Immune Response after Vaccination against SARS-COV-2 in Patients with Chronic Diseases

A special issue of Vaccines (ISSN 2076-393X). This special issue belongs to the section "COVID-19 Vaccines and Vaccination".

Deadline for manuscript submissions: closed (31 December 2022) | Viewed by 21435

Special Issue Editor


E-Mail Website
Guest Editor
1. Head of Nephrology Department, Hillel Yaffe Medical Center, Hadera 38100, Israel
2. Rappaport Faculty of Medicine, Technion Israel, Haifa 32000, Israel
Interests: immune response to COVID-19 vaccine in patients with End Stage Kidney Disease; hemodialysis; acute kidney injury

Special Issue Information

Dear Colleagues,

The enormous toll of the SARS-CoV-2 pandemic on human lives, and its effects on morbidity, mortality and global economic disruption are not yet over. Vaccination was and still is the cornerstone and most effective measure in the global effort to end the pandemic. Patient groups with certain pre-existing medical conditions may have higher risks for adverse outcomes of SARS-CoV-2. These same patient groups may also have diminished response to SARS-CoV-2 vaccination. These groups were not included in the pivotal clinical trials of the vaccines, and for them, more information is being acquired from smaller-scale clinical trials. The management of COVID-19 in these populations may require specific adjustments of vaccination and other therapies.

This Special Issue aims to update the current knowledge and future directions of SARS-CoV-2 vaccination and management in patients with chronic medical conditions (patients with solid/hematological malignancies, those who have received organ transplants, patients on dialysis, those receiving immunosuppressive medications, elderly individuals, individuals with morbid obesity, and possibly other patient groups)

In this Special Issue, original research articles and reviews are welcome. Research areas may include (but are not limited to) the following:

  • Immune response to SARS-CoV-2 vaccines in patients with chronic diseases.
  • Vaccination strategy and vaccine classes (additional and booster vaccine shots, combination of vaccine classes).
  • Efficacy of vaccination in patients with chronic medical conditions: laboratory and/or clinical evaluation.
  • SARS-CoV-2 management of the immunocompromised beyond vaccination.
  • Other related topics.

Dr. Noa Berar Yanay
Guest Editor

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Vaccines is an international peer-reviewed open access monthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2700 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • COVID-19 vaccination response
  • COVID-19 vaccination efficacy
  • chronic diseases
  • immunosuppressed patients
  • vaccination strategy
  • COVID-19 treatment
  • COVID-19 prevention

Published Papers (9 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Other

12 pages, 1221 KiB  
Article
Immune Response to CoronaVac and Its Safety in Patients with Type 2 Diabetes Compared with Healthcare Workers
by Bothamai Dechates, Thachanun Porntharukchareon, Supamas Sirisreetreerux, Phonthip Therawit, Supanat Worawitchawong, Gaidganok Sornsamdang, Kamonwan Soonklang and Kriangkrai Tawinprai
Vaccines 2023, 11(3), 684; https://doi.org/10.3390/vaccines11030684 - 17 Mar 2023
Cited by 1 | Viewed by 1061
Abstract
Background: Vaccines for SARS-CoV-2 have been critical for preventing disease. Previous research showed patients with diabetes have impaired immunity. This study aimed to determine the immunity to coronavirus after CoronaVac by comparing patients with type 2 diabetes (T2D) and healthcare workers (HCW). Materials [...] Read more.
Background: Vaccines for SARS-CoV-2 have been critical for preventing disease. Previous research showed patients with diabetes have impaired immunity. This study aimed to determine the immunity to coronavirus after CoronaVac by comparing patients with type 2 diabetes (T2D) and healthcare workers (HCW). Materials and methods: A prospective cohort study evaluated immune responses and safety after two doses of CoronaVac in T2D and HCW groups at Chulabhorn Hospital. The levels of total antibodies against the receptor-binding domain (anti-RBD) of the SARS-CoV-2 spike protein at baseline and 4 weeks after vaccination were collected. The level of anti-RBD concentrations was reported as geometric mean concentration (GMC) and compared between groups using the geometric mean ratio (GMR). Results: 81 participants were included; 27 had T2D and 54 were HCW. After complete vaccination, anti-RBD concentrations were not significantly different between T2D (57.68 binding antibody units (BAU)/mL, 95% confidence interval (CI) = 29.08; 114.44) and HCW (72.49 BAU/mL, 95% CI = 55.77; 94.22) groups. Subgroup analysis showed the GMC of anti-RBD was significantly lower in T2D patients with dyslipidaemia (50.04 BAU/mL) than in T2D patients without dyslipidaemia (341.64 BAU/mL). Conclusions: The immune response at 4 weeks after two doses of CoronaVac did not significantly differ between patients with T2D and HCW. Full article
Show Figures

Figure 1

11 pages, 1047 KiB  
Article
Evaluation of Safety and Immunogenicity of a Recombinant Receptor-Binding Domain (RBD)-Tetanus Toxoid (TT) Conjugated SARS-CoV-2 Vaccine (PastoCovac) in Recipients of Autologous Hematopoietic Stem Cell Transplantation Compared to the Healthy Controls; A Prospective, Open-Label Clinical Trial
by Maryam Barkhordar, Mohammad Ahmadvand, Leyla Sharifi Aliabadi, Seied Saeid Noorani, Fahimeh Bagheri Amiri, Ghasem Janbabai, Rahim Sorouri, Mona Asadi Milani and Mohammad Vaezi
Vaccines 2023, 11(1), 117; https://doi.org/10.3390/vaccines11010117 - 03 Jan 2023
Cited by 2 | Viewed by 1829
Abstract
Background: The urgent need for prompt SARS-CoV-2 immunization of hematopoietic stem cell transplant (HSCT) recipients in an endemic area raises many challenges regarding selecting a vaccine platform appropriate for HSCT recipients being economical for widespread use in developing countries. Methods: The [...] Read more.
Background: The urgent need for prompt SARS-CoV-2 immunization of hematopoietic stem cell transplant (HSCT) recipients in an endemic area raises many challenges regarding selecting a vaccine platform appropriate for HSCT recipients being economical for widespread use in developing countries. Methods: The trial is a prospective, single-group, open-label study to investigate the safety and serologic response of two doses of the recombinant receptor-binding domain (RBD)-Tetanus Toxoid (TT) conjugated SARS-CoV-2 vaccine (PastoCovac) early after autologous (auto) HSCT. For this reason, a total of 38 patients who completed the two-dose SARS-CoV-2 RBD-based vaccine between three to nine months after auto-HSCT and had an available anti-spike serologic test at three predefined time points of baseline and after the first and second doses and 50 healthy control individuals were included in the analysis. The primary outcome was defined as an increase in IgG Immune status ratio (ISR) to the cut-off value for the positive result (≥1.1) in the semiquantitative test. Findings: The median time between auto-HSCT and vaccination was 127 days. No participant reported any significant adverse effects (Grade 3). Pain at the injection site was the most common adverse event. The ISR increased significantly (p < 0.001) during the three-time point sampling for both patients and healthy control groups. In patients, the mean ISR increased from 1.39 (95% CI: 1.13–1.65) at baseline to 2.48 (1.93–3.03) and 3.73 (3.13–4.38) following the first and second dosages, respectively. In multivariate analysis, the higher count of lymphocytes [OR: 8.57 (95% CI: 1.51–48.75); p = 0.02] and history of obtaining COVID-19 infection before transplantation [OR: 6.24 (95% CI: 1.17–33.15); p = 0.03] remained the predictors of the stronger immune response following two doses of the RBD-TT conjugated vaccine. Moreover, we found that the immunogenicity of the COVID-19 vaccine shortly after transplantation could be influenced by pre-transplant COVID-19 vaccination. Interpretation: The RBD-TT conjugated SARS-CoV-2 vaccine was safe, highly immunogenic, and affordable early after autologous transplants. Funding: This work was mainly financed by the Hematology-Oncology-Stem Cell Transplantation Research Center (HORCSCT) of Tehran University and the Pasteur Institute of Iran. Full article
Show Figures

Figure 1

11 pages, 1416 KiB  
Article
Immune Response to SARS-CoV-2 mRNA Vaccines in an Open-Label Multicenter Study in Participants with Relapsing Multiple Sclerosis Treated with Ofatumumab
by Tjalf Ziemssen, Marie Groth, Benjamin Ettle and Tobias Bopp
Vaccines 2022, 10(12), 2167; https://doi.org/10.3390/vaccines10122167 - 16 Dec 2022
Cited by 7 | Viewed by 2236
Abstract
Background: It is unclear whether multiple sclerosis (MS) patients receiving ofatumumab mount an immune response after SARS-CoV-2 mRNA vaccination. Methods: KYRIOS is an ongoing, multicenter, open-label, prospective clinical study on immune responses in MS patients after initial or booster SARS-CoV-2 mRNA vaccination prior [...] Read more.
Background: It is unclear whether multiple sclerosis (MS) patients receiving ofatumumab mount an immune response after SARS-CoV-2 mRNA vaccination. Methods: KYRIOS is an ongoing, multicenter, open-label, prospective clinical study on immune responses in MS patients after initial or booster SARS-CoV-2 mRNA vaccination prior to (cohort 1) or during (cohort 2) ofatumumab treatment. We report one-week and one-month results of the initial vaccination. A comparison with patients vaccinated while receiving beta-interferon, glatiramer acetate, dimethyl fumarate, teriflunomide or no treatment was included (cohort 3). Results: In total, 11 patients received their initial vaccination during the study. The primary endpoint of SARS-CoV-2-specific T-cells at month 1 was reached by 80.0% of patients in cohort 1 (N = 6) and 100.0% in cohort 2 (N = 5). T-cell reactivity peaked at week 1. All cohort 1 patients reached seroconversion for SARS-CoV-2 neutralizing antibodies at week 1 and month 1. In cohort 2, neutralizing antibodies increased in all patients and exceeded the cut-off for seropositivity in 40.0% of patients at week 1 and 25.0% at month 1. Immune responses in cohort 3 were comparable to cohort 1. Conclusion: Presence of T-cell response and increase in levels of neutralizing antibodies, although less pronounced compared to controls, suggest that MS patients receiving ofatumumab are able to mount an immune response to SARS-CoV-2 mRNA vaccination. Full article
Show Figures

Figure 1

13 pages, 786 KiB  
Article
Safety and Humoral and Cellular Immunogenicity of the BNT162b2 SARS-CoV-2 Vaccine in Liver-Transplanted Adolescents Compared to Healthy Adolescents
by Palittiya Sintusek, Supranee Buranapraditkun, Siriporn Khunsri, Varattaya Saengchaisukhonkit, Preeyaporn Vichaiwattana, Donchida Srimuan, Thanunrat Thongmee and Yong Poovorawan
Vaccines 2022, 10(8), 1324; https://doi.org/10.3390/vaccines10081324 - 16 Aug 2022
Cited by 5 | Viewed by 1561
Abstract
Since BNT162b2 was approved to prevent COVID-19 in children, we aim to compare the safety and immunogenicity of the BNT162b2 vaccine in liver-transplanted (LT) and healthy adolescents. LT and healthy adolescents received two doses of 30 µg of BNT162b2. All were evaluated for [...] Read more.
Since BNT162b2 was approved to prevent COVID-19 in children, we aim to compare the safety and immunogenicity of the BNT162b2 vaccine in liver-transplanted (LT) and healthy adolescents. LT and healthy adolescents received two doses of 30 µg of BNT162b2. All were evaluated for total COVID-19 antibodies directed against the receptor-binding domain (RBD) and interferon-γ using the ELISpot at all time points; anti-nucleocapsid immunoglobulin was evaluated at week 8 and the surrogate virus-neutralizing antibody (sVN) to Omicron at day 0 and week 8. Adverse effects were recorded during days 0–7. In total, 16 LT and 27 healthy adolescents were enrolled (aged 14.78 ± 1.70 years). After completion, all LT and healthy adolescents were positive for anti-RBD immunoglobulin, with geometric mean titers of 1511.37 (95% CI 720.22–3171.59) and 6311.90 (95% CI 4955.46–8039.64)) U/mL (p < 0.001). All tested negative for anti-nucleocapsid immunoglobulin, indicating no COVID-19 infection after vaccination. However, the sVNs to Omicron were positive in only nine (33.33%) healthy adolescents and none of the LT adolescents. Interferon-γ-secreting cells were lower in LT adolescents than healthy adolescents. The LT adolescents had a lower immunogenic response to BNT162b2 than the healthy adolescents. Administrating two doses of BNT162b2 was safe, but was less effective against the Omicron variant. Full article
Show Figures

Figure 1

14 pages, 2597 KiB  
Article
BNT162b2 Booster Vaccination Induced Immunity against SARS-CoV-2 Variants among Hemodialysis Patients
by Michal Herman-Edelstein, Naomi Ben-Dor, Timna Agur, Tali Guetta, Annat Raiter, Eshcar Meisel, Weaam Alkeesh, Yaacov Ori, Benaya Rozen-Zvi and Boris Zingerman
Vaccines 2022, 10(6), 967; https://doi.org/10.3390/vaccines10060967 - 17 Jun 2022
Cited by 11 | Viewed by 1974
Abstract
Background: The emergence of new SARS-CoV-2 variants, which evade immunity, has raised the urgent need for multiple vaccine booster doses for vulnerable populations. In this study, we aimed to estimate the BNT162b2 booster effectiveness against the spread of coronavirus variants in a hemodialysis [...] Read more.
Background: The emergence of new SARS-CoV-2 variants, which evade immunity, has raised the urgent need for multiple vaccine booster doses for vulnerable populations. In this study, we aimed to estimate the BNT162b2 booster effectiveness against the spread of coronavirus variants in a hemodialysis population. Methods: We compared humoral and cell-mediated immunity in 100 dialysis patients and 66 age-matched volunteers, before and 2–3 weeks following the first booster vaccine dose. Participants were assessed for anti-spike (RBD) antibody titer, neutralizing antibodies against B.1.617.2 (Delta) and B.1.1.529 (Omicron) variants, spike-specific T-cell responses by FACS and infection outbreak after the first and second booster. Results: Anti-spike antibody titer was significantly increased following the booster, with reduced humoral and cellular response in the dialysis patients. Neutralizing antibody levels increased significantly after the booster dose, with an inferior effect (≤2 fold) against Omicron compared with the Delta variant. Furthermore, CD4+ and CD8+ T-cell activation by Delta spike protein was preserved in 70% of PBMCs from the dialysis patients. A second booster dose tended to reduce breakthrough infections in the dialysis patients. Conclusions: Until the release of an updated vaccine, BNT162b2 booster doses will improve the humoral and cell-mediated immunity against variants. These findings support the importance of repetitive booster doses for hemodialysis patients. Full article
Show Figures

Figure 1

10 pages, 847 KiB  
Article
ChAdOx1 nCoV-19 Immunogenicity and Immunological Response Following COVID-19 Infection in Patients Receiving Maintenance Hemodialysis
by Wisit Prasithsirikul, Tanawin Nopsopon, Phanupong Phutrakool, Pawita Suwanwattana, Piyawat Kantagowit, Wannarat Pongpirul, Anan Jongkaewwattana and Krit Pongpirul
Vaccines 2022, 10(6), 959; https://doi.org/10.3390/vaccines10060959 - 16 Jun 2022
Cited by 3 | Viewed by 1994
Abstract
Patients with end-stage renal disease (ESRD) receiving hemodialysis (HD) were found to have a decreased immune response following mRNA COVID-19 immunization. ChAdOx1 nCoV-19 was a promising COVID-19 vaccine that performed well in the general population, but the evidence on immunogenicity in ESRD with [...] Read more.
Patients with end-stage renal disease (ESRD) receiving hemodialysis (HD) were found to have a decreased immune response following mRNA COVID-19 immunization. ChAdOx1 nCoV-19 was a promising COVID-19 vaccine that performed well in the general population, but the evidence on immunogenicity in ESRD with HD patients was limited. Moreover, the immunological response to COVID-19 infection was inconclusive in patients with ESRD and HD. The aim of this study was to investigate the immunogenicity of ChAdOx1 nCoV-19 vaccination and the immunological response after COVID-19 infection in ESRD patients with HD. The blood samples were obtained at baseline, 1-month, and 3-month follow-up after each shot or recovery. All participants were measured for anti-spike IgG by the ELISA method, using Euroimmun. This study found a significant increase in anti-spike IgG after 1 month of two-shot ChAdOx1 nCoV-19 vaccination, followed by a significant decrease after 3 months. On the other hand, the anti-spike IgG was maintained in the post-recovery group. There was no significant difference in the change of anti-spike IgG between the one-shot ChAdOx1 nCoV-19-vaccinated and post-recovery groups for both 1-month and 3-month follow-ups. The seroconversion rate for the vaccinated group was 60.32% at 1 month after one-shot vaccination and slightly dropped to 58.73% at the 3-month follow-up, then was 92.06% at 1 month after two-shot vaccination and reduced to 82.26% at the 3-month follow-up. For the recovered group, the seroconversion rate was 95.65% at 1 month post-recovery and 92.50% at 3-month follow-up. This study demonstrated the immunogenicity of two-dose ChAdOx1 nCoV-19 in ESRD patients with HD for humoral immunity. After COVID-19 infection, the humoral immune response was strong and could be maintained for at least three months. Full article
Show Figures

Figure 1

10 pages, 809 KiB  
Article
Comparison of Immunogenicity and Safety of Inactivated, Adenovirus-Vectored, and Heterologous Adenovirus-Vectored/mRNA Vaccines in Patients with Systemic Lupus Erythematosus and Rheumatoid Arthritis: A Prospective Cohort Study
by Theerada Assawasaksakul, Tanat Lertussavavivat, Seelwan Sathitratanacheewin, Nont Oudomying, Preeyaporn Vichaiwattana, Nasamon Wanlapakorn, Yong Poovorawan, Yingyos Avihingsanon, Nawaporn Assawasaksakul, Supranee Buranapraditkun and Wonngarm Kittanamongkolchai
Vaccines 2022, 10(6), 853; https://doi.org/10.3390/vaccines10060853 - 26 May 2022
Cited by 13 | Viewed by 4584
Abstract
Background: Impaired immune responses to COVID-19 vaccines have been observed in autoimmune rheumatic disease patients. Determining the most effective and safe vaccine regimen is critically needed in such a population. We aim to compare the immunogenicity and safety of three COVID-19 vaccine regimens [...] Read more.
Background: Impaired immune responses to COVID-19 vaccines have been observed in autoimmune rheumatic disease patients. Determining the most effective and safe vaccine regimen is critically needed in such a population. We aim to compare the immunogenicity and safety of three COVID-19 vaccine regimens in patients with systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA). Methods: SLE and RA patients aged 18–65 years who received inactivated (CoronaVac or COVILO), adenovirus-vectored (AZD1222), or heterogeneous (AZD1222/BNT162b2) vaccines were enrolled. Humoral and cellular immune responses were assessed at day 28 after the second vaccination. This was performed using the serum binding antibody level against the receptor-binding domain of the SARS-CoV-2 spike protein (anti-RBD Ig) and IFNy-ELISpot assay (ELISpot), respectively. Reactogenicity was reviewed on day 7 following each vaccination. Disease activity was assessed before and on day 28 after the second vaccination. Results: The cohort consisted of 94 patients (64 SLE and 30 RA). Inactivated, AZD1222, and AZD1222/BNT162b2 vaccines were administered to 23, 43, and 28 patients, respectively. Anti-RBD titers were lowest in the inactivated vaccine group (2.84 AU/mL; 95% CI 0.96–8.44), followed by AZD1222 (233.7 AU/mL; 95% CI 99.0–505.5), and AZD1222/BNT162b2 (688.6 AU/mL; 95% CI 271–1745), p < 0.0001. After adjusting for relevant factors, the inactivated vaccine was associated with the lowest humoral response, while adenovirus-vectored/mRNA vaccine was the highest. The proportion of positive ELISpot test was also lowest in the inactivated vaccine group (27%), followed by the adenovirus-vectored vaccine (67%), and the adenovirus-vectored/mRNA vaccine (73%) (p = 0.03). All types of vaccine were well-tolerated. There was no flare of autoimmune disease post-vaccination. Conclusion: Adenovirus-vectored and adenovirus-vectored/mRNA vaccines elicited a stronger humoral and cellular immune response than inactivated vaccines, suggesting that they may be more suitable in SLE and RA patients receiving immunosuppressive therapy. Full article
Show Figures

Figure 1

Other

Jump to: Research

9 pages, 1422 KiB  
Case Report
Membranous Nephropathy following Full-Dose of Inactivated SARS-CoV-2 Virus Vaccination: A Case Report and Literature Review
by Theerachai Thammathiwat, Laor Chompuk, Suchin Worawichawong, Vijitr Boonpucknavig, Supinda Sirilak, Sutatip Pongcharoen, Watchara Pichitsiri and Talerngsak Kanjanabuch
Vaccines 2023, 11(1), 80; https://doi.org/10.3390/vaccines11010080 - 29 Dec 2022
Cited by 5 | Viewed by 2291
Abstract
Vaccination against the SARS-CoV-2 virus (COVID-19) has proven to be the most effective measure to prevent the spread and reduce infection severity. A case report of de novo membranous nephropathy (MN) following immunization with inactivated virus vaccine (CoronaVac®, Sinovac Biotech) is [...] Read more.
Vaccination against the SARS-CoV-2 virus (COVID-19) has proven to be the most effective measure to prevent the spread and reduce infection severity. A case report of de novo membranous nephropathy (MN) following immunization with inactivated virus vaccine (CoronaVac®, Sinovac Biotech) is presented here. A 53-year-old man presented with a sudden onset of leg edema a week after receiving an inactivated virus vaccine and a relapse of nephrotic syndrome (NS) with acute kidney injury (AKI) after a booster dose. Screening for serum anti-phospholipase A2 receptor antibody and secondary causes of MN were negative. Kidney biopsy revealed an early MN pattern with focal spike formation, whilst numerous subepithelial electron-dense deposits and a few small deposits in the mesangial area were observed through electron microscopy. A short course of steroids and oral cyclophosphamide was prescribed, resulting in the complete remission of NS and AKI. MN following SARS-CoV-2 vaccination should call for medical importance. Awareness of the association between vaccination and MN should be kept in mind to avoid unnecessary treatment with long-term immunosuppressive agents. Full article
Show Figures

Figure 1

11 pages, 1774 KiB  
Systematic Review
Effectiveness of COVID-19 Vaccines in Adults with Diabetes Mellitus: A Systematic Review
by Jesse M. van den Berg, Sharon Remmelzwaal, Marieke T. Blom, Beryl A. C. E. van Hoek, Karin M. A. Swart, Jetty A. Overbeek, George L. Burchell, Ron M. C. Herings and Petra J. M. Elders
Vaccines 2023, 11(1), 24; https://doi.org/10.3390/vaccines11010024 - 22 Dec 2022
Cited by 12 | Viewed by 3206
Abstract
Persons with diabetes mellitus may have an increased risk of severe illness or death from COVID-19 compared to persons without diabetes. Prior studies indicate that immune response and thus vaccine effectiveness might be lower in persons with diabetes. We aimed to systematically review [...] Read more.
Persons with diabetes mellitus may have an increased risk of severe illness or death from COVID-19 compared to persons without diabetes. Prior studies indicate that immune response and thus vaccine effectiveness might be lower in persons with diabetes. We aimed to systematically review the effectiveness of COVID-19 vaccines in adults with diabetes. Pubmed, Embase, Web of Science and Cochrane Library were searched for studies that evaluated the effectiveness of COVID-19 vaccines in adults with diabetes, published before 4 March 2022. Risk of bias in the included studies was evaluated using the ROBINS-I tool. At least two reviewers conducted the study selection, data extraction, and risk of bias assessment independently. After screening of 2196 studies, a total of 17 articles were included. Six different COVID-19 vaccines (Ad5-nCoV-S, AZD1222, BNT162b2, CoronaVac, JNJ-78436735, and mRNA-1273) were included in the synthesis. Vaccine effectiveness was reported for SARS-CoV-2 infection, symptomatic COVID-19, hospitalization, and death, and ranged from 24 to 96% in persons with diabetes, and from 33 to 97% in total study populations; effectiveness was generally lower for persons with diabetes. Odds ratios for breakthrough infection or severe COVID-19 ranged from 1.03 to 2.41 in vaccinated persons with diabetes compared to persons without diabetes. Even though the included studies were very heterogeneous, results from the synthesis indicate that effectiveness of COVID-19 vaccines might be lower in persons with diabetes. More research is needed on the comparison of vaccine effectiveness between persons with and without diabetes, and the effectiveness of repeat COVID-19 vaccinations. Full article
Show Figures

Figure 1

Back to TopTop