In recent weeks, the rate of SARS-CoV-2 infections has been progressively increasing all over the globe, even in countries where vaccination programs have been strongly implemented. In these regions in 2021, a reduction in the number of hospitalizations and deaths compared to 2020 was observed. This decrease is certainly associated with the introduction of vaccination measures. The process of the development of effective vaccines represents an important challenge. Overall, the breakthrough infections occurring in vaccinated subjects are in most cases less severe than those observed in unvaccinated individuals. This review examines the factors affecting the immunogenicity of vaccines against SARS-CoV-2 and the possible role of nutrients in modulating the response of distinct immune cells to the vaccination.
Citation: Sirio Fiorino, Andrea Carusi, Wandong Hong, Paolo Cernuschi, Claudio Giuseppe Gallo, Emanuele Ferrara, Thais Maloberti, Michela Visani, Federico Lari, Dario de Biase, Maddalena Zippi. SARS-CoV-2 vaccines: What we know, what we can do to improve them and what we could learn from other well-known viruses[J]. AIMS Microbiology, 2022, 8(4): 422-453. doi: 10.3934/microbiol.2022029
In recent weeks, the rate of SARS-CoV-2 infections has been progressively increasing all over the globe, even in countries where vaccination programs have been strongly implemented. In these regions in 2021, a reduction in the number of hospitalizations and deaths compared to 2020 was observed. This decrease is certainly associated with the introduction of vaccination measures. The process of the development of effective vaccines represents an important challenge. Overall, the breakthrough infections occurring in vaccinated subjects are in most cases less severe than those observed in unvaccinated individuals. This review examines the factors affecting the immunogenicity of vaccines against SARS-CoV-2 and the possible role of nutrients in modulating the response of distinct immune cells to the vaccination.
[1] | Fiorino S, Tateo F, Biase D, et al. (2021) SARS-CoV-2: lessons from both the history of medicine and from the biological behavior of other well-known viruses. Future Microbiol 16: 1105-1133. https://doi.org/10.2217/fmb-2021-0064 |
[2] | Fares A (2013) Factors influencing the seasonal patterns of infectious diseases. Int J Prev Med 4: 128-132. |
[3] | Price RHM, Graham C, Ramalingam S (2019) Association between viral seasonality and meteorological factors. Sci Rep 9: 929. https://doi.org/10.1038/s41598-018-37481-y |
[4] | Juthani PV, Gupta A, Borges KA, et al. (2021) Hospitalisation among vaccine breakthrough COVID-19 infections. Lancet Infect Dis 21: 1485-1486. https://doi.org/10.1016/S1473-3099(21)00558-2 |
[5] | Rivasi G, Bulgaresi M, Mossello E, et al. (2021) Course and Lethality of SARS-CoV-2 Epidemic in Nursing Homes after Vaccination in Florence, Italy. Vaccines (Basel) 9: 1174. https://doi.org/10.3390/vaccines9101174 |
[6] | COVID-19 vaccine effectiveness. Available from: https://www.cdc.gov/coronavirus/2019-ncov/vaccines/effectiveness/ |
[7] | Afrough B, Dowall S, Hewson R (2019) Emerging viruses and current strategies for vaccine intervention. Clin Exp Immunol 196: 157-166. https://doi.org/10.1111/cei.13295 |
[8] | Calina D, Docea AO, Petrakis D, et al. (2020) Towards effective COVID19 vaccines: Updates, perspectives and challenges (Review). Int J Mol Med 46: 3-16. https://doi.org/10.3892/ijmm.2020.4596 |
[9] | Castro C, Arnold JJ, Cameron CE (2005) Incorporation fidelity of the viral RNA-dependent RNA polymerase: a kinetic, thermodynamic and structural perspective. Virus Res 107: 141-149. https://doi.org/10.1016/j.virusres.2004.11.004 |
[10] | Smith DB, Bukh J, Kuiken C, et al. (2014) Expanded classification of hepatitis C virus into 7 genotypes and 67 subtypes: updated criteria and genotype assignment web resource. Hepatology 59: 318-327. https://doi.org/10.1002/hep.26744 |
[11] | Yuan M, Huang D, Lee CD, et al. (2021) Structural and functional ramifications of antigenic drift in recent SARS-CoV-2 variants. Science 373: 818-823. https://doi.org/10.1126/science.abh1139 |
[12] | Yewdell JW (2021) Antigenic drift: Understanding COVID-19. Immunity 54: 2681-2687. https://doi.org/10.1016/j.immuni.2021.11.016 |
[13] | Vasireddy D, Vanaparthy R, Mohan G, et al. (2021) Review of COVID-19 variants and COVID-19 vaccine efficacy: What the clinician should know?. J Clin Med Res 13: 317-325. https://doi.org/10.14740/jocmr4518 |
[14] | Hodgson SH, Mansatta K, Mallett G, et al. (2021) What defines an efficacious COVID-19 vaccine? A review of the challenges assessing the clinical efficacy of vaccines against SARS-CoV-2. Lancet Infect Dis 21: e26-e35. https://doi.org/10.1016/S1473-3099(20)30773-8 |
[15] | Andrews N, Tessier E, Stowe J, et al. (2021) Vaccine effectiveness and duration of protection of Comirnaty, Vaxzevria and Spikevax against mild and severe COVID-19 in the UK. medRxiv . https://doi.org/10.1101/2021.09.15.21263583 |
[16] | Bergwerk M, Gonen T, Lustig Y, et al. (2021) Covid-19 Breakthrough Infections in Vaccinated Health Care Workers. N Engl J Med 385: 1474-1484. https://doi.org/10.1056/NEJMoa2109072 |
[17] | Cohn BA, Cirillo PM, Murphy CC, et al. (2022) SARS-CoV-2 vaccine protection and deaths among US veterans during 2021. Science 375: 331-336. https://doi.org/10.1126/science.abm0620 |
[18] | Rzymski P, Camargo CA, Fal A, et al. (2021) COVID-19 vaccine boosters: The good, the bad, and the ugly. Vaccines (Basel) 9: 1299. https://doi.org/10.3390/vaccines9111299 |
[19] | Gazit S, Mizrahi B, Kalkstein N, et al. (2021) BNT162b2 mRNA vaccine effectiveness given confirmed exposure: Analysis of household members of COVID-19 patients. Clin Infect Dis 75: e73-e740. https://doi.org/10.1093/cid/ciab973 |
[20] | Chen X, Wang W, Chen X, et al. (2022) Prediction of long-term kinetics of vaccine-elicited neutralizing antibody and time-varying vaccine-specific efficacy against the SARS-CoV-2 Delta variant by clinical endpoint. BMC Med 20: 36. https://doi.org/10.1186/s12916-022-02249-9 |
[21] | Levin EG, Lustig Y, Cohen C, et al. (2021) Waning Immune Humoral Response to BNT162b2 Covid-19 Vaccine over 6 Months. N Engl J Med 385: e84. https://doi.org/10.1056/NEJMoa2114583 |
[22] | Gao YD, Ding M, Dong X, et al. (2021) Risk factors for severe and critically ill COVID-19 patients: A review. Allergy 76: 428-455. https://doi.org/10.1111/all.14657 |
[23] | WHOCOVID-19: Booster Shots (2021). Available from: https://www.who.int/emergencies/diseases/novel-coronavirus-2019/media-resources/science-in-5/episode-53---covid-19-booster-shots. |
[24] | WHOCoronavirus disease (COVID-19): Vaccines (2022). Available from: https://www.who.int/emergencies/diseases/novel-coronavirus-2019/question-and-answers-hub/q-a-detail/coronavirus-disease-(covid-19)-vaccines |
[25] | Bar-On YM, Goldberg Y, Mandel M, et al. (2021) Protection of BNT162b2 vaccine booster against Covid-19 in Israel. N Engl J Med 385: 1393-1400. https://doi.org/10.1056/NEJMoa2114255 |
[26] | Krause PR, Fleming TR, Peto R, et al. (2021) Considerations in boosting COVID-19 vaccine immune responses. Lancet 398: 1377-1380. https://doi.org/10.1016/S0140-6736(21)02046-8 |
[27] | Lam JH, Smith FL, Baumgarth N (2020) B cell activation and response regulation during viral infections. Viral Immunol 33: 294-306. https://doi.org/10.1089/vim.2019.0207 |
[28] | Laidlaw BJ, Craft JE, Kaech SM (2016) The multifaceted role of CD4(+) T cells in CD8(+) T cell memory. Nat Rev Immunol 16: 102-111. https://doi.org/10.1038/nri.2015.10 |
[29] | Kervevan J, Chakrabarti LA (2021) Role of CD4+ T cells in the control of viral infections: Recent advances and open questions. Int J Mol Sci 22: 523. https://doi.org/10.3390/ijms22020523 |
[30] | Sallusto F (2016) Heterogeneity of Human CD4(+) T cells against microbes. Annu Rev Immunol 34: 317-334. https://doi.org/10.1146/annurev-immunol-032414-112056 |
[31] | Altmann DM, Boyton RJ, Beale R (2021) Immunity to SARS-CoV-2 variants of concern. Science 371: 1103-1104. https://doi.org/10.1126/science.abg7404 |
[32] | Kramer B, Knoll R, Bonaguro L, et al. (2021) Early IFN-alpha signatures and persistent dysfunction are distinguishing features of NK cells in severe COVID-19. Immunity 54: 2650-2669 e2614. https://doi.org/10.1016/j.immuni.2021.09.002 |
[33] | Leem G, Cheon S, Lee H, et al. (2021) Abnormality in the NK-cell population is prolonged in severe COVID-19 patients. J Allergy Clin Immunol 148: 996-1006 e1018. doi:10.1016/j.jaci.2021.07.022 |
[34] | Peng Y, Mentzer AJ, Liu G, et al. (2020) Broad and strong memory CD4(+) and CD8(+) T cells induced by SARS-CoV-2 in UK convalescent individuals following COVID-19. Nat Immunol 21: 1336-1345. https://doi.org/10.1038/s41590-020-0782-6 |
[35] | Min YQ, Huang M, Sun X, et al. (2021) Immune evasion of SARS-CoV-2 from interferon antiviral system. Comput Struct Biotechnol J 19: 4217-4225. https://doi.org/10.1016/j.csbj.2021.07.023 |
[36] | Rydyznski Moderbacher C, Ramirez SI, Dan JM, et al. (2020) Antigen-specific adaptive immunity to SARS-CoV-2 in acute COVID-19 and associations with age and disease severity. Cell 183: 996-1012 e1019. https://doi.org/10.1016/j.cell.2020.09.038 |
[37] | Tan AT, Linster M, Tan CW, et al. (2021) Early induction of functional SARS-CoV-2-specific T cells associates with rapid viral clearance and mild disease in COVID-19 patients. Cell Rep 34: 108728. https://doi.org/10.1016/j.celrep.2021.108728 |
[38] | Carsetti R, Zaffina S, Piano Mortari E, et al. (2020) Different innate and adaptive immune responses to SARS-CoV-2 infection of asymptomatic, mild, and severe cases. Front Immunol 11: 610300. https://doi.org/10.3389/fimmu.2020.610300 |
[39] | Jordan SC, Shin BH, Gadsden TM, et al. (2021) T cell immune responses to SARS-CoV-2 and variants of concern (Alpha and Delta) in infected and vaccinated individuals. Cell Mol Immunol 18: 2554-2556. https://doi.org/10.1038/s41423-021-00767-9 |
[40] | Neidleman J, Luo X, George AF, et al. (2021) Distinctive features of SARS-CoV-2-specific T cells predict recovery from severe COVID-19. Cell Rep 36: 109414. https://doi.org/10.1016/j.celrep.2021.109414 |
[41] | Swadling L, Maini MK (2020) T cells in COVID-19 - united in diversity. Nat Immunol 21: 1307-1308. https://doi.org/10.1038/s41590-020-0798-y |
[42] | Mileto D, Fenizia C, Cutrera M, et al. (2021) SARS-CoV-2 mRNA vaccine BNT162b2 triggers a consistent cross-variant humoral and cellular response. Emerg Microbes Infect 10: 2235-2243. https://doi.org/10.1080/22221751.2021.2004866 |
[43] | Neidleman J, Luo X, Frouard J, et al. (2020) SARS-CoV-2-Specific T cells exhibit phenotypic features of helper function, lack of terminal differentiation, and high proliferation potential. Cell Rep Med 1: 100081. https://doi.org/10.1016/j.xcrm.2020.100081 |
[44] | Yin SW, Zhou Z, Wang JL, et al. (2021) Viral loads, lymphocyte subsets and cytokines in asymptomatic, mildly and critical symptomatic patients with SARS-CoV-2 infection: a retrospective study. Virol J 18: 126. https://doi.org/10.1186/s12985-021-01597-x |
[45] | Balachandran H, Phetsouphanh C, Agapiou D, et al. (2022) Maintenance of broad neutralizing antibodies and memory B cells 1 year post-infection is predicted by SARS-CoV-2-specific CD4+ T cell responses. Cell Rep 38: 110345. https://doi.org/10.1016/j.celrep.2022.110345 |
[46] | Gurevich M, Zilkha-Falb R, Sonis P, et al. (2022) SARS-CoV-2 memory B and T cell profiles in mild COVID-19 convalescent patients. Int J Infect Dis 115: 208-214. https://doi.org/10.1016/j.ijid.2021.12.309 |
[47] | Soresina A, Moratto D, Chiarini M, et al. (2020) Two X-linked agammaglobulinemia patients develop pneumonia as COVID-19 manifestation but recover. Pediatr Allergy Immunol 31: 565-569. https://doi.org/10.1111/pai.13263 |
[48] | Zhao Q, Meng M, Kumar R, et al. (2020) Lymphopenia is associated with severe coronavirus disease 2019 (COVID-19) infections: A systemic review and meta-analysis. Int J Infect Dis 96: 131-135. https://doi.org/10.1016/j.ijid.2020.04.086 |
[49] | De Biasi S, Meschiari M, Gibellini L, et al. (2020) Marked T cell activation, senescence, exhaustion and skewing towards TH17 in patients with COVID-19 pneumonia. Nat Commun 11: 3434. https://doi.org/10.1038/s41467-020-17292-4 |
[50] | Mathew D, Giles JR, Baxter AE, et al. (2020) Deep immune profiling of COVID-19 patients reveals distinct immunotypes with therapeutic implications. Science 369: eabc8511. https://doi.org/10.1126/science.abc8511 |
[51] | Zhang J, Lin H, Ye B, et al. (2021) One-year sustained cellular and humoral immunities of COVID-19 convalescents. Clin Infect Dis 75: e1072-e1081. https://doi.org/10.1093/cid/ciab884 |
[52] | Altman JD, Moss PA, Goulder PJ, et al. (1996) Phenotypic analysis of antigen-specific T lymphocytes. Science 274: 94-96. https://doi.org/10.1126/science.274.5284.94 |
[53] | Maini MK, Boni C, Ogg GS, et al. (1999) Direct ex vivo analysis of hepatitis B virus-specific CD8(+) T cells associated with the control of infection. Gastroenterology 117: 1386-1396. https://doi.org/10.1016/S0016-5085(99)70289-1 |
[54] | Ogg GS, McMichael AJ (1998) HLA-peptide tetrameric complexes. Curr Opin Immunol 10: 393-396. https://doi.org/10.1016/S0952-7915(98)80110-6 |
[55] | Zhu F, Eckels DD (2002) Functionally distinct helper T-cell epitopes of HCV and their role in modulation of NS3-specific, CD8+/tetramer positive CTL. Hum Immunol 63: 710-718. https://doi.org/10.1016/S0198-8859(02)00430-5 |
[56] | Mondelli M, Vergani GM, Alberti A, et al. (1982) Specificity of T lymphocyte cytotoxicity to autologous hepatocytes in chronic hepatitis B virus infection: evidence that T cells are directed against HBV core antigen expressed on hepatocytes. J Immunol 129: 2773-2778. |
[57] | Maini MK, Boni C, Lee CK, et al. (2000) The role of virus-specific CD8(+) cells in liver damage and viral control during persistent hepatitis B virus infection. J Exp Med 191: 1269-1280. https://doi.org/10.1084/jem.191.8.1269 |
[58] | Walewska-Zielecka B, Madalinski K, Jablonska J, et al. (2008) Composition of inflammatory infiltrate and its correlation with HBV/HCV antigen expression. World J Gastroenterol 14: 4040-4046. https://doi.org/10.3748/wjg.14.4040 |
[59] | Wang H, Wu B, Li L, et al. (2017) Hepatic expansion of virus-specific CD8(+)BTLA(+) T cells with regulatory properties in chronic hepatitis B virus infection. Cell Immunol 311: 36-45. https://doi.org/10.1016/j.cellimm.2016.10.002 |
[60] | Welsh RM, Selin LK (2002) No one is naive: the significance of heterologous T-cell immunity. Nat Rev Immunol 2: 417-426. https://doi.org/10.1038/nri820 |
[61] | Tough DF, Borrow P, Sprent J (1996) Induction of bystander T cell proliferation by viruses and type I interferon in vivo. Science 272: 1947-1950. https://doi.org/10.1126/science.272.5270.1947 |
[62] | Kim TS, Shin EC (2019) The activation of bystander CD8(+) T cells and their roles in viral infection. Exp Mol Med 51: 1-9. https://doi.org/10.1038/s12276-019-0316-1 |
[63] | van Aalst S, Ludwig IS, van der Zee R, et al. (2017) Bystander activation of irrelevant CD4+ T cells following antigen-specific vaccination occurs in the presence and absence of adjuvant. PLoS One 12: e0177365. https://doi.org/10.1371/journal.pone.0177365 |
[64] | Martin MD, Shan Q, Xue HH, et al. (2017) Time and antigen-stimulation history influence memory CD8 T cell bystander responses. Front Immunol 8: 634. https://doi.org/10.3389/fimmu.2017.00634 |
[65] | Zhang X, Sun S, Hwang I, et al. (1998) Potent and selective stimulation of memory-phenotype CD8+ T cells in vivo by IL-15. Immunity 8: 591-599. https://doi.org/10.1016/S1074-7613(00)80564-6 |
[66] | Raue HP, Brien JD, Hammarlund E, et al. (2004) Activation of virus-specific CD8+ T cells by lipopolysaccharide-induced IL-12 and IL-18. J Immunol 173: 6873-6881. https://doi.org/10.4049/jimmunol.173.11.6873 |
[67] | Xu D, Fu J, Jin L, et al. (2006) Circulating and liver resident CD4+CD25+ regulatory T cells actively influence the antiviral immune response and disease progression in patients with hepatitis B. J Immunol 177: 739-747. https://doi.org/10.4049/jimmunol.177.1.739 |
[68] | Wu W, Li J, Chen F, et al. (2010) Circulating Th17 cells frequency is associated with the disease progression in HBV infected patients. J Gastroenterol Hepatol 25: 750-757. https://doi.org/10.1111/j.1440-1746.2009.06154.x |
[69] | Chen G, Wu D, Guo W, et al. (2020) Clinical and immunological features of severe and moderate coronavirus disease 2019. J Clin Invest 130: 2620-2629. https://doi.org/10.1172/JCI137244 |
[70] | Datta U, Sehgal S, Pal SR, et al. (1982) Lymphocyte subpopulation in acute viral hepatitis. Gut 23: 927-930. https://doi.org/10.1136/gut.23.11.927 |
[71] | Bertoletti A, Le Bert N, Qui M, et al. (2021) SARS-CoV-2-specific T cells in infection and vaccination. Cell Mol Immunol 18: 2307-2312. https://doi.org/10.1038/s41423-021-00743-3 |
[72] | Bertoletti A, Ferrari C, Fiaccadori F, et al. (1991) HLA class I-restricted human cytotoxic T cells recognize endogenously synthesized hepatitis B virus nucleocapsid antigen. Proc Natl Acad Sci USA 88: 10445-10449. https://doi.org/10.1073/pnas.88.23.10445 |
[73] | Penna A, Chisari FV, Bertoletti A, et al. (1991) Cytotoxic T lymphocytes recognize an HLA-A2-restricted epitope within the hepatitis B virus nucleocapsid antigen. J Exp Med 174: 1565-1570. https://doi.org/10.1084/jem.174.6.1565 |
[74] | Penna A, Del Prete G, Cavalli A, et al. (1997) Predominant T-helper 1 cytokine profile of hepatitis B virus nucleocapsid-specific T cells in acute self-limited hepatitis B. Hepatology 25: 1022-1027. https://doi.org/10.1002/hep.510250438 |
[75] | Li J, Wang J, Kang AS, et al. (2020) Mapping the T cell response to COVID-19. Signal Transduct Target Ther 5: 112. https://doi.org/10.1038/s41392-020-00228-1 |
[76] | Grifoni A, Weiskopf D, Ramirez SI, et al. (2020) Targets of T Cell Responses to SARS-CoV-2 Coronavirus in Humans with COVID-19 Disease and Unexposed Individuals. Cell 181: 1489-1501 e1415. https://doi.org/10.1016/j.cell.2020.05.015 |
[77] | Ferretti AP, Kula T, Wang Y, et al. (2020) Unbiased Screens Show CD8(+) T Cells of COVID-19 Patients Recognize Shared Epitopes in SARS-CoV-2 that Largely Reside outside the Spike Protein. Immunity 53: 1095-1107 e1093. https://doi.org/10.1016/j.immuni.2020.10.006 |
[78] | Zhu C, He G, Yin Q, et al. (2021) Molecular biology of the SARs-CoV-2 spike protein: A review of current knowledge. J Med Virol 93: 5729-5741. https://doi.org/10.1002/jmv.27132 |
[79] | Zhao J, Wang L, Schank M, et al. (2021) SARS-CoV-2 specific memory T cell epitopes identified in COVID-19-recovered subjects. Virus Res 304: 198508. https://doi.org/10.1016/j.virusres.2021.198508 |
[80] | Szabo PA, Dogra P, Gray JI, et al. (2021) Longitudinal profiling of respiratory and systemic immune responses reveals myeloid cell-driven lung inflammation in severe COVID-19. Immunity 54: 797-814 e796. https://doi.org/10.1016/j.immuni.2021.03.005 |
[81] | Saris A, Reijnders TDY, Nossent EJ, et al. (2021) Distinct cellular immune profiles in the airways and blood of critically ill patients with COVID-19. Thorax 76: 1010-1019. https://doi.org/10.1136/thoraxjnl-2020-216256 |
[82] | Grau-Exposito J, Sanchez-Gaona N, Massana N, et al. (2021) Peripheral and lung resident memory T cell responses against SARS-CoV-2. Nat Commun 12: 3010. https://doi.org/10.1038/s41467-021-23333-3 |
[83] | Grant RA, Morales-Nebreda L, Markov NS, et al. (2021) Circuits between infected macrophages and T cells in SARS-CoV-2 pneumonia. Nature 590: 635-641. https://doi.org/10.1038/s41586-020-03148-w |
[84] | Urra JM, Cabrera CM, Porras L, et al. (2020) Selective CD8 cell reduction by SARS-CoV-2 is associated with a worse prognosis and systemic inflammation in COVID-19 patients. Clin Immunol 217: 108486. https://doi.org/10.1016/j.clim.2020.108486 |
[85] | Zimmermann P, Curtis N (2019) Factors that influence the immune response to vaccination. Clin Microbiol Rev 32: e00084-18. https://doi.org/10.1128/CMR.00084-18 |
[86] | Olliaro P, Torreele E, Vaillant M (2021) COVID-19 vaccine efficacy and effectiveness-the elephant (not) in the room. Lancet Microbe 2: e279-e280. https://doi.org/10.1016/S2666-5247(21)00069-0 |
[87] | Fiolet T, Kherabi Y, MacDonald CJ, et al. (2022) Comparing COVID-19 vaccines for their characteristics, efficacy and effectiveness against SARS-CoV-2 and variants of concern: a narrative review. Clin Microbiol Infect 28: 202-221. https://doi.org/10.1016/j.cmi.2021.10.005 |
[88] | Li H, Yu J, Cao B (2021) SARS-CoV-2 vaccination for immune-comprised patients: More is required. Lancet Reg Health Eur 9: 100191. https://doi.org/10.1016/j.lanepe.2021.100191 |
[89] | Weinberger B (2021) Vaccination of older adults: Influenza, pneumococcal disease, herpes zoster, COVID-19 and beyond. Immun Ageing 18: 38. https://doi.org/10.1186/s12979-021-00249-6 |
[90] | Shroff RT, Chalasani P, Wei R, et al. (2021) Immune responses to two and three doses of the BNT162b2 mRNA vaccine in adults with solid tumors. Nat Med 27: 2002-2011. https://doi.org/10.1038/s41591-021-01542-z |
[91] | Alexander JL, Kennedy NA, Lees CW, et al. (2021) SARS-CoV-2 vaccination for patients with inflammatory bowel disease-Authors' reply. Lancet Gastroenterol Hepatol 6: 523-524. https://doi.org/10.1016/S2468-1253(21)00194-1 |
[92] | Moor MB, Suter-Riniker F, Horn MP, et al. (2021) Humoral and cellular responses to mRNA vaccines against SARS-CoV-2 in patients with a history of CD20 B-cell-depleting therapy (RituxiVac): an investigator-initiated, single-centre, open-label study. Lancet Rheumatol 3: e789-e797. https://doi.org/10.1016/S2665-9913(21)00251-4 |
[93] | Boyarsky BJ, Werbel WA, Avery RK, et al. (2021) Antibody response to 2-Dose SARS-CoV-2 mRNA vaccine series in solid organ transplant recipients. JAMA 325: 2204-2206. https://doi.org/10.1001/jama.2021.7489 |
[94] | Rabinowich L, Grupper A, Baruch R, et al. (2021) Low immunogenicity to SARS-CoV-2 vaccination among liver transplant recipients. J Hepatol 75: 435-438. https://doi.org/10.1016/j.jhep.2021.04.020 |
[95] | Meng Z, Zhang J, Shi J, et al. (2020) Immunogenicity of influenza vaccine in elderly people: a systematic review and meta-analysis of randomized controlled trials, and its association with real-world effectiveness. Hum Vaccin Immunother 16: 2680-2689. https://doi.org/10.1080/21645515.2020.1747375 |
[96] | Collier DA, Ferreira I, Kotagiri P, et al. (2021) Age-related immune response heterogeneity to SARS-CoV-2 vaccine BNT162b2. Nature 596: 417-422. https://doi.org/10.1038/s41586-021-03739-1 |
[97] | Cerqueira-Silva T, Oliveira VA, Boaventura VS, et al. (2022) Influence of age on the effectiveness and duration of protection of Vaxzevria and CoronaVac vaccines: A population-based study. Lancet Reg Health Am 6: 100154. https://doi.org/10.1016/j.lana.2021.100154 |
[98] | Botton J, Dray-Spira R, Baricault B, et al. (2022) Reduced risk of severe COVID-19 in more than 1.4 million elderly people aged 75 years and older vaccinated with mRNA-based vaccines. Vaccine 40: 414-417. https://doi.org/10.1016/j.vaccine.2021.12.009 |
[99] | Ventura MT, Casciaro M, Gangemi S, et al. (2017) Immunosenescence in aging: between immune cells depletion and cytokines up-regulation. Clin Mol Allergy 15: 21. https://doi.org/10.1186/s12948-017-0077-0 |
[100] | Kirkwood KL (2018) Inflammaging. Immunol Invest 47: 770-773. https://doi.org/10.1080/08820139.2018.1552392 |
[101] | Kuderer NM, Choueiri TK, Shah DP, et al. (2020) Clinical impact of COVID-19 on patients with cancer (CCC19): a cohort study. Lancet 395: 1907-1918. https://doi.org/10.1016/S0140-6736(20)31187-9 |
[102] | Monin L, Laing AG, Munoz-Ruiz M, et al. (2021) Safety and immunogenicity of one versus two doses of the COVID-19 vaccine BNT162b2 for patients with cancer: interim analysis of a prospective observational study. Lancet Oncol 22: 765-778. https://doi.org/10.1016/S1470-2045(21)00213-8 |
[103] | Latif MB, Shukla S, Del Rio Estrada PM, et al. (2021) Immune mechanisms in cancer patients that lead to poor outcomes of SARS-CoV-2 infection. Transl Res . https://doi.org/10.1016/j.trsl.2021.12.001 |
[104] | Mansi L, Spehner L, Daguindau E, et al. (2021) Study of the SARS-CoV-2-specific immune T-cell responses in COVID-19-positive cancer patients. Eur J Cancer 150: 1-9. https://doi.org/10.1016/j.ejca.2021.03.033 |
[105] | Luqmani YA, El Hashim A (2021) The COVID-19 pandemic: A health crisis managed or a panic response with disastrous future consequences?. Med Princ Pract 31: 1-10. https://doi.org/10.1159/000520258 |
[106] | Tarke A, Sidney J, Methot N, et al. (2021) Impact of SARS-CoV-2 variants on the total CD4(+) and CD8(+) T cell reactivity in infected or vaccinated individuals. Cell Rep Med 2: 100355. https://doi.org/10.1016/j.xcrm.2021.100355 |
[107] | Thanabalan A, Kiarie EG (2021) Influence of feeding Omega-3 Polyunsaturated fatty acids to broiler breeders on indices of immunocompetence, gastrointestinal, and skeletal development in broiler chickens. Front Vet Sci 8: 653152. https://doi.org/10.3389/fvets.2021.653152 |
[108] | Hogenkamp A, van Vlies N, Fear AL, et al. (2011) Dietary fatty acids affect the immune system in male mice sensitized to ovalbumin or vaccinated with influenza. J Nutr 141: 698-702. https://doi.org/10.3945/jn.110.135863 |
[109] | Yuan J, Roshdy AR, Guo Y, et al. (2014) Effect of dietary vitamin A on reproductive performance and immune response of broiler breeders. PLoS One 9: e105677. https://doi.org/10.1371/journal.pone.0105677 |
[110] | Quigley JD, Hill TM, Dennis TS, et al. (2021) Effects of mixed tocopherols added to milk replacer and calf starter on intake, growth, and indices of stress. J Dairy Sci 104: 9769-9783. https://doi.org/10.3168/jds.2020-19929 |
[111] | Nonnecke BJ, Foote MR, Miller BL, et al. (2009) Effects of chronic environmental cold on growth, health, and select metabolic and immunologic responses of preruminant calves. J Dairy Sci 92: 6134-6143. https://doi.org/10.3168/jds.2009-2517 |
[112] | Calder PC (2013) Feeding the immune system. Proc Nutr Soc 72: 299-309. https://doi.org/10.1017/S0029665113001286 |
[113] | Friedman A, Sklan D (1995) Effect of dietary fatty acids on antibody production and fatty acid composition of lymphoid organs in broiler chicks. Poult Sci 74: 1463-1469. https://doi.org/10.3382/ps.0741463 |
[114] | Korver DR, Klasing KC (1997) Dietary fish oil alters specific and inflammatory immune responses in chicks. J Nutr 127: 2039-2046. https://doi.org/10.1093/jn/127.10.2039 |
[115] | Hayek MG, Han SN, Wu D, et al. (1999) Dietary conjugated linoleic acid influences the immune response of young and old C57BL/6NCrlBR mice. J Nutr 129: 32-38. https://doi.org/10.1093/jn/129.1.32 |
[116] | Andresen AMS, Lutfi E, Ruyter B, et al. (2019) Interaction between dietary fatty acids and genotype on immune response in Atlantic salmon (Salmo salar) after vaccination: A transcriptome study. PLoS One 14: e0219625. https://doi.org/10.1371/journal.pone.0219625 |
[117] | Carman JA, Pond L, Nashold F, et al. (1992) Immunity to Trichinella spiralis infection in vitamin A-deficient mice. J Exp Med 175: 111-120. https://doi.org/10.1084/jem.175.1.111 |
[118] | Lee GY, Han SN (2018) The role of vitamin E in immunity. Nutrients 10: 1614. https://doi.org/10.3390/nu10111614 |
[119] | Penkert RR, Surman SL, Jones BG, et al. (2016) Vitamin A deficient mice exhibit increased viral antigens and enhanced cytokine/chemokine production in nasal tissues following respiratory virus infection despite the presence of FoxP3+ T cells. Int Immunol 28: 139-152. https://doi.org/10.1093/intimm/dxv064 |
[120] | Wang Z, Wang Y, Xu B, et al. (2017) Vitamin D improves immune function in immunosuppressant mice induced by glucocorticoid. Biomed Rep 6: 120-124. https://doi.org/10.3892/br.2016.817 |
[121] | Surman SL, Jones BG, Woodland DL, et al. (2017) Enhanced CD103 expression and reduced frequencies of virus-specific CD8(+) T cells among airway lymphocytes after influenza vaccination of mice deficient in vitamins A + D. Viral Immunol 30: 737-743. https://doi.org/10.1089/vim.2017.0086 |
[122] | Surman SL, Penkert RR, Jones BG, et al. (2016) vitamin supplementation at the time of immunization with a cold-adapted influenza virus vaccine corrects poor mucosal antibody responses in mice deficient for vitamins A and D. Clin Vaccine Immunol 23: 219-227. https://doi.org/10.1128/CVI.00739-15 |
[123] | Meydani SN, Meydani M, Verdon CP, et al. (1986) Vitamin E supplementation suppresses prostaglandin E1(2) synthesis and enhances the immune response of aged mice. Mech Ageing Dev 34: 191-201. https://doi.org/10.1016/0047-6374(86)90034-5 |
[124] | Marko MG, Ahmed T, Bunnell SC, et al. (2007) Age-associated decline in effective immune synapse formation of CD4(+) T cells is reversed by vitamin E supplementation. J Immunol 178: 1443-1449. https://doi.org/10.4049/jimmunol.178.3.1443 |
[125] | Marko MG, Pang HJ, Ren Z, et al. (2009) Vitamin E reverses impaired linker for activation of T cells activation in T cells from aged C57BL/6 mice. J Nutr 139: 1192-1197. https://doi.org/10.3945/jn.108.103416 |
[126] | Quintilio W, de Freitas FA, Rodriguez D, et al. (2016) Vitamins as influenza vaccine adjuvant components. Arch Virol 161: 2787-2795. https://doi.org/10.1007/s00705-016-2994-5 |
[127] | Radhakrishnan AK, Mahalingam D, Selvaduray KR, et al. (2013) Supplementation with natural forms of vitamin E augments antigen-specific TH1-type immune response to tetanus toxoid. Biomed Res Int 2013: 782067. https://doi.org/10.1155/2013/782067 |
[128] | Kelley DS, Taylor PC, Nelson GJ, et al. (1997) Effects of dietary arachidonic acid on human immune response. Lipids 32: 449-456. https://doi.org/10.1007/s11745-997-0059-3 |
[129] | Calder PC (1998) Immunoregulatory and anti-inflammatory effects of n-3 polyunsaturated fatty acids. Braz J Med Biol Res 31: 467-490. https://doi.org/10.1590/S0100-879X1998000400002 |
[130] | Al-Khalaifah H (2020) Modulatory effect of dietary polyunsaturated fatty acids on immunity, represented by phagocytic activity. Front Vet Sci 7: 569939. https://doi.org/10.3389/fvets.2020.569939 |
[131] | Dzopalic T, Bozic-Nedeljkovic B, Jurisic V (2021) The role of vitamin A and vitamin D in modulation of the immune response with a focus on innate lymphoid cells. Cent Eur J Immunol 46: 264-269. https://doi.org/10.5114/ceji.2021.103540 |
[132] | Schwalfenberg GK (2011) A review of the critical role of vitamin D in the functioning of the immune system and the clinical implications of vitamin D deficiency. Mol Nutr Food Res 55: 96-108. https://doi.org/10.1002/mnfr.201000174 |
[133] | Mora JR, Iwata M, von Andrian UH (2008) Vitamin effects on the immune system: vitamins A and D take centre stage. Nat Rev Immunol 8: 685-698. https://doi.org/10.1038/nri2378 |
[134] | Huang Z, Liu Y, Qi G, et al. (2018) Role of vitamin A in the immune system. J Clin Med 7: 258. https://doi.org/10.3390/jcm7090258 |
[135] | Motoyama KR, Curb JD, Kadowaki T, et al. (2009) Association of serum n-6 and n-3 polyunsaturated fatty acids with lipids in 3 populations of middle-aged men. Am J Clin Nutr 90: 49-55. https://doi.org/10.3945/ajcn.2008.26761 |
[136] | Abdelmagid SA, Clarke SE, Nielsen DE, et al. (2015) Comprehensive profiling of plasma fatty acid concentrations in young healthy Canadian adults. PLoS One 10: e0116195. https://doi.org/10.1371/journal.pone.0116195 |
[137] | Mariamenatu AH, Abdu EM (2021) Overconsumption of Omega-6 Polyunsaturated fatty acids (PUFAs) versus deficiency of Omega-3 PUFAs in modern-day diets: The disturbing factor for their “balanced antagonistic metabolic functions” in the human body. J Lipids 2021: 8848161. https://doi.org/10.1155/2021/8848161 |
[138] | Han X, Ding S, Lu J, et al. (2022) Global, regional, and national burdens of common micronutrient deficiencies from 1990 to 2019: A secondary trend analysis based on the Global Burden of Disease 2019 study. E Clinical Medicine 44: 101299. https://doi.org/10.1016/j.eclinm.2022.101299 |
[139] | Zhao T, Liu S, Zhang R, et al. (2022) Global burden of vitamin A deficiency in 204 countries and territories from 1990–2019. Nutrients 14: 950. https://doi.org/10.2139/ssrn.4005132 |
[140] | Palacios C, Gonzalez L (2014) Is vitamin D deficiency a major global public health problem?. J Steroid Biochem Mol Biol 144 Pt A: 138-145. https://doi.org/10.1016/j.jsbmb.2013.11.003 |
[141] | Sundaram ME, Talbot HK, Zhu Y, et al. (2013) Vitamin D is not associated with serologic response to influenza vaccine in adults over 50 years old. Vaccine 31: 2057-2061. https://doi.org/10.1016/j.vaccine.2013.02.028 |
[142] | Lee MD, Lin CH, Lei WT, et al. (2018) Does vitamin D deficiency affect the immunogenic responses to influenza vaccination? A systematic review and meta-analysis. Nutrients 10: 409. https://doi.org/10.3390/nu10040409 |
[143] | (2000) Institute of Medicine (US) Panel on Dietary Antioxidants and Related CompoundsDietary Reference Intakes for Vitamin C, Vitamin E, Selenium, and Carotenoids. Washington (DC): National Academies Press. |
[144] | Peter S, Friedel A, Roos FF, et al. (2015) A systematic review of global alpha-tocopherol status as assessed by nutritional intakelevels and blood serum concentrations. Int J Vitam Nutr Res 85: 261-281. https://doi.org/10.1024/0300-9831/a000281 |
[145] | Ford ES, Schleicher RL, Mokdad AH, et al. (2006) Distribution of serum concentrations of alpha-tocopherol and gamma-tocopherol in the US population. Am J Clin Nutr 84: 375-383. https://doi.org/10.1093/ajcn/84.1.375 |
[146] | Malik A, Eggersdorfer M, Trilok-Kumar G (2021) Vitamin E status in healthy population in Asia: A review of current literature. Int J Vitam Nutr Res 91: 356-369. https://doi.org/10.1024/0300-9831/a000590 |
[147] | Meydani SN, Han SN, Wu D (2005) Vitamin E and immune response in the aged: molecular mechanisms and clinical implications. Immunol Rev 205: 269-284. https://doi.org/10.1111/j.0105-2896.2005.00274.x |
[148] | Murphy S, West KP, Greenough WB, et al. (1992) Impact of vitamin A supplementation on the incidence of infection in elderly nursing-home residents: a randomized controlled trial. Age Ageing 21: 435-439. https://doi.org/10.1093/ageing/21.6.435 |
[149] | Buzina-Suboticanec K, Buzina R, Stavljenic A, et al. (1998) Ageing, nutritional status and immune response. Int J Vitam Nutr Res 68: 133-141. |
[150] | Wu D, Meydani SN (2008) Age-associated changes in immune and inflammatory responses: impact of vitamin E intervention. J Leukoc Biol 84: 900-914. https://doi.org/10.1189/jlb.0108023 |
[151] | Wu D, Meydani SN (2014) Age-associated changes in immune function: impact of vitamin E intervention and the underlying mechanisms. Endocr Metab Immune Disord Drug Targets 14: 283-289. https://doi.org/10.2174/1871530314666140922143950 |
[152] | Camargo CQ, Brunetta HS, Nunes EA (2018) Effects of cotreatment with omega-3 polyunsaturated fatty acids and anticancer agents on oxidative stress parameters: a systematic review of in vitro, animal, and human studies. Nutr Rev 76: 765-777. https://doi.org/10.1093/nutrit/nuy029 |
[153] | Story MJ (2021) Zinc, omega-3 polyunsaturated fatty acids and vitamin D: An essential combination for prevention and treatment of cancers. Biochimie 181: 100-122. https://doi.org/10.1016/j.biochi.2020.11.019 |
[154] | Yuen RC, Tsao SY (2021) Embracing cancer immunotherapy with vital micronutrients. World J Clin Oncol 12: 712-724. https://doi.org/10.5306/wjco.v12.i9.712 |
[155] | Chisari FV, Ferrari C (1995) Hepatitis B virus immunopathogenesis. Annu Rev Immunol 13: 29-60. https://doi.org/10.1146/annurev.iy.13.040195.000333 |
[156] | Matchett WE, Joag V, Stolley JM, et al. (2021) Cutting Edge: Nucleocapsid vaccine elicits spike-independent SARS-CoV-2 protective immunity. J Immunol 207: 376-379. https://doi.org/10.4049/jimmunol.2100421 |
[157] | Fiorino S, Gallo C, Zippi M, et al. (2020) Cytokine storm in aged people with CoV-2: possible role of vitamins as therapy or preventive strategy. Aging Clin Exp Res 32: 2115-2131. https://doi.org/10.1007/s40520-020-01669-y |
[158] | Fiorino S, Zippi M, Gallo C, et al. (2021) The rationale for a multi-step therapeutic approach based on antivirals, drugs and nutrients with immunomodulatory activity in patients with coronavirus-SARS2-induced disease of different severities. Br J Nutr 125: 275-293. https://doi.org/10.1017/S0007114520002913 |
[159] | Husson MO, Ley D, Portal C, et al. (2016) Modulation of host defence against bacterial and viral infections by omega-3 polyunsaturated fatty acids. J Infect 73: 523-535. https://doi.org/10.1016/j.jinf.2016.10.001 |
[160] | Chiang N, Serhan CN (2020) Specialized pro-resolving mediator network: an update on production and actions. Essays Biochem 64: 443-462. https://doi.org/10.1042/EBC20200018 |
[161] | Panigrahy D, Gilligan MM, Huang S, et al. (2020) Inflammation resolution: a dual-pronged approach to averting cytokine storms in COVID-19?. Cancer Metastasis Rev 39: 337-340. https://doi.org/10.1007/s10555-020-09889-4 |
[162] | Gallo CG, Fiorino S, Posabella G, et al. (2022) The function of specialized pro-resolving endogenous lipid mediators, vitamins, and other micronutrients in the control of the inflammatory processes: Possible role in patients with SARS-CoV-2 related infection. Prostaglandins Other Lipid Mediat 159: 106619. https://doi.org/10.1016/j.prostaglandins.2022.106619 |
[163] | Sedighiyan M, Abdollahi H, Karimi E, et al. (2021) Omega-3 polyunsaturated fatty acids supplementation improve clinical symptoms in patients with Covid-19: A randomised clinical trial. Int J Clin Pract 75: e14854. https://doi.org/10.1111/ijcp.14854 |
[164] | Rogero MM, Leao MC, Santana TM, et al. (2020) Potential benefits and risks of omega-3 fatty acids supplementation to patients with COVID-19. Free Radic Biol Med 156: 190-199. https://doi.org/10.1016/j.freeradbiomed.2020.07.005 |
[165] | Ross AC, Chen Q, Ma Y (2011) Vitamin A and retinoic acid in the regulation of B-cell development and antibody production. Vitam Horm 86: 103-126. https://doi.org/10.1016/B978-0-12-386960-9.00005-8 |
[166] | Azzi L, Dalla Gasperina D, Veronesi G, et al. (2022) Mucosal immune response in BNT162b2 COVID-19 vaccine recipients. E Bio Medicine 75: 103788. https://doi.org/10.1016/j.ebiom.2021.103788 |
[167] | Hughes DA, Norton R (2009) Vitamin D and respiratory health. Clin Exp Immunol 158: 20-25. https://doi.org/10.1111/j.1365-2249.2009.04001.x |
[168] | Parekh D, Thickett DR, Turner AM (2013) Vitamin D deficiency and acute lung injury. Inflamm Allergy Drug Targets 12: 253-261. https://doi.org/10.2174/18715281113129990049 |
[169] | Remmelts HH, van de Garde EM, Meijvis SC, et al. (2012) Addition of vitamin D status to prognostic scores improves the prediction of outcome in community-acquired pneumonia. Clin Infect Dis 55: 1488-1494. https://doi.org/10.1093/cid/cis751 |
[170] | Dancer RC, Parekh D, Lax S, et al. (2015) Vitamin D deficiency contributes directly to the acute respiratory distress syndrome (ARDS). Thorax 70: 617-624. https://doi.org/10.1136/thoraxjnl-2014-206680 |
[171] | Borsche L, Glauner B, von Mendel J (2021) COVID-19 mortality risk correlates inversely with vitamin D3 status, and a mortality rate close to zero could theoretically be achieved at 50 ng/mL 25(OH)D3: results of a systematic review and meta-analysis. Nutrients 13: 3596. https://doi.org/10.3390/nu13103596 |
[172] | Ebrahimzadeh A, Mohseni S, Narimani B, et al. (2021) Association between vitamin D status and risk of covid-19 in-hospital mortality: A systematic review and meta-analysis of observational studies. Crit Rev Food Sci Nutr : 1-11. https://doi.org/10.1080/10408398.2021.2012419 |
[173] | Dissanayake HA, de Silva NL, Sumanatilleke M, et al. (2021) Prognostic and therapeutic role of vitamin D in COVID-19: systematic review and meta-analysis. J Clin Endocrinol Metab 107: 1484-1502. https://doi.org/10.1210/clinem/dgab892 |
[174] | Tentolouris N, Samakidou G, Eleftheriadou I, et al. (2021) The effect of vitamin D supplementation on mortality and intensive care unit admission of COVID-19 patients. A systematic review, meta-analysis and meta-regression. Diabetes Metab Res Rev : e3517. https://doi.org/10.1002/dmrr.3517 |
[175] | Hu Y, Kung J, Cave A, et al. (2022) Effects of Vitamin D Serum Level on Morbidity and Mortality in Patients with COVID-19: A Systematic Review and Meta-Analysis. J Pharm Pharm Sci 25: 84-92. https://doi.org/10.18433/jpps32590 |
[176] | Chen J, Mei K, Xie L, et al. (2021) Low vitamin D levels do not aggravate COVID-19 risk or death, and vitamin D supplementation does not improve outcomes in hospitalized patients with COVID-19: a meta-analysis and GRADE assessment of cohort studies and RCTs. Nutr J 20: 89. https://doi.org/10.1186/s12937-021-00744-y |
[177] | Pallast EG, Schouten EG, de Waart FG, et al. (1999) Effect of 50- and 100-mg vitamin E supplements on cellular immune function in noninstitutionalized elderly persons. Am J Clin Nutr 69: 1273-1281. https://doi.org/10.1093/ajcn/69.6.1273 |
[178] | Andreone P, Fiorino S, Cursaro C, et al. (2001) Vitamin E as treatment for chronic hepatitis B: results of a randomized controlled pilot trial. Antiviral Res 49: 75-81. https://doi.org/10.1016/S0166-3542(00)00141-8 |
[179] | Fiorino S, Loggi E, Verucchi G, et al. (2017) Vitamin E for the treatment of E-antigen-positive chronic hepatitis B in paediatric patients: results of a randomized phase 2 controlled study. Liver Int 37: 54-61. https://doi.org/10.1111/liv.13192 |
[180] | Gerner P, Posselt HG, Krahl A, et al. (2008) Vitamin E treatment for children with chronic hepatitis B: a randomized placebo controlled trial. World J Gastroenterol 14: 7208-7213. https://doi.org/10.3748/wjg.14.7208 |
[181] | Dikici B, Dagli A, Ucmak H, et al. (2007) Efficacy of vitamin E in children with immunotolerant-phase chronic hepatitis B infection. Pediatr Int 49: 603-607. https://doi.org/10.1111/j.1442-200X.2007.02419.x |
[182] | Fiorino S, Bacchi-Reggiani ML, Leandri P, et al. (2017) Vitamin E for the treatment of children with hepatitis B e antigen-positive chronic hepatitis: A systematic review and meta-analysis. World J Hepatol 9: 333-342. https://doi.org/10.4254/wjh.v9.i6.333 |