Next Article in Journal
Incidence and Risk Factors for Development of Cardiac Toxicity in Adult Patients with Newly Diagnosed Acute Myeloid Leukemia
Next Article in Special Issue
Comparison of Computed Tomography Scoring Systems in Patients with COVID-19 and Hematological Malignancies
Previous Article in Journal
Patient Experience Factors and Implications for Improvement Based on the Treatment Journey of Patients with Head and Neck Cancer
Previous Article in Special Issue
Low Spike Antibody Levels and Impaired BA.4/5 Neutralization in Patients with Multiple Myeloma or Waldenstrom’s Macroglobulinemia after BNT162b2 Booster Vaccination
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

Adult Patients with Cancer Have Impaired Humoral Responses to Complete and Booster COVID-19 Vaccination, Especially Those with Hematologic Cancer on Active Treatment: A Systematic Review and Meta-Analysis

by
Efstathia Liatsou
1,
Ioannis Ntanasis-Stathopoulos
1,
Stavros Lykos
1,
Anastasios Ntanasis-Stathopoulos
1,
Maria Gavriatopoulou
1,
Theodora Psaltopoulou
1,
Theodoros N. Sergentanis
2,† and
Evangelos Terpos
1,*,†
1
Department of Clinical Therapeutics, National and Kapodistrian University of Athens, 11528 Athens, Greece
2
Department of Public Health Policy, School of Public Health, University of West Attica, 12243 Aigaleo, Greece
*
Author to whom correspondence should be addressed.
These authors contributed equally to this study.
Submission received: 14 February 2023 / Revised: 4 April 2023 / Accepted: 5 April 2023 / Published: 12 April 2023
(This article belongs to the Special Issue COVID-19 Infection and Hematological Malignancies)

Abstract

:

Simple Summary

Taking into consideration the high risk of patients with cancer for severe COVID-19 infection, prioritization has been given to primary prevention with both primary and booster vaccination. However, robust evidence for vaccination efficacy remains limited, due to the lack of available clinical trials including patients with active cancer. The rates of both humoral and cellular immune response remain rather vague, and they are mainly based on data deriving from retrospective studies of limited internal and external validity. We aimed to gather and analyze the current available literature on the efficacy of COVID-19 vaccination among patients with different types of malignancies receiving different treatments. Our results highlight that patients with cancer present suboptimal immune responses after COVID-19 vaccination, which is more prominent among patients with hematological malignancies.

Abstract

The exclusion of patients with cancer in clinical trials evaluating COVID-19 vaccine efficacy and safety, in combination with the high rate of severe infections, highlights the need for optimizing vaccination strategies. The aim of this study was to perform a systematic review and meta-analysis of the published available data from prospective and retrospective cohort studies that included patients with either solid or hematological malignancies according to the PRISMA Guidelines. A literature search was performed in the following databases: Medline (Pubmed), Scopus, Clinicaltrials.gov, EMBASE, CENTRAL and Google Scholar. Overall, 70 studies were included for the first and second vaccine dose and 60 studies for the third dose. The Effect Size (ES) of the seroconversion rate after the first dose was 0.41 (95%CI: 0.33–0.50) for hematological malignancies and 0.56 (95%CI: 0.47–0.64) for solid tumors. The seroconversion rates after the second dose were 0.62 (95%CI: 0.57–0.67) for hematological malignancies and 0.88 (95%CI: 0.82–0.93) for solid tumors. After the third dose, the ES for seroconversion was estimated at 0.63 (95%CI: 0.54–0.72) for hematological cancer and 0.88 (95%CI: 0.75–0.97) for solid tumors. A subgroup analysis was performed to evaluate potential factors affecting immune response. Production of anti-SARS-CoV-2 antibodies was found to be more affected in patients with hematological malignancies, which was attributed to the type of malignancy and treatment with monoclonal antibodies according to the subgroup analyses. Overall, this study highlights that patients with cancer present suboptimal humoral responses after COVID-19 vaccination. Several factors including timing of vaccination in relevance with active therapy, type of therapy, and type of cancer should be considered throughout the immunization process.

1. Introduction

The COVID-19 pandemic, declared on 1 March 2020, is responsible for 6,630,000 deaths worldwide [1,2]. The overall fatality rate has been reported to be 3.3%, with a particularly high disease-specific mortality risk for patients with cancer reaching 35–43% [1]. The relatively higher transmission rate and associated greater risk of mortality highlighted the urgent demand for efficient preventative vaccination [3]. Clinical trials were designed and held in less than a year, and the BNT162b2 COVID-19 vaccine was the first to receive emergency approval from the US Food and Drug Administration (FDA), followed by mRNA-1273, Ad26.COV2.S, AZD1222, and BBIP-CorV [4]. Based both on the findings of phase III clinical trials and real-world data, the efficacy of all these vaccines has been shown irrespectively of the severity of the disease [4].
Studies have increasingly reported on the outcomes of cancer patients, highlighting severe events such as intensive care unit admission, intubation, or death following COVID-19 infection [5]. Despite improvements in current therapies and medical care, rates have been found to be higher for patients with hematological malignancies, due to their systemic immunosuppressive state caused by the malignancy itself and the systematic therapy, pre-existing comorbidities, and frequent hospitalizations [6]. In addition to preventive contaminating measures such as personal hygiene and masks, vaccinations for such vulnerable populations were prioritized [5]. However, patients with malignancies were excluded from clinical vaccine trials, whereas most data derive solely from observational studies with a limited sample size [4]. On top of that, the impaired immune system raises concerns regarding the adequate production of SARS-CoV-2-specific antibodies post vaccination [7]. Booster vaccination has been shown to restore and sustain humoral response in healthy individuals [8,9,10]. It has become common practice to offer immunocompromised patients with cancer booster vaccinations to improve SARS-CoV-2 immunity to levels obtained in healthy individuals after the standard vaccination schedule, but pertinent data are scarce [11].
The aim of this systematic review and meta-analysis is to assess the rate of seropositivity in patients with hematological and solid cancers who have been vaccinated against COVID-19 and investigate any demographic or clinical factors that might affect immune response.

2. Methods

In this study, the updated Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) reporting guideline was applied (Supplementary Materials, PRISMA_2020_checklist) [12].This Systematic Review doesn’t need a registration number of the database.

2.1. Search Strategy

We searched the Pubmed (1966–2022), Scopus (2004–2022), Clinicaltrials.gov (2008–2022), EMBASE (1980–2022), Cochrane Central Register of Controlled Trials CENTRAL (1999–2022), and Google Scholar (2004–2022) databases in our primary search along with the reference lists of electronically retrieved full-text papers. The date of our last search was set at 30 September 2022. Our search strategy included the query terms, as follows: “TUMOR OR CANCER OR MALIGNANCY OR NEOPLASIA OR LEUKEMIA OR LYMPHOMA OR SARCOMA OR NEOPLASM OR MYELOMA) AND (COVID-19 OR SARS-COV-2) AND (VACCINE OR BNT162B2 OR AZD1222 OR MRNA1273) AND (ANTIBODIES OR IMMUNORESPONSE OR RESPONSE OR HUMORAL OR SEROCONVERSION OR SEROPOSITIVITY OR IMMUNOGENICITY)”, and is schematically presented in the PRISMA flow diagram (Figure 1).

2.2. Study Selection

The database searches were imported to the COVIDENCE Systematic Review and two investigators (EL and SL) reviewed the title and abstracts. The same two authors evaluated the selected articles following the inclusion and exclusion criteria. In the case of any disagreement, a third author (INS) was included.

2.3. Study Selection

The criteria for the inclusion of studies were predetermined. Prospective or retrospective observational studies as well as randomized clinical trials that included adult patients with a diagnosis of hematologic or solid cancer after one, two or three doses of COVID-19 vaccine were considered for inclusion. Studies should have reported patients’ antibody response at specific time intervals. Case reports/series or cohort studies with an overall population of less than five patients were excluded. Studies with insufficient data on humoral response or data only on cellular response were also excluded.

2.4. Data Extraction

Data extraction was performed using Microsoft Office Excel. Extraction included the following items: (1) studies’ characteristics such as the title, digital object identifier (doi), date of publication, first author’s name, and study design; (2) demographic characteristics of patients including age, sex, history of SARS-CoV-2 infection, type of cancer, active therapy during vaccination, number of doses of vaccine received, the type of vaccine administrated, the time interval between vaccination, and the time point of antibodies evaluation; (3) the type of antibody that was evaluated and the methods applied for identification; and (4) the treatment scheme administered at the time of vaccination, time interval between therapy and vaccination, and the number of participants who were seropositive following immunization based on each treatment. The extracted data were double-checked and validated by two authors (EL and SL). A third author (INS) participated in team consensus in case of discrepancies.

2.5. Outcome Measures

The primary outcome was the humoral response of patients post vaccination in the form of the antibodies’ seropositivity rate as calculated by each study. The secondary outcomes included the rate of seroconversion after immunization according to the disease subtypes, treatment categories, and vaccine type.

2.6. Quality and Risk of Bias Assessment

The risk of bias and methodological quality of the included studies was evaluated independently by two authors (EL and SL) using the Newcastle-Ottawa Scale (NOS) (Tables S1 and S2), which evaluates the selection of the study groups, the comparability of the groups and the ascertainment of the exposure or outcome of interest [13]. A third author (INS) made the final decision on scoring in case of disagreement.

2.7. Statistical Analysis

A meta-analysis was performed using the STATA (version 2016). Dichotomous variables were assessed using the risk ratio (RR), continuous variables were assessed using the mean difference, and survival was assessed using the hazard ratio (HR). Statistical heterogeneity was assessed using the Higgins I2 statistic. The 95% confidence intervals (CI) were reported for all results. When mean values and standard deviations were not reported in the studies, values were calculated according to the equations proposed by Hozo et al. Moreover, the analyses were sub-grouped based on the cumulative dose of vaccination. Taking into account that the Ad26.COV2.s is a single shot vaccine, the emerged data related to this vaccine were analyzed in the second dose immunization group. Results were graphically displayed on forest plots. A qualitative analysis and demonstration of results was presented when the meta-analysis of the data was not feasible.

3. Results

3.1. Study Characteristics

Figure 1 shows the selection of studies. Overall, as regards the first and second vaccine shots, 70 studies were included in the meta-analysis [6,7,11,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76]. In total, 10 studies [14,16,19,24,34,64,73,76,77,78] provided immune response information for patients with solid tumors, whereas 48 studies reported immune seroconversion results from patients with hematological malignancies [7,17,20,21,22,23,25,26,27,28,29,30,32,36,37,38,39,40,41,42,43,45,46,47,48,49,51,53,54,55,56,57,59,60,61,62,63,66,68,69,70,74,75,79,80,81,82], and 12studies [15,18,24,35,44,50,52,58,65,67,71,72] included patients with both solid and hematological malignancies. In total, 8 studies calculated patients’ antibodies after the first dose of vaccination only [16,19,26,27,31,41,51,62], whereas 35 studies evaluated immunogenicity after both doses [7,14,17,20,24,25,29,30,32,34,36,40,46,47,48,50,56,57,58,63,75,76,77,78,80,81] and 27 after the second dose only [15,18,21,22,23,25,28,35,37,38,39,40,42,43,44,45,46,47,52,53,54,55,59,61,63,64,65,66,67,68,69,70,71,72,73,74,80,82]. Studies were also stratified according to the type of vaccine that patients received. Of the 70 included studies, 30 studies reported vaccination results after vaccination with BN162b2 only [14,16,18,20,22,23,24,27,28,30,34,35,37,38,39,40,43,45,47,53,57,58,63,66,77,78,80,81,82,83], 20 studies reported results from vaccination with either BNT162b2 or mRNA-1273 vaccine [7,17,25,36,40,44,46,48,52,54,59,60,62,67,68,70,72,73,74,76], 11 studies reported results from vaccination with either BNT162b2 or AZD1222 vaccine [21,25,26,29,31,32,41,50,51,56,75], 6 studies included patients that received either BNT162b2 or AD26.COV2.S or mRNA-1273 [15,42,49,61,69,71], and 3 studies had available results from vaccination with BNT162b2 or AZD122 or mRNA-1273 [19,64,65]. One study reported results on mRNA-1273 exclusively [55].

3.2. First Vaccine Dose Immune Response

Figure 2 and Figure 3 present the seroconversion rates for seroconversion among immunocompromised patients, with either hematological malignancies or solid tumors compared with immunocompetent controls after a first dose of COVID-19 vaccine, respectively. Data were available from a total of 2443 patients with hematological malignancies, 2079 patients with solid tumors and 239 immunocompetent controls who were vaccinated with the first vaccine shot against SARS-COV-2.
The rate of immune seroconversion for patients with hematologic cancer was 0.41 [95% CI: 0.33–0.50] (Figure 2), and for patients with solid tumors it was 0.56 [95% CI: 0.47–0.64] (Figure 3). The immunocompetent controls showed an immune response rate of 90% (Effect Size (ES): 0.90 [95% CI: 0.82–0.96]) (Figure S1). Risk Ratios were lower for patients with hematological malignancies (RR: 0.48 [95% CI: 0.41–0.57]) (Figure S2) in comparison with patients with solid tumors (RR: 0.57 [95% CI: 0.49–0.67]) (Figure S3).

3.3. Second Vaccine Dose Immune Response

Figures S4 and S5 present the percentages and risk ratios for seroconversion among immunocompromised patients compared with healthy controls following the second dose of vaccination after studies were stratified based on the type of vaccine. In total, 51 studies investigated response after mRNA vaccines (either BNT162b2 or mRNA-1273). A total of 8276 patients with hematological malignancies, 2230 patients with solid cancer, and 2494 controls were analyzed regarding seroconversion after the second dose against COVID-19.
The overall percentage of immune seroconversion for patients with hematological malignancies was 0.62 [95% CI: 0.57–0.67] (Figure 4) and 0.88 [95% CI: 0.82–0.93] (Figure 5) for patients with solid tumors, respectively. The immunocompetent controls showed an immune response rate of 90% (ES: 0.90 [95% CI: 0.82–0.96]) (Figure S4). Risk Ratios were lower for patients with hematological malignancies (RR: 0.59 [95% CI: 0.53–0.63]) (Figure S5) in comparison with patients with solid tumors (RR: 0.85 [95% CI: 0.78–0.92]) (Figure S6).

3.4. Subgroup Analyses on Predictive Factors for Seroconversion after Initial Complete Vaccination

The analysis of the antibody response in patients with hematological malignancies and solid tumors was further applied based on potential predictive factors such as the type of disease, active or inactive treatment, type and time intervals of active treatment, and the type of vaccine that was administered. In terms of the subtype of hematological malignancy, data were available from 4 studies with 522 patients with multiple myeloma who received one vaccine dose (ES: 0.38 [95% CI: 0.20–0.59]) (Figure S7), and 17 studies encompassing data from 1814 patients with multiple myeloma who were fully vaccinated against SARS-COV-2 (ES: 0.80 [95% CI: 0.73–0.86]) (Figure S8). Furthermore, 1023 out of 1912 patients with chronic lymphocytic leukemia responded to full COVID-19 vaccination (ES: 0.51 [95% CI: 0.44–0.58]) (Figure S9). For Hodgkin lymphoma, 110 out of 115 patients showed immune seroconversion after the second dose of the COVID-19 vaccine, and the pooled response was ES: 0.99 [95% CI: 0.94–1.00]) (Figure S10). For non-Hodgkin lymphoma, immune response was evaluated in 10 studies with a total of 934 patients who completed vaccination with a seroconversion rate of 556/934 and a pooled ES of 60% (95% CI: 49%–71%) (Figure S11). Regarding immune response in patients with myelofibrosis, data were available in three studies; 22 out of 36 fully vaccinated patients showed adequate immune response and the pooled ES was 0.61 (95% CI: 0.44–0.78) (Figure S12). Finally, 321 out of 402 patients with myelodysplastic and myeloproliferative syndromes demonstrated an immune response after full COVID-19 vaccination, with a pooled ES of 80% [95%CI: 69%–90%] (Figure S13).
We also analyzed available data on seroconversion based on the treatment status of patients. A significant difference in antibody response was found for active treatment at the time of vaccination in comparison with no treatment. More specifically, 1392 out of 2395 patients under active treatment and 1000 out of 1381 patients without treatment showed an antibody response, and the pooled responses were 48% [95% CI: 36%–61%] and 76% [95% CI: 67%–83%] for active treatment and no treatment, respectively (Figures S14 and S15, respectively). The RR was calculated from 9 studies for active treatment and 11 studies for no treatment, and immune response in cancer patients was compared with antibodies measurement in healthy controls after full vaccination. The pooled RR was 0.49 [95% CI: 0.40–0.59] for active treatment and 0.79 [95% CI: 0.70–0.8] for no treatment (Figures S16 and S17, respectively).
Furthermore, different types of treatment were evaluated. The lowest antibody titers following full immunization were observed in patients on active therapy with anti-CD20 antibodies; only 43 out of 426 patients (ES: 11% [95% CI: 0.5%–20%]) achieved detectable antibody titers (Figure S18). The RR for anti-CD20 treatment was 0.16 [95% CI: 0.09–0.28] as pooled from four studies (Figure S19). A subgroup analysis of 412 patients revealed that the time interval between anti-CD20 therapy and vaccination influenced immune response (ES: 0.60 [95% CI: 0.47–0.72], RR: 0.78 [95% CI: 0.66–0.91]) (Figures S20 and S21, respectively).Beside anti-CD20 therapy, low seropositivity rates after full vaccination were evident for those who received Bruton’s kinase inhibitors as shown in 14 studies (n = 244/691, ES: 33% [95% CI: 20%–48%], RR: 0.22 [95% CI: 0.13–0.37], ref: healthy controls) (Figures S22 and S23, respectively) and CAR-T cell therapy (n = 43/104, ES: 38% [95% CI: 22–56%], RR: 0.44 [95% CI: 0.22–0.88]) (Figures S24 and S25, respectively). Conversely, the highest proportion of antibody response among the patients on treatment was estimated for patients who received chemotherapy (n = 601/806, ES: 72% [95% CI: 62%–81%]) (Figure S26) and endocrine therapy (n = 82/107, ES: 89% [95% CI: 37%–100%]) (Figure S27). Four studies included 104 patients who received combination treatment, out of whom 73 were seropositive (ES: 59% [95% CI: 32%–84%]) (Figure S28).
A total of 1402 patients were evaluated in nine studies involving hematopoietic stem cell transplantation. There was no difference between groups for allogeneic and autologous transplants. For allogeneic transplants, 627 out of 760 patients achieved an antibody response, and the pooled response was 82% [95% CI: 78%–87%] (Figure S29). Most patients underwent transplantation more than 1 year prior to vaccination. Limited data showed reduced response rates particularly for those receiving allogeneic transplantation less than 6 or 12 months prior to vaccination. For autologous transplants, 522 out of 642 patients achieved an antibody response, and the pooled response estimate was 78% [95% CI: 67%–88%] (Figures S30 and S31).
The immune response was also evaluated in a subgroup analysis based on the type of vaccine that was received. More specifically, most patients received the BNT162B2 vaccine; 1068 patients with hematological malignancies received only one dose of the vaccine, and 5885 patients got fully vaccinated. Among them, 522 and 3647 produced adequate antibody titers with an ES of 46% [95% CI: 35%–57%] and 63% [95% CI: 56%–69%], after one and two vaccine shots, respectively (Figures S32 and S33, respectively). Immunogenicity after BNT162B2 vaccination was also investigated in patients with solid tumors, with 619 out of 1058 and 916 out of 976 developing adequate immune responses after the first and second doses, respectively (ES: 56% [95% CI: 48%–63%] and 94% [95% CI: 93%–96%]) (Figures S34 and S35, respectively). Seroconversion rates were also calculated in 11 studies including 1596 patients with hematological malignancies who were vaccinated with mRNA-1273. The ES was found to be 0.72 [95% CI: 0.57–0.85] (Figure S36). Lastly, three studies reported on full vaccination with the AZD1222 of 89 patients with hematological malignancies, 47 of whom produced a sufficient amount of antibodies with a pooled ES of 47% [95% CI: 24%–71%] (Figure S37).

3.5. Booster COVID-19 Vaccination

In total, 60 studies were included for data analysis on booster COVID-19 vaccination, 43 studies reported on the immune response of a total of 4754 patients with a diagnosis of hematological malignancy, and 22 studies determined immune seroconversion in 2440 patients with solid tumors [78,79,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133]. The median age of cancer patients was 64 years (range: 43–76). Studies were grouped according to the type of vaccine that was administered as a booster dose. The mean time of evaluation following booster vaccination was 28 days (range: 7–95).According to the studies that had available information on the specific type of hematological malignancy or solid tumor, 109 patients had ALL, 100 AML, 70 CML, 1003 CLL, 755 lymphomas, 64 MPN/MDS, 11 PV, 32 ET, 792 MM, 121 WM, 407 breast cancer, 281 urological cancer, 95 gynecological cancer, 92 melanoma, 347 lung cancer, 327 GI cancer, 19 brain cancer, 65 head and neck cancer, and 9 connective tissue cancer. Data regarding treatment status was available in 41 studies. More specifically, 3991 patients were in active treatment, and 1278 were off treatment.
SARS-COV-2 booster vaccination showed reduced seroconversion rates in patients with hematological malignancies as compared to healthy controls (ES: 0.63 [95% CI: 0.54–0.72] versus ES: 0.98 [95% CI: 0.89–1.00]) (Figure 6 and Figure 7). The pooled seropositivity RR was also lower for patients with hematological malignancies (RR: 0.67 [95% CI: 0.59–0.77]) compared to the pooled rates for patients with solid tumors (RR: 0.84 [95% CI: 0.75–0.95]) (Figure 8 and Figure 9).

3.6. Subgroup Analyses on Predictive Factors for Seroconversion after Booster Vaccination

We then divided patients into subgroups based on the treatment they received. Patients undergoing active therapy with anti-CD20 Abs showed the lowest seroconversion rate following booster immunization; only 26% [95% CI: 16–37%] of 442 patients achieved detectable antibody titers (Figure S38). Beside anti-CD20 therapy, the lowest seropositivity rate after booster vaccination was evident for those who received combination therapy of two or more therapeutic agents (n = 404, ES 45% [95% CI: 25%–65%]) (Figure S39) and CAR-T cell therapy (n = 133, ES 32% [95% CI: 22–43%], I2 = 85.1%) (Figure S40). In contrast, the highest proportion of antibody response was estimated for patients who had undergone allogenous SCT (n = 164, ES 72% [95% CI: 47–92%]) (Figure S41) and steroids (n = 252, ES 71% [95% CI: 53–87%]) (Figure S42). Studies reporting on patients who received venetoclax showed mixed results (n = 85, ES 50% [95% CI: 25–74%]) (Figure S43). Following booster immunization, the low rates of achieving an antibody response were also observed for patients with hematological malignancies who were on Bruton’s tyrosine kinase (BTK) inhibitors (n = 404, ES 45% [95% CI: 25–65%]) (Figure S44), while high rates of immune response were observed with autologous SCT (n = 126, ES 65% [95% CI: 32–93%]) (Figure S45). High rates of heterogeneity should be noted in the subgroup analyses on treatment subtypes.
When comparing antibody responses in patients with different types of hematological malignancies and solid tumors, data were available only for subtypes of hematological cancer. The lowest seropositivity rate was evident in patients with non-Hodgkin lymphoma (n = 557, ES 48% [95% CI: 35–60%]) (Figure S46), followed by those with CLL (n = 1003, ES 65% [95% CI: 49–79%]) (Figure S47) and MM (n= 792, ES: 86% [95% CI: 77–94%]) (Figure S48). Regarding immune response rates by vaccine type, data wereavailable for pooled analysis only from studies with the BNT162B2 booster vaccination. The Effect Size for antibody response was ES: 0.69 [95% CI: 0.43–0.77] (Figure S49) for patients with hematological malignancies with RR: 0.67 [95% CI: 0.59–0.77] (Figure S50), whereas for patients with solid tumors, the pooled Effect Size was found to be 0.90 [95% CI: 0.77–0.98] (Figure S51).

4. Discussion

This study showed that oncology patients had a significantly reduced antibody response compared with healthy individuals following the first, second, and booster COVID-19 vaccination. More specifically, seroconversion was less likely in patients with blood cancer compared with healthy individuals by 52%, 41%, and 33% after the first, second, and booster vaccine shot, respectively. Seroconversion was also less likely in patients with solid cancer compared with healthy controls by 43%, 15%, and 16% after each vaccine dose, respectively. It has to be noted that the anti-SARS-CoV-2 humoral response was more attenuated in patients with hematological cancer compared to patients with solid tumors. Following complete vaccination, immune response was mostly affected in patients with CLL (46% lower seroconversion compared to controls) or NHL (38% lower seroconversion), in patients on active treatment (51% lower seroconversion), and in those receiving drugs targeting CD20 (84% lower seroconversion) or BTKIs (78% lower seroconversion) or CAR-T cell therapy (60% lower seroconversion). Among patients with cancer, those with solid tumors or MPN had the highest seroconversion rates after the first vaccine dose, whereas those with HL had the highest seroconversion rates after the second dose. Similar results were derived from the pooled analysis of the booster vaccine shot; hematological patients had 33% and patients with solid cancer had 16% lower seroconversion compared to healthy controls.
According to the pooled response rate analysis, the type of vaccine administered has an effect on antibody production with the mRNA-1273 vaccine being the most effective followed by the BNT162B2 and the AZD122.This is also supported by Noori et al., where lower seropositivity rates were observed with the BNT162B2 vaccination compared to mRNA-1273 (RR: 0.89, 95% CI: 0.79–0.99) [134,135,136].
These findings can be explained by both the underlying diseases and the therapeutic approaches that impair the immune response. This immune deregulation has also become evident in studies evaluating the clinical manifestations and outcomes of COVID-19-infected cancer patients [137]. First, patients with cancer, especially those with hematological malignancies, experience long periods of neutropenia not only due to the anticancer therapy they receive, such as anti-CD20 antibodies and HSCT, but also as a result of the malignancy’s natural course itself through immunological mechanisms or direct bone marrow infiltration [138]. The humoral adaptive immune response is affected on multiple levels [138,139]. Lymphopenia is commonly found, and in that case, B cells are highly depleted, principally in cases of CLL and MM [139]. Treatments targeting CD20, CD38, or B-cell maturation antigen (BCMA) have also been associated with depleted circulating B-cells and significantly impaired IgM and IgG responses against both the ancestral Wuhan strain and the Omicron SARS-CoV-2 variants [139,140,141]. The defect in humoral response may remain evident even after a second booster vaccine dose, as it has been shown in patients with MM on anti-BCMA treatment [8].
The cellular component of the immune responses also affected, as has been shown in studies that profiled the cytometric activity of patients with cancer and COVID-19 infection [142]. More specifically, a statistically significant difference was observed in CD4+ T-cells being less frequent in patients with hematologic cancer compared to those who suffer from solid malignancies, whereas CD8+ T-cells were equally detected among those groups [142]. Similar variations have been confirmed and are consistent with findings from other studies on vaccines against viruses (influenza, hepatitis B) and bacteria (Streptococcus pneumoniae) among patients with hematological malignancies and solid organ transplant recipients [143,144]. However, only a few studies provided detailed data regarding cellular response, and high levels of heterogeneity did not permit a pooled analysis [144].
The vaccination-induced response against SARS-CoV-2 in patients with cancer has been examined in other systematic reviews and meta-analyses as well. Gagelmann et al. presented results from a total of 49 studies including patients with hematological malignancies. The authors showed an impaired antibody production, which was mainly associated with the type of malignancy, with the lowest immune response being noted in cases of chronic lymphocytic leukemia, and the administration of active treatment at the time of vaccination [145]. The large sample of patients constitutes one of the main strengths of that study; however, patients with solid tumors were not included in the study protocol, a fact that could otherwise further enlighten potential tendencies and differences in a more direct way [145]. Another systematic review by Noori et al. investigated the effect of the two-dose vaccination scheme in antibody production, and the results are consistent with our findings in terms of the defect in the humoral response following COVID-19 vaccination, especially in patients with hematological cancer. Nevertheless, our study is still the most updated regarding the systematic analysis of both the two-dose complete vaccination scheme and booster vaccination in all patients with cancer.
Our study has some limitations to be acknowledged. First, considerable heterogeneity was observed in some subgroups that were analyzed due to the misrepresentation of these subpopulations. More specifically, studies with AML, CML, WM, MF, PV, IT, and CML had limited numeric data regarding serological response; thus, no pooled data were available. The effect of other potential confounding factors, including prior COVID-19 infection and time from last infection to last vaccination, age, body mass index, and autoimmune diseases, on our results cannot be excluded [146,147,148]. However, due to a lack of reported pertinent data, we were unable to include such data in our analyses. Under this framework, subgroup analysis included a pooled analysis of the humoral response in patients with solid tumors who received chemotherapy. There is an undoubtedly wide range of different chemotherapeutic agents in various administration forms and schemes with different bioavailability and toxicity profiles and a heterogenous degree of immune suppression. However, data on each patient’s therapeutic regimen scheme was unavailable, making further analysis unfeasible. Regarding patients on ruxolitinib or immune checkpoint inhibitors or cellular therapies, as well as those receiving radiotherapy, pooled analyses were not performed due to the small number of arms within each subgroup. Patients who underwent an allogeneic stem cell transplant constitute a specific clinical group selected based on predefined clinical criteria. Current approaches to prevent and treat GVHD post transplantation include a constellation of immunosuppressive medications. Therefore, this is a highly heterogenous group of patients and the findings of immune response to vaccination should be interpreted cautiously. Furthermore, different units of antibody titers and time intervals were assessed in the included studies, which resulted in significant heterogeneity in the pooled results. Additionally, the majority of studies evaluated the efficacy of either BNT162b2 or mRNA-1273; thus, the generalizability of the pooled results on non-mRNA vaccines may be negotiable. The antibody’s titers as a surrogate endpoint could reflect the efficacy of the immune system against COVID-19 [149,150]. However, different diagnostic assays were used by each study to evaluate the immune response post vaccination [151]. The anti-spike IgG antibody was the most commonly tested in studies; however, their neutralizing activity, and thus efficacy against infection, was not consistently reported. The absence of a standard-of-care assay to determine the humoral immune response post COVID-19 vaccination may limit the clinical utility of determining antibody response in the clinical practice [151]. Regarding the efficacy assessment post vaccination, the antibody cut-off to define seropositivity as well as the measurement units are not unanimous among studies.This high level of heterogeneity made a potential subgroup analysis unfeasible as multiple subgroups with low patient numbers were derived. In addition to the above, given that COVID-19 is a viral infection, the role of CD8+ T cells has been examined and found to be protective, and it may be even more important in patients with hematological malignancies who have impaired humoral responses [152,153].

5. Conclusions

In conclusion, our results show that patients with cancer have impaired humoral responses to complete and booster COVID-19 vaccination. This is more pronounced in patients with hematologic cancer on active treatment at the time of vaccination. The high level of heterogeneity in the methods and reported outcomes among the studies necessitates the careful evaluation of subgroup analyses. Patients with cancer should be prioritized for receiving booster and updated vaccine shots, pre- and post-exposure prophylaxis with antiviral drugs, and monoclonal antibodies in order to prevent severe COVID-19 outcomes.

Supplementary Materials

The following supporting information can be downloaded at: https://0-www-mdpi-com.brum.beds.ac.uk/article/10.3390/cancers15082266/s1. Table S1. Newcastle-Ottawa Assessment Scale for cohort studies; Table S2. Newcastle-Ottawa Assessment Scale for case-control studies; Figure S1. Pooled Effect Size for immune seroconversion rates of immunocompetent controls after first vaccine dose COVID-19; Figure S2. Relative Risk for immune seroconversion of patients with hematological malignancies after first vaccine dose; Figure S3. Relative Risk for immune seroconversion of patients with solid tumors after first vaccine dose; Figure S4. Pooled Effect Size for immune seroconversion rate of immunocompetent controls after second vaccine dose; Figure S5. Relative Risk for immune response of patients with hematological malignancies after second vaccine dose; Figure S6. Relative Risk for immune response of patients with solid tumors after second vaccine dose; Figure S7. Effect Size for immune seroconversion rates in patients with multiple myeloma after first vaccine dose; Figure S8. Εffect size for immune seroconversion rates of patients with multiple myeloma after second vaccine dose; Figure S9. Effect size for immune seroconversion rates of patients with chronic lymphocytic leukemia after second vaccine dose; Figure S10. Effect size of immune seroconversion rates of patients with Hodgkin lymphoma after second vaccine dose; Figure S11. Effect size of immune seroconversion rates of patients with non-Hodgkin myeloma after second vaccine dose; Figure S12. Effect size of immune seroconversion rates of patients with myelofibrosis after second vaccine dose; Figure S13. Effect Size for immune seroconversion rates of patients with myelodysplastic and myeloproliferative syndromes after second vaccine dose; Figure S14. Effect Size for immune seroconversion rates of patients under active treatment after second vaccine dose; Figure S15. Effect Size for immune seroconversion rates of treatment naïve patients after second dose; Figure S16. Relative Risk for immune seroconversion rates of patients under active treatment after second vaccine dose; Figure S17. Relative Risk for immune seroconversion rates of treatment naïve patients after second vaccine dose; Figure S18. Effect Size for immune seroconversion rates of patients under active anti-CD20 monoclonal antibodies after second vaccine dose; Figure S19. Relative Risk of immune seroconversion rates of patients under active anti-CD20 monoclonal antibodies after second vaccine dose; Figure S20. Effect Size for immune seroconversion rates of patients having received treatment with anti-CD20 longer than one year ago; Figure S21. Relative Risk for immune seroconversion rates of patients having received treatment with anti-CD20 longer than one year ago; Figure S22. Effect Size of immune seroconversion rates of patients under active Bruton’s Tyrosine Kinase inhibitors treatment after second dose; Figure S23. Relative Risk of immune seroconversion rates of patients under active Bruton’s Tyrosine kinase inhibitors treatment after second dose; Figure S24. Effect Size for immune seroconversion rates of patients under CAR-T cells therapy after second vaccine dose; Figure S25. Relative Risk of immune seroconversion rate of patients under CAR-T cells therapy after second vaccine dose; Figure S26. Effect Size for immune seroconversion rates of patients under chemotherapy after second vaccine dose; Figure S27. Effect Size for immune seroconversion rates of patients under endocrine therapy after second vaccine dose; Figure S28. Effect Size for immune seroconversion rates of patients under combination treatment after second vaccine dose; Figure S29. Effect size for immune seroconversion rates of patients who underwent allogeneic stem cell transplantation after second vaccine dose; Figure S30. Effect Size for immune seroconversion rates of patients who underwent autologous stem cell transplantation after second vaccine dose; Figure S31. Relative Risk for immune seroconversion rates of patients who underwent allogeneic stem cell transplantation after second vaccine dose; Figure S32. Effect Size for immune seroconversion rates in patients with hematological malignancies who received first dose of BNT162B2 vaccine; Figure S33. Effect Size of immune seroconversion rates in patients with hematological malignancies who received second dose of BNT162B2; Figure S34. Effect Size for immune seroconversion rates of patients with solid tumors who received first dose of BNT162B2 vaccine; Figure S35. Effect Size for immune seroconversion rates of patients with solid tumors who received second dose of BNT162B2 vaccine; Figure S36. Effect Size for immune seroconversion rates of patients with hematological malignancies after second mRNA-1273; Figure S37. Effect Size for immune seroconversion rates of patients with hematological malignancies after second AZD1222; Figure S38. Effect Size for immune seroconversion rates of patients on active therapy with anti-CD20 monoclonal antibodies after third vaccine dose; Figure S39. Effect Size for immune seroconversion rates of patients on active therapy with combination therapy after third vaccine dose; Figure S40. Effect Size for immune seroconversion rates of patients under active CAR-T cell therapy after third vaccine dose; Figure S41. Effect Size for immune seroconversion rates of patients who underwent allogenic stem cell transplant after third vaccine dose; Figure S42. Effect Size for immune seroconversion rates of patients on treatment with steroids after third vaccine dose; Figure S43. Effect Size for immune seroconversion rates of patients under venetoclax therapy after third vaccine dose; Figure S44. Effect Size for immune seroconversion rates of patients on Bruton’s Tyrosine Kinase inhibitors after third vaccine dose; Figure S45. Effect size for immune seroconversion rates of patients who underwent autologous stem cell transplantation after third vaccine dose; Figure S46. Effect Size for immune seroconversion rates of patients with non-Hodgkin lymphoma after third vaccine dose; Figure S47. Effect size of immune seroconversion rates of patients with chronic lymphocytic leukemia after third vaccine dose; Figure S48. Effect Size of immune seroconversion rates of patients with multiple myeloma after third vaccine dose; Figure S49. Effect Size for immune seroconversion rates of patients with hematological malignancies who received BNT162B2 booster dose; Figure S50. Relative Risk of immune seroconversion rates of patients with hematological malignancies who received a BNT162B2 booster vaccine dose; Figure S51. Effect Size of immune seroconversion rates of patients with solid tumors who received a BNT162B2 booster vaccine dose. PRISMA_2020_checklist.

Author Contributions

Conceptualization, I.N.-S. and E.T.; methodology, T.N.S. and I.N.-S.; software, E.L., S.L. and T.N.S.; validation, T.N.S., A.N.-S., M.G. and T.P.; formal analysis, E.L. and T.N.S.; investigation, E.L., I.N.-S., S.L. and A.N.-S.; data curation, M.G., T.P. and T.N.S.; writing—original draft preparation, E.L. and I.N.-S.; writing—review and editing, S.L., A.N.-S., M.G., T.P., T.N.S. and E.T.; visualization, T.N.S.; supervision, T.N.S. and E.T. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data available upon reasonable request from the corresponding author.

Conflicts of Interest

The authors declare no relevant conflict of interest.

Abbreviations

ALLAcute Lymphocytic Leukemia
AMLAcute Myelocytic Leukemia
CMLChronic Myelocytic Leukemia
CLLChronic Lymphocytic Leukemia
MPN/MDSmyeloproliferative neoplasms/myelodysplastic syndrome
PVPolycythemia Vera
ETEssential Thrombocytosis
MMMultiple Myeloma
WMWaldenström’s Macroglobulinemia
GynGynecological
GIgastrointestinal
ESEffect Size
RRRelative Risk
SCTStem Cell Transplantation

References

  1. Chavez-MacGregor, M.; Lei, X.; Zhao, H.; Scheet, P.; Giordano, S.H. Evaluation of COVID-19 Mortality and Adverse Outcomes in US Patients With or Without Cancer. JAMA Oncol. 2022, 8, 69–78. [Google Scholar] [CrossRef] [PubMed]
  2. Rahman, S.; Montero, M.T.V.; Rowe, K.; Kirton, R.; Kunik, F., Jr. Epidemiology, pathogenesis, clinical presentations, diagnosis and treatment of COVID-19: A review of current evidence. Expert Rev. Clin. Pharm. 2021, 14, 601–621. [Google Scholar] [CrossRef]
  3. Mandal, A.; Singh, P.; Samaddar, A.; Singh, D.; Verma, M.; Rakesh, A.; Ranjan, R. Vaccination of cancer patients against COVID-19: Towards the end of a dilemma. Med. Oncol. 2021, 38, 92. [Google Scholar] [CrossRef]
  4. Desai, A.; Gainor, J.F.; Hegde, A.; Schram, A.M.; Curigliano, G.; Pal, S.; Liu, S.V.; Halmos, B.; Groisberg, R.; Grande, E.; et al. COVID-19 vaccine guidance for patients with cancer participating in oncology clinical trials. Nat. Rev. Clin. Oncol. 2021, 18, 313–319. [Google Scholar] [CrossRef]
  5. Aapro, M.; Lyman, G.H.; Bokemeyer, C.; Rapoport, B.L.; Mathieson, N.; Koptelova, N.; Cornes, P.; Anderson, R.; Gascón, P.; Kuderer, N.M. Supportive care in patients with cancer during the COVID-19 pandemic. ESMO Open 2021, 6, 100038. [Google Scholar] [CrossRef] [PubMed]
  6. Liu, C.; Zhao, Y.; Okwan-Duodu, D.; Basho, R.; Cui, X. COVID-19 in cancer patients: Risk, clinical features, and management. Cancer Biol. Med. 2020, 17, 519–527. [Google Scholar] [CrossRef] [PubMed]
  7. Addeo, A.; Shah, P.K.; Bordry, N.; Hudson, R.D.; Albracht, B.; Di Marco, M.; Kaklamani, V.; Dietrich, P.-Y.; Taylor, B.S.; Simand, P.-F. Immunogenicity of SARS-CoV-2 messenger RNA vaccines in patients with cancer. Cancer Cell 2021, 39, 1091–1098.e1092. [Google Scholar] [CrossRef]
  8. Ntanasis-Stathopoulos, I.; Karalis, V.; Sklirou, A.D.; Gavriatopoulou, M.; Alexopoulos, H.; Malandrakis, P.; Trougakos, I.P.; Dimopoulos, M.A.; Terpos, E. Third Dose of the BNT162b2 Vaccine Results in Sustained High Levels of Neutralizing Antibodies Against SARS-CoV-2 at 6 Months Following Vaccination in Healthy Individuals. Hemasphere 2022, 6, e747. [Google Scholar] [CrossRef] [PubMed]
  9. Terpos, E.; Karalis, V.; Sklirou, A.D.; Apostolakou, F.; Ntanasis-Stathopoulos, I.; Bagratuni, T.; Iconomidou, V.A.; Malandrakis, P.; Korompoki, E.; Papassotiriou, I.; et al. Third dose of the BNT162b2 vaccine results in very high levels of neutralizing antibodies against SARS-CoV-2: Results of a prospective study in 150 health professionals in Greece. Am. J. Hematol. 2022, 97, e147–e150. [Google Scholar] [CrossRef]
  10. Terpos, E.; Karalis, V.; Ntanasis-Stathopoulos, I.; Apostolakou, F.; Gumeni, S.; Gavriatopoulou, M.; Papadopoulos, D.; Malandrakis, P.; Papanagnou, E.D.; Korompoki, E.; et al. Sustained but Declining Humoral Immunity Against SARS-CoV-2 at 9 Months Postvaccination With BNT162b2: A Prospective Evaluation in 309 Healthy Individuals. Hemasphere 2022, 6, e677. [Google Scholar] [CrossRef]
  11. Fendler, A.; de Vries, E.G.E.; GeurtsvanKessel, C.H.; Haanen, J.B.; Wörmann, B.; Turajlic, S.; von Lilienfeld-Toal, M. COVID-19 vaccines in patients with cancer: Immunogenicity, efficacy and safety. Nat. Rev. Clin. Oncol. 2022, 19, 385–401. [Google Scholar] [CrossRef]
  12. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. BMJ 2021, 372, n71. [Google Scholar] [CrossRef] [PubMed]
  13. Wells, G.A.; Wells, G.; Shea, B.; Shea, B.; O’Connell, D.; Peterson, J.; Welch, V.; Losos, M.; Tugwell, P.; Ga, S.W.; et al. The Newcastle-Ottawa Scale (NOS) for Assessing the Quality of Nonrandomised Studies in Meta-Analyses. Available online: https://www.ohri.ca/programs/clinical_epidemiology/nosgen.pdf (accessed on 10 February 2023).
  14. Massarweh, A.; Eliakim-Raz, N.; Stemmer, A.; Levy-Barda, A.; Yust-Katz, S.; Zer, A.; Benouaich-Amiel, A.; Ben-Zvi, H.; Moskovits, N.; Brenner, B. Evaluation of seropositivity following BNT162b2 messenger RNA vaccination for SARS-CoV-2 in patients undergoing treatment for cancer. JAMA Oncol. 2021, 7, 1133–1140. [Google Scholar] [CrossRef]
  15. Thakkar, A.; Gonzalez-Lugo, J.D.; Goradia, N.; Gali, R.; Shapiro, L.C.; Pradhan, K.; Rahman, S.; Kim, S.Y.; Ko, B.; Sica, R.A. Seroconversion rates following COVID-19 vaccination among patients with cancer. Cancer Cell 2021, 39, 1081–1090.e1082. [Google Scholar] [CrossRef]
  16. Palich, R.; Veyri, M.; Marot, S.; Vozy, A.; Gligorov, J.; Maingon, P.; Marcelin, A.-G.; Spano, J.-P. Weak immunogenicity after a single dose of SARS-CoV-2 mRNA vaccine in treated cancer patients. Ann. Oncol. 2021, 32, 1051–1053. [Google Scholar] [CrossRef]
  17. Diefenbach, C.; Caro, J.; Koide, A.; Grossbard, M.; Goldberg, J.D.; Raphael, B.; Hymes, K.; Moskovits, T.; Kreditor, M.; Kaminetzky, D. Impaired humoral immunity to SARS-CoV-2 vaccination in non-Hodgkin lymphoma and CLL patients. MedRxiv 2021. [Google Scholar]
  18. Iacono, D.; Cerbone, L.; Palombi, L.; Cavalieri, E.; Sperduti, I.; Cocchiara, R.A.; Mariani, B.; Parisi, G.; Garufi, C. Serological response to COVID-19 vaccination in patients with cancer older than 80 years. J. Geriatr. Oncol. 2021, 12, 1253–1255. [Google Scholar] [CrossRef]
  19. Terpos, E.; Zagouri, F.; Liontos, M.; Sklirou, A.D.; Koutsoukos, K.; Markellos, C.; Briasoulis, A.; Papanagnou, E.-D.; Trougakos, I.P.; Dimopoulos, M.-A. Low titers of SARS-CoV-2 neutralizing antibodies after first vaccination dose in cancer patients receiving checkpoint inhibitors. J. Hematol. Oncol. 2021, 14, 1–4. [Google Scholar] [CrossRef]
  20. Pimpinelli, F.; Marchesi, F.; Piaggio, G.; Giannarelli, D.; Papa, E.; Falcucci, P.; Pontone, M.; Di Martino, S.; Laquintana, V.; La Malfa, A. Fifth-week immunogenicity and safety of anti-SARS-CoV-2 BNT162b2 vaccine in patients with multiple myeloma and myeloproliferative malignancies on active treatment: Preliminary data from a single institution. J. Hematol. Oncol. 2021, 14, 1–12. [Google Scholar] [CrossRef] [PubMed]
  21. Bird, S.; Panopoulou, A.; Shea, R.L.; Tsui, M.; Saso, R.; Sud, A.; West, S.; Smith, K.; Barwood, J.; Kaczmarek, E. Response to first vaccination against SARS-CoV-2 in patients with multiple myeloma. Lancet Haematol. 2021, 8, e389–e392. [Google Scholar] [CrossRef] [PubMed]
  22. Herzog Tzarfati, K.; Gutwein, O.; Apel, A.; Rahimi-Levene, N.; Sadovnik, M.; Harel, L.; Benveniste-Levkovitz, P.; Bar Chaim, A.; Koren-Michowitz, M. BNT162b2 COVID-19 vaccine is significantly less effective in patients with hematologic malignancies. Am. J. Hematol. 2021, 96, 1195–1203. [Google Scholar] [CrossRef] [PubMed]
  23. Maneikis, K.; Šablauskas, K.; Ringelevičiūtė, U.; Vaitekėnaitė, V.; Čekauskienė, R.; Kryžauskaitė, L.; Naumovas, D.; Banys, V.; Pečeliūnas, V.; Beinortas, T. Immunogenicity of the BNT162b2 COVID-19 mRNA vaccine and early clinical outcomes in patients with haematological malignancies in Lithuania: A national prospective cohort study. Lancet Haematol. 2021, 8, e583–e592. [Google Scholar] [CrossRef]
  24. Monin, L.; Laing, A.G.; Muñoz-Ruiz, M.; McKenzie, D.R.; Del Barrio, I.D.M.; Alaguthurai, T.; Domingo-Vila, C.; Hayday, T.S.; Graham, C.; Seow, J. Safety and immunogenicity of one versus two doses of the COVID-19 vaccine BNT162b2 for patients with cancer: Interim analysis of a prospective observational study. Lancet Oncol. 2021, 22, 765–778. [Google Scholar] [CrossRef]
  25. Lim, S.H.; Campbell, N.; Johnson, M.; Joseph-Pietras, D.; Collins, G.P.; O’Callaghan, A.; Fox, C.P.; Ahearne, M.; Johnson, P.W.; Goldblatt, D. Antibody responses after SARS-CoV-2 vaccination in patients with lymphoma. Lancet Haematol. 2021, 8, e542–e544. [Google Scholar] [CrossRef]
  26. Chowdhury, O.; Bruguier, H.; Mallett, G.; Sousos, N.; Crozier, K.; Allman, C.; Eyre, D.; Lumley, S.; Strickland, M.; Karali, C.S.; et al. Impaired antibody response to COVID-19 vaccination in patients with chronic myeloid neoplasms. Br. J. Haematol 2021, 194, 1010–1015. [Google Scholar] [CrossRef] [PubMed]
  27. Harrington, P.; de Lavallade, H.; Doores, K.J.; O’Reilly, A.; Seow, J.; Graham, C.; Lechmere, T.; Radia, D.; Dillon, R.; Shanmugharaj, Y. 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, 3573–3577. [Google Scholar] [CrossRef] [PubMed]
  28. Herishanu, Y.; Avivi, I.; Aharon, A.; Shefer, G.; Levi, S.; Bronstein, Y.; Morales, M.; Ziv, T.; Shorer Arbel, Y.; Scarfò, L. Efficacy of the BNT162b2 mRNA COVID-19 vaccine in patients with chronic lymphocytic leukemia. Blood 2021, 137, 3165–3173. [Google Scholar] [CrossRef] [PubMed]
  29. Gavriatopoulou, M.; Terpos, E.; Ntanasis-Stathopoulos, I.; Briasoulis, A.; Gumeni, S.; Malandrakis, P.; Fotiou, D.; Migkou, M.; Theodorakakou, F.; Eleutherakis-Papaiakovou, E. Poor neutralizing antibody responses in 106 patients with WM after vaccination against SARS-CoV-2: A prospective study. Blood Adv. 2021, 5, 4398–4405. [Google Scholar] [CrossRef] [PubMed]
  30. Terpos, E.; Gavriatopoulou, M.; Fotiou, D.; Giatra, C.; Asimakopoulos, I.; Dimou, M.; Sklirou, A.D.; Ntanasis-Stathopoulos, I.; Darmani, I.; Briasoulis, A. Poor neutralizing antibody responses in 132 patients with CLL, NHL and HL after vaccination against SARS-CoV-2: A prospective study. Cancers 2021, 13, 4480. [Google Scholar] [CrossRef]
  31. Zagouri, F.; Terpos, E.; Fiste, O.; Liontos, M.; Briasoulis, A.; Katsiana, I.; Skafida, E.; Markellos, C.; Kunadis, E.; Andrikopoulou, A. SARS-CoV-2 neutralizing antibodies after first vaccination dose in breast cancer patients receiving CDK4/6 inhibitors. Breast 2021, 60, 58–61. [Google Scholar] [CrossRef]
  32. Terpos, E.; Gavriatopoulou, M.; Ntanasis-Stathopoulos, I.; Briasoulis, A.; Gumeni, S.; Malandrakis, P.; Fotiou, D.; Papanagnou, E.-D.; Migkou, M.; Theodorakakou, F. The neutralizing antibody response post COVID-19 vaccination in patients with myeloma is highly dependent on the type of anti-myeloma treatment. Blood Cancer J. 2021, 11, 1–9. [Google Scholar] [CrossRef]
  33. Gounant, V.; Ferré, V.M.; Soussi, G.; Charpentier, C.; Flament, H.; Fidouh, N.; Collin, G.; Namour, C.; Assoun, S.; Bizot, A. Efficacy of severe acute respiratory syndrome coronavirus-2 vaccine in patients with thoracic cancer: A prospective study supporting a third dose in patients with minimal serologic response after two vaccine doses. J. Thorac. Oncol. 2022, 17, 239–251. [Google Scholar] [CrossRef] [PubMed]
  34. Lasagna, A.; Agustoni, F.; Percivalle, E.; Borgetto, S.; Paulet, A.; Comolli, G.; Sarasini, A.; Bergami, F.; Sammartino, J.; Ferrari, A. A snapshot of the immunogenicity, efficacy and safety of a full course of BNT162b2 anti-SARS-CoV-2 vaccine in cancer patients treated with PD-1/PD-L1 inhibitors: A longitudinal cohort study. ESMO Open 2021, 6, 100272. [Google Scholar] [CrossRef] [PubMed]
  35. Peeters, M.; Verbruggen, L.; Teuwen, L.; Vanhoutte, G.; Kerckhove, S.V.; Peeters, B.; Raats, S.; Van der Massen, I.; De Keersmaecker, S.; Debie, Y. Reduced humoral immune response after BNT162B2 COVID-19 MRNA vaccination in cancer patients under anti-neoplastic treatment. ESMO Open 2021, 100274. [Google Scholar] [CrossRef] [PubMed]
  36. Stampfer, S.D.; Goldwater, M.-S.; Jew, S.; Bujarski, S.; Regidor, B.; Daniely, D.; Chen, H.; Xu, N.; Li, M.; Green, T. Response to mRNA vaccination for COVID-19 among patients with multiple myeloma. Leukemia 2021, 35, 3534–3541. [Google Scholar] [CrossRef] [PubMed]
  37. Avivi, I.; Balaban, R.; Shragai, T.; Sheffer, G.; Morales, M.; Aharon, A.; Lowenton-Spier, N.; Trestman, S.; Perry, C.; Benyamini, N. Humoral response rate and predictors of response to BNT162b2 mRNA COVID19 vaccine in patients with multiple myeloma. Br. J. Haematol. 2021, 195, 186–193. [Google Scholar] [CrossRef]
  38. Pimpinelli, F.; Marchesi, F.; Piaggio, G.; Giannarelli, D.; Papa, E.; Falcucci, P.; Spadea, A.; Pontone, M.; Di Martino, S.; Laquintana, V. Lower response to BNT162b2 vaccine in patients with myelofibrosis compared to polycythemia vera and essential thrombocythemia. J. Hematol. Oncol. 2021, 14, 1–4. [Google Scholar] [CrossRef]
  39. Caocci, G.; Mulas, O.; Mantovani, D.; Costa, A.; Galizia, A.; Barabino, L.; Greco, M.; Murru, R.; La Nasa, G. Ruxolitinib does not impair humoral immune response to COVID-19 vaccination with BNT162b2 mRNA COVID-19 vaccine in patients with myelofibrosis. Ann. Hematol. 2022, 101, 929–931. [Google Scholar] [CrossRef]
  40. Chung, D.J.; Shah, G.L.; Devlin, S.M.; Ramanathan, L.V.; Doddi, S.; Pessin, M.S.; Hoover, E.; Marcello, L.T.; Young, J.C.; Boutemine, S.R. Disease-and Therapy-Specific Impact on Humoral Immune Responses to COVID-19 Vaccination in Hematologic Malignancies Impaired COVID-19 Vaccine Responses in Hematologic Cancers. Blood Cancer Discov. 2021, 2, 568–576. [Google Scholar] [CrossRef]
  41. Easdale, S.; Shea, R.; Ellis, L.; Bazin, J.; Davis, K.; Dallas, F.; Thistlethwayte, E.; Ethell, M.; Potter, M.; Arias, C. Serologic responses following a single dose of SARS-Cov-2 vaccination in allogeneic stem cell transplantation recipients. Transplant. Cell. Ther. 2021, 27, 880.e1–880.e4. [Google Scholar] [CrossRef]
  42. Dhakal, B.; Abedin, S.; Fenske, T.; Chhabra, S.; Ledeboer, N.; Hari, P.; Hamadani, M. Response to SARS-CoV-2 vaccination in patients after hematopoietic cell transplantation and CAR T-cell therapy. Blood J. Am. Soc. Hematol. 2021, 138, 1278–1281. [Google Scholar] [CrossRef] [PubMed]
  43. Redjoul, R.; Le Bouter, A.; Beckerich, F.; Fourati, S.; Maury, S. Antibody response after second BNT162b2 dose in allogeneic HSCT recipients. Lancet 2021, 398, 298–299. [Google Scholar] [CrossRef]
  44. Mairhofer, M.; Kausche, L.; Kaltenbrunner, S.; Ghanem, R.; Stegemann, M.; Klein, K.; Pammer, M.; Rauscher, I.; Salzer, H.J.; Doppler, S. Humoral and cellular immune responses in SARS-CoV-2 mRNA-vaccinated patients with cancer. Cancer Cell 2021, 39, 1171–1172. [Google Scholar] [CrossRef] [PubMed]
  45. Del Poeta, G.; Bomben, R.; Polesel, J.; Rossi, F.M.; Pozzo, F.; Zaina, E.; Cattarossi, I.; Varaschin, P.; Nanni, P.; Boschin, R.B. COVID-19 vaccination: Evaluation of risk for protection failure in chronic lymphocytic leukemia patients. Hematol. Oncol. 2021, 39, 712. [Google Scholar] [CrossRef] [PubMed]
  46. Jurgens, E.M.; Ketas, T.J.; Zhao, Z.; Satlin, M.J.; Small, C.B.; Sukhu, A.; Francomano, E.; Klasse, P.J.; Garcia, A.; Nguyenduy, E. Serologic response to mRNA COVID-19 vaccination in lymphoma patients. Am. J. Hematol. 2021. [Google Scholar] [CrossRef]
  47. Perry, C.; Luttwak, E.; Balaban, R.; Shefer, G.; Morales, M.M.; Aharon, A.; Tabib, Y.; Cohen, Y.C.; Benyamini, N.; Beyar-Katz, O.; et al. Efficacy of the BNT162b2 mRNA COVID-19 vaccine in patients with B-cell non-Hodgkin lymphoma. Blood Adv. 2021, 5, 3053–3061. [Google Scholar] [CrossRef]
  48. Tamari, R.; Politikos, I.; Knorr, D.A.; Vardhana, S.A.; Young, J.C.; Marcello, L.T.; Doddi, S.; Devlin, S.M.; Ramanathan, L.V.; Pessin, M.S.; et al. Predictors of Humoral Response to SARS-CoV-2 Vaccination after Hematopoietic Cell Transplantation and CAR T-cell Therapy. Blood Cancer Discov. 2021, 2, 577–585. [Google Scholar] [CrossRef]
  49. Ollila, T.A.; Lu, S.; Masel, R.; Zayac, A.; Paiva, K.; Rogers, R.D.; Olszewski, A.J. Antibody Response to COVID-19 Vaccination in Adults With Hematologic Malignant Disease. JAMA Oncol. 2021, 7, 1714–1716. [Google Scholar] [CrossRef]
  50. Fendler, A.; Shepherd, S.T.C.; Au, L.; Wilkinson, K.A.; Wu, M.; Byrne, F.; Cerrone, M.; Schmitt, A.M.; Joharatnam-Hogan, N.; Shum, B.; et al. Adaptive immunity and neutralizing antibodies against SARS-CoV-2 variants of concern following vaccination in patients with cancer: The CAPTURE study. Nat. Cancer 2021, 2, 1321–1337. [Google Scholar] [CrossRef]
  51. Kastritis, E.; Terpos, E.; Sklirou, A.; Theodorakakou, F.; Fotiou, D.; Papanagnou, E.D.; Bagratuni, T.; Kanellias, N.; Gavriatopoulou, M.; Trougakos, I.P.; et al. Antibody Response After Initial Vaccination for SARS-CoV-2 in Patients With Amyloidosis. Hemasphere 2021, 5, e614. [Google Scholar] [CrossRef] [PubMed]
  52. Ehmsen, S.; Asmussen, A.; Jeppesen, S.S.; Nilsson, A.C.; Østerlev, S.; Vestergaard, H.; Justesen, U.S.; Johansen, I.S.; Frederiksen, H.; Ditzel, H.J. Antibody and T cell immune responses following mRNA COVID-19 vaccination in patients with cancer. Cancer Cell 2021, 39, 1034–1036. [Google Scholar] [CrossRef]
  53. Gurion, R.; Rozovski, U.; Itchaki, G.; Gafter-Gvili, A.; Leibovitch, C.; Raanani, P.; Ben-Zvi, H.; Szwarcwort, M.; Taylor-Abigadol, M.; Dann, E.J.; et al. Humoral serological response to the BNT162b2 vaccine is abrogated in lymphoma patients within the first 12 months following treatment with anti-CD2O antibodies. Haematologica 2022, 107, 715–720. [Google Scholar] [CrossRef]
  54. Roeker, L.E.; Knorr, D.A.; Thompson, M.C.; Nivar, M.; Lebowitz, S.; Peters, N.; Deonarine, I., Jr.; Momotaj, S.; Sharan, S.; Chanlatte, V.; et al. COVID-19 vaccine efficacy in patients with chronic lymphocytic leukemia. Leukemia 2021, 35, 2703–2705. [Google Scholar] [CrossRef]
  55. Benjamini, O.; Rokach, L.; Itchaki, G.; Braester, A.; Shvidel, L.; Goldschmidt, N.; Shapira, S.; Dally, N.; Avigdor, A.; Rahav, G.; et al. Safety and efficacy of the BNT162b mRNA COVID-19 vaccine in patients with chronic lymphocytic leukemia. Haematologica 2022, 107, 625–634. [Google Scholar] [CrossRef] [PubMed]
  56. Parry, H.; McIlroy, G.; Bruton, R.; Ali, M.; Stephens, C.; Damery, S.; Otter, A.; McSkeane, T.; Rolfe, H.; Faustini, S.; et al. Antibody responses after first and second Covid-19 vaccination in patients with chronic lymphocytic leukaemia. Blood Cancer J. 2021, 11, 136. [Google Scholar] [CrossRef] [PubMed]
  57. Malard, F.; Gaugler, B.; Gozlan, J.; Bouquet, L.; Fofana, D.; Siblany, L.; Eshagh, D.; Adotevi, O.; Laheurte, C.; Ricard, L.; et al. Weak immunogenicity of SARS-CoV-2 vaccine in patients with hematologic malignancies. Blood Cancer J. 2021, 11, 142. [Google Scholar] [CrossRef] [PubMed]
  58. Benda, M.; Mutschlechner, B.; Ulmer, H.; Grabher, C.; Severgnini, L.; Volgger, A.; Reimann, P.; Lang, T.; Atzl, M.; Huynh, M.; et al. Serological SARS-CoV-2 antibody response, potential predictive markers and safety of BNT162b2 mRNA COVID-19 vaccine in haematological and oncological patients. Br. J. Haematol. 2021, 195, 523–531. [Google Scholar] [CrossRef]
  59. Van Oekelen, O.; Gleason, C.R.; Agte, S.; Srivastava, K.; Beach, K.F.; Aleman, A.; Kappes, K.; Mouhieddine, T.H.; Wang, B.; Chari, A.; et al. Highly variable SARS-CoV-2 spike antibody responses to two doses of COVID-19 RNA vaccination in patients with multiple myeloma. Cancer Cell 2021, 39, 1028–1030. [Google Scholar] [CrossRef] [PubMed]
  60. Greenberger, L.M.; Saltzman, L.A.; Senefeld, J.W.; Johnson, P.W.; DeGennaro, L.J.; Nichols, G.L. Antibody response to SARS-CoV-2 vaccines in patients with hematologic malignancies. Cancer Cell 2021, 39, 1031–1033. [Google Scholar] [CrossRef]
  61. Ghione, P.; Gu, J.J.; Attwood, K.; Torka, P.; Goel, S.; Sundaram, S.; Mavis, C.; Johnson, M.; Thomas, R.; McWhite, K. Impaired humoral responses to COVID-19 vaccination in patients with lymphoma receiving B-cell–directed therapies. Blood 2021, 138, 811–814. [Google Scholar] [CrossRef]
  62. Ghandili, S.; Schönlein, M.; Lütgehetmann, M.; Schulze zur Wiesch, J.; Becher, H.; Bokemeyer, C.; Sinn, M.; Weisel, K.C.; Leypoldt, L.B. Post-vaccination anti-SARS-CoV-2-antibody response in patients with multiple myeloma correlates with low CD19+ B-lymphocyte count and anti-CD38 treatment. Cancers 2021, 13, 3800. [Google Scholar] [CrossRef]
  63. Gastinne, T.; Le Bourgeois, A.; Coste-Burel, M.; Guillaume, T.; Peterlin, P.; Garnier, A.; Imbert, B.M.; Drumel, T.; Mahe, B.; Dubruille, V. Safety and antibody response after one and/or two doses of BNT162b2 Anti-SARS-CoV-2 mRNA vaccine in patients treated by CAR T cells therapy. Br. J. Haematol. 2022, 196, 360. [Google Scholar] [CrossRef] [PubMed]
  64. Linardou, H.; Spanakis, N.; Koliou, G.-A.; Christopoulou, A.; Karageorgopoulou, S.; Alevra, N.; Vagionas, A.; Tsoukalas, N.; Sgourou, S.; Fountzilas, E. Responses to SARS-CoV-2 vaccination in patients with cancer (ReCOVer study): A prospective cohort study of the Hellenic Cooperative Oncology Group. Cancers 2021, 13, 4621. [Google Scholar] [CrossRef]
  65. Mair, M.J.; Berger, J.M.; Berghoff, A.S.; Starzer, A.M.; Ortmayr, G.; Puhr, H.C.; Steindl, A.; Perkmann, T.; Haslacher, H.; Strassl, R. Humoral immune response in hematooncological patients and health care workers who received SARS-CoV-2 vaccinations. JAMA Oncol. 2022, 8, 106–113. [Google Scholar] [CrossRef]
  66. Jimenez Balarezo, M.; Roldan Galvan, E.; Fernandez Naval, C.; Villacampa, G.; Martínez Gallo, M.; Medina-Gil, D.; Peralta Garzon, S.; Pujadas, G.; Hernández Buñuel, C.; Gironella Mesa, M. Cellular and humoral immunogenicity of the mRNA-1273 SARS-CoV-2 vaccine in patients with hematologic malignancies. 2022; 6, 774–784. [Google Scholar]
  67. Zeng, C.; Evans, J.P.; Reisinger, S.; Woyach, J.; Liscynesky, C.; Boghdadly, Z.E.; Rubinstein, M.P.; Chakravarthy, K.; Saif, L.; Oltz, E.M. Impaired neutralizing antibody response to COVID-19 mRNA vaccines in cancer patients. Cell Biosci. 2021, 11, 1–6. [Google Scholar] [CrossRef]
  68. Lyski, Z.L.; Kim, M.S.; Xthona Lee, D.; Raué, H.-P.; Raghunathan, V.; Griffin, J.; Ryan, D.; Brunton, A.E.; Curlin, M.E.; Slifka, M.K. Cellular and humoral immune response to mRNA COVID-19 vaccination in subjects with chronic lymphocytic leukemia. Blood Adv. 2022, 6, 1207–1211. [Google Scholar] [CrossRef] [PubMed]
  69. Shah, M.R.; Gabel, A.; Beers, S.; Salaru, G.; Lin, Y.; Cooper, D.L. COVID-19 vaccine responses in patients with plasma cell dyscrasias after complete vaccination. Clin. Lymphoma Myeloma Leuk. 2022, 22, e321–e326. [Google Scholar] [CrossRef]
  70. Greenberg, R.S.; Ruddy, J.A.; Boyarsky, B.J.; Werbel, W.A.; Garonzik-Wang, J.M.; Segev, D.L.; Imus, P.H. Safety and antibody response to two-dose SARS-CoV-2 messenger RNA vaccination in patients with multiple myeloma. BMC Cancer 2021, 21, 1–5. [Google Scholar] [CrossRef]
  71. Singer, J.; Le, N.-S.; Mattes, D.; Klamminger, V.; Hackner, K.; Kolinsky, N.; Scherb, M.; Errhalt, P.; Kreye, G.; Pecherstorfer, M. Evaluation of antibody responses to COVID-19 vaccines among solid tumor and hematologic patients. Cancers 2021, 13, 4312. [Google Scholar] [CrossRef]
  72. Chumsri, S.; Advani, P.P.; Pai, T.S.; Li, Z.; Mummareddy, A.; Acampora, M.; Reynolds, G.A.; Wylie, N.; Boyle, A.W.; Lou, Y. Humoral Responses After SARS-CoV-2 mRNA Vaccination and Breakthrough Infection in Cancer Patients. Mayo Clin. Proc. Innov. Qual. Outcomes 2022, 6, 120–125. [Google Scholar] [CrossRef] [PubMed]
  73. Cavanna, L.; Citterio, C.; Biasini, C.; Madaro, S.; Bacchetta, N.; Lis, A.; Cremona, G.; Muroni, M.; Bernuzzi, P.; Cascio, G.L. COVID-19 vaccines in adult cancer patients with solid tumours undergoing active treatment: Seropositivity and safety. A prospective observational study in Italy. Eur. J. Cancer 2021, 157, 441–449. [Google Scholar] [CrossRef]
  74. Re, D.; Barrière, J.; Chamorey, E.; Delforge, M.; Gastaud, L.; Petit, E.; Chaminade, A.; Verrière, B.; Peyrade, F. Low rate of seroconversion after mRNA anti-SARS-CoV-2 vaccination in patients with hematological malignancies. Leuk. Lymphoma 2021, 62, 3308–3310. [Google Scholar] [CrossRef] [PubMed]
  75. Fox, T.A.; Kirkwood, A.A.; Enfield, L.; O’Reilly, M.; Arulogun, S.; D’Sa, S.; O’Nions, J.; Kavi, J.; Vitsaras, E.; Townsend, W. Low seropositivity and suboptimal neutralisation rates in patients fully vaccinated against COVID-19 with B-cell malignancies. Br. J. Haematol. 2021, 195, 706. [Google Scholar] [CrossRef]
  76. Di Noia, V.; Pimpinelli, F.; Renna, D.; Barberi, V.; Maccallini, M.T.; Gariazzo, L.; Pontone, M.; Monti, A.; Campo, F.; Taraborelli, E. Immunogenicity and Safety of COVID-19 Vaccine BNT162b2 for Patients with Solid Cancer: A Large Cohort Prospective Study from a Single InstitutionBNT162b2 Vaccine for Patients with Cancer: A Large Cohort Study. Clin. Cancer Res. 2021, 27, 6815–6823. [Google Scholar] [CrossRef] [PubMed]
  77. Shroff, R.T.; Chalasani, P.; Wei, R.; Pennington, D.; Quirk, G.; Schoenle, M.V.; Peyton, K.L.; Uhrlaub, J.L.; Ripperger, T.J.; Jergović, M.; et al. Immune responses to two and three doses of the BNT162b2 mRNA vaccine in adults with solid tumors. Nat. Med. 2021, 27, 2002–2011. [Google Scholar] [CrossRef]
  78. Nelli, F.; Giannarelli, D.; Fabbri, A.; Silvestri, M.A.; Berrios, J.R.G.; Virtuoso, A.; Marrucci, E.; Schirripa, M.; Mazzotta, M.; Onorato, A.; et al. Immunogenicity and early clinical outcome after two or three doses of SARS-CoV-2 mRNA-BNT162b2 vaccine in actively treated cancer patients: Results from the prospective observational Vax-On-Third study. Ann. Oncol. 2022, 33, 740–742. [Google Scholar] [CrossRef]
  79. Zagouri, F.; Papatheodoridi, A.; Liontos, M.; Briasoulis, A.; Sklirou, A.D.; Skafida, E.; Fiste, O.; Markellos, C.; Andrikopoulou, A.; Koutsoukos, K.; et al. Assessment of Postvaccination Neutralizing Antibodies Response against SARS-CoV-2 in Cancer Patients under Treatment with Targeted Agents. Vaccines 2022, 10, 1474. [Google Scholar] [CrossRef]
  80. Le Bourgeois, A.; Coste-Burel, M.; Guillaume, T.; Peterlin, P.; Garnier, A.; Béné, M.C.; Chevallier, P. Safety and Antibody Response After 1 and 2 Doses of BNT162b2 mRNA Vaccine in Recipients of Allogeneic Hematopoietic Stem Cell Transplant. JAMA Netw. Open 2021, 4, e2126344. [Google Scholar] [CrossRef] [PubMed]
  81. Marchesi, F.; Pimpinelli, F.; Sperandio, E.; Papa, E.; Falcucci, P.; Pontone, M.; di Martino, S.; de Latouliere, L.; Orlandi, G.; Morrone, A. The 12-week kinetics of anti-SARS-CoV-2 antibodies in different haematological cancers after vaccination with BNT162b2. Br. J. Haematol. 2022, 362–367. [Google Scholar] [CrossRef]
  82. Molica, S.; Giannarelli, D.; Lentini, M.; Zappala, D.; Mannella, A.; Loiacono, D.; Gianfelici, V.; Panduri, G.; Gariani, I.; Minchella, P.; et al. Efficacy of the BNT162b2 mRNA COVID-19 Vaccine in Patients with Chronic Lymphocytic Leukemia: A Serologic and Cellular Study. Chemotherapy 2022, 67, 91–95. [Google Scholar] [CrossRef]
  83. Tsang, H.F.; Chan, L.W.C.; Cho, W.C.S.; Yu, A.C.S.; Yim, A.K.Y.; Chan, A.K.C.; Ng, L.P.W.; Wong, Y.K.E.; Pei, X.M.; Li, M.J.W.; et al. An update on COVID-19 pandemic: The epidemiology, pathogenesis, prevention and treatment strategies. Expert Rev. Anti. Infect. Ther. 2021, 19, 877–888. [Google Scholar] [CrossRef]
  84. Terpos, E.; Fotiou, D.; Karalis, V.; Ntanasis-Stathopoulos, I.; Sklirou, A.D.; Gavriatopoulou, M.; Malandrakis, P.; Iconomidou, V.A.; Kastritis, E.; Trougakos, I.P.; et al. SARS-CoV-2 humoral responses following booster BNT162b2 vaccination in patients with B-cell malignancies. Am. J. Hematol. 2022, 97, 1300–1308. [Google Scholar] [CrossRef] [PubMed]
  85. Greenberger, L.M.; Saltzman, L.A.; Senefeld, J.W.; Johnson, P.W.; DeGennaro, L.J.; Nichols, G.L. Anti-spike antibody response to SARS-CoV-2 booster vaccination in patients with B cell-derived hematologic malignancies. Cancer Cell 2021, 39, 1297–1299. [Google Scholar] [CrossRef]
  86. Terpos, E.; Liontos, M.; Fiste, O.; Zagouri, F.; Briasoulis, A.; Sklirou, A.D.; Markellos, C.; Skafida, E.; Papatheodoridi, A.; Andrikopoulou, A.; et al. SARS-CoV-2 Neutralizing Antibodies Kinetics Postvaccination in Cancer Patients under Treatment with Immune Checkpoint Inhibition. Cancers 2022, 14. [Google Scholar] [CrossRef]
  87. Shapiro, L.C.; Thakkar, A.; Campbell, S.T.; Forest, S.K.; Pradhan, K.; Gonzalez-Lugo, J.D.; Quinn, R.; Bhagat, T.D.; Choudhary, G.S.; McCort, M.; et al. Efficacy of booster doses in augmenting waning immune responses to COVID-19 vaccine in patients with cancer. Cancer Cell 2022, 40, 3–5. [Google Scholar] [CrossRef]
  88. Saiag, E.; Grupper, A.; Avivi, I.; Elkayam, O.; Ram, R.; Herishanu, Y.; Cohen, Y.; Perry, C.; Furer, V.; Katchman, H.; et al. The effect of a third-dose BNT162b2 vaccine on anti-SARS-CoV-2 antibody levels in immunosuppressed patients. Clin. Microbiol. Infect. 2022, 28, 735.e5–735.e8. [Google Scholar] [CrossRef]
  89. Schubert, L.; Koblischke, M.; Schneider, L.; Porpaczy, E.; Winkler, F.; Jaeger, U.; Blüml, S.; Haslacher, H.; Burgmann, H.; Aberle, J.H.; et al. Immunogenicity of COVID-19 Vaccinations in Hematological Patients: 6-Month Follow-Up and Evaluation of a 3rd Vaccination. Cancers 2022, 14, 1962. [Google Scholar] [CrossRef] [PubMed]
  90. Di Giacomo, A.M.; Giacobini, G.; Anichini, G.; Gandolfo, C.; D’Alonzo, V.; Calabrò, L.; Lofiego, M.F.; Cusi, M.G.; Maio, M. SARS-CoV-2 infection in cancer patients on active therapy after the booster dose of mRNA vaccines. Eur. J. Cancer 2022, 171, 143–149. [Google Scholar] [CrossRef]
  91. Ollila, T.A.; Masel, R.H.; Reagan, J.L.; Lu, S.; Rogers, R.D.; Paiva, K.J.; Taher, R.; Burguera-Couce, E.; Zayac, A.S.; Yakirevich, I.; et al. Seroconversion and outcomes after initial and booster COVID-19 vaccination in adults with hematologic malignancies. Cancer 2022, 128, 3319–3329. [Google Scholar] [CrossRef]
  92. Mair, M.J.; Berger, J.M.; Mitterer, M.; Gansterer, M.; Bathke, A.C.; Trutschnig, W.; Berghoff, A.S.; Perkmann, T.; Haslacher, H.; Lamm, W.W.; et al. Third dose of SARS-CoV-2 vaccination in hemato-oncological patients and health care workers: Immune responses and adverse events - a retrospective cohort study. Eur. J. Cancer 2022, 165, 184–194. [Google Scholar] [CrossRef] [PubMed]
  93. Kimura, M.; Ferreira, V.H.; Kothari, S.; Pasic, I.; Mattsson, J.I.; Kulasingam, V.; Humar, A.; Mah, A.; Delisle, J.S.; Ierullo, M.; et al. Safety and Immunogenicity After a Three-Dose SARS-CoV-2 Vaccine Schedule in Allogeneic Stem Cell Transplant Recipients. Transpl. Cell Ther. 2022, 28, 706.e1–706.e10. [Google Scholar] [CrossRef] [PubMed]
  94. Shapiro, M.; Landau, R.; Shay, S.; Kaminsky, M.; Verhovsky, G. Early detection of COVID-19 outbreaks using textual analysis of electronic medical records. J. Clin. Virol. 2022, 155, 105251. [Google Scholar] [CrossRef] [PubMed]
  95. Lim, S.H.; Stuart, B.; Joseph-Pietras, D.; Johnson, M.; Campbell, N.; Kelly, A.; Jeffrey, D.; Turaj, A.H.; Rolfvondenbaumen, K.; Galloway, C.; et al. Immune responses against SARS-CoV-2 variants after two and three doses of vaccine in B-cell malignancies: UK PROSECO study. Nat. Cancer 2022, 3, 552–564. [Google Scholar] [CrossRef] [PubMed]
  96. Gössi, S.; Bacher, U.; Haslebacher, C.; Nagler, M.; Suter, F.; Staehelin, C.; Novak, U.; Pabst, T. Humoral Responses to Repetitive Doses of COVID-19 mRNA Vaccines in Patients with CAR-T-Cell Therapy. Cancers 2022, 14, 3527. [Google Scholar] [CrossRef]
  97. Lasagna, A.; Bergami, F.; Lilleri, D.; Percivalle, E.; Quaccini, M.; Alessio, N.; Comolli, G.; Sarasini, A.; Sammartino, J.C.; Ferrari, A.; et al. Immunogenicity and safety after the third dose of BNT162b2 anti-SARS-CoV-2 vaccine in patients with solid tumors on active treatment: A prospective cohort study. ESMO Open 2022, 7, 100458. [Google Scholar] [CrossRef]
  98. Canti, L.; Ariën, K.K.; Desombere, I.; Humblet-Baron, S.; Pannus, P.; Heyndrickx, L.; Henry, A.; Servais, S.; Willems, E.; Ehx, G.; et al. Antibody response against SARS-CoV-2 Delta and Omicron variants after third-dose BNT162b2 vaccination in allo-HCT recipients. Cancer Cell 2022, 40, 335–337. [Google Scholar] [CrossRef]
  99. Corradini, P.; Agrati, C.; Apolone, G.; Mantovani, A.; Giannarelli, D.; Marasco, V.; Bordoni, V.; Sacchi, A.; Matusali, G.; Salvarani, C.; et al. Humoral and T-Cell Immune Response After 3 Doses of Messenger RNA Severe Acute Respiratory Syndrome Coronavirus 2 Vaccines in Fragile Patients: The Italian VAX4FRAIL Study. Clin. Infect. Dis. 2022, 76, e426–e438. [Google Scholar] [CrossRef]
  100. Debie, Y.; Vandamme, T.; Goossens, M.E.; van Dam, P.A.; Peeters, M. Antibody titres before and after a third dose of the SARS-CoV-2 BNT162b2 vaccine in patients with cancer. Eur. J. Cancer 2022, 163, 177–179. [Google Scholar] [CrossRef]
  101. Reimann, P.; Ulmer, H.; Mutschlechner, B.; Benda, M.; Severgnini, L.; Volgger, A.; Lang, T.; Atzl, M.; Huynh, M.; Gasser, K.; et al. Efficacy and safety of heterologous booster vaccination with Ad26.COV2.S after BNT162b2 mRNA COVID-19 vaccine in haemato-oncological patients with no antibody response. Br. J. Haematol. 2022, 196, 577–584. [Google Scholar] [CrossRef] [PubMed]
  102. Di Noia, V.; Pimpinelli, F.; Renna, D.; Maccallini, M.T.; Gariazzo, L.; Riva, F.; Sperandio, E.; Giannarelli, D.; Cognetti, F. Potentiation of humoral response to the BNT162b2 vaccine after the third dose in patients with solid cancer. Ann. Oncol. 2022, 33, 563–565. [Google Scholar] [CrossRef] [PubMed]
  103. Terpos, E.; Gavriatopoulou, M.; Ntanasis-Stathopoulos, I.; Briasoulis, A.; Gumeni, S.; Malandrakis, P.; Papanagnou, E.D.; Migkou, M.; Kanellias, N.; Kastritis, E.; et al. Booster BNT162b2 optimizes SARS-CoV-2 humoral response in patients with myeloma: The negative effect of anti-BCMA therapy. Blood 2022, 139, 1409–1412. [Google Scholar] [CrossRef]
  104. Thompson, M.A.; Hallmeyer, S.; Fitzpatrick, V.E.; Liao, Y.; Mullane, M.P.; Medlin, S.C.; Copeland, K.; Weese, J.L. Real-World Third COVID-19 Vaccine Dosing and Antibody Response in Patients With Hematologic Malignancies. J. Patient Cent. Res. Rev. 2022, 9, 149–157. [Google Scholar] [CrossRef]
  105. Galitzia, A.; Barabino, L.; Murru, R.; Caocci, G.; Greco, M.; Angioni, G.; Mulas, O.; Oppi, S.; Massidda, S.; Costa, A.; et al. Patients with Chronic Lymphocytic Leukemia Have a Very High Risk of Ineffective Response to the BNT162b2 Vaccine. Vaccines 2022, 10, 1162. [Google Scholar] [CrossRef] [PubMed]
  106. Ligumsky, H.; Dor, H.; Etan, T.; Golomb, I.; Nikolaevski-Berlin, A.; Greenberg, I.; Halperin, T.; Angel, Y.; Henig, O.; Spitzer, A.; et al. Immunogenicity and safety of BNT162b2 mRNA vaccine booster in actively treated patients with cancer. Lancet Oncol. 2022, 23, 193–195. [Google Scholar] [CrossRef]
  107. Mellinghoff, S.C.; Mayer, L.; Robrecht, S.; Weskamm, L.M.; Dahlke, C.; Gruell, H.; Schlotz, M.; Vanshylla, K.; Schloser, H.A.; Thelen, M.; et al. SARS-CoV-2-specific cellular response following third COVID-19 vaccination in patients with chronic lymphocytic leukemia. Haematologica 2022, 107, 2480–2484. [Google Scholar] [CrossRef]
  108. Beaton, B.; Sasson, S.C.; Rankin, K.; Raedemaeker, J.; Wong, A.; Hastak, P.; Phetsouphanh, C.; Warden, A.; Klemm, V.; Munier, C.M.L.; et al. Patients with treated indolent lymphomas immunized with BNT162b2 have reduced anti-spike neutralizing IgG to SARS-CoV-2 variants, but preserved antigen-specific T cell responses. Am. J. Hematol. 2023, 98, 131–139. [Google Scholar] [CrossRef] [PubMed]
  109. Wagner, A.; Garner-Spitzer, E.; Schötta, A.M.; Orola, M.; Wessely, A.; Zwazl, I.; Ohradanova-Repic, A.; Weseslindtner, L.; Tajti, G.; Gebetsberger, L.; et al. SARS-CoV-2-mRNA Booster Vaccination Reverses Non-Responsiveness and Early Antibody Waning in Immunocompromised Patients—A Phase Four Study Comparing Immune Responses in Patients With Solid Cancers, Multiple Myeloma and Inflammatory Bowel Disease. Front. Immunol. 2022, 13, 889138. [Google Scholar] [CrossRef]
  110. Aleman, A.; Van Oekelen, O.; Upadhyaya, B.; Beach, K.; Kogan Zajdman, A.; Alshammary, H.; Serebryakova, K.; Agte, S.; Kappes, K.; Gleason, C.R.; et al. Augmentation of humoral and cellular immune responses after third-dose SARS-CoV-2 vaccination and viral neutralization in myeloma patients. Cancer Cell 2022, 40, 441–443. [Google Scholar] [CrossRef]
  111. Funakoshi, Y.; Yakushijin, K.; Ohji, G.; Hojo, W.; Sakai, H.; Watanabe, M.; Kitao, A.; Miyata, Y.; Saito, Y.; Kawamoto, S.; et al. Promising Efficacy of a Third Dose of mRNA SARS-CoV-2 Vaccination in Patients Treated with Anti-CD20 Antibody Who Failed 2-Dose Vaccination. Vaccines 2022, 10, 965. [Google Scholar] [CrossRef] [PubMed]
  112. Fendler, A.; Shepherd, S.T.C.; Au, L.; Wilkinson, K.A.; Wu, M.; Schmitt, A.M.; Tippu, Z.; Farag, S.; Rogiers, A.; Harvey, R.; et al. Immune responses following third COVID-19 vaccination are reduced in patients with hematological malignancies compared to patients with solid cancer. Cancer Cell 2022, 40, 114–116. [Google Scholar] [CrossRef] [PubMed]
  113. Abid, M.B.; Rubin, M.; Ledeboer, N.; Szabo, A.; Longo, W.; Mohan, M.; Shah, N.N.; Fenske, T.S.; Abedin, S.; Runaas, L.; et al. Efficacy of a third SARS-CoV-2 mRNA vaccine dose among hematopoietic cell transplantation, CAR T cell, and BiTE recipients. Cancer Cell 2022, 40, 340–342. [Google Scholar] [CrossRef] [PubMed]
  114. Ehmsen, S.; Asmussen, A.; Jeppesen, S.S.; Nilsson, A.C.; Østerlev, S.; Kragh, A.; Frederiksen, H.; Ditzel, H.J. Antibody responses following third mRNA COVID-19 vaccination in patients with cancer and potential timing of a fourth vaccination. Cancer Cell 2022, 40, 338–339. [Google Scholar] [CrossRef] [PubMed]
  115. Oosting, S.F.; van der Veldt, A.A.M.; Fehrmann, R.S.N.; GeurtsvanKessel, C.H.; van Binnendijk, R.S.; Dingemans, A.C.; Smit, E.F.; Hiltermann, T.J.N.; den Hartog, G.; Jalving, M.; et al. Immunogenicity after second and third mRNA-1273 vaccination doses in patients receiving chemotherapy, immunotherapy, or both for solid tumours. Lancet Oncol. 2022, 23, 833–835. [Google Scholar] [CrossRef]
  116. Enssle, J.C.; Campe, J.; Büchel, S.; Moter, A.; See, F.; Grießbaum, K.; Rieger, M.A.; Wolf, S.; Ballo, O.; Steffen, B.; et al. Enhanced but variant-dependent serological and cellular immune responses to third-dose BNT162b2 vaccination in patients with multiple myeloma. Cancer Cell 2022, 40, 587–589. [Google Scholar] [CrossRef]
  117. Einarsdottir, S.; Martner, A.; Nicklasson, M.; Wiktorin, H.G.; Arabpour, M.; Törnell, A.; Vaht, K.; Waldenström, J.; Ringlander, J.; Bergström, T.; et al. Reduced immunogenicity of a third COVID-19 vaccination among recipients of allogeneic hematopoietic stem cell transplantation. Haematologica 2022, 107, 1479–1482. [Google Scholar] [CrossRef] [PubMed]
  118. Bagacean, C.; Letestu, R.; Al-Nawakil, C.; Brichler, S.; Lévy, V.; Sritharan, N.; Delmer, A.; Dartigeas, C.; Leblond, V.; Roos-Weil, D.; et al. Humoral response to mRNA anti-COVID-19 vaccines BNT162b2 and mRNA-1273 in patients with chronic lymphocytic leukemia. Blood Adv. 2022, 6, 207–211. [Google Scholar] [CrossRef] [PubMed]
  119. Rottenberg, Y.; Grinshpun, A.; Ben-Dov, I.Z.; Oiknine Djian, E.; Wolf, D.G.; Kadouri, L. Assessment of Response to a Third Dose of the SARS-CoV-2 BNT162b2 mRNA Vaccine in Patients With Solid Tumors Undergoing Active Treatment. JAMA Oncol. 2022, 8, 300–301. [Google Scholar] [CrossRef]
  120. Fenioux, C.; Teixeira, L.; Fourati, S.; Melica, G.; Lelievre, J.D.; Gallien, S.; Zalcman, G.; Pawlotsky, J.M.; Tournigand, C. SARS-CoV-2 Antibody Response to 2 or 3 Doses of the BNT162b2 Vaccine in Patients Treated With Anticancer Agents. JAMA Oncol. 2022, 8, 612–617. [Google Scholar] [CrossRef] [PubMed]
  121. Gressens, S.B.; Fourati, S.; Le Bouter, A.; Le Bras, F.; Dupuis, J.; Hammoud, M.; El Gnaoui, T.; Gounot, R.; Roulin, L.; Belhadj, K.; et al. Anti-SARS-CoV-2 antibody response after 2 and 3 doses of BNT162b2 mRNA vaccine in patients with lymphoid malignancies. Clin. Microbiol. Infect. 2022, 28, 885.e7–885.e11. [Google Scholar] [CrossRef]
  122. Avivi, I.; Luttwak, E.; Saiag, E.; Halperin, T.; Haberman, S.; Sarig, A.; Levi, S.; Aharon, A.; Herishanu, Y.; Perry, C. BNT162b2 mRNA COVID-19 vaccine booster induces seroconversion in patients with B-cell non-Hodgkin lymphoma who failed to respond to two prior vaccine doses. Br. J. Haematol. 2022, 196, 1329–1333. [Google Scholar] [CrossRef]
  123. Šušol, O.; Hájková, B.; Zelená, H.; Hájek, R. Third dose of COVID-19 vaccine restores immune response in patients with haematological malignancies after loss of protective antibody titres. Br. J. Haematol. 2022, 197, 302–305. [Google Scholar] [CrossRef] [PubMed]
  124. Sherman, A.C.; Crombie, J.L.; Cheng, C.; Desjardins, M.; Zhou, G.; Ometoruwa, O.; Rooks, R.; Senussi, Y.; McDonough, M.; Guerrero, L.I.; et al. Immunogenicity of a Three-Dose Primary Series of mRNA COVID-19 Vaccines in Patients With Lymphoid Malignancies. Open Forum Infect. Dis. 2022, 9, ofac417. [Google Scholar] [CrossRef] [PubMed]
  125. Storti, P.; Marchica, V.; Vescovini, R.; Franceschi, V.; Russo, L.; Notarfranchi, L.; Raimondi, V.; Toscani, D.; Burroughs Garcia, J.; Costa, F.; et al. Immune response to SARS-CoV-2 mRNA vaccination and booster dose in patients with multiple myeloma and monoclonal gammopathies: Impact of Omicron variant on the humoral response. Oncoimmunology 2022, 11, 2120275. [Google Scholar] [CrossRef] [PubMed]
  126. Bryer, E.; Paul, S.; Chen, J.; Pleyer, C.; Wiestner, A.; Sun, C. CLL-140 Booster and BTKi Interruption Improve Response to SARS-CoV-2 Vaccine in Patients With CLL. Clin. Lymphoma Myeloma Leuk. 2022, 22, S270–S271. [Google Scholar] [CrossRef]
  127. Chang, A.; Lai, L.; Akhtar, A.; Linderman, S.; Orellana-Noia, V.; Saini, M.; Valanparambil, R.; Blum, K.; Allen, P.; Lechowicz, M. CLL-515 Antibody Responses Against SARS-CoV-2 Variants after Booster Vaccination in Patients With B Cell Non-Hodgkin Lymphoma and Chronic Lymphocytic Leukemia. Clin. Lymphoma Myeloma Leuk. 2022, 22, S281–S282. [Google Scholar] [CrossRef]
  128. St-Pierre, F.; Doukas, P.; Boyer, J.; Nieves, M.; Ma, S. CLL-211 Humoral Immune Response Following COVID-19 Vaccination in Patients With Chronic Lymphocytic Leukemia (CLL) and Indolent Non-Hodgkin Lymphoma (NHL): Results From a Large, Single-Center Observational Study. Clin. Lymphoma Myeloma Leuk. 2022, 22, S273. [Google Scholar] [CrossRef]
  129. Su, E.; Fischer, S.; Demmer-Steingruber, R.; Nigg, S.; Güsewell, S.; Albrich, W.C.; Rothermundt, C.; Silzle, T.; Kahlert, C.R. Humoral and cellular responses to mRNA-based COVID-19 booster vaccinations in patients with solid neoplasms under active treatment. ESMO Open 2022, 7, 100587. [Google Scholar] [CrossRef]
  130. Lee, H.K.; Hoechstetter, M.A.; Buchner, M.; Pham, T.T.; Huh, J.W.; Muller, K.; Zange, S.; von Buttlar, H.; Girl, P.; Wolfel, R. Comprehensive analysis of immune responses in CLL patients after heterologous COVID-19 vaccination. medRxiv 2022. [Google Scholar] [CrossRef]
  131. Haggenburg, S.; Hofsink, Q.; Lissenberg-Witte, B.I.; Broers, A.E.C.; van Doesum, J.A.; van Binnendijk, R.S.; den Hartog, G.; Bhoekhan, M.S.; Haverkate, N.J.E.; Burger, J.A.; et al. Antibody Response in Immunocompromised Patients With Hematologic Cancers Who Received a 3-Dose mRNA-1273 Vaccination Schedule for COVID-19. JAMA Oncol. 2022, 8, 1477–1483. [Google Scholar] [CrossRef]
  132. Blixt, L.; Gao, Y.; Wullimann, D.; Murén Ingelman-Sundberg, H.; Muschiol, S.; Healy, K.; Bogdanovic, G.; Pin, E.; Nilsson, P.; Kjellander, C.; et al. Hybrid immunity in immunocompromised patients with CLL after SARS-CoV-2 infection followed by booster mRNA vaccination. Blood 2022, 140, 2403–2407. [Google Scholar] [CrossRef]
  133. Lasagna, A.; Bergami, F.; Lilleri, D.; Percivalle, E.; Quaccini, M.; Serra, F.; Comolli, G.; Sarasini, A.; Sammartino, J.C.; Ferrari, A.; et al. Six-month humoral and cellular immune response to the third dose of BNT162b2 anti-SARS-CoV-2 vaccine in patients with solid tumors: A longitudinal cohort study with a focus on the variants of concern. ESMO Open 2022, 7, 100574. [Google Scholar] [CrossRef] [PubMed]
  134. Terpos, E.; Karalis, V.; Ntanasis-Stathopoulos, I.; Evangelakou, Z.; Gavriatopoulou, M.; Manola, M.S.; Malandrakis, P.; Gianniou, D.D.; Kastritis, E.; Trougakos, I.P.; et al. Comparison of Neutralizing Antibody Responses at 6 Months Post Vaccination with BNT162b2 and AZD1222. Biomedicines 2022, 10, 338. [Google Scholar] [CrossRef]
  135. Terpos, E.; Trougakos, I.P.; Karalis, V.; Ntanasis-Stathopoulos, I.; Sklirou, A.D.; Bagratuni, T.; Papanagnou, E.D.; Patseas, D.; Gumeni, S.; Malandrakis, P.; et al. Comparison of neutralizing antibody responses against SARS-CoV-2 in healthy volunteers who received the BNT162b2 mRNA or the AZD1222 vaccine: Should the second AZD1222 vaccine dose be given earlier? Am. J. Hematol. 2021, 96, e321–e324. [Google Scholar] [CrossRef]
  136. Noori, M.; Azizi, S.; Abbasi Varaki, F.; Nejadghaderi, S.A.; Bashash, D. A systematic review and meta-analysis of immune response against first and second doses of SARS-CoV-2 vaccines in adult patients with hematological malignancies. Int. Immunopharmacol. 2022, 110, 109046. [Google Scholar] [CrossRef] [PubMed]
  137. Moujaess, E.; Kourie, H.R.; Ghosn, M. Cancer patients and research during COVID-19 pandemic: A systematic review of current evidence. Crit. Rev. Oncol. Hematol. 2020, 150, 102972. [Google Scholar] [CrossRef]
  138. Ohm, J.E.; Carbone, D.P. Immune dysfunction in cancer patients. Oncology 2002, 16, 11–18. [Google Scholar]
  139. Allegra, A.; Tonacci, A.; Musolino, C.; Pioggia, G.; Gangemi, S. Secondary immunodeficiency in hematological malignancies: Focus on multiple myeloma and chronic lymphocytic leukemia. Front. Immunol. 2021, 12. [Google Scholar] [CrossRef]
  140. Rosati, M.; Terpos, E.; Bear, J.; Burns, R.; Devasundaram, S.; Ntanasis-Stathopoulos, I.; Gavriatopoulou, M.; Kastritis, E.; Dimopoulos, M.A.; Pavlakis, G.N.; et al. Low Spike Antibody Levels and Impaired BA.4/5 Neutralization in Patients with Multiple Myeloma or Waldenstrom’s Macroglobulinemia after BNT162b2 Booster Vaccination. Cancers 2022, 14. [Google Scholar] [CrossRef]
  141. Kleber, M.; Ntanasis-Stathopoulos, I.; Terpos, E. BCMA in Multiple Myeloma-A Promising Key to Therapy. J. Clin. Med. 2021, 10. [Google Scholar] [CrossRef] [PubMed]
  142. Pinato, D.J.; Tabernero, J.; Bower, M.; Scotti, L.; Patel, M.; Colomba, E.; Dolly, S.; Loizidou, A.; Chester, J.; Mukherjee, U.; et al. Prevalence and impact of COVID-19 sequelae on treatment and survival of patients with cancer who recovered from SARS-CoV-2 infection: Evidence from the OnCovid retrospective, multicentre registry study. Lancet Oncol. 2021, 22, 1669–1680. [Google Scholar] [CrossRef]
  143. Saco, T.V.; Strauss, A.T.; Ledford, D.K. Hepatitis B vaccine nonresponders: Possible mechanisms and solutions. Ann. Allergy Asthma Immunol. 2018, 121, 320–327. [Google Scholar] [CrossRef] [PubMed]
  144. Herati, R.S.; Knorr, D.A.; Vella, L.A.; Silva, L.V.; Chilukuri, L.; Apostolidis, S.A.; Huang, A.C.; Muselman, A.; Manne, S.; Kuthuru, O.; et al. PD-1 directed immunotherapy alters Tfh and humoral immune responses to seasonal influenza vaccine. Nat. Immunol. 2022, 23, 1183–1192. [Google Scholar] [CrossRef]
  145. Gagelmann, N.; Passamonti, F.; Wolschke, C.; Massoud, R.; Niederwieser, C.; Adjallé, R.; Mora, B.; Ayuk, F.; Kröger, N. Antibody response after vaccination against SARS-CoV-2 in adults with hematological malignancies: A systematic review and meta-analysis. Haematologica 2022, 107, 1840–1849. [Google Scholar] [CrossRef] [PubMed]
  146. Papadopoulos, D.; Ntanasis-Stathopoulos, I.; Gavriatopoulou, M.; Evangelakou, Z.; Malandrakis, P.; Manola, M.S.; Gianniou, D.D.; Kastritis, E.; Trougakos, I.P.; Dimopoulos, M.A.; et al. Predictive Factors for Neutralizing Antibody Levels Nine Months after Full Vaccination with BNT162b2: Results of a Machine Learning Analysis. Biomedicines 2022, 10, 204. [Google Scholar] [CrossRef] [PubMed]
  147. Rosati, M.; Terpos, E.; Agarwal, M.; Karalis, V.; Bear, J.; Burns, R.; Hu, X.; Papademetriou, D.; Ntanasis-Stathopoulos, I.; Trougakos, I.P.; et al. Distinct neutralization profile of spike variants by antibodies induced upon SARS-CoV-2 infection or vaccination. Am. J. Hematol. 2022, 97, E3–E7. [Google Scholar] [CrossRef]
  148. Gavriatopoulou, M.; Terpos, E.; Malandrakis, P.; Ntanasis-Stathopoulos, I.; Briasoulis, A.; Gumeni, S.; Fotiou, D.; Papanagnou, E.D.; Migkou, M.; Theodorakakou, F.; et al. Myeloma patients with COVID-19 have superior antibody responses compared to patients fully vaccinated with the BNT162b2 vaccine. Br. J. Haematol. 2022, 196, 356–359. [Google Scholar] [CrossRef]
  149. Meschi, S.; Matusali, G.; Colavita, F.; Lapa, D.; Bordi, L.; Puro, V.; Leoni, B.D.; Galli, C.; Capobianchi, M.R.; Castilletti, C. Predicting the protective humoral response to a SARS-CoV-2 mRNA vaccine. Clin Chem. Lab Med. 2021, 59, 2010–2018. [Google Scholar] [CrossRef]
  150. Valcourt, E.J.; Manguiat, K.; Robinson, A.; Lin, Y.C.; Abe, K.T.; Mubareka, S.; Shigayeva, A.; Zhong, Z.; Girardin, R.C.; DuPuis, A.; et al. Evaluating Humoral Immunity against SARS-CoV-2: Validation of a Plaque-Reduction Neutralization Test and a Multilaboratory Comparison of Conventional and Surrogate Neutralization Assays. Microbiol. Spectr. 2021, 9, e0088621. [Google Scholar] [CrossRef]
  151. Stocking, C.; de Miguel, L.; Suteu, G.; Dressel, A.; Soricelli, A.; Roskos, M.; Valor, S.; Mutschmann, C.; März, W. Evaluation of five widely used serologic assays for antibodies to SARS-CoV-2. Diagn. Microbiol. Infect. Dis. 2022, 102, 115587. [Google Scholar] [CrossRef]
  152. Gupta, S.; Agrawal, S.; Sandoval, A.; Su, H.; Tran, M.; Demirdag, Y. SARS-CoV-2-Specific and Functional Cytotoxic CD8 Cells in Primary Antibody Deficiency: Natural Infection and Response to Vaccine. J. Clin. Immunol. 2022, 42, 914–922. [Google Scholar] [CrossRef]
  153. Ganji, A.; Farahani, I.; Khansarinejad, B.; Ghazavi, A.; Mosayebi, G. Increased expression of CD8 marker on T-cells in COVID-19 patients. Blood Cells Mol. Dis. 2020, 83, 102437. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Flow chart.
Figure 1. Flow chart.
Cancers 15 02266 g001
Figure 2. Pooled EffectSize (ES) for the immune seroconversion rates of patients with hematological malignancies after the first dose of COVID-19 vaccine. [7,19,20,25,26,27,29,30,31,36,38,40,41,48,50,51,56,57,62,63,66,75,80].
Figure 2. Pooled EffectSize (ES) for the immune seroconversion rates of patients with hematological malignancies after the first dose of COVID-19 vaccine. [7,19,20,25,26,27,29,30,31,36,38,40,41,48,50,51,56,57,62,63,66,75,80].
Cancers 15 02266 g002
Figure 3. Pooled EffectSize (ES) for the immune seroconversion rates of patients with solid tumor after the first dose of COVID-19 vaccine [17,19,24,26,29,31,36,40,48,51,56,63,76,81].
Figure 3. Pooled EffectSize (ES) for the immune seroconversion rates of patients with solid tumor after the first dose of COVID-19 vaccine [17,19,24,26,29,31,36,40,48,51,56,63,76,81].
Cancers 15 02266 g003
Figure 4. Pooled EffectSize (ES) for the immune seroconversion rates of patients with hematological malignancies after the second dose of COVID-19 vaccine. [7,8,15,17,18,20,21,22,23,24,25,28,29,32,35,36,37,38,39,40,42,43,44,45,46,47,48,49,50,52,53,54,55,56,57,58,59,60,61,63,66,67,68,69,70,71,74,75,80,81,82,84].
Figure 4. Pooled EffectSize (ES) for the immune seroconversion rates of patients with hematological malignancies after the second dose of COVID-19 vaccine. [7,8,15,17,18,20,21,22,23,24,25,28,29,32,35,36,37,38,39,40,42,43,44,45,46,47,48,49,50,52,53,54,55,56,57,58,59,60,61,63,66,67,68,69,70,71,74,75,80,81,82,84].
Cancers 15 02266 g004
Figure 5. Pooled EffectSize (ES) for the immune seroconversion rates of patients with solid tumors after the second dose of COVID-19 vaccine. [7,15,17,18,20,22,23,24,25,28,35,36,37,38,39,40,40,42,43,44,45,46,47,48,49,52,53,54,55,57,58,59,60,61,63,66,67,68,69,71,72,74,80,81,82,85,86].
Figure 5. Pooled EffectSize (ES) for the immune seroconversion rates of patients with solid tumors after the second dose of COVID-19 vaccine. [7,15,17,18,20,22,23,24,25,28,35,36,37,38,39,40,40,42,43,44,45,46,47,48,49,52,53,54,55,57,58,59,60,61,63,66,67,68,69,71,72,74,80,81,82,85,86].
Cancers 15 02266 g005
Figure 6. Pooled EffectSize (ES) for the immune seroconversion rates of patients with hematological malignancies after the third dose of COVID-19 vaccine [28,74,85,87,88,89,91,92,93,95,96,97,98,99,100,101,104,105,107,108,109,110,111,112,113,114,114,116,117,118,122,123,124,125,126,127,128,129,130,131,132,134,135].
Figure 6. Pooled EffectSize (ES) for the immune seroconversion rates of patients with hematological malignancies after the third dose of COVID-19 vaccine [28,74,85,87,88,89,91,92,93,95,96,97,98,99,100,101,104,105,107,108,109,110,111,112,113,114,114,116,117,118,122,123,124,125,126,127,128,129,130,131,132,134,135].
Cancers 15 02266 g006
Figure 7. Pooled RelativeRisk (RR) for the immune seroconversion of patients with hematological malignancies after the third dose of COVID-19 vaccine [95,99,103,107,108,109,110,116,124,127,131].
Figure 7. Pooled RelativeRisk (RR) for the immune seroconversion of patients with hematological malignancies after the third dose of COVID-19 vaccine [95,99,103,107,108,109,110,116,124,127,131].
Cancers 15 02266 g007
Figure 8. Pooled EffectSize (ES) for the immune seroconversion rates of patients with solid tumors after the third dose of COVID-19 vaccine. [33,67,77,78,79,87,90,92,99,102,106,109,112,114,115,119,120,127,129,132,133,135].
Figure 8. Pooled EffectSize (ES) for the immune seroconversion rates of patients with solid tumors after the third dose of COVID-19 vaccine. [33,67,77,78,79,87,90,92,99,102,106,109,112,114,115,119,120,127,129,132,133,135].
Cancers 15 02266 g008
Figure 9. Pooled RelativeRisk (RR) for the immune seroconversion of patients with solid tumors after the third dose of COVID-19 vaccine [79,99,106,109,127,129,135].
Figure 9. Pooled RelativeRisk (RR) for the immune seroconversion of patients with solid tumors after the third dose of COVID-19 vaccine [79,99,106,109,127,129,135].
Cancers 15 02266 g009
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Liatsou, E.; Ntanasis-Stathopoulos, I.; Lykos, S.; Ntanasis-Stathopoulos, A.; Gavriatopoulou, M.; Psaltopoulou, T.; Sergentanis, T.N.; Terpos, E. Adult Patients with Cancer Have Impaired Humoral Responses to Complete and Booster COVID-19 Vaccination, Especially Those with Hematologic Cancer on Active Treatment: A Systematic Review and Meta-Analysis. Cancers 2023, 15, 2266. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers15082266

AMA Style

Liatsou E, Ntanasis-Stathopoulos I, Lykos S, Ntanasis-Stathopoulos A, Gavriatopoulou M, Psaltopoulou T, Sergentanis TN, Terpos E. Adult Patients with Cancer Have Impaired Humoral Responses to Complete and Booster COVID-19 Vaccination, Especially Those with Hematologic Cancer on Active Treatment: A Systematic Review and Meta-Analysis. Cancers. 2023; 15(8):2266. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers15082266

Chicago/Turabian Style

Liatsou, Efstathia, Ioannis Ntanasis-Stathopoulos, Stavros Lykos, Anastasios Ntanasis-Stathopoulos, Maria Gavriatopoulou, Theodora Psaltopoulou, Theodoros N. Sergentanis, and Evangelos Terpos. 2023. "Adult Patients with Cancer Have Impaired Humoral Responses to Complete and Booster COVID-19 Vaccination, Especially Those with Hematologic Cancer on Active Treatment: A Systematic Review and Meta-Analysis" Cancers 15, no. 8: 2266. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers15082266

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop