3,841
Views
2
CrossRef citations to date
0
Altmetric
Review

Effectiveness of vaccination against SARS-CoV-2 and the need for alternative preventative approaches in immunocompromised individuals: a narrative review of systematic reviews

, , , , , & show all
Pages 341-365 | Received 06 Feb 2023, Accepted 13 Mar 2023, Published online: 10 Apr 2023

ABSTRACT

Introduction

Vaccination against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), including administration of booster doses, continues to be the most effective method for controlling COVID-19-related complications including progression to severe illness and death. However, there is mounting evidence that more needs to be done to protect individuals with compromised immune function.

Areas covered

Here, we review the effectiveness of COVID-19 vaccination in immunocompromised patients, including those with primary immunodeficiencies, HIV, cancer (including hematological malignancies), solid organ transplant recipients, and chronic kidney disease, as reported in systematic reviews/meta-analyses published over a 12-month period in PubMed. Given the varied responses to vaccination in patients with compromised immune function, a major goal of this analysis was to try to identify specific risk-factors related to vaccine failure.

Expert opinion

COVID-19 remains a global problem, with new variants of concern emerging at regular intervals. There is an ongoing need for optimal vaccine strategies to combat the pandemic. In addition, alternative treatment approaches are needed for immunocompromised patients who may not mount an adequate immune response to current COVID-19 vaccines. Identification of high-risk patients and the introduction of newer antiviral approaches such as monoclonal antibodies will offer physicians therapeutic options for such vulnerable individuals.

1. Introduction

The coronavirus disease 2019 (COVID-19) pandemic is a result of the rapid and almost unrestricted spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) worldwide [Citation1]. This is highlighted by statistics presented by the World Health Organization (WHO), which recorded cumulative case numbers of 84.9 million (2020), 290.8 million (2021) and 733.3 million (2022), although this most recent case count is likely an underestimate of the true count by at least an order of magnitude [Citation2]. The high morbidity and mortality from COVID-19 have resulted in large-scale economic disruption and an unprecedented focus on measures to reduce its impact, with vaccination at the forefront.

The benefits that can be attributed to medical care (especially vaccination) over the last 2 years can be ascertained from a review of the numbers included on the WHO COVID-19 Dashboard. While the number of cases has risen almost 10-fold from 2020–2022, over the time period, the total number of deaths has risen to a much lower extent (3.4-fold; from 1.97 to 6.70 million). This roughly equates to one death in 43 cases in 2020 compared with less than one death in 350 cases up to 2022 [Citation2]. These numbers can be considered, at best, as rough estimates, since they only relate to confirmed cases. However, the magnitude of the improved death/case rate over the 2-year period endorses the positive impact of the global medical care response to COVID-19, including ready access to vaccines/other treatments/medical care facilities and availability of appropriate medical care.

Immunocompromised persons represent approximately 3% of the adult population [Citation3]. They are of particular concern as this vulnerable group is both at increased risk of severe COVID-19 and is less well protected by vaccination [Citation4]. This increased risk may be attributable to the underlying disease itself or as an inevitable consequence of treatment with immunosuppressant medicines. Patients with active cancer, HIV, transplant recipients requiring prolonged immunosuppression, and individuals requiring immunosuppressive treatment for autoimmune and inflammatory rheumatic diseases have been reported to be at increased risk of severe COVID-19 and poor outcomes/death [Citation5–9].

A number of vaccines for SARS-CoV-2 have been approved at unprecedented speed. This includes mRNA vaccines such as BNT162b2 (Pfizer-BioNTech) and mRNA1273 (Moderna), viral vector vaccines such as AZD1222 [ChAdOx1 nCoV-19] (Oxford-AstraZeneca) and Ad26.COV2.S (Janssen/Johnson & Johnson), and traditional inactivated whole virus vaccines such as CoronaVac (Sinovac Biotech) [Citation10]. The number of vaccine doses needed to complete the primary immunization series was usually two, except the Janssen/Johnson and Johnson vaccine (Ad26.COV2.S), which required only one dose. High seroconversion and vaccine efficacy rates against SARSCoV-2 infection have been reported regardless of the type of vaccine used or previous infection status [Citation11–14]. Despite this, vaccine trials generally excluded immunocompromised populations, resulting in a paucity of data on the efficacy and safety of vaccination in these groups that have an increased risk of developing severe COVID-19 (including intensive care unit (ICU) admission and death) [Citation15]. Thus, there is a need for more information in these high-risk patients with respect to vaccine effectiveness and outcomes, and also alternative treatment approaches such as immunotherapies to treat the disease [Citation16] and f monoclonal antibodies for pre-exposure prophylaxis of COVID-19 [Citation17].

In this article, we will review the latest evidence related to COVID-19 vaccination in immunocompromised individuals, with an emphasis on response rates in some key patient groups that have been shown to be at increased risk. This includes:

  • Persons with primary or secondary immunodeficiencies

  • Cancer patients, especially those with hematological malignancies

  • Solid organ transplant recipients

  • Patients with chronic kidney disease.

Within these difficult-to-treat populations, the risk of severe COVID-19 disease is increased, and identification of such patients would make them potential candidates for prophylaxis with monoclonal antibodies such as the approved combination tixagevimab + cilgavimab. Thus, one of the goals of this review will be to try and identify specific risk-factors in the identified immunocompromised patient cohorts.

2. Search strategy

The primary search for this review was completed by 21 October 2022 in PubMed and included the terms COVID-19 and vaccination and was restricted to all systematic reviews published in the preceding 12 months involving any COVID-19 vaccination. This strategy identified 530 records and the titles/abstracts were screened for suitability to identify all articles involving vaccination strategies against SARS-CoV-2 virus in immunocompromised persons.

Inclusion criteria for selection of systematic reviews used the ‘Population, Intervention, Comparison, Outcomes, and Time’ (PICOT) format as a framework [Citation18]. Study populations included adults with immunocompromised disease (and control groups in some studies). Interventions were any COVID-19 vaccine. Comparators were not often employed, but included healthy controls and ‘other disease groups’ on occasion. Outcomes focused on vaccine effectiveness against death, hospitalization, and development of more serious disease as reported. Time was the preceding 12 months so as to make it as current as possible.

This was augmented by searches involving the specific indications: immunodeficiency (primary and secondary), HIV, cancer, solid cancers, hematological malignancies, solid organ transplantation, chronic kidney disease, and dialysis. General reviews on the topic COVID-19 in immunocompromised persons with/without vaccination were also searched to identify the most recent concepts regarding treatment in this high-risk population and to identify any missing systematic reviews/meta-analyses. Based on these searches 41 suitable systematic reviews were found and formed the basis of this article.

3. COVID-19 in immunocompromised populations – General

From the outset of the COVID-19 pandemic, it was clear that certain subgroups such as immunocompromised individuals were at increased risk of severe disease, ICU admission, and potentially death [Citation15]. At-risk groups were identified in an analysis of the OpenSAFELY health analytics platform involving 17,278,392 primary care records for adult NHS patients in England prior to the availability of vaccines [Citation19]. A total of 10,926 COVID-19-related deaths were recorded, and the highest risk factors (Hazard Ratios [95%CI]) adjusted for age and sex) were as follows: advanced age (80+ years 38.29 [35.02–41.87]; 70–79 years 8.62 [7.84–9.46]; and 60–69 years 2.79 [2.52–3.10]), organ transplant (6.00 [4.73–7.61]), eGFR <30 ml min−1 per 1.73 m2 (3.48 [3.23–3.75]), other neurological disease (3.08 [2.85–3.33]), hematological malignancy within the last year (3.02 [2.24–4.08]) or 1–4.9 years (2.56 [2.14–3.06]), and other immunosuppressive diagnoses (2.75 [2.10–3.63]). A cluster of metabolic and related disorders were also shown to be potential risk factors for severe COVID-19 in unvaccinated subjects including obesity class III (2.66 [2.39–2.95]), diabetes [(HbA1c ≥58 mmol mol−1) (2.61 [2.46–2.77]) or no recent HbA1c measurement (2.27 [2.06–2.50])], stroke or dementia (2.57 [2.46–2.70]), and liver disease (2.39 [2.06–2.77]).

Multiple studies/systematic reviews using multivariate adjustment have highlighted the potential role of metabolic syndrome, hyperglycemia, insulin resistance, and obesity, as well as cardiovascular disorders, as risk factors for severe COVID-19 (morbidity and mortality) [Citation19–28]. Importantly, in a registry study involving 6457 consecutive patients, obesity, diabetes, and hypertension had an additive effect on COVID-19-related mortality, particularly in young and middle-aged patients, and the risk of COVID-19-related mortality in these younger patients was similar to that of older, but metabolically healthy patients [Citation29].

The rapid introduction of COVID-19 vaccination programs has had a marked impact on the dynamics of the COVID-19 pandemic, reducing both the morbidity and mortality associated with the disease. However, some groups of patients such as immunocompromised patients were excluded from vaccine trials, leading to a paucity of data on the efficacy and safety of vaccines in these at-risk groups [Citation14,Citation16]. This situation is being rectified, and two recent systematic reviews are summarized in . In the first, COVID-19 vaccines (primarily the mRNA vaccines from Pfizer/BioNTech and Moderna) decreased symptomatic COVID-19 infection with a vaccine effectiveness of 70.4% in immunocompromised patients [Citation30]. This number was lower compared to the vaccine effectiveness in the general population (94.1%) [Citation12]. A wide range of anti-SARS-CoV-2 spike protein IgG levels was reported after two doses of COVID-19 vaccines in immunocompromised patients, and the rate of response was significantly lower compared to the control group [Citation30]. Similar findings were reported by Lee and colleagues in a systematic review/meta-analysis involving 82 studies (Citation14; . Again, seroconversion rates and antibody titers after COVID-19 vaccines were significantly lower in immunocompromised patients than in controls, with solid organ transplant recipients having the lowest rates of seroconversion and patients with solid cancers the highest. Notably, immunocompromised patients generally had lower antibody titers associated with seroconversion than immunocompetent controls [Citation14]. The authors noted that antibody titers are an imperfect surrogate marker for vaccine effectiveness and, although neutralizing antibody levels are becoming more widely used, the search for a reliable correlate of protection continues [Citation14]. Given the varied responses to COVID-19 vaccination in different groups of immunocompromised patients, the following sections will cover common diseases associated with compromised immune function to try to identify specific risk-factors related to vaccine failure.

Table 1. Summary of systematic reviews published in 2022 investigating immune responses to COVID-19 vaccination in immunocompromised patients (general), and those with primary immunodeficiency (PI) or human immunodeficiency virus (HIV) infection.

4. The impact of COVID-19 vaccination in specific immunocompromised patient groups

4.1. Patients with primary immunodeficiencies

A number of individual studies have reported higher morbidity and mortality in primary immunodeficiency (PI) patients with COVID-19 compared with the general population [Citation31,Citation32]. However, there is no universal agreement on this point, as was noted in a systematic review of 22 studies involving 109,326 patients with PIs or secondary immunodeficiencies such as HIV [Citation33]. Firstly, the common view is that immunodeficient patients are at a higher risk of infection leading to a higher mortality rate. It has also been speculated that immunodeficiency might have a protective role and lower mortality rates by reducing the severity of the inflammatory response via inhibition of the cytokine storm and its consequences. It is important to note, however, that comorbidities, such as diabetes, hypertension, coronary artery disease, kidney disease, and history of lower respiratory tract infections, as well as demographic characteristics (obesity and age >70 years) might also play a role [Citation33].

Regarding the impact of COVID-19 vaccination in patients with PI, information is limited at the present time. In two studies included in a systematic review of 68 studies and 459 patients with PI, 2 doses of mRNA vaccines were found to produce an immune response in PI patients with common variable immunodeficiency (CVID; ) [Citation34]. As expected, persons with X-linked agammaglobulinemia produced few or no antibodies following vaccination. Even in patients exhibiting an immune response, the concentration of neutralizing antibodies tended to be suboptimal [Citation34].

4.2. People with human immunodeficiency virus

Systematic reviews in people living with human immunodeficiency virus (PLWH) and vaccinated against COVID-19 are summarized in [Citation35,Citation36]. The two reviews reported favorable immunogenicity and efficacy for COVID-19 vaccines in PLWH after two doses of vaccine, and seroconversion rates were only slightly lower than those reported for healthy (immunocompetent) controls.

4.3. Patients with cancer

Unvaccinated cancer patients infected with SARS-CoV-2 have been reported to have a higher risk of COVID-19-related complications including severe/critical disease, ICU admission, and a worse prognosis (including death), and this is best evidenced in patients with hematological malignancies [Citation19,Citation37]. The increased risk in cancer patients has been attributed to, at least in part, complex immunodeficiency caused by the disease as well as treatment-related factors [Citation38,Citation39]. A higher risk of death in cancer patients, particularly those with hematological malignancies, was also reported in an evaluation of the QResearch database, which comprised 1205 GP practices and 8,256,156 registered adults (aged 19–100 years) [Citation39].

Cancer patients with COVID-19, particularly those with B-cell malignancies, exhibit delayed or negligible seroconversion, prolonged viral shedding, and sustained immune-dysregulation, compared to individuals without cancer [Citation40]. Such patients have an increased likelihood of being admitted into an ICU and a higher risk of dying than patients with COVID-19 without cancer. Furthermore, severe COVID-19 can cause a substantial delay of oncological treatment in these patients, thereby worsening their longer-term prognosis [Citation41].

Whilst at the end of 2021 there was very little available clinical evidence relating to the effectiveness of COVID-19 vaccines in patients with cancer, the picture has changed significantly in 2022 with the publication of numerous systematic reviews/meta-analyses () [Citation42–52]. Based on reviews of the early clinical evidence, seroconversion rates post-vaccination were generally higher in patients with solid tumors compared with hematological malignancies [Citation53–56]. Indeed, impaired/delayed humoral antibody responses and diminished T-cell responses have been reported in cancer patients, most notably in those with hematological malignancies [Citation53–56]. However, a lack of standardization of the methodologies used, the heterogeneity of the populations studied, the changing landscape of the infecting SARS-CoV-2 strains involved, and the range of different vaccine schedules employed, somewhat confuse the overall picture.

Table 2. Summary of systematic reviews published in 2022 investigating immune responses to COVID-19 vaccination in patients with cancer.

In a review investigating seropositivity in patients with diverse health disorders following two doses of COVID-19 vaccination, rates of more than 80% were generally reported for the majority of patients with solid tumors () [Citation57]. After two doses of vaccine, seropositivity rates were 90.5% [95%CI, 87.3–93.4] in patients with solid tumors and 67.0% [95%CI, 55.4–77.0] for hematological malignancies. In cancers reporting low rates of seropositivity (e.g. sarcoma, esophageal/gastric cancer, and neurological cancer), very small numbers of patients were involved (<6) and further research is needed [Citation57].

. Rates of seropositivity after two doses of COVID-19 vaccination (Pfizer-BioNTech, CoronaVac, AstraZeneca, Moderna and Sputnik V vaccines) in different studies involving patients with solid tumors [Citation57].

In systematic reviews of COVID-19 vaccination in patients with ‘cancer’, immunological responses were consistently in the order, control patients (~99%) ≥ solid tumors (88–95%) > hematological malignancies (60–73%) () [Citation42–52] and [Citation38,Citation58,Citation59,Citation60–63]. Risk factors for a poor immunological response to COVID-19 vaccination included cancer in general and more definitively hematological malignancies, anticancer treatments interfering with humoral and cellular responses [Citation43], chemotherapy in patients with solid tumors [Citation48], anti-B cell antibodies targeting CD20 or CD39 [Citation44–48], and CAR T-cell therapy [Citation46]. While cancer patients have been clearly shown to mount a lower and/or delayed immunological response to the SARS-CoV-2 virus, most authors reported only a small number of COVID-19 cases in immunized patients, highlighting the benefits of vaccination. However, Becerril-Gaitan and colleagues noted that patients with cancer were more likely to develop COVID-19 after partial (RR 3.21 [95%CI, 0.35–29.04]) and complete (RR 2.04 [95%CI, 0.38–11.10]) immunization [Citation43]. Data are also now becoming available for booster (third) COVID-19 vaccinations in patients who remained seronegative after the standard vaccination regimen. Mai and colleagues demonstrated a substantial seroconversion rate in patients with hematological malignancies receiving a booster dose of the COVID-19 vaccine, but this rate was about half that of patients with solid tumors (44% vs. 80%) [Citation63; ]. Of note, patients with lung cancer or gastrointestinal cancer were more likely to produce a meaningful increase in antibody titers (16.8 and 25.4 times greater, respectively; both P ≤ 0.05) compared with patients with hematological malignancies. In a short report, Fendler and colleagues noted that a third dose of vaccine (Pfizer or AstraZeneca) boosted the antibody response against variants of concern (Delta or Beta) in approximately 90% of patients with solid tumors and 50% of patients with hematological malignancies who were non-responders after two doses of the vaccine [Citation64]. Al Haji and colleagues also investigated the effectiveness of booster doses of COVID-19 vaccines in a systematic review of 15 studies involving 1205 cancer patients [Citation52]. All studies reported a significant improvement in vaccine efficacy in patients with solid and hematological cancers following a third booster dose. This included patients who did not have a humoral immune response after receiving two doses of vaccine. However, studies directly comparing responses in different cancers found that booster vaccinations were more efficacious in patients with solid tumors compared to those with hematological malignancies (). Indeed, patients with hematological malignancies had poorer seroconversion rates, which were exacerbated by B-cell-depleting therapy. Population studies in cancer patients also highlighted the benefits of a third dose of COVID-19 vaccine. However, the effectiveness was heterogeneous and lower than in the general population. The authors advocate combining vaccine boosters to prevent severe disease with pharmacological interventions to prevent transmission and improve viral clearance [Citation65,Citation66].

Table 3. Summary of systematic reviews published in 2022 investigating immune responses to COVID-19 vaccination in patients with hematological malignancies.

4.3.1. Patients with hematological malignancies

As noted above, patients with hematological malignancies infected with SARS-CoV-2 have been reported to have a higher risk of COVID-19-related complications including severe/critical disease and a worse prognosis (including death), and this has been attributed to, at least in part, complex immunodeficiency caused by the disease and/or treatment [Citation37,Citation38]. An analysis of the OpenSAFELY health analytics platform, involving 17,278,392 primary care records for adult NHS patients in England prior to the availability of vaccines, highlighted an increased risk of COVID-19-related deaths (within the last year) in patients with hematological malignancies with Hazard Ratios (95%CI) adjusted for age and sex of 3.02 (95%CI, 2.24–4.08) [if diagnosed <1 year ago] and 2.56 (95%CI, 2.14–3.06) [if diagnosed between 1 and 4.9 years ago] [Citation19].

Systematic reviews in patients with hematological malignancies published in 2022 are summarized in [Citation38,Citation58,Citation59,Citation60–63]. Noori and colleagues (2022) reported significantly reduced rates of seroprotection following one and two doses of the COVID-19 vaccine compared with healthy subjects (risk ratios 0.36 and 0.62, respectively) [Citation61]. Thus, after two doses of COVID-19 vaccines, pooled seroresponse (SR) rates were approximately 60% across the overall population of patients with hematological malignancies, but the rates were significantly lower (30–51%) after only one dose of vaccine [Citation59,Citation60,Citation61]. In comparative studies, patients with hematological malignancies had a much lower antibody response compared with patients with solid tumors, those undergoing hematopoietic stem cell transplantation (HSCT), and healthy subjects [Citation60–62]. Gagelmann and colleagues reported a response rate of 64% (95%CI, 59–69) for patients with hematological malignancies, versus 82% (95%CI, 77–87) for allogenic HSCT patients, 96% (95%CI, 92–97) for patients with solid tumors, and 98% (95%CI, 96–99) for healthy subjects [Citation62]. The need for alternative treatment approaches to improve immunologic responses in patients with hematological malignancies is highlighted in the systematic reviews in . As noted by Gong et al. [Citation58] these might include improved booster vaccines/doses, anti-COVID-19 monoclonal antibodies, and convalescent serum.

4.3.2. Immune responses in different hematological malignancies

Hematological malignancies comprise a heterogeneous group of disorders in terms of pathophysiology, and they are associated with variable levels of immune dysfunction, which can be further exacerbated by the specific therapies used in their treatment [Citation67]. This is highlighted in the findings from the systematic reviews summarized in . In the largest meta-analysis to date, comparing the two major subtypes of hematological malignancies, pooled SR data demonstrated a lower seropositivity rate for lymphoid vs. myeloid cancers after complete vaccination (62.3% [95%CI, 57.7–66.9] vs. 83.6% [95%CI, 76.3–89.9] [Citation61]. Across all of the reviews, chronic lymphocytic leukemia (CLL) and non-Hodgkin lymphoma (NHL) had the poorest COVID-19 vaccine antibody responses, and this is reflected in the lowest risk ratios for seroconversion at around 0.5 [Citation60; ].

Figure 1. Risk Ratio for seroconversion after 2 doses of COVID-19 mRNA vaccines according to hematological malignancy versus healthy controls [Citation60].

Figure 1. Risk Ratio for seroconversion after 2 doses of COVID-19 mRNA vaccines according to hematological malignancy versus healthy controls [Citation60].

4.3.3. Immune responses in relation to hematological malignancy status and treatment

A number of authors undertook subgroup analyses to ascertain the relationship between the humoral response to COVID-19 vaccination and the current status of the patient’s malignancy and its treatment. Pooled antibody responses to COVID-19 vaccination were reported to be higher in patients whose disease was in remission vs. those with stable or progressive disease (72 vs. 48%; P = 0.014), in individuals who had previously had COVID-19 (87 vs. 66%; P = 0.005), and in patients not being actively treated (76 vs. 35%; P < 0.001) [Citation62].

All systematic reviews/meta-analyses summarized in consistently reported that serologic responses to COVID-19 vaccination were abrogated by cancer treatment, and Noori et al. calculated an RR of 0.58 [95%CI, 0.47–0.73]. In this large analysis of patients with hematological malignancies, the lowest rates of pooled antibody responses after two doses of the COVID-19 vaccine in relation to their anticancer treatment are shown in [Citation61].

Regarding targeted anti-CD20 therapy, lower seropositivity rates were observed in patients who were vaccinated within 12 months of anti-CD20 therapy compared with those vaccinated ≥12 months after completion of their anticancer treatment (15–19% vs. 59–61%, respectively [Citation59,Citation62]. Other targeted anticancer treatments associated with particularly poor antibody response rates to COVID-19 vaccination include Bruton tyrosine kinase inhibitors (BTK; 26.8%), chimeric antigen receptor T-cell therapy (CAR T-cell; 28.4%), and B-cell lymphoma 2 inhibitors (BCL2; 33.3%) [Citation61; ].

. Pooled antibody response after two doses of the COVID-19 vaccine in subgroups of patients with hematological malignancies according to their anticancer treatment [Citation61]

Based on the above findings, the conclusions from these large systematic reviews were consistent and include the following:

  • Patients with hematological malignancies receiving COVID-19 vaccination mount a lower antibody response compared to healthy subjects and those with solid tumors

  • The weakest antibody responses were demonstrated in patients with CLL and NHL

  • Anticancer treatments such as BTK inhibitors, CAR T-cell therapy, anti-CD20 therapy, and other B-cell depleting modalities are also associated with weak antibody responses

  • Additional studies are needed to ascertain whether booster immunization strategies with COVID-19 vaccines will improve the situation

  • Failing this, new approaches for treating these high-risk patients will be required including prophylaxis with pre-exposure monoclonal antibodies.

4.4. Patients undergoing solid organ transplantation

In the analysis of the large OpenSAFELY health analytics platform study, organ transplantation was associated with one of the highest risk levels for death with a Hazard Ratio [95%CI]) adjusted for age and sex of 6.00 [4.73–7.61]) [Citation19]. An and colleagues also reported worse clinical outcomes (mortality, morbidity, and severity) in a meta-analysis of 47 studies, which included solid organ transplant recipients (kidney, liver, heart, lung, or multi-organ). Mortality was 17.4%, which is about 8 times more than that reported in the general population [Citation68]. In contrast, a systematic review/meta-analysis of 31 studies and 590,375 patients (5759 solid organ transplant recipients) with confirmed COVID-19 found no increased risk of mortality in organ transplant patients compared with controls when appropriate adjustments were made for demographic and clinical features at baseline [Citation69]. In a meta-analysis of mortality risk factors in kidney transplant patients with COVID-19 (13 studies, 4440 patients), non-survivors were significantly older, had co-morbidities such as pneumonia, dyspnea, diabetes, cardiovascular, active cancer, and acute kidney injury, and were deceased donor kidney recipients [Citation70].

Publications assessing the effectiveness of COVID-19 vaccines in solid organ transplant recipients have increased significantly in 2022, including numerous systematic reviews/meta-analyses () [Citation71–81]. The systematic reviews/meta-analyses summarized in consistently reported a weaker immune response in patients undergoing solid organ transplantation and a number of risk factors were identified such as:

  • Older age (>65 years) was reported to be associated with lower seroconversion rates in almost all studies

  • Concomitant treatments including not only immunosuppressive regimens (mostly mycophenolate mofetil but also corticosteroids, calcineurin inhibitors and belatacept; and multiple regimens); recent rituximab or antithymocyte globulin exposure

  • Receipt of an organ from a deceased donor

  • Patients with concomitant underlying disorders such as diabetes and reduced renal function

  • In one study, the BNT162b2 vaccine (Pfizer) was less effective than the mRNA-1273 (Moderna) vaccine [Citation74]

  • A number of studies compared antibody responses to COVID-19 by type of organ transplantation and reported and order (best response > to lesser response) of liver > heart > kidney > kidney-pancreas > lung [Citation76,Citation78,Citation81]

  • Finally, a number of studies highlighted the benefits of extra doses of COVID-19 vaccine, including multiple booster doses [e.g. Citation72].

Table 4. Summary of systematic reviews published in 2022 investigating immune responses to COVID-19 vaccination in patients undergoing solid organ transplantation.

4.5. Patients with chronic kidney disease

From the outset of the COVID-19 pandemic, persons with chronic kidney disease were reported to have a higher rate of complications, including more severe morbidity and deaths, than the general population [Citation19,Citation82–84]. Furthermore, large multicenter and population-based studies highlighted a mortality rate of between 20 and 30% in patients receiving dialysis for kidney failure, and a 4-fold increased mortality rate in dialysis versus non-dialysis patients with kidney disease [Citation19,Citation85–87]. In an umbrella review (incorporating 103 reviews), patients with chronic kidney disease/kidney transplantation were at increased risk of developing COVID-19, and these patients had a notably higher incidence of acute kidney injury associated with advanced age, male gender, coronary artery disease, diabetes, and hypertension as risk factors [Citation88]. Separately, in a systematic review involving 13 studies and 18,822 patients with chronic kidney disease and COVID-19, individuals with diabetes had an increased mortality rate compared with non-diabetic patients (RR 1.41 [95%CI, 1.15–1.72; P < 0.001]) [Citation89]. In 3160 patients with kidney failure (dialysis or renal transplant recipients) and COVID-19, there was an increased risk of mortality associated with obesity [Citation90].

Systematic reviews evaluating the effectiveness of COVID-19 vaccination in patients with chronic kidney disease and undergoing dialysis, are summarized in [Citation71,Citation80,Citation91–94]. Key findings include:

  • Seroconversion rates were markedly lower in patients undergoing dialysis for chronic kidney disease receiving one dose of COVID-19 vaccination, but the response improved significantly with a second dose of vaccine.

  • Of the various forms of renal replacement therapy, kidney transplant patients had the worst response to COVID-19 vaccination

  • Risk factors for lower seroconversion rates following COVID-19 vaccination included older age, current immunosuppressive therapy or chemotherapy, lower serum albumin levels, lower white blood cell/lymphocyte counts, length of time on dialysis, and concomitant diabetes.

Table 5. Summary of systematic reviews published in 2022 investigating immune responses to COVID-19 vaccination in patients with renal insufficiency on dialysis.

The poorer response in kidney transplant recipients has been noted in studies that investigated renal replacement therapy in general [Citation71,Citation80,Citation84]. The need to optimize treatment schedules in patients with chronic kidney disease, including the value of booster doses of the COVID-19 vaccine, is an important priority. Indeed, El Karoui & De Vriese noted that progressive waning of immunity and emergence of SARS-CoV-2 variants with a high potential of immune escape highlight the need for booster vaccinations in dialysis patients. Persistent poor vaccine responders are potential candidates for primary prophylaxis with neutralizing monoclonal antibodies [Citation95].

5. Risk factors for COVID-19 vaccine failure

COVID-19 vaccines have proven highly effective in producing high levels of short-term protection against the SARS-CoV-2 virus, reducing the risk of severe infections, hospital admissions and death [Citation96–98]. However, there is evidence that the protective effects of current vaccines against symptomatic disease decreases over time and this has led to the introduction of COVID-19 vaccine booster programs [Citation98]. It was previously reported that COVID-19 vaccine effectiveness against the Delta variant peaked in the early weeks after the second dose and decreased to 47.3% [95%CI, 45–49.6] and 69.7% [95%CI, 68.7–70.5] by ≥20 weeks for ChAdOx1-S (AstraZeneca) and BNT162b (Pfizer/BioNTech) vaccines, respectively. The vaccine remained effective against severe disease outcomes for up to 20 weeks in most groups, although there was greater waning in older adults and those with underlying medical conditions [Citation99]. Vaccine effectiveness of a booster dose was very similar (≥90%) independent of the vaccine used in the primary course, and there was no clear evidence of waning against severe disease up to 10 weeks after the booster. Limited waning was seen 10 or more weeks after the booster. This study provides real-world evidence of substantially increased protection from booster vaccination against mild and severe disease irrespective of the primary course [Citation99].

Primary vaccination with two doses of BNT162b2 (Pfizer/BioNTech), ChAdOx1 nCoV-19 (AstraZeneca), or mRNA-1273 (Moderna) vaccines provided limited protection against COVID-19 caused by the Omicron variant of SARS-CoV-2. A BNT162b2 or mRNA-1273 booster after either the ChAdOx1 nCoV-19 or BNT162b2 primary course increased protection, but the protection waned over time [Citation100]. Significant waning of vaccine effectiveness against symptomatic COVID-19 has been reported and was highest in older adults and in clinical risk groups (patients with chronic diseases, who were immunocompromised or had severe respiratory disease) [Citation101].

Breakthrough infections in persons vaccinated against COVID-19 have been a reality since 2021 and have continued to increase with the predominance of Omicron variants [Citation101]. Vo and colleagues undertook a nationwide retrospective cohort study in US Veterans Affairs Hospitals to identify key risk factors for severe breakthrough of SARS-CoV-2 infections in 110,760 fully vaccinated individuals. The most strongly associated risk factors for severe disease are shown in [Citation102].

. The main demographic, clinical, or vaccination-related risk factors (odds ratios all >2.0) associated with COVID-19 breakthrough despite being fully vaccinated [Citation102].

Findings from our literature search identified the following risk factors for a poor immunological response to COVID-19 vaccination:

  • All immunocompromised persons who have not been fully vaccinated, and in some instances who have not received at least one booster dose as well

  • Older age (>65 years) was reported to be associated with lower seroconversion rates in almost all studies

  • Patients with PI, especially with X-linked agammaglobulinemia (who produce few or no antibodies following vaccination)

  • Cancer patients in general, and more definitively, those with hematological malignancies (the weakest antibody responses were demonstrated in patients with CLL and NHL)

  • Anticancer treatments interfering with humoral and cellular responses (e.g. anti-CD20 therapy and other B-cell depleting modalities)

  • Solid organ transplant recipients having the lowest responses to vaccination by type of organ transplantation were in the order (worst response > to best response) lung > kidney-pancreas > kidney > heart > liver; also, patients receiving an organ from a deceased donor

  • Solid organ transplant recipients on concomitant treatments such as immunosuppressive regimens (not only mycophenolate mofetil, but also corticosteroids, calcineurin inhibitors, and belatacept; and multiple regimens), recent rituximab or antithymocyte globulin exposure, and CAR T-cell recipients

  • Patients with concomitant underlying disorders such as diabetes and reduced kidney function

  • In one study, the BNT162b2 vaccine (Pfizer) was less effective than the mRNA-1273 (Moderna) vaccine [Citation74]

  • Chronic kidney disease risk factors for lower seroconversion rates following COVID-19 vaccination included older age, current immunosuppressive therapy, or chemotherapy, lower serum albumin levels, lower white blood cell/lymphocyte counts, length of time on dialysis and concomitant diabetes.

Excess bodyweight/obesity has been speculated to be a risk factor for vaccine failure, but large studies have shown that the protection provided by COVID-19 vaccines against severe disease outcomes was high across all BMI categories when comparing vaccinated versus unvaccinated persons [Citation103,Citation104]. Patients with immune-mediated diseases such as rheumatoid arthritis and inflammatory bowel disease need special mention since the two conditions can be very similar in terms of pathogenesis (excessive release of inflammatory cytokines to B cell activation and excessive macrophage activation, which can lead to damage to various organs) [Citation105]. In some cases, patients with autoimmune/autoinflammatory diseases (e.g. systemic lupus erythematosus, rheumatoid arthritis, type 1 diabetes, and multiple sclerosis, among others) are more susceptible to SARS-CoV-2 infection, either because of the active autoimmune diseases per se or because of the medications used to treat them [Citation105,Citation106]. Despite blunted immunogenicity observed with some agents, vaccination is generally beneficial in patients with immune-mediated diseases. The greatest reduction in vaccine response has been observed in patients taking B cell-depleting agents, followed by TNF inhibitors, and the CTLA-4 fusion protein abatacept [Citation106]. The importance of COVID-19 vaccination is clearly highlighted in a number of systematic reviews in patients with rheumatic diseases [Citation107,Citation108], inflammatory bowel disease [Citation109,Citation110], and immune-mediated diseases in general [Citation111].

6. Treatment options for patients poorly responsive to COVID-19 vaccination

The pharmacological management of COVID-19 has continued to evolve since SARS-CoV-2 emerged in late 2019, driven in part by global efforts to accelerate vaccine, anti-viral, and monoclonal antibody development [Citation112]. While vaccination has clearly benefitted a large proportion of the world’s population with almost 70% receiving at least one dose of the COVID-19 vaccine [Citation113], there are many individuals who remain unvaccinated, have not completed a primary series plus booster, or who mounted a weak immune response to vaccination (especially immunocompromised persons). These patients are at a higher risk of severe disease and will likely require additional therapeutic support [Citation17,Citation114]. There is therefore an unmet need for alternative approaches.

A variety of medicines have been recommended for the prevention or treatment of COVID-19 in at-risk patients [Citation115]. Broadly speaking, these include antiviral drugs, monoclonal antibodies, convalescent plasma, immunomodulatory agents, and others (e.g. anticoagulants). It is important to note that this field of medical treatment is evolving rapidly with new therapies being made available and others being discontinued. A full review of therapeutic approaches is outside the scope of this review, and we provide an overview for the reader to provide context. For a fuller review see Zhang et al. 2023 [Citation116].

Antiviral agents mainly include polymerase inhibitors (e.g. remdesivir), protease inhibitors (e.g. ritonavir and nirmatrelvir), inhibitors of nucleoside and nucleotide reverse transcriptase (e.g. molnupiravir) entry and uncoating inhibitors (e.g. amantadine and enfuvirtide), and others [Citation116,Citation117]. Antivirals are generally considered most effective when prescribed early in the course of the disease to reduce the risk of severe disease [Citation118].

Host-related therapies are typically immunomodulatory therapies such as corticosteroids, Janus kinase inhibitors (e.g. baricitinib and tofacitinib), and antibody therapies (e.g. anakinra and tocilizumab), which are prescribed following the onset of severe illness to reduce harmful inflammation [Citation116,Citation117].

Monoclonal antibodies have demonstrated efficacy as both pre-/post-exposure prophylaxis and treatment for COVID-19 [Citation119–125]. Antibodies have high specificity, although mutations in the targeted regions (usually of the spike protein) can prevent binding, resulting in viral evasion and resistance [Citation119]. Because of the high frequency of non-susceptible Omicron variants currently circulating in the US, casirivimab/imdevimab, sotrovimab, and bamlanivimab/etesevimab are no longer currently authorized [Citation126–128], whereas tixagevimab/cilgavimab has received Emergency Use Authorization for pre-exposure immunoprophylaxis from the FDA [Citation129].

In Europe, a number of agents have received authorization for use against COVID-19 from the EMA including the monoclonal antibodies tixagevimab/cilgavimab, sotrovimab, casirivimab/imdevimab, and regdanvimab; the antiviral drug ritonavir/nirmatrelvir and remdesivir (molnupiravir is currently under review); and the immune modulators anakinra and tocilizumab [Citation130,Citation131]. This is a dynamic area as new variants emerge, and for the latest information of availability and usage, the reader is referred to the applicable FDA [Citation126] and EMA [Citation130] websites.

A subset of people develop chronic/persistent symptoms after an acute episode of COVID-19, and such symptoms can last for weeks and up to many months. These post-acute sequelae of COVID-19 are often referred to as ‘Long COVID’ [Citation132–134]. Multivariate regression analysis showed that long COVID was more common in females and individuals presenting with at least one co-morbidity [Citation132]. While the response to COVID-19 vaccines has been variable in patients with Long COVID, there is mounting evidence that they are protective, even against breakthrough infections [Citation133,Citation134]. However, evidence against Omicron strains of the virus is limited and different therapeutic options may be required long term to optimally manage patients at risk of developing Long COVID [Citation132–135]. The individualized nature of long COVID supports the need for different therapeutic approaches. Furthermore, improved diagnostic procedures to identify patients at-risk, possibly including genomic biomarkers, could support a more effective and predictive personalized approach to patient management.

7. Limitations

As noted in a recent Cochrane review, there has been an explosion in COVID-19-related research to an extent that it is highly challenging for a clinician to keep up-to-date with the published literature [Citation67]. Practically, we decided to limit our search to the most recent 12 months and solely to systematic reviews/meta-analyses (clinical best evidence). Furthermore, we focused our searches and discussions on patient groups known to be associated with a compromised immune status or receiving immunosuppressant therapy (primary and secondary immunodeficiencies (e.g. HIV), cancer (especially hematological malignancies), chronic kidney disease, and solid organ transplant recipients). This scoping review approach offers a high-level view of the dynamic changes occurring with COVID-19 vaccines in the management of immunocompromised patients at risk from SARS-CoV-2 infection. Our primary focus was to identify any subgroups of patients that had a reduced response to COVID-19 vaccination and who may therefore be candidates for alternative treatment approaches better suited to their needs. However, it should be noted that a limitation of our review is that we did not assess the methodological quality of the included systematic reviews.

This review has a number of other notable limitations related to dynamic changes that are characteristic of the COVID-19 pandemic, and these result in significant heterogeneity across studies. Factors that were generally poorly reported and could lead to bias included the virus strains involved (the changing pattern of ‘variants of concern’ potentially influencing vaccine effectiveness); details of vaccines/dosages (including effects of booster doses) and timing of results relative to the time of administration; the majority of data relate to the use of mRNA vaccines and are markedly less for viral vector vaccines and inactivated whole virus vaccines; the data derive almost exclusively from observational studies (which is typical for infection prevention literature); and there was a lack of consistency with respect to serological testing to quantify antibody responses. Consequently, despite the fact that this review represents data from a very large number of patients, the findings should be viewed with caution given the considerable heterogeneity in the identified studies.

8. Conclusions

Our analysis of 12-month data from late 2021 takes an aerial view of the rapidly expanding literature related to vaccination in immunocompromised persons at risk from COVID-19. The goal was to identify factors relating to a suboptimal response to vaccination and provide evidence of which patients may benefit from preexposure prophylaxis. Findings from our literature review identified a number of risk factors for a poor immunological response to COVID-19 vaccination with key ones being non-fully vaccinated immunocompromised persons, older age (especially >75 years), patients with hematological malignancies, patients receiving treatments interfering with humoral and cellular responses (e.g. anti-CD20 therapy and other B-cell depleting modalities), some solid organ transplant recipients, and those receiving immunosuppressive therapy.

Development of models to estimate the probability of a patient progressing to severe disease and to guide treatment and prophylaxis planning will be a step forward to meet the unmet need for many patients not adequately protected from the SARS-CoV-2 virus at the present time. This will require more detailed reporting of virus ‘variants of concern,’ full details of vaccines/dosages (including effects of booster doses), and timing of results relative to the time of administration.

9. Expert opinion

The emergence of SARS-CoV-2 and the subsequent COVID-19 pandemic have led to an unprecedented research effort related to zoonotic infections in general and coronavirus infections in particular. The response of the global community, including the pharmaceutical industry, particularly in regard to disease management initiatives has been impressive after the catastrophic early phases of the pandemic. From protective measures, such as social distancing, mask wearing, and hand sanitization, to vaccine development and newer antiviral approaches including monoclonal antibodies (e.g. tixagevimab/cilgavimab, casirivimab/imdevimab and sotrovimab), we have come a long way in a relatively short time. However, it is important to note that the journey is clearly still ongoing. While the Omicron variant has led to a milder form of the disease, the morbidity and mortality associated with COVID-19 remains too high, and optimization of treatment for all—including poorer communities, vulnerable patients such as immunocompromised individuals and those not benefiting from current vaccines—needs to be prioritized. Beyond this, we have no idea what future variants of SARS-CoV-2 will look like in terms of pathogenicity and virulence. Efforts to maintain the currency of vaccination programs based on variants of interest as well personalization of pharmacological approaches (including immunotherapy) need to be ongoing.

The findings of this review of systematic reviews in immunocompromised populations provide further knowledge about COVID-19 vaccination status and effectiveness in an at-risk/vulnerable group of patients. By necessity, it takes a broad view of the dynamic changes that are occurring during the pandemic, when many variables such as virus strains, vaccine types, and vaccination status are all changing and impact our view of the disease. The administration of booster doses of COVID-19 vaccines has helped maintain vaccine effectiveness against different outcomes, such as infection, symptomatic disease, and hospitalization in the short term, but there is emerging evidence of a waning level of protection against Omicron strains on COVID-19.

The COVID-19 pandemic is both complex and unpredictable, and development of effective vaccines and alternative treatments is a key strategy to manage the disease. In terms of vaccine research, a number of developments are being investigated such as:

  • variant-specific COVID-19 vaccines and a number of companies are developing Omicron-specific vaccines

  • development of a multi-antigenic vaccine that has antigens of different variants to increase the breadth of the immune response

  • a ‘prime-boost strategy’ in which a priming response is initiated with an effective mRNA vaccine and boosted with a different vaccine such as an intranasal product—this again is intended to produce a broad immunological response with higher protection against infection and transmission. There is a long list of intranasal vaccines under development

  • development of bivalent vaccines to produce a potent, more durable, and broader immune responses is also being investigated. One such approach involves a modified, bivalent booster vaccine containing equal amounts of messenger RNAs (mRNAs) encoding the ancestral SARS-CoV-2 and beta variant spike proteins. This bivalent vaccine elicited superior and more durable neutralizing antibody responses against the Beta, Delta, and Omicron variants as compared with mRNA-1273 alone

  • Another potential development is a bivalent vaccine against SARS-CoV-2 and influenza viruses, but this is still in the early phases of development.

COVID-19 has changed the world as we know it, and the pandemic has caused immense economic and healthcare disruption. The societal consequences have been enormous, but the global response to manage the disease, especially via vaccine development programmes and the introduction of new treatment modalities, has been impressive. However, the disease burden from SARS-CoV-2 infection is a concern as the number of new cases worldwide continues to be high. While COVID-19 vaccination is helping to reduce the number of deaths and serious cases of the disease, for the time being, there is little evidence that levels of immunity in the community are sufficient to consider the pandemic to be over. This is possibly driven by the emergence of new viral variants, which remains a major concern given the possibility of a more virulent mutation of SARS-CoV-2 evolving. This is already causing problems for some treatment choices, with the emergence of sub-variants such as SARS-CoV-2 Omicron BA.2 in the US. This strain has resulted in the FDA recommending that sotrovimab is no longer used and also restricting the use of bamlanivimab/etesevimab and casirivimab/imdevimab to patients likely to have been infected with or exposed to variants that are susceptible to these treatments.

Now, more than ever, we need to maintain our focus on providing the best clinical protection against the SARS-CoV-2 virus. This could include optimization of vaccine strategies (type of vaccine and dosage schedules) and provision of alternative immunotherapeutic approaches for immunocompromised individuals who do not respond adequately to COVID-19 vaccination.

Declaration of interest

V Shetty and N Azmi are employees of AstraZeneca. S Clissold is a professional scientific writer (ContentEdNet, Singapore). The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript, apart from those disclosed.

Reviewer disclosures

A reviewer of this manuscript has disclosed that they have provided consultancies for epidemiological research and data analyses for Moderna, Pfizer, GSK, Seqirus, and Astra Zeneca. Peer reviewers of this manuscript have received an honorarium for their review work.

Authors’ contributions

All authors meet the International Committee of Medical Journal Editors (ICMJE) criteria for authorship for this article, which was developed in line with Good Publishing Practice-3 guidelines. All authors contributed to the review’s conception and search strategy, focus, and interpretation, drafting, and/or revision of the manuscript and have given their approval for this version of the manuscript to be submitted for publication.

Additional information

Funding

This manuscript was funded by AstraZeneca, Singapore.

References

  • World Health Organization. Health topics/Coronavirus disease (COVID-19). Available at: https://www.who.int/health-topics/coronavirus#tab=tab_1 Cited 2022 Aug 10.
  • World Health Organization. WHO coronavirus (COVID-19) dashboard. Cited 2022 Aug 10. Available at: https://covid19.who.int/.
  • Centers for Disease Control and Prevention. Covid-19 vaccines for moderately to severely immunocompromised people. CDC. [Cited 2021 Sept 27]. www.cdc.gov/coronavirus/2019-ncov/vaccines/ recommendations/immuno.html
  • Manuel O, Estabrook M. on behalf of the American Society of Transplantation Infectious Diseases Community of Practice. RNA respiratory viral infections in solid organ transplant recipients: guidelines from the American Society of Transplantation Infectious Diseases Community of Practice. Clin Transplant. 2019;33:e13511.
  • Mohammed AH, Blebil A, Dujaili J, et al. The risk and impact of COVID-19 pandemic on immunosuppressed patients: cancer, HIV, and solid organ transplant recipients. AIDS Rev. 2020;22:151–157.
  • Madan A, Siglin J, Khan A. Comprehensive review of implications of COVID-19 on clinical outcomes of cancer patients and management of solid tumors during the pandemic. Cancer Med. 2020;9:9205–9218.
  • Pereira MR, Mohan S, Cohen DJ, et al. COVID-19 in solid organ transplant recipients: initial report from the US epicenter. Am J Transplant. 2020;20:1800–1808.
  • Curtis JR, Johnson SR, Anthony DD, et al. American college of rheumatology guidance for COVID-19 vaccination in patients with rheumatic and musculoskeletal diseases: version 3. Arthritis Rheumatol. 2021;73:e60–75.
  • Ssentongo P, Heilbrunn ES, Ssentongo AE, et al. Epidemiology and outcomes of COVID-19 in HIV-infected individuals: a systematic review and meta-analysis. Sci Rep. 2021;11:6283.
  • Tregoning JS, Flight KE, Higham SL, et al. Progress of the COVID-19 vaccine effort: viruses, vaccines and variants versus efficacy, effectiveness and escape. Nat Rev Immunol. 2021 Oct;21(10):626–636.
  • Polack FP, Thomas SJ, Kitchin N, et al. C4591001 clinical trial group. safety and efficacy of the BNT162b2 mRNA Covid-19 vaccine. N Engl J Med. 2020 Dec 31;383(27):2603–2615.
  • LR B, El SHM, Essink B, et al. COVE study group. efficacy and safety of the mRNA-1273 SARS-CoV-2 vaccine. N Engl J Med. 2021 Feb 4;384(5):403–416.
  • Voysey M, Clemens SAC, Madhi SA, et al. Oxford COVID Vaccine Trial Group. Safety and efficacy of the ChAdOx1 nCoV-19 vaccine (AZD1222) against SARS-CoV-2: an interim analysis of four randomised controlled trials in Brazil, South Africa, and the UK. Lancet. 2021 Jan 9;397(10269):99–111.
  • Lee ARYB, Wong SY, Chai LYA, et al. Efficacy of covid-19 vaccines in immunocompromised patients: systematic review and meta-analysis. BMJ. 2022 Mar 2;376: e068632.
  • Jrc S, PD K, Pham H, et al. Factors associated with severe outcomes among immunocompromised adults hospitalized for COVID-19 - COVID-NET, 10 States, March 2020-February 2022. MMWR Morb Mortal Wkly Rep. 2022 Jul 8;71(27):878–884.
  • Goldman JD, Robinson PC, Uldrick TS, et al. in immunocompromised populations: implications for prognosis and repurposing of immunotherapies. J Immunother Cancer. 2021 Jun;9(6):e002630.
  • Levin MJ, Ustianowski A, De Wit S, Launay O, Avila M, Templeton A, Yuan Y, Seegobin S, Ellery A, Levinson DJ, Ambery P, Arends RH, Beavon R, Dey K, Garbes P, Kelly EJ, Koh GCKW, Near KA, Padilla KW, Psachoulia K, Sharbaugh A, Streicher K, Pangalos MN, Esser MT; PROVENT Study Group. Intramuscular AZD7442 (Tixagevimab-Cilgavimab) for prevention of Covid-19. N Engl J Med. 2022 Jun 9;386(23):2188–2200.
  • Riva JJ, Malik KM, Burnie SJ, et al. What is your research question? An introduction to the PICOT format for clinicians. J Can Chiropr Assoc. 2012;56(3):167–171.
  • Williamson EJ, Walker AJ, Bhaskaran K, et al. Factors associated with COVID-19-related death using OpenSAFELY. Nature. 2020 Aug;584(7821):430–436.
  • Földi M, Farkas N, Kiss S, Zádori N, Váncsa S, Szakó L, Dembrovszky F, Solymár M, Bartalis E, Szakács Z, Hartmann P, Pár G, Erőss B, Molnár Z, Hegyi P, Szentesi A; KETLAK Study Group. Obesity is a risk factor for developing critical condition in COVID-19 patients: a systematic review and meta-analysis. Obes Rev. 2020 Oct;21(10):e13095.
  • Földi M, Farkas N, Kiss S, et al. Visceral adiposity elevates the risk of critical condition in COVID-19: a systematic review and meta-analysis. Obesity (Silver Spring). 2021 Mar;29(3):521–528.
  • Huang Y, Lu Y, Huang YM, Wang M, Ling W, Sui Y, Zhao HL. Obesity in patients with COVID-19: a systematic review and meta-analysis. Metabolism. 2020;113:154378. Dec.
  • Popkin BM, Du S, Green WD, et al. Individuals with obesity and COVID-19: a global perspective on the epidemiology and biological relationships. Obes Rev. 2020 Nov;21(11):e13128.
  • Gao M, Piernas C, Astbury NM, et al. Associations between body-mass index and COVID-19 severity in 6·9 million people in England: a prospective, community-based, cohort study. Lancet Diabetes Endocrinol. 2021 Jun;9(6):350–359.
  • Stefan N, Birkenfeld AL, Schulze MB. Global pandemics interconnected - obesity, impaired metabolic health and COVID-19. Nat Rev Endocrinol. 2021 Mar;17(3):135–149.
  • Sawadogo W, Tsegaye M, Gizaw A, et al. Overweight and obesity as risk factors for COVID-19-associated hospitalisations and death: systematic review and meta-analysis. BMJ Nutr Prev Health. 2022 Jan 19;5(1):10–18.
  • Sardu C, Marfella R, Prattichizzo F, et al. Effect of hyperglycemia on COVID-19 outcomes: vaccination efficacy, disease severity, and molecular mechanisms. J Clin Med. 2022 Mar 12;11(6):1564.
  • Ng SM, Pan J, Mouyis K, et al. Quantifying the Excess Risk of Adverse COVID-19 outcomes in unvaccinated individuals with diabetes mellitus, hypertension, ischaemic heart disease or myocardial injury: a meta-analysis. Front Cardiovasc Med. 2022 Apr 26;9: 871151.
  • Stefan N, Sippel K, Heni M, et al., Hrabe de Angelis M, Ludwig DS, Schulze MB, Jensen BEO, Birkenfeld AL. Obesity and Impaired metabolic health increase risk of COVID-19-related mortality in young and middle-aged adults to the level observed in older people: the LEOSS registry. Front Med. 2022 May 11; 9:875430. (Lausanne)
  • Marra AR, Kobayashi T, Suzuki H, et al. Short-term effectiveness of COVID-19 vaccines in immunocompromised patients: a systematic literature review and meta-analysis. J Infect. 2022 Mar;84(3):297–310.
  • Delavari S, Abolhassani H, Abolnezhadian F, et al. Impact of SARS-CoV-2 pandemic on patients with primary immunodeficiency. J Clin Immunol. 2021 Feb;41(2):345–355. Epub 2020 Dec 1
  • Shields AM, Tadros S, Al-Hakim A, et al. Impact of vaccination on hospitalization and mortality from COVID-19 in patients with primary and secondary immunodeficiency: the United Kingdom experience. Front Immunol. 2022 Sep 23;13: 984376.
  • SeyedAlinaghi S, Karimi A, Barzegary A, et al. mortality in patients with immunodeficiency and its predictors: a systematic review. Eur J Med Res. 2022 Oct 8;27(1):195.
  • Drzymalla E, Green RF, Knuth M, et al. COVID-19-related health outcomes in people with primary immunodeficiency: a systematic review. Clin Immunol. 2022Oct;243:109097.
  • Kang L, Shang W, Gao P, et al. and Safety of COVID-19 vaccines among people living with HIV: a systematic review and meta-analysis. Vaccines (Basel). 2022 Sep 19;10(9):1569.
  • Yin J, Chen Y, Li Y, et al. Immunogenicity and efficacy of COVID-19 vaccines in people living with HIV: a systematic review and meta-analysis. Int J Infect Dis. 2022 Oct 12;124: 212–223.
  • Di Felice G, Visci G, Teglia F, et al. Effect of cancer on outcome of COVID-19 patients: a systematic review and meta-analysis of studies of unvaccinated patients. Elife. 2022 Feb 16;11: e74634.
  • Piechotta V, Mellinghoff SC, Hirsch C, et al. Effectiveness, immunogenicity, and safety of COVID-19 vaccines for individuals with hematological malignancies: a systematic review. Blood Cancer J. 2022 May 31;12(5):86.
  • Clift AK, Coupland CAC, Keogh RH, et al. Living risk prediction algorithm (QCOVID) for risk of hospital admission and mortality from coronavirus 19 in adults: national derivation and validation cohort study. BMJ. 2020 Oct 20;371: m3731.
  • Monin L, Laing AG, Muñoz-Ruiz M, et al. Safety and immunogenicity of one versus two doses of the COVID-19 vaccine BNT162b2 for patients with cancer: interim analysis of a prospective observational study. Lancet Oncol. 2021 Jun;22(6):765–778.
  • Oosting SF, van der VAAM, GeurtsvanKessel CH, et al. mRNA-1273 COVID-19 vaccination in patients receiving chemotherapy, immunotherapy, or chemoimmunotherapy for solid tumours: a prospective, multicentre, non-inferiority trial. Lancet Oncol. 2021 Dec;22(12):1681–1691.
  • Cavanna L, Citterio C, Toscani I. COVID-19 vaccines in cancer patients. seropositivity and safety. systematic review and meta-analysis. Vaccines (Basel). 2021 Sep 20;9(9):1048.
  • Becerril-Gaitan A, Vaca-Cartagena BF, Ferrigno AS, et al. Immunogenicity and risk of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection after coronavirus disease 2019 (COVID-19) vaccination in patients with cancer: a systematic review and meta-analysis. Eur J Cancer. 2022Jan;160:243–260.
  • Corti C, Antonarelli G, Scotté F, et al. Seroconversion rate after vaccination against COVID-19 in patients with cancer-a systematic review. Ann Oncol. 2022 Feb;33(2):158–168.
  • Guven DC, Sahin TK, Kilickap S, et al. Antibody responses to COVID-19 vaccination in cancer: a systematic review. Front Oncol. 2021 Nov 4;11: 759108.
  • Sakuraba A, Luna A, Micic D. Serologic response following SARS-COV2 vaccination in patients with cancer: a systematic review and meta-analysis. J Hematol Oncol. 2022 Feb 5;15(1):15.
  • Javadinia SA, Alizadeh K, Mojadadi MS, et al. Vaccination in patients with malignancy; A systematic review and meta-analysis of the efficacy and safety. Front Endocrinol (Lausanne). 2022 May 2;13: 860238.
  • Yang W, Zhang D, Li Z, et al. Predictors of poor serologic response to COVID-19 vaccine in patients with cancer: a systematic review and meta-analysis. Eur J Cancer. 2022Sep;172:41–50.
  • Tang K, Wei Z, Wu X. Impaired serological response to COVID-19 vaccination following anticancer therapy: a systematic review and meta-analysis. J Med Virol. 2022 Oct;94(10):4860–4868.
  • Martins-Branco D, Nader-Marta G, Tecic Vuger A, et al. Immune response to anti-SARS-CoV-2 prime-vaccination in patients with cancer: a systematic review and meta-analysis. J Cancer Res Clin Oncol. 2022Jul;22:1–6.
  • Yin J, Chen Y, Li Y, et al. Seroconversion rate after COVID-19 vaccination in patients with solid cancer: a systematic review and meta-analysis. Hum Vaccin Immunother. 2022Sep;26:2119763.
  • Al Hajji Y, Taylor H, Starkey T, et al. Antibody response to a third booster dose of SARS-CoV-2 vaccination in adults with haematological and solid cancer: a systematic review. Br J Cancer. 2022Oct;12:1–10.
  • Thakkar A, Gonzalez-Lugo JD, Goradia N, et al. Seroconversion rates following COVID-19 vaccination among patients with cancer. Cancer Cell. 2021 Aug 9;39(8):1081–1090.e2.
  • Guven DC, Sahin TK, Akın S, et al. Impact of therapy in patients with hematologic malignancies on seroconversion rates after SARS-CoV-2 vaccination. Oncologist. 2022 Apr 5;27(4):e357–e361.
  • Kakkassery H, Carpenter E, Patten PEM, et al. Immunogenicity of SARS-CoV-2 vaccines in patients with cancer. Trends Mol Med. 2022 Aug 3;S1471–4914(22):00186–1.
  • Mehrabi Nejad MM, Moosaie F, Dehghanbanadaki H, et al. Immunogenicity of COVID-19 mRNA vaccines in immunocompromised patients: a systematic review and meta-analysis. Eur J Med Res. 2022;27(1):23.
  • Cho K, Park S, Kim EY, et al. Immunogenicity of COVID-19 vaccines in patients with diverse health conditions: a comprehensive systematic review. J Med Virol. 2022 Sep;94(9):4144–4155.
  • Gong IY, Vijenthira A, Betschel SD, et al. vaccine response in patients with hematologic malignancy: a systematic review and meta-analysis. Am J Hematol. 2022 Apr;97(4):E132–E135.
  • Teh JSK, Coussement J, Neoh ZCF, et al. Immunogenicity of COVID-19 vaccines in patients with hematologic malignancies: a systematic review and meta-analysis. Blood Adv. 2022 Apr 12;6(7):2014–2034.
  • Rinaldi I, Pratama S, Wiyono L, et al. Efficacy and safety profile of COVID-19 mRNA vaccine in patients with hematological malignancies: systematic review and meta-analysis. Front Oncol. 2022 Aug 5;12: 951215.
  • Noori M, Azizi S, Abbasi Varaki F, et al. A systematic review and meta-analysis of immune response against first and second doses of SARS-CoV-2 vaccines in adult patients with hematological malignancies. Int Immunopharmacol. 2022Sep;110:109046.
  • Gagelmann N, Passamonti F, Wolschke C, et al. Antibody response after vaccination against SARS-CoV-2 in adults with hematological malignancies: a systematic review and meta-analysis. Haematologica. 2022 Aug 1;107(8):1840–1849.
  • Mai AS, Lee ARYB, Tay RYK, et al. Booster doses of COVID-19 vaccines for patients with haematological and solid cancer: a systematic review and individual patient data meta-analysis. Eur J Cancer. 2022Sep;172:65–75.
  • Fendler A, Shepherd STC, Au L, et al. Immune responses following third COVID-19 vaccination are reduced in patients with hematological malignancies compared to patients with solid cancer. Cancer Cell. 2022;40(4):438.
  • Lee LYW, Starkey T, Ionescu MC, et al. Vaccine effectiveness against COVID-19 breakthrough infections in patients with cancer (UKCCEP): a population-based test-negative case-control study. Lancet Oncol. 2022 Jun;23(6):748–757.
  • Lee LYW, Ionescu MC, Starkey T, et al. UK Coronavirus Cancer Programme. COVID-19: third dose booster vaccine effectiveness against breakthrough coronavirus infection, hospitalisations and death in patients with cancer: a population-based study. Eur J Cancer. 2022 Nov;175:1–10.
  • Kreuzberger N, Hirsch C, Andreas M, et al. Immunity after COVID-19 vaccination in people with higher risk of compromised immune status: a scoping review. Cochrane Database Syst Rev. 2022 Aug 9;8(8):CD015021.
  • An W, Wang Q, Kim TE, et al. Clinical characteristics and outcome of coronavirus disease 2019 infection in patients with solid organ transplants: a systematic review and meta-analysis. J Infect Public Health. 2022 Mar;15(3):365–372.
  • Gatti M, Rinaldi M, Bussini L, et al. ORCHESTRA study group. Clinical outcome in solid organ transplant recipients affected by COVID-19 compared to general population: a systematic review and meta-analysis. Clin Microbiol Infect. 2022 Aug;28(8):1057–1065.
  • Udomkarnjananun S, Kerr SJ, Townamchai N, et al. Mortality risk factors of COVID-19 infection in kidney transplantation recipients: a systematic review and meta-analysis of cohorts and clinical registries. Sci Rep. 2021 Oct 8;11(1):20073.
  • Akyol M, Çevik E, Ucku D, et al. Immunogenicity of SARS-CoV-2 mRNA vaccine in dialysis and kidney transplant patients: a systematic review. Tuberk Toraks. 2021 Dec;69(4):547–560.
  • Manothummetha K, Chuleerarux N, Sanguankeo A, et al. And risk factors associated with poor humoral immune response of SARS-CoV-2 vaccines in recipients of solid organ transplant: a systematic review and meta-analysis. JAMA Network Open. 2022 Apr 1;5(4):e226822.
  • Shoar S, ACC P-R, Patarroyo-Aponte G, et al. Immune response to SARS-CoV-2 vaccine among heart transplant recipients: a systematic review. Clin Med Insights Circ Respir Pulm Med. 2022 Jun 5;16: 11795484221105327.
  • Verleye A, Wijtvliet V, Abrams S, Hellemans R, Bougrea R, Massart A, Pipeleers L, Wissing KM, Ariën KK, De Winter BY, Van Damme P, Abramowicz D, Ledeganck KJ. Seroconversion rate after primary vaccination with two doses of BNT162b2 versus mRNA-1273 in solid organ transplant recipients: a systematic review and meta-analysis. Nephrol Dial Transplant. 2022 Jul 26;37(8):1566–1575.
  • Zong K, Peng D, Yang H, et al. Risk factors for weak antibody response of SARS-CoV-2 vaccine in adult solid organ transplant recipients: a systemic review and meta-analysis. Front Immunol. 2022 Jun 14;13: 888385.
  • Li J, Ayada I, Wang Y, et al. Factors associated with COVID-19 vaccine response in transplant recipients: a systematic review and meta-analysis. Transplantation. 2022 Oct 1;106(10):2068–2075.
  • Ge C, Du K, Luo M, et al. Serologic response and safety of COVID-19 vaccination in HSCT or CAR T-cell recipients: a systematic review and meta-analysis. Exp Hematol Oncol. 2022 Aug 16;11(1):46.
  • Meshram HS, Kute V, Rane H, et al. Humoral and cellular response of COVID-19 vaccine among solid organ transplant recipients: a systematic review and meta-analysis. Transpl Infect Dis. 2022Aug;4:e13926.
  • Sakuraba A, Luna A, Micic D. A systematic review and Meta-analysis of serologic response following coronavirus disease 2019 (COVID-19) vaccination in solid organ transplant recipients. Viruses. 2022 Aug 19;14(8):1822.
  • Ma BM, Tam AR, Chan KW, et al. Immunogenicity and safety of COVID-19 vaccines in patients receiving renal replacement therapy: a systematic review and meta-analysis. Front Med (Lausanne). 2022 Mar 9;9: 827859.
  • Luo D, Chen X, Du J, et al. Immunogenicity of COVID-19 vaccines in chronic liver disease patients and liver transplant recipients: a systematic review and meta-analysis. Liver Int. 2023 Jan;43(1):34–48.
  • El Karoui K, Hourmant M, Ayav C, et al. Vaccination and COVID-19 dynamics in dialysis patients. Clin J Am Soc Nephrol. 2022 Mar;17(3):395–402.
  • Cancarevic I, Nassar M, Daoud A, et al. Mortality rate of COVID-19 infection in end stage kidney disease patients on maintenance hemodialysis: a systematic review and meta-analysis. World J Virol. 2022 Sep 25;11(5):352–361.
  • Sanders JF, Bemelman FJ, Messchendorp AL, et al. The RECOVAC Immune-response Study: the Immunogenicity, Tolerability, and Safety of COVID-19 vaccination in patients with chronic kidney disease, on dialysis, or living with a kidney transplant. Transplantation. 2022 Apr 1;106(4):821–834.
  • Hilbrands LB, Duivenvoorden R, Vart P, et al. COVID-19-related mortality in kidney transplant and dialysis patients: results of the ERACODA collaboration. Nephrol Dial Transplant. 2020 Nov 1;35(11):1973–1983.
  • Jager KJ, Kramer A, Chesnaye NC, et al. Results from the ERA-EDTA Registry indicate a high mortality due to COVID-19 in dialysis patients and kidney transplant recipients across Europe. Kidney Int. 2020 Dec;98(6):1540–1548.
  • De Meester J, De Bacquer D, Naesens M, et al. NBVN kidney registry group. incidence, characteristics, and outcome of COVID-19 in adults on kidney replacement therapy: a regionwide registry study. J Am Soc Nephrol. 2021 Feb;32(2):385–396.
  • Yang L, Li J, Wei W, et al. Kidney health in the COVID-19 pandemic: an umbrella review of meta-analyses and systematic reviews. Front Public Health. 2022 Sep 12;10: 963667.
  • Iryaningrum MR, Supriyadi R, Lawrensia S, et al. Diabetes and mortality among patients with chronic kidney disease and COVID-19: a systematic review, meta-analysis, and meta-regression. Indian J Nephrol. 2022 July-August;32(4):327–333.
  • Tantisattamo E, Imhof C, Jager KJ, et al. ERACODA collaborators. Association of obesity with 3-month mortality in kidney failure patients with COVID-19. Clin Kidney J. 2022 Mar 21;15(7):1348–1360.
  • Chen JJ, Lee TH, Tian YC, et al. Immunogenicity rates after SARS-CoV-2 vaccination in people with end-stage kidney disease: a systematic review and meta-analysis. JAMA Network Open. 2021 Oct 1;4(10):e2131749.
  • Mehta N, Shah S, Paudel K, et al. Safety and efficacy of coronavirus disease-19 vaccines in chronic kidney disease patients under maintenance hemodialysis: a systematic review. Health Sci Rep. 2022 Jun 16;5(4):e700.
  • Peiyao R, Mengjie Y, Xiaogang S, et al. Immunogenicity and safety of SARS-CoV-2 vaccine in hemodialysis patients: a systematic review and meta-analysis. Front Public Health. 2022 Sep 23;10: 951096.
  • Falahi S, Sayyadi H, Kenarkoohi A. Immunogenicity of COVID-19 mRNA vaccines in hemodialysis patients: systematic review and meta-analysis. Health Sci Rep. 2022 Oct 3;5(6):e874.
  • El Karoui K, De Vriese ASCOVID-19. in dialysis: clinical impact, immune response, prevention, and treatment. Kidney Int. 2022 May;101(5):883–894.
  • Lopez Bernal J, Andrews N, Gower C, et al. Effectiveness of Covid-19 Vaccines against the B.1.617.2 (Delta) variant. N Engl J Med. 2021 Aug 12;385(7):585–594.
  • Vasileiou E, Simpson CR, Shi T, et al. Interim findings from first-dose mass COVID-19 vaccination roll-out and COVID-19 hospital admissions in Scotland: a national prospective cohort study. Lancet. 2021 May 1;397(10285):1646–1657.
  • Ismail SJ, Tunis MC, Zhao L, et al. Navigating inequities: a roadmap out of the pandemic. BMJ Glob Health. 2021;6(1):e004087.
  • Andrews N, Stowe J, Kirsebom F, et al. Effectiveness of COVID-19 booster vaccines against COVID-19-related symptoms, hospitalization and death in England. Nat Med. 2022 Apr;28(4):831–837.
  • Andrews N, Stowe J, Kirsebom F, et al. Covid-19 vaccine effectiveness against the Omicron (B.1.1.529) Variant. N Engl J Med. 2022 Apr 21;386(16):1532–1546.
  • Andrews N, Tessier E, Stowe J, et al. Duration of protection against mild and severe disease by Covid-19 vaccines. N Engl J Med. 2022 Jan 27;386(4):340–350.
  • Vo AD, La J, Wu JT-Y, et al. Factors associated with severe COVID-19 among vaccinated adults treated in US veterans affairs hospitals. JAMA Network Open. 2022 Oct 3;5(10):e2240037.
  • Butsch WS, Hajduk A, Cardel MI, et al. COVID-19 vaccines are effective in people with obesity: a position statement from The Obesity Society. Obesity (Silver Spring). 2021 Oct;29(10):1575–1579.
  • Piernas C, Patone M, Astbury NM, et al. Associations of BMI with COVID-19 vaccine uptake, vaccine effectiveness, and risk of severe COVID-19 outcomes after vaccination in England: a population-based cohort study. Lancet Diabetes Endocrinol. 2022 Aug;10(8):571–580.
  • Kocivnik N, Velnar T. (2022). A review pertaining to SARS-CoV-2 and autoimmune diseases: what is the connection? Life (Basel, Switzerland). 12(11): 1918.
  • Garcillán B, Salavert M, Regueiro JR, et al. Response to vaccines in patients with immune-mediated inflammatory diseases: a narrative review. Vaccines (Basel). 2022;10(2):297.
  • Tang K-T, Hsu B-C, Chen D-Y. Immunogenicity, effectiveness, and safety of COVID-19 vaccines in rheumatic patients: an updated systematic review and meta-analysis. Biomedicines. 2022;10(4):834.
  • Sood A, Tran M, Murthy V, et al. Immunogenicity and safety of SARS-CoV-2 vaccination in patients with rheumatic diseases: a systematic review and meta-analysis. J Clin Rheumatol. 2022;28(8):381–389.
  • Sung K-Y, Chang T-E, Wang Y-P, et al. SARS-CoV-2 vaccination in patients with inflammatory bowel disease: a systemic review and meta-analysis. J Chin Med Assoc. 2022;85(4):421–430.
  • Jena A, James D, Singh AK, et al. Effectiveness and durability of COVID-19 vaccination in 9447 patients with IBD: a systematic review and meta-analysis. Clin Gastroenterol Hepatol. 2022;20(7):1456–1479.e18.
  • Lee ARYB, Wong SY, Tay SH. Booster COVID-19 vaccines for immune-mediated inflammatory disease patients: a systematic review and meta-analysis of efficacy and safety. Vaccines (Basel). 2022 Apr 22;10(5):668.
  • Duly K, Farraye FA, Bhat S. COVID-19 vaccine use in immunocompromised patients: a commentary on evidence and recommendations. Am J Health Syst Pharm. 2022 Jan 5;79(2):63–71.
  • Mathieu E, Ritchie H, Rodés-Guirao L, et al. OurWorldInData: coronavirus Pandemic (COVID-19)- 2020. [Cited 2022 Oct 26]. Available at: https://ourworldindata.org/coronavirus
  • European Medicines Agency. Evusheld: EPAR- assessment report, 2022. [Cited 2022 Oct 26]. Available at: https://www.ema.europa.eu/en/medicines/human/EPAR/evusheld
  • Lafont E, Pere H, Lebeaux D, et al. SARS-CoV-2 treatment is associated with decreased mortality in immunocompromised patients with COVID-19. J Antimicrob Chemother. 2022 Sep 30;77(10):2688–2692.
  • Zhang X, Yuan H, Yang Z, et al. SARS-CoV-2: an updated review highlighting its evolution and treatments. Vaccines (Basel). 2022;10:2145.
  • Yuan Y, Jiao B, Qu L, et al. The development of COVID-19 treatment. Front Immunol. 2023;14:1125246.
  • Gottlieb RL, Vaca CE, Paredes R, et al. GS-US-540–9012 (PINETREE) Investigators. early remdesivir to prevent progression to severe covid-19 in outpatients. N Engl J Med. 2022 Jan 27;386(4):305–315. Epub 2021 Dec 22
  • Widyasari K, Kim J. A review of the currently available antibody therapy for the treatment of coronavirus disease 2019 (COVID-19). Antibodies (Basel). 2023;12(1):5.
  • RECOVERY Collaborative Group. Casirivimab and imdevimab in patients admitted to hospital with COVID-19 (RECOVERY): a randomised, controlled, open-label, platform trial. Lancet. 2022 Feb 12;399(10325):665–676.
  • Gupta A, Gonzalez-Rojas Y, Juarez E, et al. Early treatment for Covid-19 with SARS-CoV-2 neutralizing antibody sotrovimab. N Engl J Med. 2021 Nov 18;385(21):1941–1950. Epub 2021 Oct 27
  • Medicines and Healthcare Products Regulatory Agency. Ronapreve 120 mg/ mL solution for injection or infusion. Summary of product characteristics. 2021 (https://assets.publishing.service.gov.uk/ government/uploads/system/uploads/ attachment_data/file/1012415/revisedgb spcronapreveclean120mg ml12aug2021docx.pdf). government/uploads/system/uploads/ attachment_data/file/1012415/revisedgb spcronapreveclean120mg ml12aug2021docx.pdf).
  • Food and Drug Administration. Fact sheet for health care providers: emergency use authorization (EUA) of REGEN-COV (casirivimab and imdevimab). 2021 https://www.fda.gov/media/145611/download).a
  • Food and Drug Administration. Fact sheet for health care providers: emergency use authorization (EUA) of bamlanivimab and etesevimab. Jan 2022. (https://www.fda.gov/media/145802/download).b
  • Food and Drug Administration. Fact sheet for healthcare providers: emergency use authorization (EUA) of sotrovimab. Feb 2022 (https://www.fda.gov/ media/149534/download).c
  • Food and Drug Administration. Coronavirus (COVID-19) Drugs. Cited Jan 2023. Available at: https://www.fda.gov/drugs/emergency-preparedness-drugs/coronavirus-covid-19-drugs
  • FDA updates Sotrovimab emergency use authorization. Cited January 2023. Available at: https://www.fda.gov/drugs/drug-safety-and-availability/fda-updates-sotrovimab-emergency-use-authorization.
  • Cavazzoni P. Coronavirus (COVID-19) update: FDA limits use of certain monoclonal antibodies to treat COVID-19 due to the omicron variant. Available at: https://www.fda.gov/news-events/press-announcements/coronavirus-covid-19-update-fda-limits-use-certain-monoclonal-antibodies-treat-covid-19-due-omicron.
  • Fact sheet for healthcare providers: emergency Use Authorization for Evusheld™ (tixagevimab co-packaged with cilgavimab). Cited Jan 2023. Available at: https://www.fda.gov/media/154701/download.
  • European Medicines Agency. COVID-19 treatments: under evaluation. Cited Jan 2023. Available at: https://www.ema.europa.eu/en/human-regulatory/overview/public-health-threats/coronavirus-disease-covid-19/treatments-vaccines/treatments-covid-19/covid-19-treatments-under-evaluation#covid-19-treatments-under-marketing-authorisation-evaluation-section.
  • Evusheld Summary of Product Characteristics. Cited January 2023. Available at: https://www.ema.europa.eu/en/documents/product-information/evusheld-epar-product-information_en.pdf.
  • Proal AD, VanElzakker MB. Long COVID or post-acute sequelae of COVID-19 (PASC): an overview of biological factors that may contribute to persistent symptoms. Front Microbiol. 2021 Jun 23;12: 698169.
  • Yomogida K, Zhu S, Rubino F, et al. Sequelae of SARS-CoV-2 infection among adults aged ≥18 years - Long Beach, California, April 1-December 10, 2020. MMWR Morb Mortal Wkly Rep. 2021 Sep 17;70(37):1274–1277.
  • Brannock MD, Chew RF, Preiss AJ, et al. N3C and RECOVER consortia. long COVID risk and Pre-COVID vaccination: an EHR-based cohort study from the recover program. medRxiv [Preprint]. 2022 7;2022(Oct). DOI:10.1101/2022.10.06.22280795.
  • Gao P, Liu J, Liu M. Effect of COVID-19 Vaccines on reducing the risk of long COVID in the real world: a systematic review and meta-analysis. Int J Environ Res Public Health. 2022 Sep 29;19(19):12422.