COVID-19 vs. Cancer Immunosurveillance: A Game of Thrones within an Inflamed Microenviroment
Abstract
:Simple Summary
Abstract
1. Introduction
2. Immune Response against SARS-CoV-2
2.1. Innate Anti-COVID-19 Response
2.2. Adaptive Anti-COVID-19 Response
2.3. Long-Term Impacts of COVID-19-Mediated Immunomodulation
3. Cancer Immunosurveillance, Immunoediting and Inflammation
3.1. The Basic Concepts of Cancer Immune-Surveillance and Immune-Escape
3.2. Cancer Immunosurveillance vs. Inflammation: The Inflamed Tumor Microenviron Ment
4. COVID-19 and Cancer
4.1. COVID-19 Impact on Cancer Disease: The Current Status of Knowledge
4.2. Immunologic Interplay between COVID-19 and Cancer Onset/Progression
5. Conclusions
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- WHO Coronavirus (COVID-19) Dashboard|WHO Coronavirus (COVID-19) Dashboard with Vaccination Data. Available online: https://covid19.who.int (accessed on 28 July 2022).
- Gebhard, C.; Regitz-Zagrosek, V.; Neuhauser, H.K.; Morgan, R.; Klein, S.L. Impact of sex and gender on COVID-19 outcomes in Europe. Biol. Sex Differ. 2020, 11, 29. [Google Scholar] [CrossRef] [PubMed]
- Parasher, A. COVID-19: Current understanding of its Pathophysiology, Clinical presentation and Treatment. Postgrad. Med. J. 2021, 97, 312–320. [Google Scholar] [CrossRef]
- Triggle, C.R.; Bansal, D.; Ding, H.; Islam, M.; Farag, E.A.B.A.; Hadi, H.A.; Sultan, A.A. A Comprehensive Review of Viral Characteristics, Transmission, Pathophysiology, Immune Response, and Management of SARS-CoV-2 and COVID-19 as a Basis for Controlling the Pandemic. Front. Immunol. 2021, 12, 631139. [Google Scholar] [CrossRef] [PubMed]
- Glowacka, I.; Bertram, S.; Müller, M.A.; Allen, P.; Soilleux, E.; Pfefferle, S.; Steffen, I.; Tsegaye, T.S.; He, Y.; Gnirss, K.; et al. Evidence that TMPRSS2 Activates the Severe Acute Respiratory Syndrome Coronavirus Spike Protein for Membrane Fusion and Reduces Viral Control by the Humoral Immune Response. J. Virol. 2011, 85, 4122–4134. [Google Scholar] [CrossRef] [PubMed]
- Sinha, P.; Matthay, M.A.; Calfee, C.S. Is a “Cytokine Storm” Relevant to COVID-19? JAMA Intern. Med. 2020, 180, 1152–1154. [Google Scholar] [CrossRef]
- Andreakos, E.; Zanoni, I.; Galani, I.E. Lambda interferons come to light: Dual function cytokines mediating antiviral immunity and damage control. Curr. Opin. Immunol. 2019, 56, 67–75. [Google Scholar] [CrossRef]
- Park, A.; Iwasaki, A. Type I and Type III Interferons—Induction, Signaling, Evasion, and Application to Combat COVID-19. Cell Host Microbe 2020, 27, 870–878. [Google Scholar] [CrossRef]
- Chu, H.; Chan, J.F.-W.; Wang, Y.; Yuen, T.T.-T.; Chai, Y.; Hou, Y.; Shuai, H.; Yang, D.; Hu, B.; Huang, X.; et al. Comparative Replication and Immune Activation Profiles of SARS-CoV-2 and SARS-CoV in Human Lungs: An Ex Vivo Study With Implications for the Pathogenesis of COVID-19. Clin. Infect. Dis. 2020, 71, 1400–1409. [Google Scholar] [CrossRef]
- Blanco-Melo, D.; Nilsson-Payant, B.E.; Liu, W.-C.; Uhl, S.; Hoagland, D.; Møller, 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. [Google Scholar] [CrossRef]
- Rockx, B.; Kuiken, T.; Herfst, S.; Bestebroer, T.; Lamers, M.M.; Munnink, B.B.O.; De Meulder, D.; Van Amerongen, G.; van den Brand, J.; Okba, N.M.A.; et al. Comparative pathogenesis of COVID-19, MERS, and SARS in a nonhuman primate model. Science 2020, 368, 1012–1015. [Google Scholar] [CrossRef] [Green Version]
- Moore, B.J.B.; June, C.H. Cytokine release syndrome in severe COVID-19. Science 2020, 368, 473–474. [Google Scholar] [CrossRef] [PubMed]
- Winterbourn, C.C.; Kettle, A.J.; Hampton, M.B. Reactive Oxygen Species and Neutrophil Function. Annu. Rev. Biochem. 2016, 85, 765–792. [Google Scholar] [CrossRef] [PubMed]
- Liu, Y.; Du, X.; Chen, J.; Jin, Y.; Peng, L.; Wang, H.H.; Luo, M.; Chen, L.; Zhao, Y. Neutrophil-to-lymphocyte ratio as an independent risk factor for mortality in hospitalized patients with COVID-19. J. Infect. 2020, 81, e6–e12. [Google Scholar] [CrossRef] [PubMed]
- Li, X.; Xu, S.; Yu, M.; Wang, K.; Tao, Y.; Zhou, Y.; Shi, J.; Zhou, M.; Wu, B.; Yang, Z.; et al. Risk factors for severity and mortality in adult COVID-19 inpatients in Wuhan. J. Allergy Clin. Immunol. 2020, 146, 110–118. [Google Scholar] [CrossRef] [PubMed]
- Henderson, L.A.; Canna, S.W.; Schulert, G.S.; Volpi, S.; Lee, P.Y.; Kernan, K.F.; Caricchio, R.; Mahmud, S.; Hazen, M.M.; Halyabar, O.; et al. On the Alert for Cytokine Storm: Immunopathology in COVID -19. Arthritis Rheumatol. 2020, 72, 1059–1063. [Google Scholar] [CrossRef] [PubMed]
- Varghese, P.M.; Tsolaki, A.G.; Yasmin, H.; Shastri, A.; Ferluga, J.; Vatish, M.; Madan, T.; Kishore, U. Host-pathogen interaction in COVID-19: Pathogenesis, potential therapeutics and vaccination strategies. Immunobiology 2020, 225, 152008. [Google Scholar] [CrossRef]
- Boechat, J.; Chora, I.; Morais, A.; Delgado, L. The immune response to SARS-CoV-2 and COVID-19 immunopathology—Current perspectives. Pulmonology 2021, 27, 423–437. [Google Scholar] [CrossRef]
- Gustine, J.N.; Jones, D. Immunopathology of Hyperinflammation in COVID-19. Am. J. Pathol. 2021, 191, 4–17. [Google Scholar] [CrossRef]
- Brandstadter, J.D.; Yang, Y. Natural Killer Cell Responses to Viral Infection. J. Innate Immun. 2011, 3, 274–279. [Google Scholar] [CrossRef]
- Zheng, M.; Gao, Y.; Wang, G.; Song, G.; Liu, S.; Sun, D.; Xu, Y.; Tian, Z. Functional exhaustion of antiviral lymphocytes in COVID-19 patients. Cell. Mol. Immunol. 2020, 17, 533–535. [Google Scholar] [CrossRef] [Green Version]
- Giamarellos-Bourboulis, E.J.; Netea, M.G.; Rovina, N.; Akinosoglou, K.; Antoniadou, A.; Antonakos, N.; Damoraki, G.; Gkavogianni, T.; Adami, M.-E.; Katsaounou, P.; et al. Complex Immune Dysregulation in COVID-19 Patients with Severe Respiratory Failure. Cell Host Microbe 2020, 27, 992–1000. [Google Scholar] [CrossRef] [PubMed]
- Cifaldi, L.; Prencipe, G.; Caiello, I.; Bracaglia, C.; Locatelli, F.; De Benedetti, F.; Strippoli, R. Inhibition of Natural Killer Cell Cytotoxicity by Interleukin-6: Implications for the Pathogenesis of Macrophage Activation Syndrome. Arthritis Rheumatol. 2015, 67, 3037–3046. [Google Scholar] [CrossRef]
- Soresina, A.; Moratto, D.; Chiarini, M.; Paolillo, C.; Baresi, G.; Foca, E.; Bezzi, M.; Baronio, B.; Giacomelli, M.; Badolato, R. Two X-linked agammaglobulinemia patients develop pneumonia as COVID-19 manifestation but recover. Pediatr. Allergy Immunol. 2020, 31, 565–569. [Google Scholar] [CrossRef]
- Quinti, I.; Lougaris, V.; Milito, C.; Cinetto, F.; Pecoraro, A.; Mezzaroma, I.; Mastroianni, C.M.; Turriziani, O.; Bondioni, M.P.; Filippini, M.; et al. A possible role for B cells in COVID-19? Lesson from patients with agammaglobulinemia. J. Allergy Clin. Immunol. 2020, 146, 211–213. [Google Scholar] [CrossRef] [PubMed]
- Zhao, J.; Yuan, Q.; Wang, H.; Liu, W.; Liao, X.; Su, Y.; Wang, X.; Yuan, J.; Li, T.; Li, J.; et al. Antibody Responses to SARS-CoV-2 in Patients With Novel Coronavirus Disease 2019. Clin. Infect. Dis. 2020, 71, 2027–2034. [Google Scholar] [CrossRef] [PubMed]
- Xu, Z.; Shi, L.; Wang, Y.; Zhang, J.; Huang, L.; Zhang, C.; Liu, S.; Zhao, P.; Liu, H.; Zhu, L.; et al. Pathological findings of COVID-19 associated with acute respiratory distress syndrome. Lancet Respir. Med. 2020, 8, 420–422. [Google Scholar] [CrossRef]
- Kuri-Cervantes, L.; Pampena, M.B.; Meng, W.; Rosenfeld, A.M.; Ittner, C.A.G.; Weisman, A.R.; Agyekum, R.S.; Mathew, D.; Baxter, A.E.; Vella, L.A.; et al. Comprehensive mapping of immune perturbations associated with severe COVID-19. Sci. Immunol. 2020, 5, eabd7114. [Google Scholar] [CrossRef]
- Jiang, Y.; Li, Y.; Zhu, B. T-cell exhaustion in the tumor microenvironment. Cell Death Dis. 2015, 6, e1792. [Google Scholar] [CrossRef]
- Moon, C. Fighting COVID-19 exhausts T cells. Nat. Rev. Immunol. 2020, 20, 277. [Google Scholar] [CrossRef]
- Chen, N.; Zhou, M.; Dong, X.; Qu, J.; Gong, F.; Han, Y.; Qiu, Y.; Wang, J.; Liu, Y.; Wei, Y.; et al. Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: A descriptive study. Lancet 2020, 395, 507–513. [Google Scholar] [CrossRef] [Green Version]
- Qin, C.; Zhou, L.; Hu, Z.; Zhang, S.; Yang, S.; Tao, Y.; Xie, C.; Ma, K.; Shang, K.; Wang, W.; et al. Dysregulation of Immune Response in Patients With Coronavirus 2019 (COVID-19) in Wuhan, China. Clin. Infect. Dis. 2020, 71, 762–768. [Google Scholar] [CrossRef] [PubMed]
- Carsana, L.; Sonzogni, A.; Nasr, A.; Rossi, R.S.; Pellegrinelli, A.; Zerbi, P.; Rech, R.; Colombo, R.; Antinori, S.; Corbellino, M.; et al. Pulmonary post-mortem findings in a series of COVID-19 cases from northern Italy: A two-centre descriptive study. Lancet Infect. Dis. 2020, 20, 1135–1140. [Google Scholar] [CrossRef]
- Sette, A.; Crotty, S. Pre-existing immunity to SARS-CoV-2: The knowns and unknowns. Nat. Rev. Immunol. 2020, 20, 457–458. [Google Scholar] [CrossRef] [PubMed]
- Wang, X.; Gui, J. Cell-mediated immunity to SARS-CoV-2. Pediatr. Investig. 2020, 4, 281–291. [Google Scholar] [CrossRef]
- Hotez, P.J.; Bottazzi, M.E.; Corry, D.B. The potential role of Th17 immune responses in coronavirus immunopathology and vaccine-induced immune enhancement. Microbes Infect. 2020, 22, 165–167. [Google Scholar] [CrossRef]
- Li, H.; Liu, L.; Zhang, D.; Xu, J.; Dai, H.; Tang, N.; Su, X.; Cao, B. SARS-CoV-2 and viral sepsis: Observations and hypotheses. Lancet 2020, 395, 1517–1520. [Google Scholar] [CrossRef]
- Mehta, P.; McAuley, D.F.; Brown, M.; Sanchez, E.; Tattersall, R.S.; Manson, J.J.; on behalf of theHLH Across Speciality Collaboration, UK. COVID-19: Consider cytokine storm syndromes and immunosuppression. Lancet 2020, 395, 1033–1034. [Google Scholar] [CrossRef]
- Karlsson, A.C.; Humbert, M.; Buggert, M. The known unknowns of T cell immunity to COVID-19. Sci. Immunol. 2020, 5, eabe8063. [Google Scholar] [CrossRef]
- Yong, S.J. Long COVID or post-COVID-19 syndrome: Putative pathophysiology, risk factors, and treatments. Infect. Dis. 2021, 53, 737–754. [Google Scholar] [CrossRef]
- Peluso, M.J.; Deitchman, A.N.; Torres, L.; Iyer, N.S.; Munter, S.E.; Nixon, C.C.; Donatelli, J.; Thanh, C.; Takahashi, S.; Hakim, J.; et al. Long-term SARS-CoV-2-specific immune and inflammatory responses in individuals recovering from COVID-19 with and without post-acute symptoms. Cell Rep. 2021, 36, 109518. [Google Scholar] [CrossRef]
- Mao, Y.; Wang, W.; Ma, J.; Wu, S.; Sun, F. Reinfection rates among patients previously infected by SARS-CoV-2: Systematic review and meta-analysis. Chin. Med. J. 2021, 135, 145–152. [Google Scholar] [CrossRef]
- Subramanian, A.; Nirantharakumar, K.; Hughes, S.; Myles, P.; Williams, T.; Gokhale, K.M.; Taverner, T.; Chandan, J.S.; Brown, K.; Simms-Williams, N.; et al. Symptoms and risk factors for long COVID in non-hospitalized adults. Nat. Med. 2022, 28, 1706–1714. [Google Scholar] [CrossRef]
- Burnet, F.M. The concept of immunological surveillance. Prog. Exp. Tumor Res. 1970, 13, 1–27. [Google Scholar] [PubMed]
- Cellular and Humoral Aspects of the Hypersensitive States: A Symposium at the New York Academy of Medicine. J. Am. Med. Assoc. 1959, 170, 883. [CrossRef]
- Dunn, G.P.; Bruce, A.T.; Ikeda, H.; Old, L.J.; Schreiber, R.D. Cancer immunoediting: From immunosurveillance to tumor escape. Nat. Immunol. 2002, 3, 991–998. [Google Scholar] [CrossRef] [PubMed]
- Kyriazi, A.A.; Papiris, E.; Kalyvianakis, K.K.; Sakellaris, G.; Baritaki, S. Dual Effects of Non-Coding RNAs (ncRNAs) in Cancer Stem Cell Biology. Int. J. Mol. Sci. 2020, 21, 6658. [Google Scholar] [CrossRef]
- Novikova, I.V.; Hennelly, S.P.; Sanbonmatsu, K.Y. Tackling Structures of Long Noncoding RNAs. Int. J. Mol. Sci. 2013, 14, 23672–23684. [Google Scholar] [CrossRef]
- Cabili, M.N.; Trapnell, C.; Goff, L.; Koziol, M.; Tazon-Vega, B.; Regev, A.; Rinn, J.L. Integrative annotation of human large intergenic noncoding RNAs reveals global properties and specific subclasses. Genes Dev. 2011, 25, 1915–1927. [Google Scholar] [CrossRef]
- Yaguchi, T.; Sumimoto, H.; Kudo-Saito, C.; Tsukamoto, N.; Ueda, R.; Iwata-Kajihara, T.; Nishio, H.; Kawamura, N.; Kawakami, Y. The mechanisms of cancer immunoescape and development of overcoming strategies. Int. J. Hematol. 2011, 93, 294–300. [Google Scholar] [CrossRef]
- Rosen, J.M.; Jordan, C.T. The Increasing Complexity of the Cancer Stem Cell Paradigm. Science 2009, 324, 1670–1673. [Google Scholar] [CrossRef] [Green Version]
- Kudo-Saito, C.; Shirako, H.; Takeuchi, T.; Kawakami, Y. Cancer Metastasis Is Accelerated through Immunosuppression during Snail-Induced EMT of Cancer Cells. Cancer Cell 2009, 15, 195–206. [Google Scholar] [CrossRef] [PubMed]
- Dyck, L.; Mills, K.H.G. Immune checkpoints and their inhibition in cancer and infectious diseases. Eur. J. Immunol. 2017, 47, 765–779. [Google Scholar] [CrossRef] [PubMed]
- Takahashi, T.; Tagami, T.; Yamazaki, S.; Uede, T.; Shimizu, J.; Sakaguchi, N.; Mak, T.W.; Sakaguchi, S. Immunologic Self-Tolerance Maintained by Cd25+Cd4+Regulatory T Cells Constitutively Expressing Cytotoxic T Lymphocyte–Associated Antigen 4. J. Exp. Med. 2000, 192, 303–310. [Google Scholar] [CrossRef]
- Marques, A.C.; Ponting, C. Intergenic lncRNAs and the evolution of gene expression. Curr. Opin. Genet. Dev. 2014, 27, 48–53. [Google Scholar] [CrossRef] [PubMed]
- Petermann, K.B.; Rozenberg, G.I.; Zedek, D.; Groben, P.; McKinnon, K.; Buehler, C.; Kim, W.Y.; Shields, J.M.; Penland, S.; Bear, J.E.; et al. CD200 is induced by ERK and is a potential therapeutic target in melanoma. J. Clin. Investig. 2007, 117, 3922–3929. [Google Scholar] [CrossRef]
- Trovato, R.; Fiore, A.; Sartori, S.; Canè, S.; Giugno, R.; Cascione, L.; Paiella, S.; Salvia, R.; De Sanctis, F.; Poffe, O.; et al. Immunosuppression by monocytic myeloid-derived suppressor cells in patients with pancreatic ductal carcinoma is orchestrated by STAT3. J. Immunother. Cancer 2019, 7, 255. [Google Scholar] [CrossRef]
- Gabrilovich, D.I.; Nagaraj, S. Myeloid-derived suppressor cells as regulators of the immune system. Nat. Rev. Immunol. 2009, 9, 162–174. [Google Scholar] [CrossRef]
- DeNardo, D.G.; Ruffell, B. Macrophages as regulators of tumour immunity and immunotherapy. Nat. Rev. Immunol. 2019, 19, 369–382. [Google Scholar] [CrossRef]
- Hussain, S.P.; Harris, C.C. Inflammation and cancer: An ancient link with novel potentials. Int. J. Cancer 2007, 121, 2373–2380. [Google Scholar] [CrossRef]
- Szalayova, G.; James, T.A.; Rincon, M. A framework for the role of acute inflammation in tumor progression. Breast Cancer Res. Treat. 2015, 151, 235–238. [Google Scholar] [CrossRef]
- Hobson, J.; Gummadidala, P.; Silverstrim, B.; Grier, D.; Bunn, J.; James, T.; Rincon, M. Acute inflammation induced by the biopsy of mouse mammary tumors promotes the development of metastasis. Breast Cancer Res. Treat. 2013, 139, 391–401. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Bates, R.C.; Mercurio, A.M. Tumor Necrosis Factor-α Stimulates the Epithelial-to-Mesenchymal Transition of Human Colonic Organoids. Mol. Biol. Cell 2003, 14, 1790–1800. [Google Scholar] [CrossRef] [PubMed]
- Abulaiti, A.; Shintani, Y.; Funaki, S.; Nakagiri, T.; Inoue, M.; Sawabata, N.; Minami, M.; Okumura, M. Interaction between non-small-cell lung cancer cells and fibroblasts via enhancement of TGF-β signaling by IL-6. Lung Cancer 2013, 82, 204–213. [Google Scholar] [CrossRef]
- Yadav, A.; Kumar, B.; Datta, J.; Teknos, T.N.; Kumar, P. IL-6 Promotes Head and Neck Tumor Metastasis by Inducing Epithelial–Mesenchymal Transition via the JAK-STAT3-SNAIL Signaling Pathway. Mol. Cancer Res. 2011, 9, 1658–1667. [Google Scholar] [CrossRef]
- Eldesoky, A.; Shouma, A.; Mosaad, Y.; Elhawary, A. Clinical relevance of serum vascular endothelial growth factor and Interleukin-6 in patients with colorectal cancer. Saudi J. Gastroenterol. 2011, 17, 170–173. [Google Scholar] [CrossRef]
- Huang, S.-P.; Wu, M.-S.; Shun, C.-T.; Wang, H.-P.; Lin, M.-T.; Kuo, M.-L.; Lin, J.-T. Interleukin-6 increases vascular endothelial growth factor and angiogenesis in gastric carcinoma. J. Biomed. Sci. 2004, 11, 517–527. [Google Scholar] [CrossRef]
- Moore, K.W.; de Waal Malefyt, R.; Coffman, R.L.; O’Garra, A. Interleukin-10 and the Interleukin-10 Receptor. Annu. Rev. Immunol. 2001, 19, 683–765. [Google Scholar] [CrossRef]
- Roers, A.; Siewe, L.; Strittmatter, E.; Deckert, M.; Schlüter, D.; Stenzel, W.; Gruber, A.D.; Krieg, T.; Rajewsky, K.; Muller, W. T Cell–specific Inactivation of the Interleukin 10 Gene in Mice Results in Enhanced T Cell Responses but Normal Innate Responses to Lipopolysaccharide or Skin Irritation. J. Exp. Med. 2004, 200, 1289–1297. [Google Scholar] [CrossRef]
- Tian, G.; Li, J.-L.; Wang, D.-G.; Zhou, D. Targeting IL-10 in Auto-immune Diseases. Cell Biophys. 2014, 70, 37–49. [Google Scholar] [CrossRef]
- Murray, P.J. Understanding and exploiting the endogenous interleukin-10/STAT3-mediated anti-inflammatory response. Curr. Opin. Pharmacol. 2006, 6, 379–386. [Google Scholar] [CrossRef] [PubMed]
- Li, L.; Yu, R.; Cai, T.; Chen, Z.; Lan, M.; Zou, T.; Wang, B.; Wang, Q.; Zhao, Y.; Cai, Y. Effects of immune cells and cytokines on inflammation and immunosuppression in the tumor microenvironment. Int. Immunopharmacol. 2020, 88, 106939. [Google Scholar] [CrossRef] [PubMed]
- Hsu, P.; Santner-Nanan, B.; Hu, M.; Skarratt, K.; Lee, C.H.; Stormon, M.; Wong, M.; Fuller, S.J.; Nanan, R. IL-10 Potentiates Differentiation of Human Induced Regulatory T Cells via STAT3 and Foxo1. J. Immunol. 2015, 195, 3665–3674. [Google Scholar] [CrossRef]
- Alas, S.; Emmanouilides, C.; Bonavida, B. Inhibition of interleukin 10 by rituximab results in down-regulation of bcl-2 and sensitization of B-cell non-Hodgkin’s lymphoma to apoptosis. Clin. Cancer Res. 2001, 7, 709–723. [Google Scholar] [PubMed]
- Braun, D.A.; Fribourg, M.; Sealfon, S.C. Cytokine Response Is Determined by Duration of Receptor and Signal Transducers and Activators of Transcription 3 (STAT3) Activation. J. Biol. Chem. 2013, 288, 2986–2993. [Google Scholar] [CrossRef]
- Yang, C.; He, L.; He, P.; Liu, Y.; Wang, W.; He, Y.; Du, Y.; Gao, F. Increased drug resistance in breast cancer by tumor-associated macrophages through IL-10/STAT3/bcl-2 signaling pathway. Med. Oncol. 2015, 32, 14–352. [Google Scholar] [CrossRef]
- Popa, C.; Netea, M.G.; van Riel, P.L.C.M.; van der Meer, J.W.M.; Stalenhoef, A.F.H. The role of TNF-α in chronic inflammatory conditions, intermediary metabolism, and cardiovascular risk. J. Lipid Res. 2007, 48, 751–762. [Google Scholar] [CrossRef]
- Woo, C.-H.; Eom, Y.-W.; Yoo, M.-H.; You, H.-J.; Han, H.J.; Song, W.K.; Yoo, Y.J.; Chun, J.-S.; Kim, J.-H. Tumor Necrosis Factor-α Generates Reactive Oxygen Species via a Cytosolic Phospholipase A2-linked Cascade. J. Biol. Chem. 2000, 275, 32357–32362. [Google Scholar] [CrossRef]
- Meulmeester, E.; Ten Dijke, P. The dynamic roles of TGF-β in cancer. J. Pathol. 2011, 223, 205–218. [Google Scholar] [CrossRef]
- Chang, J.H.; Jiang, Y.; Pillarisetty, V.G. Role of immune cells in pancreatic cancer from bench to clinical application. Medicine 2016, 95, e5541. [Google Scholar] [CrossRef]
- Fuxe, J.; Karlsson, M.C. TGF-β-induced epithelial-mesenchymal transition: A link between cancer and inflammation. Semin. Cancer Biol. 2012, 22, 455–461. [Google Scholar] [CrossRef] [PubMed]
- Xu, J.; Lamouille, S.; Derynck, R. TGF-beta-induced epithelial to mesenchymal transition. Cell Res. 2009, 19, 156–172. [Google Scholar] [CrossRef] [PubMed]
- Bui, J.D.; Schreiber, R.D. Cancer immunosurveillance, immunoediting and inflammation: Independent or interdependent processes? Curr. Opin. Immunol. 2007, 19, 203–208. [Google Scholar] [CrossRef]
- Chow, M.T.; Möller, A.; Smyth, M.J. Inflammation and immune surveillance in cancer. Semin. Cancer Biol. 2012, 22, 23–32. [Google Scholar] [CrossRef] [PubMed]
- Massagué, J. TGFbeta in Cancer. Cell 2008, 134, 215–230. [Google Scholar] [CrossRef]
- Suganuma, M.; Okabe, S.; Marino, M.W.; Sakai, A.; Sueoka, E.; Fujiki, H. Essential role of tumor necrosis factor alpha (TNF-alpha) in tumor promotion as revealed by TNF-alpha-deficient mice. Cancer Res. 1999, 59, 4516–4518. [Google Scholar]
- Luo, J.-L.; Maeda, S.; Hsu, L.-C.; Yagita, H.; Karin, M. Inhibition of NF-κB in cancer cells converts inflammation- induced tumor growth mediated by TNFα to TRAIL-mediated tumor regression. Cancer Cell 2004, 6, 297–305. [Google Scholar] [CrossRef] [PubMed]
- Waldhauer, I.; Steinle, A. NK cells and cancer immunosurveillance. Oncogene 2008, 27, 5932–5943. [Google Scholar] [CrossRef]
- Ebata, K.; Shimizu, Y.; Nakayama, Y.; Minemura, M.; Murakami, J.; Kato, T.; Yasumura, S.; Takahara, T.; Sugiyama, T.; Saito, S. Immature NK Cells Suppress Dendritic Cell Functions during the Development of Leukemia in a Mouse Model. J. Immunol. 2006, 176, 4113–4124. [Google Scholar] [CrossRef]
- Khazaie, K.; von Boehmer, H. The impact of CD4+CD25+ Treg on tumor specific CD8+ T cell cytotoxicity and cancer. Semin. Cancer Biol. 2006, 16, 124–136. [Google Scholar] [CrossRef]
- Erdman, S.E.; Sohn, J.J.; Rao, V.P.; Nambiar, P.R.; Ge, Z.; Fox, J.G.; Schauer, D.B. CD4+CD25+ Regulatory Lymphocytes Induce Regression of Intestinal Tumors in ApcMin/+ Mice. Cancer Res. 2005, 65, 3998–4004. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Olver, S.; Groves, P.; Buttigieg, K.; Morris, E.S.; Janas, M.L.; Kelso, A.; Kienzle, N. Tumor-Derived Interleukin-4 Reduces Tumor Clearance and Deviates the Cytokine and Granzyme Profile of Tumor-Induced CD8+ T Cells. Cancer Res. 2006, 66, 571–580. [Google Scholar] [CrossRef] [PubMed]
- Schüler, T.; Blankenstein, T. Cutting Edge: CD8+ Effector T Cells Reject Tumors by Direct Antigen Recognition but Indirect Action on Host Cells. J. Immunol. 2003, 170, 4427–4431. [Google Scholar] [CrossRef]
- Liu, C.; Zhao, Y.; Okwan-Duodu, E.A.D.; Basho, R.; Cui, X. COVID-19 in cancer patients: Risk, clinical features, and management. Cancer Biol. Med. 2020, 17, 519–527. [Google Scholar] [CrossRef]
- Zarifkar, P.; Kamath, A.; Robinson, C.; Morgulchik, N.; Shah, S.; Cheng, T.; Dominic, C.; Fehintola, A.; Bhalla, G.; Ahillan, T.; et al. Clinical Characteristics and Outcomes in Patients with COVID-19 and Cancer: A Systematic Review and Meta-analysis. Clin. Oncol. 2021, 33, e180–e191. [Google Scholar] [CrossRef]
- Zhang, L.; Zhu, F.; Xie, L.; Wang, C.; Wang, J.; Chen, R.; Jia, P.; Guan, H.Q.; Peng, L.; Chen, Y.; et al. Clinical characteristics of COVID-19-infected cancer patients: A retrospective case study in three hospitals within Wuhan, China. Ann. Oncol. 2020, 31, 894–901. [Google Scholar] [CrossRef]
- Chavez-MacGregor, M.; Lei, X.; Zhao, H.; Scheet, P.; Giordano, S.H. Evaluation of COVID-19 Mortality and Adverse Outcomes in US Patients With or Without Cancer. JAMA Oncol. 2022, 8, 69–78. [Google Scholar] [CrossRef]
- Ioannidis, J.P.; Axfors, C.; Contopoulos-Ioannidis, D.G. Population-level COVID-19 mortality risk for non-elderly individuals overall and for non-elderly individuals without underlying diseases in pandemic epicenters. Environ. Res. 2020, 188, 109890. [Google Scholar] [CrossRef]
- Aboueshia, M.; Hussein, M.H.; Attia, A.S.; Swinford, A.; Miller, P.; Omar, M.; Toraih, E.A.; Saba, N.; Safah, H.; Duchesne, J.; et al. Cancer and COVID-19: Analysis of patient outcomes. Futur. Oncol. 2021, 17, 3499–3510. [Google Scholar] [CrossRef]
- Jee, J.; Foote, M.B.; Lumish, M.; Stonestrom, A.J.; Wills, B.; Narendra, V.; Avutu, V.; Murciano-Goroff, Y.R.; Chan, J.E.; Derkach, A.; et al. Chemotherapy and COVID-19 Outcomes in Patients With Cancer. J. Clin. Oncol. 2020, 38, 3538–3546. [Google Scholar] [CrossRef] [PubMed]
- Dai, M.; Liu, D.; Liu, M.; Zhou, F.; Li, G.; Chen, Z.; Zhang, Z.; You, H.; Wu, M.; Zheng, Q.; et al. Patients with cancer appear more vulnerable to SARS-COV-2: A multicenter study during the COVID-19 outbreak. Cancer Discov. 2020, 10, 783–791. [Google Scholar] [CrossRef] [PubMed]
- Okwan-Duodu, D.; Pollack, B.P.; Lawson, D.; Khan, M.K. Role of Radiation Therapy as Immune Activator in the Era of Modern Immunotherapy for Metastatic Malignant Melanoma. Am. J. Clin. Oncol. 2015, 38, 119–125. [Google Scholar] [CrossRef] [PubMed]
- Bersanelli, M. Controversies about COVID-19 and anticancer treatment with immune checkpoint inhibitors. Immunotherapy 2020, 12, 269–273. [Google Scholar] [CrossRef]
- Saha, A.; Anirvan, P. Cancer progression in COVID-19: Integrating the roles of renin angiotensin aldosterone system, angiopoietin-2, heat shock protein-27 and epithelial mesenchymal transition. Ecancermedicalscience 2020, 14, 1099. [Google Scholar] [CrossRef]
- Imanishi, Y.; Hu, B.; Jarzynka, M.J.; Guo, P.; Elishaev, E.; Bar-Joseph, I.; Cheng, S.-Y. Angiopoietin-2 Stimulates Breast Cancer Metastasis through the α5β1 Integrin-Mediated Pathway. Cancer Res. 2007, 67, 4254–4263. [Google Scholar] [CrossRef]
- Huang, C.-Y.; Wei, P.-L.; Chen, W.-Y.; Chang, W.-C.; Chang, Y.-J. Silencing Heat Shock Protein 27 Inhibits the Progression and Metastasis of Colorectal Cancer (CRC) by Maintaining the Stability of Stromal Interaction Molecule 1 (STIM1) Proteins. Cells 2018, 7, 262. [Google Scholar] [CrossRef]
- Da Costa, P.L.; Sirois, P.; Tannock, I.F.; Chammas, R. The role of kinin receptors in cancer and therapeutic opportunities. Cancer Lett. 2014, 345, 27–38. [Google Scholar] [CrossRef]
- Katsogiannou, M.; Andrieu, C.; Rocchi, P. Heat shock protein 27 phosphorylation state is associated with cancer progression. Front. Genet. 2014, 5, 346. [Google Scholar] [CrossRef]
- El-Arif, G.; Khazaal, S.; Farhat, A.; Harb, J.; Annweiler, C.; Wu, Y.; Cao, Z.; Kovacic, H.; Khattar, Z.A.; Fajloun, Z.; et al. Angiotensin II Type I Receptor (AT1R): The Gate towards COVID-19-Associated Diseases. Molecules 2022, 27, 2048. [Google Scholar] [CrossRef]
- Tawinwung, S.; Ninsontia, C.; Chanvorachote, P. Angiotensin II Increases Cancer Stem Cell-like Phenotype in Lung Cancer Cells. Anticancer Res. 2015, 35, 4789–4797. [Google Scholar] [PubMed]
- Arrieta, O.; Villarreal-Garza, C.; Vizcaíno, G.; Pineda, B.; Hernández-Pedro, N.; Guevara-Salazar, P.; Wegman-Ostrosky, T.; Villanueva-Rodríguez, G.; Gamboa-Domínguez, A. Association between AT1 and AT2 angiotensin II receptor expression with cell proliferation and angiogenesis in operable breast cancer. Tumor Biol. 2015, 36, 5627–5634. [Google Scholar] [CrossRef] [PubMed]
- Bhowmick, N.A.; Oft, J.; Dorff, T.; Pal, S.; Agarwal, N.; Figlin, R.A.; Posadas, E.M.; Freedland, S.J.; Gong, J. COVID-19 and androgen-targeted therapy for prostate cancer patients. Endocr. Relat. Cancer 2020, 27, R281–R292. [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] [PubMed]
- Ohadi, L.; Hosseinzadeh, F.; Dadkhahfar, S.; Nasiri, S. Oncolytic effect of SARS-CoV-2 in a patient with mycosis fungoides: A case report. Clin. Case Rep. 2022, 10, e05682. [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.; Calveri, M.M.; Calabrese, A.; Pizzarelli, G.; Bongiovanni, I.; Andreoli, M.; Cattaneo, C.; Rignanese, G. Oncolytic effect of SARS-CoV2 in a patient with NK lymphoma: Spontaneous remission in NK/T-cell Lymphoma during COVID-19. Acta Bio Med. Atenei Parm. 2020, 91, e2020047. [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. Pediatr. 2021, 48, 2652–2654. [Google Scholar] [CrossRef] [PubMed]
- Antwi-Amoabeng, D.; Ulanja, M.B.; Beutler, B.D.; Reddy, S.V. Multiple Myeloma Remission Following COVID-19: An Observation in Search of a Mechanism (a Case Report). Pan Afr. Med. J. 2021, 39, 117. [Google Scholar] [CrossRef]
- Kandeel, E.Z.; Refaat, L.; Abdel-Fatah, R.; Samra, M.; Bayoumi, A.; Abdellateif, M.S.; Abdel-Hady, H.; Ali, M.; Khafagy, M. Could COVID-19 induce remission of acute leukemia? Hematology 2021, 26, 870–873. [Google Scholar] [CrossRef]
- Shin, D.H.; Gillard, A.; Van Wieren, A.; Gomez-Manzano, C.; Fueyo, J. Remission of liquid tumors and SARS-CoV-2 infection: A literature review. Mol. Ther. Oncolytics 2022, 26, 135–140. [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]
- Li, M.-Y.; Li, L.; Zhang, Y.; Wang, X.-S. Expression of the SARS-CoV-2 cell receptor gene ACE2 in a wide variety of human tissues. Infect. Dis. Poverty 2020, 9, 23–29. [Google Scholar] [CrossRef] [PubMed]
- Domingo, E.; Escarmís, C.; Sevilla, N.; Moya, A.; Elena, S.; Quer, J.; Novella, I.S.; Holland, J.J. Basic concepts in RNA virus evolution. FASEB J. 1996, 10, 859–864. [Google Scholar] [CrossRef] [PubMed]
- Guillerey, C.; Smyth, M.J. NK cells and cancer immunoediting. In Natural Killer Cells; Springer: Cham, Switzerland, 2015; pp. 115–145. [Google Scholar] [CrossRef]
- Dunn, G.P.; Koebel, C.M.; Schreiber, R.D. Interferons, immunity and cancer immunoediting. Nat. Rev. Immunol. 2006, 6, 836–848. [Google Scholar] [CrossRef]
- Andrade, B.S.; Siqueira, S.; Soares, W.D.A.; Rangel, F.D.S.; Santos, N.; Freitas, A.D.S.; da Silveira, P.R.; Tiwari, S.; Alzahrani, K.; Góes-Neto, A.; et al. Long-COVID and Post-COVID Health Complications: An Up-to-Date Review on Clinical Conditions and Their Possible Molecular Mechanisms. Viruses 2021, 13, 700. [Google Scholar] [CrossRef]
- Apor, E.; O’Brien, J.; Stephen, M.; Castillo, J.J. Systemic lupus erythematosus is associated with increased incidence of hematologic malignancies: A meta-analysis of prospective cohort studies. Leuk. Res. 2014, 38, 1067–1071. [Google Scholar] [CrossRef] [PubMed]
- Llorca, J.; Lopez-Diaz, M.J.; Gonzalez-Juanatey, C.; Ollier, W.E.; Martin, J.; Gonzalez-Gay, M.A. Persistent Chronic Inflammation Contributes to the Development of Cancer in Patients with Rheumatoid Arthritis from a Defined Population of Northwestern Spain. Semin. Arthritis Rheum. 2007, 37, 31–38. [Google Scholar] [CrossRef]
- Madhok, R.; Crilly, A.; Watson, J.; A Capell, H. Serum interleukin 6 levels in rheumatoid arthritis: Correlations with clinical and laboratory indices of disease activity. Ann. Rheum. Dis. 1993, 52, 232–234. [Google Scholar] [CrossRef]
- Ludwig, H.; Nachbaur, D.M.; Fritz, E.; Krainer, M.; Huber, H. Interleukin-6 Is a Prognostic Factor in Multiple Myeloma. Blood 1991, 77, 2794–2795. [Google Scholar] [CrossRef]
- Kawano, M.; Hirano, T.; Matsuda, T.; Taga, T.; Horii, Y.; Iwato, K.; Asaoku, H.; Tang, B.; Tanabe, O.; Tanaka, H.; et al. Autocrine generation and requirement of BSF-2/IL-6 for human multiple myelomas. Nature 1988, 332, 83–85. [Google Scholar] [CrossRef]
- Hirano, T.; Taga, T.; Yasukawa, K.; Nakajima, K.; Nakano, N.; Takatsuki, F.; Shimizu, M.; Murashima, A.; Tsunasawa, S.; Sakiyama, F. Human B-cell differentiation factor defined by an anti-peptide antibody and its possible role in autoantibody production. Proc. Natl. Acad. Sci. USA 1987, 84, 228–231. [Google Scholar] [CrossRef] [PubMed]
- Hobisch, A.; Rogatsch, H.; Hittmair, A.; Fuchs, D.; Bartsch, G.; Klocker, H.; Culig, Z. Immunohistochemical localization of interleukin-6 and its receptor in benign, premalignant and malignant prostate tissue. J. Pathol. 2000, 191, 239–244. [Google Scholar] [CrossRef]
- Hirano, T. IL-6 in inflammation, autoimmunity and cancer. Int. Immunol. 2021, 33, 127–148. [Google Scholar] [CrossRef] [PubMed]
- Greisen, S.R.; Deleuran, B. Checkpoint Molecules in Rheumatology—or the Benefits of Being Exhausted. Curr. Rheumatol. Rep. 2021, 23, 22. [Google Scholar] [CrossRef]
- Masetti, R.; Tiri, A.; Tignanelli, A.; Turrini, E.; Argentiero, A.; Pession, A.; Esposito, S. Autoimmunity and cancer. Autoimmun. Rev. 2021, 20, 102882. [Google Scholar] [CrossRef]
- Dreyer, L.; Faurschou, M.; Mogensen, M.; Jacobsen, S. High incidence of potentially virus-induced malignancies in systemic lupus erythematosus: A long-term followup study in a Danish cohort. Arthritis Care Res. 2011, 63, 3032–3037. [Google Scholar] [CrossRef]
- Ward, M.D.; Buehler, M.J.; Jaffe, M.H.; Berkelman, R.L. 1993 Revised Classification System for HIV Infection and Expanded Surveillance Case Definition for AIDS among Adolescents and Adults. MMWR Recomm. Rep. 1992, 41, 1–19. [Google Scholar]
- Yarchoan, R.; Uldrick, T.S. HIV-Associated Cancers and Related Diseases. N. Engl. J. Med. 2018, 378, 1029–1041. [Google Scholar] [CrossRef]
- Shmakova, A.; Germini, D.; Vassetzky, Y. HIV-1, HAART and cancer: A complex relationship. Int. J. Cancer 2020, 146, 2666–2679. [Google Scholar] [CrossRef]
- Bruyand, M.; Ryom, L.; Shepherd, L.; Fatkenheuer, G.; Grulich, A.; Reiss, P.; de Wit, S.; Monforte, A.D.; Furrer, H.; Pradier, C.; et al. Cancer Risk and Use of Protease Inhibitor or Nonnucleoside Reverse Transcriptase Inhibitor–Based Combination Antiretroviral Therapy. JAIDS J. Acquir. Immune Defic. Syndr. 2015, 68, 568–577. [Google Scholar] [CrossRef]
- Krasemann, S.; Haferkamp, U.; Pfefferle, S.; Woo, M.S.; Heinrich, F.; Schweizer, M.; Appelt-Menzel, A.; Cubukova, A.; Barenberg, J.; Leu, J.; et al. The blood-brain barrier is dysregulated in COVID-19 and serves as a CNS entry route for SARS-CoV-2. Stem Cell Rep. 2022, 17, 307–320. [Google Scholar] [CrossRef] [PubMed]
- Peirouvi, T.; Aliaghaei, A.; Farsani, B.E.; Ziaeipour, S.; Ebrahimi, V.; Forozesh, M.; Ghadipasha, M.; Mahmoudiasl, G.-R.; Aryan, A.; Moghimi, N.; et al. COVID-19 disrupts the blood–testis barrier through the induction of inflammatory cytokines and disruption of junctional proteins. Agents Actions 2021, 70, 1165–1175. [Google Scholar] [CrossRef] [PubMed]
- Program, W.T.C.H. Minimum Latency & Types of Cancer. 2012; pp. 1–9. Available online: https://www.cdc.gov/wtc/pdfs/policies/WTCHP-Minimum-Cancer-Latency-PP-01062015-508.pdf (accessed on 31 August 2022).
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 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
Liapis, I.; Baritaki, S. COVID-19 vs. Cancer Immunosurveillance: A Game of Thrones within an Inflamed Microenviroment. Cancers 2022, 14, 4330. https://doi.org/10.3390/cancers14174330
Liapis I, Baritaki S. COVID-19 vs. Cancer Immunosurveillance: A Game of Thrones within an Inflamed Microenviroment. Cancers. 2022; 14(17):4330. https://doi.org/10.3390/cancers14174330
Chicago/Turabian StyleLiapis, Ioannis, and Stavroula Baritaki. 2022. "COVID-19 vs. Cancer Immunosurveillance: A Game of Thrones within an Inflamed Microenviroment" Cancers 14, no. 17: 4330. https://doi.org/10.3390/cancers14174330
APA StyleLiapis, I., & Baritaki, S. (2022). COVID-19 vs. Cancer Immunosurveillance: A Game of Thrones within an Inflamed Microenviroment. Cancers, 14(17), 4330. https://doi.org/10.3390/cancers14174330