Next Article in Journal
State of the Art in Robotic Surgery with Hugo RAS System: Feasibility, Safety and Clinical Applications
Previous Article in Journal
Feasibility of a Virtual-Reality-Enabled At-Home Telerehabilitation Program for Stroke Survivors: A Case Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

COVID-19 Vaccines and Assisted Reproductive Techniques: A Systematic Review

by
Elena Satorres-Pérez
,
Alicia Martínez-Varea
* and
José Morales-Roselló
Department of Obstetrics and Gynaecology, La Fe University and Polytechnic Hospital, Avenida Fernando Abril Martorell 106, 46026 Valencia, Spain
*
Author to whom correspondence should be addressed.
J. Pers. Med. 2023, 13(8), 1232; https://doi.org/10.3390/jpm13081232
Submission received: 18 July 2023 / Revised: 2 August 2023 / Accepted: 3 August 2023 / Published: 4 August 2023
(This article belongs to the Section Epidemiology)

Abstract

:
Objective: To review the current knowledge concerning COVID-19 vaccination and assisted reproductive techniques (ART). Methods: A systematic review in Pubmed-Medline, the Cochrane Database, the Web of Science, and the National Guideline was performed. Studies were selected if they were primary studies, included vaccinated (case) and unvaccinated (control) patients, and described fertility treatment response. Results: A total of 24 studies were selected. Outcomes related to the association between COVID-19 vaccination and ART were collected. The vast majority of studies found no statistical differences concerning oocyte stimulation response, embryo quality, implantation rates, or pregnancy outcome (clinical or biochemical pregnancy rates and losses) when comparing cases and controls. Similarly, no differences were found when comparing different types of vaccines or distinct ART (artificial insemination, in vitro fertilization, and embryo transfer of frozen embryos). Conclusions: Patients receiving ART and health care professionals should be encouraged to complete and recommend COVID-19 vaccination, as the available evidence regarding assisted reproductive outcomes is reassuring.

Graphical Abstract

1. Introduction

SARS-CoV-2 is a single, positive-strand RNA virus [1]. Until the first identification of human coronaviruses in the 1960s, coronavirus infections were known to be inoffensive to humans [2]. Since then, the total number of human coronaviruses identified has increased throughout the years. While most infections cause minor respiratory symptoms, some may cause major problems, especially in high-risk patients such as the elderly, infants, immunodeficient patients, and individuals with chronic respiratory diseases [3,4].
SARS-CoV-2 was first identified in Wuhan (China) in late 2019. By the end of January 2020, 7734 cases had been confirmed in China, and 90 other cases had also been reported in 19 different countries. The mortality rate was estimated to be 2.2% [5]. According to the World Health Organization, by 19 July 2023, there will have been 768,237,788 confirmed cases of COVID-19, including 6,951,677 deaths reported [6].
Since the declaration of the COVID-19 pandemic in March 2020, researchers have aimed to answer healthcare professionals’ and patients’ questions concerning the infection: its origin, transmission, mechanism, and management.
To control the sanitary crisis, diverse vaccines were developed worldwide. The FDA has approved four formulations and has helped, through massive vaccination, to constrain the pandemic [7]. According to the World Health Organization, as of 23 July 2023, a total of 13,474,348,801 vaccine doses had been administered [6]. It is interesting to discern the different kinds of developed vaccines and their mechanisms of action:
  • Inactivated vaccines have been widely used and are well-established for infections such as influenza [8] and poliovirus [9]. These vaccines are produced by disarming pathogens through heat, radiation, or chemicals such as formalin or formaldehyde in order to maintain their immunogenicity but block their capability to replicate and infect [10].
  • Quite the reverse, COVID mRNA vaccines are the first to be approved with this formulation. They use the host’s own cellular function to synthesize a viral protein product and establish protective immunity [11,12].
  • Recombinant subunit vaccines contain fragments (proteins, polysaccharides, etc.) of the pathogen. These parts are enough to trigger the host’s immunity, although they could be less immunogenic than other types of vaccination. Examples of subunit vaccines are hepatitis B, clostridium tetani, or papillomavirus [10].
  • Non-replicating viral vector vaccines use an innovative approach to create host immunity. They use the capacity of other viruses with heterologous antigens to infect cells and induce an antigen-specific humoral and cellular immune response [13].
The global sanitary crisis aroused many questions and uncertainties about the long-term consequences and adverse effects of suffering from the infection. Additionally, the rapid development of the above-mentioned vaccines awakened distrust in a large part of the population. Fertility treatment patients were not to be an exception; it is widely known that fertility and reproductive treatments constitute a stressful event for most patients who have difficulties achieving pregnancy [14,15]. During the initial phases of the pandemic, many fertility treatments were postponed, with a subsequent psychological impact on women and their partners and an increase in their stress and anxiety [16,17,18].
Despite the directions provided by international organizations, physician and patient hesitancy towards vaccination during pregnancy and preconception has remained an issue. Some studies report that <50% were willing to get vaccinated during these periods [19,20]. Published surveys and inquiries suggest that around 30% of infertile couples undergoing assisted reproductive treatment delayed the primary vaccination [21].
Nevertheless, according to the Centers for Disease Control and Prevention, in 2020, there were 326,468 ART cycles performed at 449 reporting clinics in the United States, resulting in 75,023 live births (deliveries of one or more living infants) and 79,942 live-born infants [22]. Comparing these ciphers to those reported in 2019, it is evident that COVID did not impede infertile patients from initiating ART. 330,773 new cycles were reported in 2019, of which 95,030 resulted in pregnancies, 77,998 live-birth deliveries (delivery of one or more living infants), and 83,946 infants [23]. It is evident that infertility treatments did not decrease despite the COVID-19 sanitary crisis; however, it probably stopped thousands of individuals from accepting and receiving vaccination.
Some of the main fears arose from uncertainty: Could COVID-19, prior to or during assisted reproductive techniques (ART), negatively influence the results of the treatment? Does COVID-19 vaccination during ART have deleterious effects on the treatment? What if previous immunity to SARS-CoV-2—through infection or vaccination—influences ART success?
Published investigations, as well as previous systematic reviews and meta-analyses, have aimed to answer these questions and clarify the association between COVID-19 and fertility. Many investigators defend that sperm production can be altered, as male gonads can be vulnerable to infection [24,25,26]. This can be explained by the presence of angiotensin-converting enzyme 2 (ACE2) receptors, which are abundant in testes, seminiferous duct cells, spermatogonia, Leydig, and Sertoly cells [27,28]. It is known that SARS-CoV-2 enters the host cell by ligating precisely to these ACE2 receptors. Furthermore, fever is one of the most frequent symptoms among those suffering from COVID-19. This state can cause dysregulation of sperm production and development, as a stable scrotal temperature is essential for this process [29].
In addition, numerous published studies to date have questioned if COVID-19 could also affect female fertility and disrupt its functions. ACE2 receptors are also present in endometrium cells and are involved in follicular and ovulation regulation and development, angiogenesis, and luteal degeneration [27]. Furthermore, the cytokine storm and consequent appearance of reactive oxygen species secondary to inflammation and the immune response activated by COVID-19 infection could cause ovarian damage and disrupt oocyte development and normal embryo implantation [30]. Nevertheless, most studies conclude that no evidence suggests COVID-19 infection could significantly alter female fertility [31].
Nevertheless, there is scarce evidence concerning COVID-19 vaccination and fertility, and valuable data is even harder to find referring to patients undergoing ART. This review aims to summarize the existing evidence concerning this group of patients to resolve some of the unanswered questions and guide healthcare professionals and patients in their decision-making.

2. Methods

2.1. Information Sources

This systematic review was developed following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guide. Publications from the following sources were included: Pubmed-Medline, Cochrane Database, Web of Science, and National Guideline.

2.2. Eligibility Criteria

All observational and experimental studies concerning the COVID-19 vaccine and ART were considered adequate for this review. Subsequently, exclusive criteria were applied: (1) secondary studies (reviews, metaanalyses), (2) studies not measuring ART outcome, (3) studies not evaluating COVID-19 vaccination, and (4) studies without full text available.
As the present article is a systematic review, only primary investigations have been analyzed. Other secondary sources, such as systematic reviews and meta-analyses, are included and debated in the discussion section.

2.3. Search Strategy

In order to define our search, a PICO question was defined: P (patient = patient undergoing ART), I (intervention = COVID-19 vaccines), C (comparison = not vaccinated), and O (outcome = fertility treatment response). Then, MeSH terms were defined: COVID-19, vaccine, ART, in vitro fertilization (IVF), embryo, SARS-CoV-2, and coronavirus.

2.4. Data Collection Process

The literature search was executed carefully according to PRISMA guidelines [32]. Formerly, titles and abstracts of all articles were screened to exclude articles that were not focused on the theme, and subsequently, an intense lecture on each article was performed. Thus, potential exclusion criteria were detected. Finally, a discussion was conducted between collaborators, and a final selection was made by consensus. The selection process is shown in the following flow diagram, Figure 1.

2.5. Bias Assessment

The described eligibility criteria were strictly defined and applied to reduce the risk of bias in our systematic review. The research was based on multiple information sources, including English, French, and Spanish articles. This scrutiny was performed in an objective and reproducible manner. Furthermore, and as previously detailed, two authors analyzed and screened the selected articles to contrast and refine the final selection.

3. Results

3.1. Search Results

A total of 59 articles were initially collected from four different databases. Twenty-three records were excluded after reading the title and abstract, as the main subject was unrelated to the investigated matter. Thus, 36 reports were assessed for eligibility and submitted to an intense evaluation. Finally, 24 documents were selected to constitute this review after excluding 12 articles for not meeting the inclusion criteria. Figure 1 details the justifications for article exclusion.

3.2. Characteristics of Included Studies

Altogether, 3469 ART cycles were gathered and analyzed. Control groups consisting of unvaccinated patients summed up to 15,124 individuals, while cases were divided between vaccinated patients (n = 6656), those with a passed COVID-19 infection (n = 71), and those accomplishing both situations (n = 34). The mean total sample size was 143,204 patients per article; however, it should be highlighted that the sample size was rather discordant (from 32 up to 10,541). Most of the included studies had retrospective cohort designs (66.6%), while the rest displayed prospective cohort designs. Furthermore, most were single-centered studies (75%) based on six different countries: China (11), Israel (6), Spain (3), the United States of America (2), Jordan (1), and Austria (1).
All the selected articles except three, detailed the type of vaccine administered to the included patients. The most prevalent vaccine among the included sample was the inactivated virus vaccine (50%), followed by messenger RNA (mRNA) vaccines (41.67%), adenovirus vector vaccines (20.8%), and finally recombinant subunit vaccines (12.5%). Only five studies compared results between different vaccines, as discussed further [33,34,35,36,37].
Regarding ART techniques, most studies analyzed IVF cycles (83.3%). A minority included data concerning embryo transfer (ET) exclusively (16.6%) and artificial insemination (AI) procedures (4.2%).
Detailed study characteristics are described in Table 1. Table 2 shows the variables that were assessed in the included studies.

3.3. Ovarian Stimulation and Oocyte Retrieval

Most studies analyzed the number of oocytes retrieved (n = 17) and their quality (n = 14). The vast majority of studies agree that no evidence was found to suggest that COVID-19 vaccination negatively affects cycle stimulation characteristics, as no differences between case and control groups were detected in any of the surrogate parameters for ovarian follicle quality [34,36,38,40,41,46,47,48,49,50,51,52,53,54,56].
Nevertheless, one Spanish study published in 2023 revealed a higher number of oocytes retrieved in vaccinated patients, though their quality was significantly lower [42]. Similarly, a Chinese study including 536 patients undergoing IVF/intracytoplasmatic sperm injection (ICSI) found lower-quality oocytes in vaccinated women [33].

3.4. Fertilization, Embryo Development, and Transfer

A total of 12 studies evaluated the number of fertilized oocytes. The most common finding was that COVID-19 vaccination did not affect this parameter [34,36,40,46,49,50,51,52,53,56], although two other of the included studies differed [33,47].
A Chinese study including women who received inactivated or recombinant COVID-19 vaccination obtained lower fertilization rates in women who had received the recombinant vaccine, especially in those who received ART treatment less than six weeks after vaccination [47]. This, however, did not substantially affect the primary study endpoints: average fertilization rate and clinical pregnancy rate. On the contrary, an American study displays that in their studied sample, vaccinated patients had higher mean and standard deviation (SD) fertilization rates than unvaccinated patients (77.45% (41.45%) vs. 68.66% (20.51%); p = 0.03) [33].
Regarding embryo quality and euploidy, only two studies have found significant differences between groups. Dong et al. [34] affirm no significant differences in laboratory results (high-quality embryo rate and blastocyst formation rate) among groups (p > 0.05). Nonetheless, the embryo transfer stage (cleavage or blastocyte) and type (fresh or frozen) did have significant differences among the four groups: a higher rate of cleavage vs. blastocyst was found among groups with both partners receiving two doses of COVID-19 vaccine or neither of them vaccinated compared to groups receiving only one of the members of the couple vaccinated. Contrarily, Jacobs et al. describe that the number of embryos at the cleavage stage was significantly lower in the vaccinated group; however, with no significant differences in pregnancy rates [47]. The rest of the included studies revealed that vaccination status did not affect embryo quality [33,36,38,39,42,44,45,46,47,50,52,53,56] or euploidy [38,46].
Nine studies have evaluated the number of embryos transferred [33,34,39,40,44,45,46,50,52,53]. Two studies detail those available for posterior cryopreservation [40,47], and another study points out the state of each embryo when transferred [50]. All studies conclude that there is no evidence to suggest that COVID-19 vaccination negatively affects these embryological variables.

3.5. Implantation, Pregnancy, and Loss

It is convenient to clarify the definition of “implantation rate,” meaning the total number of early gestational sacs/the total number of transferred embryos × 100%. Besides, the clinical pregnancy rate is the number of pregnancy cycles/the total number of transfer cycles × 100%.
All ten studies that assessed implantation rates agree that no statistically significant differences were found when comparing couples with completed vaccinations vs. those unvaccinated [33,34,35,40,42,43,45,46,52,53,55]. Equally, clinical pregnancy rates and data concerning ongoing pregnancy, biochemical loss, and clinical loss support that COVID-19 vaccination is safe and effective and has no impact on fertility [33,34,38,39,40,41,42,43,44,45,46,47,49,50,51,52,53,55,56].

3.6. Comparison between Vaccines

Five articles compare the results of different types of vaccines, Table 3. Among them, only one found statistical differences when comparing inactivated virus vaccines and recombinant subunit vaccines regarding oocyte maturation and fertilization rates [33].

4. Discussion

The global COVID-19 pandemic is currently under control, given that the WHO decreed the end of the international emergency due to COVID on 5 May 2023 [57]. Massive vaccination has indeed helped to dominate the crisis, as COVID-19 vaccination has proved not only to protect against severe symptoms of the disease but also to be an essential tool in decreasing the spread of the virus and the rate of infection [58,59,60].
Many studies have already demonstrated high security in vaccination during pregnancy [61,62] and vaccinated women with SARS-CoV-2 infection are associated with decreased hospital admission due to COVID-19 as well as reduced progression to severe COVID-19 [63].
This systematic review concerning COVID-19 vaccination and ART shows excellent vaccine safety evidence for couples requiring ART. The vast majority of studies are consistent about unaltered results in oocyte maturation and retrieval, fertilization, embryo development and transfer, implantation, pregnancy, and loss.
The gathered information suggests that all evaluated vaccines (inactivated, adenovirus vector, recombinant, and mRNA) constitute a safe option, with no significant differences in oocyte stimulation response, embryo quality, or pregnancy outcomes. This is applicable among infertile couples in which both, only one, or neither of the partners were vaccinated against COVID-19.
Three systematic reviews and meta-analyses concerning COVID-19 and ART results have been previously published. Chen et al. [33] thoroughly reviewed the literature and compared 15 studies on the safety of COVID-19 vaccines in 536 women undergoing fresh embryo transfer after IVF/ICSI treatment. Coinciding with the findings of the present systematic review, they concluded that implantation rates were not significantly lower in vaccinated patients.
Likewise, a systematic review and meta-analysis published in April 2023 analyzed 18,877 individual cases undergoing IVF [64], vaccinated with either an mRNA or inactivated virus vaccine. Their results conclude that vaccination against COVID-19 did not adversely affect the different stages of the process (number of oocytes and MII/mature oocytes obtained; implantation, blastocysts, and fertilization rates) or the final outcome (biochemical pregnancy rates) of IVF.
A third article evaluates ART cycle results before and after the COVID pandemic by analyzing seven previous studies of 33,883 ART cycles [65]. The findings of this review describe that no statistical differences were found before and after the COVID-19 pandemic for all of the studied outcomes (both clinical and laboratory). Nevertheless, it is essential to highlight that this review does not assess vaccination status but evaluates whether the COVID-19 infection itself and the subsequent sociosanitary changes secondary to the pandemic state (such as quarantine measures and changes in the frequency of medical visits) could affect the laboratory and clinical outcomes of women undergoing ART.
The present systematic review detected several limitations in the gathered evidence. Firstly, only observational studies have been found, as no placebo-controlled studies in vaccinated patients have been developed. Future investigations should design, in this manner, higher-quality studies in the furtherance of strengthening the present evidence. Furthermore, paternal factors are barely evaluated, despite being an essential element in fertility and ART. This factor should be solidly considered and treated as confounding if only female vaccination or immunity were assessed. In addition, only five studies compared different types of vaccines. However, none included all varieties, and limited evidence is published concerning ART techniques rather than IVF (IA, timed intercourse, ovulation induction techniques). Similarly, long-term pregnancy outcomes are extremely scant. Many studies have proven the safety of COVID-19 vaccines before conception and during pregnancy [66]. However, ART pregnancies are not studied independently. All these aspects need to be approached and deserve attention.
To the best of our knowledge, this is the first systematic review concerning COVID-19 vaccination and ART, including the four types of vaccines developed. An ulterior meta-analysis would strengthen the assembled evidence and give a clearer view of the subject.
In conclusion, this systematic review reveals that there is no scientific evidence of any association between COVID-19 vaccination and adverse outcomes in ART. Thus, patients and healthcare professionals should be reassured about completing vaccination schedules before starting ART. Nonetheless, more data is warranted to confirm that long-term pregnancy outcomes are not altered.

Author Contributions

E.S.-P., A.M.-V., and J.M.-R. performed the work’s conceptualization. E.S.-P. and A.M.-V. carried out the methodology and design. A.M.-V. supervised the research work. E.S.-P. and A.M.-V. wrote the original draft. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Patient consent was waived due to the retrospective nature of this review study.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Holmes, K.V. SARS coronavirus: A new challenge for prevention and therapy. J. Clin. Investig. 2003, 111, 1605–1609. [Google Scholar] [CrossRef] [PubMed]
  2. Wan, Y.; Shang, J.; Graham, R.; Baric, R.S.; Li, F. Receptor recognition by the novel coronavirus from Wuhan: An analysis based on decade-long structural studies of SARS coronavirus. J. Virol. 2020, 94, e00127–e220. [Google Scholar] [PubMed] [Green Version]
  3. Jean, A.; Quach, C.; Yung, A.; Semret, M. Severity and outcome associated with human coronavirus OC43 infections among children. Pediatr. Infect. Dis. J. 2013, 32, 325–329. [Google Scholar] [CrossRef] [PubMed]
  4. SeyedAlinaghi, S.; Karimi, A.; Barzegary, A.; Mojdeganlou, H.; Vahedi, F.; Mirghaderi, S.P.; Shobeiri, P.; Ramezani, M.; Yousefi Konjdar, P.; Mirzapour, P.; et al. COVID-19 mortality in patients with immunodeficiency and its predictors: A systematic review. Eur. J. Med. Res. 2022, 27, 195. [Google Scholar] [CrossRef] [PubMed]
  5. Bassetti, M.; Vena, A.; Roberto Giacobbe, D. The Novel Chinese Coronavirus (2019-nCoV) Infections: Challenges for fighting the storm. Eur. J. Clin. Investig. 2020, 50, e13209. [Google Scholar] [CrossRef] [Green Version]
  6. WHO. WHO Health Coronavirus (COVID-19) Dashboard. July 2023. Available online: https://covid19.who.int (accessed on 3 July 2023).
  7. FDA. Updated COVID-19 Vaccines for Use in the United States Beginning in Fall. 2023. Available online: https://www.fda.gov/vaccines-blood-biologics/updated-covid-19-vaccines-use-united-states-beginning-fall-2023 (accessed on 16 June 2023).
  8. Keshavarz, M.; Mirzaei, H.; Salemi, M.; Momeni, F.; Mousavi, M.J.; Sadeghalvad, M.; Arjeini, Y.; Solaymani-Mohammadi, F.; Sadri Nahand, J.; Namdari, H.; et al. Influenza vaccine: Where are we and where do we go? Rev. Med. Virol. 2019, 29, e2014. [Google Scholar] [CrossRef]
  9. Bandyopadhyay, A.S.; Garon, J.; Seib, K.; Orenstein, W.A. Polio vaccination: Past, present and future. Future Microbiol. 2015, 10, 791–808. [Google Scholar] [CrossRef] [Green Version]
  10. Vetter, V.; Denizer, G.; Friedland, L.R.; Krishnan, J.; Shapiro, M. Understanding modern-day vaccines: What you need to know. Ann. Med. 2018, 50, 110–120. [Google Scholar] [CrossRef]
  11. Jain, S.; Venkataraman, A.; Wechsler, M.E.; Peppas, N.A. Messenger RNA-based vaccines: Past, present, and future directions in the context of the COVID-19 pandemic. Adv. Drug Deliv. Rev. 2021, 179, 114000. [Google Scholar]
  12. Meo, S.A.; Bukhari, I.A.; Akram, J.; Meo, A.S.; Klonoff, D.C. COVID-19 vaccines: Comparison of biological, pharmacological characteristics and adverse effects of Pfizer/BioNTech and Moderna Vaccines. Eur. Rev. Med. Pharmacol. Sci. 2021, 25, 1663–1669. [Google Scholar]
  13. Fan, Y.J.; Chan, K.H.; Hung, I.F. Safety and Efficacy of COVID-19 Vaccines: A Systematic Review and Meta-Analysis of Different Vaccines at Phase 3. Vaccines 2021, 9, 989. [Google Scholar] [CrossRef]
  14. Seibel, M.M.; Taymor, M.L. Emotional aspects of infertility. Fertil. Steril. 1982, 37, 137–145. [Google Scholar] [CrossRef] [PubMed]
  15. Stanhiser, J.; Steiner, A.Z. Psychosocial Aspects of Fertility and Assisted Reproductive Technology. Obstet. Gynecol. Clin. N. Am. 2018, 45, 563–574. [Google Scholar] [CrossRef]
  16. Lablanche, O.; Salle, B.; Perie, M.A.; Labrune, E.; Langlois-Jacques, C.; Fraison, E. Psychological effect of COVID-19 pandemic among women undergoing infertility care, a French cohort—PsyCovART Psychological effect of COVID-19: PsyCovART. J. Gynecol. Obstet. Hum. Reprod. 2022, 51, 102251. [Google Scholar] [CrossRef] [PubMed]
  17. Pappa, K.; Xydias, E.M.; Ziogas, A.C.; Bimba, K.; Tsakos, E.; Kolibianakis, E. The Impact of Coronavirus Disease 2019 Pandemic on Female Patients Seeking or Undergoing Fertility Treatment in a Single in vitro Fertilisation Clinic in Greece: An International, Cross-sectional Study. J. Hum. Reprod. Sci. 2023, 16, 50. [Google Scholar] [CrossRef] [PubMed]
  18. Arbağ, E.; Aluş Tokat, M.; Özöztürk, S. Emotions, thoughts, and coping strategies of women with infertility problems on changes in treatment during Covid-19 pandemic: A qualitative study. Women’s Stud. Int. Forum 2023, 98, 102735. [Google Scholar] [CrossRef]
  19. Battarbee, A.N.; Stockwell, M.S.; Varner, M.; Newes-Adeyi, G.; Daugherty, M.; Gyamfi-Bannerman, C.; Tita, A.T.; Vorwaller, K.; Vargas, C.; Subramaniam, A.; et al. Attitudes Toward COVID-19 Illness and COVID-19 Vaccination among Pregnant Women: A Cross-Sectional Multicenter Study during August-December 2020. Am. J. Perinatol. 2022, 39, 75–83. [Google Scholar] [CrossRef]
  20. Goncu Ayhan, S.; Oluklu, D.; Atalay, A.; Menekse Beser, D.; Tanacan, A.; Moraloglu Tekin, O.; Sahin, D. COVID-19 vaccine acceptance in pregnant women. Int. J. Gynaecol. Obstet. 2021, 36, 197–211. [Google Scholar] [CrossRef]
  21. Wang, X.; Wang, H.; Du, A.; Wang, J.; Shi, J.; Zhang, Y.; Zhang, Y.; Ma, J.; Meng, W.; Lv, J.; et al. COVID-19 vaccine hesitancy and associated factors among infertile couples undergoing assisted reproductive treatment. Front. Immunol. 2022, 13, 973600. [Google Scholar] [CrossRef]
  22. US Department of Health and Human Services. Assisted Reproductive Technology. National Summary Report 2020; Centers for Disease Control and Prevention: Atlanta, GA, USA, 2023. [Google Scholar]
  23. US Department of Health and Human Services. Assisted Reprotuction Technology. National Summary Report 2019; Centers for Disease Control and Prevention: Atlanta, GA, USA, 2021. [Google Scholar]
  24. Săndulescu, M.S.; Văduva, C.C.; Siminel, M.A.; Dijmărescu, A.L.; Vrabie, S.C.; Camen, I.V.; Tache, D.E.; Neamţu, S.D.; Nagy, R.D.; Carp-Velişcu, A.; et al. Impact of COVID-19 on fertility and assisted reproductive technology (ART): A systematic review. Rom. J. Morphol. Embryol. 2022, 63, 503–510. [Google Scholar] [CrossRef]
  25. Khalili, M.A.; Leisegang, K.; Majzoub, A.; Finelli, R.; Panner Selvam, M.K.; Henkel, R.; Mojgan, M.; Agarwal, A. Male Fertility and the COVID-19 Pandemic: Systematic Review of the Literature. World J. Mens. Health 2020, 38, 506–520. [Google Scholar] [CrossRef] [PubMed]
  26. Wang, S.; Zhang, A.; Pan, Y.; Liu, L.; Niu, S.; Zhang, F.; Liu, X. Association between COVID-19 and Male Fertility: Systematic Review and Meta-Analysis of Observational Studies. World J. Mens. Health 2023, 41, 311–329. [Google Scholar] [CrossRef]
  27. Li, F.; Lu, H.; Zhang, Q.; Li, X.; Wang, T.; Liu, Q.; Yang, Q.; Qiang, L. Impact of COVID-19 on female fertility: A systematic review and meta-analysis protocol. BMJ Open 2021, 11, e045524. [Google Scholar] [CrossRef] [PubMed]
  28. Huang, H.H.; Wang, P.H.; Yang, Y.P.; Chou, S.J.; Chu, P.W.; Wu, G.J.; Chang, C.C. A review of severe acute respiratory syndrome coronavirus 2 infection in the reproductive system. J. Chin. Med. Assoc. 2020, 83, 895–897. [Google Scholar] [CrossRef] [PubMed]
  29. Meng, T.T.; Dong, R.J.; Li, T.G. Relationship between COVID-19 and the male reproductive system. Eur. Rev. Med. Pharmacol. Sci. 2021, 25, 1109–1113. [Google Scholar]
  30. Zou, X.; Chen, K.; Zou, J.; Han, P.; Hao, J.; Han, Z. Single-cell RNA-seq data analysis on the receptor ACE2 expression reveals the potential risk of different human organs vulnerable to 2019-nCoV infection. Front. Med. 2020, 14, 185–192. [Google Scholar] [CrossRef] [Green Version]
  31. Zaçe, D.; La Gatta, E.; Petrella, L.; Di Pietro, M.L. The impact of COVID-19 vaccines on fertility-A systematic review and meta-analysis. Vaccine 2020, 40, 6023–6034. [Google Scholar] [CrossRef]
  32. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. BMJ 2021, 372, n71. [Google Scholar] [CrossRef]
  33. Chen, H.; Zhang, X.; Lin, G.; Gong, F.; Hocher, B. Safety of COVID-19 vaccination in women undergoing IVF/ICSI treatment—Clinical study and systematic review. Front. Immunol. 2023, 13, 1054273. [Google Scholar] [CrossRef]
  34. Dong, M.; Wu, S.; Zhang, X.; Zhao, N.; Qi, J.; Zhao, D.; Sang, Y.; Tan, J. Effects of COVID-19 vaccination status, vaccine type, and vaccination interval on IVF pregnancy outcomes in infertile couples. J. Assist. Reprod. Genet. 2022, 39, 1849–1859. [Google Scholar] [CrossRef]
  35. Wang, C.; Tang, D.; Liu, J.; Zhang, S.; Xu, Y.; Qiao, J.; Cao, Y. Association Between COVID-19 Vaccination and Artificial Insemination Outcomes for Couples Experiencing Infertility. JAMA Netw. Open 2022, 5, e2247216. [Google Scholar] [CrossRef] [PubMed]
  36. Requena, A.; Vergara, V.; González-Ravina, C.; Ruiz, M.E.; Cruz, M. The type of SARS-CoV-2 vaccine does not affect ovarian function in assisted reproduction cycle. Fertil. Steril. 2023, 119, 618–623. [Google Scholar] [CrossRef] [PubMed]
  37. Zhao, Y.; Zhao, Y.; Ai, A.; Jin, L. Association of inactivated COVID-19 vaccination with in vitro fertilization and early pregnancy outcomes. J. Med. Virol. 2023, 95, e28432. [Google Scholar] [CrossRef]
  38. Aharon, D.; Lederman, M.; Ghofranian, A.; Hernandez-Nieto, C.; Canon, C.; Hanley, W.; Gounko, D.; Lee, J.A.; Stein, D.; Buyuk, E.; et al. In Vitro Fertilization and Early Pregnancy Outcomes After Coronavirus Disease 2019 (COVID-19) Vaccination. Obstet. Gynecol. 2022, 139, 490–497. [Google Scholar] [CrossRef]
  39. Aizer, A.; Noach-Hirsh, M.; Dratviman-Storobinsky, O.; Nahum, R.; Machtinger, R.; Yung, Y.; Haas, J.; Orvieto, R. The effect of coronavirus disease 2019 immunity on frozen-thawed embryo transfer cycles outcome. Fertil. Steril. 2022, 117, 974–979. [Google Scholar] [CrossRef]
  40. Albeitawi, S.; Al-Alami, Z.M.; Hamadneh, J.; Alqam, H.; Qublan, H.; Al Natsheh, M. COVID-19 infection and vaccine have no impact on in-vitro fertilization (IVF) outcome. Sci. Rep. 2022, 2, 21702. [Google Scholar] [CrossRef]
  41. Avraham, S.; Kedem, A.; Zur, H.; Youngster, M.; Yaakov, O.; Yerushalmi, G.M.; Gat, I.; Gidoni, Y.; Hochberg, A.; Baum, M.; et al. Coronavirus disease 2019 vaccination and infertility treatment outcomes. Fertil. Steril. 2022, 117, 1291–1299. [Google Scholar] [CrossRef]
  42. Bosch, A.; Albero, S.; Castillo, J.C.; Ten, J.; Guerrero, J.; Ortiz, J.A.; Bernabeu, A.; Bernabeu, R. Does mRNA COVID-19 vaccination in oocyte donors impact ovarian stimulation parameters or IVF outcomes for recipients? Reprod. Biomed. Online 2023, 46, 697–704. [Google Scholar] [CrossRef]
  43. Brandão, P.; Pellicer, A.; Meseguer, M.; Remohí, J.; Garrido, N.; García-Velasco, J.A. COVID-19 mRNA vaccines have no effect on endometrial receptivity after euploid embryo transfer. Reprod. Biomed. Online 2022, 45, 688–695. [Google Scholar] [CrossRef]
  44. Cao, M.; Wu, Y.; Lin, Y.; Xu, Z.; Liang, Z.; Huang, Q.; Li, S.; Liu, H.; An, C.; Luo, Y.; et al. Inactivated Covid-19 vaccine did not undermine live birth and neonatal outcomes of women with frozen-thawed embryo transfer. Hum. Reprod. 2022, 37, 2942–2951. [Google Scholar] [CrossRef]
  45. Huang, J.; Liu, Y.; Zeng, H.; Tian, L.; Hu, Y.; He, J.; Nie, L.; Li, Y.; Fang, Z.; Deng, W.; et al. Effect of inactivated COVID-19 vaccination on pregnancy outcomes following frozen-thawed embryo transfer: A retrospective cohort study. Int. Immunopharmacol. 2023, 114, 109552. [Google Scholar] [CrossRef]
  46. Huang, J.; Xia, L.; Tian, L.; Fan, H.; Xu, D.; Ai, X.; Wu, X.; Chen, J.; Xing, G.; Huang, L.; et al. Impact of inactivated SARS-CoV-2 vaccination on embryo ploidy: A retrospective cohort study of 133 PGT-A cycles in China. Biol. Res. 2022, 55, 26. [Google Scholar] [CrossRef] [PubMed]
  47. Jacobs, E.; Summers, K.; Sparks, A.; Mejia, R. Fresh Embryo Transfer Cycle Characteristics and Outcomes Following In Vitro Fertilization via Intracytoplasmic Sperm Injection Among Patients With and Without COVID-19 Vaccination. JAMA Netw. Open 2022, 5, e228625. [Google Scholar] [CrossRef] [PubMed]
  48. Karavani, G.; Chill, H.H.; Dick, A.; Meirman, C.; Gutman-Ido, E.; Herzberg, S.; Ben-Meir, A.; Imbar, T. Pfizer SARS-CoV-2 BNT162b2 mRNA vaccination (BNT162b2) has no adverse effect on elective oocyte cryopreservation outcomes. Reprod. Biomed. Online 2022, 45, 987–994. [Google Scholar] [CrossRef] [PubMed]
  49. Odeh-Natour, R.; Shapira, M.; Estrada, D.; Freimann, S.; Tal, Y.; Atzmon, Y.; Bilgory, A.; Aslih, N.; Abu-Raya, Y.S.; Shalom-Paz, E. Does mRNA SARS-CoV-2 vaccine in the follicular fluid impact follicle and oocyte performance in IVF treatments? Am. J. Reprod. Immunol. 2022, 87, e13530. [Google Scholar] [CrossRef]
  50. Orvieto, R.; Noach-Hirsh, M.; Segev-Zahav, A.; Haas, J.; Nahum, R. Does mRNA SARS-CoV-2 vaccine influence patients’ performance during IVF-ET cycle? Reprod. Biol. Endocrinol. 2021, 19, 69. [Google Scholar] [CrossRef]
  51. Chillon, T.S.; Weiss, G.; Demircan, K.; Minich, W.B.; Schenk, M.; Schomburg, L. Antibodies to SARS-CoV-2 in follicular fluids and their association with assisted reproduction. Front. Immunol. 2023, 14, 1120328. [Google Scholar] [CrossRef]
  52. Xia, W.; Zhao, J.; Hu, Y.; Fang, L.; Wu, S. Investigate the effect of COVID-19 inactivated vaccine on sperm parameters and embryo quality in in vitro fertilization. Andrologia 2022, 54, e14483. [Google Scholar] [CrossRef]
  53. Wu, Y.; Cao, M.; Lin, Y.; Xu, Z.; Liang, Z.; Huang, Q.; Li, S.; Li, L.; Meng, Y.; An, C.; et al. Inactivated COVID-19 vaccination does not affect in vitro fertilization outcomes in women. Hum. Reprod. 2022, 37, 2054–2062. [Google Scholar] [CrossRef]
  54. Bentov, Y.; Beharier, O.; Moav-Zafrir, A.; Kabessa, M.; Godin, M.; Greenfield, C.S.; Ketzinel-Gilad, M.; Ash Broder, E.; Holzer, H.E.G.; Wolf, D.; et al. Ovarian follicular function is not altered by SARS-CoV-2 infection or BNT162b2 mRNA COVID-19 vaccination. Hum. Reprod. 2021, 36, 2506–2513. [Google Scholar] [CrossRef]
  55. Wang, Y.; Ren, X.; Wang, Z.; Feng, X.; Li, M.; Liu, P. Receipt of inactivated COVID-19 vaccine had no adverse influence on embryo implantation, clinical pregnancy and miscarriage in early pregnancy. Sci. China Life Sci. 2022, 65, 2332–2334. [Google Scholar] [CrossRef] [PubMed]
  56. Yin, J.; Wang, Y.; Tao, L.; Chen, L.; Liu, P.; Li, R. Impact of vaccination against COVID-19 on the outcomes of in vitro fertilization-embryo transfer: A retrospective cohort study. Chin. Med. J. 2023, 136, 207–212. [Google Scholar] [CrossRef] [PubMed]
  57. WHO. Statement on the Fifteenth Meeting of the IHR (2005) Emergency Committee on the COVID-19 Pandemic. 2023. Available online: https://www.who.int/news/item/05-05-2023-statement-on-the-fifteenth-meeting-of-the-international-health-regulations-(2005)-emergency-committee-regarding-the-coronavirus-disease-(covid-19)-pandemic (accessed on 3 July 2023).
  58. Vitiello, A.; Ferrara, F.; Troiano, V.; La Porta, R. COVID-19 vaccines and decreased transmission of SARS-CoV-2. Inflammopharmacology 2021, 29, 1357–1360. [Google Scholar] [CrossRef]
  59. Sharif, N.; Alzahrani, K.J.; Ahmed, S.N.; Dey, S.K. Efficacy, Immunogenicity and Safety of COVID-19 Vaccines: A Systematic Review and Meta-Analysis. Front. Immunol. 2021, 12, 714170. [Google Scholar] [CrossRef] [PubMed]
  60. Novillo, B.; Martínez-Varea, A. COVID-19 Vaccines during Pregnancy and Breastfeeding: A Systematic Review. J. Pers. Med. 2022, 13, 40. [Google Scholar] [CrossRef] [PubMed]
  61. Carbone, L.; Trinchillo, M.G.; Di Girolamo, R.; Raffone, A.; Saccone, G.; Iorio, G.G.; Gabrielli, O.; Maruotti, G.M. COVID-19 vaccine and pregnancy outcomes: A systematic review and meta-analysis. Int. J. Gynaecol. Obstet. 2022, 159, 651–661. [Google Scholar] [CrossRef]
  62. Graña, C.; Ghosn, L.; Evrenoglou, T.; Jarde, A.; Minozzi, S.; Bergman, H.; Buckley, B.S.; Probyn, K.; Villanueva, G.; Henschke, N.; et al. Efficacy and safety of COVID-19 vaccines. Cochrane Database Syst. Rev. 2022, 12, CD015477. [Google Scholar] [CrossRef]
  63. Martínez-Varea, A.; Satorres, E.; Florez, S.; Domenech, J.; Desco-Blay, J.; Monfort-Pitarch, S.; Hueso, M.; Perales-Marín, A.; Diago-Almela, V. Comparison of Maternal-Fetal Outcomes among Unvaccinated and Vaccinated Pregnant Women with COVID-19. J. Pers. Med. 2022, 12, 2008. [Google Scholar] [CrossRef]
  64. Zhang, L.; Sun, X.; Wang, R.; Ma, F. Effect of COVID-19 vaccination on the outcome of in vitro fertilization: A systematic review and meta-analysis. Front. Public Health 2023, 11, 1151999. [Google Scholar] [CrossRef]
  65. Hu, W.; Zhu, Y.; Wu, Y.; Wang, F.; Qu, F.B. Impact of COVID-19 pandemic on the pregnancy outcomes of women undergoing assisted reproductive techniques (ARTs): A systematic review and meta-analysis. J. Zhejiang Univ. Sci. 2022, 23, 655–665. [Google Scholar] [CrossRef]
  66. Juliá-Burchés, C.; Martínez-Varea, A. An Update on COVID-19 Vaccination and Pregnancy. J. Pers. Med. 2023, 13, 797. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Flow diagram representing the article selection process.
Figure 1. Flow diagram representing the article selection process.
Jpm 13 01232 g001
Table 1. Characteristics of the included studies.
Table 1. Characteristics of the included studies.
Article DetailsSample (Cycles)Type of VaccineART
Technique
ArticleCountry and Date of PublicationCentreDesignCasesPassed COVID-19VaccinatedPassed and VaccinatedControlInactivated VirusmRNAAdenovirusRecombinatedIVFETAI
Aharon et al. [38]EEUU, 2022Single centreRetrospective cohort study120502220983 x x
Aizer A et al. [39]Israel, 2022Single centreRetrospective cohort study672442200408 x x
Albeitawi S et al. [40]Jordan, 2022Multicentric studyRetrospective cohort study15118663433xxx x
Avrahan S et al. [41]Israel, 2022Multicentric studyRetrospective cohort study40002000200 x x
Bosch A et al. [42]Spain, 2023Multicentric studyRetrospective cohort study23001150115 x x
Brandao P et al. [43]Spain, 2022Multicentric studyRetrospective cohort study4162089003272 x x
Cao M et al. [44]China 2022Single centreRetrospective cohort study210105’2O1589x x
Chen H et al. [33]china 2023Single centreRetrospective cohort study5360268 0268x xx
Dong M et al. [34]China, 2022 Single centreProspective cohort study73502210514x xxxx
Huang J et al. [45]China 2022Single centreRetrospective cohort study121002650265x x
Huang J et al. [46]China 2022Single centreRetrospective cohort study133066067x x
Jacobs E et al. [47]EEUU, 2022Single centreProspective cohort study28001420138 x
Karavani G et al. [48]israel, 2022Single centreRetrospective cohort study138083055 x x
Odeh-Natour R et al. [49]Israel, 2022Single centreProspective cohort study59037022 x
Orvieto R et al. [50]Israel, 2021Single centreProspective cohort study36036036 x x
Requena A et al. [36]Spain, 2023Multicentric studyRetrospective cohort study1700051001190 xx x
Chillon T et al. [51]Austria, 2023Single centreProspective cohort study89045044 x
Wang C et al. [35]China 2022Multicentric studyProspective cohort study41850212902056 x xx x
Xia W et al. [52]China 2022Single centreProspective cohort study26001050155x x
Wu Y et al. [53]China 2022Single centreRetrospective cohort study2400001343 x x
Bentov Y et al. [54]Israel, 2021Single centreProspective cohort study3299014 x x
Wang Y et al. [55]China 2022Single centreRetrospective cohort study1496046001036x x
Yin J et al. [56]China 2023Single centreRetrospective cohort study10,541083501670x x x
Zhao Yan et al. [37]China 2022Single centreRetrospective cohort study377871728343050x
Assisted reproductive techniques (ART), embryo transfer (ET), and artificial insemination (AI).
Table 2. Variables studied in the included studies. * SD: statistical differences.
Table 2. Variables studied in the included studies. * SD: statistical differences.
Article Country and Date of PublicationEndometrial ThicknessNumber of Oocytes RetrievedQuality of Oocytes Number of Fertilized OocytesNumber and Quality of Embryos Number of Embryos TransferredNumber of Frozen EmbryosEuploidyImplantation Rate Clinical
Pregnancy Rate
Ongoing PregnancyBiochemical LossClinical LossOther
Aharon et al. [38]EEUU, 2022 NSNS NS NS NSNSNSNS
Aizer A et al. [39]Israel, 2022NS NSNS NSNSNSNS
Albeitawi S et al. [40]Jordan, 2022NSNSNSNSLower in vaccinatedNSNS NSNS
Avrahan S et al. [41]Israel, 2022 NS NS
Bosch A et al. [42]Spain, 2023 Higher in vaccinatedLower in vaccinated NS NSNS
Brandao P et al. [43]Spain, 2022 NSNS
Cao M et al. [44]China, 2022NS NSNS NSNSNSNSEctopic, brith height and weight
Chen H et al. [33]China, 2023NSNSLower in vaccinatedLower in vaccinatedNSNS NSNS NS
Dong M et al. [34]China, 2022NSNSNSNS* SD in 4 groupsNSNS NSNS
Huang J et al. [45]China, 2022NS NSNS NSNS NS Birth
Huang J et al. [46]China, 2022 NSNSNSNS NSNSNS NS
Jacobs E et al. [47]EEUU, 2022 NSNSHigher in vaccinatedNS NS NSNS NS
Karavani G et al. [48]Israel, 2022 NSNS
Odeh-Natour R et al. [49]Israel, 2022NSNSNSNS NS
Orvieto R et al. [50]Israel, 2021 NSNSNSNSNS NS
Requena A et al. [36]Spain, 2023 NSNSNSNS
Chillon T et al. [51]Austria, 2023 NS NS NSNSNSNSBirth
Wang C et al. [35]China 2022 NS
Xia W et al. [52]China, 2022 NSNSNSNSNS NSNSNS Sperm quality, NS
Wu Y et al. [53]China, 2022Lower in vaccinatedNS NSNSNS NSNSNSNSNSEctopic
Bentov Y et al. [54]Israel, 2021 NSNS Igg in folicular fluid, NS
Wang Y et al. [55]China, 2022NS NSNS NSEctopic
Yin J et al. [56]China 2023 NSNSNSNS NSNS
Zhao Yan et al. [37]China 2022 NS NS
Table 3. Comparison between different types of vaccines.
Table 3. Comparison between different types of vaccines.
Article Country and Date of PublicationVaccine
Inactivated Virus Vaccines ADENOVIRUS Vector Recombinant Subunit mRNA
Dong M et al. [34]China, 2022NSNSNS
Chen H et al. [33]China, 2023Lower oocyte maturation and fertilization rate NS
Wang C et al. [55]China, 2022NSNSNS
Requena A et al. [36]Spain, 2023 NS NS
Yin J et al. [56]China, 2023NSNS
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Satorres-Pérez, E.; Martínez-Varea, A.; Morales-Roselló, J. COVID-19 Vaccines and Assisted Reproductive Techniques: A Systematic Review. J. Pers. Med. 2023, 13, 1232. https://doi.org/10.3390/jpm13081232

AMA Style

Satorres-Pérez E, Martínez-Varea A, Morales-Roselló J. COVID-19 Vaccines and Assisted Reproductive Techniques: A Systematic Review. Journal of Personalized Medicine. 2023; 13(8):1232. https://doi.org/10.3390/jpm13081232

Chicago/Turabian Style

Satorres-Pérez, Elena, Alicia Martínez-Varea, and José Morales-Roselló. 2023. "COVID-19 Vaccines and Assisted Reproductive Techniques: A Systematic Review" Journal of Personalized Medicine 13, no. 8: 1232. https://doi.org/10.3390/jpm13081232

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