Biological and Exploitable Crossroads for the Immune Response in Cancer and COVID-19
Abstract
:1. Introduction
2. Time Makes the Difference: Acute and Chronic Features of Immune Response
3. Cytokines
4. Molecular Pathways
5. Therapeutic Implications
6. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Scagliotti, G.; Novello, S.; Veltri, A.; Boccuzzi, A.; Perboni, A.; Terzolo, M. Patients With Lung Cancer and Coronavirus Disease 2019 Epidemic: An Experience From an Italian University Hospital. JTO Clin. Res. Rep. 2020, 1, 100067. [Google Scholar] [CrossRef] [PubMed]
- Smeltzer, M.P.; Scagliotti, G.V.; Wakelee, H.A.; Mitsudomi, T.; Roy, U.B.; Clark, R.C.; Arndt, R.; Pruett, C.D.; Kelly, K.L.; Ujhazy, P.; et al. International Association for the Study of Lung Cancer Study of the Impact of Coronavirus Disease 2019 on International Lung Cancer Clinical Trials. J. Thorac. Oncol. 2022, 17, 651–660. [Google Scholar] [CrossRef] [PubMed]
- Richards, M.; Anderson, M.; Carter, P.; Ebert, B.L.; Mossialos, E. The impact of the COVID-19 pandemic on cancer care. Nat. Cancer 2020, 1, 565–567. [Google Scholar] [CrossRef] [PubMed]
- Lee, L.Y.; Cazier, J.B.; Angelis, V.; Arnold, R.; Bisht, V.; Campton, N.A.; Chackathayil, J.; Cheng, V.W.; Curley, H.M.; Fittall, M.W.; et al. COVID-19 mortality in patients with cancer on chemotherapy or other anticancer treatments: A prospective cohort study. Lancet 2020, 395, 1919–1926. [Google Scholar] [CrossRef]
- Kuderer, N.M.; Choueiri, T.K.; Shah, D.P.; Shyr, Y.; Rubinstein, S.M.; Rivera, D.R.; Shete, S.; Hsu, C.Y.; Desai, A.; de Lima Lopes, G.; et al. Clinical impact of COVID-19 on patients with cancer (CCC19): A cohort study. Lancet 2020, 395, 1907–1918. [Google Scholar] [CrossRef]
- Yuan, Y.; Cao, D.; Zhang, Y.; Ma, J.; Qi, J.; Wang, Q.; Lu, G.; Wu, Y.; Yan, J.; Shi, Y.; et al. Cryo-EM structures of MERS-CoV and SARS-CoV spike glycoproteins reveal the dynamic receptor binding domains. Nat. Commun. 2017, 8, 15092. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Walls, A.C.; Park, Y.J.; Tortorici, M.A.; Wall, A.; McGuire, A.T.; Veesler, D. Structure, Function, and Antigenicity of the SARS-CoV-2 Spike Glycoprotein. Cell 2020, 183, 1735. [Google Scholar] [CrossRef] [PubMed]
- Hui, K.P.Y.; Cheung, M.C.; Perera, R.A.P.M.; Ng, K.C.; Bui, C.H.T.; Ho, J.C.W.; Ng, M.M.T.; Kuok, D.I.T.; Shih, K.C.; Tsao, S.W.; et al. Tropism, replication competence, and innate immune responses of the coronavirus SARS-CoV-2 in human respiratory tract and conjunctiva: An analysis in ex-vivo and in-vitro cultures. Lancet Respir. Med. 2020, 8, 687–695. [Google Scholar] [CrossRef]
- Mathew, D.; Giles, J.R.; Baxter, A.E.; Oldridge, D.A.; Greenplate, A.R.; Wu, J.E.; Alanio, C.; Kuri-Cervantes, L.; Pampena, M.B.; D’Andrea, K.; et al. Deep immune profiling of COVID-19 patients reveals distinct immunotypes with therapeutic implications. Science 2020, 369, eabc8511. [Google Scholar] [CrossRef]
- Gold, M.S.; Sehayek, D.; Gabrielli, S.; Zhang, X.; McCusker, C.; Ben-Shoshan, M. COVID-19 and comorbidities: A systematic review and meta-analysis. Postgrad. Med. 2020, 132, 749–755. [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] [PubMed]
- Marfella, R.; Sardu, C.; D’Onofrio, N.; Prattichizzo, F.; Scisciola, L.; Messina, V.; La Grotta, R.; Balestrieri, M.L.; Maggi, P.; Napoli, C.; et al. Glycaemic control is associated with SARS-CoV-2 breakthrough infections in vaccinated patients with type 2 diabetes. Nat. Commun. 2022, 13, 2318. [Google Scholar] [CrossRef] [PubMed]
- Marfella, R.; D’Onofrio, N.; Sardu, C.; Scisciola, L.; Maggi, P.; Coppola, N.; Romano, C.; Messina, V.; Turriziani, F.; Siniscalchi, M.; et al. Does poor glycaemic control affect the immunogenicity of the COVID-19 vaccination in patients with type 2 diabetes: The CAVEAT study. Diabetes. Obes. Metab. 2022, 24, 160–165. [Google Scholar] [CrossRef]
- Castelo-Branco, L.; Tsourti, Z.; Gennatas, S.; Rogado, J.; Sekacheva, M.; Viñal, D.; Lee, R.; Croitoru, A.; Vitorino, M.; Khallaf, S.; et al. COVID-19 in patients with cancer: First report of the ESMO international, registry-based, cohort study (ESMO-CoCARE). ESMO Open 2022, 7, 100499. [Google Scholar] [CrossRef]
- Lennon, H.; Sperrin, M.; Badrick, E.; Renehan, A.G. The Obesity Paradox in Cancer: A Review. Curr. Oncol. Rep. 2016, 18, 56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Petrelli, F.; Cortellini, A.; Indini, A.; Tomasello, G.; Ghidini, M.; Nigro, O.; Salati, M.; Dottorini, L.; Iaculli, A.; Varricchio, A.; et al. Obesity paradox in patients with cancer: A systematic review and meta-analysis of 6,320,365 patients. medRxiv 2020. [Google Scholar] [CrossRef]
- Romero Starke, K.; Reissig, D.; Petereit-Haack, G.; Schmauder, S.; Nienhaus, A.; Seidler, A. The isolated effect of age on the risk of COVID-19 severe outcomes: A systematic review with meta-analysis. BMJ Glob. Health 2021, 6, e006434. [Google Scholar] [CrossRef]
- Napoli, C.; Tritto, I.; Benincasa, G.; Mansueto, G.; Ambrosio, G. Cardiovascular involvement during COVID-19 and clinical implications in elderly patients. A review. Ann. Med. Surg. 2020, 57, 236–243. [Google Scholar] [CrossRef]
- Napoli, C.; Tritto, I.; Mansueto, G.; Coscioni, E.; Ambrosio, G. Immunosenescence exacerbates the COVID-19. Arch Gerontol. Geriatr. 2020, 90, 104174. [Google Scholar] [CrossRef] [PubMed]
- Sciacchitano, S.; De Vitis, C.; D’Ascanio, M.; Giovagnoli, S.; De Dominicis, C.; Laghi, A.; Anibaldi, P.; Petrucca, A.; Salerno, G.; Santino, I.; et al. Gene signature and immune cell profiling by high-dimensional, single-cell analysis in COVID-19 patients, presenting Low T3 syndrome and coexistent hematological malignancies. J. Transl. Med. 2021, 19, 139. [Google Scholar] [CrossRef] [PubMed]
- Montisci, A.; Vietri, M.T.; Palmieri, V.; Sala, S.; Donatelli, F.; Napoli, C. Cardiac Toxicity Associated with Cancer Immunotherapy and Biological Drugs. Cancers 2021, 13, 4797. [Google Scholar] [CrossRef]
- Montisci, A.; Palmieri, V.; Liu, J.E.; Vietri, M.T.; Cirri, S.; Donatelli, F.; Napoli, C. Severe Cardiac Toxicity Induced by Cancer Therapies Requiring Intensive Care Unit Admission. Front. Cardiovasc. Med. 2021, 8, 713694. [Google Scholar] [CrossRef]
- Bracci, L.; Schiavoni, G.; Sistigu, A.; Belardelli, F. Immune-based mechanisms of cytotoxic chemotherapy: Implications for the design of novel and rationale-based combined treatments against cancer. Cell Death Differ. 2014, 21, 15–25. [Google Scholar] [CrossRef] [Green Version]
- Zitvogel, L.; Apetoh, L.; Ghiringhelli, F.; Kroemer, G. Immunological aspects of cancer chemotherapy. Nat. Rev. Immunol. 2008, 8, 59–73. [Google Scholar] [CrossRef]
- Galluzzi, L.; Zitvogel, L.; Kroemer, G. Immunological Mechanisms Underneath the Efficacy of Cancer Therapy. Cancer Immunol. Res. 2016, 4, 895–902. [Google Scholar] [CrossRef] [Green Version]
- Galluzzi, L.; Buqué, A.; Kepp, O.; Zitvogel, L.; Kroemer, G. Immunological Effects of Conventional Chemotherapy and Targeted Anticancer Agents. Cancer Cell 2015, 28, 690–714. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rasmussen, L.; Arvin, A. Chemotherapy-induced immunosuppression. Environ. Health Perspect. 1982, 43, 21–25. [Google Scholar] [CrossRef]
- Mansueto, G.; Niola, M.; Napoli, C. Can COVID 2019 induce a specific cardiovascular damage or it exacerbates pre-existing cardiovascular diseases? Pathol. Res. Pract. 2020, 216, 153086. [Google Scholar] [CrossRef] [PubMed]
- Xie, Y.; Xu, E.; Bowe, B.; Al-Aly, Z. Long-term cardiovascular outcomes of COVID-19. Nat. Med. 2022, 28, 583–590. [Google Scholar] [CrossRef]
- Zheng, Y.Y.; Ma, Y.T.; Zhang, J.Y.; Xie, X. COVID-19 and the cardiovascular system. Nat. Rev. Cardiol. 2020, 17, 259–260. [Google Scholar] [CrossRef]
- Menna, P.; Paz, O.G.; Chello, M.; Covino, E.; Salvatorelli, E.; Minotti, G. Anthracycline cardiotoxicity. Expert. Opin. Drug. Saf. 2012, 11 (Suppl. S1), S21–S36. [Google Scholar] [CrossRef]
- Pondé, N.F.; Lambertini, M.; de Azambuja, E. Twenty years of anti-HER2 therapy-associated cardiotoxicity. ESMO Open 2016, 1, e000073. [Google Scholar] [CrossRef] [Green Version]
- Furlan, A.; Forner, G.; Cipriani, L.; Vian, E.; Rigoli, R.; Gherlinzoni, F.; Scotton, P. COVID-19 in B Cell-Depleted Patients After Rituximab: A Diagnostic and Therapeutic Challenge. Front. Immunol. 2021, 12, 763412. [Google Scholar] [CrossRef]
- Boekel, L.; Wolbink, G.J. Rituximab during the COVID-19 pandemic: Time to discuss treatment options with patients. Lancet Rheumatol. 2022, 4, e154–e155. [Google Scholar] [CrossRef]
- Yu, M.; Cheng, Y.; Zhang, R.; Wen, T.; Huai, S.; Wei, X.; Zhang, L. Evaluating treatment strategies for non-small cell lung cancer during COVID-19: A propensity score matching analysis. Medicine 2022, 101, e30051. [Google Scholar] [CrossRef]
- Turnquist, C.; Ryan, B.M.; Horikawa, I.; Harris, B.T.; Harris, C.C. Cytokine Storms in Cancer and COVID-19. Cancer Cell 2020, 38, 598–601. [Google Scholar] [CrossRef]
- Shalapour, S.; Karin, M. Immunity, inflammation, and cancer: An eternal fight between good and evil. J. Clin. Investig. 2015, 125, 3347–3355. [Google Scholar] [CrossRef] [Green Version]
- Germolec, D.R.; Shipkowski, K.A.; Frawley, R.P.; Evans, E. Markers of Inflammation. Methods Mol. Biol. 2018, 1803, 57–79. [Google Scholar] [CrossRef]
- Chen, L.; Deng, H.; Cui, H.; Fang, J.; Zuo, Z.; Deng, J.; Li, Y.; Wang, X.; Zhao, L. Inflammatory responses and inflammation-associated diseases in organs. Oncotarget 2018, 9, 7204–7218. [Google Scholar] [CrossRef] [Green Version]
- Altmann, D.M.; Boyton, R.J. SARS-CoV-2 T cell immunity: Specificity, function, durability, and role in protection. Sci. Immunol. 2020, 5, eabd6160. [Google Scholar] [CrossRef]
- Napoli, C.; Benincasa, G.; Criscuolo, C.; Faenza, M.; Liberato, C.; Rusciano, M. Immune reactivity during COVID-19: Implications for treatment. Immunol. Lett. 2021, 231, 28–34. [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.e1039. [Google Scholar] [CrossRef]
- Hadjadj, J.; Yatim, N.; Barnabei, L.; Corneau, A.; Boussier, J.; Smith, N.; Péré, H.; Charbit, B.; Bondet, V.; Chenevier-Gobeaux, C.; et al. Impaired type I interferon activity and inflammatory responses in severe COVID-19 patients. Science 2020, 369, 718–724. [Google Scholar] [CrossRef]
- Rydyznski Moderbacher, C.; Ramirez, S.I.; Dan, J.M.; Grifoni, A.; Hastie, K.M.; Weiskopf, D.; Belanger, S.; Abbott, R.K.; Kim, C.; Choi, J.; et al. Antigen-Specific Adaptive Immunity to SARS-CoV-2 in Acute COVID-19 and Associations with Age and Disease Severity. Cell 2020, 183, 996–1012.e1019. [Google Scholar] [CrossRef]
- Sette, A.; Crotty, S. Adaptive immunity to SARS-CoV-2 and COVID-19. Cell 2021, 184, 861–880. [Google Scholar] [CrossRef]
- Prager, I.; Watzl, C. Mechanisms of natural killer cell-mediated cellular cytotoxicity. J. Leukoc. Biol. 2019, 105, 1319–1329. [Google Scholar] [CrossRef]
- Janeway, C.A. How the immune system protects the host from infection. Microbes. Infect. 2001, 3, 1167–1171. [Google Scholar] [CrossRef]
- Kim, K.D.; Zhao, J.; Auh, S.; Yang, X.; Du, P.; Tang, H.; Fu, Y.X. Adaptive immune cells temper initial innate responses. Nat. Med. 2007, 13, 1248–1252. [Google Scholar] [CrossRef]
- Jordan, S.C. Innate and adaptive immune responses to SARS-CoV-2 in humans: Relevance to acquired immunity and vaccine responses. Clin. Exp. Immunol. 2021, 204, 310–320. [Google Scholar] [CrossRef]
- Grifoni, A.; Weiskopf, D.; Ramirez, S.I.; Mateus, J.; Dan, J.M.; Moderbacher, C.R.; Rawlings, S.A.; Sutherland, A.; Premkumar, L.; Jadi, R.S.; et al. Targets of T Cell Responses to SARS-CoV-2 Coronavirus in Humans with COVID-19 Disease and Unexposed Individuals. Cell 2020, 181, 1489–1501.e1415. [Google Scholar] [CrossRef]
- Sekine, T.; Perez-Potti, A.; Rivera-Ballesteros, O.; Strålin, K.; Gorin, J.B.; Olsson, A.; Llewellyn-Lacey, S.; Kamal, H.; Bogdanovic, G.; Muschiol, S.; et al. Robust T Cell Immunity in Convalescent Individuals with Asymptomatic or Mild COVID-19. Cell 2020, 183, 158–168.e114. [Google Scholar] [CrossRef] [PubMed]
- Liao, M.; Liu, Y.; Yuan, J.; Wen, Y.; Xu, G.; Zhao, J.; Cheng, L.; Li, J.; Wang, X.; Wang, F.; et al. Single-cell landscape of bronchoalveolar immune cells in patients with COVID-19. Nat. Med. 2020, 26, 842–844. [Google Scholar] [CrossRef] [PubMed]
- Zhou, R.; To, K.K.; Wong, Y.C.; Liu, L.; Zhou, B.; Li, X.; Huang, H.; Mo, Y.; Luk, T.Y.; Lau, T.T.; et al. Acute SARS-CoV-2 Infection Impairs Dendritic Cell and T Cell Responses. Immunity 2020, 53, 864–877.e865. [Google Scholar] [CrossRef] [PubMed]
- Bergamaschi, L.; Mescia, F.; Turner, L.; Hanson, A.L.; Kotagiri, P.; Dunmore, B.J.; Ruffieux, H.; De Sa, A.; Huhn, O.; Morgan, M.D.; et al. Longitudinal analysis reveals that delayed bystander CD8+ T cell activation and early immune pathology distinguish severe COVID-19 from mild disease. Immunity 2021, 54, 1257–1275.e1258. [Google Scholar] [CrossRef]
- Notarbartolo, S.; Ranzani, V.; Bandera, A.; Gruarin, P.; Bevilacqua, V.; Putignano, A.R.; Gobbini, A.; Galeota, E.; Manara, C.; Bombaci, M.; et al. Integrated longitudinal immunophenotypic, transcriptional and repertoire analyses delineate immune responses in COVID-19 patients. Sci. Immunol. 2021, 6, abg5021. [Google Scholar] [CrossRef]
- Braun, J.; Loyal, L.; Frentsch, M.; Wendisch, D.; Georg, P.; Kurth, F.; Hippenstiel, S.; Dingeldey, M.; Kruse, B.; Fauchere, F.; et al. SARS-CoV-2-reactive T cells in healthy donors and patients with COVID-19. Nature 2020, 587, 270–274. [Google Scholar] [CrossRef]
- Tan, A.T.; Linster, M.; Tan, C.W.; Le Bert, N.; Chia, W.N.; Kunasegaran, K.; Zhuang, Y.; Tham, C.Y.L.; Chia, A.; Smith, G.J.D.; et al. Early induction of functional SARS-CoV-2-specific T cells associates with rapid viral clearance and mild disease in COVID-19 patients. Cell Rep. 2021, 34, 108728. [Google Scholar] [CrossRef]
- Schulien, I.; Kemming, J.; Oberhardt, V.; Wild, K.; Seidel, L.M.; Killmer, S.; Sagar; Daul, F.; Salvat Lago, M.; Decker, A.; et al. Characterization of pre-existing and induced SARS-CoV-2-specific CD8. Nat. Med. 2021, 27, 78–85. [Google Scholar] [CrossRef]
- Grimaldi, V.; Benincasa, G.; Moccia, G.; Sansone, A.; Signoriello, G.; Napoli, C. Evaluation of circulating leucocyte populations both in subjects with previous SARS-CoV-2 infection and in healthy subjects after vaccination. J. Immunol. Methods 2022, 502, 113230. [Google Scholar] [CrossRef]
- 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]
- Stephen-Victor, E.; Das, M.; Karnam, A.; Pitard, B.; Gautier, J.F.; Bayry, J. Potential of regulatory T-cell-based therapies in the management of severe COVID-19. Eur. Respir. J. 2020, 56, 2002182. [Google Scholar] [CrossRef] [PubMed]
- Neumann, J.; Prezzemolo, T.; Vanderbeke, L.; Roca, C.P.; Gerbaux, M.; Janssens, S.; Willemsen, M.; Burton, O.; Van Mol, P.; Van Herck, Y.; et al. Increased IL-10-producing regulatory T cells are characteristic of severe cases of COVID-19. Clin. Transl. Immunol. 2020, 9, e1204. [Google Scholar] [CrossRef] [PubMed]
- Tan, M.; Liu, Y.; Zhou, R.; Deng, X.; Li, F.; Liang, K.; Shi, Y. Immunopathological characteristics of coronavirus disease 2019 cases in Guangzhou, China. Immunology 2020, 160, 261–268. [Google Scholar] [CrossRef] [PubMed]
- Virgin, H.W.; Wherry, E.J.; Ahmed, R. Redefining chronic viral infection. Cell 2009, 138, 30–50. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Huang, C.; Wang, Y.; Li, X.; Ren, L.; Zhao, J.; Hu, Y.; Zhang, L.; Fan, G.; Xu, J.; Gu, X.; et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 2020, 395, 497–506. [Google Scholar] [CrossRef] [Green Version]
- McGonagle, D.; Sharif, K.; O’Regan, A.; Bridgewood, C. The Role of Cytokines including Interleukin-6 in COVID-19 induced Pneumonia and Macrophage Activation Syndrome-Like Disease. Autoimmun. Rev. 2020, 19, 102537. [Google Scholar] [CrossRef]
- Murthy, H.; Iqbal, M.; Chavez, J.C.; Kharfan-Dabaja, M.A. Cytokine Release Syndrome: Current Perspectives. Immunotargets Ther. 2019, 8, 43–52. [Google Scholar] [CrossRef] [Green Version]
- Behrens, E.M.; Koretzky, G.A. Review: Cytokine Storm Syndrome: Looking Toward the Precision Medicine Era. Arthritis Rheumatol. 2017, 69, 1135–1143. [Google Scholar] [CrossRef] [Green Version]
- Del Valle, D.M.; Kim-Schulze, S.; Huang, H.H.; Beckmann, N.D.; Nirenberg, S.; Wang, B.; Lavin, Y.; Swartz, T.H.; Madduri, D.; Stock, A.; et al. An inflammatory cytokine signature predicts COVID-19 severity and survival. Nat. Med. 2020, 26, 1636–1643. [Google Scholar] [CrossRef]
- Chen, G.; Wu, D.; Guo, W.; Cao, Y.; Huang, D.; Wang, H.; Wang, T.; Zhang, X.; Chen, H.; Yu, H.; et al. Clinical and immunological features of severe and moderate coronavirus disease 2019. J Clin Invest 2020, 130, 2620–2629. [Google Scholar] [CrossRef]
- Liu, Y.; Zhang, C.; Huang, F.; Yang, Y.; Wang, F.; Yuan, J.; Zhang, Z.; Qin, Y.; Li, X.; Zhao, D.; et al. Elevated plasma levels of selective cytokines in COVID-19 patients reflect viral load and lung injury. Natl. Sci. Rev. 2020, 7, 1003–1011. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Narazaki, M.; Kishimoto, T. The Two-Faced Cytokine IL-6 in Host Defense and Diseases. Int. J. Mol. Sci. 2018, 19, 3528. [Google Scholar] [CrossRef] [Green Version]
- Arnaldez, F.I.; O’Day, S.J.; Drake, C.G.; Fox, B.A.; Fu, B.; Urba, W.J.; Montesarchio, V.; Weber, J.S.; Wei, H.; Wigginton, J.M.; et al. The Society for Immunotherapy of Cancer perspective on regulation of interleukin-6 signaling in COVID-19-related systemic inflammatory response. J. Immunother. Cancer 2020, 8, e000930. [Google Scholar] [CrossRef]
- Kumari, N.; Dwarakanath, B.S.; Das, A.; Bhatt, A.N. Role of interleukin-6 in cancer progression and therapeutic resistance. Tumour. Biol. 2016, 37, 11553–11572. [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] [PubMed]
- Liu, Z.; Li, J.; Chen, D.; Gao, R.; Zeng, W.; Chen, S.; Huang, Y.; Huang, J.; Long, W.; Li, M.; et al. Dynamic Interleukin-6 Level Changes as a Prognostic Indicator in Patients With COVID-19. Front. Pharmacol. 2020, 11, 1093. [Google Scholar] [CrossRef]
- Mehta, A.K.; Gracias, D.T.; Croft, M. TNF activity and T cells. Cytokine 2018, 101, 14–18. [Google Scholar] [CrossRef]
- Pasquereau, S.; Kumar, A.; Herbein, G. Targeting TNF and TNF Receptor Pathway in HIV-1 Infection: From Immune Activation to Viral Reservoirs. Viruses 2017, 9, 64. [Google Scholar] [CrossRef]
- Popivanova, B.K.; Kitamura, K.; Wu, Y.; Kondo, T.; Kagaya, T.; Kaneko, S.; Oshima, M.; Fujii, C.; Mukaida, N. Blocking TNF-alpha in mice reduces colorectal carcinogenesis associated with chronic colitis. J. Clin. Investig. 2008, 118, 560–570. [Google Scholar] [CrossRef] [PubMed]
- Zhaorigetu, S.; Yanaka, N.; Sasaki, M.; Watanabe, H.; Kato, N. Silk protein, sericin, suppresses DMBA-TPA-induced mouse skin tumorigenesis by reducing oxidative stress, inflammatory responses and endogenous tumor promoter TNF-alpha. Oncol. Rep. 2003, 10, 537–543. [Google Scholar] [PubMed]
- Berberoglu, U.; Yildirim, E.; Celen, O. Serum levels of tumor necrosis factor alpha correlate with response to neoadjuvant chemotherapy in locally advanced breast cancer. Int. J. Biol. Markers 2004, 19, 130–134. [Google Scholar] [CrossRef] [PubMed]
- Michalaki, V.; Syrigos, K.; Charles, P.; Waxman, J. Serum levels of IL-6 and TNF-alpha correlate with clinicopathological features and patient survival in patients with prostate cancer. Br. J. Cancer 2004, 90, 2312–2316. [Google Scholar] [CrossRef]
- Ferrajoli, A.; Keating, M.J.; Manshouri, T.; Giles, F.J.; Dey, A.; Estrov, Z.; Koller, C.A.; Kurzrock, R.; Thomas, D.A.; Faderl, S.; et al. The clinical significance of tumor necrosis factor-alpha plasma level in patients having chronic lymphocytic leukemia. Blood 2002, 100, 1215–1219. [Google Scholar] [CrossRef] [PubMed]
- Szlosarek, P.W.; Grimshaw, M.J.; Kulbe, H.; Wilson, J.L.; Wilbanks, G.D.; Burke, F.; Balkwill, F.R. Expression and regulation of tumor necrosis factor alpha in normal and malignant ovarian epithelium. Mol. Cancer Ther. 2006, 5, 382–390. [Google Scholar] [CrossRef] [Green Version]
- Mulchandani, R.; Lyngdoh, T.; Kakkar, A.K. Deciphering the COVID-19 cytokine storm: Systematic review and meta-analysis. Eur. J. Clin. Investig. 2021, 51, e13429. [Google Scholar] [CrossRef]
- Pestka, S.; Krause, C.D.; Walter, M.R. Interferons, interferon-like cytokines, and their receptors. Immunol. Rev. 2004, 202, 8–32. [Google Scholar] [CrossRef]
- Lee, A.J.; Ashkar, A.A. The Dual Nature of Type I and Type II Interferons. Front. Immunol. 2018, 9, 2061. [Google Scholar] [CrossRef] [Green Version]
- Witte, K.; Witte, E.; Sabat, R.; Wolk, K. IL-28A, IL-28B, and IL-29: Promising cytokines with type I interferon-like properties. Cytokine Growth Factor Rev. 2010, 21, 237–251. [Google Scholar] [CrossRef] [PubMed]
- Snell, L.M.; McGaha, T.L.; Brooks, D.G. Type I Interferon in Chronic Virus Infection and Cancer. Trends Immunol. 2017, 38, 542–557. [Google Scholar] [CrossRef]
- McNab, F.; Mayer-Barber, K.; Sher, A.; Wack, A.; O’Garra, A. Type I interferons in infectious disease. Nat. Rev. Immunol. 2015, 15, 87–103. [Google Scholar] [CrossRef]
- Musella, M.; Manic, G.; De Maria, R.; Vitale, I.; Sistigu, A. Type-I-interferons in infection and cancer: Unanticipated dynamics with therapeutic implications. Oncoimmunology 2017, 6, e1314424. [Google Scholar] [CrossRef] [Green Version]
- Aricò, E.; Castiello, L.; Capone, I.; Gabriele, L.; Belardelli, F. Type I Interferons and Cancer: An Evolving Story Demanding Novel Clinical Applications. Cancers 2019, 11, 1943. [Google Scholar] [CrossRef] [Green Version]
- Schreiber, R.D.; Old, L.J.; Smyth, M.J. Cancer immunoediting: Integrating immunity’s roles in cancer suppression and promotion. Science 2011, 331, 1565–1570. [Google Scholar] [CrossRef] [Green Version]
- Zitvogel, L.; Galluzzi, L.; Kepp, O.; Smyth, M.J.; Kroemer, G. Type I interferons in anticancer immunity. Nat. Rev. Immunol. 2015, 15, 405–414. [Google Scholar] [CrossRef]
- Gupta, R. The double edged interferon riddle in COVID-19 pathogenesis. Crit. Care 2020, 24, 631. [Google Scholar] [CrossRef] [PubMed]
- Lopez, L.; Sang, P.C.; Tian, Y.; Sang, Y. Dysregulated Interferon Response Underlying Severe COVID-19. Viruses 2020, 12, 1433. [Google Scholar] [CrossRef]
- Israelow, B.; Song, E.; Mao, T.; Lu, P.; Meir, A.; Liu, F.; Alfajaro, M.M.; Wei, J.; Dong, H.; Homer, R.J.; et al. Mouse model of SARS-CoV-2 reveals inflammatory role of type I interferon signaling. J. Exp. Med. 2020, 217, e20201241. [Google Scholar] [CrossRef]
- Liu, T.; Zhang, L.; Joo, D.; Sun, S.C. NF-κB signaling in inflammation. Signal Transduct. Target. Ther. 2017, 2, 17023. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Oeckinghaus, A.; Hayden, M.S.; Ghosh, S. Crosstalk in NF-κB signaling pathways. Nat. Immunol. 2011, 12, 695–708. [Google Scholar] [CrossRef]
- Yu, H.; Lin, L.; Zhang, Z.; Zhang, H.; Hu, H. Targeting NF-κB pathway for the therapy of diseases: Mechanism and clinical study. Signal Transduct. Target. Ther. 2020, 5, 209. [Google Scholar] [CrossRef]
- Moynagh, P.N. The NF-kappaB pathway. J Cell Sci 2005, 118, 4589–4592. [Google Scholar] [CrossRef] [Green Version]
- Neufeldt, C.J.; Cerikan, B.; Cortese, M.; Frankish, J.; Lee, J.Y.; Plociennikowska, A.; Heigwer, F.; Prasad, V.; Joecks, S.; Burkart, S.S.; et al. SARS-CoV-2 infection induces a pro-inflammatory cytokine response through cGAS-STING and NF-κB. Commun. Biol. 2022, 5, 45. [Google Scholar] [CrossRef]
- Su, C.M.; Wang, L.; Yoo, D. Activation of NF-κB and induction of proinflammatory cytokine expressions mediated by ORF7a protein of SARS-CoV-2. Sci. Rep. 2021, 11, 13464. [Google Scholar] [CrossRef]
- Catrysse, L.; van Loo, G. Inflammation and the Metabolic Syndrome: The Tissue-Specific Functions of NF-κB. Trends Cell Biol. 2017, 27, 417–429. [Google Scholar] [CrossRef]
- Apicella, M.; Campopiano, M.C.; Mantuano, M.; Mazoni, L.; Coppelli, A.; Del Prato, S. COVID-19 in people with diabetes: Understanding the reasons for worse outcomes. Lancet Diabetes Endocrinol. 2020, 8, 782–792. [Google Scholar] [CrossRef]
- Popkin, B.M.; Du, S.; Green, W.D.; Beck, M.A.; Algaith, T.; Herbst, C.H.; Alsukait, R.F.; Alluhidan, M.; Alazemi, N.; Shekar, M. Individuals with obesity and COVID-19: A global perspective on the epidemiology and biological relationships. Obes. Rev. 2020, 21, e13128. [Google Scholar] [CrossRef]
- Gilmore, T.D. Multiple mutations contribute to the oncogenicity of the retroviral oncoprotein v-Rel. Oncogene 1999, 18, 6925–6937. [Google Scholar] [CrossRef] [Green Version]
- Gilmore, T.D.; Kalaitzidis, D.; Liang, M.C.; Starczynowski, D.T. The c-Rel transcription factor and B-cell proliferation: A deal with the devil. Oncogene 2004, 23, 2275–2286. [Google Scholar] [CrossRef] [Green Version]
- Neri, A.; Chang, C.C.; Lombardi, L.; Salina, M.; Corradini, P.; Maiolo, A.T.; Chaganti, R.S.; Dalla-Favera, R. B cell lymphoma-associated chromosomal translocation involves candidate oncogene lyt-10, homologous to NF-kappa B p50. Cell 1991, 67, 1075–1087. [Google Scholar] [CrossRef]
- Bredel, M.; Scholtens, D.M.; Yadav, A.K.; Alvarez, A.A.; Renfrow, J.J.; Chandler, J.P.; Yu, I.L.; Carro, M.S.; Dai, F.; Tagge, M.J.; et al. NFKBIA deletion in glioblastomas. N. Engl. J. Med. 2011, 364, 627–637. [Google Scholar] [CrossRef]
- Greenman, C.; Stephens, P.; Smith, R.; Dalgliesh, G.L.; Hunter, C.; Bignell, G.; Davies, H.; Teague, J.; Butler, A.; Stevens, C.; et al. Patterns of somatic mutation in human cancer genomes. Nature 2007, 446, 153–158. [Google Scholar] [CrossRef] [Green Version]
- Pflueger, D.; Terry, S.; Sboner, A.; Habegger, L.; Esgueva, R.; Lin, P.C.; Svensson, M.A.; Kitabayashi, N.; Moss, B.J.; MacDonald, T.Y.; et al. Discovery of non-ETS gene fusions in human prostate cancer using next-generation RNA sequencing. Genome Res. 2011, 21, 56–67. [Google Scholar] [CrossRef] [Green Version]
- Boehm, J.S.; Zhao, J.J.; Yao, J.; Kim, S.Y.; Firestein, R.; Dunn, I.F.; Sjostrom, S.K.; Garraway, L.A.; Weremowicz, S.; Richardson, A.L.; et al. Integrative genomic approaches identify IKBKE as a breast cancer oncogene. Cell 2007, 129, 1065–1079. [Google Scholar] [CrossRef] [Green Version]
- Terzić, J.; Grivennikov, S.; Karin, E.; Karin, M. Inflammation and colon cancer. Gastroenterology 2010, 138, 2101–2114.e2105. [Google Scholar] [CrossRef]
- Pikarsky, E.; Porat, R.M.; Stein, I.; Abramovitch, R.; Amit, S.; Kasem, S.; Gutkovich-Pyest, E.; Urieli-Shoval, S.; Galun, E.; Ben-Neriah, Y. NF-kappaB functions as a tumour promoter in inflammation-associated cancer. Nature 2004, 431, 461–466. [Google Scholar] [CrossRef]
- Hagemann, T.; Lawrence, T.; McNeish, I.; Charles, K.A.; Kulbe, H.; Thompson, R.G.; Robinson, S.C.; Balkwill, F.R. “Re-educating” tumor-associated macrophages by targeting NF-kappaB. J. Exp. Med. 2008, 205, 1261–1268. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Darnell, J.E. STATs and gene regulation. Science 1997, 277, 1630–1635. [Google Scholar] [CrossRef]
- O’Shea, J.J.; Schwartz, D.M.; Villarino, A.V.; Gadina, M.; McInnes, I.B.; Laurence, A. The JAK-STAT pathway: Impact on human disease and therapeutic intervention. Annu. Rev. Med. 2015, 66, 311–328. [Google Scholar] [CrossRef] [Green Version]
- Owen, K.L.; Brockwell, N.K.; Parker, B.S. JAK-STAT Signaling: A Double-Edged Sword of Immune Regulation and Cancer Progression. Cancers 2019, 11, 2002. [Google Scholar] [CrossRef] [Green Version]
- Aittomäki, S.; Pesu, M. Therapeutic targeting of the Jak/STAT pathway. Basic Clin. Pharmacol. Toxicol. 2014, 114, 18–23. [Google Scholar] [CrossRef]
- Vainchenker, W.; Constantinescu, S.N. JAK/STAT signaling in hematological malignancies. Oncogene 2013, 32, 2601–2613. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vainchenker, W.; Delhommeau, F.; Constantinescu, S.N.; Bernard, O.A. New mutations and pathogenesis of myeloproliferative neoplasms. Blood 2011, 118, 1723–1735. [Google Scholar] [CrossRef]
- Pardanani, A.; Lasho, T.L.; Finke, C.; Hanson, C.A.; Tefferi, A. Prevalence and clinicopathologic correlates of JAK2 exon 12 mutations in JAK2V617F-negative polycythemia vera. Leukemia 2007, 21, 1960–1963. [Google Scholar] [CrossRef]
- Koskela, H.L.; Eldfors, S.; Ellonen, P.; van Adrichem, A.J.; Kuusanmäki, H.; Andersson, E.I.; Lagström, S.; Clemente, M.J.; Olson, T.; Jalkanen, S.E.; et al. Somatic STAT3 mutations in large granular lymphocytic leukemia. N. Engl. J. Med. 2012, 366, 1905–1913. [Google Scholar] [CrossRef] [Green Version]
- Xu, Y.H.; Lu, S. A meta-analysis of STAT3 and phospho-STAT3 expression and survival of patients with non-small-cell lung cancer. Eur. J. Surg. Oncol. 2014, 40, 311–317. [Google Scholar] [CrossRef]
- Mirtti, T.; Leiby, B.E.; Abdulghani, J.; Aaltonen, E.; Pavela, M.; Mamtani, A.; Alanen, K.; Egevad, L.; Granfors, T.; Josefsson, A.; et al. Nuclear Stat5a/b predicts early recurrence and prostate cancer-specific death in patients treated by radical prostatectomy. Hum. Pathol. 2013, 44, 310–319. [Google Scholar] [CrossRef] [Green Version]
- Macha, M.A.; Matta, A.; Kaur, J.; Chauhan, S.S.; Thakar, A.; Shukla, N.K.; Gupta, S.D.; Ralhan, R. Prognostic significance of nuclear pSTAT3 in oral cancer. Head Neck. 2011, 33, 482–489. [Google Scholar] [CrossRef]
- Messina, J.L.; Yu, H.; Riker, A.I.; Munster, P.N.; Jove, R.L.; Daud, A.I. Activated stat-3 in melanoma. Cancer Control 2008, 15, 196–201. [Google Scholar] [CrossRef] [Green Version]
- Sonnenblick, A.; Uziely, B.; Nechushtan, H.; Kadouri, L.; Galun, E.; Axelrod, J.H.; Katz, D.; Daum, H.; Hamburger, T.; Maly, B.; et al. Tumor STAT3 tyrosine phosphorylation status, as a predictor of benefit from adjuvant chemotherapy for breast cancer. Breast Cancer Res. Treat. 2013, 138, 407–413. [Google Scholar] [CrossRef]
- Kusaba, T.; Nakayama, T.; Yamazumi, K.; Yakata, Y.; Yoshizaki, A.; Inoue, K.; Nagayasu, T.; Sekine, I. Activation of STAT3 is a marker of poor prognosis in human colorectal cancer. Oncol. Rep. 2006, 15, 1445–1451. [Google Scholar] [CrossRef]
- Chen, D.Y.; Khan, N.; Close, B.J.; Goel, R.K.; Blum, B.; Tavares, A.H.; Kenney, D.; Conway, H.L.; Ewoldt, J.K.; Chitalia, V.C.; et al. SARS-CoV-2 Disrupts Proximal Elements in the JAK-STAT Pathway. J. Virol. 2021, 95, e0086221. [Google Scholar] [CrossRef] [PubMed]
- Huang, P.W.; Chang, J.W. Immune checkpoint inhibitors win the 2018 Nobel Prize. Biomed. J. 2019, 42, 299–306. [Google Scholar] [CrossRef]
- Gajewski, T.F.; Schreiber, H.; Fu, Y.X. Innate and adaptive immune cells in the tumor microenvironment. Nat. Immunol. 2013, 14, 1014–1022. [Google Scholar] [CrossRef] [Green Version]
- Zhao, Q.; Meng, M.; Kumar, R.; Wu, Y.; Huang, J.; Deng, Y.; Weng, Z.; Yang, L. Lymphopenia is associated with severe coronavirus disease 2019 (COVID-19) infections: A systemic review and meta-analysis. Int. J. Infect. Dis. 2020, 96, 131–135. [Google Scholar] [CrossRef]
- Diao, B.; Wang, C.; Tan, Y.; Chen, X.; Liu, Y.; Ning, L.; Chen, L.; Li, M.; Wang, G.; Yuan, Z.; et al. Reduction and Functional Exhaustion of T Cells in Patients With Coronavirus Disease 2019 (COVID-19). Front. Immunol. 2020, 11, 827. [Google Scholar] [CrossRef]
- Sabbatino, F.; Conti, V.; Franci, G.; Sellitto, C.; Manzo, V.; Pagliano, P.; De Bellis, E.; Masullo, A.; Salzano, F.A.; Caputo, A.; et al. PD-L1 Dysregulation in COVID-19 Patients. Front. Immunol. 2021, 12, 695242. [Google Scholar] [CrossRef]
- Kraus, W.L. PARPs and ADP-Ribosylation: 50 Years… and Counting. Mol. Cell 2015, 58, 902–910. [Google Scholar] [CrossRef] [Green Version]
- Grignani, G.; Merlini, A.; Sangiolo, D.; D’Ambrosio, L.; Pignochino, Y. Delving into PARP inhibition from bench to bedside and back. Pharmacol. Ther. 2020, 206, 107446. [Google Scholar] [CrossRef]
- Alhammad, Y.M.O.; Kashipathy, M.M.; Roy, A.; Gagné, J.-P.; McDonald, P.; Gao, P.; Nonfoux, L.; Battaile, K.P.; Johnson, D.K.; Holmstrom, E.D.; et al. The SARS-CoV-2 Conserved Macrodomain Is a Mono-ADP-Ribosylhydrolase. J. Virol. 2021, 95, e01969-20. [Google Scholar] [CrossRef]
- Fehr, A.R.; Singh, S.A.; Kerr, C.M.; Mukai, S.; Higashi, H.; Aikawa, M. The impact of PARPs and ADP-ribosylation on inflammation and host-pathogen interactions. Genes Dev. 2020, 34, 341–359. [Google Scholar] [CrossRef]
- Cohen, M.S. Interplay between compartmentalized NAD. Genes Dev. 2020, 34, 254–262. [Google Scholar] [CrossRef] [PubMed]
- Hottiger, M.O. Nuclear ADP-Ribosylation and Its Role in Chromatin Plasticity, Cell Differentiation, and Epigenetics. Annu. Rev. Biochem. 2015, 84, 227–263. [Google Scholar] [CrossRef] [PubMed]
- Ryu, K.W.; Kim, D.S.; Kraus, W.L. New facets in the regulation of gene expression by ADP-ribosylation and poly(ADP-ribose) polymerases. Chem. Rev. 2015, 115, 2453–2481. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gupte, R.; Liu, Z.; Kraus, W.L. PARPs and ADP-ribosylation: Recent advances linking molecular functions to biological outcomes. Genes Dev. 2017, 31, 101–126. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lord, C.J.; Ashworth, A. PARP inhibitors: Synthetic lethality in the clinic. Science 2017, 355, 1152–1158. [Google Scholar] [CrossRef] [PubMed]
- Lippitz, B.E. Cytokine patterns in patients with cancer: A systematic review. Lancet Oncol. 2013, 14, e218–e228. [Google Scholar] [CrossRef]
- Salesi, M.; Shojaie, B.; Farajzadegan, Z.; Salesi, N.; Mohammadi, E. TNF-α Blockers Showed Prophylactic Effects in Preventing COVID-19 in Patients with Rheumatoid Arthritis and Seronegative Spondyloarthropathies: A Case-Control Study. Rheumatol. Ther. 2021, 8, 1355–1370. [Google Scholar] [CrossRef]
- Wise, J. COVID-19: Arthritis drugs improve survival in intensive care patients, shows study. BMJ 2021, 372, n61. [Google Scholar] [CrossRef]
- Quesada, J.R.; Reuben, J.; Manning, J.T.; Hersh, E.M.; Gutterman, J.U. Alpha interferon for induction of remission in hairy-cell leukemia. N. Engl. J. Med. 1984, 310, 15–18. [Google Scholar] [CrossRef]
- Foon, K.A.; Sherwin, S.A.; Abrams, P.G.; Longo, D.L.; Fer, M.F.; Stevenson, H.C.; Ochs, J.J.; Bottino, G.C.; Schoenberger, C.S.; Zeffren, J. Treatment of advanced non-Hodgkin’s lymphoma with recombinant leukocyte A interferon. N. Engl. J. Med. 1984, 311, 1148–1152. [Google Scholar] [CrossRef]
- Eggermont, A.M.; Suciu, S.; Santinami, M.; Testori, A.; Kruit, W.H.; Marsden, J.; Punt, C.J.; Salès, F.; Gore, M.; MacKie, R.; et al. Adjuvant therapy with pegylated interferon alfa-2b versus observation alone in resected stage III melanoma: Final results of EORTC 18991, a randomised phase III trial. Lancet 2008, 372, 117–126. [Google Scholar] [CrossRef] [Green Version]
- Smits, E.L.; Anguille, S.; Berneman, Z.N. Interferon α may be back on track to treat acute myeloid leukemia. Oncoimmunology 2013, 2, e23619. [Google Scholar] [CrossRef] [Green Version]
- Inoue, M.; Hisasue, S.; Nagae, M.; China, T.; Saito, K.; Isotani, S.; Yamaguchi, R.; Ide, H.; Muto, S.; Horie, S. Interferon-α Treatment for Growing Teratoma Syndrome of the Testis. Case Rep. Nephrol. Urol. 2013, 3, 40–45. [Google Scholar] [CrossRef]
- Radesi-Sarghi, S.; Arbion, F.; Dartigeas, C.; Delain, M.; Benboubker, L.; Hérault, O.; Colombat, P.; Gyan, E. Interferon alpha with or without rituximab achieves a high response rate and durable responses in relapsed FL: 17 years’ experience in a single centre. Ann. Hematol. 2014, 93, 147–156. [Google Scholar] [CrossRef]
- Monk, P.D.; Marsden, R.J.; Tear, V.J.; Brookes, J.; Batten, T.N.; Mankowski, M.; Gabbay, F.J.; Davies, D.E.; Holgate, S.T.; Ho, L.P.; et al. Safety and efficacy of inhaled nebulised interferon beta-1a (SNG001) for treatment of SARS-CoV-2 infection: A randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Respir. Med. 2021, 9, 196–206. [Google Scholar] [CrossRef]
- Peiffer-Smadja, N.; Yazdanpanah, Y. Nebulised interferon beta-1a for patients with COVID-19. Lancet Respir. Med. 2021, 9, 122–123. [Google Scholar] [CrossRef]
- Nakhlband, A.; Fakhari, A.; Azizi, H. Interferon-beta offers promising avenues to COVID-19 treatment: A systematic review and meta-analysis of clinical trial studies. Naunyn Schmiedebergs Arch Pharmacol. 2021, 394, 829–838. [Google Scholar] [CrossRef] [PubMed]
- Wong, C.K.H.; Wan, E.Y.F.; Luo, S.; Ding, Y.; Lau, E.H.Y.; Ling, P.; Hu, X.; Lau, E.C.H.; Wong, J.; Zheng, X.; et al. Clinical outcomes of different therapeutic options for COVID-19 in two Chinese case cohorts: A propensity-score analysis. eClinicalMedicine 2021, 32, 100743. [Google Scholar] [CrossRef]
- Wang, N.; Zhan, Y.; Zhu, L.; Hou, Z.; Liu, F.; Song, P.; Qiu, F.; Wang, X.; Zou, X.; Wan, D.; et al. Retrospective Multicenter Cohort Study Shows Early Interferon Therapy Is Associated with Favorable Clinical Responses in COVID-19 Patients. Cell Host Microbe 2020, 28, 455–464.e452. [Google Scholar] [CrossRef]
- AIFA. Medicine Usable for Treatment of COVID-19 Disease. Available online: https://www.aifa.gov.it/en/aggiornamento-sui-farmaci-utilizzabili-per-il-trattamento-della-malattia-covid19 (accessed on 22 July 2022).
- Yanaihara, N.; Hirata, Y.; Yamaguchi, N.; Noguchi, Y.; Saito, M.; Nagata, C.; Takakura, S.; Yamada, K.; Okamoto, A. Antitumor effects of interleukin-6 (IL-6)/interleukin-6 receptor (IL-6R) signaling pathway inhibition in clear cell carcinoma of the ovary. Mol. Carcinog. 2016, 55, 832–841. [Google Scholar] [CrossRef] [PubMed]
- Goumas, F.A.; Holmer, R.; Egberts, J.H.; Gontarewicz, A.; Heneweer, C.; Geisen, U.; Hauser, C.; Mende, M.M.; Legler, K.; Röcken, C.; et al. Inhibition of IL-6 signaling significantly reduces primary tumor growth and recurrencies in orthotopic xenograft models of pancreatic cancer. Int. J. Cancer 2015, 137, 1035–1046. [Google Scholar] [CrossRef] [PubMed]
- Grivennikov, S.; Karin, E.; Terzic, J.; Mucida, D.; Yu, G.Y.; Vallabhapurapu, S.; Scheller, J.; Rose-John, S.; Cheroutre, H.; Eckmann, L.; et al. IL-6 and Stat3 are required for survival of intestinal epithelial cells and development of colitis-associated cancer. Cancer Cell 2009, 15, 103–113. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sarfraz, A.; Sarfraz, Z.; Sarfraz, M.; Aftab, H.; Pervaiz, Z. Tocilizumab and COVID-19: A meta-analysis of 2120 patients with severe disease and implications for clinical trial methodologies. Turk. J. Med. Sci. 2021, 51, 890–897. [Google Scholar] [CrossRef] [PubMed]
- Durán-Méndez, A.; Aguilar-Arroyo, A.D.; Vivanco-Gómez, E.; Nieto-Ortega, E.; Pérez-Ortega, D.; Jiménez-Pérez, C.; Hernández-Skewes, K.Y.; Montiel-Bravo, G.; Roque-Reyes, O.J.; Romero-Lechuga, F.; et al. Tocilizumab reduces COVID-19 mortality and pathology in a dose and timing-dependent fashion: A multi-centric study. Sci. Rep. 2021, 11, 19728. [Google Scholar] [CrossRef] [PubMed]
- Bose, P.; Verstovsek, S. JAK2 inhibitors for myeloproliferative neoplasms: What is next? Blood 2017, 130, 115–125. [Google Scholar] [CrossRef] [Green Version]
- Becker, H.; Engelhardt, M.; von Bubnoff, N.; Wäsch, R. Ruxolitinib. Recent Results Cancer Res. 2014, 201, 249–257. [Google Scholar] [CrossRef]
- Dougados, M.; van der Heijde, D.; Chen, Y.C.; Greenwald, M.; Drescher, E.; Liu, J.; Beattie, S.; Witt, S.; de la Torre, I.; Gaich, C.; et al. Baricitinib in patients with inadequate response or intolerance to conventional synthetic DMARDs: Results from the RA-BUILD study. Ann. Rheum. Dis. 2017, 76, 88–95. [Google Scholar] [CrossRef]
- Schwartz, D.M.; Bonelli, M.; Gadina, M.; O’Shea, J.J. Type I/II cytokines, JAKs, and new strategies for treating autoimmune diseases. Nat. Rev. Rheumatol. 2016, 12, 25–36. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- WHO. WHO Recommends Two New Drugs to Treat COVID-19. Available online: https://www.who.int/news/item/14-01-2022-who-recommends-two-new-drugs-to-treat-covid-19 (accessed on 22 July 2022).
- Iastrebner, M.; Castro, J.; García Espina, E.; Lettieri, C.; Payaslian, S.; Cuesta, M.C.; Gutiérrez Fernández, P.; Mandrile, A.; Contreras, A.P.; Gervasoni, S.; et al. Ruxolitinib in severe COVID-19: Results of a multicenter, prospective, single arm, open-label clinical study to investigate the efficacy and safety of ruxolitinib in patients with COVID-19 and severe acute respiratory syndrome. Rev. Fac. Cien. Med. Univ. Nac. Cordoba 2021, 78, 294–302. [Google Scholar] [CrossRef]
- Han, M.K.; Antila, M.; Ficker, J.H.; Gordeev, I.; Guerreros, A.; Bernus, A.L.; Roquilly, A.; Sifuentes-Osornio, J.; Tabak, F.; Teijeiro, R.; et al. Ruxolitinib in addition to standard of care for the treatment of patients admitted to hospital with COVID-19 (RUXCOVID): A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Rheumatol. 2022, 4, e351–e361. [Google Scholar] [CrossRef]
- Marconi, V.C.; Ramanan, A.V.; de Bono, S.; Kartman, C.E.; Krishnan, V.; Liao, R.; Piruzeli, M.L.B.; Goldman, J.D.; Alatorre-Alexander, J.; de Cassia Pellegrini, R.; et al. Efficacy and safety of baricitinib for the treatment of hospitalised adults with COVID-19 (COV-BARRIER): A randomised, double-blind, parallel-group, placebo-controlled phase 3 trial. Lancet Respir. Med. 2021, 9, 1407–1418. [Google Scholar] [CrossRef]
- Lin, Z.; Niu, J.; Xu, Y.; Qin, L.; Ding, J.; Zhou, L. Clinical efficacy and adverse events of baricitinib treatment for coronavirus disease-2019 (COVID-19): A systematic review and meta-analysis. J. Med. Virol. 2022, 94, 1523–1534. [Google Scholar] [CrossRef] [PubMed]
- Iglesias Gómez, R.; Méndez, R.; Palanques-Pastor, T.; Ballesta-López, O.; Borrás Almenar, C.; Megías Vericat, J.E.; López-Briz, E.; Font-Noguera, I.; Menéndez Villanueva, R.; Román Iborra, J.A.; et al. Baricitinib against severe COVID-19: Effectiveness and safety in hospitalised pretreated patients. Eur. J. Hosp. Pharm. 2022, 29, e41–e45. [Google Scholar] [CrossRef]
- Zidi, I.; Mestiri, S.; Bartegi, A.; Amor, N.B. TNF-alpha and its inhibitors in cancer. Med. Oncol. 2010, 27, 185–198. [Google Scholar] [CrossRef]
- Keystone, E.C. Advances in targeted therapy: Safety of biological agents. Ann. Rheum. Dis. 2003, 62 (Suppl. S2), ii34–ii36. [Google Scholar] [CrossRef] [Green Version]
- Askling, J.; Fored, C.M.; Baecklund, E.; Brandt, L.; Backlin, C.; Ekbom, A.; Sundström, C.; Bertilsson, L.; Cöster, L.; Geborek, P.; et al. Haematopoietic malignancies in rheumatoid arthritis: Lymphoma risk and characteristics after exposure to tumour necrosis factor antagonists. Ann. Rheum. Dis. 2005, 64, 1414–1420. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Haynes, K.; Beukelman, T.; Curtis, J.R.; Newcomb, C.; Herrinton, L.J.; Graham, D.J.; Solomon, D.H.; Griffin, M.R.; Chen, L.; Liu, L.; et al. Tumor necrosis factor α inhibitor therapy and cancer risk in chronic immune-mediated diseases. Arthritis Rheum. 2013, 65, 48–58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kokkotis, G.; Kitsou, K.; Xynogalas, I.; Spoulou, V.; Magiorkinis, G.; Trontzas, I.; Trontzas, P.; Poulakou, G.; Syrigos, K.; Bamias, G. Systematic review with meta-analysis: COVID-19 outcomes in patients receiving anti-TNF treatments. Aliment. Pharmacol. Ther. 2022, 55, 154–167. [Google Scholar] [CrossRef]
- Hachem, H.; Godara, A.; Schroeder, C.; Fein, D.; Mann, H.; Lawlor, C.; Marshall, J.; Klein, A.; Poutsiaka, D.; Breeze, J.L.; et al. Rapid and sustained decline in CXCL-10 (IP-10) annotates clinical outcomes following TNF-α antagonist therapy in hospitalized patients with severe and critical COVID-19 respiratory failure. medRxiv 2021. [Google Scholar] [CrossRef]
- Horby, P.; Lim, W.S.; Emberson, J.R.; Mafham, M.; Bell, J.L.; Linsell, L.; Staplin, N.; Brightling, C.; Ustianowski, A.; Elmahi, E.; et al. Dexamethasone in Hospitalized Patients with COVID-19. N. Engl. J. Med. 2021, 384, 693–704. [Google Scholar] [CrossRef] [PubMed]
- Conforti, F.; Wang, Y.; Rodriguez, J.A.; Alberobello, A.T.; Zhang, Y.W.; Giaccone, G. Molecular Pathways: Anticancer Activity by Inhibition of Nucleocytoplasmic Shuttling. Clin. Cancer Res. 2015, 21, 4508–4513. [Google Scholar] [CrossRef] [Green Version]
- Chari, A.; Vogl, D.T.; Gavriatopoulou, M.; Nooka, A.K.; Yee, A.J.; Huff, C.A.; Moreau, P.; Dingli, D.; Cole, C.; Lonial, S.; et al. Oral Selinexor-Dexamethasone for Triple-Class Refractory Multiple Myeloma. N. Engl. J. Med. 2019, 381, 727–738. [Google Scholar] [CrossRef]
- Lapalombella, R.; Sun, Q.; Williams, K.; Tangeman, L.; Jha, S.; Zhong, Y.; Goettl, V.; Mahoney, E.; Berglund, C.; Gupta, S.; et al. Selective inhibitors of nuclear export show that CRM1/XPO1 is a target in chronic lymphocytic leukemia. Blood 2012, 120, 4621–4634. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Abdul Razak, A.R.; Mau-Soerensen, M.; Gabrail, N.Y.; Gerecitano, J.F.; Shields, A.F.; Unger, T.J.; Saint-Martin, J.R.; Carlson, R.; Landesman, Y.; McCauley, D.; et al. First-in-Class, First-in-Human Phase I Study of Selinexor, a Selective Inhibitor of Nuclear Export, in Patients With Advanced Solid Tumors. J. Clin. Oncol. 2016, 34, 4142–4150. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wu, M.; Gui, H.; Feng, Z.; Xu, H.; Li, G.; Li, M.; Chen, T.; Wu, Y.; Huang, J.; Bai, Z.; et al. KPT-330, a potent and selective CRM1 inhibitor, exhibits anti-inflammation effects and protection against sepsis. Biochem. Biophys. Res. Commun. 2018, 503, 1773–1779. [Google Scholar] [CrossRef]
- Sinniah, A.; Yazid, S.; Flower, R.J. The Anti-allergic Cromones: Past, Present, and Future. Front. Pharmacol. 2017, 8, 827. [Google Scholar] [CrossRef] [Green Version]
- Granucci, E.J.; Griciuc, A.; Mueller, K.A.; Mills, A.N.; Le, H.; Dios, A.M.; McGinty, D.; Pereira, J.; Elmaleh, D.; Berry, J.D.; et al. Cromolyn sodium delays disease onset and is neuroprotective in the SOD1. Sci. Rep. 2019, 9, 17728. [Google Scholar] [CrossRef] [Green Version]
- Hori, Y.; Takeda, S.; Cho, H.; Wegmann, S.; Shoup, T.M.; Takahashi, K.; Irimia, D.; Elmaleh, D.R.; Hyman, B.T.; Hudry, E. A Food and Drug Administration-approved asthma therapeutic agent impacts amyloid β in the brain in a transgenic model of Alzheimer disease. J. Biol. Chem. 2015, 290, 1966–1978. [Google Scholar] [CrossRef] [Green Version]
- Ng, G.; Ohlsson, A. Cromolyn sodium for the prevention of chronic lung disease in preterm infants. Cochrane Database Syst. Rev. 2017, 1, CD003059. [Google Scholar] [CrossRef]
- Topalian, S.L.; Hodi, F.S.; Brahmer, J.R.; Gettinger, S.N.; Smith, D.C.; McDermott, D.F.; Powderly, J.D.; Carvajal, R.D.; Sosman, J.A.; Atkins, M.B.; et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N. Engl. J. Med. 2012, 366, 2443–2454. [Google Scholar] [CrossRef] [PubMed]
- Yi, M.; Zheng, X.; Niu, M.; Zhu, S.; Ge, H.; Wu, K. Combination strategies with PD-1/PD-L1 blockade: Current advances and future directions. Mol. Cancer 2022, 21, 28. [Google Scholar] [CrossRef] [PubMed]
- Ettinger, D.S.; Wood, D.E.; Aisner, D.L.; Akerley, W.; Bauman, J.R.; Bharat, A.; Bruno, D.S.; Chang, J.Y.; Chirieac, L.R.; D’Amico, T.A.; et al. Non-Small Cell Lung Cancer, Version 3.2022, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Cancer Netw. 2022, 20, 497–530. [Google Scholar] [CrossRef] [PubMed]
- Keilholz, U.; Ascierto, P.A.; Dummer, R.; Robert, C.; Lorigan, P.; van Akkooi, A.; Arance, A.; Blank, C.U.; Chiarion Sileni, V.; Donia, M.; et al. ESMO consensus conference recommendations on the management of metastatic melanoma: Under the auspices of the ESMO Guidelines Committee. Ann. Oncol. 2020, 31, 1435–1448. [Google Scholar] [CrossRef] [PubMed]
- Audisio, M.; Tucci, M.; Di Stefano, R.F.; Parlagreco, E.; Ungaro, A.; Turco, F.; Audisio, A.; Di Prima, L.; Ortega, C.; Di Maio, M.; et al. New emerging targets in advanced urothelial carcinoma: Is it the primetime for personalized medicine? Crit. Rev. Oncol. Hematol. 2022, 174, 103682. [Google Scholar] [CrossRef] [PubMed]
- Herrmann, M.; Schulte, S.; Wildner, N.H.; Wittner, M.; Brehm, T.T.; Ramharter, M.; Woost, R.; Lohse, A.W.; Jacobs, T.; Schulze Zur Wiesch, J. Analysis of Co-inhibitory Receptor Expression in COVID-19 Infection Compared to Acute. Front. Immunol. 2020, 11, 1870. [Google Scholar] [CrossRef] [PubMed]
- Curtin, N.; Bányai, K.; Thaventhiran, J.; Le Quesne, J.; Helyes, Z.; Bai, P. Repositioning PARP inhibitors for SARS-CoV-2 infection(COVID-19); a new multi-pronged therapy for acute respiratory distress syndrome? Br. J. Pharmacol. 2020, 177, 3635–3645. [Google Scholar] [CrossRef] [PubMed]
- Heer, C.D.; Sanderson, D.J.; Voth, L.S.; Alhammad, Y.M.O.; Schmidt, M.S.; Trammell, S.A.J.; Perlman, S.; Cohen, M.S.; Fehr, A.R.; Brenner, C. Coronavirus infection and PARP expression dysregulate the NAD metabolome: An actionable component of innate immunity. J. Biol. Chem. 2020, 295, 17986–17996. [Google Scholar] [CrossRef] [PubMed]
Study Title | Study Type | Interventions | Phase | Clinical Trial Identifier |
---|---|---|---|---|
COVID-19 Prevention and Treatment in Cancer; a Sequential Multiple Assignment Randomized Trial; (C-SMART) | Interventional | Drug: interferon alfa Drug: selinexor Drug: lenzilumab | III | NCT04534725 |
Rintatolimod and IFN Alpha-2b for the Treatment of COVID-19 in Cancer Patients | Interventional | Other: best practice Biological: recombinant interferon alfa-2b Drug: rintatolimod | I///II | NCT04379518 |
Leflunomide for the Treatment of Severe COVID-19 in Patients With a Concurrent Malignancy | Interventional | Other: best practice Drug: leflunomide Drug: placebo administration | I///II | NCT04532372 |
Viral Specific T Cell Therapy for COVID-19 Related Pneumonia in Cancer Patients | Interventional | Biological: SARS-CoV-2 antigen-specific cytotoxic T-lymphocytes | I | NCT04742595 |
A Trial of the Safety and Immunogenicity of the COVID-19 Vaccine (mRNA-1273) in Participants With Hematologic Malignancies and Various Regimens of Immunosuppression, and in Participants With Solid Tumors on PD1/PDL1 Inhibitor Therapy, Including Booster Doses of Vaccine | Interventional | Biological: mRNA 1273 injection | II | NCT04847050 |
(COVID-19) Longitudinal Neutralizing Antibody Titers in Cancer Patients Receiving Different Anticancer Therapies | Observational | - | - | NCT05384509 |
Antibodies Production After COVID-19 Vaccination Among Patients with Medical History of Cancer and Anti-CD-20 Treatment | Observational | - | - | NCT04779996 |
Immunity Against Severe Acute Respiratory Syndrome Coronavirus 2 Disease (COVID-19] in the Oncology Outpatient Setting (COVIDOUT) | Observational | - | - | NCT04779346 |
A Study on the Immune Response to COVID-19 Vaccination in Cancer Patients—the IOSI-COVID-19-001 Study | Observational | - | - | NCT04800146 |
ASCO Survey on COVID-19 in Oncology (ASCO) Registry | Observational | - | - | NCT04659135 |
Investigation of the B- and T-cell Repertoire and Immune Response in Patients with Acute and Resolved COVID-19 Infection | Observational | - | - | NCT04362865 |
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
Vitali, L.; Merlini, A.; Galvagno, F.; Proment, A.; Sangiolo, D. Biological and Exploitable Crossroads for the Immune Response in Cancer and COVID-19. Biomedicines 2022, 10, 2628. https://doi.org/10.3390/biomedicines10102628
Vitali L, Merlini A, Galvagno F, Proment A, Sangiolo D. Biological and Exploitable Crossroads for the Immune Response in Cancer and COVID-19. Biomedicines. 2022; 10(10):2628. https://doi.org/10.3390/biomedicines10102628
Chicago/Turabian StyleVitali, Letizia, Alessandra Merlini, Federica Galvagno, Alessia Proment, and Dario Sangiolo. 2022. "Biological and Exploitable Crossroads for the Immune Response in Cancer and COVID-19" Biomedicines 10, no. 10: 2628. https://doi.org/10.3390/biomedicines10102628
APA StyleVitali, L., Merlini, A., Galvagno, F., Proment, A., & Sangiolo, D. (2022). Biological and Exploitable Crossroads for the Immune Response in Cancer and COVID-19. Biomedicines, 10(10), 2628. https://doi.org/10.3390/biomedicines10102628