Open Access Open Access  Restricted Access Subscription Access

Vaccine Development: Perspectives From Life-History Traits


Affiliations
1 Department of Biology, Indian Institute of Science Education and Research, Tirupati 517 507, India., India
 

Major breakthroughs in vaccinology arise from our understanding of the fundamentals of immunology and infection biology of the disease. The pathogenicity cycle of a microorganism, from its introduction into the host until the resultant effects of infection, is closely linked to the host's immune response towards the pathogen. Additionally, questions in vaccinology, such as what determines the most efficacious route for vaccine delivery and whether mimicking infection from a different route of entry will generate a better immune response, remain open and may not be fully addressed during vaccine development. This article highlights the importance of the life-history traits of the pathogens and enumerates their relevance in designing vaccines. We revisit the path from life-history traits to vaccine development conside­ring the pathogenicity cycle, which may prove critical in designing effective future vaccines and predicting vaccine behaviour in humans.

Keywords

Immune Response, Life-History Traits, Patho-Genesis Cycle, Route of Administration, Vaccine Development.
User
Notifications
Font Size

  • Mukherjee, S., Before virus, after virus: a reckoning. Cell, 2020, 183, 308–314.
  • Greenwood, B., The contribution of vaccination to global health: past, present and future. Philos. Trans. R. Soc. London, Ser. B, 2014, 369, 20130433.
  • Morrison, W. I., Taylor, G., Gaddum, R. M. and Ellis, S. A., Con-tribution of advances in immunology to vaccine development. In Advances in Veterinary Medicine, Academic Press, California, USA, 1999, pp. 181–195.
  • Stern, P. L., Key steps in vaccine development. Ann. Allergy, Asthma Immunol., 2020, 125, 17–27.
  • Hird, T. R. and Grassly, N. C., Systematic review of mucosal im-munity induced by oral and inactivated poliovirus vaccines against virus shedding following oral poliovirus challenge. PLoS Pathog., 2012, 8, e1002599.
  • Priya, S., In Conservation: T. Jacob John. Curr. Sci., 2018, 114(3), 436–438.
  • Bricker, T. L. et al., A single intranasal or intramuscular immunization with chimpanzee adenovirus-vectored SARS-CoV-2 vaccine pro-tects against pneumonia in hamsters. Cell Rep., 2021, 36, 109400.
  • Hassan, A. O. et al., A SARS-CoV-2 infection model in mice demonstrates protection by neutralizing antibodies. Cell, 2020, 182, 744–753.e4.
  • Travis, C. R., As plain as the nose on your face: the case for a nasal (mucosal) route of vaccine administration for COVID-19 disease prevention. Front. Immunol., 2020, 11, 591897.
  • Begon, M., Townsend, C. and Harper, J., Ecology: From Individuals to Ecosystems, 2005.
  • Demars, A. et al., Route of infection strongly impacts the host– pathogen relationship. Front. Immunol., 2019, 10, 1589.
  • Leggett, H. C., Cornwallis, C. K. and West, S. A., Mechanisms of pathogenesis, infective dose and virulence in human parasites. PLoS Pathog., 2012, 8, e1002512.
  • Behrens, S. et al., Infection routes matter in population-specific responses of the red flour beetle to the entomopathogen Bacillus thuringiensis. BMC Genomics, 2014, 15, 445.
  • Darrah, P. A. et al., Prevention of tuberculosis in macaques after intravenous BCG immunization. Nature, 2020, 577, 95–102.
  • Hiremath, G. S. and Omer, S. B., A meta-analysis of studies com-paring the respiratory route with the subcutaneous route of mea-sles vaccine administration. Hum. Vaccines, 2005, 1, 30–36.
  • Manjaly Thomas, Z.-R. and McShane, H., Aerosol immunisation for TB: matching route of vaccination to route of infection. Trans. R. Soc. Trop. Med. Hyg., 2015, 109, 175–181.
  • Park, J.-G. et al., Immunogenicity and protective efficacy of an in-tranasal live-attenuated vaccine against SARS-CoV-2. iScience, 2021, 24, 102941.
  • Garcia-Revilla, J., Deierborg, T., Venero, J. L. and Boza-Serrano, A., Hyperinflammation and fibrosis in severe COVID-19 patients: Galectin-3, a target molecule to consider. Front. Immunol., 2020, 11, 2069.
  • Polidoro, R. B., Hagan, R. S., de Santis Santiago, R. and Schmidt, N. W., Overview: systemic inflammatory response derived from lung injury caused by SARS-CoV-2 infection explains severe out-comes in COVID-19. Front. Immunol., 2020, 11, 1626.
  • Beverley, P. C. L., Sridhar, S., Lalvani, A. and Tchilian, E. Z., Harnessing local and systemic immunity for vaccines against tubercu-losis. Mucosal Immunol., 2014, 7, 20–26.
  • Vetter, V., Denizer, G., Friedland, L. R., Krishnan, J. and Shapiro, M., Understanding modern-day vaccines: what you need to know. Ann. Med., 2018, 50, 110–120.
  • Pulendran, B. and Ahmed, R., Immunological mechanisms of vac-cination. Nature Immunol., 2011, 12, 509–517.
  • Lee, Y. J., Lee, J. Y., Jang, Y. H., Seo, S.-U., Chang, J. and Seong, B. L., Non-specific effect of vaccines: immediate protection against respiratory syncytial virus infection by a live attenuated influenza vaccine. Front. Microbiol., 2018, 9, 83.
  • Christensen, D., Vaccine adjuvants: why and how. Hum. Vaccines Immunother., 2016, 12, 2709–2711.
  • Pulendran, B. S., Arunachalam, P. and O’Hagan, D. T., Emerging concepts in the science of vaccine adjuvants. Nature Rev. Drug Discov., 2021, 20, 454–475.
  • Chumakov, K. et al., Old vaccines for new infections: exploiting innate immunity to control COVID-19 and prevent future pandemics. Proc. Natl. Acad. Sci. USA, 2021, 118, e2101718118.
  • Töpfer, E., Boraschi, D. and Italiani, P., Innate immune memory: the latest frontier of adjuvanticity. J. Immunol. Res., 2015, 2015, 1–7.
  • Netea, M. G. et al., Trained immunity: a program of innate immune memory in health and disease. Science, 2016, 352, aaf1098.
  • Blok, B. A., Arts, R. J. W., van Crevel, R., Benn, C. S. and Netea, M. G., Trained innate immunity as underlying mechanism for the long-term, nonspecific effects of vaccines. J. Leukoc. Biol., 2015, 98, 347–356.
  • Lee, P. P., Disseminated Bacillus Calmette-Guérin and susceptibility to mycobacterial infections-implications on Bacillus Calmette-Guérin vaccinations. Ann. Acad. Med. Singap., 2015, 44, 297–301.
  • Barría, M. I. et al., Localized mucosal response to intranasal live attenuated influenza vaccine in adults. J. Infect. Dis., 2013, 207, 115–124.
  • Song, L. et al., Mucosal and systemic immune responses to influenza H7N9 antigen HA1–2 co-delivered intranasally with flagellin or polyethyleneimine in mice and chickens. Front. Immunol., 2017, 8, 326.
  • Kim, S.-H. and Jang, Y.-S., The development of mucosal vaccines for both mucosal and systemic immune induction and the roles played by adjuvants. Clin. Exp. Vaccine Res., 2017, 6, 15.
  • Lemoine, C. et al., Technological approaches for improving vac-cination compliance and coverage. Vaccines (Basel), 2020, 8(2), 304.
  • Zhu, Q. and Berzofsky, J. A., Oral vaccines: directed safe passage to the front line of defense. Gut Microb., 2013, 4, 246–252.
  • Vela Ramirez, J. E., Sharpe, L. A. and Peppas, N. A., Current state and challenges in developing oral vaccines. Adv. Drug Deliv. Rev., 2017, 114, 116–131.
  • Kurup, V. M. and Thomas, J., Edible vaccines: promises and chal-lenges. Mol. Biotechnol., 2020, 62, 79–90.
  • Abeysundara, A. T., Aponso, M. M. W. and Silva, G. D., A review on edible vaccines: a novel approach to oral immunization as a re-placement of conventional vaccines. Int. J. Food Sci. Nutr., 2017, 2, 19–22.
  • Egunsola, O. et al., Immunogenicity and safety of reduced-dose intradermal versus intramuscular influenza vaccines: a systematic review and meta-analysis. JAMA Netw. Open, 2021, 4, e2035693.
  • Cook, I. F., Subcutaneous vaccine administration – an outmoded practice. Hum. Vaccines Immunother., 2020, 17(5), 1329–1341.
  • Birkhoff, M., Leitz, M. and Marx, D., Advantages of intranasal vaccination and considerations on device selection. Indian J. Pharm. Sci., 2009, 71, 729–731.
  • Rosenbaum, P. et al., Vaccine inoculation route modulates early immunity and consequently antigen specific immune response. Front. Immunol., 2021, 12, 645210.
  • Ols, S. et al., Route of vaccine administration alters antigen traf-ficking but not innate or adaptive immunity. Cell Rep., 2020, 30, 3964–3971.
  • Malik, B., Rath, G. and Goyal, A. K., Are the anatomical sites for vaccine administration selected judiciously? Int. Immunopharma-col., 2014, 19, 17–26.
  • Zimmermann, P. and Curtis, N., Factors that influence the im-mune response to vaccination. Clin. Microbiol. Rev., 2019, 32, e00084-18.
  • Zhang, L., Wang, W. and Wang, S., Effect of vaccine administra-tion modality on immunogenicity and efficacy. Expert Rev. Vac-cines, 2015, 14, 1509–1523.
  • Pasternak, J. A., Hamonic, G., Forsberg, N. M., Wheler, C. L., Dyck, M. K. and Wilson, H. L., Intrauterine delivery of subunit vaccines induces a systemic and mucosal immune response in rab-bits. Am. J. Reprod. Immunol., 2017, 78, e12732.
  • Chang, M.-H. and Chen, D.-S., Prevention of hepatitis B. Cold Spring Harb. Perspect. Med., 2015, 5, a021493.
  • Dutta, A. et al., Sterilizing immunity to influenza virus infection requires local antigen-specific T cell response in the lungs. Sci. Rep., 2016, 6, 32973.
  • Messer, R. J., Dittmer, U., Peterson, K. E. and Hasenkrug, K. J., Essential role for virus-neutralizing antibodies in sterilizing im-munity against friend retrovirus infection. Proc. Natl. Acad. Sci. USA, 2004, 101, 12260–12265.
  • Jangra, S. et al., Sterilizing immunity against SARS-CoV-2 infection in mice by a single-shot and modified imidazoquinoline TLR7/8 agonist-adjuvanted recombinant spike protein vaccine. Preprint. Microbiology, 2020.
  • Herati, R. S. and Wherry, E. J., What is the predictive value of animal models for vaccine efficacy in humans?. Consideration of strate-gies to improve the value of animal models. Cold Spring Harb. Perspect. Biol., 2018, 10, a031583.
  • Cunningham, A. L. et al., Vaccine development: from concept to early clinical testing. Vaccine, 2016, 34, 6655–6664.
  • Gerdts, V. et al., Large animal models for vaccine development and testing. ILAR J., 2015, 56, 53–62.
  • Chirkova, T., Ha, B., Rimawi, B. H., Oomens, A. G. P., Hartert, T. V. and Anderson, L. J., In vitro model for the assessment of human immune responses to subunit RSV vaccines. PLoS ONE, 2020, 15, e0229660.
  • Ming, M. et al., An in vitro functional assay to measure the bio-logical activity of TB vaccine candidate H4-IC31. Vaccine, 2019, 37, 2960–2966.
  • Sanchez-Schmitz, G. et al., Microphysiologic human tissue con-structs reproduce autologous age-specific BCG and HBV primary immunization in vitro. Front. Immunol., 2018, 9, 2634.
  • Ye, W., Luo, C., Li, C., Huang, J. and Liu, F., Organoids to study immune functions, immunological diseases and immunotherapy. Cancer Lett., 2020, 477, 31–40.
  • Wagar, L. E. et al., Modeling human adaptive immune responses with tonsil organoids. Nature Med., 2021, 27, 125–135.
  • Lamers, M. M. et al., SARS-CoV-2 productively infects human gut enterocytes. Science, 2020, 369, 50–54.
  • He, Y. and Xiang, Z., Databases and in silico tools for vaccine design. In In Silico Models for Drug Discovery (ed. Kortagere, S.), Hu-mana Press, Totowa, NJ, USA, 2013, pp. 115–127.
  • María, R. R., Arturo, C. J., Alicia, J. A., Paulina, M. G. and Gerardo, A. O., The impact of bioinformatics on vaccine design and development. In Vaccines (eds Afrin, F., Hemeg, H. and Ozbak, H.), InTech.Open, 2017; doi:10.5772/intechopen.69273.
  • Abdelmageed, M. I. et al., Design of a multiepitope-based peptide vaccine against the E protein of human COVID-19: an immuno-informatics approach. BioMed. Res. Int., 2020, 2020, 1–12.
  • Ong, E., Wong, M. U., Huffman, A. and He, Y., COVID-19 coro-navirus vaccine design using reverse vaccinology and machine learning. Front. Immunol., 2020, 11, 1581.
  • Russo, G. et al., In silico trial to test COVID-19 candidate vaccines: a case study with UISS platform. BMC Bioinformat., 2020, 21, 527.
  • Le, D., Miller, J. D. and Ganusov, V. V., Mathematical modeling provides kinetic details of the human immune response to vaccina-tion. Front. Cell. Infect. Microbiol., 2015, 4, 177.
  • Barbarossa, M. V. and Röst, G., Mathematical models for vaccination, waning immunity and immune system boosting: a general frame-work. arXiv, 7 January 2015.
  • Bonin, C. R. B., Fernandes, G. C., dos Santos, R. W. and Lobosco, M., A qualitatively validated mathematical–computational model of the immune response to the yellow fever vaccine. BMC Immunol., 2018, 19, 15.
  • Ball, P., The lightning-fast quest for COVID vaccines – and what it means for other diseases. Nature, 2020, 589, 16–18.
  • Theeten, H., Van Herck, K., Van Der Meeren, O., Crasta, P., Van Damme, P. and Hens, N., Long-term antibody persistence after vaccination with a 2-dose Havrix TM (inactivated hepatitis A vac-cine): 20 years of observed data, and long-term model-based pre-dictions. Vaccine, 2015, 33, 5723–5727.
  • Amanna, I. J. and Slifka, M. K., Successful vaccines. In Vaccination Strategies against Highly Variable Pathogens (eds Hangartner, L. and Burton, D. R.), Springer International Publishing, Springer Nature, Switzerland, 2018, pp. 1–30.
  • Safaeian, M. et al., Durable antibody responses following one dose of the bivalent human papillomavirus L1 virus-like particle vaccine in the Costa Rica vaccine trial. Cancer Prev. Res., 2013, 6, 1242–1250.
  • Schiller, J. and Lowy, D., Explanations for the high potency of HPV prophylactic vaccines. Vaccine, 2018, 36, 4768–4773.
  • Kennedy, R. B., Ovsyannikova, I. G., Jacobson, R. M. and Poland, G. A., The immunology of smallpox vaccines. Curr. Opin. Immunol., 2009, 21, 314–320.
  • Bonaldo, M. C., Sequeira, P. C. and Galler, R., The yellow fever 17D virus as a platform for new live attenuated vaccines. Hum. Vaccines Immunother., 2014, 10, 1256–1265.
  • Moliva, J. I., Turner, J. and Torrelles, J. B., Immune responses to bacillus Calmette-Guérin vaccination: why do they fail to protect against Mycobacterium tuberculosis? Front. Immunol., 2017, 8, 407.
  • Mohn, K. G.-I., Smith, I., Sjursen, H. and Cox, R. J., Immune re-sponses after live attenuated influenza vaccination. Hum. Vaccines Immunother., 2018, 14, 571–578.
  • Panapasa, J. A., Cox, R. J., Mohn, K. G. I., Aqrawi, L. A. and Brokstad, K. A., The expression of B & T cell activation markers in children’s tonsils following live attenuated influenza vaccine. Hum. Vaccines Immunother., 2015, 11, 1663–1672.
  • Fox, J. P., Modes of action of poliovirus vaccines and relation to resulting immunity. Clin. Infect. Dis., 1984, 6, S352–S355.
  • Connor, R. I. et al., Mucosal immunity to poliovirus. Mucosal Immunol., 2022, 15, 1–9.
  • Herzog, C., Van Herck, K. and Van Damme, P., Hepatitis A vaccina-tion and its immunological and epidemiological long-term effects – a review of the evidence. Hum. Vaccines Immunother., 2021, 17, 1496–1519.
  • Overduin, L. A., van Dongen, J. J. M. and Visser, L. G., The cellular immune response to rabies vaccination: a systematic review. Vac-cines (Basel), 2019, 7, 110.
  • Vila-Corcoles, A. and Ochoa-Gondar, O., Preventing pneumococcal disease in the elderly: recent advances in vaccines and implica-tions for clinical practice. Drugs Aging, 2013, 30, 263–276.
  • Daniels, C. C., Rogers, P. D. and Shelton, C. M., A review of pneumococcal vaccines: current polysaccharide vaccine recom-mendations and future protein antigens. J. Pediatr. Pharmacol. Ther., 2016, 21, 27–35.
  • Zhang, Z. et al., Humoral and cellular immune memory to four COVID-19 vaccines. Cell, 2022, 185, 2434–2451.
  • Hielscher, F. et al., NVX-CoV2373-induced cellular and humoral immunity towards parental SARS-CoV-2 and VOCs compared to BNT162b2 and mRNA-1273-regimens. J. Clin. Virol., 2022, 157, 105321.
  • Dunkle, L. M. et al., Efficacy and safety of NVX-CoV2373 in adults in the United States and Mexico. N. Engl. J. Med., 2022, 386, 531–543.
  • Sadarangani, M., Marchant, A. and Kollmann, T. R., Immunological mechanisms of vaccine-induced protection against COVID-19 in humans. Nature Rev. Immunol., 2021, 21, 475–484.
  • Ferlito, C. et al., Tetanus–diphtheria vaccination in adults: the long-term persistence of antibodies is not dependent on polyclonal B-cell activation and the defective response to diphtheria toxoid re-vaccination is associated to HLADRB1∗01. Vaccine, 2018, 36, 6718–6725.
  • Weinberger, B., Schirmer, M., Matteucci Gothe, R., Siebert, U., Fuchs, D. and Grubeck-Loebenstein, B., Recall responses to tetanus and diphtheria vaccination are frequently insufficient in elderly persons. PLoS ONE, 2013, 8, e82967.
  • Honorati, M. C. and Facchini, A., Immune response against HBsAg vaccine. World J. Gastroenterol., 1998, 4, 464–466.
  • Wang, R.-X., Boland, G. J., van Hattum, J. and de Gast, G. C., Long term persistence of T cell memory to HBsAg after hepatitis B vaccination. World J. Gastroenterol., 2004, 10, 260–263.
  • Ozaki, T., Mochizuki, H., Ichikawa, Y., Fukuzawa, Y., Yoshida, S. and Morimoto, M., Persistence of hepatitis B surface antibody levels after vaccination with a recombinant hepatitis B vaccine: a 3-year follow-up study. J. Oral Sci., 2000, 42, 147–150.
  • Herrin, D. M. et al., Comparison of adaptive and innate immune responses induced by licensed vaccines for human papillomavirus. Hum. Vaccines Immunother., 2014, 10, 3446–3454.
  • Mariani, L. and Venuti, A., HPV vaccine: an overview of immune response, clinical protection and new approaches for the future. J. Transl. Med., 2010, 8, 105.
  • Goel, R. R. et al., Distinct antibody and memory B cell responses in SARS-CoV-2 naïve and recovered individuals after mRNA vac-cination. Sci. Immunol., 2021, 6, eabi6950.
  • Mateus, J. et al., Low-dose mRNA-1273 COVID-19 vaccine generates durable memory enhanced by cross-reactive T cells. Science, 2021, 374, eabj9853.
  • Andreano, E. et al., B cell analyses after SARS-CoV-2 mRNA third vaccination reveals a hybrid immunity like antibody response. Nature Commun., 2023, 14, 53.
  • Dey, A. et al., Immunogenic potential of DNA vaccine candidate, ZyCoV-D against SARS-CoV-2 in animal models. Vaccine, 2021, 39, 4108–4116.
  • Khobragade, A. et al., Efficacy, safety, and immunogenicity of the DNA SARS-CoV-2 vaccine (ZyCoV-D): the interim efficacy results of a phase 3, randomised, double-blind, placebo-controlled study in India. Lancet, 2022, 399, 1313–1321.
  • Ewer, K. J. et al., T cell and antibody responses induced by a single dose of ChAdOx1 nCoV-19 (AZD1222) vaccine in a phase 1/2 clinical trial. Nature Med., 2021, 27, 270–278.
  • Shen, C.-F. et al., Innate immune responses of vaccines determine early neutralizing antibody production after ChAdOx1nCoV-19 vaccination. Front. Immunol., 2022, 13, 807454.
  • Viola, A., Munari, F., Sánchez-Rodríguez, R., Scolaro, T. and Castegna, A., The metabolic signature of macrophage responses. Front. Immunol., 2019, 10, 1462.
  • Schluns, K. S. and Lefrançois, L., Cytokine control of memory T-cell development and survival. Nature Rev. Immunol., 2003, 3, 269– 279.
  • Vazquez, M. I., Catalan-Dibene, J. and Zlotnik, A., B cells responses and cytokine production are regulated by their immune microenvi-ronment. Cytokine, 2015, 74, 318–326.

Abstract Views: 97

PDF Views: 67




  • Vaccine Development: Perspectives From Life-History Traits

Abstract Views: 97  |  PDF Views: 67

Authors

Deevitha Balasubramanian
Department of Biology, Indian Institute of Science Education and Research, Tirupati 517 507, India., India
Raju Mukherjee
Department of Biology, Indian Institute of Science Education and Research, Tirupati 517 507, India., India

Abstract


Major breakthroughs in vaccinology arise from our understanding of the fundamentals of immunology and infection biology of the disease. The pathogenicity cycle of a microorganism, from its introduction into the host until the resultant effects of infection, is closely linked to the host's immune response towards the pathogen. Additionally, questions in vaccinology, such as what determines the most efficacious route for vaccine delivery and whether mimicking infection from a different route of entry will generate a better immune response, remain open and may not be fully addressed during vaccine development. This article highlights the importance of the life-history traits of the pathogens and enumerates their relevance in designing vaccines. We revisit the path from life-history traits to vaccine development conside­ring the pathogenicity cycle, which may prove critical in designing effective future vaccines and predicting vaccine behaviour in humans.

Keywords


Immune Response, Life-History Traits, Patho-Genesis Cycle, Route of Administration, Vaccine Development.

References





DOI: https://doi.org/10.18520/cs%2Fv124%2Fi9%2F1039-1052