Abstract
Inborn errors of immunity (IEI) are a heterogeneous group of disorders affecting immune host defense and immunoregulation. Considering the predisposition to develop severe and chronic infections, it is crucial to understand the clinical evolution of COVID-19 in IEI patients. This review analyzes clinical outcomes following SARS-CoV-2 infection, as well as response to COVID-19 vaccines in patients with IEI.
Keywords: COVID-19, COVID-19 vaccine, inborn errors of immunity, primary immune deficiency, SARS-COV-2
Introduction
As of April 2022, COVID-19 has spread worldwide infecting more than 500 million individuals and causing more than 6 million deaths. Elderly subjects and individuals with underlying comorbidities, including specific groups of immunocompromised patients, often suffered from severe disease and had worse clinical outcomes (1, 2). For patients with inborn errors of immunity (IEI), some studies have reported contrasting data on infection fatality rates that were either comparable to those in the general population (3–5) or increased, with a higher rate of hospitalization, younger age at death, and prolonged viral shedding (6–10). These discrepancies are mainly due to an assortment bias due to the differences in the nature of IEI included in the studies. Importantly, studies conducted on patients with IEI and life-threatening COVID-19 have revealed how impairment in selected immune genes may predispose a patient to critical SARS-CoV-2 infection and multisystem inflammatory syndrome (MIS) (11–18). In this context, an anti-SARS- CoV-2 vaccination campaign was implemented to protect patients with IEI. However, cellular and humoral responses after standard immunization doses are often suboptimal in this group of patients (19–23) and additional vaccination boosts and preexposure prophylaxis with monoclonal antibodies (moAbs) against the SARS-CoV-2 spike protein are indicated (20, 22, 23). Here we review clinical outcomes after SARS-CoV-2 infection and response to COVID-19 vaccines in patients with IEI.
Patients with IEI and SARS-CoV-2 Infection
Since the beginning of the pandemic, multiple cases of SARS-CoV-2 infection have been reported in patients with a broad variety of IEI. The majority of the patients presented with clinical features of classic acute COVID-19 infection, including respiratory symptoms, pneumonia, ARDS, fever, headaches, anosmia, fatigue, and gastrointestinal symptoms, while a smaller group developed features consistent with multisystem inflammatory syndrome (MIS) 4–6 wk after exposure to SARS-CoV-2 (4, 7, 15, 16, 24). In the next paragraphs, we summarize outcomes reported up to March 2022 in patients with COVID-19 and various IEI.
Combined Immunodeficiencies
We identified in the literature 132 patients with a diagnosis of either severe combined immunodeficiency (SCID; n = 22) or combined immunodeficiency (CID; n = 110) and COVID-19 (3–11, 25–40). The overall mortality in this group was 12% (n = 16) with a total of 27 patients (20%) admitted to the intensive care unit (ICU). In the SCID group, 14 patients had received stem cell therapy (SCT) before infection with SARS-CoV-2 (3, 4, 6–8, 11, 28, 36, 39, 40) and their survival rate was 93% (13/14), with only one death reported in a patient that was infected on day +40 post-SCT (8). By contrast, out of eight untransplanted SCID patients who were infected with SARS-CoV-2, only four survived, with death being primarily due to respiratory failure and ARDS (26). Recently Al-Saud et al. (11) reported of a previously healthy 5-mo-old boy with critical COVID-19 pneumonia requiring mechanical ventilation that after immune workup was diagnosed with T-B + NK SCID due to JAK3 deficiency and was successfully transplanted from a matched sibling donor. This case underlines the importance of genetic evaluation in critical cases of COVID-19 in healthy pediatric patients. SARS-CoV-2 infection has been reported in 11 patients with hyper-IgE syndromes (HIES): 2 patients with PGM3 deficiency (4), 7 with heterozygous dominant negative signal transducer and activator of transcription 3 (DNSTAT3) mutations (4, 8), and 2 without known genetic diagnosis (9, 26). In this group, three adults (2 DNSTAT3 and 1 without genetic diagnosis) with lung disease before SARS-CoV-2 infection died of respiratory failure due to ARDS and shock, while the other nine patients had milder disease and recovered. Despite preexisting lung damage, most patients with DNSTAT3 variants had a relatively good outcome. Interestingly elevated IL-6 levels have been associated with poor survival in COVID-19 (41, 42); thus it is reasonable to postulate that impaired IL-6/STAT3 signaling could dampen the hyperinflammation characteristic of severe COVID-19 and ameliorate disease outcomes in patients with DNSTAT3 variants.
Fatal outcomes due to sepsis and respiratory failure have also been reported in a patient with CID due to STIM1 deficiency and Nijmegen Breakage Syndrome (8, 26). In contrast, three patients with RELB deficiency (3) and two with ARPC1B deficiency had only mild disease (4, 6, 24).
Primary Antibody Deficiencies
The majority of patients with IEI infected with SARS-CoV-2 reported in the literature were affected with primary antibody deficiency (PAD; n = 446) (3–9, 26, 28, 30, 32, 33, 35, 37, 38, 43–68), including 290 with common variable immunodeficiency (CVID; 65% of all PAD), 71 with X-linked agammaglobulinemia (XLA; 16%), and 9 with autosomal recessive agammaglobulinemia (2%). The remaining 76 patients had other B-cell defects, including activated PI3Kdelta syndrome (APDS1: n = 6; APDS2: n = 1), nuclear factor-kappa B subunit 1 (NFKB1) deficiency (n = 2), nuclear factor-kappa B subunit 2 (NFKB2) deficiency (n = 4), TRNT1 (CCA-adding transfer RNA nucleotidyl transferase) enzyme deficiency (n = 1), specific polysaccharide antibody deficiency (SPAD; n = 13), isolated IgA deficiency (n = 17), IgG subclasses deficiency (n = 3), or unspecified antibody deficiency (n = 29). CVID and XLA patients displayed similar ICU admission and mortality rates (15% and 8% respectively); however, the latter group had a significantly lower median age as compared with the CVID group (69). These data caution on the validity of reports early in the pandemic that suggested that XLA patients may not be susceptible to severe COVID-19 disease, because BTK deficiency may decrease virus-driven IL-6 production by monocyte and consequently attenuate the cytokine storm typical COVID -19.
The overall ICU admission rate in the PAD group was 13% with a mortality rate of 7%. Deaths were often due to respiratory failure sometimes associated with septic shock and multiorgan failure, especially in patients with preexisting comorbidities. In particular, a meta-analysis of 88 CVID cases showed that patients with chronic lung involvement have an increased risk for severe COVID-19 in comparison to those without lung diseases. Different from the general population, age and metabolic comorbidities did not represent a risk factor for a severe course in this group of patients (70). Among the other causes of PAD, subjects with APDS (n = 7) and IgA deficiency (n = 17) had the best outcomes with no deaths or need for ICU care, while patients with NFKB2 deficiency had variable clinical severity (4, 37, 43, 57).
Primary Immune Regulatory Disorders
Sixty-nine patients with primary immune regulatory disorders (PIRD) and SARS-CoV-2 infection have been reported (3, 4, 7–9, 15, 16, 28, 29, 35, 71–77). The mortality rate was 16% (n = 11) while the rate of ICU admission was 30% (n = 21), similar to what had already been reported in the meta-analysis by Bucciol et al (78). Thirty-one patients were affected by APECED due to biallelic LOF mutations in the autoimmune regulator (AIRE) gene (8, 71–73, 75, 76). In this group, the mortality rate (16%, n = 5) was comparable to the overall PIRD group while the ICU admission rate (42%, n = 13) was higher. This finding is most likely due to the presence of neutralizing autoantibodies directed against type-I interferons (IFNs) and preexisting autoimmune pneumonitis typical of the disease (71, 72, 75, 79). Respiratory failure was the main cause of death in APECED, but early therapeutic intervention with anti-SARS-CoV-2 mAbs may be beneficial (80).
COVID-19 has been reported in 12 patients with either CTLA-4 haploinsufficiency or lipopolysaccharide-responsive and beige-like anchor protein (LRBA) deficiency, with a mortality rate of 17% (n = 2). One LRBA and one CTLA-4 patient were status post-SCT, 4 and 2 yr before SARS-CoV-2 infection, and both survived (3, 4, 8, 10, 30). Five other patients were affected with X-linked lymphoproliferative syndrome (XLP) 1 (n = 1) or XLP2 (n = 4). One previously healthy child was diagnosed with X-linked inhibitor of apoptosis (XIAP) deficiency after presenting with severe MIS-C (15). Two XLP2 patients died after being infected with SARS-CovV- post-SCT: one was infected on day +6 post-SCT and eventually died due to fungal infection in the context of poor engraftment, while the other developed severe graft-versus-host disease (GVHD) and succumbed to sepsis and HLH (4, 7, 15). The remaining two patients with XLP had mild COVID-19 (9).
Congenital Defects of Phagocytes
Among 34 patients with phagocyte defects reported, 22 had chronic granulomatous disease (CGD) (3, 4, 6, 7, 10, 15, 28, 30, 33, 38, 81). Two patients with CGD required ICU admission and eventually died due to Bulkolderia sepsis and uncontrolled MIS-C, respectively (4, 6). Among the remaining patients with congenital defects of phagocytes, two patients were affected by GATA2 haploinsufficiency (4, 10): both survived but one was hospitalized. One patient had DNAJC21 deficiency with early childhood bone marrow failure and mental disability and was hospitalized due to incomplete HLH but recovered (4).
Innate Immune Defects
Fifty-eight patients with innate immune defects and COVID-19 have been reported (4, 5, 7, 12, 18, 29, 30, 82–88). More than 50% of the subjects in this cohort were admitted to the ICU with a mortality rate of 10% (n = 6). Four patients succumbed to respiratory failure, one patient died of septic shock, and another developed sinus thrombosis, seizures, and multiorgan failure in association with a hyperinflammatory status (87). Worse outcomes in this group of IEI underline the fundamental role of innate immunity in early antiviral response against coronavirus; however, a strong selection bias must be taken into account given that many of the subjects were identified in studies focused on identifying innate immune defects in critically ill COVID-19 patients (12, 18, 88). Zhang et al. (18) identified that around 3.5% of severe COVID-19 cases resulted from IEI in genes involved in the type I IFN signaling pathway (i.e., TLR3, UNC93B1, TBK1, IRF3). Subsequently, TLR7 LOF variants were identified in 21 male subjects with critical COVID-19 with a case fatality rate of 15% (n = 3). Virtually all patients required ICU care despite having few comorbidities before SARS-CoV-2 infection. An impaired signaling in response to specific TLR7 ligands, as well as SARS-CoV-2, leading to reduced production of type I IFN by B cells, myeloid cells, and plasmacytoid dendritic cells was identified as a major pathogenetic factor in this group (12, 88). Of note, two patients with TLR3 and IRF3 deficiency benefit from the administration of subcutaneous pegylated IFNα2 (85). Consistent with a crucial role of the TLR signaling pathway in early responses to SARS-CoV-2, several cases of severe infection have been reported in patients with MyD88 (n = 5) and IRAK4 (n = 3) deficiency, thus supporting the role of these molecules in the control of viral replication in lung tissue as previously shown in a murine model of SARS-CoV-1. Among the five MyD88 cases described, three required ICU care (5, 7, 29).
Six patients with STAT1 GOF were identified (4, 8, 9, 29, 30, 84): five experienced mild disease course, and only one was hospitalized but recovered. This favorable outcome may support the concept that STAT1 GOF mutations may to some degree protect from SARS-CoV-2 infection/severe COVID-19 via the preemptive overactivation of IFN-I signaling, despite their otherwise immune deficient status (89).
Autoinflammatory Disorders
Ninety-one patients with a previous diagnosis of autoinflammatory disease were infected with SARS-CoV-2. The majority of these patients had familial Mediterranean fever (FMF; n = 73) (4, 6, 28, 90–93). In the FMF group, the rate of ICU admission was 5% (n = 4) and the infection fatality rate was 4% (n = 3), lower compared to the overall group infection fatality rate of 6% (n = 5) and ICU admission rate of 7% (n = 6). Of the remaining patients, seven had type I interferon-mediated diseases (IFNopathy): two SAVI (STING-associated vasculopathy with onset in infancy), four CANDLE (chronic atypical neutrophilic dermatosis with lipodystrophy and elevated temperature) or PRAAS (proteasome-associated autoinflammatory syndrome), and one SAMD9L-associated autoinflammatory disease (SAAD). All patients with IFNopathy had mild disease (94) except for the patient with SAAD and interstitial lung disease that developed severe pneumonia, and systemic hyperinflammation and required mechanical ventilation for 8 days before recovery. Critical disease was also observed in a child with unspecified autoinflammatory disease that died of MIS-C and vasculitis and in another patient with IL-1 receptor antagonist deficiency syndrome that succumbed to respiratory failure (6, 28). In contrast, one child with IL-18-mediated disease, two with neonatal-onset multisystem inflammatory disease (NOMID), and five with Aicardi-Goutieres syndrome (2 treated with JAK inhibitors) experienced mild COVID-19 infection and did not even require hospitalization (4, 5, 10, 94). Finally, one subject with DADA2 deficiency was hospitalized but recovered (8).
Complement Deficiencies
Among 42 reported patients with complement deficiency, 41 had a diagnosis of hereditary angioedema with or without C1inhibitor deficiency (HEA) and 1 patient had C3 deficiency. All subjects suffered from either asymptomatic or mild COVID-19 with no ICU admissions or deaths reported (7, 9, 95). Forty percent of HAE patients developed edema attacks (mainly extremities and abdominal edema, rare laryngeal edema) including one subject that had not suffered from angioedema attacks in the previous 14 yr (95). Sustained complement activation has been associated with negative COVID-19 outcomes (96, 97). In this regard, it is surprising that patients with HAE that present with bradykinin overproduction (which had been proposed as a possible mediator involved in the respiratory complications of COVID-19 infection) display only mild COVID-19 clinical manifestations.
Good Syndrome
Thirteen cases of SARS-CoV-2 infection have been reported in patients with Good syndrome (thymoma with hypogammaglobulinemia) (5–7, 9, 59, 65, 98–100). Most subjects had severe and critical disease, with an ICU admission rate of 62% (n = 8) and a mortality rate of 31% (n = 4). Higher median age in this group of patients should be considered as a confounder. Hyperinflammatory status, septic shock, and respiratory and cardiac failure were the cause of death (6, 7). Neutralizing autoantibodies to type I IFN are common in Good syndrome and likely represent a risk factor for severe COVID-19; however, 2 of the 13 patients lacked these autoantibodies and yet suffered from critical or severe disease (65).
MIS-C and MIS-A
MIS in children (MIS-C) or adults (MIS-A) occurs 4–6 wk after exposure to SARS-CoV-2 (101–105) and manifests with multiorgan disease, with the majority of cases displaying cardiovascular involvement, associated myocarditis (88%), and shock in half of the patients. Gastrointestinal, cutaneous, respiratory, and neurologic involvement is also common. Older age at onset and the severe hyperinflammatory status with overlapping features with HLH represent distinctive features of MIS-C compared to Kawasaki disease (102–105). At least 15 patients with IEI and MIS-C have been described (4, 6, 7, 15, 16) with fatal outcomes in 3 and one-third of the patients requiring intensive care unit care for cardiovascular and respiratory support. These patients were between 1 and 17 yr of age, and 85% of them were males. The genetic diagnosis was known in nine patients (1 IRAK4 deficiency, 1 SOCS1 deficiency, 2 XIAIP, 1 CD40L, 1 Chediak-Higashi, and 3 CGD) while five patients carried a diagnosis of autoinflammatory conditions (6), innate immune defects, and disorders of immune regulation without a specific genetic diagnosis.
Risk Factors and Therapeutic Strategies of SARS-CoV-2 Inborn Errors of Immunity
Specific risk factors among patients with IEI have been associated with worse prognoses. Shields et al. (10) reported that higher age, use of prophylactic antibiotics, and comorbidities such as diabetes, cardiovascular disease, chronic kidney, and lung disease correlate with increased hospitalization and mortality rates. In line with these findings in the study by Meyts et al. (4), all adult patients that died of COVID19 (n = 7) carried significant preexisting multiple organ impairment. By contrast, another multicenter study (9) did not confirm that increased age and body mass index have a negative impact on COVID-19 outcomes; nonetheless, this and other groups convincingly showed increased mortality in patients with low lymphocyte counts (T, B, and NK cells) and low levels of IgG, IgM, and IgA (9, 57, 69).
NIH therapeutic guidelines for immunocompromised patients including IEI include antivirals (in particular, Paxlovid and Remdesivir) and anti-SARS-CoV-2 monoclonal antibodies as primary or secondary prevention of COVID-19. In addition, immunomodulators such as IL-1 inhibitors (e.g., Anakinra), IL-6 inhibitors (e.g., Tocilizumab), and kinase inhibitors (JAK inhibitors and BTK inhibitors) are currently under investigation in clinical trials of patients with severe COVID-19 (106–109). Early treatment with type I interferons to promptly limit viral penetration has been attempted despite the timing of administration being a challenge (85, 110). Convalescent plasma appeared to be safe and effective in multiple patients with IEI; however, caution is required due to the presence of anti-type I interferon antibodies in the plasma preparations (25, 36, 49, 59, 66). Furthermore, adequate IgG levels during immunoglobulin replacement therapy and/or effective immunosuppressive therapies, including monoclonals, that target specific hyperactivated immune pathways dampening underlying inflammation before SARS-CoV-2 infection (i.e., tocilizumab, sirolimus, JAK inhibitors) have been associated with mild disease course (57, 84, 90, 109). This evidence is not definitive given the paucity of data, and the clinical immunologist should be aware that guidelines for patients with rheumatological diseases treated with immunomodulatory medication recommend withholding the immunosuppression during symptomatic COVID-19 up to 2 wk after resolution of symptoms (111). However, in patients with IEI, immunodeficiency and immune dysregulation often coexist; therefore, genetic diagnosis and the patient’s clinical and immunological features should be taken into account to identify the best treatment during SARS-CoV-2 infection.
Anti-SARS-CoV-2 Vaccination in Inborn Errors of Immunity
Although inactivated or nonviable vaccines are generally considered safe in patients with primary immunodeficiencies, the efficacy of these vaccines is variable, mainly depending on the specific IEI and the associated defect in the immune response (112). Of note, many patients with humoral immunodeficiencies can mount a protective T-cell-dependent antibody response to protein-conjugated vaccines (112).
Measured response to SARS-CoV-2 infection in otherwise healthy subjects has revealed a strong interplay between the cellular and humoral adaptative immune systems, with a prominent CD4+ T-cell response, which in turn helps induce antibodies against the Spike (S) and Nucleocapsid (N) proteins of SARS-CoV-2 (113). Similarly, immune response to the available messenger RNA (mRNA) COVID-19 vaccines has been found to induce a significant T-cell and neutralizing anti-S antibody response (114).
As for the studies evaluating the effect of SARS-CoV-2 infection in IEI, the majority of reports focus on COVID-19 vaccination in CVID patients (19–23, 115–124). Table 1 summarizes the data on seroconversion in CVID patients following the COVID-19 vaccine. Of note, CVID patients showed a relative ability to mount a specific humoral response after two vaccine doses, although variable across the studies. Combining the seroconversion data reported in Table 1, anti-Spike antibodies were detected in 196/329 (60%) CVID patients after two vaccine doses. However, these results need to be interpreted with some caveats. All the studies that included vaccinated healthy controls (HC) (19, 22, 23, 116–118) demonstrated a lower magnitude of antibody response in CVID patients compared to HC. In addition, the development of the humoral immune response was slower in CVID than in HC, since most CVID patients needed two vaccine doses to produce virus-specific antibodies. Moreover, the studies that assessed the antibody neutralizing activity (21, 117) showed that only a limited number of CVID patients can develop virus-neutralizing antibodies. Overall, these data suggest that both the quantity and quality of the humoral immune response to COVID-19 vaccination may be suboptimal in CVID subjects. In this regard, the evaluation of SARS-CoV-2-specific B- and T-cell immunity seems to be critically important to analyze vaccine-induced protection in these patients. Recent studies have shown that, in CVID patients, immunization with two doses of mRNA vaccine did not generate Spike-specific classical memory B cells (mB) but atypical mB with low binding capacity to Spike protein (22, 23, 118). Interestingly, the B-cell response in convalescent CVID patients was consistent with that observed in immunized immunocompetent individuals, who generated Spike-specific mB (22), suggesting that natural infection may induce more robust germinal center responses than the mRNA vaccine. Moreover, immunization after SARS-CoV-2 infection generated Spike-specific classical mB with low binding capacity to Spike protein and Spike-specific antibodies in CVID patients (22). This observation that the immune response elicited by SARS-CoV-2 natural infection was enhanced by subsequent immunization underlies the need to immunize convalescent COVID-19 CVID patients after recovery (22). Spike-specific T-cell responses were induced in immunized CVID patients, although with a variable frequency (22, 23, 116, 118) and less efficiently than observed for specific T cells generated after influenza virus immunization (125). For CVID patients, it has been postulated that while the influenza virus vaccine induces specific T cells after multiple exposures to viral antigen, the limited Spike-specific T-cell response may be a consequence of a limited antigenic stimulation by a new pathogen, which has never been encountered before (126). In this context, CVID patients might require additional vaccine doses or combinations of different SARS-CoV-2 vaccines to obtain possible protection. Recently, Gernez et al. (119) assessed the efficacy of a third dose of the COVID-19 mRNA vaccine on the humoral responses in CVID patients. All 10 patients with CVID mounted a virus-specific antibody response, and one-half (n = 5) had functional antibodies that had 50% or greater ACE2 blocking activity, thus showing that a third “booster” dose is required to achieve a better immune response against COVID-19. However, further studies are needed to evaluate the effect of the third vaccine dose on the efficacy and persistence of humoral and cellular responses to SARS-CoV-2.
TABLE 1.
Antibody responses to COVID-19 vaccine in patients with CVID
Country | Seroconversion Data | Factors Associated with Increased Risk of Failure | Major Adverse Events | Reference |
---|---|---|---|---|
USA | 6/6 (100%) | NA | None | 118 |
USA | 7/8 (87.5%) | NA | None | 20 |
USA | 12/15 (80%)* | NA | None (see Table 2) | 21 |
USA | 10/10 (100%) after 3 doses† | NA | None | 113 |
Israel | 10/12 (83.3%) | Lower response in older pt | None | 114 |
Israel | 11/15 (73.3%) | B ≤1% or B <6% and smB ≤2% of B | NA | 109 |
Italy | 3/4 (75%) | B <1% | None | 116 |
Italy | 11/33 (33%)‡ | Low RBD-specific smB | NA | 112 |
Italy | 8/34 (23.5%)‡ | All CVID patients lacked mB and activated mB with high binding capacity | NA | 23 |
Italy | 14/38 (36.8%)‡ | smB ≤2% of B; low IgA, IgM | NA | 22 |
Italy | 13/14 (92%)‡ | NA | None | 110 |
Sweden | 28/41 (68.3%)‡ | NA | None | 19 |
Wales | 43/60 (71.7%) | Low IgA + IgM, low B, ChAdOx1-S recipients | NA | 115 |
Spain | 15/18 (83%),‡ 9/18 (50%) neutralizing Ab | B cell lymphopenia; autoimmune/lymphoproliferation | None | 111 |
Austria | 15/31 (48.4%) | Low IgG pre-IVIG, low B, low smB, reduced Tmem activation | NA | 117 |
Ab, antibody; B, B cells; CVID, common variable immunodeficiency; NA, not available; IVIG, intravenous immunoglobulin; pt; patients; RBD, receptor-binding domain; smB, switched memory B cells; Tmem, vaccine antigen-specific T-memory cells. *Only 8.3% with neutralizing activity (SARS-CoV-2 ACE2 blocking activity) after 2 doses. †50% with neutralizing activity (SARS-CoV-2 ACE2 blocking activity) after 3 doses. ‡Low magnitude of antibody response compared to healthy controls.
The recently reported ‘‘breakthrough’’ COVID-19 cases in vaccinated individuals, which are the result of both the emergence of new viral variants and waning immunity over time, represent a current health challenge. A recent study has demonstrated that patients with primary immunodeficiencies, together with other immunocompromised patients as organ transplant recipients, have a higher incidence rate of breakthrough infections after two mRNA vaccine doses compared to healthy vaccinated individuals (127).
Another important aspect regarding the studies that assessed the antibody responses to the COVID-19 vaccine in patients with CVID is the definition of the clinical and immunological factors associated with increased risk of seroconversion failure after two vaccine doses. These risk factors are specified in Table 1 and included older patients’ age (120), history of autoimmunity (117), B-cell lymphopenia (including B cells ≤1%; switched memory B cells ≤2% of the total B cells; low SARS-CoV-2-specific memory B cells) (20, 22, 23, 115, 117, 118, 121–123), low levels of IgA and IgM (22, 121), and T-cell lymphopenia (20) with reduced T-memory activation (123).
Besides CVID, data on the immunogenicity of the SARS-CoV-2 vaccine in patients with IEI are few and often limited to anecdotal cases or heterogeneous cohorts. Table 2 summarizes the available literature (19–21, 23, 89, 120, 121, 124, 128). Except for patients with XLA and combined immunodeficiency, most patients with IEI can produce anti-Spike antibodies following two vaccine doses. Moreover, Spike protein-specific T-cell responses evaluated using SARS-CoV-2 IFN-γ release assays or ELISpot assays tested positive in most patients with XLA, demonstrating, as expected, that also these patients can mount SARS-CoV-2-specific T-cell responses. Of note, all the studies showed that the COVID-19 vaccine has a good safety profile with no major adverse events reported in IEI patients. However, because of the increased risk of thrombosis with thrombocytopenia syndrome, the use of the Johnson & Johnson vaccine should be avoided in IEI patients with thrombocytopenia or with anti-phospholipid antibodies (129, 130).
TABLE 2.
Humoral and cellular responses to COVID-19 vaccine in IEI other than CVID*
Country | SARS-CoV-2 Tests Performed | IEI | Evidence of Vaccine Response | Major Adverse Events | Reference |
---|---|---|---|---|---|
USA | Anti-S Ab; anti-N Ab | XLA (n = 1) | No | None | 118 |
WAS (n = 1) | Anti-S Ab + | ||||
DiGeorge syndrome (n = 1) | Anti-S Ab + | ||||
USA | Anti-S Ab; IGRA | Agammaglobulinemia (n = 2) | IGRA + (2/2, 100%) | Most frequent adverse event was sore arm. No major adverse events (only one patient reported a flare of enteropathy one week after vaccination). | 21 |
Hypogammaglobulinemia (n = 4) | Anti-S Ab + (2/4, 50%) | ||||
IGRA + (3/4, 75%) | |||||
SpAD (n = 2) | Anti-S Ab + (2/2, 100%) | ||||
IGRA + (2/2, 100%) | |||||
Good syndrome (n = 4) | Anti-S Ab + (0/4) | ||||
IGRA + (1/4, 25%) | |||||
Hyper IgM syndrome (n = 2) | Anti-S Ab + (0/2) | ||||
IGRA + (2/2, 100%) | |||||
CTLA-4 deficiency (n = 1) | IGRA + | ||||
PIK3R1 mutation causing APDS2 (n = 1) | IGRA not performed | ||||
Ataxia telangiectasia (n = 1) | IGRA not performed | ||||
ATP6AP1 gene/immunodeficiency (n = 1) | IGRA + | ||||
USA | Anti-S Ab; anti-N Ab | STAT3 DN (n = 26) | Anti-S ab + in | None | 20 |
APECED (n = 14) | |||||
Other immune regulation disorders (n = 10, including STAT1 GOF, STAT3 GOF, RAG deficiency with autoimmunity, unspecified CID with autoimmunity, recurrent autoimmune cytopenias, MAGT1 deficiency and thymoma) | 27/46 patients (58.7%) after 1 dose | ||||
Antibody deficiency (n = 13, including CVID, XLA, and other forms of hypogammaglobulinemia) | |||||
Other forms of IEI (n = 14, including WAS, WHIM syndrome, prolidase deficiency, SASH3 deficiency, CD40L deficiency, RALD, ADA deficiency and FOXN1 deficiency) | 63/74 (85.1%) after 2 dose | ||||
Post-HCT patients (n = 6) | |||||
USA | Anti-S Ab; anti-N Ab | MAGT1 deficiency (n = 1) | Anti-S Ab + after 2 doses | None | 122 |
Israel | Anti-S Ab; anti-N Ab; neutralizing Ab; specific anti-RBD B cells; ELISpot assay | XLA (n = 4) | Anti-S ab + in 18/26 (70%) | None | 114 |
Predominantly antibody deficiency (n = 17) | |||||
ALPS-like (n = 2) | |||||
STAT1-GOF (n = 1) | S-peptide–specific T-cell response + in 19/26 (73%) | ||||
STAT3 DN (n = 1) | |||||
C4 deficiency (n = 1) | |||||
Sweden | Anti-S Ab | XLA (4) | Anti-S Ab + in 0/4 | None | 19 |
Idiopathic T-cell Lymphopenia (n = 11) | Anti-S Ab + in 10/11 (90.9%) | ||||
Monogenic diseases (n = 9) | Anti-S Ab + in 7/9 (77.8%) | ||||
Other PID (n = 10) | Anti-S Ab + in 10/10 (100%) | ||||
Wales | Anti-S Ab | CID without molecular diagnosis (n = 8) | Anti-S Ab + in | NA | 115 |
0/8 | |||||
SpAD (n = 8) | 7/8 (87.5%) | ||||
DiGeorge syndrome (n = 4) | 4/4 | ||||
XLA (n = 3) | 0/3 | ||||
STAT1 GOF (n = 2) | 1/2 (50%) | ||||
APECED (n = 1) | 1/1 | ||||
CD40L deficiency (n = 1) | 0/1 | ||||
CGD (n = 1) | 1/1 | ||||
CTLA- 4 deficiency (n = 1) | 1/1 | ||||
Complement C2 deficiency (n = 1) | 1/1 | ||||
ADA2 deficiency (n = 1) | 1/1 | ||||
IFNGR1 deficiency (n = 1) | 1/1 | ||||
NEMO deficiency (n = 1) | 1/1 | ||||
CHH (n = 1) | 1/1 | ||||
STAT3 DN. Posthematopoietic stem cell transplantation (n = 1) | 1/1 | ||||
Idiopathic T-cell lymphopenia (n = 1) | 0/1 | ||||
WAS (n = 1) | 1/1 | ||||
Italy | Anti-S Ab; anti-RBD ab; Spike-specific T-cells | XLA (n = 7) | Anti-S ab + in 0/7 | NA | 23 |
Anti-RBD ab + in 0/7 | |||||
Spike-specific T-cells in 5/6† | |||||
Czech Republic | Anti-S Ab; anti-N Ab; IGRA | STAT1 GOF (n = 7) | Anti-S Ab + in 6/7 | None | 89 |
IGRA + in 6/6† |
Ab, antibody; ALPS, autoimmune lymphoproliferative syndrome; ADA, adenosine deaminase; ADA2, adenosine deaminase 2; AIRE, autoimmune regulator; ALPS, autoimmune lymphoproliferative syndrome; anti-S, anti-Spike; anti-N, anti-Nucleocapsid; APECED, autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy; APDS2, activated PI3K Delta syndrome 2; CD40L, CD40 ligand; CGD, chronic granulomatous disease; CHH, cartilage hair hypoplasia; CID, combined immunodeficiency; CTLA-4, cytotoxic T-Lymphocyte antigen 4; CVID, common variable immunodeficiency; DN, dominant negative; FOXN1, forkhead box N1; GOF, gain-of-function; IFNGR1, interferon-gamma receptor; IGRA, spike protein–specific T-cell responses evaluated using SARS-CoV-2 IFN-γ release assay; MAGT1, magnesium transporter 1; NA, not available; NEMO, NF-kappa B essential modulator; PIK3R1, phosphoinositide-3-kinase regulatory subunit 1; RAG, recombination-activating genes; RALD, ras-associated autoimmune leukoproliferative disorder; RBD, receptor-binding domain; SASH3, SAM and SH3 domain containing 3; SpAD, specific antibody deficiency; STAT, signal transducer and activator of transcription; WAS, Wiskott-Aldrich syndrome; WHIM, warts, hypogammaglobulinemia, infections, and myelokathexis; XLA, X-linked agammaglobulinemia. *CVID have been included in the analysis only for the studies in which disaggregated data were not available. †Not performed in one patient.
Conclusions
The large number of cases of SARS-CoV-2 infection reported in patients with IEI has led to the identification of differences in the risk of developing severe disease among different forms of IEI. In some cases, such as in patients with impaired production of or response to type I IFN, the genetic defect favors active replication of the virus and spreading to the lower respiratory tract. In other forms of IEI (combined immunodeficiencies, antibody defects, patients within 1 yr of HSCT), the inadequacy of adaptive immune responses may impede control of the infection. Finally, hyperinflammation associated with some forms of PIRD may exacerbate some of the features of COVID-19, with an increased risk of multisystem involvement. In this regard, knowledge of the underlying gene defect may be essential to prompt timely and appropriate preventive and therapeutic interventions to counteract the risk of severe disease in patients with IEI. At the same time, in case of severe SARS-CoV-2 infection, especially in children and in adults with no apparent comorbidities, it is fundamental to consider an IEI as an underlying cause and further investigations, including genetic tests and autoantibodies measurement, should be performed (131).
The results reported in the studies published so far indicate that COVID-19 vaccines are efficacious and safe in patients with IEI, supporting the current recommendation that these patients should be vaccinated against COVID-19. However, additional studies are needed to assess the durability and robustness of the immune responses to COVID-19 vaccines in IEI patients. Larger patient cohorts are required to drive definite conclusions on vaccine efficacy in this heterogeneous population. There is an urgent need for further studies evaluating both humoral and cellular responses in different IEI cohorts longitudinally over time. By combining these data with analysis of the frequency and severity of breakthrough cases among patients with IEI and in the general population, it will be possible to obtain a more solid indication of the possible need for additional vaccine boosts in patients with IEI.
Acknowledgments
This project was supported by grants from the National Institutes of Health Intramural Research Program, National Institute of Allergy and Infectious Diseases (AI001270).
No conflicts of interest, financial or otherwise, are declared by the authors.
Author Contributions: O.M.D. and R.C. conceived and designed research; O.M.D. and R.C. drafted manuscript; O.M.D., R.C., and L.D.N. edited and revised manuscript; O.M.D., R.C., and L.D.N. approved final version of manuscript.
References
- 1. Vijenthira A, Gong IY, Fox TA, Booth S, Cook G, Fattizzo B, Martin-Moro F, Razanamahery J, Riches JC, Zwicker J, Patell R, Vekemans MC, Scarfo L, Chatzikonstantinou T, Yildiz H, Lattenist R, Mantzaris I, Wood WA, Hicks LK. Outcomes of patients with hematologic malignancies and COVID-19: a systematic review and meta-analysis of 3377 patients. Blood 136: 2881–2892, 2020. doi: 10.1182/blood.2020008824. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Azzi Y, Bartash R, Scalea J, Loarte-Campos P, Akalin E. COVID-19 and solid organ transplantation: a review article. Transplantation 105: 37–55, 2021. doi: 10.1097/TP.0000000000003523. [DOI] [PubMed] [Google Scholar]
- 3. Marcus N, Frizinsky S, Hagin D, Ovadia A, Hanna S, Farkash M, Maoz-Segal R, Agmon-Levin N, Broides A, Nahum A, Rosenberg E, Kuperman AA, Dinur-Schejter Y, Berkun Y, Toker O, Goldberg S, Confino-Cohen R, Scheuerman O, Badarneh B, Epstein-Rigbi N, Etzioni A, Dalal I, Somech R. Minor clinical impact of COVID-19 pandemic on patients with primary immunodeficiency in Israel. Front Immunol 11: 614086, 2020. doi: 10.3389/fimmu.2020.614086. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Meyts I, Bucciol G, Quinti I, Neven B, Fischer A, Seoane E, , et al. Coronavirus disease 2019 in patients with inborn errors of immunity: an international study. J Allergy Clin Immunol 147: 520–531, 2021. doi: 10.1016/j.jaci.2020.09.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Milito C, Lougaris V, Giardino G, Punziano A, Vultaggio A, Carrabba M, , et al. Clinical outcome, incidence, and SARS-CoV-2 infection-fatality rates in Italian patients with inborn errors of immunity. J Allergy Clin Immunol Pract 9: 2904–2906.e2, 2021. doi: 10.1016/j.jaip.2021.04.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Castano-Jaramillo LM, Yamazaki-Nakashimada MA, O’Farrill-Romanillos PM, Muzquiz Zermeno D, Scheffler Mendoza SC, Venegas Montoya E, Garcia Campos JA, Sanchez-Sanchez LM, Gamez Gonzalez LB, Ramirez Lopez JM, Bustamante Ogando JC, Vasquez-Echeverri E, Medina Torres EA, Lopez-Herrera G, Blancas Galicia L, Berron Ruiz L, Staines-Boone AT, Espinosa-Padilla SE, Segura Mendez NH, Lugo Reyes SO. COVID-19 in the context of inborn errors of immunity: a case series of 31 patients from Mexico. J Clin Immunol 41: 1463–1478, 2021. doi: 10.1007/s10875-021-01077-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Goudouris ES, Pinto-Mariz F, Mendonca LO, Aranda CS, Guimaraes RR, Kokron C, , et al. Outcome of SARS-CoV-2 infection in 121 patients with inborn errors of immunity: a cross-sectional study. J Clin Immunol 41: 1479–1489, 2021. doi: 10.1007/s10875-021-01066-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Karakoc Aydiner E, Bilgic Eltan S, Babayeva R, Aydiner O, Kepenekli E, Kolukisa B, Sefer AP, Yalcin Gungoren E, Karabiber E, Yucel EO, Ozdemir O, Kiykim A, Artac H, Yakici N, Yalcin K, Cokugras H, Celkan TT, Orhan F, Yesilipek MA, Baris S, Ozen A. Adverse COVID-19 outcomes in immune deficiencies: inequality exists between subclasses. Allergy 77: 282–295, 2022. doi: 10.1111/all.15025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Milota T, Sobotkova M, Smetanova J, Bloomfield M, Vydlakova J, Chovancova Z, Litzman J, Hakl R, Novak J, Malkusova I, Hanzlikova J, Jilek D, Hutyrova B, Novak V, Krcmova I, Sediva A, Kralickova P. Risk factors for severe COVID-19 and hospital admission in patients with inborn errors of immunity–results from a multicenter nationwide study. Front Immunol 13: 835770, 2022. doi: 10.3389/fimmu.2022.835770. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Shields AM, Burns SO, Savic S, Richter AG, UK PIN COVID-19 Consortium. COVID-19 in patients with primary and secondary immunodeficiency: the United Kingdom experience. J Allergy Clin Immunol 147: 870–875.e1, 2021. doi: 10.1016/j.jaci.2020.12.620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Al-Saud B, Hazzazi KM, Mohammed R, Al Najjar A, Al Hazmi T, Monies D, Alkuraya FS. SARS-CoV-2-related acute respiratory distress syndrome uncovers a patient with severe combined immunodeficiency disease. J Clin Immunol 41: 1507–1510, 2021. doi: 10.1007/s10875-021-01063-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Asano T, Boisson B, Onodi F, Matuozzo D, Moncada-Velez M, Maglorius Renkilaraj MR, , et al. X-linked recessive TLR7 deficiency in ∼1% of men under 60 years old with life-threatening COVID-19. Sci Immunol 6: eabl4348, 2021. doi: 10.1126/sciimmunol.abl4348. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Bastard P, Gervais A, Le Voyer T, Rosain J, Philippot Q, Manry J, , et al. Autoantibodies neutralizing type I IFNs are present in ∼4% of uninfected individuals over 70 years old and account for ∼20% of COVID-19 deaths. Sci Immunol 6: eabl4340, 2021. doi: 10.1126/sciimmunol.abl4340. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Bastard P, Rosen LB, Zhang Q, Michailidis E, Hoffmann HH, Zhang Y, , et al. Autoantibodies against type I IFNs in patients with life-threatening COVID-19. Science 370: eabd4585, 2020. doi: 10.1126/science.abd4585. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Chou J, Platt CD, Habiballah S, Nguyen AA, Elkins M, Weeks S, Peters Z, Day-Lewis M, Novak T, Armant M, Williams L, Rockowitz S, Sliz P, Williams DA, Randolph AG, Geha RS, Taking on COVID-19 Together Study Investigators. Mechanisms underlying genetic susceptibility to multisystem inflammatory syndrome in children (MIS-C). J Allergy Clin Immunol 148: 732–738.e1, 2021.doi: 10.1016/j.jaci.2021.06.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Lee PY, Platt CD, Weeks S, Grace RF, Maher G, Gauthier K, Devana S, Vitali S, Randolph AG, McDonald DR, Geha RS, Chou J. Immune dysregulation and multisystem inflammatory syndrome in children (MIS-C) in individuals with haploinsufficiency of SOCS1. J Allergy Clin Immunol 146: 1194–1200.e1, 2020. doi: 10.1016/j.jaci.2020.07.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Levy M, Recher M, Hubert H, Javouhey E, Flechelles O, Leteurtre S, Angoulvant F. Multisystem inflammatory syndrome in children by COVID-19 vaccination status of adolescents in France. JAMA 327: 281–283, 2022. doi: 10.1001/jama.2021.23262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Zhang Q, Bastard P, Liu Z, Le Pen J, Moncada-Velez M, Chen J, , et al. Inborn errors of type I IFN immunity in patients with life-threatening COVID-19. Science 370: eabd4570, 2020. doi: 10.1126/science.abd4570. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Bergman P, Blennow O, Hansson L, Mielke S, Nowak P, Chen P, , et al. Safety and efficacy of the mRNA BNT162b2 vaccine against SARS-CoV-2 in five groups of immunocompromised patients and healthy controls in a prospective open-label clinical trial. EBioMedicine 74: 103705, 2021. doi: 10.1016/j.ebiom.2021.103705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Delmonte OM, Bergerson JR, Burbelo PD, Durkee-Shock JR, Dobbs K, Bosticardo M, Keller MD, McDermott DH, Rao VK, Dimitrova D, Quiros-Roldan E, Imberti L, Ferre EM, Schmitt M, Lafeer C, Pfister J, Shaw D, Draper D, Truong M, Ulrick J, DiMaggio T, Urban A, Holland SM, Lionakis MS, Cohen JI, Ricotta EE, Notarangelo LD, Freeman AF. Antibody responses to the SARS-CoV-2 vaccine in individuals with various inborn errors of immunity. J Allergy Clin Immunol 148: 1192–1197, 2021. doi: 10.1016/j.jaci.2021.08.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Pham MN, Murugesan K, Banaei N, Pinsky BA, Tang M, Hoyte E, Lewis DB, Gernez Y. Immunogenicity and tolerability of COVID-19 messenger RNA vaccines in primary immunodeficiency patients with functional B-cell defects. J Allergy Clin Immunol 149: 907–911.e3, 2022. doi: 10.1016/j.jaci.2021.11.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Pulvirenti F, Fernandez Salinas A, Milito C, Terreri S, Piano Mortari E, Quintarelli C, Di Cecca S, Lagnese G, Punziano A, Guercio M, Bonanni L, Auria S, Villani F, Albano C, Locatelli F, Spadaro G, Carsetti R, Quinti I. B cell response induced by SARS-CoV-2 infection is boosted by the BNT162b2 vaccine in primary antibody deficiencies. Cells 10: 2915, 2021. doi: 10.3390/cells10112915. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Salinas AF, Mortari EP, Terreri S, Quintarelli C, Pulvirenti F, Di Cecca S, Guercio M, Milito C, Bonanni L, Auria S, Romaggioli L, Cusano G, Albano C, Zaffina S, Perno CF, Spadaro G, Locatelli F, Carsetti R, Quinti I. SARS-CoV-2 vaccine induced atypical immune responses in antibody defects: everybody does their best. J Clin Immunol 41: 1709–1722, 2021. doi: 10.1007/s10875-021-01133-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Castano-Jaramillo LM, Yamazaki-Nakashimada MA, Scheffler Mendoza SC, Bustamante-Ogando JC, Espinosa-Padilla SE, Lugo Reyes SO. A male infant with COVID-19 in the context of ARPC1B deficiency. Pediatr Allergy Immunol 32: 199–201, 2021. doi: 10.1111/pai.13322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Alkan G, Artac H, Oz SK, Emiroglu M. Management of COVID-19 pneumonia in a child with NEMO deficiency. Immunol Res 69: 391–393, 2021. doi: 10.1007/s12026-021-09184-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Babaei M, Kanannejad Z, Sepahi N, Alyasin S. The effect of COVID-19 pandemic on patients with primary immunodeficiency: a cohort study. Iran J Med Sci 47: 162–166, 2022. doi: 10.30476/IJMS.2022.92862.2415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Bradley RE, Ponsford MJ, Scurr MJ, Godkin A, Jolles S, Immunodeficiency Centre for Wales. Persistent COVID-19 infection in wiskott-aldrich syndrome cleared following therapeutic vaccination: a case report. J Clin Immunol 42: 32–35, 2022. doi: 10.1007/s10875-021-01158-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Delavari S, Abolhassani H, Abolnezhadian F, Babaha F, Iranparast S, Ahanchian H, Moazzen N, Nabavi M, Arshi S, Fallahpour M, Bemanian MH, Shokri S, Momen T, Sadeghi-Shabestari M, Molatefi R, Shirkani A, Vosughimotlagh A, Safarirad M, Sharifzadeh M, Pashangzadeh S, Salami F, Shirmast P, Rezaei A, Moeini Shad T, Mohraz M, Rezaei N, Hammarstrom L, Yazdani R, Aghamohamamdi A. Impact of SARS-CoV-2 pandemic on patients with primary immunodeficiency. J Clin Immunol 41: 345–355, 2021. doi: 10.1007/s10875-020-00928-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Deya-Martinez A, Garcia-Garcia A, Gonzalez-Navarro EA, Yiyi L, Vlagea A, Jordan I, Fumado V, Fortuny C, Espanol M, Launes C, Esteve-Sole A, Juan M, Pascal M, Alsina L. COVID-19 in children and young adults with moderate/severe inborn errors of immunity in a high burden area in pre-vaccine era. Clin Immunol 230: 108821, 2021. doi: 10.1016/j.clim.2021.108821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Esenboga S, Ocak M, Akarsu A, Bildik HN, Cagdas D, Iskit AT, Tezcan I. COVID-19 in patients with primary immunodeficiency. J Clin Immunol 41: 1515–1522, 2021. doi: 10.1007/s10875-021-01065-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Gabryszewski SJ, England RN, Sun D, Gentile TL, Hochgertel W, Jyonouchi S, Silverman M, Zaoutis T, Sullivan KE, Henrickson SE. Self-limited COVID-19 in a patient with artemis hypomorphic SCID. J Clin Immunol 41: 1745–1747, 2021. doi: 10.1007/s10875-021-01093-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Giardino G, Milito C, Lougaris V, Punziano A, Carrabba M, Cinetto F, , et al. The impact of SARS-CoV-2 infection in patients with inborn errors of immunity: the Experience of the Italian Primary Immunodeficiencies Network (IPINet). J Clin Immunol. In press. doi: 10.1007/s10875-022-01264-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Gotzinger F, Santiago-Garcia B, Noguera-Julian A, Lanaspa M, Lancella L, Calo Carducci FI, Gabrovska N, Velizarova S, Prunk P, Osterman V, Krivec U, Lo Vecchio A, Shingadia D, Soriano-Arandes A, Melendo S, Lanari M, Pierantoni L, Wagner N, L'Huillier AG, Heininger U, Ritz N, Bandi S, Krajcar N, Roglic S, Santos M, Christiaens C, Creuven M, Buonsenso D, Welch SB, Bogyi M, Brinkmann F, Tebruegge M, ptbnet COVID-19 Study Group. COVID-19 in children and adolescents in Europe: a multinational, multicentre cohort study. Lancet Child Adolesc Health 4: 653–661, 2020. doi: 10.1016/S2352-4642(20)30177-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Hariharan SV, Muthusamy S, Asokan SK. Persistent viral shedding after SARS-CoV-2 infection in an infant with severe combined immunodeficiency. Indian J Pediatr 89: 94, 2022. doi: 10.1007/s12098-021-03935-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Ho HE, Mathew S, Peluso MJ, Cunningham-Rundles C. Clinical outcomes and features of COVID-19 in patients with primary immunodeficiencies in New York City. J Allergy Clin Immunol Pract 9: 490–493.e2, 2021. doi: 10.1016/j.jaip.2020.09.052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Keitel V, Bode JG, Feldt T, Walker A, Muller L, Kunstein A, Klindt C, Killer A, Senff T, Timm J, Ostermann P, Damagnez M, Lubke N, Adams O, Schaal H, Antoch G, Neubert J, Albrecht P, Meuth S, Elben S, Mohring A, Fischer JC, Bolke E, Hoenig M, Schulz AS, Luedde T, Jensen B. Case report: convalescent plasma achieves SARS-CoV-2 viral clearance in a patient with persistently high viral replication over 8 weeks due to severe combined immunodeficiency (SCID) and graft failure. Front Immunol 12: 645989, 2021. doi: 10.3389/fimmu.2021.645989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Lang-Meli J, Fuchs J, Mathe P, Ho HE, Kern L, Jaki L, Rusignuolo G, Mertins S, Somogyi V, Neumann-Haefelin C, Trinkmann F, Muller M, Thimme R, Umhau M, Quinti I, Wagner D, Panning M, Cunningham-Rundles C, Laubner K, Warnatz K. Case series: convalescent plasma therapy for patients with COVID-19 and primary antibody deficiency. J Clin Immunol 42: 253–265, 2022. doi: 10.1007/s10875-021-01193-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Moazzen N, Ahanchian H, Aelami MH, Asiyon H, Astaneh M, Naeimi AM, Rezaei N. COVID-19 in children with inborn errors of immunity: clinical scenarios. Am J Clin Exp Immunol 10: 77–85, 2021. [PMC free article] [PubMed] [Google Scholar]
- 39. van Oers NS, Hanners NW, Sue PK, Aquino V, Li QZ, Schoggins JW, Wysocki CA. SARS-CoV-2 infection associated with hepatitis in an infant with X-linked severe combined immunodeficiency. Clin Immunol 224: 108662, 2021. doi: 10.1016/j.clim.2020.108662. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Zamperlini-Netto G, Fernandes JF, Garcia JL, Ribeiro AA, Camargo LF, de Moraes Terra C, Hamerschlak N. COVID-19 after hematopoietic stem cell transplantation: report of two children. Bone Marrow Transplant 56: 713–715, 2021. doi: 10.1038/s41409-020-01041-8. [DOI] [PubMed] [Google Scholar]
- 41. Abers MS, Delmonte OM, Ricotta EE, Fintzi J, Fink DL, de Jesus AA, , et al. An immune-based biomarker signature is associated with mortality in COVID-19 patients. JCI Insight 6: e144455, 2021. doi: 10.1172/jci.insight.144455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Guirao JJ, Cabrera CM, Jimenez N, Rincon L, Urra JM. High serum IL-6 values increase the risk of mortality and the severity of pneumonia in patients diagnosed with COVID-19. Mol Immunol 128: 64–68, 2020. doi: 10.1016/j.molimm.2020.10.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Abraham RS, Marshall JM, Kuehn HS, Rueda CM, Gibbs A, Guider W, Stewart C, Rosenzweig SD, Wang H, Jean S, Peeples M, King T, Hunt WG, Honegger JR, Ramilo O, Mustillo PJ, Mejias A, Ardura MI, Shimamura M. Severe SARS-CoV-2 disease in the context of a NF-kappaB2 loss-of-function pathogenic variant. J Allergy Clin Immunol 147: 532–544.e1, 2021. doi: 10.1016/j.jaci.2020.09.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Al Yazidi LS, Al Rawahi H, Al Busaidi I, Al Tamemi S. Covid-19 and primary immunodeficiency: one-year experience. J Paediatr Child Health 57: 594, 2021. doi: 10.1111/jpc.15433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Aljaberi R, Wishah K. Positive outcome in a patient with coronavirus disease 2019 and common variable immunodeficiency after intravenous immunoglobulin. Ann Allergy Asthma Immunol 125: 349–350, 2020. doi: 10.1016/j.anai.2020.06.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Almontasheri A, Al-Husayni F, Alsuraihi AK, Binyahib H, Albanna AS. The clinical course of COVID-19 pneumonia in a 19-Year-old man on intravenous immunoglobulin replacement therapy for X-linked agammaglobulinemia. Am J Case Rep 22: e929447, 2021. doi: 10.12659/AJCR.929447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Buckland MS, Galloway JB, Fhogartaigh CN, Meredith L, Provine NM, Bloor S, , et al. Treatment of COVID-19 with remdesivir in the absence of humoral immunity: a case report. Nat Commun 11: 6385, 2020. doi: 10.1038/s41467-020-19761-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Cohen B, Rubinstein R, Gans MD, Deng L, Rubinstein A, Eisenberg R. COVID-19 infection in 10 common variable immunodeficiency patients in New York City. J Allergy Clin Immunol Pract 9: 504–507.e1, 2021. doi: 10.1016/j.jaip.2020.11.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Delgado-Fernandez M, Garcia-Gemar GM, Fuentes-Lopez A, Munoz-Perez MI, Oyonarte-Gomez S, Ruiz-Garcia I, Martin-Carmona J, Sanz-Canovas J, Castano-Carracedo MA, Reguera-Iglesias JM, Ruiz-Mesa JD. Treatment of COVID-19 with convalescent plasma in patients with humoral immunodeficiency–three consecutive cases and review of the literature. Enferm Infecc Microbiol Clin (Engl Ed) 6: S0213-005X(21)00035-5, 2021. doi: 10.1126/sciimmunol.abl4348. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Devassikutty FM, Jain A, Edavazhippurath A, Joseph MC, Peedikayil MM, Scaria V, Sandhya P, Govindaraj GM. X-linked agammaglobulinemia and COVID-19: two case reports and review of literature. Pediatr Allergy Immunol Pulmonol 34: 115–118, 2021. doi: 10.1089/ped.2021.0002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Drabe CH, Hansen AE, Rasmussen LD, Larsen OD, Moller A, Mogensen TH, Helweg-Larsen J, Katzenstein TL. Low morbidity in Danish patients with common variable immunodeficiency disorder infected with severe acute respiratory syndrome coronavirus 2. Infect Dis (Lond) 53: 953–958, 2021. doi: 10.1080/23744235.2021.1957144. [DOI] [PubMed] [Google Scholar]
- 52. Fill L, Hadney L, Graven K, Persaud R, Hostoffer R. The clinical observation of a patient with common variable immunodeficiency diagnosed as having coronavirus disease 2019. Ann Allergy Asthma Immunol 125: 112–114, 2020. doi: 10.1016/j.anai.2020.04.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Guchelaar NA, van Laar JA, Hermans MA, van der Houwen TB, Atmaca S, van Maaren MS, Brkic Z, van Daele PL, Dalm V, van Hagen PM, Rombach SM. Characteristics of COVID-19 infection and antibody formation in patients known at a tertiary immunology department. J Transl Autoimmun 4: 100084, 2021. doi: 10.1016/j.jtauto.2021.100084. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Gupta S, Su H, Narsai T, Agrawal S. SARS-CoV-2-Associated T-cell responses in the presence of humoral immunodeficiency. Int Arch Allergy Immunol 182: 195–209, 2021. doi: 10.1159/000514193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Iaboni A, Wong N, Betschel SD. A patient with X-linked agammaglobulinemia and COVID-19 infection treated with remdesivir and convalescent plasma. J Clin Immunol 41: 923–925, 2021. doi: 10.1007/s10875-021-00983-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Kinoshita H, Durkee-Shock J, Jensen-Wachspress M, Kankate VV, Lang H, Lazarski CA, Keswani A, Webber KC, Montgomery-Recht K, Walkiewicz M, Notarangelo LD, Burbelo PD, Fuss I, Cohen JI, Bollard CM, Keller MD. Robust antibody and T cell responses to SARS-CoV-2 in patients with antibody deficiency. J Clin Immunol 41: 1146–1153, 2021. doi: 10.1007/s10875-021-01046-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Kuster JK, Unlu S, Makin TA, Par-Young J, Simonov M, Shafi S, Balanda M, Randolph C, Steele R, Hsu FI, Price C, Kohli-Pamnani A, Borish L, Lawrence MG, Kang I, Shin JJ. Low IgG trough and lymphocyte subset counts are associated with hospitalization for COVID-19 in patients with primary antibody deficiency. J Allergy Clin Immunol Pract 10: 633–636.e3, 2022. doi: 10.1016/j.jaip.2021.11.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Loh SY, Bassett J, Hoodless EJ, Walshaw M. Possible COVID-19 reinfection in a patient with X-linked agammaglobulinaemia. BMJ Case Rep 14: e240765, 2021. doi: 10.1136/bcr-2020-240765. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. London J, Boutboul D, Lacombe K, Pirenne F, Heym B, Zeller V, Baudet A, Ouedrani A, Berezne A. Severe COVID-19 in patients with B cell alymphocytosis and response to convalescent plasma therapy. J Clin Immunol 41: 356–361, 2021. doi: 10.1007/s10875-020-00904-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Milosevic I, Jovanovic J, Stevanovic O. Atypical course of COVID-19 in patient with Bruton agammaglobulinemia. J Infect Dev Ctries 14: 1248–1251, 2020. doi: 10.3855/jidc.13840. [DOI] [PubMed] [Google Scholar]
- 61. Mira E, Yarce OA, Ortega C, Fernandez S, Pascual NM, Gomez C, Alvarez MA, Molina IJ, Lama R, Santamaria M. Rapid recovery of a SARS-CoV-2-infected X-linked agammaglobulinemia patient after infusion of COVID-19 convalescent plasma. J Allergy Clin Immunol Pract 8: 2793–2795, 2020. doi: 10.1016/j.jaip.2020.06.046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Mullur J, Wang A, Feldweg A. A fatal case of coronavirus disease 2019 in a patient with common variable immunodeficiency. Ann Allergy Asthma Immunol 126: 90–92, 2021. doi: 10.1016/j.anai.2020.08.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Palomba E, Carrabba M, Zuglian G, Alagna L, Saltini P, Fortina V, Hu C, Bandera A, Fabio G, Gori A, Muscatello A. Treatment of SARS-CoV-2 relapse with remdesivir and neutralizing antibodies cocktail in a patient with X-linked agammaglobulinaemia. Int J Infect Dis 110: 338–340, 2021. doi: 10.1016/j.ijid.2021.07.064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Quinti I, Lougaris V, Milito C, Cinetto F, Pecoraro A, Mezzaroma I, Mastroianni CM, Turriziani O, Bondioni MP, Filippini M, Soresina A, Spadaro G, Agostini C, Carsetti R, Plebani A. A possible role for B cells in COVID-19? Lesson from patients with agammaglobulinemia. J Allergy Clin Immunol 146: 211–213.e4, 2020. doi: 10.1016/j.jaci.2020.04.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Steiner S, Schwarz T, Corman VM, Gebert L, Kleinschmidt MC, Wald A, Gläser S, Kruse JM, Zickler D, Peric A, Meisel C, Meyer T, Staudacher OL, Wittke K, Kedor C, Bauer S, Besher NA, Kalus U, Pruß A, Drosten C, Volk HD, Scheibenbogen C, Hanitsch LG. SARS-CoV-2 T cell response in severe and fatal COVID-19 in primary antibody deficiency patients without specific humoral immunity. Front Immunol 13: 840126, 2022. doi: 10.3389/fimmu.2022.840126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Van Damme KF, Tavernier S, Van Roy N, De Leeuw E, Declercq J, Bosteels C, Maes B, De Bruyne M, Bogaert D, Bosteels V, Hoste L, Naesens L, Maes P, Grifoni A, Weiskopf D, Sette A, Depuydt P, Van Braeckel E, Haerynck F, Lambrecht BN. Case report: convalescent plasma, a targeted therapy for patients with CVID and severe COVID-19. Front Immunol 11: 596761, 2020. doi: 10.3389/fimmu.2020.596761. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Weifenbach N, Jung A, Lotters S. COVID-19 infection in CVID patients: what we know so far. Immun Inflamm Dis 9: 632–634, 2021. doi: 10.1002/iid3.450. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Zhou X, Zhou J, Zhao J. Recurrent pneumonia in a patient with new coronavirus infection after discharge from hospital for insufficient antibody production: a case report. BMC Infect Dis 20: 500, 2020. doi: 10.1186/s12879-020-05231-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Tangye SG, Bucciol G, Meyts I. Mechanisms underlying host defense and disease pathology in response to severe acute respiratory syndrome (SARS)-CoV2 infection: insights from inborn errors of immunity. Curr Opin Allergy Clin Immunol 21: 515–524, 2021. doi: 10.1097/ACI.0000000000000786. [DOI] [PubMed] [Google Scholar]
- 70. Milito C, Soccodato V, Auria S, Pulvirenti F, Quinti I. COVID-19 in complex common variable immunodeficiency patients affected by lung diseases. Curr Opin Allergy Clin Immunol 21: 535–544, 2021. doi: 10.1097/ACI.0000000000000789. [DOI] [PubMed] [Google Scholar]
- 71. Bastard P, Orlova E, Sozaeva L, Levy R, James A, Schmitt MM, , et al. Preexisting autoantibodies to type I IFNs underlie critical COVID-19 pneumonia in patients with APS-1. J Exp Med 218: e20210554, 2021. doi: 10.1084/jem.20210554. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Beccuti G, Ghizzoni L, Cambria V, Codullo V, Sacchi P, Lovati E, Mongodi S, Iotti GA, Mojoli F. A COVID-19 pneumonia case report of autoimmune polyendocrine syndrome type 1 in Lombardy, Italy: letter to the editor. J Endocrinol Invest 43: 1175–1177, 2020. doi: 10.1007/s40618-020-01323-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Carpino A, Buganza R, Matarazzo P, Tuli G, Pinon M, Calvo PL, Montin D, Licciardi F, De Sanctis L. Autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy in two siblings: same mutations but very different phenotypes. Genes (Basel) 12: 169, 2021. doi: 10.3390/genes12020169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Lange M, Linden T, Muller HL, Flasskuehler MA, Koester H, Lehmberg K, Ledig S, Ehl S, Heep A, Beske F. Primary haemophagocytic lymphohistiocytosis (Chediak-Higashi Syndrome) triggered by acute SARS-CoV-2 infection in a six-week-old infant. Br J Haematol 195: 198–200, 2021. doi: 10.1111/bjh.17669. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Lemarquis A, Campbell T, Aranda-Guillen M, Hennings V, Brodin P, Kampe O, Blennow K, Zetterberg H, Wenneras C, Eriksson K, Landegren N, Bryceson Y, Berg S, Ekwall O. Severe COVID-19 in an APS1 patient with interferon autoantibodies treated with plasmapheresis. J Allergy Clin Immunol 148: 96–98, 2021. doi: 10.1016/j.jaci.2021.03.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Meisel C, Akbil B, Meyer T, Lankes E, Corman VM, Staudacher O, Unterwalder N, Kolsch U, Drosten C, Mall MA, Kallinich T, Schnabel D, Goffinet C, von Bernuth H. Mild COVID-19 despite autoantibodies against type I IFNs in autoimmune polyendocrine syndrome type 1. J Clin Invest 131: e150867, 2021. doi: 10.1172/JCI150867. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Wahlster L, Weichert-Leahey N, Trissal M, Grace RF, Sankaran VG. COVID-19 presenting with autoimmune hemolytic anemia in the setting of underlying immune dysregulation. Pediatr Blood Cancer 67: e28382, 2020. doi: 10.1002/pbc.28382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Bucciol G, Tangye SG, Meyts I. Coronavirus disease 2019 in patients with inborn errors of immunity: lessons learned. Curr Opin Pediatr 33: 648–656, 2021. doi: 10.1097/MOP.0000000000001062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Ferre EM, Rose SR, Rosenzweig SD, Burbelo PD, Romito KR, Niemela JE, , et al. Redefined clinical features and diagnostic criteria in autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy. JCI Insight 1: e88782, 2016. doi: 10.1172/jci.insight.88782. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Ferre EM, Schmitt MM, Ochoa S, Rosen LB, Shaw ER, Burbelo PD, Stoddard JL, Rampertaap S, DiMaggio T, Bergerson JR, Rosenzweig SD, Notarangelo LD, Holland SM, Lionakis MS. SARS-CoV-2 spike protein-directed monoclonal antibodies may ameliorate COVID-19 complications in APECED patients. Front Immunol 12: 720205, 2021. doi: 10.3389/fimmu.2021.720205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Vignesh P, Mondal S, Sudhakar M, Sharma YK, Bansal A, Singh M, Sachdeva MU. SARS-CoV-2 infection in a child with severe congenital neutropenia. J Clin Immunol 41: 1165–1168, 2021. doi: 10.1007/s10875-021-01054-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Abolhassani H, Landegren N, Bastard P, Materna M, Modaresi M, Du L, Aranda-Guillen M, Sardh F, Zuo F, Zhang P, Marcotte H, Marr N, Khan T, Ata M, Al-Ali F, Pescarmona R, Belot A, Beziat V, Zhang Q, Casanova JL, Kampe O, Zhang SY, Hammarstrom L, Pan-Hammarstrom Q. Inherited IFNAR1 deficiency in a child with both critical COVID-19 pneumonia and multisystem inflammatory syndrome. J Clin Immunol 42: 471–483, 2022. doi: 10.1007/s10875-022-01215-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Akahane D, Otsuki S, Hasegwa D, Watanabe H, Gotoh A. Dexamethasone treatment for COVID-19-related lung injury in an adult with WHIM syndrome. J Clin Immunol 42: 270–273, 2022. doi: 10.1007/s10875-021-01185-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Guisado Hernandez P, Blanco Lobo P, Villaoslada I, de Felipe B, Lucena JM, Martin Gutierrez G, Castro MJ, Gutierrez Valencia A, Sanchez Codez MI, Gaboli M, Neth O, Olbrich P. SARS-CoV-2 infection in a pediatrics STAT1 GOF patient under Ruxolitinib therapy-a matter of balance? J Clin Immunol 41: 1502–1506, 2021. doi: 10.1007/s10875-021-01081-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Levy R, Bastard P, Lanternier F, Lecuit M, Zhang SY, Casanova JL. IFN-alpha2a therapy in two patients with inborn errors of TLR3 and IRF3 infected with SARS-CoV-2. J Clin Immunol 41: 26–27, 2021. doi: 10.1007/s10875-020-00933-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Levy R, Zhang P, Bastard P, Dorgham K, Melki I, Hadchouel A, Hartoularos GC, Neven B, Castelle M, Roy C, Toin T, Berteloot L, Bizien L, Abid H, Burgard M, Houhou-Fidouh N, Rozenberg F, Jouanguy E, Ye CJ, Gorochov G, Zhang Q, Casanova JL. Monoclonal antibody-mediated neutralization of SARS-CoV-2 in an IRF9-deficient child. Proc Natl Acad Sci U S A 118: e2114390118, 2021. doi: 10.1073/pnas.2114390118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Schmidt A, Peters S, Knaus A, Sabir H, Hamsen F, Maj C, Fazaal J, Sivalingam S, Savchenko O, Mantri A, Holzinger D, Neudorf U, Muller A, Ludwig KU, Krawitz PM, Engels H, Nothen MM, Bagci S. TBK1 and TNFRSF13B mutations and an autoinflammatory disease in a child with lethal COVID-19. NPJ Genom Med 6: 55, 2021. doi: 10.1038/s41525-021-00220-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. van der Made CI, Simons A, Schuurs-Hoeijmakers J, van den Heuvel G, Mantere T, Kersten S, van Deuren RC, Steehouwer M, van Reijmersdal SV, Jaeger M, Hofste T, Astuti G, Corominas Galbany J, van der Schoot V, van der Hoeven H, Hagmolen Of Ten Have W, Klijn E, van den Meer C, Fiddelaers J, de Mast Q, Bleeker-Rovers CP, Joosten LA, Yntema HG, Gilissen C, Nelen M, van der Meer JW, Brunner HG, Netea MG, van de Veerdonk FL, Hoischen A. Presence of genetic variants among young men with severe COVID-19. JAMA 324: 663–673, 2020. doi: 10.1001/jama.2020.13719. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Bloomfield M, Parackova Z, Hanzlikova J, Lastovicka J, Sediva A. Immunogenicity and safety of COVID-19 mRNA vaccine in STAT1 GOF patients. J Clin Immunol 42: 266–269, 2022. doi: 10.1007/s10875-021-01163-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Bourguiba R, Delplanque M, Vinit C, Ackermann F, Savey L, Grateau G, Hentgen V, Georgin-Lavialle S. Clinical course of COVID-19 in a cohort of 342 familial Mediterranean fever patients with a long-term treatment by colchicine in a French endemic area. Ann Rheum Dis 80: 539–540, 2021. doi: 10.1136/annrheumdis-2020-218707. [DOI] [PubMed] [Google Scholar]
- 91. Güven SC, Erden A, Karakaş Ö, Armağan B, Usul E, Omma A, Küçükşahin O. COVID-19 outcomes in patients with familial Mediterranean fever: a retrospective cohort study. Rheumatol Int 41: 715–719, 2021. doi: 10.1007/s00296-021-04812-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Haslak F, Varol SE, Gunalp A, Kaynar O, Yildiz M, Adrovic A, Sahin S, Kes G, Ayzit-Kilinc A, Akdeniz B, Onal P, Apaydin G, Aygun D, Arslan H, Kilic-Baskan A, Hepkaya E, Meral O, Barut K, Cokugras HC, Kasapcopur O. Comparisons of clinical features and outcomes of COVID-19 between patients with pediatric onset inflammatory rheumatic diseases and healthy children. J Clin Med 11: 2102, 2022. doi: 10.3390/jcm11082102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Kobak S. COVID-19 infection in a patient with FMF: does colchicine have a protective effect? Ann Rheum Dis 80: e39–e39, 2021. doi: 10.1136/annrheumdis-2020-217882. [DOI] [PubMed] [Google Scholar]
- 94. Alehashemi S, van Gelderen E, Rastegar A, de Jesus AA, Goldbach-Mansky R. Post-SARS-CoV-2 vaccine monitoring of disease flares in autoinflammatory diseases. J Clin Immunol 42: 732–735, 2022. doi: 10.1007/s10875-022-01225-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Grumach AS, Goudouris E, Dortas Junior S, Marcelino FC, Alonso ML, Martins RO, Arpon MA, Valle SO. COVID-19 affecting hereditary angioedema patients with and without C1 inhibitor deficiency. J Allergy Clin Immunol Pract 9: 508–510, 2021. doi: 10.1016/j.jaip.2020.11.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Holter JC, Pischke SE, de Boer E, Lind A, Jenum S, Holten AR, , et al. Systemic complement activation is associated with respiratory failure in COVID-19 hospitalized patients. Proc Natl Acad Sci USA 117: 25018–25025, 2020. doi: 10.1073/pnas.2010540117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Ramlall V, Thangaraj PM, Meydan C, Foox J, Butler D, Kim J, May B, De Freitas JK, Glicksberg BS, Mason CE, Tatonetti NP, Shapira SD. Immune complement and coagulation dysfunction in adverse outcomes of SARS-CoV-2 infection. Nat Med 26: 1609–1615, 2020. doi: 10.1038/s41591-020-1021-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Cos Esquius ML, Lopez Montesinos I, Gimeno Martinez R, Eguia Nunez J, Caballero-Rabasco MA, Sanchez Gonzalez B, Lopez Garcia A, Mellibovsky L. Severe COVID-19 pneumonia in Good syndrome with a favorable outcome. Clin Immunol 235: 108789, 2022. doi: 10.1016/j.clim.2021.108789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Duarte M, Faria L, Patronillo C, da Costa Fernandes S, Seara V. A case of severe COVID-19 in a patient with Good’s Syndrome. Eur J Case Rep Intern Med 8: 002976, 2021. doi: 10.12890/2021_002976. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Pozzi MR, Baronio M, Janetti MB, Gazzurelli L, Moratto D, Chiarini M, Plebani A, Lougaris V. Fatal SARS-CoV-2 infection in a male patient with Good's syndrome. Clin Immunol 223: 108644, 2021. doi: 10.1016/j.clim.2020.108644. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. eldstein LR, Rose EB, Horwitz SM, Collins JP, Newhams MM, Son MB, , et al. Multisystem inflammatory syndrome in U.S. children and adolescents. N Engl J Med 383: 334–346, 2020. doi: 10.1056/NEJMoa2021680. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Morris SB, Schwartz NG, Patel P, Abbo L, Beauchamps L, Balan S, Lee EH, Paneth-Pollak R, Geevarughese A, Lash MK, Dorsinville MS, Ballen V, Eiras DP, Newton-Cheh C, Smith E, Robinson S, Stogsdill P, Lim S, Fox SE, Richardson G, Hand J, Oliver NT, Kofman A, Bryant B, Ende Z, Datta D, Belay E, Godfred-Cato S. Case series of multisystem inflammatory syndrome in adults associated with SARS-CoV-2 Infection–United Kingdom and United States, March-August 2020. MMWR Morb Mortal Wkly Rep 69: 1450–1456, 2020. doi: 10.15585/mmwr.mm6940e1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Sacco K, Castagnoli R, Vakkilainen S, Liu C, Delmonte OM, Oguz C, , et al. Immunopathological signatures in multisystem inflammatory syndrome in children and pediatric COVID-19. Nat Med 28: 1050–1062, 2022. doi: 10.1038/s41591-022-01724-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Toubiana J, Cohen JF, Brice J, Poirault C, Bajolle F, Curtis W, Moulin F, Matczak S, Leruez M, Casanova JL, Chalumeau M, Taylor M, Allali S. Distinctive features of kawasaki disease following SARS-CoV-2 infection: a controlled study in Paris, France. J Clin Immunol 41: 526–535, 2021. doi: 10.1007/s10875-020-00941-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Verdoni L, Mazza A, Gervasoni A, Martelli L, Ruggeri M, Ciuffreda M, Bonanomi E, D'Antiga L. An outbreak of severe Kawasaki-like disease at the Italian epicentre of the SARS-CoV-2 epidemic: an observational cohort study. Lancet 395: 1771–1778, 2020. doi: 10.1016/S0140-6736(20)31103-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Alunno A, Najm A, Machado PM, Bertheussen H, Burmester GR, Carubbi F, De Marco G, Giacomelli R, Hermine O, Isaacs JD, Kone-Paut I, Magro-Checa C, McInnes IB, Meroni PL, Quartuccio L, Ramanan AV, Ramos-Casals M, Rodriguez Carrio J, Schulze-Koops H, Stamm TA, Tas SW, Terrier B, McGonagle DG, Mariette X. 2021 update of the EULAR points to consider on the use of immunomodulatory therapies in COVID-19. Ann Rheum Dis 81: 34–40, 2022. doi: 10.1136/annrheumdis-2021-221366. [DOI] [PubMed] [Google Scholar]
- 107. Chauvineau-Grenier A, Bastard P, Casanova JL, Rossi B. Autoantibodies neutralizing type I INFs may be associated with efficacy of Tocilizumab in COVID-19 pneumonia. J Clin Immunol, 2022. doi: 10.1007/s10875-022-01295-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Chen CY, Chen WC, Hsu CK, Chao CM, Lai CC. Clinical efficacy and safety of Janus kinase inhibitors for COVID-19: a systematic review and meta-analysis of randomized controlled trials. Int Immunopharmacol 99: 108027, 2021. doi: 10.1016/j.intimp.2021.108027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Stack M, Sacco K, Castagnoli R, Livinski AA, Notarangelo LD, Lionakis MS. BTK inhibitors for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2): a systematic review. Clin Immunol 230: 108816, 2021. doi: 10.1016/j.clim.2021.108816. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Vinh DC, Abel L, Bastard P, Cheng MP, Condino-Neto A, Gregersen PK, Haerynck F, Cicalese MP, Hagin D, Soler-Palacin P, Planas AM, Pujol A, Notarangelo LD, Zhang Q, Su HC, Casanova JL, Meyts I, COVID Human Genetic Effort. Harnessing type I IFN immunity against SARS-CoV-2 with early administration of IFN-beta. J Clin Immunol 41: 1425–1442, 2021. doi: 10.1007/s10875-021-01068-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Grainger R, Kim AH, Conway R, Yazdany J, Robinson PC. COVID-19 in people with rheumatic diseases: risks, outcomes, treatment considerations. Nat Rev Rheumatol 18: 191–204, 2022. doi: 10.1038/s41584-022-00755-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Sobh A, Bonilla FA. Vaccination in primary immunodeficiency disorders. J Allergy Clin Immunol Pract 4: 1066–1075, 2016. doi: 10.1016/j.jaip.2016.09.012. [DOI] [PubMed] [Google Scholar]
- 113. Sette A, Crotty S. Adaptive immunity to SARS-CoV-2 and COVID-19. Cell 184: 861–880, 2021. doi: 10.1016/j.cell.2021.01.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Dan J, da Silva Antunes R, Grifoni A, Weiskopf D, Crotty S, Sette A. Observations and perspectives on adaptive immunity to SARS-CoV-2. Clin Infect Dis 75: S24–S29, 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Abo-Helo N, Muhammad E, Ghaben-Amara S, Panasoff J, Cohen S. Specific antibody response of patients with common variable immunodeficiency to BNT162b2 coronavirus disease 2019 vaccination. Ann Allergy Asthma Immunol 127: 501–503, 2021. doi: 10.1016/j.anai.2021.07.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Amodio D, Ruggiero A, Sgrulletti M, Pighi C, Cotugno N, Medri C, Morrocchi E, Colagrossi L, Russo C, Zaffina S, Di Matteo G, Cifaldi C, Di Cesare S, Rivalta B, Pacillo L, Santilli V, Giancotta C, Manno EC, Ciofi Degli Atti M, Raponi M, Rossi P, Finocchi A, Cancrini C, Perno CF, Moschese V, Palma P. Humoral and cellular response following vaccination with the BNT162b2 mRNA COVID-19 vaccine in patients affected by primary immunodeficiencies. Front Immunol 12: 727850, 2021. doi: 10.3389/fimmu.2021.727850. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Arroyo-Sanchez D, Cabrera-Marante O, Laguna-Goya R, Almendro-Vazquez P, Carretero O, Gil-Etayo FJ, Suarez-Fernandez P, Perez-Romero P, Rodriguez de Frias E, Serrano A, Allende LM, Pleguezuelo D, Paz-Artal E. Immunogenicity of anti-SARS-CoV-2 vaccines in common variable immunodeficiency. J Clin Immunol 42: 240–252, 2022. doi: 10.1007/s10875-021-01174-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Fernandez Salinas A, Piano Mortari E, Terreri S, Milito C, Zaffina S, Perno CF, Locatelli F, Quinti I, Carsetti R. Impaired memory B-cell response to the Pfizer-BioNTech COVID-19 vaccine in patients with common variable immunodeficiency. J Allergy Clin Immunol 149: 76–77, 2022. doi: 10.1016/j.jaci.2021.08.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Gernez Y, Murugesan K, Cortales CR, Banaei N, Hoyte L, Pinsky BA, Lewis DB, Pham MN. Immunogenicity of a third COVID-19 messenger RNA vaccine dose in primary immunodeficiency disorder patients with functional B-cell defects. J Allergy Clin Immunol Pract 10: 1385–1388.e2, 2022.doi: 10.1016/j.jaip.2022.02.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Hagin D, Freund T, Navon M, Halperin T, Adir D, Marom R, Levi I, Benor S, Alcalay Y, Freund NT. Immunogenicity of Pfizer-BioNTech COVID-19 vaccine in patients with inborn errors of immunity. J Allergy Clin Immunol 148: 739–749, 2021. doi: 10.1016/j.jaci.2021.05.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Ponsford MJ, Evans K, Carne EM, Jolles S, Bramhall K, Grant L, McGuire F, Matthews A, Bradley R, Wijetilleka S, Pritchard A, Price CR, Farewell D, Cousins R, El-Shanawany T, Immunodeficiency Centre for Wales and Division of Population Medicine. COVID-19 vaccine uptake and efficacy in a national immunodeficiency cohort. J Clin Immunol 42: 728–731, 2022. doi: 10.1007/s10875-022-01223-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Romano C, Esposito S, Donnarumma G, Marrone A. Detection of neutralizing anti-severe acute respiratory syndrome coronavirus 2 antibodies in patients with common variable immunodeficiency after immunization with messenger RNA vaccines. Ann Allergy Asthma Immunol 127: 499–501, 2021. doi: 10.1016/j.anai.2021.07.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Sauerwein KT, Geier CB, Stemberger RF, Akyaman H, Illes P, Fischer MB, Eibl MM, Walter JE, Wolf HM. Antigen-specific CD4(+) T-cell activation in primary antibody deficiency after BNT162b2 mRNA COVID-19 vaccination. Front Immunol 13: 827048, 2022. doi: 10.3389/fimmu.2022.827048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Squire J, Joshi A. Seroconversion after coronavirus disease 2019 vaccination in patients with immune deficiency. Ann Allergy Asthma Immunol 127: 383–384, 2021. doi: 10.1016/j.anai.2021.05.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Friedmann D, Goldacker S, Peter HH, Warnatz K. Preserved cellular immunity upon influenza vaccination in most patients with common variable immunodeficiency. J Allergy Clin Immunol Pract 8: 2332–2340.e5, 2020.doi: 10.1016/j.jaip.2020.04.019. [DOI] [PubMed] [Google Scholar]
- 126. Quinti I, Locatelli F, Carsetti R. The immune response to SARS-CoV-2 vaccination: insights learned from adult patients with common variable immune deficiency. Front Immunol 12: 815404, 2021. doi: 10.3389/fimmu.2021.815404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Di Fusco M, Moran MM, Cane A, Curcio D, Khan F, Malhotra D, Surinach A, Miles A, Swerdlow D, McLaughlin JM, Nguyen JL. Evaluation of COVID-19 vaccine breakthrough infections among immunocompromised patients fully vaccinated with BNT162b2. J Med Econ 24: 1248–1260, 2021. doi: 10.1080/13696998.2021.2002063. [DOI] [PubMed] [Google Scholar]
- 128. Jalil M, Rowane M, Rajan J, Hostoffer R. Successful anti-SARS-CoV-2 spike protein antibody response to vaccination in MAGT1 deficiency. Allergy Rhinol (Providence) 12: 21526567211056239, 2021. doi: 10.1177/21526567211056239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. See I, Lale A, Marquez P, Streiff MB, Wheeler AP, Tepper NK, Woo EJ, Broder KR, Edwards KM, Gallego R, Geller AI, Jackson KA, Sharma S, Talaat KR, Walter EB, Akpan IJ, Ortel TL, Urrutia VC, Walker SC, Yui JC, Shimabukuro TT, Mba-Jonas A, Su JR, Shay DK. Case series of thrombosis with thrombocytopenia syndrome after COVID-19 vaccination-United States, December 2020 to August 2021. Ann Intern Med 175: 513–522, 2022. doi: 10.7326/M21-4502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. See I, Su JR, Lale A, Woo EJ, Guh AY, Shimabukuro TT, Streiff MB, Rao AK, Wheeler AP, Beavers SF, Durbin AP, Edwards K, Miller E, Harrington TA, Mba-Jonas A, Nair N, Nguyen DT, Talaat KR, Urrutia VC, Walker SC, Creech CB, Clark TA, DeStefano F, Broder KR. US case reports of cerebral venous sinus thrombosis with thrombocytopenia after Ad26.COV2.S vaccination, March 2 to April 21, 2021. JAMA 325: 2448–2456, 2021. doi: 10.1001/jama.2021.7517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Zhang Q, Bastard P, Effort CH, Cobat A, Casanova JL, COVID Human Genetic Effort. Human genetic and immunological determinants of critical COVID-19 pneumonia. Nature 603: 587–598, 2022. doi: 10.1038/s41586-022-04447-0. [DOI] [PMC free article] [PubMed] [Google Scholar]