 229–240. [Google Scholar] [CrossRef]
- Hajeer, A.; Jawdat, D.; Massadeh, S.; Aljawini, N.; Abedalthagafi, M.S.; Arabi, Y.M.; Alaamery, M. Association of KIR gene polymorphisms with COVID-19 disease. Clin. Immunol. 2022, 234, 108911. [Google Scholar] [CrossRef] [PubMed]
- De Winter, F.H.R.; Hotterbeekx, A.; Huizing, M.T.; Konnova, A.; Fransen, E.; Jongers, B.; Jairam, R.K.; Van Averbeke, V.; Moons, P.; Roelant, E.; et al. Blood Cytokine Analysis Suggests That SARS-CoV-2 Infection Results in a Sustained Tumour Promoting Environment in Cancer Patients. Cancers 2021, 13, 5718. [Google Scholar] [CrossRef]
- Habibzadeh, P.; Dastsooz, H.; Eshraghi, M.; Los, M.J.; Klionsky, D.J.; Ghavami, S. Autophagy: The potential link between SARS-CoV-2 and cancer. Cancers 2021, 13, 5721. [Google Scholar] [CrossRef]
- Challenor, S.; Tucker, D. SARS-CoV-2-induced remission of Hodgkin lymphoma. Br. J. Haematol. 2021, 192, 415. [Google Scholar] [CrossRef]
- Pasin, F.; Mascalchi Calveri, M.; Calabrese, A.; Pizzarelli, G.; Bongiovanni, I.; Andreoli, M.; Cattaneo, C.; Rignanese, G. Oncolytic effect of SARS-CoV2 in a patient with NK lymphoma. Acta Biomed. 2020, 91. ahead of print. [Google Scholar] [CrossRef]
- Sollini, M.; Gelardi, F.; Carlo-Stella, C.; Chiti, A. Complete remission of follicular lymphoma after SARS-CoV-2 infection: From the “flare phenomenon” to the “abscopal effect”. Eur. J. Nucl. Med. Mol. Imaging 2021, 48, 2652–2654. [Google Scholar] [CrossRef]
- Ottaiano, A.; Scala, S.; D’Alterio, C.; Trotta, A.; Bello, A.; Rea, G.; Picone, C.; Santorsola, M.; Petrillo, A.; Nasti, G. Unexpected tumor reduction in metastatic colorectal cancer patients during SARS-Cov-2 infection. Ther. Adv. Med. Oncol. 2021, 13, 17588359211011455. [Google Scholar] [CrossRef]
- Donia, A.; Shahid, R.; Nawaz, M.; Yaqub, T.; Bokhari, H. Can we develop oncolytic SARS-CoV-2 to specifically target cancer cells? Ther. Adv. Med. Oncol. 2021, 13, 17588359211061988. [Google Scholar] [CrossRef] [PubMed]
- Buckner, T.W.; Dunphy, C.; Fedoriw, Y.D.; van Deventer, H.W.; Foster, M.C.; Richards, K.L.; Park, S.I. Complete spontaneous remission of diffuse large B-cell lymphoma of the maxillary sinus after concurrent infections. Clin. Lymphoma Myeloma Leuk. 2012, 12, 455–458. [Google Scholar] [CrossRef]
- Lam, M.H.; Wing, Y.K.; Yu, M.W.; Leung, C.M.; Ma, R.C.; Kong, A.P.; So, W.Y.; Fong, S.Y.; Lam, S.P. Mental morbidities and chronic fatigue in severe acute respiratory syndrome survivors: Long-term follow-up. Arch. Intern. Med. 2009, 169, 2142–2147. [Google Scholar] [CrossRef] [Green Version]
- Zhang, P.; Li, J.; Liu, H.; Han, N.; Ju, J.; Kou, Y.; Chen, L.; Jiang, M.; Pan, F.; Zheng, Y.; et al. Long-term bone and lung consequences associated with hospital-acquired severe acute respiratory syndrome: A 15-year follow-up from a prospective cohort study. Bone Res. 2020, 8, 8. [Google Scholar] [CrossRef] [Green Version]
- Allen, M.B. COVID-19, cancer post-pandemic risk, and the radiation oncology physicist. J. Appl. Clin. Med. Phys. 2022, 23, e13628. [Google Scholar] [CrossRef]
- Maringe, C.; Spicer, J.; Morris, M.; Purushotham, A.; Nolte, E.; Sullivan, R.; Rachet, B.; Aggarwal, A. The impact of the COVID-19 pandemic on cancer deaths due to delays in diagnosis in England, UK: A national, population-based, modelling study. Lancet Oncol. 2020, 21, 1023–1034. [Google Scholar] [CrossRef]
- Bian, T.; Gibbs, J.D.; Örvell, C.; Imani, F. Respiratory syncytial virus matrix protein induces lung epithelial cell cycle arrest through a p53 dependent pathway. PLoS ONE 2012, 8, e38052. [Google Scholar] [CrossRef]
- Frere, J.J.; Serafini, R.A.; Pryce, K.D.; Zazhytska, M.; Oishi, K.; Golynker, I.; Panis, M.; Zimering, J.; Horiuchi, S.; Hoagland, D.A.; et al. SARS-CoV-2 infection in hamsters and humans results in lasting and unique systemic perturbations post recovery. Sci. Transl. Med. 2022, eabq3059. [Google Scholar] [CrossRef]
In Favour | Against |
---|---|
In senescent and non-senescent cultured human cells SARS-CoV-2 induced senescence and exacerbates the SASP [34,35]. | Acceleration of the biological blood clock reversed in some patients [39]. |
Senescence mechanisms could be mediated by DNA damage and activation of the DNA damage response pathway [36]. | No differences on epigenetic age or telomere length [41]. |
DNA methylation changes [37]. | Persons with LC have not significative changes on cytokine levels of IFN-γ, TNF and IL-6 [47]. |
Acceleration of the biological blood clock [38] and reduction on the telomere length in Vero E6 cells [40]. | No detection of viral RNA of patients recovering from COVID-19 [62]. |
Significant inflammatory response associated wih anti-inflammatory response and mitochondrial stress [45]. | The presence of SARS-CoV-2 RNA months after infection does not necessary indicates the presence of live virus [63]. |
Neutrophil dysfunction [46]. | Non-culture of SARS-CoV-2 from samples with persistence of viral antigen [68]. |
Persons with LC have significant higher levels of IL–17 and IL–2 [47]. | No integration into human genome of SARS-CoV-2 [73,74,75]. |
Residual virus reactivation [49,50]. | In-silico study model not evidence of a genetic correlation with any of the female-specific cancers [90]. |
SARS–CoV–2 promotes inflammasome activation and inflammatory pathways [54,55]. | So far, studies on SARS survivors have not reported an increase in cancer incidence [115,116]. |
Detection of residual SARS-CoV-2 in multiple tissues from patients recovering from COVID-19 [62]. | |
Presence of SARS-CoV-2 RNA and/or antigens months after infection [63,68,69]. | |
Indirect evidence suggesting the viral reservoir hypothesis based on the potential benefits of protease inhibitors in persons having LC [71]. | |
Some tumour suppressor genes could be inhibited by SARS-CoV-2 [79,83,85]. | |
Cross-trait meta-analysis identified some conditions that share multiple mechanistic pathways with some female cancers [90]. | |
Bioinformatics studies suggest that some transcriptional factors show a correlation with respiratory illness and progression of malignancies [91]. | |
Bioinformatic studies show the up-regulation of some tumour–related genes [92]. | |
SARS–CoV–2 mimics the histone proteins [100]. |
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. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Amiama-Roig, A.; Pérez-Martínez, L.; Rodríguez Ledo, P.; Verdugo-Sivianes, E.M.; Blanco, J.-R. Should We Expect an Increase in the Number of Cancer Cases in People with Long COVID? Microorganisms 2023, 11, 713. https://doi.org/10.3390/microorganisms11030713
Amiama-Roig A, Pérez-Martínez L, Rodríguez Ledo P, Verdugo-Sivianes EM, Blanco J-R. Should We Expect an Increase in the Number of Cancer Cases in People with Long COVID? Microorganisms. 2023; 11(3):713. https://doi.org/10.3390/microorganisms11030713
Chicago/Turabian StyleAmiama-Roig, Ana, Laura Pérez-Martínez, Pilar Rodríguez Ledo, Eva M. Verdugo-Sivianes, and José-Ramón Blanco. 2023. "Should We Expect an Increase in the Number of Cancer Cases in People with Long COVID?" Microorganisms 11, no. 3: 713. https://doi.org/10.3390/microorganisms11030713