Skip to main content
Erschienen in: Infectious Diseases and Therapy 4/2022

Open Access 25.05.2022 | COVID-19 | Review

Immune Response to COVID-19 and mRNA Vaccination in Immunocompromised Individuals: A Narrative Review

verfasst von: Norka I. Napuri, Daniel Curcio, David L. Swerdlow, Amit Srivastava

Erschienen in: Infectious Diseases and Therapy | Ausgabe 4/2022

Abstract

Immunocompromised individuals are at high risk of poor coronavirus disease 2019 (COVID-19) outcomes and demonstrate a lower immune response to COVID-19 vaccines, including to the novel mRNA vaccines that have been shown to elicit high neutralizing antibody levels. This review synthesized available data on the immune response to COVID-19 and critically assessed mRNA COVID-19 vaccine immunogenicity in this vulnerable subpopulation. Patients with various immunocompromising conditions exhibit diverse responses to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and COVID-19 severity and mortality, and available vaccines elicit lower immune responses, particularly in solid organ transplant recipients. Strategies to improve vaccine responses in immunocompromised individuals are being implemented in vaccine recommendations, including the use of a third and fourth vaccine dose beyond the two-dose series. Additional doses may enhance vaccine effectiveness and help provide broad coverage against emerging SARS-CoV-2 variants. Continued investigation of vaccines and dosing regimens will help refine approaches to help protect this vulnerable subpopulation from COVID-19.
Hinweise

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Key Summary Points
Despite prioritization of immunocompromised individuals within COVID-19 vaccine protocols, this population remains vulnerable to adverse effects of SARS-CoV-2 infection, including high morbidity and mortality.
Patients with various immunocompromising conditions exhibit diverse responses to SARS-CoV-2 infection, and available vaccines elicit lower immune responses, particularly in solid organ transplant recipients.
Additional COVID-19 vaccine doses may enhance vaccine effectiveness and protect against emerging SARS-CoV-2 variants.
Continued investigation of COVID-19 vaccine dosing and intervals, heterologous vaccination regimens, and breadth of coverage of emerging variants is needed to optimize vaccination recommendations to protect this vulnerable population.

Introduction

Coronavirus disease 2019 (COVID-19) manifests in a range of outcomes, from asymptomatic infection to critical outcomes, including respiratory failure, septic shock, and death [1]. Among those who recover, long-term sequelae may occur, characterized by persistent symptoms or organ dysfunction.
Multiple COVID-19 vaccines across different platforms are under investigation; several are available via emergency use authorization (EUA), including two mRNA vaccines, BNT162b2 and mRNA-1273 (Table 1; [29]), which represent the first use of an mRNA vaccine platform outside of clinical trial settings [1015]. BNT162b2 is now licensed in the United States for those ≥ 16 years old [16] and has EUA for prevention of COVID-19 in those 5–15 years old [17, 18]. mRNA-1273 recently received US licensure for those 18 years and older [19]. Both mRNA vaccines elicit strong humoral responses (i.e., neutralizing antibody production) and strong cellular responses via CD4+ and CD8+ T-cell induction and Th1 cytokine expression [20, 21]. mRNA vaccine-elicited neutralizing antibody titers (NATs) are the highest among the various vaccine platforms [22, 23]. High efficacy for both vaccines was shown in phase 3 trials, and real-world data from their pragmatic use have demonstrated high effectiveness, including against circulating variants [2, 6, 2430].
Table 1
Summary of mRNA COVID-19 vaccines
Vaccine
Characteristics
Dosing
Reported populations
BNT162b2 [36, 8, 9]
Encodes the full length of the SARS-CoV-2 spike, modified by two proline mutations locking it in a prefusion conformation
2 doses at a 21-day interval
Third dose ≥ 28 days after dose 2 in immunocompromised individuals or solid organ transplant recipients 5 years and older
Booster dose ≥ 5 months after primary series in individuals 12 years and older (18 and older for heterologous booster; same interval and dose)
5 years and older
Healthy or with stable chronic medical conditions (e.g., HIV, HBV, HCV infection)a
mRNA-1273 [2, 7]
LNP-encapsulated and expressing the prefusion-stabilized spike
2 doses at a 1-month interval
Third dose ≥ 1 month after dose 2 in immunocompromised individuals or solid organ transplant recipients
Booster dose ≥ 5 months after primary series (same interval and dose for heterologous booster)
18 years and older
In a medically stable condition
COVID-19 coronavirus disease 2019, HBV hepatitis B virus, HCV hepatitis C virus, LNP lipid nanoparticle, SARS-CoV-2 severe acute respiratory syndrome coronavirus 2
aThe studies in participants 5–15 years of age only included healthy participants [3, 5]
In those with immunocompromising conditions, immune responses to infection, treatment, and vaccination are often altered [31]. Data on COVID-19 outcomes in this population are emerging. Results from a UK study of 17 million patients from 2020 suggest that immunocompromised individuals have increased risk of COVID-19–related death [32]. Case reports also indicate that immunocompromised individuals have higher risk of prolonged severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, shedding, and viral evolution [3336]. The severity of COVID-19 outcomes in this population suggests that cytokine storm–mediated effects are occurring in infected patients, although immunosuppression leading to a depressed immunosuppressive immunologic phenotype may be an alternative explanation for this observation [37]. Additionally, because immunocompromised individuals were excluded from initial COVID-19 vaccine trials, vaccination responses in this population are being investigated in observational studies. Therefore, this review synthesized available data on the immune response to COVID-19 and, using a targeted literature search, critically assessed mRNA COVID-19 vaccine immunogenicity in immunocompromised individuals, and considers the available tolerability and safety data from these studies. Data supporting the benefit of additional vaccine doses beyond the two-dose series used initially in this population are also reviewed.

Methods

A targeted search of PubMed and preprint servers (medRxiv and SSRN) was conducted on May 10, 2021, using the following search terms: (coronavirus OR “Severe acute respiratory syndrome” OR covid-19 OR nCoV OR COVID OR SARS OR MERS OR middle east respiratory syndrome) AND (“immune disorders” OR “immunocompromised condition” OR “autoimmune disease” OR “autoimmune disorder” OR “immunosuppressive therapy” OR “immunodeficienc*” OR “chronic inflammatory disease” OR “hematologic malignancy” OR “cancer” OR “hemodialysis” OR transplant OR HIV-1). Reviews were excluded. The following data were extracted from the identified publications: article type, study population (e.g., immunocompromised condition), country, vaccine used (if applicable; type, dosing, and interval), treatments, main immunologic findings (e.g., anti-spike protein immunoglobulin G [IgG] titers, neutralizing antibodies, and cell response), safety and tolerability profile of the vaccine, and any other relevant findings.
This article is based on previously conducted studies and does not contain any new studies with human participants or animals performed by any of the authors.

Outcomes in Specific Immunocompromised Conditions

Malignancy

COVID-19 Characteristics

Large studies and systematic assessments have synthesized the available literature on COVID-19 characteristics in cancer patients. In a US and European community-based survey that included 20,000 cancer patients, risk of COVID-19 test positivity or hospitalization was 60% greater in cancer patients; associations between cancer and COVID-19 increased among older individuals, men, and those receiving cancer treatment [38]. Other analyses support these findings, including reports of increased risk of severe disease, intensive care unit (ICU) admission, and mortality in cancer patients with COVID-19 [3942]. The type of cancer may also confer higher risk of COVID-19-associated death, particularly among patients with lung or hematologic malignancies [31, 40, 42]. Worse outcomes may be associated with recent receipt of cancer therapy, patient age, and male sex.

Vaccination Response

Four studies evaluating COVID-19 vaccine immune responses in 348 patients with hematologic malignancies, including multiple myeloma (MM), chronic lymphocytic leukemia (CLL), and myeloproliferative neoplasms (MPNs), were identified (Table 2) [4346]. Responses were evaluated after two BNT162b2, mRNA-1273, or AZD1222 (a COVID-19 adenovirus vaccine) doses (2 studies; assessed 2–3 weeks post-dose) or one BNT162b2 dose (2 studies; assessed 21 days post-dose). All studies measured IgG-anti-spike protein levels; one also evaluated NATs and T-cell responses.
Table 2
Overview of studies assessing COVID-19 vaccine responses among immunocompromised patients
Reference
Country
Population
Men, %
Median age, years
Therapeutic regimena
Vaccine
Timing of serology assessment
Immunoassays
Antibody response
Vaccine safety
Hematologic malignancies
 Agha et al. [43]
USA
Patients with various hematologic malignancies (n = 67); most commonly MM (43%)
52
71
Cancer-directed therapy within 3 months of treatment
 Observation: 57%
 Active treatment: 43%
BNT162b2
mRNA-1273
PD2 (time period NR)
IgG-anti-spike protein levels
54% (PD2)
NR
 Bird et al. [44]
UK
Patients with MM (n = 93)
59
67
On therapy: 71%
Most common therapy
 Immunomodulatory drug: 47%
 PI: 19%
 Steroid: 45%
 Anti-CD38 mAb: 23%
 Other: 11%
BNT162b2
AZ1222
≥ 21 days PD1
IgG-anti-spike protein levels
56% (PD1)
NR
 Herishanu et al. [45]
Israel
Patients with CLL (n = 167)
67
71
Protocol of currently treated patients
 BTKi: 67%
 Venetoclax ± anti-CD20 mAb: 29%
 Other: 4%
BNT162b2
2–3 weeks PD2
IgG-anti-spike protein levels
40% (PD2)
Mild local reactions: 31% PD1; 34% PD2 (most commonly injection site pain)
Mild systemic events: 13% PD1; 23% PD2 (most commonly weakness)
No significant differences in AE rates between patients actively treated vs. not actively treated at time of vaccination
 Harrington et al. [46]
UK
Patients with myeloproliferative neoplasms (n = 21)
33
58
Peg-IFN: 19%
Surveillance: 33%
Hydroxycarbamide: 19%
Ruxolitinib: 29%
BNT162b2
21 days PD1
IgG-anti-spike protein levels
Neutralizing antibodies
Cell response
86% (PD1)
Localized inflammation: 57%
Systemic events: 48%
Solid tumor malignancies
 Barrière et al. [47]
France
Patients with solid tumors (n = 122); HV (n = 29)
53
69
Chemotherapy ± targeted therapy: 86%
BNT162b2
3–4 weeks PD2
IgG-anti-spike protein levels
48% (PD1)
95% (PD2)
No SAE reported
 Palich et al. [48]
France
Patients with solid tumors (n = 110), which was most commonly breast (34%); HV (n = 25)
40
66
Chemotherapy: 35%
Targeted therapy: 24%
Immunotherapy: 16%
Hormonal therapy: 15%
Radiotherapy: 6%
Surveillance: 16%
BNT162b2
4 weeks PD1
IgG-anti-spike protein levels
58% (PD1)
NR
Chronic inflammatory diseases
 Geisen et al. [52]
Germany
Patients with CID (n = 26); HVs (n = 42)
36
50.5
Biological DMARD: 77%
Conventional DMARD: 31%
Corticosteroid: 27%
BNT162b2
mRNA-1273
7 days PD2
IgG-anti-spike protein levels
Neutralizing antibodies
87% (PD2)
Although side effects were comparable in study groups, patients with CID had marginal propensity towards mild AEs vs. HVs
 Braun-Moscovici et al. [53]
Israel
Patients with CID (n = 264 vaccinated); most commonly RA (37%)
24
57.6
Biological DMARD: 67%
Conventional DMARD: 61%
Colchicine: 0.2%
Nintedanib: 0.1%
Corticosteroid: 35%
BNT162b2
4–6 weeks PD2
IgG-anti-spike protein levels
86% (PD2)
Local reactions: 58%
Fatigue: 30%
Myalgia: 12%
Headache: 20%
Low-grade fever: 3%
 Deepak et al. [54]
USA
Patients with CID (n = 133) compared with immunocompetent controls (n = 53); most commonly IBD (32%) and RA (29%)
26
45.5b
Biologic therapy: 59%
Conventional DMARD: 63%
Prednisone: 13%
Janus kinase inhibitors: 8%
NSAID: 20%
No immunosuppression: 7%
BNT162b2
mRNA-1273
Within 20 days PD2
IgG-anti-spike protein levels
Neutralizing antibodies
89% (PD2)
NR
 Boyarsky et al. [55]
USA
Patients with rheumatic and musculoskeletal diseases (n = 123); most commonly inflammatory arthritis (28%)
5
50
None: 28%
Conventional DMARD: 19%
Biologic DMARD: 14%
Corticosteroid monotherapy: 3%
Combination therapy: 37%
BNT162b2
mRNA-1273
Median 22 days PD1
IgG-anti-spike protein levels
74% (PD1)
NR
 Wong et al. [56]
USA
IBD (n = 48)
48
35.2 (mean)
Biologics: 85%
Oral corticosteroid: 6%
Non therapy: 10%
BNT162b2
mRNA-1273
NR
IgG-anti-spike protein levels
67% (PD1)
100% (PD2)
NR
Hemodialysis
 Schrezenmeier et al. [61]
Germany
Dialysis patients with stage 5 CKD (vaccinated, n = 36; unvaccinated n = 18); controls (n = 44)
69
74
NR
BNT162b2
1–10 weeks PD2
IgG-anti-spike protein levels
Neutralizing antibodies
Cell response
56% (1 week PD2)
89% (3 week PD2)
NR
 Goupil et al. [62]
Canada
Hemodialysis patients (n = 154); controls (n = 40)b
63
70–76 (mean)
Immunosuppressive medications: 16%
BNT162b2
4 weeks PD1
IgG-anti-spike protein levels
43% (PD1; without prior SARS-CoV-2 infection)
84% (PD1; with prior SARS-CoV-2 infection)
NR
 Simon et al. [63]
Austria
Hemodialysis patients (n = 81); controls (n = 80)
55
67 (mean)
RAAS inhibitor: 43%
Immunosuppressants: 11%
Vitamin D: 75%
EPO: 91%
BNT162b2
3 weeks PD2
Neutralizing antibodies
53% (PD2)
No grade 4 AEs
Hemodialysis patients had significantly fewer local and systemic events compared with control group
 Grupper et al. [64]
Israel
Hemodialysis patients (n = 56); HV (n = 95)
75
74
NR
BNT162b2
PD2
IgG-anti-spike protein levels
96% (PD2)
NR
Solid organ transplant
 Boyarsky et al. [75]
USA
SOT (n = 436)
39
55.9
Tacrolimus: 83%
Corticosteroids: 54%
Mycophenolate: 66%
Azathioprine: 9%
Sirolimus: 4%
Everolimus: 2%
BNT162b2
mRNA-1273
17–24 days PD1
Anti-spike protein levels
17% (PD1)
NR
 Sattler et al. [76]
Germany
Kidney transplant (n = 39); HV (n = 39)c
72
57.4 (mean)
CS + TAC + MMF: 56%
CS + CyA + MMF: 33%
mTORi + MMF ± CS: 8%
mTORi + CyA + MMF: 3%
CMV seropositive pre-treatment: 67%
BNT162b2
7–22 days PD2
IgG-anti-spike protein levels
Neutralizing antibodies
Cell response
3% (PD2)
NR
 Itzhaki Ben Zadok et al. [77]
Israel
Heart transplant (n = 42)
83
61
Calcineurin inhibitor: 81%
mTOR inhibitor: 57%
Oral steroid: 69%
Anti-metabolite: 55%
BNT162b2
14–19 days PD2
IgG-anti-spike protein levels
15% (PD1)
49% (PD2)
Injection site pain: 71%
Fatigue: 14%
Arthralgia: 12%
Myalgia: 10%
 Basic-Jukic and Ivo [78]
Croatia
Kidney transplant (< 50 patients received vaccination at the time of the report)
NR
NR
NR
BNT162b2
N/A
NR
NR
NR
 Rusk et al. [79]
USA
Case report of patient with bilateral lung transplant (n = 1)
0
55
Various post-transplant medications including prednisone, mycophenolate, tacrolimus, and alendronate
BNT162b2
16 days PD2
IgG-anti-spike protein levels
0% (PD2)
Mild injection site pain PD1 and PD2
 Grupper et al. [80]
Israel
Kidney transplant (n = 136); HV (n = 25)
82
58.6 (mean)
Calcineurin inhibitor backbone: 90%
Prednisone: 89%
MMF: 76%
Combination regimen: 79%
Prednisone + tacrolimus + MMF: 60%
BNT162b2
10–20 days PD2
IgG-anti-spike protein levels
38% (PD2)
Local pain: 52%
Systemic symptoms: 19%
 Rozen-Zvi et al. [81]
Israel
Kidney transplant (n = 308)
64
57.5
Mycophenolic acid: 73%
Tacrolimus: 93%
Cyclosporine: 8%
mTOR inhibitor: 8%
Corticosteroid: 8%
Rituximab: 2%
Antithymocyte globulin: 5%
BNT162b2
4 weeks PD2
IgG-anti-spike protein levels
36% (PD2)
NR
 Narasimhan et al. [82]
USA
Lung transplant (n = 73)
74
65
Anti-metabolite: 99%
BNT162b2
mRNA-1273
Up to ~ 50 days PD2
IgG-anti-spike protein levels
Cell response
25% (PD2)
NR
 Peled et al. [83]
Israel
Heart transplant (n = 77)
65
62
Mycophenolic acid: 75%
Everolimus: 26%
Prednisone: 75%
BNT162b2
3 weeks PD2
IgG-anti-spike protein levels
Neutralizing antibodies
18% (PD2)
Local reaction: 49–56% (most commonly pain)
Systemic event: 37–49% (most commonly fatigue or headache)
 Rincon-Arevalo et al. [84]
Germany
Kidney transplant (n = 40); HV (n = 35)d
70
62
MMF: 98%
Steroid: 93%
Calcineurin inhibitor: 93%
BNT162b2
7 days PD2
IgG-anti-spike protein levels
Neutralizing antibodies
Cell response
2.5% (PD2)
NR
HIV
 Levy et al. [94]
Israel
PLWH (n = 143); HV (n = 261)
92
49.8 (mean)
ART: 100%
BNT162b2
2–3 weeks PD2
IgG-anti-spike protein levels
Neutralizing antibodies
97% (PD2)
Local reaction: 26–41% (most commonly mild pain)
Systemic event: 20–48% (most commonly fatigue and headache PD1 and fatigue and fever PD2)
AE adverse event, ART antiretroviral therapy, BTKi Bruton’s tyrosine kinase inhibitor, CID chronic inflammatory disease, CKD chronic kidney disease, CLL chronic lymphocytic leukemia, COVID-19 coronavirus disease 2019, CS + CyA + MMF corticosteroids + cyclosporin A + mycophenolate mofetil, CS + Tac + MMF corticosteroids + tacrolimus + mycophenolate mofetil, DMARD disease-modifying antirheumatic drug, EC50 50% effective concentration, EPO erythropoietin, HV healthy volunteer, IBD inflammatory bowel disease, IgG immunoglobulin G, mTORi + CyA + MMF mTOR inhibitor + cyclosporin A + mycophenolate mofetil, mTORi + MMF ± CS mTOR inhibitor + mycophenolate mofetil ± corticosteroids, N/A not applicable, NR not reported, NSAID nonsteroidal anti-inflammatory drug, mAb monoclonal antibody, MM multiple myeloma, MMF mycophenolate mofetil or mycophenolate sodium, mTOR mammalian target of rapamycin, PD1 post-dose 1, PD2 post-dose 2, Peg-IFN pegylated interferon, PI protease inhibitor, PLWH people living with HIV, RA rheumatoid arthritis, RAAS renin–angiotensin–aldosterone system, SARS-CoV-2 severe acute respiratory syndrome coronavirus 2, SAE serious adverse event, SOT solid organ transplant, TKI tyrosine kinase inhibitor
aMay not total 100% because patients could receive ≥ 1 class of treatment
bIncluded participants with and without prior SARS-CoV-2 infection
cStudy also included 26 patients with kidney failure undergoing hemodialysis
dStudy also included patients on maintenance hemodialysis (n = 41), and peritoneal dialysis (n = 4)
After two mRNA vaccine doses, humoral responses ranged from 40% in CLL patients to 54% in patients with various hematologic malignancies [43, 45]. In a study in CLL patients that included comparison to sex- and age-matched healthy controls (HCs), antibody-mediated response to mRNA vaccination was significantly reduced in patients (52% vs. 100%; adjusted odds ratio [OR] = 0.010; P < 0.001) [45]. In the study assessing NATs and cellular responses, 86% of MPN patients had positive neutralizing antibodies and 80% memory T-cell responses, with CD4+ and CD8+ T-cell responses in 75% and 35% of patients, respectively, after one BNT162b2 dose [46]. In this study, no significant differences in humoral or T-cell responses were observed between those receiving active treatment and those undergoing active surveillance.
Two studies comprising a total of 232 patients with solid tumors were identified; both compared BNT162b2 responses to HCs (Table 2) [47, 48]. Responses were evaluated after one dose (1 study; assessed 4 weeks post-dose) or two doses (1 study; assessed 3–4 weeks post-dose); both only evaluated IgG-anti-spike protein levels. Across both studies, approximately 50% (58/122; 64/110) of patients had IgG-anti-spike protein responses post-dose 1. In the study assessing response post-dose 2, the percentage of responders among evaluable patients increased to 95% (n = 40) [47]. After adjustment for potential confounders, one study found older age (OR = 3.58; P = 0.008) and chemotherapy receipt (OR = 4.34; P = 0.003) were strongly associated with lack of seroconversion among patients [48].
Three studies in patients with malignancies reported on the safety and tolerability profile of BNT162b2 [4547]. In a study in patients with CLL who received BNT162b2, mild local reactions were reported in 31% and 34% of patients after dose 1 and 2, respectively, and were most commonly related to injection site pain [45]. Systemic events were reported in 13% and 23% of patients after dose 1 and 2, respectively, and were most commonly related to weakness. No significant differences in adverse event (AE) frequencies were observed between patients receiving active CLL treatment compared with those who were not. Finally, in a study in patients with MPNs, localized inflammation was reported in 57% and systemic events (e.g., flu-like illness, fatigue, gastrointestinal symptoms) in 48% of patients after receipt of one BNT162b2 dose [46]. In a study in patients with various solid tumors, no serious AEs (SAEs) were reported among BNT162b2 recipients [47].

Chronic Inflammatory Diseases

COVID-19 Characteristics

Chronic inflammatory diseases (CIDs) are associated with increased infection risk, although immunosuppressive treatments might limit the cytokine storm associated with severe COVID-19 [49]. Consistent with this hypothesis, a cohort of 6792 US patients with and without CIDs found that CID patients with COVID-19 were more likely to be hospitalized, but having CID was not an independent risk factor for severe COVID-19 [49]. Patients receiving corticosteroids had increased severe disease risk, while biologic or non-biologic immunosuppressive treatments were not significantly associated with this risk. Consistent with this finding, a prospective study of 103 inflammatory arthritis (IA) patients found that COVID-19 outcomes were worse in those receiving corticosteroids compared with those receiving anti-cytokines [50]. The type of CID might also be associated with severe COVID-19. In a study of 228 hospitalized patients with IA or connective tissue diseases, the latter was associated with severe disease and poor outcomes [51].

Vaccination Response

Five studies describing immunogenicity responses in 594 CID patients receiving mRNA vaccines were identified; a range of CIDs were included, most commonly rheumatoid arthritis, IA, and inflammatory bowel disease (IBD) (Table 2) [5256]. Immune responses were evaluated after two BNT162b2 or mRNA-1273 doses (4 studies; assessed 1–6 weeks post-dose) or one BNT162b2 or mRNA-1273 dose (1 study; assessed ~ 3 weeks post-dose). All studies evaluated IgG-anti-spike protein levels; two evaluated NATs. Cellular immune responses were not assessed.
Humoral responses in CID patients were observed after one (67–74%) or two (86–100%) doses [5256]. In two studies that included HCs, IgG-anti-spike protein levels and NATs were lower in CID patients versus HCs, with B-cell–depleting and corticosteroid therapies associated with reduced responses [52, 54]. In patients who received B-cell–depleting therapies, the lack of vaccine response was predominantly observed in those who had recently received therapy. One study of patients with IA treated with disease-modifying antirheumatic drugs (DMARDs) found that B-cell–depleting treatments were associated with significantly lower immune responses to vaccination [53]. Two other studies also analyzed immune responses to vaccination according to CID treatment. In patients with CID who received BNT162b2 or mRNA-1273, 94% of those receiving anti-tumor necrosis factor (TNF) therapy had immune responses to vaccination [55]. In a study in patients with IBD, receipt of anti-TNF or vedolizumab therapy was associated with decreased humoral responses compared with HCs (anti-TNF: anti–receptor-binding domain [RBD] total immunoglobulin; vedolizumab: anti-RBD total immunoglobulin, anti-RBD IgG, and anti-spike protein IgG) [56].
Two studies in patients with CID described the safety and tolerability profile after vaccination [52, 53]. In one study, 58% of patients experienced a local reaction, and fatigue (30%) and headache (20%) were the most common systemic events after receipt of BNT162b2 [53]. One patient reported new-onset arthritis 2 months after vaccination, but no patients experienced a flare of inflammatory rheumatic disease at up to 2 months of follow-up. In the second study, in which patients received either BNT162b2 or mRNA-1273, patients with CID experienced generally milder AEs compared with HCs, with mild fatigue (54% vs. 43%) and myalgia (42% vs. 32%) more frequent in patients, and fever more common in HCs (0% vs. 14%) [52].

Hemodialysis

COVID-19 Characteristics

Hemodialysis recipients may be at greater COVID-19 risk because of immunosuppression and the older age, comorbidities, and frequent healthcare visits common in this population [5759]. In a European registry study of 4298 kidney transplant and dialysis patients, COVID-19 incidence was slightly increased in dialysis patients versus the general population, which was attributed to the older age or more frequent SARS-CoV-2 testing undertaken in patients [59]. Other studies support these findings. A systematic review of 396,062 hemodialysis patients (COVID-19 cases, n = 3261) found a 15-fold increase in COVID-19 incidence and greater mortality risk in patients versus the general population [57]. Mortality risk may be lower in hemodialysis patients from Asia, which is potentially associated with their younger age and diagnostic approaches in this region allowing for timely intervention [57]. In another systematic review of > 1.1 million patients with chronic kidney disease (CKD), COVID-19 incidence was higher in patients receiving maintenance dialysis [60].

Vaccination Response

Four studies assessed responses to BNT162b2 vaccination in 325 hemodialysis recipients (Table 2) [6164]. All included comparator groups (vaccinated vs. unvaccinated patients or HCs). Three reported immune responses post-dose 2 (assessed 1–10 weeks post-dose); the fourth assessed responses 4 weeks post-dose 1. All evaluated IgG-anti-spike protein levels. One study also assessed NATs and cellular immune responses.
Humoral responses in hemodialysis recipients were 43–84% and 53–96% after one and two BNT162b2 doses, respectively; responses were lower in patients versus HCs [6164]. One study of 154 patients receiving hemodialysis found that humoral responses were dependent on past SARS-CoV-2 infection, with patients previously infected mounting a delayed antibody response post-dose 1 [62]. Delayed responses were also reported in some patients who received two doses [63]. In this study, men had lower median titers versus women, while increasing age was inversely associated with titer level. In a study of two doses, the neutralizing antibody capacity was 78% (28/36) and SARS-CoV-2 T cells were detected in 68% of patients (21/31) post-dose 2, supportive of vaccine-induced B- and T-cell responses [61]. Another study found older age to be significantly associated with decreased antibody levels in hemodialysis recipients [64]. One study found that receipt of immunosuppressive medication did not appear to affect the immune response to vaccination [62].
One study in hemodialysis recipients described the safety and tolerability profile of BNT162b2 [63]. No grade 4 AEs were reported, and hemodialysis recipients had significantly fewer local and systemic events compared with HCs after either vaccine dose.

Solid Organ Transplant Recipients

COVID-19 Characteristics

The weakened T-cell–mediated immune response of solid organ transplant recipients (SOTRs) makes them vulnerable to many viral infections, although increased vulnerability to Middle East respiratory syndrome (MERS) and severe acute respiratory syndrome (SARS) has not been reported [65]. Immunosuppressive treatments and comorbidities common in this population may also increase vulnerability to infection, causing substantial morbidity and mortality [66, 67]. Systematic reviews have assessed morbidity in SOTRs. One that included 22 studies found that among SOTRs with COVID-19, 81% were hospitalized, of which 29% required ICU admission and 26% mechanical ventilation [65]. These rates are consistent with those in kidney and liver transplant recipients [6870]. Among 415 heart transplant recipients, 34% had severe and 14% critical COVID-19, while 23% required ICU admission and 16% mechanical ventilation [71]. In a systematic review of SOTRs that included 37 articles, all-cause mortality was 19%, which varied by transplant type [65]. However, mortality is generally consistent across transplantation type, including kidney and liver transplant recipients [6870, 72, 73]. COVID-19–associated mortality of 24–26% was reported in two studies of 39 and 415 heart transplant recipients, respectively; diabetes mellitus and stage 3 or higher CKD were associated with increased mortality in one study [71, 74]. A systematic review in 5559 kidney transplant recipients found those who died from COVID-19 were older, had a shorter time since transplantation, or were living in Asia–Pacific or Europe (vs. the United States) [68].

Vaccination Response

Nine studies and one case report of vaccine responses in approximately 1200 SOTRs were identified and included kidney, heart, and lung transplantation recipients (Table 2) [7584]. Responses were evaluated after two (8 studies; assessed 7 to ~ 50 days post-dose) or one dose (1 study; assessed 17–24 days post-dose) of BNT162b2 or mRNA-1273; one study did not specify schedule or response period. Nine evaluated IgG-anti-spike protein levels, three NATs, and three cellular responses.
Among these studies, < 50% of SOTRs had immune response to vaccination, with limited development of neutralizing antibodies and diminished cellular responses [7577, 79, 8184]. Two patients developed COVID-19 after the second BNT162b2 dose in a study of < 50 kidney transplant recipients [78]. Patient age, longer maintenance dialysis duration and lower estimated glomerular filtration rate (for kidney transplant recipients) pre-transplantation, and specific COVID-19 vaccine administered were factors associated with poor responses [75, 77, 80, 81, 83]. Factors associated with poor responses also included use of maintenance antimetabolite immunosuppressive therapy and type of immunosuppressant regimen received [7577, 80, 81, 83]. In two studies, those receiving antimetabolite immunosuppressive therapy were less likely to develop an antibody response [75, 77]. In two studies in kidney transplant recipients, multivariate analyses found receipt of high-dose corticosteroids, maintenance triple immunosuppressive medications, mycophenolate mofetil (MMF)–containing regimens, and mammalian target of rapamycin (mTOR) inhibitors were associated with poor humoral responses [80, 81]. Another study in heart transplant recipients found that patients with a high antibody response to vaccination had a lower use of MMF and higher use of mTOR inhibitors compared with patients with low antibody responses [83]. However, a study in kidney transplant recipients found immune responses did not differ significantly between patients receiving tacrolimus and cyclosporin A [76].
Three studies in SOTRs described safety and tolerability after BNT162b2 administration [77, 79, 83]. In two studies in heart transplant recipients, local reactions occurred in 49–76% and systemic events in 37–49% of patients, which were most commonly injection site pain, fatigue, headache, arthralgia, and myalgia [77, 83]. In a study in kidney transplant recipients, local pain was experienced by 52% and systemic symptoms by 19% of patients after vaccination, and there was a similar reactogenicity rate between patients and HCs [80].

HIV

COVID-19 Characteristics

Because HIV infection is associated with abnormal humoral and T-cell–mediated immune responses, people living with HIV (PLWH) may be at increased risk of adverse COVID-19 outcomes [85, 86]. However, determining effects of SARS-CoV-2 infection in PLWH is complicated by the potential benefit of antiretroviral (ART) and protease inhibitor therapies, some of which have been investigated as COVID-19 treatments; PLWH may also be less susceptible to the cytokine storm associated with severe COVID-19 because of dysfunctional immune systems [87]. Systematic reviews have assessed epidemiology and effects of COVID-19 in PLWH. Two systematic reviews with approximately 850,000 PLWH reported 0.7–0.9% COVID-19 and 5.3–8.8% mortality rates; the hospitalization rate was 28%, which included 2.5% who were severe-critical and 3.5% requiring ICU care [88, 89]. Some systematic reviews found no apparent correlation with concurrent HIV and SARS-CoV-2 infection and severe disease and adverse outcomes, although conflicting findings were reported [85, 86, 8991]. One review of > 89,000 patients with coinfection found those with advanced HIV stages had less severe COVID-19 and lower mortality versus patients with earlier stages, although some studies provided conflicting findings [92]. Other systematic reviews, which included 23–63 studies, found that men with coinfection had higher morbidity and mortality; higher morbidity and mortality from COVID-19 was also reported in patients who had multiple comorbidities, were older, or had high viral load [85, 87, 93]. In other systematic reviews, which included 14–82 studies, the effect of ART in reducing risk of infection and COVID-19–associated mortality was inconclusive [8789, 91].

Vaccination Response

One study was identified assessing response to BNT162b2 in PLWH, which evaluated IgG-anti-spike protein and neutralizing antibody responses (Table 2) [94]. Immunocompetent healthcare workers were included as controls; responses were assessed 2–3 weeks post-dose 2. Overall, 18% (26/143) of patients had AIDS and all were receiving ART. The mean (range) of nadir CD4+ T cells was 345 (2–900)/µL, and 10% and 18% of PLWH had nadir CD4+ T cells < 100 cells/µL and < 200 cells/µL, respectively.
In this study, anti–receptor-binding domain IgG was positive and NATs developed in 97% of patients each after dose 2 [94]. In an adjusted linear regression analysis, PLWH had significantly lower IgG concentrations than HCs (P = 0.008). The population included in the study had a high CD4+ cell count at baseline; of the three patients with a CD4+ count < 200 cells/µL, all had high immune responses to BNT162b2. However, a statistically significant decrease from baseline in CD4+ counts after dose 1 and 2 (P < 0.0001) was found, which persisted up to 4 months after dose 2 and was not associated with clinical changes.
In PLWH, local reactions were more common after the first BNT162b2 dose (41% vs. 26%), while systemic events were more common after the second dose (20% vs. 48%) [94]. Injection site pain was the most common local reaction, which was mild and resolved within 24 h. Fatigue and headache were the most common systemic events after dose 1, and fatigue and fever after dose 2. No immediate or delayed hypersensitivity reactions were observed.

Additional COVID-19 Vaccine Doses for Immunocompromised Individuals

With COVID-19 vaccination programs initiated since December 2020, the need for additional doses was soon debated, including whether required for all individuals, timing post-primary series, and whether booster and primary vaccine matching is needed [95]. Differing recommendations regarding the need for a third and fourth dose have resulted, with several countries/regions issuing recommendations for additional doses in immunocompromised individuals (Table 3; [96102]).
Table 3
Recommendations for additional COVID-19 vaccine doses in immunocompromised individuals
Country or region
Issuing body
Date of recommendation
Recommendation
France [98, 99]
Government of France
April 11, 2021
Third dose 4 weeks PD2 in severely immunocompromised individuals
 
Government of France
January 28, 2022
Administration of a fourth dose in severely immunocompromised individuals within 3 months of the third dose
Israel [100]
 
December 30, 2021
Fourth dose in individuals who have undergone organ or stem cell transplants, with hematologic cancers, autoimmune diseases, and who are receiving specific immunosuppressive medications
Those with breast, lung, or colon cancer do not qualify
Global [96]
WHO
October 26, 2021
Additional dose recommended in moderately to severely immunocompromised individuals 1–3 months after the primary series. This recommendation applies to individuals with active cancer, solid organ and stem cell transplant recipients, individuals with immunodeficiency, HIV, and those receiving immunosuppressive therapies
Homologous additional doses are recommended, but heterologous platforms may be considered because of vaccine supply and access
USA [97]
CDC
February 17, 2022
Individuals 5–11 years old who are moderately or severely immunocompromised should receive a primary series of three doses of BNT162b2, with the second dose given 3 weeks after the first dose and the third dose given ≥ 4 weeks after the second dose; a fourth dose is not recommended at this time
Individuals 12 years and older (for BNT162b2) and 18 years and older (for mRNA-1273) who are moderately or severely immunocompromised should receive a third dose ≥ 4 weeks after receiving their second dose; a booster dose ≥ 3 months after the third dose is recommended; mRNA COVID-19 vaccines are preferred
This recommendation extends to individuals receiving cancer treatment, who have received an organ transplant and are using immunosuppressive therapies, who received a stem cell transplant in the past 2 years or are taking immunosuppressive therapies, with moderate or severe primary immunodeficiency, with advanced or untreated HIV infection, or receiving active treatment with high-dose corticosteroids or other immunosuppressive therapies
EU [101]
EMA
October 4, 2021
An additional dose of BNT162b2 or mRNA-1273 may be given to individuals with severely weakened immune systems ≥ 28 days after the second dose
UK [102, 103]
MHRA
September 9, 2021
One dose of BNT162b2 may be administered as a third dose ≥ 8 weeks after the second dose of an mRNA or adenovirus-vectored COVID-19 vaccine if the potential benefits are greater than any potential risks
 
UK Health Security Agency
February 11, 2022
For those 12 years and older, a reinforcing dose 3 months after the third dose is recommended
CDC US Centers for Disease Control and Prevention, COVID-19 coronavirus disease 2019, EMA European Medicines Agency, MHRA Medicines and Healthcare Products Regulatory Agency, PD2 post-dose 2, SOT solid organ transplant, WHO World Health Organization
Current US recommendations were based on the US Food and Drug Administration (FDA) granting EUA for mRNA vaccines allowing administration of a third dose in SOTRs and other individuals with comparable immunocompromised status on August 12, 2021 [103]. On August 16, 2021, the US Centers for Disease Control and Prevention (CDC) recommended that moderately to severely immunocompromised adults receive an additional mRNA vaccine dose [104]; since then, the recommendation has been extended to include immunocompromised children 5 years and older [105]. The initial recommendation was based on limited serologic evidence from two small, randomized controlled trials. In a study of 50 hemodialysis recipients, five of whom had prior COVID-19, three BNT162b2 doses were administered to those without prior infection and two to those previously infected [106]. In the latter, 89% (40/45) had antibody responses post-dose 2 and 93% (42/45) post-dose 3; two who did not respond to dose 2 had robust responses post-dose 3. Antibody responses after three doses in those not previously infected were comparable to those measured post-dose 2 in patients with a COVID-19 history. In a study of 120 SOTRs who received two mRNA-1273 doses, a third mRNA-1273 dose or placebo was administered 2 months later [107]. One month post-dose 3, 55% of mRNA-1273 and 18% of placebo recipients had serologic responses (P < 0.001); the median percent virus neutralization was 71% and 13%, respectively. In their recommendations, the CDC cited evidence that immunocompromised individuals (e.g., those with hematologic cancers or on hemodialysis or receiving certain immunosuppressive therapies [108]) do not always have the same degree of immune response to vaccination [105].
Additional evidence has accumulated in systematic reviews of data regarding the immune response to COVID-19 vaccination and vaccine efficacy among immunocompromised subpopulations [109]. While the majority of the data come from mRNA vaccines, there is some evidence from other platforms, such as viral vector and inactivated whole virus vaccines, showing diminished immune responses to vaccination and reduced vaccine efficacy among immunocompromised individuals, thereby making the case for administering additional doses to help protect against COVID-19 [96]. In line with this accumulating evidence, various public health agencies have updated their recommendations. For instance, the CDC recommends that moderately or severely immunocompromised adults who have received one dose of the Ad26.COV2.S viral vector vaccine should receive two subsequent doses of an mRNA vaccine (dose 2 at ≥ 4 weeks post-dose 1 and dose 3 at ≥ 2 months post-dose 2) [97]. Similarly, the UK Joint Committee on Vaccination and Immunisation (JCVI) recommends a preference for mRNA vaccines for the third vaccine dose in immunocompromised individuals, while the AZD1222 adenovirus vaccine may be an option for those 16 years and older who have received this vaccine previously and in whom the mRNA vaccines are contraindicated [102].
Emerging evidence for increased seropositivity rates in immunocompromised individuals following a third mRNA vaccine dose is summarized in Fig. 1. In studies that included SOTRs and hemodialysis patients, 33–50% of immunocompromised individuals who were seronegative to the initial two-dose series developed antibody responses to a third dose administered approximately 1–3 months post-dose 2 [110113]. The MELODY (Mass Evaluation of Lateral flow immunOassays for the Detection of SARS-CoV-2 antibodY responses in immunosuppressed people) study, which opened in December 2021, aims to evaluate antibody responses to a third COVID-19 vaccine dose in SOTRs, individuals with hematologic malignancies, and individuals with autoimmune disease [114]. These data may help inform vaccination strategies and future recommendations in immunocompromised individuals.

Discussion

Our literature review shows reduced antibody responses to one or two mRNA vaccine doses in patients with a range of immunocompromising conditions, although this diminished response appears more pronounced in some conditions, particularly SOTRs (Fig. 2). Although responses to two doses versus one dose were higher, even two-dose regimens generally provided diminished responses. Our review also found a generally consistent safety and tolerability profile of mRNA vaccines in immunocompromised individuals compared with the profile in other adult populations.
The findings from our review suggest that despite prioritizing immunocompromised individuals within COVID-19 vaccine protocols, this population remains vulnerable to adverse effects of SARS-CoV-2 infection, including high morbidity and mortality. One approach to improve protection against COVID-19 in immunocompromised individuals is to administer additional vaccine doses. This is supported by emerging clinical data showing enhanced antibody response without detrimental safety and tolerability effects post-dose 3 in immunocompromised adults [106, 110113]. Additional doses in this population are now recommended in some countries and regions.
Importantly, the two-dose schedule for mRNA vaccines administered approximately 3–4 weeks apart was implemented in initial clinical trials and within mass vaccination programs in response to the rapidly evolving pandemic. As we learn more regarding transmission, infection, and disease, as well as the characteristics of SARS-CoV-2 variants and the evolving epidemiology among various populations, it is anticipated that vaccine schedules and recommendations, including among immunocompromised individuals, will evolve.
We have considered transferable lessons from other vaccines used in immunocompromised individuals in the context of COVID-19 vaccination in this population, as this population is also recommended to receive immunization against several infectious diseases to reduce infection-associated morbidity and mortality [115]. Unfortunately, there is low vaccine uptake and limited data on safety, efficacy, effectiveness, and immunogenicity of vaccines in immunocompromised individuals. Accordingly, strategies to overcome these limitations have been investigated. For instance, to protect immunocompromised individuals against influenza, the timing of vaccination, administration of two doses in an influenza season, vaccine formulation modifications, and vaccination of close contacts of the patient and healthcare workers have been proposed [116].
Further investigation to optimize COVID-19 vaccination recommendations for immunocompromised individuals is required, including consideration of timing, immunosuppression protocols, dosing and intervals, heterologous primary and booster vaccination regimens, and breadth of coverage of emerging SARS-CoV-2 variants including omicron. Additionally, identification of an absolute correlate of protection for COVID-19 vaccines will be extremely helpful in the further development and implementation of these vaccines, although neutralizing antibody responses are the most promising candidate [117], and were used extensively in the studies included within our review. Given the small study sizes identified within our review effort, larger prospective trials in this population will be informative, and several such studies in both adults and children are ongoing (Table 4). With the remarkable global uptake of COVID-19 vaccinations, with > 10 billion doses administered as of February 28, 2022 [118], it is also anticipated that real-world effectiveness data in these vulnerable populations will emerge.
Table 4
Clinical trials evaluating mRNA COVID-19 vaccines in immunocompromised individualsa
Vaccine
Clinical trial identifier
Details
Status
BNT162b2
NCT04952766
Phase 4 study assessing humoral response against SARS-CoV-2 after vaccination in immunocompromised adults compared with healthy volunteers
Recruiting (estimated primary completion March 2022)
BNT162b2
NCT04895982
Phase 2 study assessing a three-dose schedule in immunocompromised children and adults
Recruiting
BNT162b2
NCT04969601
Phase 1/2 study assessing a two-dose schedule in children 1–15 years of age with acute leukemia and in their siblings (12–15 years of age)
Recruiting (estimated primary completion May 2022)
BNT162b2 mRNA-1273
NCT04805125
Phase 3 study using Swiss cohorts of immunocompromised patients to assess the comparative effectiveness and safety of BNT162b2 and mRNA-1273
Ongoing (estimated completion July 2022)
BNT162b2
NCT04844489
Study to assess the humoral and cellular responses to vaccination against SARS-CoV-2 variants in immunocompromised individuals
Recruiting
mRNA-1273
NCT04847050
Phase 2 study assessing the safety and immunogenicity of vaccination in adults with hematologic malignancies, various regimens of immunosuppression, and in participants with solid tumors on PD1/PDL1 inhibitor therapy
Recruiting (estimated primary completion June 2023)
COVID-19 coronavirus disease 2019, PD1 post-dose 1, PD2 post-dose 2, SARS-CoV-2 severe acute respiratory syndrome coronavirus 2
aAs of January 13, 2022
Limitations of the data set from our literature review should be noted. For instance, several immunocompromising conditions were included, and within each study, the assessed populations were generally heterogeneous, limiting assessment of immune responses to disease and vaccination between subgroups (e.g., by type of cancer or CID or by stage of disease including patients undergoing flares or in remission). A comprehensive assessment of the association between these factors and immune responses would require large data sets over time.

Conclusion

Patients with various immunocompromising conditions exhibit diverse responses to SARS-CoV-2 infection and COVID-19 severity and mortality, and available vaccines elicit lower immune responses but have a similar safety and tolerability profile to that observed in other adult populations. Additional doses may enhance vaccine effectiveness and protect against emerging variants. Continued investigation will help refine approaches to help protect this vulnerable population.

Acknowledgements

Funding

This work was supported by Pfizer Inc. Pfizer Inc also funded the journal’s Rapid Services Fee.

Medical writing, editorial, and other assistance

Editorial/medical writing support was provided by Tricia Newell, PhD, and Allison Gillies, PhD, of ICON (Blue Bell, PA, USA) and was funded by Pfizer Inc.

Authorship

All named authors meet the International Committee of Medical Journal Editors (ICMJE) criteria for authorship for this article, take responsibility for the integrity of the work as a whole, and have given their approval for this version to be published.

Author contributions

All authors contributed to the conceptualization of this work. The literature search and data analysis were performed by Daniel Curcio, Amit Srivastava, and Tricia Newell. The first draft of the manuscript was written by Tricia Newell under direction from the authors, and all authors commented on previous versions of the manuscript. All authors read and approved the final manuscript.

Disclosures

Norka I. Napuri, Daniel Curcio, and Amit Srivastava are employees of Pfizer Inc and David L. Swerdlow is a retired employee of Pfizer Inc, and all authors may hold stock or stock options. David L. Swerdlow was an employee of Pfizer Inc at the initiation of the manuscript.

Compliance with ethics guidelines

This article is based on previously conducted studies and does not contain any new studies with human participants or animals performed by any of the authors.

Data availability

This article is based on published literature and therefore does not contain any applicable data sets.
Open AccessThis article is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License, which permits any non-commercial use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by-nc/​4.​0/​.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
2.
Zurück zum Zitat Baden LR, El Sahly HM, Essink B, et al. Efficacy and safety of the mRNA-1273 SARS-CoV-2 vaccine. N Engl J Med. 2021;384:403–16.PubMedCrossRef Baden LR, El Sahly HM, Essink B, et al. Efficacy and safety of the mRNA-1273 SARS-CoV-2 vaccine. N Engl J Med. 2021;384:403–16.PubMedCrossRef
3.
Zurück zum Zitat Walter EB, Talaat KR, Sabharwal C, et al. Evaluation of the BNT162b2 Covid-19 vaccine in children 5 to 11 years of age. N Engl J Med. 2022;386:35–46.PubMedCrossRef Walter EB, Talaat KR, Sabharwal C, et al. Evaluation of the BNT162b2 Covid-19 vaccine in children 5 to 11 years of age. N Engl J Med. 2022;386:35–46.PubMedCrossRef
5.
Zurück zum Zitat Frenck RW Jr, Klein NP, Kitchin N, et al. Safety, immunogenicity, and efficacy of the BNT162b2 Covid-19 vaccine in adolescents. N Engl J Med. 2021;385:239–50.PubMedCrossRef Frenck RW Jr, Klein NP, Kitchin N, et al. Safety, immunogenicity, and efficacy of the BNT162b2 Covid-19 vaccine in adolescents. N Engl J Med. 2021;385:239–50.PubMedCrossRef
6.
Zurück zum Zitat Polack FP, Thomas SJ, Kitchin N, et al. Safety and efficacy of the BNT162b2 mRNA Covid-19 vaccine. N Engl J Med. 2020;383:2603–15.PubMedCrossRef Polack FP, Thomas SJ, Kitchin N, et al. Safety and efficacy of the BNT162b2 mRNA Covid-19 vaccine. N Engl J Med. 2020;383:2603–15.PubMedCrossRef
9.
Zurück zum Zitat Walsh EE, Frenck RW Jr, Falsey AR, et al. Safety and immunogenicity of two RNA-based Covid-19 vaccine candidates. N Engl J Med. 2020;383:2439–50.PubMedCrossRef Walsh EE, Frenck RW Jr, Falsey AR, et al. Safety and immunogenicity of two RNA-based Covid-19 vaccine candidates. N Engl J Med. 2020;383:2439–50.PubMedCrossRef
16.
Zurück zum Zitat 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;397:1646–57.PubMedPubMedCentralCrossRef 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;397:1646–57.PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat Sahin U, Muik A, Derhovanessian E, et al. COVID-19 vaccine BNT162b1 elicits human antibody and TH1 T cell responses. Nature. 2020;586:594–9.PubMedCrossRef Sahin U, Muik A, Derhovanessian E, et al. COVID-19 vaccine BNT162b1 elicits human antibody and TH1 T cell responses. Nature. 2020;586:594–9.PubMedCrossRef
21.
Zurück zum Zitat Jackson LA, Anderson EJ, Rouphael NG, et al. An mRNA vaccine against SARS-CoV-2—preliminary report. N Engl J Med. 2020;383:1920–31.PubMedCrossRef Jackson LA, Anderson EJ, Rouphael NG, et al. An mRNA vaccine against SARS-CoV-2—preliminary report. N Engl J Med. 2020;383:1920–31.PubMedCrossRef
22.
Zurück zum Zitat Khoury DS, Cromer D, Reynaldi A, et al. Neutralizing antibody levels are highly predictive of immune protection from symptomatic SARS-CoV-2 infection. Nat Med. 2021;27:1205–11.PubMedCrossRef Khoury DS, Cromer D, Reynaldi A, et al. Neutralizing antibody levels are highly predictive of immune protection from symptomatic SARS-CoV-2 infection. Nat Med. 2021;27:1205–11.PubMedCrossRef
23.
Zurück zum Zitat Earle KA, Ambrosino DM, Fiore-Gartland A, et al. Evidence for antibody as a protective correlate for COVID-19 vaccines. Vaccine. 2021;39:4423–8.PubMedPubMedCentralCrossRef Earle KA, Ambrosino DM, Fiore-Gartland A, et al. Evidence for antibody as a protective correlate for COVID-19 vaccines. Vaccine. 2021;39:4423–8.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Haas EJ, Angulo FJ, McLaughlin JM, et al. Impact and effectiveness of mRNA BNT162b2 vaccine against SARS-CoV-2 infections and COVID-19 cases, hospitalisations, and deaths following a nationwide vaccination campaign in Israel: an observational study using national surveillance data. Lancet. 2021;397:1819–29.PubMedPubMedCentralCrossRef Haas EJ, Angulo FJ, McLaughlin JM, et al. Impact and effectiveness of mRNA BNT162b2 vaccine against SARS-CoV-2 infections and COVID-19 cases, hospitalisations, and deaths following a nationwide vaccination campaign in Israel: an observational study using national surveillance data. Lancet. 2021;397:1819–29.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Abu-Raddad LJ, Chemaitelly H, Butt AA, National Study Group for COVID-19 Vaccination. Effectiveness of the BNT162b2 Covid-19 vaccine against the B.1.1.7 and B.1.351 variants. N Engl J Med. 2021;385:187–9.PubMedCrossRef Abu-Raddad LJ, Chemaitelly H, Butt AA, National Study Group for COVID-19 Vaccination. Effectiveness of the BNT162b2 Covid-19 vaccine against the B.1.1.7 and B.1.351 variants. N Engl J Med. 2021;385:187–9.PubMedCrossRef
26.
Zurück zum Zitat Lopez Bernal J, Andrews N, Gower C, 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.PubMedCrossRef Lopez Bernal J, Andrews N, Gower C, 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.PubMedCrossRef
27.
Zurück zum Zitat Sheikh A, McMenamin J, Taylor B, Robertson C. SARS-CoV-2 Delta VOC in Scotland: demographics, risk of hospital admission, and vaccine effectiveness. Lancet. 2021;397:2461–2.PubMedPubMedCentralCrossRef Sheikh A, McMenamin J, Taylor B, Robertson C. SARS-CoV-2 Delta VOC in Scotland: demographics, risk of hospital admission, and vaccine effectiveness. Lancet. 2021;397:2461–2.PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Thompson MG, Burgess JL, Naleway AL, et al. Prevention and attenuation of Covid-19 with the BNT162b2 and mRNA-1273 vaccines. N Engl J Med. 2021;385:320–9.PubMedCrossRef Thompson MG, Burgess JL, Naleway AL, et al. Prevention and attenuation of Covid-19 with the BNT162b2 and mRNA-1273 vaccines. N Engl J Med. 2021;385:320–9.PubMedCrossRef
30.
Zurück zum Zitat Hatcher SM, Endres-Dighe SM, Angulo FJ, et al. COVID-19 vaccine effectiveness: a review of the first 6 months of COVID-19 vaccine availability (1 January–30 June 2021). Vaccines. 2022;10:393.PubMedPubMedCentralCrossRef Hatcher SM, Endres-Dighe SM, Angulo FJ, et al. COVID-19 vaccine effectiveness: a review of the first 6 months of COVID-19 vaccine availability (1 January–30 June 2021). Vaccines. 2022;10:393.PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Fung M, Babik JM. COVID-19 in immunocompromised hosts: what we know so far. Clin Infect Dis. 2021;72:340–50.PubMedCrossRef Fung M, Babik JM. COVID-19 in immunocompromised hosts: what we know so far. Clin Infect Dis. 2021;72:340–50.PubMedCrossRef
32.
33.
Zurück zum Zitat Truong TT, Ryutov A, Pandey U, et al. Increased viral variants in children and young adults with impaired humoral immunity and persistent SARS-CoV-2 infection: a consecutive case series. EBioMedicine. 2021;67:103355.PubMedPubMedCentralCrossRef Truong TT, Ryutov A, Pandey U, et al. Increased viral variants in children and young adults with impaired humoral immunity and persistent SARS-CoV-2 infection: a consecutive case series. EBioMedicine. 2021;67:103355.PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Hensley MK, Bain WG, Jacobs J, et al. Intractable coronavirus disease 2019 (COVID-19) and prolonged severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) replication in a chimeric antigen receptor-modified T-cell therapy recipient: a case study. Clin Infect Dis. 2021;73:e815–21.PubMedPubMedCentralCrossRef Hensley MK, Bain WG, Jacobs J, et al. Intractable coronavirus disease 2019 (COVID-19) and prolonged severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) replication in a chimeric antigen receptor-modified T-cell therapy recipient: a case study. Clin Infect Dis. 2021;73:e815–21.PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Baang JH, Smith C, Mirabelli C, et al. Prolonged severe acute respiratory syndrome coronavirus 2 replication in an immunocompromised patient. J Infect Dis. 2021;223:23–7.PubMedCrossRef Baang JH, Smith C, Mirabelli C, et al. Prolonged severe acute respiratory syndrome coronavirus 2 replication in an immunocompromised patient. J Infect Dis. 2021;223:23–7.PubMedCrossRef
36.
Zurück zum Zitat Choi B, Choudhary MC, Regan J, et al. Persistence and evolution of SARS-CoV-2 in an immunocompromised host. N Engl J Med. 2020;383:2291–3.PubMedCrossRef Choi B, Choudhary MC, Regan J, et al. Persistence and evolution of SARS-CoV-2 in an immunocompromised host. N Engl J Med. 2020;383:2291–3.PubMedCrossRef
37.
Zurück zum Zitat Remy KE, Mazer M, Striker DA, et al. Severe immunosuppression and not a cytokine storm characterizes COVID-19 infections. JCI Insight. 2020;5:e140329.PubMedCentralCrossRef Remy KE, Mazer M, Striker DA, et al. Severe immunosuppression and not a cytokine storm characterizes COVID-19 infections. JCI Insight. 2020;5:e140329.PubMedCentralCrossRef
38.
Zurück zum Zitat Lee KA, Ma W, Sikavi DR, et al. Cancer and risk of COVID-19 through a general community survey. Oncologist. 2021;26:e182–5.CrossRef Lee KA, Ma W, Sikavi DR, et al. Cancer and risk of COVID-19 through a general community survey. Oncologist. 2021;26:e182–5.CrossRef
39.
Zurück zum Zitat Tian Y, Qiu X, Wang C, et al. Cancer associates with risk and severe events of COVID-19: a systematic review and meta-analysis. Int J Cancer. 2021;148:363–74.PubMedCrossRef Tian Y, Qiu X, Wang C, et al. Cancer associates with risk and severe events of COVID-19: a systematic review and meta-analysis. Int J Cancer. 2021;148:363–74.PubMedCrossRef
40.
Zurück zum Zitat Giannakoulis VG, Papoutsi E, Siempos II. Effect of cancer on clinical outcomes of patients with COVID-19: a meta-analysis of patient data. JCO Glob Oncol. 2020;6:799–808.PubMedCrossRef Giannakoulis VG, Papoutsi E, Siempos II. Effect of cancer on clinical outcomes of patients with COVID-19: a meta-analysis of patient data. JCO Glob Oncol. 2020;6:799–808.PubMedCrossRef
41.
Zurück zum Zitat Zarifkar P, Kamath A, Robinson C, et al. Clinical characteristics and outcomes in patients with COVID-19 and cancer: a systematic review and meta-analysis. Clin Oncol (R Coll Radiol). 2021;33:e180–91.CrossRef Zarifkar P, Kamath A, Robinson C, et al. Clinical characteristics and outcomes in patients with COVID-19 and cancer: a systematic review and meta-analysis. Clin Oncol (R Coll Radiol). 2021;33:e180–91.CrossRef
42.
Zurück zum Zitat Zhang H, Han H, He T, et al. Clinical characteristics and outcomes of COVID-19-infected cancer patients: a systematic review and meta-analysis. J Natl Cancer Inst. 2021;113:371–80.PubMedCrossRef Zhang H, Han H, He T, et al. Clinical characteristics and outcomes of COVID-19-infected cancer patients: a systematic review and meta-analysis. J Natl Cancer Inst. 2021;113:371–80.PubMedCrossRef
43.
Zurück zum Zitat Agha ME, Blake M, Chilleo C, Wells A, Haidar G. Suboptimal response to coronavirus disease 2019 messenger RNA vaccines in patients with hematologic malignancies: a need for vigilance in the postmasking era. Open Forum Infect Dis. 2021;8:ofab353.PubMedPubMedCentralCrossRef Agha ME, Blake M, Chilleo C, Wells A, Haidar G. Suboptimal response to coronavirus disease 2019 messenger RNA vaccines in patients with hematologic malignancies: a need for vigilance in the postmasking era. Open Forum Infect Dis. 2021;8:ofab353.PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat Bird S, Panopoulou A, Shea RL, et al. Response to first vaccination against SARS-CoV-2 in patients with multiple myeloma. Lancet Haematol. 2021;8:e389–92.PubMedPubMedCentralCrossRef Bird S, Panopoulou A, Shea RL, et al. Response to first vaccination against SARS-CoV-2 in patients with multiple myeloma. Lancet Haematol. 2021;8:e389–92.PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Herishanu Y, Avivi I, Aharon A, et al. Efficacy of the BNT162b2 mRNA COVID-19 vaccine in patients with chronic lymphocytic leukemia. Blood. 2021;137:3165–73.PubMedPubMedCentralCrossRef Herishanu Y, Avivi I, Aharon A, et al. Efficacy of the BNT162b2 mRNA COVID-19 vaccine in patients with chronic lymphocytic leukemia. Blood. 2021;137:3165–73.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Harrington P, de Lavallade H, Doores KJ, et al. Single dose of BNT162b2 mRNA vaccine against SARS-CoV-2 induces high frequency of neutralising antibody and polyfunctional T-cell responses in patients with myeloproliferative neoplasms. Leukemia. 2021;35:3573–7.PubMedPubMedCentralCrossRef Harrington P, de Lavallade H, Doores KJ, et al. Single dose of BNT162b2 mRNA vaccine against SARS-CoV-2 induces high frequency of neutralising antibody and polyfunctional T-cell responses in patients with myeloproliferative neoplasms. Leukemia. 2021;35:3573–7.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Barrière J, Chamorey E, Adjtoutah Z, et al. Impaired immunogenicity of BNT162b2 anti-SARS-CoV-2 vaccine in patients treated for solid tumors. Ann Oncol. 2021;32:1053–5.PubMedCrossRef Barrière J, Chamorey E, Adjtoutah Z, et al. Impaired immunogenicity of BNT162b2 anti-SARS-CoV-2 vaccine in patients treated for solid tumors. Ann Oncol. 2021;32:1053–5.PubMedCrossRef
48.
Zurück zum Zitat Palich R, Veyri M, Marot S, et al. Weak immunogenicity after a single dose of SARS-CoV-2 mRNA vaccine in treated cancer patients. Ann Oncol. 2021;32:1051–3.PubMedCrossRef Palich R, Veyri M, Marot S, et al. Weak immunogenicity after a single dose of SARS-CoV-2 mRNA vaccine in treated cancer patients. Ann Oncol. 2021;32:1051–3.PubMedCrossRef
49.
50.
Zurück zum Zitat Haberman RH, Castillo R, Chen A, et al. COVID-19 in patients with inflammatory arthritis: a prospective study on the effects of comorbidities and disease-modifying antirheumatic drugs on clinical outcomes. Arthritis Rheumatol. 2020;72:1981–9.PubMedPubMedCentralCrossRef Haberman RH, Castillo R, Chen A, et al. COVID-19 in patients with inflammatory arthritis: a prospective study on the effects of comorbidities and disease-modifying antirheumatic drugs on clinical outcomes. Arthritis Rheumatol. 2020;72:1981–9.PubMedPubMedCentralCrossRef
51.
Zurück zum Zitat Pablos JL, Galindo M, Carmona L, et al. Clinical outcomes of hospitalised patients with COVID-19 and chronic inflammatory and autoimmune rheumatic diseases: a multicentric matched cohort study. Ann Rheum Dis. 2020;79:1544–9.PubMedCrossRef Pablos JL, Galindo M, Carmona L, et al. Clinical outcomes of hospitalised patients with COVID-19 and chronic inflammatory and autoimmune rheumatic diseases: a multicentric matched cohort study. Ann Rheum Dis. 2020;79:1544–9.PubMedCrossRef
52.
Zurück zum Zitat Geisen UM, Berner DK, Tran F, et al. Immunogenicity and safety of anti-SARS-CoV-2 mRNA vaccines in patients with chronic inflammatory conditions and immunosuppressive therapy in a monocentric cohort. Ann Rheum Dis. 2021;80:1306–11.PubMedCrossRef Geisen UM, Berner DK, Tran F, et al. Immunogenicity and safety of anti-SARS-CoV-2 mRNA vaccines in patients with chronic inflammatory conditions and immunosuppressive therapy in a monocentric cohort. Ann Rheum Dis. 2021;80:1306–11.PubMedCrossRef
53.
Zurück zum Zitat Braun-Moscovici Y, Kaplan M, Braun M, et al. Disease activity and humoral response in patients with inflammatory rheumatic diseases after two doses of the Pfizer mRNA vaccine against SARS-CoV-2. Ann Rheum Dis. 2021;80:1317–21.PubMedCrossRef Braun-Moscovici Y, Kaplan M, Braun M, et al. Disease activity and humoral response in patients with inflammatory rheumatic diseases after two doses of the Pfizer mRNA vaccine against SARS-CoV-2. Ann Rheum Dis. 2021;80:1317–21.PubMedCrossRef
54.
Zurück zum Zitat Deepak P, Kim W, Paley MA, et al. Effect of immunosuppression on the immunogenicity of mRNA vaccines to SARS-CoV-2: a prospective cohort study. Ann Intern Med. 2021;174:1572–85.PubMedCrossRef Deepak P, Kim W, Paley MA, et al. Effect of immunosuppression on the immunogenicity of mRNA vaccines to SARS-CoV-2: a prospective cohort study. Ann Intern Med. 2021;174:1572–85.PubMedCrossRef
55.
Zurück zum Zitat Boyarsky BJ, Ruddy JA, Connolly CM, et al. Antibody response to a single dose of SARS-CoV-2 mRNA vaccine in patients with rheumatic and musculoskeletal diseases. Ann Rheum Dis. 2021;80:1098–9.PubMedCrossRef Boyarsky BJ, Ruddy JA, Connolly CM, et al. Antibody response to a single dose of SARS-CoV-2 mRNA vaccine in patients with rheumatic and musculoskeletal diseases. Ann Rheum Dis. 2021;80:1098–9.PubMedCrossRef
56.
Zurück zum Zitat Wong SY, Dixon R, Martinez Pazos V, et al. Serologic response to messenger RNA coronavirus disease 2019 vaccines in inflammatory bowel disease patients receiving biologic therapies. Gastroenterology. 2021;161:715–8.e4.CrossRef Wong SY, Dixon R, Martinez Pazos V, et al. Serologic response to messenger RNA coronavirus disease 2019 vaccines in inflammatory bowel disease patients receiving biologic therapies. Gastroenterology. 2021;161:715–8.e4.CrossRef
57.
Zurück zum Zitat Chen CY, Shao SC, Chen YT, et al. Incidence and clinical impacts of COVID-19 infection in patients with hemodialysis: systematic review and meta-analysis of 396,062 hemodialysis patients. Healthcare (Basel). 2021;9:47.CrossRef Chen CY, Shao SC, Chen YT, et al. Incidence and clinical impacts of COVID-19 infection in patients with hemodialysis: systematic review and meta-analysis of 396,062 hemodialysis patients. Healthcare (Basel). 2021;9:47.CrossRef
58.
Zurück zum Zitat Bruchfeld A. The COVID-19 pandemic: consequences for nephrology. Nat Rev Nephrol. 2021;17:81–2.PubMedCrossRef Bruchfeld A. The COVID-19 pandemic: consequences for nephrology. Nat Rev Nephrol. 2021;17:81–2.PubMedCrossRef
59.
Zurück zum Zitat 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;98:1540–8.PubMedPubMedCentralCrossRef 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;98:1540–8.PubMedPubMedCentralCrossRef
60.
Zurück zum Zitat Chung EYM, Palmer SC, Natale P, et al. Incidence and outcomes of COVID-19 in people with CKD: a systematic review and meta-analysis. Am J Kidney Dis. 2021;78:804–15.PubMedPubMedCentralCrossRef Chung EYM, Palmer SC, Natale P, et al. Incidence and outcomes of COVID-19 in people with CKD: a systematic review and meta-analysis. Am J Kidney Dis. 2021;78:804–15.PubMedPubMedCentralCrossRef
61.
Zurück zum Zitat Schrezenmeier E, Bergfeld L, Hillus D, et al. Immunogenicity of COVID-19 Tozinameran vaccination in patients on chronic dialysis. Front Immunol. 2021;12:690698.PubMedPubMedCentralCrossRef Schrezenmeier E, Bergfeld L, Hillus D, et al. Immunogenicity of COVID-19 Tozinameran vaccination in patients on chronic dialysis. Front Immunol. 2021;12:690698.PubMedPubMedCentralCrossRef
62.
Zurück zum Zitat Goupil R, Benlarbi M, Beaubien-Souligny W, et al. Short-term antibody response after 1 dose of BNT162b2 vaccine in patients receiving hemodialysis. CMAJ. 2021;193:E793–800.PubMedPubMedCentralCrossRef Goupil R, Benlarbi M, Beaubien-Souligny W, et al. Short-term antibody response after 1 dose of BNT162b2 vaccine in patients receiving hemodialysis. CMAJ. 2021;193:E793–800.PubMedPubMedCentralCrossRef
63.
Zurück zum Zitat Simon B, Rubey H, Treipl A, et al. Haemodialysis patients show a highly diminished antibody response after COVID-19 mRNA vaccination compared with healthy controls. Nephrol Dial Transplant. 2021;36:1709–16.PubMedCrossRef Simon B, Rubey H, Treipl A, et al. Haemodialysis patients show a highly diminished antibody response after COVID-19 mRNA vaccination compared with healthy controls. Nephrol Dial Transplant. 2021;36:1709–16.PubMedCrossRef
64.
Zurück zum Zitat Grupper A, Sharon N, Finn T, et al. Humoral response to the Pfizer BNT162b2 vaccine in patients undergoing maintenance hemodialysis. Clin J Am Soc Nephrol. 2021;16:1037–42.PubMedPubMedCentralCrossRef Grupper A, Sharon N, Finn T, et al. Humoral response to the Pfizer BNT162b2 vaccine in patients undergoing maintenance hemodialysis. Clin J Am Soc Nephrol. 2021;16:1037–42.PubMedPubMedCentralCrossRef
66.
Zurück zum Zitat Prabhu M, Murphy EA, Sukhu AC, et al. Antibody response to coronavirus disease 2019 (COVID-19) messenger RNA vaccination in pregnant women and transplacental passage into cord blood. Obstet Gynecol. 2021;138:278–80.PubMedPubMedCentral Prabhu M, Murphy EA, Sukhu AC, et al. Antibody response to coronavirus disease 2019 (COVID-19) messenger RNA vaccination in pregnant women and transplacental passage into cord blood. Obstet Gynecol. 2021;138:278–80.PubMedPubMedCentral
67.
Zurück zum Zitat Mahalingasivam V, Craik A, Tomlinson LA, et al. A systematic review of COVID-19 and kidney transplantation. Kidney Int Rep. 2021;6:24–45.PubMedCrossRef Mahalingasivam V, Craik A, Tomlinson LA, et al. A systematic review of COVID-19 and kidney transplantation. Kidney Int Rep. 2021;6:24–45.PubMedCrossRef
68.
Zurück zum Zitat Kremer D, Pieters TT, Verhaar MC, et al. A systematic review and meta-analysis of COVID-19 in kidney transplant recipients: lessons to be learned. Am J Transplant. 2021;21:3936–45.PubMedPubMedCentralCrossRef Kremer D, Pieters TT, Verhaar MC, et al. A systematic review and meta-analysis of COVID-19 in kidney transplant recipients: lessons to be learned. Am J Transplant. 2021;21:3936–45.PubMedPubMedCentralCrossRef
69.
Zurück zum Zitat Kulkarni AV, Tevethia HV, Premkumar M, et al. Impact of COVID-19 on liver transplant recipients—a systematic review and meta-analysis. EClinicalMedicine. 2021;38:101025.PubMedPubMedCentralCrossRef Kulkarni AV, Tevethia HV, Premkumar M, et al. Impact of COVID-19 on liver transplant recipients—a systematic review and meta-analysis. EClinicalMedicine. 2021;38:101025.PubMedPubMedCentralCrossRef
70.
Zurück zum Zitat Jayant K, Reccia I, Virdis F, et al. COVID-19 in hospitalized liver transplant recipients: an early systematic review and meta-analysis. Clin Transplant. 2021;35:e14246.PubMedPubMedCentralCrossRef Jayant K, Reccia I, Virdis F, et al. COVID-19 in hospitalized liver transplant recipients: an early systematic review and meta-analysis. Clin Transplant. 2021;35:e14246.PubMedPubMedCentralCrossRef
71.
Zurück zum Zitat Diaz-Arocutipa C, Carvallo-Castaneda D, Luis-Ybanez O, et al. COVID-19 in heart transplant recipients during February–August 2020: a systematic review. Clin Transplant. 2021;35:e14390.PubMedPubMedCentralCrossRef Diaz-Arocutipa C, Carvallo-Castaneda D, Luis-Ybanez O, et al. COVID-19 in heart transplant recipients during February–August 2020: a systematic review. Clin Transplant. 2021;35:e14390.PubMedPubMedCentralCrossRef
72.
Zurück zum Zitat Phanish M, Ster IC, Ghazanfar A, et al. Systematic review and meta-analysis of COVID-19 and kidney transplant recipients, the South West London kidney transplant network experience. Kidney Int Rep. 2021;6:574–85.PubMedCrossRef Phanish M, Ster IC, Ghazanfar A, et al. Systematic review and meta-analysis of COVID-19 and kidney transplant recipients, the South West London kidney transplant network experience. Kidney Int Rep. 2021;6:574–85.PubMedCrossRef
73.
Zurück zum Zitat Samidoust P, Nikoupour H, Hemmati H, Samidoust A. Clinical manifestations and characterization of COVID-19 in liver transplant recipients: a systematic review of case reports and case series. Ethiop J Health Sci. 2021;31:429–38.PubMedPubMedCentral Samidoust P, Nikoupour H, Hemmati H, Samidoust A. Clinical manifestations and characterization of COVID-19 in liver transplant recipients: a systematic review of case reports and case series. Ethiop J Health Sci. 2021;31:429–38.PubMedPubMedCentral
74.
Zurück zum Zitat Granger C, Guedeney P, Arnaud C, et al. Clinical manifestations and outcomes of coronavirus disease-19 in heart transplant recipients: a multicentre case series with a systematic review and meta-analysis. Transpl Int. 2021;34:721–31.PubMedCrossRef Granger C, Guedeney P, Arnaud C, et al. Clinical manifestations and outcomes of coronavirus disease-19 in heart transplant recipients: a multicentre case series with a systematic review and meta-analysis. Transpl Int. 2021;34:721–31.PubMedCrossRef
75.
Zurück zum Zitat Boyarsky BJ, Werbel WA, Avery RK, et al. Immunogenicity of a single dose of SARS-CoV-2 messenger RNA vaccine in solid organ transplant recipients. JAMA. 2021;325:1784–6.PubMedPubMedCentralCrossRef Boyarsky BJ, Werbel WA, Avery RK, et al. Immunogenicity of a single dose of SARS-CoV-2 messenger RNA vaccine in solid organ transplant recipients. JAMA. 2021;325:1784–6.PubMedPubMedCentralCrossRef
76.
Zurück zum Zitat Sattler A, Schrezenmeier E, Weber UA, et al. Impaired humoral and cellular immunity after SARS-CoV-2 BNT162b2 (tozinameran) prime-boost vaccination in kidney transplant recipients. J Clin Investig. 2021;131:14.CrossRef Sattler A, Schrezenmeier E, Weber UA, et al. Impaired humoral and cellular immunity after SARS-CoV-2 BNT162b2 (tozinameran) prime-boost vaccination in kidney transplant recipients. J Clin Investig. 2021;131:14.CrossRef
77.
Zurück zum Zitat Itzhaki Ben Zadok O, Shaul AA, Ben-Avraham B, et al. Immunogenicity of the BNT162b2 mRNA vaccine in heart transplant recipients—a prospective cohort study. Eur J Heart Fail. 2021;23:1555–9.PubMedCrossRef Itzhaki Ben Zadok O, Shaul AA, Ben-Avraham B, et al. Immunogenicity of the BNT162b2 mRNA vaccine in heart transplant recipients—a prospective cohort study. Eur J Heart Fail. 2021;23:1555–9.PubMedCrossRef
78.
Zurück zum Zitat Basic-Jukic N, Ivo J. SARS-CoV-2 infection after two doses of mRNA vaccine in renal transplant recipients. Transplant Infect Dis. 2021;2021:e13628. Basic-Jukic N, Ivo J. SARS-CoV-2 infection after two doses of mRNA vaccine in renal transplant recipients. Transplant Infect Dis. 2021;2021:e13628.
79.
Zurück zum Zitat Rusk DS, Strachan CC, Hunter BR. Lack of immune response after mRNA vaccination to SARS-CoV-2 in a solid organ transplant patient. J Med Virol. 2021;93:5623–5.PubMedPubMedCentralCrossRef Rusk DS, Strachan CC, Hunter BR. Lack of immune response after mRNA vaccination to SARS-CoV-2 in a solid organ transplant patient. J Med Virol. 2021;93:5623–5.PubMedPubMedCentralCrossRef
80.
Zurück zum Zitat Grupper A, Rabinowich L, Schwartz D, et al. Reduced humoral response to mRNA SARS-CoV-2 BNT162b2 vaccine in kidney transplant recipients without prior exposure to the virus. Am J Transplant. 2021;21:2719–26.PubMedPubMedCentralCrossRef Grupper A, Rabinowich L, Schwartz D, et al. Reduced humoral response to mRNA SARS-CoV-2 BNT162b2 vaccine in kidney transplant recipients without prior exposure to the virus. Am J Transplant. 2021;21:2719–26.PubMedPubMedCentralCrossRef
81.
Zurück zum Zitat Rozen-Zvi B, Yahav D, Agur T, et al. Antibody response to SARS-CoV-2 mRNA vaccine among kidney transplant recipients: a prospective cohort study. Clin Microbiol Infect. 2021;27:1173.e1–4. Rozen-Zvi B, Yahav D, Agur T, et al. Antibody response to SARS-CoV-2 mRNA vaccine among kidney transplant recipients: a prospective cohort study. Clin Microbiol Infect. 2021;27:1173.e1–4.
82.
Zurück zum Zitat Narasimhan M, Mahimainathan L, Clark AE, et al. Serological response in lung transplant recipients after two doses of SARS-CoV-2 mRNA vaccines. Vaccines (Basel). 2021;9:708.CrossRef Narasimhan M, Mahimainathan L, Clark AE, et al. Serological response in lung transplant recipients after two doses of SARS-CoV-2 mRNA vaccines. Vaccines (Basel). 2021;9:708.CrossRef
83.
Zurück zum Zitat Peled Y, Ram E, Lavee J, et al. BNT162b2 vaccination in heart transplant recipients: clinical experience and antibody response. J Heart Lung Transplant. 2021;40:759–62.PubMedPubMedCentralCrossRef Peled Y, Ram E, Lavee J, et al. BNT162b2 vaccination in heart transplant recipients: clinical experience and antibody response. J Heart Lung Transplant. 2021;40:759–62.PubMedPubMedCentralCrossRef
84.
Zurück zum Zitat Rincon-Arevalo H, Choi M, Stefanski AL, et al. Impaired humoral immunity to SARS-CoV-2 BNT162b2 vaccine in kidney transplant recipients and dialysis patients. Sci Immunol. 2021;6:eabj1031.PubMedCrossRef Rincon-Arevalo H, Choi M, Stefanski AL, et al. Impaired humoral immunity to SARS-CoV-2 BNT162b2 vaccine in kidney transplant recipients and dialysis patients. Sci Immunol. 2021;6:eabj1031.PubMedCrossRef
85.
Zurück zum Zitat Mirzaei H, McFarland W, Karamouzian M, Sharifi H. COVID-19 among people living with HIV: a systematic review. AIDS Behav. 2021;25:85–92.PubMedCrossRef Mirzaei H, McFarland W, Karamouzian M, Sharifi H. COVID-19 among people living with HIV: a systematic review. AIDS Behav. 2021;25:85–92.PubMedCrossRef
86.
Zurück zum Zitat Hariyanto TI, Rosalind J, Christian K, Kurniawan A. Human immunodeficiency virus and mortality from coronavirus disease 2019: a systematic review and meta-analysis. South Afr J HIV Med. 2021;22:1220.PubMedPubMedCentralCrossRef Hariyanto TI, Rosalind J, Christian K, Kurniawan A. Human immunodeficiency virus and mortality from coronavirus disease 2019: a systematic review and meta-analysis. South Afr J HIV Med. 2021;22:1220.PubMedPubMedCentralCrossRef
87.
Zurück zum Zitat Patel RH, Acharya A, Chand HS, Mohan M, Byrareddy SN. Human immunodeficiency virus and severe acute respiratory syndrome coronavirus 2 coinfection: a systematic review of the literature and challenges. AIDS Res Hum Retroviruses. 2021;37:266–82.PubMedPubMedCentralCrossRef Patel RH, Acharya A, Chand HS, Mohan M, Byrareddy SN. Human immunodeficiency virus and severe acute respiratory syndrome coronavirus 2 coinfection: a systematic review of the literature and challenges. AIDS Res Hum Retroviruses. 2021;37:266–82.PubMedPubMedCentralCrossRef
88.
Zurück zum Zitat Lee KW, Yap SF, Ngeow YF, Lye MS. COVID-19 in people living with HIV: a systematic review and meta-analysis. Int J Environ Res Public Health. 2021;18:3554.PubMedPubMedCentralCrossRef Lee KW, Yap SF, Ngeow YF, Lye MS. COVID-19 in people living with HIV: a systematic review and meta-analysis. Int J Environ Res Public Health. 2021;18:3554.PubMedPubMedCentralCrossRef
89.
Zurück zum Zitat Liang M, Luo N, Chen M, et al. Prevalence and mortality due to COVID-19 in HIV co-infected population: a systematic review and meta-analysis. Infect Dis Ther. 2021;10:1267–85.PubMedPubMedCentralCrossRef Liang M, Luo N, Chen M, et al. Prevalence and mortality due to COVID-19 in HIV co-infected population: a systematic review and meta-analysis. Infect Dis Ther. 2021;10:1267–85.PubMedPubMedCentralCrossRef
90.
Zurück zum Zitat Sarkar S, Khanna P, Singh AK. Impact of COVID-19 in patients with concurrent co-infections: a systematic review and meta-analyses. J Med Virol. 2021;93:2385–95.PubMedCrossRef Sarkar S, Khanna P, Singh AK. Impact of COVID-19 in patients with concurrent co-infections: a systematic review and meta-analyses. J Med Virol. 2021;93:2385–95.PubMedCrossRef
91.
Zurück zum Zitat 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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
92.
Zurück zum Zitat SeyedAlinaghi S, Karimi A, MohsseniPour M, et al. The clinical outcomes of COVID-19 in HIV-positive patients: a systematic review of current evidence. Immun Inflamm Dis. 2021;9:1160–85.PubMedPubMedCentralCrossRef SeyedAlinaghi S, Karimi A, MohsseniPour M, et al. The clinical outcomes of COVID-19 in HIV-positive patients: a systematic review of current evidence. Immun Inflamm Dis. 2021;9:1160–85.PubMedPubMedCentralCrossRef
93.
Zurück zum Zitat Costenaro P, Minotti C, Barbieri E, Giaquinto C, Donà D. SARS-CoV-2 infection in people living with HIV: a systematic review. Rev Med Virol. 2021;31:1–12.PubMedCrossRef Costenaro P, Minotti C, Barbieri E, Giaquinto C, Donà D. SARS-CoV-2 infection in people living with HIV: a systematic review. Rev Med Virol. 2021;31:1–12.PubMedCrossRef
94.
Zurück zum Zitat Levy I, Wieder-Finesod A, Litchevsky V, et al. Immunogenicity and safety of the BNT162b2 mRNA COVID-19 vaccine in people living with HIV-1. Clin Microbiol Infect. 2021;27:1851–5.PubMedPubMedCentralCrossRef Levy I, Wieder-Finesod A, Litchevsky V, et al. Immunogenicity and safety of the BNT162b2 mRNA COVID-19 vaccine in people living with HIV-1. Clin Microbiol Infect. 2021;27:1851–5.PubMedPubMedCentralCrossRef
106.
Zurück zum Zitat Ducloux D, Colladant M, Chabannes M, Yannaraki M, Courivaud C. Humoral response after 3 doses of the BNT162b2 mRNA COVID-19 vaccine in patients on hemodialysis. Kidney Int. 2021;100:702–4.PubMedPubMedCentralCrossRef Ducloux D, Colladant M, Chabannes M, Yannaraki M, Courivaud C. Humoral response after 3 doses of the BNT162b2 mRNA COVID-19 vaccine in patients on hemodialysis. Kidney Int. 2021;100:702–4.PubMedPubMedCentralCrossRef
107.
Zurück zum Zitat Hall VG, Ferreira VH, Ku T, et al. Randomized trial of a third dose of mRNA-1273 vaccine in transplant recipients. N Engl J Med. 2021;385:1244–6.PubMedCrossRef Hall VG, Ferreira VH, Ku T, et al. Randomized trial of a third dose of mRNA-1273 vaccine in transplant recipients. N Engl J Med. 2021;385:1244–6.PubMedCrossRef
109.
Zurück zum Zitat Lee A, Wong SY, Chai LYA, et al. Efficacy of covid-19 vaccines in immunocompromised patients: systematic review and meta-analysis. BMJ. 2022;376: e068632.PubMedCrossRef Lee A, Wong SY, Chai LYA, et al. Efficacy of covid-19 vaccines in immunocompromised patients: systematic review and meta-analysis. BMJ. 2022;376: e068632.PubMedCrossRef
110.
Zurück zum Zitat Kamar N, Abravanel F, Marion O, et al. Three doses of an mRNA Covid-19 vaccine in solid-organ transplant recipients. N Engl J Med. 2021;385:661–2.PubMedCrossRef Kamar N, Abravanel F, Marion O, et al. Three doses of an mRNA Covid-19 vaccine in solid-organ transplant recipients. N Engl J Med. 2021;385:661–2.PubMedCrossRef
111.
Zurück zum Zitat Werbel WA, Boyarsky BJ, Ou MT, et al. Safety and immunogenicity of a third dose of SARS-CoV-2 vaccine in solid organ transplant recipients: a case series. Ann Intern Med. 2021;174:1330–2.PubMedCrossRef Werbel WA, Boyarsky BJ, Ou MT, et al. Safety and immunogenicity of a third dose of SARS-CoV-2 vaccine in solid organ transplant recipients: a case series. Ann Intern Med. 2021;174:1330–2.PubMedCrossRef
112.
Zurück zum Zitat Longlune N, Nogier MB, Miedouge M, et al. High immunogenicity of a messenger RNA-based vaccine against SARS-CoV-2 in chronic dialysis patients. Nephrol Dial Transplant. 2021;36:1704–9.PubMedCrossRef Longlune N, Nogier MB, Miedouge M, et al. High immunogenicity of a messenger RNA-based vaccine against SARS-CoV-2 in chronic dialysis patients. Nephrol Dial Transplant. 2021;36:1704–9.PubMedCrossRef
115.
Zurück zum Zitat Rubin LG, Levin MJ, Ljungman P, et al. 2013 IDSA clinical practice guideline for vaccination of the immunocompromised host. Clin Infect Dis. 2014;58:e44–100.PubMedCrossRef Rubin LG, Levin MJ, Ljungman P, et al. 2013 IDSA clinical practice guideline for vaccination of the immunocompromised host. Clin Infect Dis. 2014;58:e44–100.PubMedCrossRef
117.
Zurück zum Zitat Krammer F. A correlate of protection for SARS-CoV-2 vaccines is urgently needed. Nat Med. 2021;27:1147–8.PubMedCrossRef Krammer F. A correlate of protection for SARS-CoV-2 vaccines is urgently needed. Nat Med. 2021;27:1147–8.PubMedCrossRef
Metadaten
Titel
Immune Response to COVID-19 and mRNA Vaccination in Immunocompromised Individuals: A Narrative Review
verfasst von
Norka I. Napuri
Daniel Curcio
David L. Swerdlow
Amit Srivastava
Publikationsdatum
25.05.2022
Verlag
Springer Healthcare
Schlagwort
COVID-19
Erschienen in
Infectious Diseases and Therapy / Ausgabe 4/2022
Print ISSN: 2193-8229
Elektronische ISSN: 2193-6382
DOI
https://doi.org/10.1007/s40121-022-00648-2

Weitere Artikel der Ausgabe 4/2022

Infectious Diseases and Therapy 4/2022 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Mehr Lebenszeit mit Abemaciclib bei fortgeschrittenem Brustkrebs?

24.05.2024 Mammakarzinom Nachrichten

In der MONARCHE-3-Studie lebten Frauen mit fortgeschrittenem Hormonrezeptor-positivem, HER2-negativem Brustkrebs länger, wenn sie zusätzlich zu einem nicht steroidalen Aromatasehemmer mit Abemaciclib behandelt wurden; allerdings verfehlte der numerische Zugewinn die statistische Signifikanz.

ADT zur Radiatio nach Prostatektomie: Wenn, dann wohl länger

24.05.2024 Prostatakarzinom Nachrichten

Welchen Nutzen es trägt, wenn die Strahlentherapie nach radikaler Prostatektomie um eine Androgendeprivation ergänzt wird, hat die RADICALS-HD-Studie untersucht. Nun liegen die Ergebnisse vor. Sie sprechen für länger dauernden Hormonentzug.

„Überwältigende“ Evidenz für Tripeltherapie beim metastasierten Prostata-Ca.

22.05.2024 Prostatakarzinom Nachrichten

Patienten mit metastasiertem hormonsensitivem Prostatakarzinom sollten nicht mehr mit einer alleinigen Androgendeprivationstherapie (ADT) behandelt werden, mahnt ein US-Team nach Sichtung der aktuellen Datenlage. Mit einer Tripeltherapie haben die Betroffenen offenbar die besten Überlebenschancen.

So sicher sind Tattoos: Neue Daten zur Risikobewertung

22.05.2024 Melanom Nachrichten

Das größte medizinische Problem bei Tattoos bleiben allergische Reaktionen. Melanome werden dadurch offensichtlich nicht gefördert, die Farbpigmente könnten aber andere Tumoren begünstigen.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.