Next Article in Journal
Prevalence, Risk Factors and Impacts Related to Mould-Affected Housing: An Australian Integrative Review
Next Article in Special Issue
The Emerging Role of the Aging Process and Exercise Training on the Crosstalk between Gut Microbiota and Telomere Length
Previous Article in Journal
A “Ballpark” Assessment of Social Distancing Efficiency in the Early Stages of the COVID-19 Pandemic
Previous Article in Special Issue
Acute Effects of Different Activity Types and Work-To-Rest Ratio on Post-Activation Performance Enhancement in Young Male and Female Taekwondo Athletes
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Why Physical Activity Should Be Considered in Clinical Trials for COVID-19 Vaccines: A Focus on Risk Groups

by
Miguel Junior Sordi Bortolini
1,
Bernardo Petriz
2,
José Roberto Mineo
3 and
Rafael de Oliveira Resende
4,5,6,*
1
Laboratory of Translational Immunology, Health and Sports Sciences Center, Federal University of Acre, Rio Branco 69920-900, Brazil
2
Laboratory of Molecular Exercise Physiology, University Center UDF, Brasília 70390-045, Brazil
3
Laboratory of Immunoparasitology “Dr. Mário Endsfeldz Camargo”, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia 38405-317, Brazil
4
Laboratory on Thymus Research, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro 21040-900, Brazil
5
National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Rio de Janeiro 21040-900, Brazil
6
Laboratory of Allergy and Clinical Immunology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia 38405-317, Brazil
*
Author to whom correspondence should be addressed.
Int. J. Environ. Res. Public Health 2022, 19(3), 1853; https://doi.org/10.3390/ijerph19031853
Submission received: 30 October 2021 / Revised: 19 January 2022 / Accepted: 20 January 2022 / Published: 7 February 2022
(This article belongs to the Special Issue New Advances in Sports Performance and Health across the Lifespan)

Abstract

:
Since the World Health Organization declared the global COVID-19 state of emergency in early 2020, several vaccine candidates have emerged to control SARS-CoV-2, and some of them have been approved and implemented in vaccination campaigns worldwide. Although clinical trials for these vaccines have been carried out using highly controlled methods with accurate immunological tests, clinical questionnaires did not include questions concerning the physical activity profile among volunteers. It has been well established that physical activity plays a pivotal role in the immune response after vaccination, led by the activation of cytokines, antibodies, and cells. This concept should have been considered when evaluating the efficacy of COVID-19 vaccine candidates, particularly in elderly and obese people. Here, we discuss data from the literature providing strong evidence regarding the importance of analyzing physical activity parameters to improve the accuracy of clinical trials on assessing the efficacy of vaccine candidates.

1. Introduction

Vaccines are a vital tool for modern society. Their administration is essential for the prevention of infectious diseases, as they represent the most reasonable cost-effectiveness ratio among global interventions to eradicate diseases [1,2]. It is not by chance that the investigation of mechanisms to prevent diseases, including those caused by lethal pathogens, has led many teams around the world to develop new methods and assessment protocols for vaccines.
The global population has been facing the worst health crisis in the contemporary era since COVID-19 was declared a health emergency by the World Health Organization (WHO) in 2020. Although new technologies have been put in place to develop effective vaccines against SARS-CoV-2 to control the pandemic [3,4,5,6], clinical trials aimed at evaluating their safety and efficacy have been based on laboratory tests that assess seroconversion and immune cell responses to the viral antigens along with clinical questionnaires containing questions regarding the lifestyle and/or health conditions of volunteers.
COVID-19 was first described as severe pneumonia with an undefined etiology when the first cases were reported [7]. Since then, the puzzle of this disease has been partially solved, although some pieces still need to be filled in. Currently, it is known that SARS-CoV-2 can reach human cells via the interaction between the viral spike (S) protein and angiotensin-converting enzyme-2 (ACE2) receptor in association with transmembrane serine-protease-2 (TMPRSS2) receptor in human cells [8]. This interaction allows for viral internalization and replication, triggering a set of inflammatory cells (e.g., macrophages, neutrophils, and T cells) and molecules (e.g., type I IFN, TLR-4, TLR-7, TLR-8, CXCL-9, and CXCL-6) [9]. These can cause clinical symptoms such as cough, myalgia, headache, fever, anosmia, ageusia, and nasal congestion to varying degrees among patients. In some cases, no clinical symptoms are claimed, with no abnormalities in the image diagnosis [10]. In contrast, in other cases, there can be a cytokine storm, which is characterized by an increased expression of tumor necrosis factor alpha (TNF-α), interleukin (IL)-1, and IL-6, responsible for its progression to severe disease [11]. Notably, the worst prognosis is found in people with comorbidities, particularly those who are obese or elderly. As the induction of an inappropriate immune response characterizes these individuals, they are likely to recover from COVID-19 if they have an active lifestyle [12].
On the other hand, social isolation and stress caused by the pandemic have led most people to reduce or withdraw from physical activity, contributing to immune dysregulation [13,14,15,16,17]. A study compared the risk of intensive care unit admission, hospitalization, and mortality between inactive/less-active people and active people [18]. Inactive people had the most significant risk factor for all situations and active people had the lowest. In addition, physical inactivity, advanced age, and a history of organ transplantation were associated with the most substantial risk for severe COVID-19 outcomes. Indeed, a disturbed immune response to SARS-CoV-2 could be minimized if physical activity was maintained during the pandemic.
The development of COVID-19 vaccine candidates during an ongoing global emergency is unprecedented [19]. Basic information on clinical evaluation may not be adequately obtained due to the overall pressure of the pandemic. Until recently, vaccines took years or decades to be applied in vaccination programs from their early phases. Nonetheless, only 10 months after the SARS-CoV-2 structure was solved and the first experimental data were revealed, at least five vaccines were licensed by regulatory agencies worldwide, and mass vaccination was started in many countries to control the pandemic.
Advances in the research on molecular and physiological responses to exercise have given prominence to the field of exercise immunology. In this sense, it is known that physical activity contributes significantly to stimulating crucial components of the immune system [20,21], promoting the host’s defense against parasites [22,23,24], treating cancer [25], and regulating autoimmune and hypersensitivity components [26,27,28]. It has also been reported that physical exercise is essential for vaccine effectiveness [29,30,31,32], which is crucial for the new COVID-19 vaccines. There is strong clinical evidence concerning the association of SARS-CoV-2 infection with an increased risk of intensive care unit admission, hospitalization, and mortality for inactive or less active people compared to active people [18]. Therefore, we believe that populations with a greater propensity for experiencing more severe COVID-19 could benefit from physical activity assessments to identify vaccine efficacy more accurately for those groups. Thus, we summarized the importance of physical activity in assessing the efficacy and effectiveness of COVID-19 vaccines based on immunological principles.

2. Physical Activity and Immune Response

The immune response to vaccination is individual, and pathways can be activated in different ways among individuals [33,34,35,36]. This feature has also been discussed for COVID-19 vaccines, particularly in older people, who are more vulnerable and at higher risk of worsening disease [37]. Several factors may influence the immune response to vaccination, including genetics, nutrition, perinatal status, pathogen class, the environment, and behavioral aspects [32]. It is imperative to highlight physical fitness and physical activity among these factors. In a recent study concerning the efficacy of the tetravalent influenza virus vaccine among athletes and non-athletes, CD4+ T cells were found to be 1.5 times higher among athletes than the control group one week after vaccination [38].
Similarly, they found that athletes had higher cytotoxic T-lymphocyte-associated protein 4 (CTLA4) expression and specific-H1N1 and H3N2 neutralizing antibodies. Additionally, influenza virus-specific antibodies were assessed after vaccination in exercised and inactive older people, with higher influenza virus-specific IgM and IgG titers observed in the first group [39]. Moreover, vaccine efficacy was improved in active elderly patients, represented by higher numbers of monocytes and plasma blasts in the peripheral blood, increased expression of genes associated with phagocytosis, and lower levels of CXCL10 and eotaxin chemokines [40].
Remarkably, the skeletal muscle constitutes an endocrine organ [41], known to release more than 10,000 proteins (e.g., IL-6, IL-15, and IL-7 myokines), 90% of which reach the bloodstream when stimulated by acute and chronic exercise stimuli [42,43,44,45]. These myokines are also associated with lymphocyte memory and were shown to be upregulated after strength exercises, which is essential to improving the protective effect of vaccines [46]. It has also been demonstrated that immunosenescence components may be downregulated in people who exercise, with evident improvement in adaptive immunity [47]. In addition, higher concentrations of interferon gamma (IFN-γ) and TNF-α were found along with decreased IL-4 and IL-10 levels after 12 weeks of swimming in a murine model [24].
Adjuvants are essential for stimulating the immune system after vaccination [48], and physical activity could play this functional role in improving vaccine effectiveness [49,50,51,52]. In a randomized controlled clinical trial, subjects underwent a 45 min session of aerobic exercise before influenza vaccination [31], which led to higher antibody titers 20 weeks after vaccination in young women compared to men, who had a higher IFN-γ response 8 weeks after vaccination, suggesting a sex-specific effect of muscle stress on antibody and cytokine responses to vaccination. In a herpes simplex virus type 1 (HSV-1) murine model, IFN release was also enhanced, as well as humoral immunity (IgG2a/IgG1 ratio), after short-term aerobic exercise associated with vaccination [53]. From this perspective, 133 healthy young adults were randomized to an exercise or control task group [49], and each received a full or half dose of pneumococcal vaccine. Before vaccination, the exercise group completed a 15 min arm and shoulder exercise while the control group rested quietly. The exercise group presented increased antibody levels and better responses than those who rested and received a half dose, and similarly for those who received the full dose. Moreover, higher IgG and IgM titers and enhanced cytokine production were found after vaccination with bacterial and viral antigens associated with bouts of exercise, reinforcing physical activity as a potential vaccination adjuvant [54].
An important issue that must be addressed is the effect of acute and chronic exercise on vaccination responses in addition to age and gender. Although in general, antibody titers were shown to be similar between trained and non-trained older people 4 weeks after influenza vaccination, when comparing gender, trained women presented a significantly higher antibody response against H1N1 influenza than men, indicating that moderate acute aerobic exercise elicits a stronger immune response in women compared to men [50]. In contrast, in adult athletes, specific-T cells and neutralizing antibodies increased after vaccination with tetravalent influenza [38], suggesting that a high frequency and high intensity of training enhances vaccination response.
Furthermore, B and T lymphocytes have been related to physical activity. For example, B cell content was shown to increase up to 88% after acute exercise, among which immature cells (CD27CD10+ and CD27IgD) were the most prominent, followed by memory (CD27+CD38 and CD27+IgD) and naïve (CD27CD10 and CD27IgD+) B cells, when healthy adult subjects engaged in 30-min continuous cycling [55]. In addition, regular physical exercise, cognitive scores on memory tests, processing speed, attention, verbal fluency, and executive function were associated with increased numbers of circulating naïve B and T cells after 12 months of moderate- to high-intensity aerobic exercise training [56].
The roles of myeloid (mDC) and plasmacytoid (pDC) dendritic cells are well established as the main antigen-presenting cells in the innate immune system [57], and the synergic immunogenicity process has been specifically addressed to both cell lineages [58,59]. Even when mDCs are not directly infected, they can capture exogenous antigens from the infectious agent and present them on MHC class I molecules by a cross-presentation mechanism. Further, the ability of pDC to present antigens aimed at activating CD4+ and CD8+ T cells in addition to cross-presentation by antigen transfer to bystander mDCs mediated by pDC-derived exosomes were also reported [60].
Interestingly, mDCs can be increased after acute exercise, and they can also increase after strenuous/prolonged exercise. In this context, the total number of DCs increases after a bout of exercise, suggesting preferential mobilization of plasmacytoid DCs during exercise. However, pDCs are shown to decrease immediately post-exercise [61]. Although no CD209+ mDC subset was found in the peripheral blood before, during, or after the exercise, CD205− cells were the most responsive among the mDC subsets [62].
Concerning the investigation of mDC maturation in animals with induced immunosuppression subjected to physical activity, the percentage of DCs increased in trained groups with and without immunosuppressants, associated with higher CD80, CD86, and MHC-II expression levels when compared to untrained animals. Moreover, IFN-γ and IL-12 cytokines may be upregulated in exercised mice, with physical exercise suggested as an inducer of differentiation and maturation of DCs, even within an immunosuppressive environment, in association with the Th1 profile [63]. Regarding the Th2 response, in an experimental study based on the OVA-induced asthma model, moderate physical exercise modulated allergic pulmonary inflammation, increasing Treg and M2 recruitment and pDC activation [28].
Exosomes are another topic of significant debate. This vesicular structure derived from an invagination of the plasma membrane is known to mediate cell-to-cell communication in a broad range of tissues. Notably, physical exercise plays an essential role in releasing exosomes, particularly from T cells, monocytes, and antigen-presenting cells [64,65]. Thus, we speculate that the release of exosomes from muscles during exercise could be another way the immune response to microbes, by either natural infection or vaccination, is improved.
Several studies have shown evidence of the critical crosstalk between the gut microbiota and the immune system. Throughout life, the continuous communication between the microbial consortia and the immune cells of the gut mucosa (e.g., dendritic cells and macrophages) modulates the development of the immune system, giving it the ability to differentiate harmless bacteria from pathogens through the recognition of microbe-associated molecular patterns (MAMPS) [66,67]. Furthermore, the possible role of epithelial-derived exosomes in modulating gut microbiota homeostasis through the delivery of antimicrobial agents is also under discussion [68]. Since the modulating effects of exercise on gut microbiota are already recognized [69], as well as their role in muscle exosome release [70], it is essential to consider the hypothesis of a similar stimulation for epithelial and gut microbes.

3. COVID-19 Vaccine Candidates and Physical Activity

Vaccine candidates for COVID-19 began to emerge a few months after the onset of the pandemic. The starting point for the development of these vaccines was, in addition to the discovery of SARS-CoV-2 as the causative pathogen, solving the genomic and structural sequence of the virus. After that, some laboratories, supported by biotechnological and pharmaceutical companies, initiated robust efforts based on established platforms, such as the use of attenuated virus and peptide macromolecules, while others exploited technologies not previously used in this context, such as messenger RNA, DNA, and a viral plasmid vector. To evaluate the efficacy of the vaccines, subjects were recruited for clinical trials from different parts of the globe, especially where the numbers of cases remained higher. However, so far, among the leading COVID-19 vaccine clinical trials [71,72,73,74,75,76], none have been concerned with collecting data regarding the current physical fitness or physical practice of people recruited for the studies.
To understand the possible role of physical conditioning and physical activity on the immune response to vaccination, we highlight the main findings regarding the significance of physical exercise before vaccination (Figure 1). A model is presented to compare vaccine efficacy and the subject’s physical condition (Figure 1A–D) based on antibody/cell-mediated cytokine levels, MHC, CD80/CD86 expression, and lymphocyte/dendritic cell proliferation data reported in the literature. When regularly active trained subjects had an exercise session shortly before vaccination, the efficacy was mostly improved (Figure 1A). However, if trained subjects curtailed physical activity just before vaccination, vaccine efficacy could decrease (Figure 1B). When subjects with a sedentary lifestyle exercised a few minutes before vaccination, the efficacy was lower compared to physically active people (Figure 1C) but higher than when they did not perform an exercise before vaccination (Figure 1D). In this context, we hypothesized that active people would be more likely to experience a better immune response than inactive people with most COVID-19 vaccines, and the same would be found for people who exercise just before vaccination.
To assess the physical activity profile among study participants, we can use indirect or direct tools (Figure 2). Among the former, clinical questionnaires are a good assessment tool, as they allow people’s clinical history to be known using questions that address their physical activity routine. However, this method has some limitations, as it can produce inaccurate results depending on how the questions are answered. In this sense, the use of markers and direct assays (e.g., accelerometry) can provide a more accurate picture of the individual’s physical state in terms of their physical activity practice. In order to provide more accurate representative data, a combination of both forms is also recommended.

4. Conclusions

There is evidence that physical activity regulates the inflammatory response. This process is mediated by cellular and humoral immune responses, including distinct cytokine profiles, neutralizing antibody isotypes, and MHC-II and CD80/CD86 expression, leading to increased vaccine efficacy by acting as a potential adjuvant. Due to the global public health emergency scenario, several vaccine candidates have been developed in the short term to control COVID-19 incidence [77,78], and people are currently being vaccinated. In some countries, an administration of a third dose has been authorized [79]. Since physical inactivity and two other factors present the most significant risk for severe outcomes of COVID-19, inactive people, particularly with comorbidities such as obesity, immunosuppression, or immunodeficiency, should be prioritized in third dose vaccination. Although physical exercise has not been evaluated with regard to the COVID-19 vaccines available to date, many potential vaccine candidates are emerging from pharmaceutical companies in Europe, Asia, and Latin America to protect the global population against the spread of the disease. Furthermore, with the emergence of new variants of SARS-CoV-2, the use of a booster dose has been adopted, and those who do not practice regular physical activity, in addition to those who are elderly or immunosuppressed, should be included among the priority groups, as their capacity to produce neutralizing antibodies is considerably reduced. The alternative of developing new vaccine compositions has become clear, and new tests must be conducted to evaluate those formulations. In this regard, as part of the clinical protocol, a complete immunological profile and lifestyle activities should be assessed before and after vaccination, with direct or indirect methods that allow people’s physical activity level to be investigated. People who exercise display robust responses to the vaccines, and this strategy should encourage people to practice physical activity, especially during the COVID-19 pandemic.

Author Contributions

Conceptualization, M.J.S.B.; writing—original draft preparation, M.J.S.B.; data analysis, M.J.S.B., R.d.O.R.; writing—review and editing, R.d.O.R., B.P. and J.R.M.; supervision, J.R.M. and R.d.O.R.; funding acquisition, R.d.O.R. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Fiocruz and Mercosur Fund for Structural Convergence (FOCEM; #03/11).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We thank to Dea Villa Verde for major English revisions of this manuscript and Gabriel Oliveira Marques for graphical support.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. WHO; UNICEF; The World Bank. State of the World’s Vaccines and Immunization, 3rd ed.; World Health Organization: Geneva, Switzerland, 2009.
  2. Leidner, A.J.; Murthy, N.; Chesson, H.W.; Biggerstaff, M.; Stoecker, C.; Harris, A.M.; Acosta, A.; Dooling, K.; Bridges, C.B. Cost-effectiveness of adult vaccinations: A systematic review. Vaccine 2019, 37, 226–234. [Google Scholar] [CrossRef] [PubMed]
  3. Brüssow, H. Efforts towards a COVID-19 vaccine. Environ. Microbiol. 2020, 22, 4071–4084. [Google Scholar] [CrossRef] [PubMed]
  4. Awadasseid, A.; Wu, Y.; Tanaka, Y.; Zhang, W. Current advances in the development of SARS-CoV-2 vaccines. Int. J. Biol. Sci. 2021, 17, 8–19. [Google Scholar] [CrossRef] [PubMed]
  5. Kis, Z.; Shattock, R.; Shah, N.; Kontoravdi, C. Emerging technologies for low-cost, rapid vaccine manufacture. Biotechnol. J. 2019, 14, e1800376. [Google Scholar] [CrossRef] [Green Version]
  6. Hume, H.K.C.; Lua, L.H.L. Platform technologies for modern vaccine manufacturing. Vaccine 2017, 35, 4480–4485. [Google Scholar] [CrossRef]
  7. Khan, N. New Virus Discovered by Chinese Scientists Investigating Pneumonia Outbreak. Available online: https://www.wsj.com/articles/new-virus-discovered-by-chinese-scientists-investigating-pneumonia-outbreak-11578485668 (accessed on 8 June 2021).
  8. Hoffmann, M.; Kleine-Weber, H.; Schroeder, S.; Kruger, N.; Herrler, T.; Erichsen, S.; Schiergens, T.S.; Herrler, G.; Wu, N.H.; Nitsche, A.; et al. SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell 2020, 181, 271–280.e8. [Google Scholar] [CrossRef]
  9. Wang, D.; Hu, B.; Hu, C.; Zhu, F.; Liu, X.; Zhang, J.; Wang, B.; Xiang, H.; Cheng, Z.; Xiong, Y.; et al. Clinical characteristics of 138 hospitalized patients with 2019 novel coronavirus-Infected pneumonia in Wuhan, China. JAMA 2020, 323, 1061–1069. [Google Scholar] [CrossRef]
  10. Gao, Z.; Xu, Y.; Sun, C.; Wang, X.; Guo, Y.; Qiu, S.; Ma, K. A systematic review of asymptomatic infections with COVID-19. J. Microbiol. Immunol. Infect. 2021, 54, 12–16. [Google Scholar] [CrossRef]
  11. Ragab, D.; Eldin, H.S.; Taeimah, M.; Khattab, R.; Salem, R. The COVID-19 cytokine storm: What we know so far. Front. Immunol. 2020, 11, 1446. [Google Scholar] [CrossRef]
  12. Da Silveira, M.P.; da Silva Fagundes, K.K.; Bizuti, M.R.; Starck, E.; Rossi, R.C.; de Resende, E.S.D.T. Physical exercise as a tool to help the immune system against COVID-19: An integrative review of the current literature. Clin. Exp. Med. 2021, 21, 15–28. [Google Scholar] [CrossRef]
  13. Ingram, J.; Maciejewski, G.; Hand, C.J. Changes in diet, sleep, and physical activity are associated with differences in negative mood during COVID-19 lockdown. Front. Psychol. 2020, 11, 588604. [Google Scholar] [CrossRef] [PubMed]
  14. Werneck, A.O.; Collings, P.J.; Barboza, L.L.; Stubbs, B.; Silva, D.R. Associations of sedentary behaviors and physical activity with social isolation in 100,839 school students: The Brazilian Scholar Health Survey. Gen. Hosp. Psychiatry 2019, 59, 7–13. [Google Scholar] [CrossRef] [PubMed]
  15. Ruiz-Roso, M.B.; Knott-Torcal, C.; Matilla-Escalante, D.C.; Garcimartin, A.; Sampedro-Nunez, M.A.; Davalos, A.; Marazuela, M. COVID-19 lockdown and changes of the dietary pattern and physical activity habits in a cohort of patients with type 2 diabetes mellitus. Nutrients 2020, 12, 2327. [Google Scholar] [CrossRef] [PubMed]
  16. Ruiz-Roso, M.B.; de Carvalho Padilha, P.; Matilla-Escalante, D.C.; Brun, P.; Ulloa, N.; Acevedo-Correa, D.; Peres, W.A.F.; Martorell, M.; Carrilho, T.R.B.; de Oliveira Cardoso, L.; et al. Changes of physical activity and ultra-processed food consumption in adolescents from different countries during Covid-19 pandemic: An observational study. Nutrients 2020, 12, 2289. [Google Scholar] [CrossRef] [PubMed]
  17. Damiot, A.; Pinto, A.J.; Turner, J.E.; Gualano, B. Immunological implications of physical inactivity among older adults during the COVID-19 pandemic. Gerontology 2020, 66, 431–438. [Google Scholar] [CrossRef]
  18. Sallis, R.; Young, D.R.; Tartof, S.Y.; Sallis, J.F.; Sall, J.; Li, Q.; Smith, G.N.; Cohen, D.A. Physical inactivity is associated with a higher risk for severe COVID-19 outcomes: A study in 48,440 adult patients. Br. J. Sports Med. 2021, 55, 1099–1105. [Google Scholar] [CrossRef]
  19. Flanagan, K.L.; Best, E.; Crawford, N.W.; Giles, M.; Koirala, A.; Macartney, K.; Russell, F.; Teh, B.W.; Wen, S.C. Progress and pitfalls in the quest for effective SARS-CoV-2 (COVID-19) vaccines. Front. Immunol. 2020, 11, 579250. [Google Scholar] [CrossRef]
  20. Walsh, N.P.; Gleeson, M.; Pyne, D.B.; Nieman, D.C.; Dhabhar, F.S.; Shephard, R.J.; Oliver, S.J.; Bermon, S.; Kajeniene, A. Position statement. Part two: Maintaining immune health. Exerc. Immunol. Rev. 2011, 17, 64–103. [Google Scholar]
  21. Walsh, N.P.; Gleeson, M.; Shephard, R.J.; Gleeson, M.; Woods, J.A.; Bishop, N.C.; Fleshner, M.; Green, C.; Pedersen, B.K.; Hoffman-Goetz, L.; et al. Position statement. Part one: Immune function and exercise. Exerc. Immunol. Rev. 2011, 17, 6–63. [Google Scholar]
  22. Bortolini, M.J.; Silva, M.V.; Alonso, F.M.; Medeiros, L.A.; Carvalho, F.R.; Costa, L.F.; Silva, N.M.; Penha-Silva, N.; Mineo, T.W.; Mineo, J.R. Strength and aerobic physical exercises are able to increase survival of Toxoplasma gondii-infected C57BL/6 mice by interfering in the IFN-gamma expression. Front. Physiol. 2016, 7, 641. [Google Scholar] [CrossRef] [Green Version]
  23. Schebeleski-Soares, C.; Occhi-Soares, R.C.; Franzoi-de-Moraes, S.M.; de Oliveira Dalalio, M.M.; Almeida, F.N.; de Ornelas Toledo, M.J.; de Araujo, S.M. Preinfection aerobic treadmill training improves resistance against Trypanosoma cruzi infection in mice. Appl. Physiol. Nutr. Metab. 2009, 34, 659–665. [Google Scholar] [CrossRef] [PubMed]
  24. Terra, R.; Alves, P.J.; da Silva, S.A.G.; Salerno, V.P.; Dutra, P.M. Exercise improves the Th1 response by modulating cytokine and NO production in BALB/c mice. Int. J. Sports Med. 2013, 34, 661–666. [Google Scholar] [CrossRef] [PubMed]
  25. Wang, Q.; Zhou, W. Roles and molecular mechanisms of physical exercise in cancer prevention and treatment. J. Sport Health Sci. 2020, 10, 201–210. [Google Scholar] [CrossRef] [PubMed]
  26. Benatti, F.B.; Pedersen, B.K. Exercise as an anti-inflammatory therapy for rheumatic diseases-myokine regulation. Nat. Rev. Rheumatol. 2015, 11, 86–97. [Google Scholar] [CrossRef] [PubMed]
  27. Nyenhuis, S.M.; Greiwe, J.; Zeiger, J.S.; Nanda, A.; Cooke, A. Exercise and Fitness in the Age of Social Distancing during the COVID-19 Pandemic. J. Allergy Clin. Immunol. Pract. 2020, 8, 2152–2155. [Google Scholar] [CrossRef]
  28. Fernandes, P.; de Mendonca Oliveira, L.; Bruggemann, T.R.; Sato, M.N.; Olivo, C.R.; Arantes-Costa, F.M. Physical exercise induces immunoregulation of TREG, M2, and pDCs in a lung allergic inflammation model. Front. Immunol. 2019, 10, 854. [Google Scholar] [CrossRef]
  29. Kohut, M.L.; Cooper, M.M.; Nickolaus, M.S.; Russell, D.R.; Cunnick, J.E. Exercise and psychosocial factors modulate immunity to influenza vaccine in elderly individuals. J. Gerontol. A Biol. Sci. Med. Sci. 2002, 57, M557–M562. [Google Scholar] [CrossRef] [Green Version]
  30. Stenger, T.; Ledo, A.; Ziller, C.; Schub, D.; Schmidt, T.; Enders, M.; Gärtner, B.C.; Sester, M.; Meyer, T. Timing of vaccination after training: Immune response and side effects in athletes. Med. Sci. Sports Exerc. 2020, 52, 1603–1609. [Google Scholar] [CrossRef]
  31. Edwards, K.M.; Burns, V.E.; Allen, L.M.; McPhee, J.S.; Bosch, J.A.; Carroll, D.; Drayson, M.; Ring, C. Eccentric exercise as an adjuvant to influenza vaccination in humans. Brain Behav. Immun. 2007, 21, 209–217. [Google Scholar] [CrossRef]
  32. Zimmermann, P.; Curtis, N. Factors that influence the immune response to vaccination. Clin. Microbiol. Rev. 2019, 32. [Google Scholar] [CrossRef] [Green Version]
  33. Dugan, H.L.; Henry, C.; Wilson, P.C. Aging and influenza vaccine-induced immunity. Cell. Immunol. 2020, 348, 103998. [Google Scholar] [CrossRef] [PubMed]
  34. Guichelaar, T.; Hoeboer, J.; Widjojoatmodjo, M.N.; Reemers, S.S.; van Els, C.A.; Otten, R.; van Remmerden, Y.; Boes, J.; Luytjes, W. Impaired immune response to vaccination against infection with human respiratory syncytial virus at advanced age. J. Virol. 2014, 88, 9744–9750. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Gershon, A.A.; Brooks, D.; Stevenson, D.D.; Chin, W.K.; Oldstone, M.B.A.; Gershon, M.D. High constitutive interleukin 10 level interferes with the immune response to varicella-zoster virus in elderly recipients of live attenuated zoster vaccine. J. Infect. Dis. 2019, 219, 1338–1346. [Google Scholar] [CrossRef] [PubMed]
  36. Beyer, W.E.; Nauta, J.J.; Palache, A.M.; Giezeman, K.M.; Osterhaus, A.D. Immunogenicity and safety of inactivated influenza vaccines in primed populations: A systematic literature review and meta-analysis. Vaccine 2011, 29, 5785–5792. [Google Scholar] [CrossRef] [PubMed]
  37. Andrew, M.K.; McElhaney, J.E. Age and frailty in COVID-19 vaccine development. Lancet 2021, 396, 1942–1944. [Google Scholar] [CrossRef]
  38. Ledo, A.; Schub, D.; Ziller, C.; Enders, M.; Stenger, T.; Gartner, B.C.; Schmidt, T.; Meyer, T.; Sester, M. Elite athletes on regular training show more pronounced induction of vaccine-specific T-cells and antibodies after tetravalent influenza vaccination than controls. Brain Behav. Immun. 2020, 83, 135–145. [Google Scholar] [CrossRef]
  39. Bachi, A.L.; Suguri, V.M.; Ramos, L.R.; Mariano, M.; Vaisberg, M.; Lopes, J.D. Increased production of autoantibodies and specific antibodies in response to influenza virus vaccination in physically active older individuals. Results Immunol. 2013, 3, 10–16. [Google Scholar] [CrossRef] [Green Version]
  40. Wong, G.C.L.; Narang, V.; Lu, Y.; Camous, X.; Nyunt, M.S.Z.; Carre, C.; Tan, C.; Xian, C.H.; Chong, J.; Chua, M.; et al. Hallmarks of improved immunological responses in the vaccination of more physically active elderly females. Exerc. Immunol. Rev. 2019, 25, 20–33. [Google Scholar]
  41. Pedersen, B.K.; Febbraio, M.A. Muscle as an endocrine organ: Focus on muscle-derived interleukin-6. Physiol. Rev. 2008, 88, 1379–1406. [Google Scholar] [CrossRef] [Green Version]
  42. Whitham, M.; Febbraio, M.A. The ever-expanding myokinome: Discovery challenges and therapeutic implications. Nat. Rev. Drug Discov. 2016, 15, 719–729. [Google Scholar] [CrossRef]
  43. Crescioli, C. Targeting age-dependent functional and metabolic decline of human skeletal muscle: The geroprotective role of exercise, myokine IL-6, and vitamin D. Int. J. Mol. Sci. 2020, 21, 1010. [Google Scholar] [CrossRef] [Green Version]
  44. Riechman, S.E.; Balasekaran, G.; Roth, S.M.; Ferrell, R.E. Association of interleukin-15 protein and interleukin-15 receptor genetic variation with resistance exercise training responses. J. Appl. Physiol. 2004, 97, 2214–2219. [Google Scholar] [CrossRef]
  45. Haugen, F.; Norheim, F.; Lian, H.; Wensaas, A.J.; Dueland, S.; Berg, O.; Funderud, A.; Skalhegg, B.S.; Raastad, T.; Drevon, C.A. IL-7 is expressed and secreted by human skeletal muscle cells. Am. J. Physiol. Cell Physiol. 2010, 298, C807–C816. [Google Scholar] [CrossRef] [Green Version]
  46. Raeber, M.E.; Zurbuchen, Y.; Impellizzieri, D.; Boyman, O. The role of cytokines in T-cell memory in health and disease. Immunol. Rev. 2018, 283, 176–193. [Google Scholar] [CrossRef]
  47. Katharina, A.; Christian, P.; Karsten, K. Current knowledge and new challenges in exercise immunology. Ger. J. Sports Med. 2019, 70, 250–260. [Google Scholar] [CrossRef]
  48. Ho, N.I.; Huis In’t Veld, L.G.M.; Raaijmakers, T.K.; Adema, G.J. Adjuvants enhancing cross-oresentation by dendritic cells: The key to more effective vaccines? Front. Immunol. 2018, 9, 2874. [Google Scholar] [CrossRef]
  49. Edwards, K.M.; Pung, M.A.; Tomfohr, L.M.; Ziegler, M.G.; Campbell, J.P.; Drayson, M.T.; Mills, P.J. Acute exercise enhancement of pneumococcal vaccination response: A randomised controlled trial of weaker and stronger immune response. Vaccine 2012, 30, 6389–6395. [Google Scholar] [CrossRef] [Green Version]
  50. Ranadive, S.M.; Cook, M.; Kappus, R.M.; Yan, H.; Lane, A.D.; Woods, J.A.; Wilund, K.R.; Iwamoto, G.; Vanar, V.; Tandon, R.; et al. Effect of acute aerobic exercise on vaccine efficacy in older adults. Med. Sci. Sports Exerc. 2014, 46, 455–461. [Google Scholar] [CrossRef] [Green Version]
  51. Pascoe, A.R.; Singh, M.A.F.; Edwards, K.M. The effects of exercise on vaccination responses: A review of chronic and acute exercise interventions in humans. Brain Behav. Immun. 2014, 39, 33–41. [Google Scholar] [CrossRef]
  52. De Araujo, A.L.; Silva, L.C.; Fernandes, J.R.; Matias Mde, S.; Boas, L.S.; Machado, C.M.; Garcez-Leme, L.E.; Benard, G. Elderly men with moderate and intense training lifestyle present sustained higher antibody responses to influenza vaccine. Age 2015, 37, 105. [Google Scholar] [CrossRef] [Green Version]
  53. Shamsi, M.M.; Najedi, S.; Hassan, Z.M.; Isanejad, A.; Mahdavi, M. Short term exercise training enhances cell-mediated responses to HSV-1 vaccine in mice. Microb. Pathog. 2017, 110, 457–463. [Google Scholar] [CrossRef] [PubMed]
  54. Campbell, J.P.; Turner, J.E. Debunking the myth of exercise-induced immune suppression: Redefining the impact of exercise on immunological health across the lifespan. Front. Immunol. 2018, 9, 648. [Google Scholar] [CrossRef] [PubMed]
  55. Turner, J.E.; Spielmann, G.; Wadley, A.J.; Aldred, S.; Simpson, R.J.; Campbell, J.P. Exercise-induced B cell mobilisation: Preliminary evidence for an influx of immature cells into the bloodstream. Physiol. Behav. 2016, 164, 376–382. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Poinsatte, K.; Smith, E.E.; Torres, V.O.; Ortega, S.B.; Huebinger, R.M.; Cullum, C.M.; Monson, N.L.; Zhang, R.; Stowe, A.M. T and B cell subsets differentially correlate with amyloid deposition and neurocognitive function in patients with amnestic mild cognitive impairment after one year of physical activity. Exerc. Immunol. Rev. 2019, 25, 34–49. [Google Scholar] [PubMed]
  57. MacDonald, K.P.; Munster, D.J.; Clark, G.J.; Dzionek, A.; Schmitz, J.; Hart, D.N. Characterization of human blood dendritic cell subsets. Blood 2002, 100, 4512–4520. [Google Scholar] [CrossRef] [PubMed]
  58. Lou, Y.; Liu, C.; Kim, G.J.; Liu, Y.J.; Hwu, P.; Wang, G. Plasmacytoid dendritic cells synergize with myeloid dendritic cells in the induction of antigen-specific antitumor immune responses. J. Immunol. 2007, 178, 1534–1541. [Google Scholar] [CrossRef] [Green Version]
  59. Chen, H.; Tan, J.; Li, X.; Li, H.; Wu, W.; Wu, Y.; Zhang, J.; Gu, L. Myeloid and plasmacytoid dendritic cell combined vaccines loaded with heat-treated tumor cell lysates enhance antitumor activity in murine lung cancer. Oncol. Lett. 2021, 21, 90. [Google Scholar] [CrossRef]
  60. Fu, C.; Peng, P.; Loschko, J.; Feng, L.; Pham, P.; Cui, W.; Lee, K.P.; Krug, A.B.; Jiang, A. Plasmacytoid dendritic cells cross-prime naive CD8 T cells by transferring antigen to conventional dendritic cells through exosomes. Proc. Natl. Acad. Sci. USA 2020, 117, 23730–23741. [Google Scholar] [CrossRef]
  61. Nickel, T.; Emslander, I.; Sisic, Z.; David, R.; Schmaderer, C.; Marx, N.; Schmidt-Trucksass, A.; Hoster, E.; Halle, M.; Weis, M.; et al. Modulation of dendritic cells and toll-like receptors by marathon running. Eur. J. Appl. Physiol. 2012, 112, 1699–1708. [Google Scholar] [CrossRef]
  62. Brown, F.F.; Campbell, J.P.; Wadley, A.J.; Fisher, J.P.; Aldred, S.; Turner, J.E. Acute aerobic exercise induces a preferential mobilisation of plasmacytoid dendritic cells into the peripheral blood in man. Physiol. Behav. 2018, 194, 191–198. [Google Scholar] [CrossRef] [Green Version]
  63. Abdalla, D.R.; Gomes, B.B.M.; Murta, E.F.C.; Michelin, M.A. Bone marrow-derived dendritic cells under influence of experimental breast cancer and physical activity. Oncol. Lett. 2017, 13, 1406–1410. [Google Scholar] [CrossRef] [Green Version]
  64. Estebanez, B.; Jimenez-Pavon, D.; Huang, C.J.; Cuevas, M.J.; Gonzalez-Gallego, J. Effects of exercise on exosome release and cargo in in vivo and ex vivo models: A systematic review. J. Cell. Physiol. 2021, 236, 3336–3353. [Google Scholar] [CrossRef]
  65. Brahmer, A.; Neuberger, E.; Esch-Heisser, L.; Haller, N.; Jorgensen, M.M.; Baek, R.; Mobius, W.; Simon, P.; Kramer-Albers, E.M. Platelets, endothelial cells and leukocytes contribute to the exercise-triggered release of extracellular vesicles into the circulation. J. Extracell. Vesicles 2019, 8, 1615820. [Google Scholar] [CrossRef]
  66. Macpherson, A.J.; Harris, N.L. Interactions between commensal intestinal bacteria and the immune system. Nat. Rev. Immunol. 2004, 4, 478–485. [Google Scholar] [CrossRef]
  67. Lundin, A.; Bok, C.M.; Aronsson, L.; Bjorkholm, B.; Gustafsson, J.A.; Pott, S.; Arulampalam, V.; Hibberd, M.; Rafter, J.; Pettersson, S. Gut flora, Toll-like receptors and nuclear receptors: A tripartite communication that tunes innate immunity in large intestine. Cell. Microbiol. 2008, 10, 1093–1103. [Google Scholar] [CrossRef]
  68. Hu, G.; Gong, A.Y.; Roth, A.L.; Huang, B.Q.; Ward, H.D.; Zhu, G.; Larusso, N.F.; Hanson, N.D.; Chen, X.M. Release of luminal exosomes contributes to TLR4-mediated epithelial antimicrobial defense. PLoS Pathog. 2013, 9, e1003261. [Google Scholar] [CrossRef] [Green Version]
  69. Bermon, S.; Petriz, B.; Kajeniene, A.; Prestes, J.; Castell, L.; Franco, O.L. The microbiota: An exercise immunology perspective. Exerc. Immunol. Rev. 2015, 21, 70–79. [Google Scholar]
  70. Oliveira, G.P., Jr.; Porto, W.F.; Palu, C.C.; Pereira, L.M.; Petriz, B.; Almeida, J.A.; Viana, J.; Filho, N.N.A.; Franco, O.L.; Pereira, R.W. Effects of Acute Aerobic Exercise on Rats Serum Extracellular Vesicles Diameter, Concentration and Small RNAs Content. Front. Physiol. 2018, 9, 532. [Google Scholar] [CrossRef] [Green Version]
  71. Ramasamy, M.N.; Minassian, A.M.; Ewer, K.J.; Flaxman, A.L.; Folegatti, P.M.; Owens, D.R.; Voysey, M.; Aley, P.K.; Angus, B.; Babbage, G.; et al. Safety and immunogenicity of ChAdOx1 nCoV-19 vaccine administered in a prime-boost regimen in young and old adults (COV002): A single-blind, randomised, controlled, phase 2/3 trial. Lancet 2021, 396, 1979–1993. [Google Scholar] [CrossRef]
  72. Xia, S.; Duan, K.; Zhang, Y.; Zhao, D.; Zhang, H.; Xie, Z.; Li, X.; Peng, C.; Zhang, Y.; Zhang, W.; et al. Effect of an inactivated vaccine against SARS-CoV-2 on safety and immunogenicity outcomes: Interim analysis of 2 randomized clinical trials. JAMA 2020, 324, 951–960. [Google Scholar] [CrossRef]
  73. Voysey, M.; Clemens, S.A.C.; Madhi, S.A.; Weckx, L.Y.; Folegatti, P.M.; Aley, P.K.; Angus, B.; Baillie, V.L.; Barnabas, S.L.; Bhorat, Q.E.; et al. Safety and efficacy of the ChAdOx1 nCoV-19 vaccine (AZD1222) against SARS-CoV-2: An interim analysis of four randomised controlled trials in Brazil, South Africa, and the UK. Lancet 2021, 397, 99–111. [Google Scholar] [CrossRef]
  74. Walsh, E.E.; Frenck, R.W., Jr.; Falsey, A.R.; Kitchin, N.; Absalon, J.; Gurtman, A.; Lockhart, S.; Neuzil, K.; Mulligan, M.J.; Bailey, R.; et al. Safety and immunogenicity of two RNA-based Covid-19 vaccine candidates. N. Engl. J. Med. 2020, 383, 2439–2450. [Google Scholar] [CrossRef] [PubMed]
  75. Anderson, E.J.; Rouphael, N.G.; Widge, A.T.; Jackson, L.A.; Roberts, P.C.; Makhene, M.; Chappell, J.D.; Denison, M.R.; Stevens, L.J.; Pruijssers, A.J.; et al. Safety and immunogenicity of SARS-CoV-2 mRNA-1273 vaccine in older adults. N. Engl. J. Med. 2020, 383, 2427–2438. [Google Scholar] [CrossRef] [PubMed]
  76. Zhang, Y.; Zeng, G.; Pan, H.; Li, C.; Hu, Y.; Chu, K.; Han, W.; Chen, Z.; Tang, R.; Yin, W.; et al. Safety, tolerability, and immunogenicity of an inactivated SARS-CoV-2 vaccine in healthy adults aged 18–59 years: A randomised, double-blind, placebo-controlled, phase 1/2 clinical trial. Lancet Infect. Dis. 2021, 21, 181–192. [Google Scholar] [CrossRef]
  77. Mallapaty, S.; Ledford, H. COVID-vaccine results are on the way—And scientists’ concerns are growing. Nature 2020, 586, 16–17. [Google Scholar] [CrossRef]
  78. Jamrozik, E.; Selgelid, M.J. COVID-19 human challenge studies: Ethical issues. Lancet Infect. Dis. 2020, 20, e198–e203. [Google Scholar] [CrossRef]
  79. Callaway, E. COVID vaccine boosters: The most important questions. Nature 2021, 596, 178–180. [Google Scholar] [CrossRef]
Figure 1. How acute and chronic physical exercise could interfere with immune system and vaccine efficacy, based on published studies. (A) subjects who exercise regularly and performed acute exercise just before vaccination; (B) subjects who exercise regularly and did not perform acute exercise just before vaccination; (C) subjects with a sedentary lifestyle who performed acute exercise just before vaccination; (D) subjects with a sedentary lifestyle who did not perform acute exercise just before vaccination. (E) List of main influences of physical exercise on immune system. Pre-vaccination is defined as days or months before vaccination; vaccination is defined as the moment of injection; chronic exercise is defined as a regular physical exercise in months or years; acute exercise is defined as moderate/vigorous physical exercise a few minutes or hours before vaccination.
Figure 1. How acute and chronic physical exercise could interfere with immune system and vaccine efficacy, based on published studies. (A) subjects who exercise regularly and performed acute exercise just before vaccination; (B) subjects who exercise regularly and did not perform acute exercise just before vaccination; (C) subjects with a sedentary lifestyle who performed acute exercise just before vaccination; (D) subjects with a sedentary lifestyle who did not perform acute exercise just before vaccination. (E) List of main influences of physical exercise on immune system. Pre-vaccination is defined as days or months before vaccination; vaccination is defined as the moment of injection; chronic exercise is defined as a regular physical exercise in months or years; acute exercise is defined as moderate/vigorous physical exercise a few minutes or hours before vaccination.
Ijerph 19 01853 g001
Figure 2. Proposed model for analyzing physical activity level among volunteers with indirect (questionnaire) or direct (accelerometer) method in clinical trials for vaccines. (A) Assessment of volunteer’s level of physical activity by questionnaire. Researcher should apply the same questionnaire at least twice: at first dose and same day as last dose. (B) Evaluation of physical activity level by accelerometry. Researcher should proceed similarly to questionnaire method. If vaccine is single dose, second assessment of both cases must be carried out 30 days later. Accelerometer assessment requires several days.
Figure 2. Proposed model for analyzing physical activity level among volunteers with indirect (questionnaire) or direct (accelerometer) method in clinical trials for vaccines. (A) Assessment of volunteer’s level of physical activity by questionnaire. Researcher should apply the same questionnaire at least twice: at first dose and same day as last dose. (B) Evaluation of physical activity level by accelerometry. Researcher should proceed similarly to questionnaire method. If vaccine is single dose, second assessment of both cases must be carried out 30 days later. Accelerometer assessment requires several days.
Ijerph 19 01853 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Bortolini, M.J.S.; Petriz, B.; Mineo, J.R.; Resende, R.d.O. Why Physical Activity Should Be Considered in Clinical Trials for COVID-19 Vaccines: A Focus on Risk Groups. Int. J. Environ. Res. Public Health 2022, 19, 1853. https://doi.org/10.3390/ijerph19031853

AMA Style

Bortolini MJS, Petriz B, Mineo JR, Resende RdO. Why Physical Activity Should Be Considered in Clinical Trials for COVID-19 Vaccines: A Focus on Risk Groups. International Journal of Environmental Research and Public Health. 2022; 19(3):1853. https://doi.org/10.3390/ijerph19031853

Chicago/Turabian Style

Bortolini, Miguel Junior Sordi, Bernardo Petriz, José Roberto Mineo, and Rafael de Oliveira Resende. 2022. "Why Physical Activity Should Be Considered in Clinical Trials for COVID-19 Vaccines: A Focus on Risk Groups" International Journal of Environmental Research and Public Health 19, no. 3: 1853. https://doi.org/10.3390/ijerph19031853

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop