Next Article in Journal
Identifying Hotspots of People Diagnosed of Tuberculosis with Addiction to Alcohol, Tobacco, and Other Drugs through a Geospatial Intelligence Application in Communities from Southern Brazil
Next Article in Special Issue
Commentary on COVID-19 Vaccine Hesitancy in sub-Saharan Africa
Previous Article in Journal
Acute Viral Gastrointestinal (GI) Infections in the Tropics—A Role for Cartridge-Based Multiplex PCR Panels?
Previous Article in Special Issue
Dengue and COVID-19: Managing Undifferentiated Febrile Illness during a “Twindemic”
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

Characteristics of COVID-19 Breakthrough Infections among Vaccinated Individuals and Associated Risk Factors: A Systematic Review

1
Department of Internal Medicine, Division of Infectious Disease, Johns Hopkins University, Baltimore, MD 21205, USA
2
Division of Research and Academic Affairs, Larkin Community Hospital, South Miami, FL 33143, USA
3
Department of Bioengineering and Therapeutic Sciences, University of California San Francisco (UCSF), San Francisco, CA 94158, USA
4
Department of Bioengineering, University of California Berkeley, Berkeley, CA 94704, USA
5
Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD 21218, USA
6
Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
7
Department of Plant and Microbial Biology, University of California, Berkeley, CA 94720, USA
8
University of California San Francisco (UCSF) Library, San Francisco, CA 94143, USA
9
Department of Internal Medicine, Larkin Community Hospital, South Miami, FL 33143, USA
*
Author to whom correspondence should be addressed.
Trop. Med. Infect. Dis. 2022, 7(5), 81; https://doi.org/10.3390/tropicalmed7050081
Submission received: 17 April 2022 / Revised: 17 May 2022 / Accepted: 20 May 2022 / Published: 22 May 2022
(This article belongs to the Special Issue COVID-19: Current Status and Future Prospects)

Abstract

:
We sought to assess breakthrough SARS-CoV-2 infections in vaccinated individuals by variant distribution and to identify the common risk associations. The PubMed, Web of Science, ProQuest, and Embase databases were searched from 2019 to 30 January 2022. The outcome of interest was breakthrough infections (BTIs) in individuals who had completed a primary COVID-19 vaccination series. Thirty-three papers were included in the review. BTIs were more common among variants of concern (VOC) of which Delta accounted for the largest number of BTIs (96%), followed by Alpha (0.94%). In addition, 90% of patients with BTIs recovered, 11.6% were hospitalized with mechanical ventilation, and 0.6% resulted in mortality. BTIs were more common in healthcare workers (HCWs) and immunodeficient individuals with a small percentage found in fully vaccinated healthy individuals. VOC mutations were the primary cause of BTIs. Continued mitigation approaches (e.g., wearing masks and social distancing) are warranted even in fully vaccinated individuals to prevent transmission. Further studies utilizing genomic surveillance and heterologous vaccine regimens to boost the immune response are needed to better understand and control BTIs.

1. Introduction

The severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) that emerged in December 2019 in Wuhan, China, and was the cause of coronavirus 2019 disease (COVID-19), continues to cause morbidity as part of the ongoing pandemic. As of 4 March 2022, 440,807,756 confirmed cases of COVID-19, including 5,978,096 deaths have been reported [1]. Mortality due to COVID-19 has substantially decreased since the introduction of vaccines and mass vaccination efforts worldwide. As of 27 February 2022, a total of 10,585,766,316 vaccine doses have been administered around the world [1].
However, emerging variants of SARS-CoV-2 and waning immunity in vaccinated individuals continue to hinder efforts to control the disease. Breakthrough infections (BTIs) are defined by the United States (U.S.) Centers for Disease Control and Prevention (CDC) as a positive COVID-19 test by a reverse transcription-polymerase chain reaction (RT-PCR) or rapid antigen test > 14 days following the final dose of the recommended vaccination regimen [2]. In the U.S. as of 22 January 2022, the rate of BTIs was 846.73 per 100,000 in individuals who had completed a primary series of vaccination and 642.19 per 100,000 in those who had completed a primary series and a booster dose [3]. The death rate was 0.96 per 100,000 individuals vaccinated with a primary series and 9.68 in unvaccinated people [3].
The CDC and SARS-CoV-2 Interagency Group (SIG) variant classification define four classes of SARS-CoV-2 variants: variants being monitored (VBM), variants of interest (VOI), variants of concern (VOC), and variants of high consequence (VHC) [4]. VBM includes variants that were associated with an increased rate of transmission but are no longer detected and do not pose a threat to public health in the U.S. These currently include Alpha (B.1.1.7 and Q lineages), Beta (B.1.351, descendent lineages), Gamma (P.1, descendent lineages), Epsilon (B.1.427, B.1.429), Eta (B.1.525), Iota (B.1.526), Kappa (B.1.617.1), 1.617.3, Mu (B.1.621, B.1.621.1), and Zeta (P.2). VOI are associated with increased transmissibility and higher levels of infection. Iota (B.1.526) and B.1.525, identified in the United States, and Zeta (P. 2), first detected in Brazil, belong to this class [4]. Increased transmissibility and disease severity is seen in VOC. These include Alpha (B.1.1.7), first detected in the United Kingdom; Gamma (P.1), first detected in Brazil; Beta (B.1.351) from South Africa; and Epsilon (B.1.427 and B.1.429), detected in the United States [4]. Among all the variants, the Delta (B.1.617.2) variant was reported to have the most transmissibility and severity based on the hospitalization rate until the advent of Omicron (B.1.1.529), which was detected for the first time in South Africa on 24 November 2021 [5,6]. Recent reports suggest that certain VOC might result in a less robust immune response, among other factors, following vaccination against COVID-19, especially in patients with immunosuppression [7,8].
On a mass scale, BTIs pose a serious challenge in tackling the pandemic as these patients may serve as a source of viral spread [9]. With the emergence of VOC such as Omicron and its variants and waning immunity in certain populations after vaccination, a better understanding of BTIs and their attributes, particularly with variant profiles, is essential. These data can help guide public health efforts in determining specific populations that could benefit the most from booster doses of COVID-19 vaccines and help to assess vaccine effectiveness against specific variants.
We explored the current literature on BTIs of SARS-CoV-2 among vaccinated individuals, with a particular focus on the type of vaccine, the SARS-CoV-2 variant involved, common etiology, and immune parameters. This study aimed to identify any associated risk factors and to determine the extent to which BTIs are due to an immune evasion by VOCs as opposed to the failure of vaccines to elicit a satisfactory immune response.

2. Materials and Methods

2.1. Search Strategy and Selection Criteria

This systematic review was performed in accordance with the standards of the Preferred Reporting Items for Systematic Review and Meta-Analysis (PRISMA) Statement [10]. Approval from the Institutional Review Board was not needed. PubMed, Web of Science, ProQuest, and Embase databases were systematically searched from 2019 until 30 January 2022. A medical subject headings (MeSH) term and keyword search of each database was performed using the Boolean operators OR and AND. Keywords used included: SARS-CoV-2, SARS-CoV-2 variants, and breakthrough infections. The full search strategy for each database is provided in the Supplementary Materials.
Studies were included if they:
  • Were conducted on adult patients with confirmed COVID-19 diagnosis.
  • Reported COVID-19 breakthrough infections.
  • Were written in the English language.
  • Were peer-reviewed.
  • Were either clinical trials, observational studies consisting of prospective cohort, retrospective cohort, case-control studies, case reports, or case series.
  • Studies were excluded if they:
  • Contained incomplete data.
  • Were animal studies.
  • Presented outcomes of no interest.

2.2. Data Extraction and Analysis

Two authors (A.I. and S.G.) independently performed the title and abstract screening. Relevant articles were then retrieved for full-text screening which was performed by two independent authors (P.S.S. and S.G.). All conflicts were resolved by a third author (A.I.). The references of the included articles were also reviewed to identify any articles missed by electronic database search.
The primary outcome of this systematic review was BTIs in vaccinated individuals; the variants causing these BTIs were also noted. The secondary outcomes were clinical and symptom severity in the vaccinated BTIs.

3. Results

Our initial search generated 848 studies; 139 duplicates were removed; 528 studies were excluded by title and abstract screening; and 181 studies were screened for full text. We then identified 33 eligible studies describing infection with COVID-19 in those with prior vaccination (Table 1). Figure 1 depicts in detail the flow of the article selection following the PRISMA guidelines.
The total number of participants in the review who were vaccinated with two doses of vaccine was 651,595. Among these, 25,743 (3.95%) presented with BTIs. The age of the patients ranged from <15 to >83 years with a mean age of 52 years. Out of the 25,743 patients with BTIs, 11,648 (44.24%) were male and 14,068 (54.65%) were female patients. The gender of three patients was reported as “others” and the gender of 18 patients (0.07%) was unknown. BTIs presented from <4 to 185 days with a mean of 52.33 days after full vaccination (defined as completing a primary series of vaccination as recommended for the vaccine type excluding the booster).

Study Type and Geographical Distribution

All 33 studies were observational; 19 were cohort studies, 7 were case reports, 6 were case-control studies, 1 was a longitudinal study, and 1 was a case series. The majority of the studies were conducted in the United States of America (USA) (9), followed by India (7), Italy (4), Germany (3), Israel (2), Brazil (2), Saudi Arabia, Vietnam, Mexico, Netherlands, South Korea, French Guiana, and Singapore.
Most individuals received the mRNA COVID-19 vaccinations Pfizer/BioNTech (23 studies) and Moderna (9 studies). Other vaccinations included were Covishield/AstraZeneca (10 studies), Johnson & Johnson/Janssen vaccine (4 studies), Covaxin (4 studies), Sinovac (3 studies), CanSino, and Sinopharm. Two studies did not specify which mRNA vaccine the patients received.
Among the reviewed studies, 96% of BTIs occurred with the Delta variant (B.1.617.2) and 0.94% of BTIs were due to the Alpha variant (B.1.1.7). Other variants included Gamma -P.1 (0.21%), Beta-B.1.351 (0.15%), and Kappa-B.1.617.1 (0.14%). In addition, 70 patients (0.27%) had BTIs due to non-VOCs; 19 patients were reported as other; and 17 had BTIs due to the Iota (B.1.526) variant. The serum samples of nine patients with BTIs revealed the Epsilon (B.1.427 and B.1.429) variant and four patients with the Mu (B.1.621) variant. The B.1.1.306, B.1.617.3, and 20G variants were seen in two patients each, whereas the Eta (B.1.525) and B.1.560 variants were seen in one patient each. The variant distribution for four patients was reported as unknown. Among the reported mutations, the most commonly identified were the N501Y, E484K, and the L452R mutations. Of interest, the AY.1 lineage of the Delta variant was also identified in a subset of BTIs.
A total of 8.4% of patients had pre-existing comorbidities, which included chronic bronchitis, smoking, obesity, dyslipidemia, type 2 diabetes mellitus, and immunosuppressive conditions. Moreover, 591 (2.3%) of the reported BTIs occurred in healthcare workers (HCW). The symptoms in the BTIs ranged from asymptomatic to severe pneumonia as well as intensive care unit (ICU) admission with mechanical ventilation. The majority of patients recovered without any complications. However, 11.6% of patients were hospitalized requiring oxygen supplementation, intubation, or ECMO, and 0.6% died.

4. Discussion

This systematic review aimed to assess the existing evidence on BTIs of SARS-CoV-2. The results shed light on the distribution of variant type, clinical outcomes, and symptom severity in BTIs, and the associative factors. SARS-CoV-2 structure and function. An understanding of BTIs begins with consideration of the characteristics of SARS-CoV-2, which comprises two groups of proteins: structural proteins (SP) and non-structural proteins (NSP). SPs are encoded by four genes, including E (envelope), M (membrane), S (spike), and N (nucleocapsid) genes [44]. NSPs are mostly enzymes or functional proteins that play a role in viral replication and methylation and may induce host responses to infection [44]. These genes are encoded in several groups, namely ORF1a (NSP1–11), ORF1b (NSP12–16), ORF3a, ORF6, ORF7a, ORF7b, ORF8, and ORF10 [44]. Importantly, not all genetic mutations lead to an increase in viral infectivity. VOCs mostly carry mutations in the spike gene, and the ORF1a frame is the critical region for mutations in the E, M, and S genes [44]. As of February 2022, over 8,600,000 sequences and eight variants of interest or concern have been identified in the global SARS-CoV-2 sequence database operated by the Global Initiative on Sharing Avian Influenza Data (GISAID) [45].
SARS-CoV-2 viral entry into the cells is facilitated by the spike protein, which attaches to the angiotensin-converting enzyme 2 (ACE2) receptor on the cell’s surface. The spike protein is split into two subunits, S1 and S2. Mutations in the S1 region, which is the receptor-binding domain (RBD) site, lower the affinity to neutralizing antibodies and show increased affinity to ACE2 receptors [46,47]. These include the N501Y (N asparagine replaced with Y tyrosine), K417N (lysine K replaced with asparagine N), and E484K (glutamic acid E replaced with lysine K) mutations in the Alpha variant. In the Beta variant, in addition to the N501Y mutation, the E484K mutations were seen, whereas both the E484K and K417T mutations were seen in the Gamma variant. The Delta and Kappa variants share the E484Q (glutamic acid E replaced with glutamine Q) and L452R (leucine L altered by arginine R) mutations. Another mutation unique to the Delta variant is T478K (threonine T replaced by lysine K) [48,49,50]. In addition to the above, mutations at the non-receptor binding site, D614G, increase the density of the spike proteins, thus leading to more functional spikes and increased replication and infectivity [51,52,53].

4.1. COVID-19 Vaccines and Efficacy

As of February 2022, the vaccines recommended by the World Health Organization (WHO) as part of its emergency use listing include the Comirnaty vaccine by Pfizer/BioNTech, the ChAdOx1-S nCov-19 vaccines by AstraZeneca, the Janssen/Ad26.COV 2.S vaccine by Johnson & Johnson, mRNA 1273 by Moderna, Sinopharm COVID-19 vaccine, the CoronaVac vaccine by Sinovac, BBV152 Covaxin by Bharat Biotech, Covishield (ChAdOx1-S [recombinant]) and the Covovax (NVX–CoV2373) vaccine by the Serum Institute of India, the Nuvaxovid (NVX–CoV2373) vaccine by Novavax, and the Inactivated COVID-19 Vaccine (Vero Cell) by the Beijing Institute of Biological Products [54]. The United States Food and Drug Administration (FDA) has approved three different vaccinations against SARS-CoV-2: BNT162b2 (Pfizer-BioNTech), mRNA-1273 (Moderna), and Ad26.COV2. S (Janssen) [55]. The final list of studies included these vaccines, in addition to Ad5-nCoV by CanSino, which was not yet approved for emergency use by WHO or FDA [54].
The Pfizer–BioNTech vaccine is estimated to be 90% effective after the second dose in individuals aged 80 years or older and at least 97% effective in preventing symptomatic COVID-19 cases, hospitalizations, and deaths [56]. The mRNA-1273 vaccine by Moderna is highly effective against SARS-CoV-2 after six months and has an efficacy of 94.1% against COVID-19 14 days after the first dose [57]. The Pfizer-BioNTech and Moderna vaccines contain synthetic nucleoside-modified mRNA encapsulated in lipid nanoparticles (LNP). The mRNA is translated in the cytoplasm of the cells by ribosomes into viral spike proteins activating the host immune response [58]. The AstraZeneca vaccine has a 76% efficacy in preventing symptomatic SARS-CoV-2 infection, specifically during the 15 days after the second dose (with a 29-day interval between the two doses). The vaccine utilizes an inactivated adenovirus DNA as a vector that carries the SARS-CoV-2 spike protein gene, which is then transcribed into mRNA, ultimately activating the immune system and antibody production in a manner similar to the Pfizer-BioNTech and Moderna vaccines [59]. The Sinopharm vaccine is an inactivated vaccine that stimulates the host’s immune system. It has an efficacy of 79% against symptomatic SARS-CoV-2 infection 14 days or more after the second dose (with a 21-day interval between the two doses). The Ad5-nCoV by CanSino is an adenovirus-based viral vector vaccine with an efficacy rate of 57.5% against symptomatic COVID-19 infection [60]. Ad.26.COV2.S or JNJ-78436725 Janssen vaccine is known to elicit a durable immune response for a minimum of eight months post-vaccination with minimal reductions in antibody levels [61]. The vaccine efficacy is 85.4% against critical illness and 93.1 % against hospitalization [62]. This recombinant vaccine contains an adenovirus serotype 26 (Ad26) vector that expresses a SARS-CoV-2 spike protein, which is then translated into mRNA that stimulates cellular immune responses and antibody formation against the S antigen [63]. The Sinovac vaccine is an inactivated virus vaccine, which is 51% efficacious against symptomatic SARS-CoV-2 infection, and Covaxin is an inactivated vaccine that induces a robust immune response using an adjuvant called Alhydroxiquim-II [64]. It has an efficacy of 78% against severe COVID-19 disease [64].

4.2. SARS-CoV-2 Variants and Breakthrough Infections

However, despite the above vaccine efficacy rates, BTIs occur. Most BTIs in our review were due to the Delta variant. This confirms the results of other studies in the literature where lowered effectiveness of the vaccines has been due to the highly transmissible Delta variant (which is 60% more transmissible than the Alpha variant) [7,64,65]. B.1.617.1 also partially impairs neutralizing antibodies elicited by BNT162b2 and ChAdOx1 nCoV-19 (Covishield) vaccines [20]. The T478K mutation in the Delta variant may also facilitate an escape by antibodies generated by vaccines or natural infection [45,66]. The AY.4 lineage of the Delta variant was seen predominantly in hospitalized patients vaccinated by the CanSino vaccine where around 67% of vaccinated individuals developed milder symptoms of COVID-19 [23]. Despite the asymptomatic or mild disease, the BTIs were associated with low levels of neutralizing antibodies, high viral load, and prolonged positivity on PCR tests, thus potentially contributing to ongoing transmission from fully vaccinated individuals [66]. Another study that analyzed the viral loads of over 16,000 infections during the predominantly Delta wave in Israel, found lower viral loads in BTIs in fully vaccinated individuals compared to infections in the unvaccinated. However, this effect started to decline after 2 months [23].
Moderate reductions in vaccine efficacy with the E484K, L452R, S477N, and N501Y mutations during the Delta variant surge were also observed in New York City between November 2020 and August 2021 [34]. However, the immune escape mutations in the spike protein gene were evenly distributed among the partially and fully vaccinated cases [34]. BTIs in which Delta was the predominant variant also revealed lowered humoral and cell-mediated immunity with Eotaxin, SCF, SDF-1a, and PIGF-1; low memory B cell cytokines (IL-1b, TNF, IFNc) and chemokines (Eotaxin, SCF, SDF-1a, PIGF-1); increased levels of plasmablast cells; and a higher frequency of CD4+ and IL-2 cells after vaccination with the BNT162b2 vaccine [42]. Compared to plasma antibodies, memory B cells were found to have a higher neutralizing effect against VOCs potentially implying that the lowered memory B cells with the Delta variant may have led to BTIs [67]. Data also shows that there is a 3-fold and 16-fold reduction in neutralization against the Delta and Beta variants as compared with the Alpha variant with BNT162b2 vaccinated sera, and a 5-fold and 9-fold reduction against the same with ChAdOx1 nCoV-19 [68].
The N501Y mutation predominantly seen in the studies yielded by our review also lowers the neutralization capacity of the vaccines [25,69]. Infections with the N501Y mutation in the Alpha variant led to low neutralizing antibodies against the AZD1222 vaccine compared to non-Alpha variants [14].
Similarly, both the E484K and S477 mutations, found in P.1 and P.6 respectively, are reported to escape neutralization by a range of mAbs [70]. E484K is also associated with a decrease in the neutralizing activity of convalescent and post-vaccination (BNT162b2) sera [71,72,73]. E484K causes resistance to many class 2 RBD-directed antibodies, including bamlanivimab [74,75]. The most potent mRNA vaccine-elicited monoclonal antibodies were over 10-fold less effective against pseudotyped viruses carrying the E484K mutation [18]. In the study by Olsen et al., BTIs in fully vaccinated patients due to the E484K variant mutations in the Alpha variant had a significantly lower cycle threshold (a proxy for higher virus load) and significantly higher hospitalization rate [40]. Other variants (e.g., B.1.429 and B.1.427, P.1, P.2 (Zeta), and R.1) also increased rapidly, although the magnitude was less than that in Alpha [40]. Additionally, patients infected with the B.1.617.1 or B.1.617.2 variants also had a high rate of hospitalization despite vaccination51. In addition to the above, the L452R mutation, where Leucine-452 that is located at the point of interaction with the ACE2 receptor in the RBD receptor is replaced by arginine, also causes greater receptor affinity and escape from neutralizing antibodies [20,24,76].
Although most BTIs reported in the final 33 studies occurred before full vaccine-induced immunity, a few reinfections were also reported despite the presence of neutralizing antibodies [28]. Schulte et al. reported the case of an HCW who developed infection with the Eta (B.1.525) variant despite the presence of neutralizing antibodies seven weeks after vaccination [77]. The authors hypothesized that this could be attributed to the absence of an N-specific antibody and spike-based neutralization post-vaccination, which prevents antibody responses to the nucleocapsid, thus demonstrating the need for protective measures such as masks even after full vaccination [78]. As per their study, neutralization assays demonstrated differences against variants by a factor of 4. Variant B.1.525 is the best at neutralizing, followed by the B.3 and B.1.1.7 variants. The B.1.351 variant neutralizes the least. The study concluded that differences in spike proteins play a crucial role in neutralization [78]. Another study showed similar results, with higher neutralization against B.1.525 and B.1.1.7 and weaker neutralization against B.1.351 compared to B.1 [79].

4.3. Breakthrough Infections in at-Risk Populations

4.3.1. Immunosuppression

Laboratory and clinical investigations among the final 33 studies showed that post-vaccine antibody responses against SARS-CoV-2 variants are less than antibody responses against wild SARS-CoV-2 but are still protective against severe disease and death [80,81]. This phenomenon is applicable for immunocompetent patients who are mounting high antibody responses that can overcome the mutations in the spike protein but inadequate for solid organ transplant recipients and those with immunosuppression who mount a suboptimal antibody response against wild SARS-CoV-2 [82]. In patients with solid organ transplantation, lower antibody response and waning immunity render those patients at higher risk of BTIs after vaccination. In addition, immunosuppressive medications such as calcineurin inhibitors, mycophenolic acid, and antiproliferative drugs were reported to increase the risk of SARS-CoV-2 BTIs by lowering the immunogenicity of vaccines and in developing an adequate immune response [17,83].
In a study by Deng et al., BTIs occurred in fully vaccinated individuals over four weeks of follow-up [76]. Fourteen patients were identified and 42.8% were solid organ transplant (SOT) recipients. Another study by Almaghrabi et al. demonstrated that BTIs after COVID-19 mRNA vaccination were highest in immunocompromised patients with primary immunodeficiencies, active malignancies, and transplantation [84]. In one study, patients with cancer undergoing chemotherapy had lower levels of antibodies compared to healthy controls following the second dose of the BNT162b2 vaccine [43]. Sun et al. demonstrated that full vaccination was associated with a reduced rate of BTIs regardless of the immune status [85]. However, even among these, the rate of BTIs was still higher in the immunocompromised group thus necessitating the need for alternate strategies such as monoclonal antibodies and non-pharmaceutical personal protective measures such as masks, social distancing, and avoiding large gatherings [85]. Immunosuppressed individuals also had a higher risk factor for BTIs when controlled for age, gender, and comorbidities [85]. To combat this, the third dose of the vaccine was initially recommended for immunocompromised patients [86]. However, studies still revealed a substantially lower immune response compared to the general population, thus paving the way for treatment with monoclonal antibodies [87,88].

4.3.2. Aging

Our study revealed that the aging of the immune system or immunosenescence, which decreases the number of naive T & B cells, can also lead to reduced vaccine efficacy, particularly in older individuals, thus predisposing them to BTIs [84,89]. A recent study that described humoral and cell-mediated responses after two doses of mRNA vaccination against SARS-CoV-2 VOCs in relation to different age groups showed that patients above eighty years old had lower cell-mediated responses compared to younger patients [90]. Another multicenter study in the USA that examined the factors affecting COVID-19 immunity in individuals who were administered two doses of the BNT162b2 vaccine, found that antibody titers were negatively correlated with increasing age [11]. Sun et al. who analyzed the risk of BTIs in immunocompromised patients, found that although full vaccination was associated with a 28% reduced risk of BTIs, older individuals still had a higher rate of BTIs [85].

4.3.3. Occupational Risk

Lastly, the results showed that reinfections were seen due to prolonged exposure, predominantly in healthcare workers despite vaccination [16,20,29,31,41,69,91]. Although occupational exposure other than healthcare settings was not reported in the studies in our review, prolonged exposure to COVID-19 has also been known to occur in retail workers, meat and poultry workers, shelter staff, call center staff, and transit operators [92]. As per the WHO prior to the availability of COVID-19 vaccines, HCWs accounted for 14% of COVID-19 cases [93]. Several studies have also reported milder infection in HCWs, and this could be due to the availability of frequent testing and detection [94]. Although our review reported no comorbidities among HCWs, around 6% of HCWs in previous studies who presented with severe infection had comorbidities such as obesity [94]. The risk of BTIs among HCWs is said to have declined after the introduction of COVID-19 vaccinations, with a greater proportion of infections from community exposure. Despite this, BTIs due to waning immunity and the emergence of variants still present a risk to patients and coworkers, highlighting the need for ongoing screening and testing in this population [95].

4.3.4. Ct (Cycle Threshold) Values & Viral Loads

The Ct (cycle threshold) value is the number of cycles it takes for the RT-PCR test to detect the virus. Ct levels are inversely proportional to the amount of target nucleic acid in the sample. The higher the amount of the viral nucleic acid in the sample, the lower the Ct value. An important issue for controlling the spread of variants is to determine if the BTI is associated with high viral loads that may result in a secondary spread. Previous studies reported that low viral loads and a high Ct value were detected following vaccination [23,96]. In contrast, a study by Deng et al. detected relatively high viral loads (median Ct of 19.6) even in non-immunosuppressed vaccinated subjects exhibiting asymptomatic or mild infection [28]. This finding is consistent with other studies that reported that individuals with BTIs with the Alpha variant had a significantly lower Ct value compared to non-Alpha patients [40]. Although this could be viewed as an enhanced transmissibility potential of Alpha, no clear correlation between Ct values and transmission rates has been confirmed.

4.3.5. Heterogenous Vaccination Regimens

Numerous studies have shown a stronger immune response where mix and match vaccine regimens are used [19,97,98]. Individuals who receive different types of COVID-19 vaccines for their first, second, and subsequent booster doses show more potent immune responses. One study in our review described the transmission of infection from a fully vaccinated spouse, thus hypothesizing that this was due to a lack of immune response against the nucleocapsid protein, against which the mRNA vaccines are not effective. A study by Nordstrom et al., found that those who received a mixed vaccine regimen were 68% less likely to develop an infection compared to unvaccinated people, whereas those who received two doses of the same vaccine (Astra Zeneca) were 50% less likely to do so [82]. Another study also showed similar results where the vaccine efficacy against SARS-CoV-2 infection was 88% when ChAdOx1 and an mRNA vaccine were combined [83]. Additionally, there is some evidence that heterologous vaccination may also confer greater protection, with combined cellular and humoral immunity in immunocompromised individuals [84].

5. Limitations

Importantly, our study has several notable limitations. Given the nature of surveillance, testing, and reporting, oftentimes not all cases are documented. There may also have been some overlap in status (e.g., some individuals who had been vaccinated may have been previously infected at some point). We describe the cases that have been documented in the scientific literature. Additionally, we must consider the possibility of asymptomatic viral transmission among vaccinated individuals; these numbers are not reflected in these studies. Thus, it could be possible that the extent of SARS-CoV-2 transmissibility among vaccinated individuals is greater than expected as per our current understanding. Data reported from hospital settings where exposure to infection is higher, may not reflect the infection rates in the general population. Also, data in several studies were collected from electronic medical records and hence may be prone to error. Similarly, the history of exposure in those with BTIs may not always be accurate and the source of infection is not always known. Among the data from the immunocompromised patients, there were no specific mentions of which condition may have had a greater contribution towards the lowered immunity.

6. Conclusions

BTIs remain a critical challenge in controlling the epidemic. Whether individuals with BTIs contribute substantially to the onward transmission of SARS-CoV-2 in the population currently remains unclear. In our review, we found that BTIs do not reflect selection towards specific immunity-evading variants, rather, they reflect the most prevalent variant in the community at that time. Hence a standardized surveillance reporting protocol for suspected BTIs is necessary to better assess the nature and extent of the burden of reinfections in vaccinated individuals. Studies on BTIs could be helpful to understand the neutralizing response to SARS-CoV-2 infection and the corresponding immunity. However, the absence of systematic genomic sequencing of positive cases worldwide impedes advances in public health surveillance to manage the pandemic at the individual and collective levels. Further investigations, including a genetic comparison of SARS-CoV-2 strains, would be beneficial to understanding the frequency and pathophysiology of SARS-CoV-2 reinfections. Although COVID-19 vaccines have proven to be highly effective, the possibility of BTIs remains a reality, particularly in the context of emerging variants of concern. Many factors contribute to BTIs including the transmission dynamics of SARS-CoV-2 variants and their biological capacity to survive, behavioral characteristics of individuals, and vaccination status. Future studies should explore the role of combining different types of vaccines, post-exposure prophylaxis, and close monitoring for disease progression including disease progression and transmission in high-risk individuals such as HCWs or immunocompromised patients.

Supplementary Materials

The following are available online at https://www.mdpi.com/article/10.3390/tropicalmed7050081/s1, The full search strategy for each database.

Author Contributions

Conceptualization, S.G., and N.D.; Methodology, P.T., S.G., and N.D.; Formal Analysis, S.G., A.I., S.P., and P.S.S.; Data Curation, S.G., A.I., S.P., P.S.S., P.S., and E.X.; Writing—Original Draft Preparation: S.G., A.I., N.D., S.P., and P.S.S.; Writing—Review and Editing: S.G., A.I., and N.D.; Supervision, S.G., S.M., J.M., and G.M.; Project Administration, S.G., S.M., J.M., and G.M. 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

Not Applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. World Health Organization. WHO Coronavirus (COVID-19) Dashboard. 2022. Available online: https://covid19.who.int/ (accessed on 4 February 2022).
  2. CDC COVID-19 Vaccine Breakthrough Case Investigations Team. COVID-19 Vaccine Breakthrough Infections Reported to CDC—United States, 1 January—30 April 2021. MMWR Morb. Mortal. Wkly. Rep. 2021, 70, 792–793. [Google Scholar] [CrossRef] [PubMed]
  3. Centers for Disease Control and Prevention. Rates of COVID-19 Cases and Deaths by Vaccination Status. 2022. Available online: https://covid.cdc.gov/covid-data-tracker/#rates-by-vaccine-status (accessed on 4 March 2022).
  4. Centers for Disease Control and Prevention. Investigative Criteria for Suspected Cases of SARS-CoV-2 Reinfection (ICR). 2020. Available online: https://www.cdc.gov/coronavirus/2019-ncov/php/invest-criteria.html (accessed on 9 January 2022).
  5. Krause, P.R.; Fleming, T.R.; Longini, I.M.; Peto, R.; Briand, S.; Heymann, D.L.; Beral, V.; Snape, M.D.; Rees, H.; Ropero, A.M.; et al. SARS-CoV-2 Variants and Vaccines. N. Engl. J. Med. 2021, 385, 179–186. [Google Scholar] [CrossRef] [PubMed]
  6. World Health Organization. Classification of Omicron (B.1.1.529): SARS-CoV-2 Variant of Concern. 2021. Available online: https://www.who.int/news/item/26-11-2021-classification-of-omicron-(b.1.1.529)-SARS-CoV-2-variant-of-concern (accessed on 27 November 2021).
  7. Lopez Bernal, J.; Andrews, N.; Gower, C.; Gallagher, E.; Simmons, R.; Thelwall, S.; Stowe, J.; Tessier, E.; Groves, N.; Dabrera, G.; et al. Effectiveness of COVID-19 Vaccines against the B.1.617.2 (Delta) Variant. N. Engl. J. Med. 2021, 385, 585–594. [Google Scholar] [CrossRef] [PubMed]
  8. Fendler, A.; Shepherd, S.; Au, L.; Wilkinson, K.; Wu, M.; Byrne, F.; Cerrone, M.; Schmitt, A.M.; Joharatnam-Hogan, N.; Shum, B.; et al. Adaptive immunity and neutralizing antibodies against SARS-CoV-2 variants of concern following vaccination in patients with cancer: The Capture study. Nat. Cancer 2021, 2, 1321–1337. [Google Scholar] [CrossRef] [PubMed]
  9. Seyed Alinaghi, S.; Oliaei, S.; Kianzad, S.; Afsahi, A.M.; MohsseniPour, M.; Barzegary, A.; Barzegary, A.; Mirzapour, P.; Behnezhad, F.; Noori, T.; et al. Reinfection risk of novel coronavirus (COVID-19): A systematic review of current evidence. World J. Virol. 2020, 9, 79–90. [Google Scholar] [CrossRef]
  10. 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. Syst. Rev. 2021, 10, 89. [Google Scholar] [CrossRef]
  11. Bergwerk, M.; Gonen, T.; Lustig, Y.; Amit, S.; Lipsitch, M.; Cohen, C.; Mandelboim, M.; Levin, E.G.; Rubin, C.; Indenbaum, V.; et al. COVID-19 Breakthrough Infections in Vaccinated Health Care Workers. N. Engl. J. Med. 2021, 385, 1474–1484. [Google Scholar] [CrossRef]
  12. Estofolete, C.F.; Banho, C.A.; Campos, G.R.F.; Marques, B.C.; Sacchetto, L.; Ullmann, L.S.; Possebon, F.S.; Machado, L.F.; Syrio, J.D.; Araújo Junior, J.P.; et al. Case Study of Two Post Vaccination SARS-CoV-2 Infections with P1 Variants in CoronaVac Vaccinees in Brazil. Viruses 2021, 13, 1237. [Google Scholar] [CrossRef]
  13. Fabiani, M.; Margiotti, K.; Viola, A.; Mesoraca, A.; Giorlandino, C. Mild Symptomatic SARS-CoV-2 P.1 (B.1.1.28) Infection in a Fully Vaccinated 83-Year-Old Man. Pathogens 2021, 10, 614. [Google Scholar] [CrossRef]
  14. Philomina, J.B.; Jolly, B.; John, N.; Bhoyar, R.C.; Majeed, N.; Senthivel, V.; Cp, F.; Rophina, M.; Vasudevan, B.; Imran, M.; et al. Genomic survey of SARS-CoV-2 vaccine breakthrough infections in healthcare workers from Kerala, India. J. Infect. 2021, 83, 237–279. [Google Scholar] [CrossRef]
  15. Hacisuleyman, E.; Hale, C.; Saito, Y.; Blachere, N.E.; Bergh, M.; Conlon, E.G.; Schaefer-Babajew, D.J.; DaSilva, J.; Muecksch, F.; Gaebler, C.; et al. Vaccine Breakthrough Infections with SARS-CoV-2 Variants. N. Engl. J. Med. 2021, 384, 2212–2218. [Google Scholar] [CrossRef] [PubMed]
  16. Kroidl, I.; Mecklenburg, I.; Schneiderat, P.; Müller, K.; Girl, P.; Wölfel, R.; Sing, A.; Dangel, A.; Wieser, A.; Hoelscher, M. Vaccine breakthrough infection and onward transmission of SARS-CoV-2 Beta (B.1.351) variant, Bavaria, Germany, February to March 2021. Eurosurveillance 2021, 26, 2100673. [Google Scholar] [CrossRef] [PubMed]
  17. Almaghrabi, R.S.; Alhamlan, F.S.; Dada, A.; Al-Tawfiq, J.A.; Al Hroub, M.K.; Saeedi, M.F.; Alamri, M.; Alhothaly, B.; Alqasabi, A.; Al-Qahtani, A.A.; et al. Outcome of SARS-CoV-2 variant breakthrough infection in fully immunized solid organ transplant recipients. J. Infect. Public Health 2022, 15, 51–55. [Google Scholar] [CrossRef] [PubMed]
  18. Olsen, R.J.; Christensen, P.A.; Long, S.W.; Subedi, S.; Hodjat, P.; Olson, R.; Nguyen, M.; Davis, J.J.; Yerramilli, P.; Saavedra, M.O.; et al. Trajectory of Growth of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Variants in Houston, Texas, January through May 2021, Based on 12,476 Genome Sequences. Am. J. Pathol. 2021, 191, 1754–1773. [Google Scholar] [CrossRef]
  19. Schmidt, T.; Klemis, V.; Schub, D.; Schneitler, S.; Reichert, M.C.; Wilkens, H.; Sester, U.; Sester, M.; Mihm, J. Cellular immunity predominates over humoral immunity after homologous and heterologous mRNA and vector-based COVID-19 vaccine regimens in solid organ transplant recipients. Am. J. Transplant. 2021, 21, 3990–4002. [Google Scholar] [CrossRef]
  20. Chau, N.V.V.; Ngoc, N.M.; Nguyet, L.A.; Quang, V.M.; Ny, N.T.H.; Khoa, D.B.; Phong, N.T.; Toan, L.M.; Hong, N.T.; Tuyen, N.T.K.; et al. An observational study of breakthrough SARS-CoV-2 Delta variant infections among vaccinated healthcare workers in Vietnam. EClinicalMedicine 2021, 41, 101143. [Google Scholar] [CrossRef]
  21. Connor, B.A.; Couto-Rodriguez, M.; Barrows, J.E.; Gardner, M.; Rogova, M.; O’Hara, N.B.; Nagy-Szakal, D. Monoclonal Antibody Therapy in a Vaccine Breakthrough SARS-CoV-2 Hospitalized Delta (B1.617.2) Variant Case. Int. J. Infect. Dis. 2021, 110, 232–234. [Google Scholar] [CrossRef]
  22. Gharpure, R.; Sami, S.; Vostok, J.; Johnson, H.; Hall, N.; Foreman, A.; Sabo, R.T.; Schubert, P.L.; Shephard, H.; Brown, V.R.; et al. Multistate Outbreak of SARS-CoV-2 Infections, Including Vaccine Breakthrough Infections, Associated with Large Public Gatherings, United States. Emerg. Infect. Dis. 2022, 28, 35–43. [Google Scholar] [CrossRef]
  23. Galán-Huerta, K.A.; Flores-Treviño, S.; Salas-Treviño, D.; Bocanegra-Ibarias, P.; Rivas-Estilla, A.M.; Pérez-Alba, E.; Lozano-Sepúlveda, S.A.; Arellanos-Soto, D.; Camacho-Ortiz, A. Prevalence of SARS-CoV-2 Variants of Concern and Variants of Interest in COVID-19 Breakthrough Infections in a Hospital in Monterrey, Mexico. Viruses 2022, 14, 154. [Google Scholar] [CrossRef]
  24. Deng, X.; Evdokimova, M.; O’Brien, A.; Rowe, C.L.; Clark, N.M.; Harrington, A.; Reid, G.E.; Uprichard, S.L.; Baker, S.C. Breakthrough Infections with Multiple Lineages of SARS-CoV-2 Variants Reveals Continued Risk of Severe Disease in Immunosuppressed Patients. Viruses 2021, 13, 1743. [Google Scholar] [CrossRef]
  25. De Souza, W.M.; Muraro, S.P.; Souza, G.F.; Amorim, M.R.; Sesti-Costa, R.; Mofatto, L.S.; Forato, J.; Barbosa, P.P.; Toledo-Teixeira, D.A.; Bispo-Dos-Santos, K.; et al. Clusters of SARS-CoV-2 Lineage B.1.1.7 Infection after Vaccination with Adenovirus-Vectored and Inactivated Vaccines. Viruses 2021, 13, 2127. [Google Scholar] [CrossRef] [PubMed]
  26. Gupta, N.; Kaur, H.; Yadav, P.D.; Mukhopadhyay, L.; Sahay, R.R.; Kumar, A.; Nyayanit, D.A.; Shete, A.M.; Patil, S.; Majumdar, T.; et al. Clinical Characterization and Genomic Analysis of Samples from COVID-19 Breakthrough Infections during the Second Wave among the Various States of India. Viruses 2021, 13, 1782. [Google Scholar] [CrossRef] [PubMed]
  27. Kale, P.; Gupta, E.; Bihari, C.; Patel, N.; Rooge, S.; Pandey, A.; Bajpai, M.; Khillan, V.; Chattopadhyay, P.; Devi, P.; et al. Vaccine Breakthrough Infections by SARS-CoV-2 Variants after ChAdOx1 nCoV-19 Vaccination in Healthcare Workers. Vaccines 2021, 10, 54. [Google Scholar] [CrossRef]
  28. Schulte, B.; Marx, B.; Korencak, M.; Emmert, D.; Aldabbagh, S.; Eis-Hübinger, A.M.; Streeck, H. Case Report: Infection With SARS-CoV-2 in the Presence of High Levels of Vaccine-Induced Neutralizing Antibody Responses. Front. Med. 2021, 8, 704719. [Google Scholar] [CrossRef] [PubMed]
  29. Malhotra, S.; Mani, K.; Lodha, R.; Bakhshi, S.; Mathur, V.P.; Gupta, P.; Kedia, S.; Sankar, J.; Kumar, P.; Kumar, A.; et al. SARS-CoV-2 Reinfection Rate and Estimated Effectiveness of the Inactivated Whole Virion Vaccine BBV152 Against Reinfection Among Health Care Workers in New Delhi, India. JAMA Netw. Open 2022, 5, e2142210. [Google Scholar] [CrossRef]
  30. Shastri, J.; Parikh, S.; Aggarwal, V.; Agrawal, S.; Chatterjee, N.; Shah, R.; Devi, P.; Mehta, P.; Pandey, R. Severe SARS-CoV-2 Breakthrough Reinfection with Delta Variant After Recovery from Breakthrough Infection by Alpha Variant in a Fully Vaccinated Health Worker. Front. Med. 2021, 8, 737007. [Google Scholar] [CrossRef] [PubMed]
  31. Rovida, F.; Cassaniti, I.; Paolucci, S.; Percivalle, E.; Sarasini, A.; Piralla, A.; Giardina, F.; Sammartino, J.C.; Ferrari, A.; Bergami, F.; et al. SARS-CoV-2 vaccine breakthrough infections with the alpha variant are asymptomatic or mildly symptomatic among health care workers. Nat. Commun. 2021, 12, 6032. [Google Scholar] [CrossRef]
  32. Rümke, L.W.; Groenveld, F.C.; van Os, Y.M.G.; Praest, P.; Tanja, A.A.N.; de Jong, D.T.C.M.; Symons, J.; Schuurman, R.; Reinders, T.; Hofstra, L.M.; et al. In-depth Characterization of Vaccine Breakthrough Infections With SARS-CoV-2 Among Health Care Workers in a Dutch Academic Medical Center. Open Forum Infect. Dis. 2021, 9, ofab553. [Google Scholar] [CrossRef]
  33. Yi, S.; Kim, J.M.; Choe, Y.J.; Hong, S.; Choi, S.; Ahn, S.B.; Kim, M.; Park, Y.J. SARS-CoV-2 Delta Variant Breakthrough Infection and Onward Secondary Transmission in Household. J. Korean Med. Sci. 2022, 37, e12. [Google Scholar] [CrossRef]
  34. Robilotti, E.V.; Whiting, K.; Lucca, A.; Poon, C.; Guest, R.; McMillen, T.; Jani, K.; Solovyov, A.; Kelson, S.; Browne, K.; et al. Clinical and Genomic Characterization of SARS-CoV-2 infections in mRNA Vaccinated Health Care Personnel in New York City. Clin. Infect. Dis. 2021, ciab886. [Google Scholar] [CrossRef]
  35. Vignier, N.; Bérot, V.; Bonnave, N.; Peugny, S.; Ballet, M.; Jacoud, E.; Michaud, C.; Gaillet, M.; Djossou, F.; Blanchet, D.; et al. Breakthrough Infections of SARS-CoV-2 Gamma Variant in Fully Vaccinated Gold Miners, French Guiana, 2021. Emerg. Infect. Dis. 2021, 27, 2673–2676. [Google Scholar] [CrossRef] [PubMed]
  36. Tober-Lau, P.; Schwarz, T.; Hillus, D.; Spieckermann, J.; Helbig, E.T.; Lippert, L.J.; Thibeault, C.; Koch, W.; Bergfeld, L.; Niemeyer, D.; et al. Outbreak of SARS-CoV-2 B.1.1.7 Lineage after Vaccination in Long-Term Care Facility, Germany, February–March 2021. Emerg. Infect. Dis. 2021, 27, 2169–2173. [Google Scholar] [CrossRef] [PubMed]
  37. Servellita, V.; Morris, M.K.; Sotomayor-Gonzalez, A.; Gliwa, A.S.; Torres, E.; Brazer, N.; Zhou, A.; Hernandez, K.T.; Sankaran, M.; Wang, B.; et al. Predominance of antibody-resistant SARS-CoV-2 variants in vaccine breakthrough cases from the San Francisco Bay Area, California. Nat. Microbiol. 2022, 7, 277–288. [Google Scholar] [CrossRef] [PubMed]
  38. Singer, S.R.; Angulo, F.J.; Swerdlow, D.L.; McLaughlin, J.M.; Hazan, I.; Ginish, N.; Anis, E.; Mendelson, E.; Mor, O.; Zuckerman, N.S.; et al. Effectiveness of BNT162b2 mRNA COVID-19 vaccine against SARS-CoV-2 variant Beta (B.1.351) among persons identified through contact tracing in Israel: A prospective cohort study. EClinicalMedicine 2021, 42, 101190. [Google Scholar] [CrossRef] [PubMed]
  39. Thangaraj, J.W.V.; Yadav, P.; Kumar, C.G.; Shete, A.; Nyayanit, D.A.; Rani, D.S.; Kumar, A.; Kumar, M.S.; Sabarinathan, R.; Saravana Kumar, V.; et al. Predominance of delta variant among the COVID-19 vaccinated and unvaccinated individuals, India, May 2021. J. Infect. 2022, 84, 94–118. [Google Scholar] [CrossRef] [PubMed]
  40. Motozono, C.; Toyoda, M.; Zahradnik, J.; Saito, A.; Nasser, H.; Tan, T.S.; Ngare, I.; Kimura, I.; Uriu, K.; Kosugi, Y.; et al. SARS-CoV-2 spike L452R variant evades cellular immunity and increases infectivity. Cell Host Microbe 2021, 29, 1124–1136. [Google Scholar] [CrossRef]
  41. Singh, U.B.; Rophina, M.; Chaudhry, R.; Senthivel, V.; Bala, K.; Bhoyar, R.C.; Jolly, B.; Jamshed, N.; Imran, M.; Gupta, R.; et al. Variants of concern responsible for SARS-CoV-2 vaccine breakthrough infections from India. J. Med. Virol. 2022, 94, 1696–1700. [Google Scholar] [CrossRef]
  42. Tay, M.Z.; Rouers, A.; Fong, S.W.; Goh, Y.S.; Chan, Y.H.; Chang, Z.W.; Xu, W.; Tan, C.W.; Chia, W.N.; Torres-Ruesta, A.; et al. Decreased memory B cell frequencies in COVID-19 delta variant vaccine breakthrough infection. EMBO Mol. Med. 2022, 14, e15227. [Google Scholar] [CrossRef]
  43. Sun, J.; Zheng, Q.; Madhira, V.; Olex, A.L.; Anzalone, A.J.; Vinson, A.; Singh, J.A.; French, E.; Abraham, A.G.; Mathew, J.; et al. Association Between Immune Dysfunction and COVID-19 Breakthrough Infection After SARS-CoV-2 Vaccination in the US. JAMA Intern. Med. 2022, 182, 153–162. [Google Scholar] [CrossRef]
  44. Wu, F.; Zhao, S.; Yu, B.; Chen, Y.M.; Wang, W.; Song, Z.G.; Hu, Y.; Tao, Z.W.; Tian, J.H.; Pei, Y.Y.; et al. A new coronavirus associated with human respiratory disease in China. Nature 2020, 579, 265–269. [Google Scholar] [CrossRef] [Green Version]
  45. GISAID. Tracking of Variants. 2022. Available online: https://www.gisaid.org/hcov19-variants (accessed on 5 March 2022).
  46. Piccoli, L.; Park, Y.J.; Tortorici, M.A.; Czudnochowski, N.; Walls, A.C.; Beltramello, M.; Silacci-Fregni, C.; Pinto, D.; Rosen, L.E.; Bowen, J.E.; et al. Mapping Neutralizing and Immunodominant Sites on the SARS-CoV-2 Spike Receptor-Binding Domain by Structure-Guided High-Resolution Serology. Cell 2020, 183, 1024–1042.e21. [Google Scholar] [CrossRef] [PubMed]
  47. Starr, T.N.; Greaney, A.J.; Addetia, A.; Hannon, W.W.; Choudhary, M.C.; Dingens, A.S.; Li, J.Z.; Bloom, J.D. Prospective mapping of viral mutations that escape antibodies used to treat COVID-19. Science 2021, 371, 850–854. [Google Scholar] [CrossRef] [PubMed]
  48. Zhou, D.; Dejnirattisai, W.; Supasa, P.; Liu, C.; Mentzer, A.J.; Ginn, H.M.; Zhao, Y.; Duyvesteyn, H.M.E.; Tuekprakhon, A.; Nutalai, R.; et al. Evidence of escape of SARS-CoV-2 variant B.1.351 from natural and vaccine-induced sera. Cell 2021, 184, 2348–2361.e6. [Google Scholar] [CrossRef] [PubMed]
  49. Cele, S.; Gazy, I.; Jackson, L.; Hwa, S.; Tegally, H.; Lustig, G.; Giandhari, J.; Pillay, S.; Wilkinson, E.; Naidoo, Y.; et al. Escape of SARS-CoV-2 501Y.V2 from neutralization by convalescent plasma. Nature 2021, 593, 142–146. [Google Scholar] [CrossRef]
  50. Wang, P.; Casner, R.G.; Nair, M.S.; Wang, M.; Yu, J.; Cerutti, G.; Liu, L.; Kwong, P.D.; Huang, Y.; Shapiro, L.; et al. Increased resistance of SARS-CoV-2 variant P.1 to antibody neutralization. Cell Host Microbe 2021, 29, 747–751.e4. [Google Scholar] [CrossRef]
  51. Starr, T.N.; Greaney, A.J.; Hilton, S.K.; Ellis, D.; Crawford, K.H.D.; Dingens, A.S.; Navarro, M.J.; Bowen, J.E.; Tortorici, M.A.; Walls, A.C.; et al. Deep Mutational Scanning of SARS-CoV-2 Receptor Binding Domain Reveals Constraints on Folding and ACE2 Binding. Cell 2020, 182, 1295–1310.e20. [Google Scholar] [CrossRef]
  52. Zhang, L.; Jackson, C.B.; Mou, H.; Ojha, A.; Peng, H.; Quinlan, B.D.; Rangarajan, E.S.; Pan, A.; Vanderheiden, A.; Suthar, M.S.; et al. SARS-CoV-2 spike-protein D614G mutation increases virion spike density and infectivity. Nat. Commun. 2020, 11, 6013. [Google Scholar] [CrossRef]
  53. Hou, Y.J.; Chiba, S.; Halfmann, P.; Ehre, C.; Kuroda, M.; Dinnon, K.H., 3rd; Leist, S.R.; Schäfer, A.; Nakajima, N.; Takahashi, K.; et al. SARS-CoV-2 D614G variant exhibits efficient replication ex vivo and transmission in vivo. Science 2020, 370, 1464–1468. [Google Scholar] [CrossRef]
  54. World Health Organization. Coronavirus Disease (COVID-19): Vaccines. 2022. Available online: https://www.who.int/news-room/questions-and-answers/item/coronavirus-disease-(covid-19)-vaccines (accessed on 4 March 2022).
  55. Lythgoe, M.P.; Middleton, P. Comparison of COVID-19 Vaccine Approvals at the US Food and Drug Administration, European Medicines Agency, and Health Canada. JAMA Netw. Open. 2021, 4, e2114531. [Google Scholar] [CrossRef]
  56. Lopez Bernal, J.; Andrews, N.; Gower, C.; Robertson, C.; Stowe, J.; Tessier, E.; Simmons, R.; Cottrell, S.; Roberts, R.; O’Doherty, M.; et al. Effectiveness of the Pfizer-BioNTech and Oxford-AstraZeneca vaccines on COVID-19 related symptoms, hospital admissions, and mortality in older adults in England: Test negative case-control study. BMJ 2021, 373, n1088. [Google Scholar] [CrossRef]
  57. Doria-Rose, N.; Suthar, M.S.; Makowski, M.; O’Connell, S.; McDermott, A.B.; Flach, B.; Ledgerwood, J.E.; Mascola, J.R.; Graham, B.S.; Lin, B.C.; et al. mRNA-1273 Study Group. Antibody Persistence through 6 Months after the Second Dose of mRNA-1273 Vaccine for COVID-19. N. Engl. J. Med. 2021, 384, 2259–2261. [Google Scholar] [CrossRef] [PubMed]
  58. Zhou, X.; Jiang, X.; Qu, M.; Aninwene, G.E.; Jucaud, V.; Moon, J.J.; Gu, Z.; Sun, W.; Khademhosseini, A. Engineering Antiviral Vaccines. ACS Nano 2020, 14, 12370–12389. [Google Scholar] [CrossRef] [PubMed]
  59. Mascellino, M.T.; Di Timoteo, F.; De Angelis, M.; Oliva, A. Overview of the Main Anti-SARS-CoV-2 Vaccines: Mechanism of Action, Efficacy and Safety. Infect. Drug Resist. 2021, 14, 3459–3476. [Google Scholar] [CrossRef] [PubMed]
  60. Halperin, S.A.; Ye, L.; MacKinnon-Cameron, D.; Smith, B.; Cahn, P.E.; Ruiz-Palacios, G.M.; Ikram, A.; Lanas, F.; Guerrero, M.L.; Navarro, S.R.M.; et al. CanSino COVID-19 Global Efficacy Study Group. Final efficacy analysis, interim safety analysis, and immunogenicity of a single dose of recombinant novel coronavirus vaccine (adenovirus type 5 vector) in adults 18 years and older: An international, multicentre, randomised, double-blinded, placebo-controlled phase 3 trial. Lancet 2022, 399, 237–248. [Google Scholar] [CrossRef]
  61. Barouch, D.H.; Stephenson, K.E.; Sadoff, J.; Yu, J.; Chang, A.; Gebre, M.; McMahan, K.; Liu, J.; Chandrashekar, A.; Patel, S.; et al. Durable Humoral and Cellular Immune Responses 8 Months after Ad26.COV2.S Vaccination. N. Engl. J. Med. 2021, 385, 951–953. [Google Scholar] [CrossRef]
  62. World Health Organization. The Janssen Ad26.COV2.S COVID-19 Vaccine: What You Need to Know. 2021. Available online: https://www.who.int/news-room/feature-stories/detail/the-j-j-covid-19-vaccine-what-you-need-to-know (accessed on 4 March 2022).
  63. Grifoni, A.; Weiskopf, D.; Ramirez, S.I.; Mateus, J.; Dan, J.M.; Moderbacher, C.R.; Rawlings, S.A.; Sutherland, A.; Premkumar, L.; Jadi, R.S.; et al. Targets of T Cell Responses to SARS-CoV-2 Coronavirus in Humans with COVID-19 Disease and Unexposed Individuals. Cell 2020, 181, 1489–1501.e15. [Google Scholar] [CrossRef]
  64. National Institute of Health. Adjuvant Developed with NIH Funding Enhances Efficacy of India’s COVID-19 Vaccine. 2021. Available online: https://www.nih.gov/news-events/news-releases/adjuvant-developed-nih-funding-enhances-efficacy-indias-covid-19-vaccine (accessed on 4 March 2022).
  65. Liu, Y.; Rocklöv, J. The reproductive number of the Delta variant of SARS-CoV-2 is far higher compared to the ancestral SARS-CoV-2 virus. J. Travel Med. 2021, 28, taab124. [Google Scholar] [CrossRef]
  66. Levine-Tiefenbrun, M.; Yelin, I.; Alapi, H.; Katz, R.; Herzel, E.; Kuint, J.; Chodick, G.; Gazit, S.; Patalon, T.; Kishony, R.; et al. Viral loads of Delta-variant SARS-CoV-2 breakthrough infections after vaccination and booster with BNT162b2. Nat. Med. 2021, 27, 2108–2110. [Google Scholar] [CrossRef]
  67. Sokal, A.; Barba-Spaeth, G.; Fernández, I.; Broketa, M.; Azzaoui, I.; de La Selle, A.; Vandenberghe, A.; Fourati, S.; Roeser, A.; Meola, A.; et al. mRNA vaccination of naive and COVID-19-recovered individuals elicits potent memory B cells that recognize SARS-CoV-2 variants. Immunity 2021, 54, 2893–2907.e5. [Google Scholar] [CrossRef]
  68. Abdool Karim, S.S.; de Oliveira, T. New SARS-CoV-2 Variants—Clinical, Public Health, and Vaccine Implications. N. Engl. J. Med. 2021, 384, 1866–1868. [Google Scholar] [CrossRef]
  69. Emary, K.R.W.; Golubchik, T.; Aley, P.K. Efficacy of ChAdOx1 nCoV-19 (AZD1222) vaccine against SARS-CoV-2 variant of concern 202012/01 (B.1.1.7): An exploratory analysis of a randomized controlled trial. Lancet 2021, 397, 1351–1362. [Google Scholar] [CrossRef]
  70. Liu, Z.; VanBlargan, L.A.; Bloyet, L.M.; Rothlauf, P.W.; Chen, R.E.; Stumpf, S.; Zhao, H.; Errico, J.M.; Theel, E.S.; Liebeskind, M.J.; et al. Identification of SARS-CoV-2 spike mutations that attenuate monoclonal and serum antibody neutralization. Cell Host Microbe 2021, 29, 477–488.e4. [Google Scholar] [CrossRef] [PubMed]
  71. Jangra, S.; Ye, C.; Rathnasinghe, R.; Stadlbauer, D.; Personalized Virology Initiative Study Group; Krammer, F.; Simon, V.; Martinez-Sobrido, L.; García-Sastre, A.; Schotsaert, M.; et al. SARS-CoV-2 spike E484K mutation reduces antibody neutralisation. Lancet Microbe 2021, 2, e283–e284. [Google Scholar] [CrossRef]
  72. Li, Q.; Nie, J.; Wu, J.; Zhang, L.; Ding, R.; Wang, H.; Zhang, Y.; Li, T.; Liu, S.; Zhang, M.; et al. SARS-CoV-2 501Y.V2 variants lack higher infectivity but do have immune escape. Cell 2021, 184, 2362–2371.e9. [Google Scholar] [CrossRef]
  73. Greaney, A.J.; Starr, T.N.; Barnes, C.O.; Weisblum, Y.; Schmidt, F.; Caskey, M.; Gaebler, C.; Cho, A.; Agudelo, M.; Finkin, S.; et al. Mapping mutations to the SARS-CoV-2 RBD that escape binding by different classes of antibodies. Nat. Commun. 2021, 12, 4196. [Google Scholar] [CrossRef] [PubMed]
  74. Starr, T.N.; Greaney, A.J.; Dingens, A.S.; Bloom, J.D. Complete map of SARS-CoV-2 RBD mutations that escape the monoclonal antibody LY-CoV555 and its cocktail with LY-CoV016. Cell Rep. Med. 2021, 2, 100255. [Google Scholar] [CrossRef] [PubMed]
  75. Baj, A.; Novazzi, F.; Pasciuta, R.; Genoni, A.; Ferrante, F.D.; Valli, M.; Partenope, M.; Tripiciano, R.; Ciserchia, A.; Catanoso, G.; et al. Breakthrough Infections of E484K-Harboring SARS-CoV-2 Delta Variant, Lombardy, Italy. Emerg. Infect. Dis. 2021, 27, 3180–3182. [Google Scholar] [CrossRef]
  76. García, L.F. Immune Response, Inflammation, and the Clinical Spectrum of COVID-19. Front. Immunol. 2020, 11, 1441. [Google Scholar] [CrossRef]
  77. Zani, A.; Caccuri, F.; Messali, S.; Bonfanti, C.; Caruso, A. Serosurvey in BNT162b2 vaccine-elicited neutralizing antibodies against authentic B.1, B.1.1.7, B.1.351, B.1.525 and P.1 SARS-CoV-2 variants. Emerg. Microbes Infect. 2021, 10, 1241–1243. [Google Scholar] [CrossRef]
  78. Edridge, A.W.D.; Kaczorowska, J.; Hoste, A.C.R.; Bakker, M.; Klein, M.; Loens, K.; Jebbink, M.F.; Matser, A.; Kinsella, C.M.; Rueda, P.; et al. Seasonal coronavirus protective immunity is short-lasting. Nat. Med. 2020, 26, 1691–1693. [Google Scholar] [CrossRef]
  79. Becker, M.; Dulovic, A.; Junker, D.; Ruetalo, N.; Kaiser, P.D.; Pinilla, Y.T.; Heinzel, C.; Haering, J.; Traenkle, B.; Wagner, T.R.; et al. Immune response to SARS-CoV-2 variants of concern in vaccinated individuals. Nat. Commun. 2021, 12, 3109. [Google Scholar] [CrossRef] [PubMed]
  80. Wang, Z.; Schmidt, F.; Weisblum, Y.; Muecksch, F.; Barnes, C.O.; Finkin, S.; Schaefer-Babajew, D.; Cipolla, M.; Gaebler, C.; Lieberman, J.A.; et al. mRNA vaccine-elicited antibodies to SARS-CoV-2 and circulating variants. Nature 2021, 592, 616–622. [Google Scholar] [CrossRef] [PubMed]
  81. Hall, V.G.; Ferreira, V.H.; Ierullo, M.; Ku, T.; Marinelli, T.; Majchrzak-Kita, B.; Yousuf, A.; Kulasingam, V.; Humar, A.; Kumar, D.; et al. Humoral and cellular immune response and safety of two-dose SARS-CoV-2 mRNA-1273 vaccine in solid organ transplant recipients. Am. J. Transplant. 2021, 21, 3980–3989. [Google Scholar] [CrossRef] [PubMed]
  82. Holden, I.K.; Bistrup, C.; Nilsson, A.C.; Hansen, J.F.; Abazi, R.; Davidsen, J.R.; Poulsen, M.K.; Lindvig, S.O.; Justesen, U.S.; Johansen, I.S.; et al. Immunogenicity of SARS-CoV-2 mRNA vaccine in solid organ transplant recipients. J. Intern. Med. 2021, 290, 1264–1267. [Google Scholar] [CrossRef] [PubMed]
  83. Chavarot, N.; Morel, A.; Leruez-Ville, M.; Vilain, E.; Divard, G.; Burger, C.; Serris, A.; Sberro-Soussan, R.; Martinez, F.; Amrouche, L.; et al. Weak antibody response to three doses of mRNA vaccine in kidney transplant recipients treated with belatacept. Am. J. Transplant. 2021, 21, 4043–4051. [Google Scholar] [CrossRef]
  84. Shroff, R.T.; Chalasani, P.; Wei, R.; Pennington, D.; Quirk, G.; Schoenle, M.V.; Peyton, K.L.; Uhrlaub, J.L.; Ripperger, T.J.; Jergović, M.; et al. Immune responses to two and three doses of the BNT162b2 mRNA vaccine in adults with solid tumors. Nat. Med. 2021, 27, 2002–2011. [Google Scholar] [CrossRef] [PubMed]
  85. Kamar, N.; Abravanel, F.; Marion, O.; Couat, C.; Izopet, J.; Del Bello, A. Three doses of an mRNA COVID-19 vaccine in solid-organ transplant recipients. N. Engl. J. Med. 2021, 385, 661–662. [Google Scholar] [CrossRef]
  86. Food and Drug Administration. Coronavirus (COVID-19) Update: FDA Authorizes New Long-Acting Monoclonal Antibodies for Pre-exposure Prevention of COVID-19 in Certain Individuals. 2021. Available online: https://www.fda.gov/news-events/press-announcements/coronavirus-covid-19-update-fda-authorizes-new-long-acting-monoclonal-antibodies-pre-exposure (accessed on 4 March 2022).
  87. O’Brien, M.P.; Forleo-Neto, E.; Musser, B.J.; Isa, F.; Chan, K.C.; Sarkar, N.; Bar, K.J.; Barnabas, R.V.; Barouch, D.H.; Cohen, M.S.; et al. Subcutaneous REGEN-COV Antibody Combination to Prevent COVID-19. N. Engl. J. Med. 2021, 385, 1184–1195. [Google Scholar] [CrossRef]
  88. Chen, R.E.; Zhang, X.; Case, J.B.; Winkler, E.S.; Liu, Y.; VanBlargan, L.A.; Liu, J.; Errico, J.M.; Xie, X.; Suryadevara, N.; et al. Resistance of SARS-CoV-2 variants to neutralization by monoclonal and serum-derived polyclonal antibodies. Nat. Med. 2021, 27, 717–726. [Google Scholar] [CrossRef]
  89. Richman, D.D. COVID-19 vaccines: Implementation, limitations, and opportunities. Glob. Health Med. 2021, 3, 1–5. [Google Scholar] [CrossRef]
  90. Nomura, Y.; Sawahata, M.; Nakamura, Y.; Kurihara, M.; Koike, R.; Katsube, O.; Hagiwara, K.; Niho, S.; Masuda, N.; Tanaka, T.; et al. Age and Smoking Predict Antibody Titres at 3 Months after the Second Dose of the BNT162b2 COVID-19 Vaccine. Vaccines 2021, 9, 1042. [Google Scholar] [CrossRef] [PubMed]
  91. De Gier, B.; de Oliveira Bressane Lima, P.; van Gaalen, R.D.; de Boer, P.T.; Alblas, J.; Ruijten, M.; van Gageldonk-Lafeber, A.B.; Waegemaekers, T.; Schreijer, A.; van den Hof, S.; et al. Occupation- and age-associated risk of SARS-CoV-2 test positivity, the Netherlands, June to October 2020. Eurosurveillance 2020, 25, 2001884. [Google Scholar] [CrossRef] [PubMed]
  92. World Health Organization. Prevention, Identification, and Management of Health Worker Infection in the Context of COVID-19. 2020. Available online: https://www.who.int/publications/i/item/10665-336265 (accessed on 4 March 2022).
  93. Chou, R.; Dana, T.; Buckley, D.I.; Selph, S.; Fu, R.; Totten, A.M. Epidemiology of and Risk Factors for Coronavirus Infection in Health Care Workers: A Living Rapid Review. Ann. Intern. Med. 2020, 173, 120. [Google Scholar] [CrossRef] [PubMed]
  94. Goldberg, L.; Levinsky, Y.; Marcus, N.; Hoffer, V.; Gafner, M.; Hadas, S.; Kraus, S.; Mor, M.; Scheuerman, O. SARS-CoV-2 Infection among Health Care Workers Despite the Use of Surgical Masks and Physical Distancing-the Role of Airborne Transmission. Open Forum Infect. Dis. 2021, 8, ofab036. [Google Scholar] [CrossRef]
  95. Teran, R.A.; Walblay, K.A.; Shane, E.L.; Xydis, S.; Gretsch, S.; Gagner, A.; Samala, U.; Choi, H.; Zelinski, C.; Black, S.R.; et al. Postvaccination SARS-CoV-2 infections among skilled nursing facility residents and staff members—Chicago, Illinois, December 2020–March 2021. Am. J. Transplant. 2021, 21, 2290–2297. [Google Scholar] [CrossRef]
  96. Nordström, P.; Ballin, M.; Nordström, A. Effectiveness of heterologous ChAdOx1 nCoV-19 and mRNA prime-boost vaccination against symptomatic COVID-19 infection in Sweden: A nationwide cohort study. Lancet Reg. Health Eur. 2021, 11, 100249. [Google Scholar] [CrossRef]
  97. Gram, M.A.; Nielsen, J.; Schelde, A.B.; Nielsen, K.F.; Moustsen-Helms, I.R.; Sørensen, A.K.B.; Valentiner-Branth, P.; Emborg, H.D. Vaccine effectiveness against SARS-CoV-2 infection, hospitalization, and death when combining a first dose ChAdOx1 vaccine with a subsequent mRNA vaccine in Denmark: A nationwide population-based cohort study. PLoS Med. 2021, 18, e1003874. [Google Scholar] [CrossRef]
  98. Bignardi, E.; Brogna, C.; Capasso, C.; Brogna, B. A fatal case of COVID-19 breakthrough infection due to the delta variant. Clin. Case Rep. 2022, 10, e05232. [Google Scholar] [CrossRef]
Figure 1. Study selection and screening following the PRISMA 2020 guidelines.
Figure 1. Study selection and screening following the PRISMA 2020 guidelines.
Tropicalmed 07 00081 g001
Table 1. Included studies and patient characteristics of reinfection in vaccinated individuals.
Table 1. Included studies and patient characteristics of reinfection in vaccinated individuals.
StudyType of StudyNumber of Fully Vaccinated Individuals
(Breakthrough Infections)
CountryGenderAge (Years)Number of Days since VaccinationVaccine ReceivedSymptomsComorbiditiesVariants
[Reported Mutations]
Ct (Cycle Threshold)
Value
Complications & Outcome
Bergwerk et al., 2021 [11]Case control11,453 (39)IsraelFemales: 25; Males:14Mean: 42Median: 39 (Range: 11–102)BNT162b2
(Pfizer-BioNTech)
Upper respiratory congestion (36%), myalgia (28%), loss of smell or taste(28%); fever or rigors (21%);
Asymptomatic (33%)
Immunosuppressed (1), CLL*(1), ITP*(1),
metabolic syndrome (6), thyroid disorder (3), other (migraines, fibromyalgia, osteoporosis, PCOS*) (4)
Alpha (B.1.1.7): 85% of samples<30 (74%); >30 (26%)Recovery
Estofolete et al., 2021 [12]Case Report2 (2)BrazilMale60106Corona Vac
(Sinovac)
Anosmia, malaise, myalgia, dyspneaType 2 diabetes mellitus, hypertension, obesity degree I (BMI*: 32.3 kg/m2)Gamma (P.1)
[K417T, E4844K, N501Y]
UnknownHospitalization with supplemental oxygen → Recovery
55122Sore throat, headache, malaise, chills, coryza, sneezing, dyspnea, hypoxiaNone
Fabiani et al., 2021 [13]Case Report1 (1)ItalyMale8323BNT162b2(Pfizer-BioNTech)Slight headache, mild coldNoneGamma (P.1)
[K417T, E484K, N501Y, D614G]
13Recovery
Philomina et al., 2021 [14]Retrospective cohort6 (6)IndiaFemale2535AZD1222/Covishield (SII)Influenza-like illnessUnknownB.1.1.306
[E484K]
16.45Recovery
Male5030Fever, malaise, anosmia, headacheAlpha (B.1.1.7)
[N501Y]
20
Female5328Rhinitis21
2526Fever, loose stools, abdominal pain, dry cough, myalgia, rhinitis, anosmia24
3225Mild nasal congestion, headache26
3317Loss of smell, loose stools, rhinitisB.1.1
B.1.560
[S477N]
14
Hacisuleyman et al., 2021 [15]Prospective cohort417 (2)USAFemale5119mRNA-1273
(Moderna)
Sore throat, congestion, headache, anosmiaNoneAlpha (B.1.1.7)
[E484K
D614G
T95I, del142–144]
24.2Recovery
6536BNT162b2 (Pfizer–BioNTech)Fatigue, sinus congestion, headacheAlpha (B.1.1.7)
[S477N
T95I, del142–144
R190T
F2201
R237K
R246T
D614G]
33.3
Kroidl et al., 2021 [16]Case report1 (1)GermanyUnknownEarly ‘60s26BNT162b2 (Pfizer–BioNTech)Headache, congested noseNoneBeta (B.1.351)UnknownRecovery
Almaghrabi et al., 2022 [17]Case series4 (4)Saudi ArabiaMale6873BNT162b2
(Pfizer-BioNTech)
Fever, chills, vomitingLiver transplant, diabetes mellitus,
hypertension, immunosuppressive medication
Alpha (B.1.1.7)
[E484K]
25Severe pneumonia →Mechanical ventilation →Death
69150Shortness of breath, hypoxiaRenal transplant, diabetes mellitus,
hypertension, immunosuppressive medication
Alpha (B.1.1.7)30Pneumonia → Mechanical ventilation, septic shock →Death
4139Mild coughs, shortness of breathRenal transplant, immunosuppressive medicationsBeta (B.1.351)29ICU* admission with HFNC*→ Recovery
Female4821ChAdOx1 nCoV-19 vaccine (AstraZeneca)Fever, hypoxiaRenal transplant, post-transplant lymphoma, immunosuppressive medicationsDelta (B.1.617.2)19Hospital acquired infection, HFNC*→ Recovery
Baj et al., 2021 [18]Retrospective cohort Study4 (4)ItalyFemale8077mRNA-1273 (Moderna)Fatigue, headache, myalgia, dyspneaUnknownDelta (B.1.617.2)[E484K]22Recovery
Male7767Pfizer-BioNTech (BNT162b2)Fever19
Female8387BNT162b2
(Pfizer-BioNTech)
Fever, fatigue, ageusia, anosmia18
Female8145mRNA-1273 (Moderna)Dyspnea, fever, myalgia, fatigue21Hospital admission → Recovery
Bignardi et al., 2022 [19]Case report1 (1)ItalyMale61120mRNA vaccine -type not specifiedDyspnea, cough, feverHypertension, obesityDelta (B.1.617.2)UnknownPneumonia → Death
Chau et al., 2021 [20]Cohort 866 (62) aVietnamFemales:
29 Males:33
Median: 41.5 (IQR: 32–50)49–56 (97%)Oxford-AstraZeneca Fever (27%), cough (37%), sore throat (34%), runny nose (36%), loss of smell (39%), loss of taste (8%), muscle pain (27%), headache (19%), chest pain (3%), nausea (8%), shortness of breath (4%), pneumonia (5%), asymptomatic (21%)Overweight (6), obesity (3), hypertension (3), hepatitis B (3), diabetes mellitus (2), pregnancy (1)Delta (B.1.617.2)31.9 (IQR: 23.3–34.9)Recovery
Connor et al., 2021 [21]Case report2 (2)USAMale6360BNT162b2 (Pfizer-BioNTech)Nasal congestion, headache, dry coughHypertension, benign prostatic hypertrophy, overweightB.1.617.2
B.1.619
[35 mutations detected, including 9 in the spike protein]
31.3Recovery
2590Upper respiratory symptoms, headachesNoneB.1.617.2
[7/12 shared S-gene mutations]
25.2Recovery
Gharpure et al., 2022 [22]Cohort1128 (918)USAFemales: 90
Males: 822
19–49 (66%), 50–64 (30%), 65–74 (4%), >75 (0.4%)>14BNT162b2
(Pfizer-BioNTech): 504
mRNA-1273 (Moderna): 293
Johnson & Johnson: 121
Abdominal pain (6%), chills (35%),congestion (58%),cough (73%), diarrhea(20%), shortness of breath (10%), fatigue (41%), fever (43%), headache (47%), loss of appetite (16%), loss of smell or taste (50%), muscle pain (39%), sore throat (42%), vomiting (3%)Active cancer (3), autoimmune disease (11), cardiovascular disease (36), chronic kidney disease (3), chronic lung disease (22), pregnancy (3), diabetes mellitus (21), HIV* infection (6), solid organ transplant (1), other immunosuppressive conditions (41)Delta (B.1.617.2): 98%Delta (AY.3 sublineage:0.3%,
Delta (AY.4 sublineage): 0.8%
Gamma (P.1): 0.8%
UnknownHospitalized (7), ICU* (2) → Recovery
Galan Huerta et al., 2022 [23]Case-control 53 (53)MexicoFemales: 28; Males: 25Mean: 59.7 (50–70)7AstraZeneca/Oxford: 8 (15%)
BNT162b2 (Pfizer/BioNTech): 8 (15%)
Convidecia(CanSino): 24 (45%)
CoronoVac (Sinovac): 10 (19%)
Unspecified: 3
Mostly mild or asymptomaticHospitalized: hypertension (11), Type 2 diabetes mellitus (13), obesity (1), smoking (4); Ambulatory:
hypertension (5), Type 2 diabetes mellitus (5), obesity (2), smoking (1)
Delta (B.1.617.2) (AY.1, AY.2, AY.3, AY.4 lineage): 67.92%
Gamma (P.1, P.1.1, P.1.2): 7.55%
Mu (B.1.621): 7.55%
Alpha (B.1.1.7): 5.66%
Hospitalized: 19.58 (17.19–22.49); Ambulatory: 18.81 (15.72–21.24)Hospitalized:
High-flow O2 (14), intubation (10), ICU* admission (1), death (4); Ambulatory: all recovered (30)
Deng et al., 2021 [24]Case control 14 (14)USAFemale60Range: (14–109)BNT162b2
(Pfizer-BioNTech)
RhinorrheaNoneAlpha 20I/S: 501Y.V118.8Recovery
Male58Chill, subjective feverNoneAlpha
(20I/S: 501Y.V1)
19.1
Female48Weakness, congestion loss of taste/smell, fatigueSmokerAlpha
(20I/S: 501Y.V1)
20.9
51mRNA vaccine type not providedHeadache, cough, rhinorrhea ageusia, anosmiaImmunosuppressive medication, non-alcoholic steatohepatitisGamma
(20J/S: 501Y.V3)
17.1
37mRNA-1273 (Moderna)AsymptomaticNone20G19.5
50BNT162b2
(Pfizer-BioNTech)
AsymptomaticNoneUnknown34.2
81Johnson & JohnsonShortness of breath, coughHeart disease, cerebrovascular diseaseAlpha
(20I/S: 501Y.V1)
18.8Hospitalization → recovery
Male65BNT162b2
(Pfizer-BioNTech)
Diarrhea, myalgia, chills, feverImmunosuppressive medication, kidney, and heart transplantAlpha
(20I/S: 501Y.V1)
20.1Pneumonia → recovery
55Cough, acute hypoxic respiratory failure, sepsisImmunosuppressive medication, kidney transplantAlpha
(20I/S: 501Y.V1)
22.3Intensive Care Unit (ICU) → Death
70Cough, weakness, fever, dyspneaImmunosuppressive medication, liver transplantGamma
(20J/S: 501Y.V3)
19.6Hospitalization → recovery
68mRNA-1273 (Moderna)Acute hypoxia, acute pneumonia, hemoptysis Immunosuppressive medication, lung transplantGamma
(20J/S: 501Y.V3)
21.4
Female60Shortness of breath, fever, chills, body aches, hypoxiaImmunosuppressive medication, lung transplantGamma
(20J/S: 501Y.V3)
15.7Intensive Care Unit (ICU) → Recovery
Male65BNT162b2
(Pfizer-BioNTech)
Diarrhea, nausea, weakness cough, dyspneaImmunosuppressive medication, liver transplantEpsilon
(CAL.20C)
22.1Hospitalization → Recovery
Female76Fever, chills, acute respiratory failure None20G18.3Intensive Care Unit (ICU) → Recovery
De Souza et al., 2021 [25]Case control 42 (22)BrazilFemales: 17
Males: 5
77 (IQR: 51–87)5–27CoronaVac (SinoVac)Asymptomatic (75%)
Mild COVID-19 symptoms (25%)
UnknownAlpha (B.1.1.7)UnknownDeath: 1%
Recovery: 99%
Gupta et al., 2021 [26]Case control 592 (592)IndiaFemales: 207; Males: 385Mean 44 (31–56)39 (19–58)Covaxin: 71 (10.5%)Covishield
(AstraZeneca): 604 (89.2%)
Covilo (Sinopharm): 2 (0.3%)
Symptomatic (71%) with one or more symptoms, fever (69%), body ache, headache and nausea (56%), cough (45%), sore throat (37%), loss of smell and taste (22%), diarrhea (6%), breathlessness (6%), ocular irritation, redness (1%);
Asymptomatic (29%)
Type 2 diabetes mellitus, hypertension, obesity, chronic cardiac, renal, and pulmonary diseases Delta (B.1.617.2): 384 Alpha (B.1.1.7): 28)
Kappa (B.1.617.1: 22
B.1.617.3: 2
B.1.36: 2
B.1.1.294: 1
B.1.36.16: 1
B.1.1.306: 1
Delta (AY.2): 2
<30Fully vaccinated: hospitalized (53), Recovered (589), Death (3)
Kale et al., 2021 [27]Cohort 1639 (156)IndiaFemale: 86 Males: 70Median: 34 (IQR21–67)>14ChAdOx1 nCoV-19/Covishield (SII)Fever, muscle achesUnknownDelta (B.1.617.2): 32
Kappa (B.1.617.1):11
Alpha (B.1.1.7): 1
23.2 (IQR 0.0–33.1) Recovery; Hospitalization (0.22%)
Schulte et al., 2021 [28]Case report1 (1)GermanyMale4249BNT162b2
(Pfizer-BioNTech)
AsymptomaticNoneB.1.5259.44Recovery
Malhotra et al., 2022 [29]Retrospective cohort 1079 (17)IndiaUnknown<25:72 (6.6%); 25–44: 660 (60.6%);≥ 45: 357 (32.8%)>15BBV152/Covaxin (Bharat Biotech)Symptomatic:-Fever, rhinorrhea, sore throat, cough, chest pain, wheezing, difficulty breathing, shortness of breath, anosmia, dysgeusia, fatigue, myalgia, headache, abdominal pain, nausea, diarrhea.
Asymptomatic: 3
Hypertension; chronic heart, lung, or kidney disease; cancer; hypothyroidismGamma (B.1.617.2)UnknownRecovery
Shastri et al., 2021 [30]Case report1(1) bIndiaFemale6128ChAdOx1 nCoV-19(Covishield)1st infection episode: Abdominal pain, fever, myalgia, fatigue
2nd infection episode: Body ache, fatigue, headache, cough, breathlessness, fever, rhinorrhea, vomiting
Prediabetes, bronchial asthma, hypertensionAlpha (B.1.1.7):1st
Delta (B.1.617.2):2nd
1st infection: 35.2
2nd infection: 20.4
Recovery
Rovida et al., 2021 [31]Cohort3702 (33)ItalyFemales:26
Males: 7
Unknown 47 (Range 7–90)BNT162b2
(Pfizer-BioNTech)
Asymptomatic (48%), fever (6%), asthenia (6%), headache (6%), arthralgia (9%), pharyngodynia (3%), rhinitis (27%), cough (9%), cough (9%), anosmia (9%), ageusia (3%), nausea (9%), diarrhea (10%)UnknownAlpha
(B.1.1.7)
UnknownRecovery
Rumke et al., 2022 [32]Cohort14 (14)NetherlandsFemale4543BNT162b2
(Pfizer-BioNTech)
Anosmia, arthralgia, fever, headache, myalgia, peripheral neuropathy, rhinosinusitisNoneAlpha (B.1.1.7)18.5Recovery
6278Anosmia, rhinosinusitisNoneAlpha (B.1.1.7) 23.7
2764RhinitisNoneAlpha (B.1.1.7)
[A771V]
23.5
5261Cough, dyspnea, feverAsthmaAlpha (B.1.1.7)19.6
3574Anosmia, cough, rhinitisNoneAlpha (B.1.1.7)
[H245Y]
21.5
3580Anosmia, rhinosinusitisNoneAlpha (B.1.1.7)
[S494P]
24.7
Male5880AsymptomaticNoneAlpha (B.1.1.7)31.9
Female26111Fever, rhinitisNoneAlpha (B.1.1.7)
[V382L]
19.8
3838Ad26.COV2.S (Johnson & Johnson)Cough, fever, pharyngitis, rhinosinusitisNoneAlpha (B.1.1.7)
[D88V]
18.9
5737Cough, dyspneaNoneAlpha (B.1.1.7)
[V483I, A706V]
21.5
5020AsymptomaticNoneAlpha (B.1.1.7)
[S12F, D905N]
23.6
5445Cough, fever, headache, myalgia, otitisAtopic dermatitisDelta (B.617.2)31.3
3818AsymptomaticHashimoto thyroiditisAlpha (B.1.1.7)29.0
4852Anosmia, fever, headache, myalgia, sinusitisNoneDelta (B.617.2)
[G142D]
29.2
Yi et al., 2022 [33]Cohort24 (24)South Korea Females:1878.9 (Range 34–99)Mean: 40
(Range, 80–117)
BNT162b2
(Pfizer-BioNTech)
Asymptomatic (48%)
Symptomatic (48%)
UnknownDelta (B.1.617.2):1318.1 (symptomatic); 20 (asymptomatic)Recovery (96%), Death (4%)
Robilotti et al., 202 [34]Cohort12,046
(80: pre-Delta)
USAFemales: 60
Males: 20
Median37 (Range:22–65)Median: 56 (Range:1–100)BNT162b2
(Pfizer-BioNTech): 91%
mRNA-1273 (Moderna): 9%
Asymptomatic (20%)
Headache (55%)
Fatigue (45%)
Body aches (28%)
Fever (including subjective) (19%)
Loss of smell/taste (28%)
Chills (20%)
Sore throat (21%)
Rhinorrhea, nasal congestion, sneezing (53%)
GI symptoms (nausea, vomiting, diarrhea or abdominal pain) (18%)
Cough (31%)
Shortness of breath (8%)
UnknownAlpha (B.1.1.7)
[E484K
K417T/N
S477N
N501Y]
UnknownRecovery
(179: post-Delta)Females: 127
Males: 52
Median 33 (Range 21–63)Median: 185 (Range 8–235)BNT162b2
(Pfizer-BioNTech): 79%
mRNA-1273
(Moderna): 21%
Asymptomatic (8%)
Headache (45%)
Fatigue (55%)
Body aches (37%)
Fever (including subjective) (32%)
Loss of smell/taste (28%)
Chills (27%)
Sore throat (44%)
Rhinorrhea, nasal congestion, sneezing (52%)
GI symptoms (nausea, vomiting, diarrhea or abdominal pain) (17%)
Cough (53%)
Shortness of breath (9%)
UnknownDelta (B.1.617.2)
[L452R
T478K
E484Q]
UnknownRecovery
Vignier et al., 2021 [35]Cohort 25 (15)French GuianaMales: 15Median: 53.3>14BNT162b2
(Pfizer-BioNTech): 56.8%
Symptomatic: Fever, dyspnea (87%)Hypertension, diabetes mellitus, obesity, cardiac insufficiencyGamma (P.1) 18–35Recovery
Tober-lau et al., 2021 [36]Longitudinal 20 (16)GermanyFemales: 12
Males: 4
>65 years4–5BNT162b2
(Pfizer-BioNTech)
Asymptomatic mostly.
Diarrhea, fatigue,
cough or shortness of breath (31.25%)
Hypertension,
Type 2 diabetes mellitus,
chronic kidney disease
dementia
Alpha (B.1.1.7)UnknownHospitalization (31.25%)
Supplemental oxygen (6.3%)
Death (12.5%)
Servellita et al., 2022 [37]Cohort1373
(125) c
USAFemales: 68
Males: 57
Mean: 49 (Range 22–97)Median: 73.5 (range 15–140)BNT162b2
(Pfizer-BioNTech): 51%,
mRNA-1273
(Moderna): 31%
Johnson & Johnson: 10%
Asymptomatic (26%)
COVID-19 pneumonia (15.4%)
Immunocompromised (23%)Delta (B.1.617.2:31%, Alpha (B.1.1.7): 18.3%, Gamma (P.1): 15.6%,
Iota (B.1.526): 11.9%, Epsilon (B.1.427/B.1.429): 6.4%,
Beta (B.1.351): 3.7%, Other: 12.8%
[L452R/Q, E484K/Q and/or F490S]
23.1Recovery (100%) ICU (2.6%), Hospitalizations (15.4%)
Singer et al., 2021 [38]Prospective cohort343 (31)IsraelFemales: 17
Males: 14
Median: 58 (21–87)>7BNT162b2
(Pfizer-BioNTech)
Asymptomatic (05%)UnknownBeta (B.1.351)UnknownRecovery
Thangaraj et al., 2022 [39]Prospective cohort113 (113)IndiaFemales: 44
Males:66 Others:3
Median:54 (42–64)>14Covaxin: 27.4%
Covishield: 70.8%
Unknown: 1.8%
Symptomatic (88.5%)Unspecified comorbidities (46%)Delta (B.1.617.2):74.3% B.1.617.1: 0.9%
AY.1: 0.9%
Alpha (B.1.1.7): 0.9%
Beta (B.1.351): 0.9%
<30Recovery
Olsen et al., 2021 [40]Cohort12,476
(207)
USAFemales: 53% Males: 47% dMedian: 52.5 d>14BNT162b2
(Pfizer-BioNTech): 87%
mRNA-1273
(Moderna): 13%
UnknownBMI > 30 (42.7%)Alpha (B.1.1.7): 126; Gamma (P.1): 5 Epsilon (B.1.429): 3
B.1526: 1 B.1526.1:1
Eta (B.1.525): 1 non-VOC: 70
23.9Hospitalization (34.8%)
Singh et al., 2022 [41]Cohort63 (36)IndiaFemales: 13
Male: 23
Median: 37 (21–92)UnknownAZD1222/Covishield (SII): 15.87%
BBV152/Covaxin: 84.13%
High-grade unremitting fever, shortness of breath, headacheNoneDelta (B.1.617.2): 63.9%
B.1.617.1: 11.1%
Alpha (B.1.1.7) 2.8%
Range: 11.3–31Recovery
Tay et al., 2022 [42]Prospective case-control55 (55)SingaporeFemales: 19
Males: 36
Median 46 (IQR 36.5–59.5)82 (IQR 51.5–99)BNT162b2
(Pfizer-BioNTech)
Asymptomatic (21.8%)
Mild symptoms (78.2%)
Chronic venous, asthma, other chronic lung diseases, rheumatologic disease, chronic liver disease, diabetes mellitus, chronic kidney disease, malignancies, or HIV (6)Delta (B.1.617.2): 87.3%
Unknown: 7.3%
Non-Delta:5.5%
UnknownRecovery
Sun et al., 2021 [43]Retrospective cohort604,035 (22,917)USAFemales: 13,040
Males: 9877
Median: 51 (IQR 34–66)138 (85–178)BNT162b2
(Pfizer-BioNTech)
mRNA-1273 (Moderna)
UnknownImmunocompromised (1451).Delta (B.1.617.2)UnknownRecovery (93.5%); Hospitalization: 11.5%
Severe outcomes (0.65%)
* Abbreviations: CLL—Chronic Lung Disease; ITP—Idiopathic thrombocytopenic purpura; PCOS—Polycystic ovarian syndrome; BMI—Body mass index; HIV: Human Immunodeficiency Virus; ICU: Intensive Care Unit; HFNC: High flow nasal cannula. a Only 62 participants included in the study; b Patient had 2 breakthrough infections; c Variant breakdown provided for 109 patients; d Patient data corresponds to total number of patients.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Gopinath, S.; Ishak, A.; Dhawan, N.; Poudel, S.; Shrestha, P.S.; Singh, P.; Xie, E.; Tahir, P.; Marzaban, S.; Michel, J.; et al. Characteristics of COVID-19 Breakthrough Infections among Vaccinated Individuals and Associated Risk Factors: A Systematic Review. Trop. Med. Infect. Dis. 2022, 7, 81. https://doi.org/10.3390/tropicalmed7050081

AMA Style

Gopinath S, Ishak A, Dhawan N, Poudel S, Shrestha PS, Singh P, Xie E, Tahir P, Marzaban S, Michel J, et al. Characteristics of COVID-19 Breakthrough Infections among Vaccinated Individuals and Associated Risk Factors: A Systematic Review. Tropical Medicine and Infectious Disease. 2022; 7(5):81. https://doi.org/10.3390/tropicalmed7050081

Chicago/Turabian Style

Gopinath, Shilpa, Angela Ishak, Naveen Dhawan, Sujan Poudel, Prakriti Singh Shrestha, Prabhjeet Singh, Emily Xie, Peggy Tahir, Sima Marzaban, Jack Michel, and et al. 2022. "Characteristics of COVID-19 Breakthrough Infections among Vaccinated Individuals and Associated Risk Factors: A Systematic Review" Tropical Medicine and Infectious Disease 7, no. 5: 81. https://doi.org/10.3390/tropicalmed7050081

Article Metrics

Back to TopTop