scholarly journals Cellular and humoral immunogenicity of the mRNA-1273 SARS-CoV-2 vaccine in patients with hematologic malignancies

Author(s):  
Moraima Jiménez ◽  
Elisa Roldan ◽  
Candela Fernández- Naval ◽  
Guillermo Villacampa ◽  
Monica Martinez-Gallo ◽  
...  

Recent studies have demonstrated a suboptimal humoral response to SARS-CoV-2 mRNA vaccines in patients diagnosed with hematologic malignancies, however data about cellular immunogenicity is scarce. In this study we aimed to evaluate both the humoral and cellular immunogenicity one month after the second dose of the mRNA-1273 vaccine. Antibody titers were measured by the Elecsys and LIAISON Anti-SARS-CoV-2 S assay while T-cell response was assessed by Interferon-Gamma-Release-immuno-Assay technology. Overall, 76.3% (184/241) of patients developed humoral immunity and the cellular response rate was 79% (184/233). Hypogammaglobulinemia, lymphopenia, active hematological treatment and anti-CD20 therapy during the last 6 months were associated with an inferior humoral response. Conversely, age over 65 years, active disease, lymphopenia and immunosuppressive treatment for GvHD were associated with an impaired cellular response. A significant dissociation between humoral and cellular response was observed in patients treated with anti-CD20 therapy, being the humoral response of 17.5% whereas the cellular response was 71.1%. In these patients B-cell aplasia was confirmed while T cell counts were preserved. In contrast, humoral response was observed in 77.3% of patients under immunosuppressive treatment for GvHD, while only 52.4% had cellular response. The cellular and humoral response to the SARS-CoV-2 mRNA-1273 vaccine in patients with hematological malignancies is highly influenced by the presence of treatments like anti-CD20 therapy and immunosuppressive agents. This observation has implications for the further management of these patients.

2022 ◽  
Author(s):  
Paolo Corradini ◽  
Chiara Agrati ◽  
Giovanni Apolone ◽  
Alberto Mantovani ◽  
Diana Giannarelli ◽  
...  

Background: Patients with solid or hematological tumors, neurological and immune-inflammatory disorders represent potentially fragile subjects with increased risk to experience severe COVID-19 and inadequate response to SARS-CoV2 vaccination. Methods: We designed a prospective Italian multicentric study to assess humoral and T-cell response to SARS-CoV2 vaccination in patients (n=378) with solid tumors (ST), hematological malignancies (HM), neurological (ND) and immuno-rheumatological diseases (ID). The immunogenicity of primary vaccination schedule and of the booster dose were analyzed. Results: Overall, patient seroconversion rate after two doses was 62.1%. A significant lower rate was observed in HM (52.4%) and ID (51.9%) patients compared to ST (95.6%) and ND (70.7%); a lower median level of antibodies was detected in HM and ID versus the others (p<0.0001). A similar rate of patients with a positive SARS-CoV2 T-cell response was observed in all disease groups, with a higher level observed in the ND group. The booster dose improved humoral responses in all disease groups, although with a lower response in HM patients, while the T-cell response increased similarly in all groups. In the multivariable logistic model, the independent predictors for seroconversion were disease subgroups, type of therapies and age. Notably, the ongoing treatment known to affect the immune system was associated with the worst humoral response to vaccination (p<0.0001), but had no effects on the T-cell responses. Conclusions: Immunosuppressive treatment more than disease type per se is a risk factor for low humoral response after vaccination. The booster dose can improve both humoral and T-cell response.


Blood ◽  
2009 ◽  
Vol 113 (16) ◽  
pp. 3809-3812 ◽  
Author(s):  
Shannon P. Hilchey ◽  
Ollivier Hyrien ◽  
Tim R. Mosmann ◽  
Alexandra M. Livingstone ◽  
Jonathan W. Friedberg ◽  
...  

Abstract The incorporation of rituximab, a chimeric anti-CD20 monoclonal antibody, into the therapeutic armamentarium for patients with follicular lymphoma (FL) has significantly improved treatment outcome for such patients. Despite the almost universal application of this therapy, however, its exact mechanism of action has not been completely defined. One proposed mechanism is that of a “vaccinal” effect, whereby FL cell kill by rituximab results in the elicitation of an FL-specific T-cell response. The demonstration that rituximab can even elicit such a response in patients has, to our knowledge, never been shown. We analyzed the response against the immunoglobulin expressed by the FL before and after rituximab monotherapy in 5 FL patients and found an increase in FL idiotype–specific T cells after rituximab in 4 of 5 patients. Our data thus provide “proof of principle” for the ability of passive immunotherapy with rituximab to elicit an active FL-specific cellular response.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 3724-3724
Author(s):  
Clare Sun ◽  
Erika M Gaglione ◽  
Lauren T Vaughn ◽  
Rui Mu ◽  
Chingiz Underbayev ◽  
...  

Abstract Humoral and cellular adaptive immunity likely contribute to protection against coronavirus disease 2019 (COVID-19). Neutralizing antibodies and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-specific T cells have been detected in convalescent and immunized immunocompetent individuals. Chronic lymphocytic leukemia (CLL) and its treatment, particularly anti-CD20 monoclonal antibodies and Bruton tyrosine kinase inhibitors (BTKis), blunt the antibody response to vaccines. To understand the immunogenicity of COVID-19 vaccination in patients with CLL, assessment of the T-cell response is urgently needed. Between December 22, 2020 and May 7, 2021, 57 patients with CLL were immunized with either 2 doses of BNT162b2 (n = 30) or mRNA-1273 (n = 25) or 1 dose of Ad26.COV2.S (n = 2). Qualitative and semi-quantitative anti-spike antibodies were measured with serology tests authorized by the FDA under Emergency Use Authorization. A positive humoral response to vaccination was defined as the detection of anti-spike antibodies. Ultra-deep TCRß sequencing (Adaptive Biotechnologies) was performed on total peripheral blood mononuclear cells collected before and after vaccination. These data were analyzed in the immunoSEQ Analyzer. Differential abundance was calculated using the beta-binomial model and two-sided α=.05. SARS-CoV-2 spike-specific T cells were identified in the T-MAP COVID ImmuneCODE database. A positive cellular response to vaccination was defined as significant expansion of ≥1 spike-specific clonotype. Anti-spike antibodies were detected in 61% (35/57) of patients at a median (interquartile range, IQR) of 45 (30-56) days after the last dose of vaccine. The median (IQR) antibody titer was 19.1 U/mL (3.6-150.9) among 27 patients with humoral response. There were 4 patients with titers above the upper limit of quantification (&gt;250 U/mL) and 4 patients who had qualitative testing only. The rate of humoral response was 71% (15/21) in treatment naïve (TN) patients, 57% (16/28) in patients treated with BTKi, and 0% (0/4) in patients treated with venetoclax and anti-CD20 monoclonal antibody (mAb). Among 16 BTKi-treated patients with anti-spike antibodies, 2 interrupted BTKi during the vaccination period. One patient treated with venetoclax monotherapy and 3 previously treated patients had detectable anti-spike antibodies. The immediate prior therapies were acalabrutinib &gt;1 year before vaccination for 2 patients and chemoimmunotherapy &gt;8 years before vaccination for 1 patient. Vaccination with mRNA-1273 induced numerically higher titers compared to BNT162b2 (median 85.5 U/mL versus 11.0 U/mL; P=.1), but the rate of seroconversion was not significantly different (P=.4). No patients reported a history of SARS-CoV-2 infection and anti-nucleocapsid antibodies were negative in 100% (50/50) of patients tested. Circulating CD8 + T cells increased from a median (IQR) of 13.2% (7.8-18.8) at baseline to 14.3% (8.8-20.6) after vaccination (P=.015). CD3 + and CD4 + T cells did not significantly change. TCRß sequencing results are available in 7 patients (Table). The median (IQR) number of productive templates, which corresponds to the number of T cells sequenced in each sample, was 447,805 (377,738-503,097). Cellular response was observed in 57% (4/7) of patients. A total of 10 expanded spike-specific clonotypes were identified and ranged between 1 and 6 clonotypes per patient. The cumulative frequency of spike-specific clonotypes after vaccination ranged between 0.0036% and 1.55% per patient. None of these clonotypes were found at baseline despite the large number of productive templates generated in each sample. Spike-specific T cells were detected in 50% (2/4) of patients with anti-spike antibodies and 67% (2/3) of patients without seroconversion. In conclusion, patients with CLL have impaired humoral and cellular responses to COVID-19 vaccination. Seroconversion occurred less often in patients treated with BTKi than TN patients and was absent in patients treated with venetoclax and anti-CD20 mAb. Cellular responses were seen in the absence of humoral responses. TCRß sequencing is ongoing in additional patients. Updated data will be presented at the meeting. Figure 1 Figure 1. Disclosures Sun: Genmab: Research Funding. Wiestner: Merck: Research Funding; Nurix: Research Funding; Genmab: Research Funding; Verastem: Research Funding; Acerta Pharma: Research Funding; Pharmacyclics: Research Funding.


Blood ◽  
2020 ◽  
Vol 136 (20) ◽  
pp. 2290-2295 ◽  
Author(s):  
Thomas Hueso ◽  
Cécile Pouderoux ◽  
Hélène Péré ◽  
Anne-Lise Beaumont ◽  
Laure-Anne Raillon ◽  
...  

Abstract Anti-CD20 monoclonal antibodies are widely used for the treatment of hematological malignancies or autoimmune disease but may be responsible for a secondary humoral deficiency. In the context of COVID-19 infection, this may prevent the elicitation of a specific SARS-CoV-2 antibody response. We report a series of 17 consecutive patients with profound B-cell lymphopenia and prolonged COVID-19 symptoms, negative immunoglobulin G (IgG)-IgM SARS-CoV-2 serology, and positive RNAemia measured by digital polymerase chain reaction who were treated with 4 units of COVID-19 convalescent plasma. Within 48 hours of transfusion, all but 1 patient experienced an improvement of clinical symptoms. The inflammatory syndrome abated within a week. Only 1 patient who needed mechanical ventilation for severe COVID-19 disease died of bacterial pneumonia. SARS-CoV-2 RNAemia decreased to below the sensitivity threshold in all 9 evaluated patients. In 3 patients, virus-specific T-cell responses were analyzed using T-cell enzyme-linked immunospot assay before convalescent plasma transfusion. All showed a maintained SARS-CoV-2 T-cell response and poor cross-response to other coronaviruses. No adverse event was reported. Convalescent plasma with anti–SARS-CoV-2 antibodies appears to be a very promising approach in the context of protracted COVID-19 symptoms in patients unable to mount a specific humoral response to SARS-CoV-2.


2021 ◽  
Author(s):  
Daniel Re ◽  
Barbara Seitz-Polski ◽  
Michel Carles ◽  
Vesna Brglez ◽  
Daisy Graça ◽  
...  

Abstract BACKGROUND: Immunocompromised patients such as patients with hematological malignancies have impaired immune response to two doses of BNT162b2 (Pfizer / BioNtech) vaccine against SARS-CoV-2. Evaluation of a repeated immune stimulation with a third vaccine dose is needed.METHODS: a vaccine monitoring observatory was conducted in outpatients who were treated for lymphoid malignancies (LM) to monitor both immune and cellular response measured the day of administration of the dose 3 of the mRNA vaccine BNT162b2 and again three to four weeks. Elecsys ® Anti-SARS-CoV-2 immunoassay was used to asses to the level of SARS-CoV-2 anti-Spike (S) antibodies (Abs) titer and SARS-CoV-2-specific T-cell responses were assessed by a whole blood Interferon-Gamma Release Immuno Assay (IGRA) (QuantiFERON Human IFN-gamma SARS-CoV-2, Qiagen®).RESULTS: Among the 43 assessable patients (suffering from chronic lymphocytic leukemia (CLL) (n=15), indolent and aggressive B cell non-Hodgkin lymphoma (NHL) (n=14), and multiple myeloma (MM) (n=16)), 18 (41,8%) had no anti-S Abs before the dose 3 of BNT162b2 vaccine (n=9 CLL, n=8 NHL, n=1 MM), and they all 18 remained negative after the dose 3. Amongst the 25 patients with positive anti-S titers before dose 3, all patients remained positive and 23 patients increased their anti-S titer after dose 3. Patients with CLL and/or with previous anti-CD20 therapy treated within 12 months of administration of dose 3 had no significant increase of the humoral response. Among 22 available patients, dose 3 of BNT162b2 vaccine significantly increased the median IFN-gamma secretion. On eight (36.4%) patients who were double-negative for both immune and cellular response, five (22.7%) patients remained double-negative after dose 3.CONCLUSIONSDose 3 of BNT162b2 vaccine stimulated humoral immune response among patients with LM, in particular patients with MM (who had higher anti-S baseline titer after dose 2) and those with no anti-CD20 treatment history within a year. T-cell response was increased among patients in particular with no active chemotherapy regimen. Our data support the use of an early third vaccine dose among immunocompromised patients followed for LM though some of them will still have vaccine failure.


Vaccines ◽  
2021 ◽  
Vol 9 (12) ◽  
pp. 1431
Author(s):  
Josien Lanfermeijer ◽  
Marieke M. Nühn ◽  
Maarten E. Emmelot ◽  
Martien C. M. Poelen ◽  
Cécile A. C. M. van Els ◽  
...  

Waning of the mumps virus (MuV)-specific humoral response after vaccination has been suggested as a cause for recent mumps outbreaks in vaccinated young adults, although it cannot explain all cases. Moreover, CD8+ T cells may play an important role in the response against MuV; however, little is known about the characteristics and dynamics of the MuV-specific CD8+ T-cell response after MuV infection. Here, we had the opportunity to follow the CD8+ T-cell response to three recently identified HLA-A2*02:01-restricted MuV-specific epitopes from 1.5 to 36 months post-MuV infection in five previously vaccinated and three unvaccinated individuals. The infection-induced CD8+ T-cell response was dominated by T cells specific for the ALDQTDIRV and LLDSSTTRV epitopes, while the response to the GLMEGQIVSV epitope was subdominant. MuV-specific CD8+ T-cell frequencies in the blood declined between 1.5 and 9 months after infection. This decline was not explained by changes in the expression of inhibitory receptors or homing markers. Despite the ongoing changes in the frequencies and phenotype of MuV-specific CD8+ T cells, TCRβ analyses revealed a stable MuV-specific T-cell repertoire over time. These insights in the maintenance of the cellular response against mumps may provide hallmarks for optimizing vaccination strategies towards a long-term cellular memory response.


2019 ◽  
Vol 17 (5) ◽  
pp. 350-359
Author(s):  
Liliana Acevedo-Saenz ◽  
Federico Perdomo-Celis ◽  
Carlos J. Montoya ◽  
Paula A. Velilla

Background: : The diversity of the HIV proteome influences the cellular response and development of an effective vaccine, particularly due to the generation of viral variants with mutations located within CD8+ T-cell epitopes. These mutations can affect the recognition of the epitopes, that may result in the selection of HIV variants with mutated epitopes (autologous epitopes) and different CD8+ T-cell functional profiles. Objective:: To determine the phenotype and functionality of CD8+ T-cell from HIV-infected Colombian patients in response to autologous and consensus peptides derived from HIV-1 clade B protease and reverse transcriptase (RT). Methods:: By flow cytometry, we compared the ex vivo CD8+ T-cell responses from HIV-infected patients to autologous and consensus peptides derived from HIV-1 clade B protease and RT, restricted by HLA-B*35, HLA-B*44 and HLA-B*51 alleles. Results:: Although autologous peptides restricted by HLA-B*35 and HLA-B*44 did not show any differences compared with consensus peptides, we observed the induction of a higher polyfunctional profile of CD8+ T-cells by autologous peptides restricted by HLA-B*51, particularly by the production of interferon-γ and macrophage inflammatory protein-1β. The response by different memory CD8+ T-cell populations was comparable between autologous vs. consensus peptides. In addition, the magnitude of the polyfunctional response induced by the HLA-B*51-restricted QRPLVTIRI autologous epitope correlated with low viremia. Conclusion:: Autologous peptides should be considered for the evaluation of HIV-specific CD8+ Tcell responses and to reveal some relevant epitopes that could be useful for therapeutic strategies aiming to promote polyfunctional CD8+ T-cell responses in a specific population of HIV-infected patients.


Vaccines ◽  
2021 ◽  
Vol 9 (6) ◽  
pp. 606
Author(s):  
Giuseppe Cappellano ◽  
Hugo Abreu ◽  
Chiara Casale ◽  
Umberto Dianzani ◽  
Annalisa Chiocchetti

The first vaccines ever made were based on live-attenuated or inactivated pathogens, either whole cells or fragments. Although these vaccines required the co-administration of antigens with adjuvants to induce a strong humoral response, they could only elicit a poor CD8+ T-cell response. In contrast, next-generation nano/microparticle-based vaccines offer several advantages over traditional ones because they can induce a more potent CD8+ T-cell response and, at the same time, are ideal carriers for proteins, adjuvants, and nucleic acids. The fact that these nanocarriers can be loaded with molecules able to modulate the immune response by inducing different effector functions and regulatory activities makes them ideal tools for inverse vaccination, whose goal is to shut down the immune response in autoimmune diseases. Poly (lactic-co-glycolic acid) (PLGA) and liposomes are biocompatible materials approved by the Food and Drug Administration (FDA) for clinical use and are, therefore, suitable for nanoparticle-based vaccines. Recently, another candidate platform for innovative vaccines based on extracellular vesicles (EVs) has been shown to efficiently co-deliver antigens and adjuvants. This review will discuss the potential use of PLGA-NPs, liposomes, and EVs as carriers of peptides, adjuvants, mRNA, and DNA for the development of next-generation vaccines against endemic and emerging viruses in light of the recent COVID-19 pandemic.


2007 ◽  
Vol 81 (10) ◽  
pp. 4928-4940 ◽  
Author(s):  
Maya F. Kotturi ◽  
Bjoern Peters ◽  
Fernando Buendia-Laysa ◽  
John Sidney ◽  
Carla Oseroff ◽  
...  

ABSTRACT CD8+ T-cell responses control lymphocytic choriomeningitis virus (LCMV) infection in H-2b mice. Although antigen-specific responses against LCMV infection are well studied, we found that a significant fraction of the CD8+ CD44hi T-cell response to LCMV in H-2b mice was not accounted for by known epitopes. We screened peptides predicted to bind major histocompatibility complex class I and overlapping 15-mer peptides spanning the complete LCMV proteome for gamma interferon (IFN-γ) induction from CD8+ T cells derived from LCMV-infected H-2b mice. We identified 19 novel epitopes. Together with the 9 previously known, these epitopes account for the total CD8+ CD44hi response. Thus, bystander T-cell activation does not contribute appreciably to the CD8+ CD44hi pool. Strikingly, 15 of the 19 new epitopes were derived from the viral L polymerase, which, until now, was not recognized as a target of the cellular response induced by LCMV infection. The L epitopes induced significant levels of in vivo cytotoxicity and conferred protection against LCMV challenge. Interestingly, protection from viral challenge was best correlated with the cytolytic potential of CD8+ T cells, whereas IFN-γ production and peptide avidity appear to play a lesser role. Taken together, these findings illustrate that the LCMV-specific CD8+ T-cell response is more complex than previously appreciated.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2402-2402
Author(s):  
Yunfeng Cheng ◽  
Yong Tang ◽  
Spencer Green ◽  
Keyvan Keyvanfar ◽  
Tullia Bruno ◽  
...  

Abstract Aplastic anemia is a bone-marrow-failure syndrome characterized by low blood-cell counts and a fatty bone marrow. In most cases, no obvious etiological factor can be identified, but clinical responses to immunosuppressive treatment (IST) strongly suggest an immune pathophysiology. Our previous study of T-cell receptor (TCR) Vβ (variable region of β-chain) repertoire usage by flow cytometry suggested that aplastic anemia results from antigen-specific lymphocyte attack on hematopoietic progenitors (Risitano et al. Lancet2004; 364:355). In the current work, 7 patients were investigated for Vβ pattern expression before first immunosuppresive treatment, at the remission, and again on relapse. The TCR Vβ repertoire was analyzed for CD4+ and CD8+ subsets, separately, by flow cytometry, using a monoclonal antibody set of 22 different Vβ chains. Most patients had very different patterns of Vβ usage from healthy individuals, and all but one showed expansion of at least one Vβ family before immunosuppressive treatment (Vβ family expansions were defined as 2 standard deviations (SD) from the means in controls). The median number of expanded Vβ families was 4 per patient among CD8CD28dim effector cells. At remission, almost all the initially expanded Vβ subfamilies decreased to less than 2SD of controls. At relapse, most of the expanded Vβ subsets were increased again. However, 5/7 patients showed new expanded Vβ subsets at recurrence of cytopenias, suggesting antigenic spread of new epitopes recognized by immune systems. Although no common pattern of specific expanded Vβ subsets could be identified among different patients, some Vβ subfamilies appeared to be more frequently involved (Vβ 5.1 and Vβ 5.2 were expanded in 4 of 7 patients both at initial presentation and relapse ). These data suggest that monitoring Vβ subsets in aplastic anemia, and potentially in other immune-mediated human diseases of a similar pathophysiology could be used to guide individual therapeutic decisions and in the development of new treatments.


Sign in / Sign up

Export Citation Format

Share Document