ABSTRACT
Aim: and objectives: The aim was to contribute to the editorial principles on the possible use of Artificial Intelligence (AI)- based tools for scientific writing. The objectives included: A. Enlist the inclusion and exclusion criteria to test ChatGPT use in scientific writing B. Develop evaluation criteria to assess the quality of articles written by human authors and ChatGPT C. Compare prospectively written manuscripts by human authors and ChatGPT Design: Prospective exploratory study Intervention: Human authors and ChatGPT were asked to write short journal articles on three topics: 1) Promotion of early childhood development in Pakistan 2) Interventions to improve gender-responsive health services in low-and-middle-income countries, and 3) The pitfalls in risk communication for COVID-19. We content analyzed the articles using an evaluation matrix. Outcome measures: The completeness, credibility, and scientific content of an article. Completeness meant that structure (IMRaD) and organization was maintained. Credibility required that others work is duly cited, with an accurate bibliography. Scientific content required specificity, data accuracy, cohesion, inclusivity, confidentiality, limitations, readability, and time efficiency. Results: The articles by human authors scored better than ChatGPT in completeness and credibility. Similarly, human-written articles scored better for most of the items in scientific content except for time efficiency where ChatGPT scored better. The methods section was absent in ChatGPT articles, and a majority of references in its bibliography were unverifiable. Conclusions: ChatGPT generates content that is believable but may not be true. The creators of this powerful model must step up and provide solutions to manage its glitches and potential misuse. In parallel, the academic departments, editors, and publishers must expect a growing utilization of ChatGPT and similar tools. Disallowing ChatGPT as a co-author may not be enough on their part. They must adapt the editorial policies, use measures to detect AI-based writing, and stop its likely implications for human health and life.
Subject(s)
COVID-19 , Lymphoma, B-Cell , Epilepsy, ReflexABSTRACT
ACE2, a member of the angiotensin converting enzyme family, plays an irreplaceable role in the renin-angiotensin system. And the variations of ACE2 are regarded as the key factor to human diseases such as the novel coronavirus pneumonia, cardiovascular disease, and tumors. Here, we summarized the mutation, expression, modification and function of the human ACE2 based on comprehensive bioinformatics analysis. Especially, the relationship between ACE2 expression and diseases, especially tumor was further discussed. ACE2 is highly conserved in different genera and families. We explored the correlation between ACE2 and disease based on the datasets of GCBI and GEO (Gene expression omnibus), and found the expression of ACE2 is related to heart failure. High prevalence of ACE2 mutations is observed in diffuse large B-cell lymphoma, uterine carcinosarcoma (UCS), and stomach adenocarcinoma (STAD). We first identified that highly expressed of ACE2 was linked to poor prognosis of overall survival for tumors of brain lower grade glioma (LGG). Specially, the expression level of ACE2 in kidney-related tumor tissues is much higher than that of normal kidney tissues. ACE2 is negatively correlated with the infiltration level of cancer-associated fibroblasts in most kinds of cancers, such as uterine corpus endometrial carcinoma (UCEC), esophageal carcinoma (ESCA), ovarian serous cystadenocarcinoma (OV) and kidney renal clear cell carcinoma (KIRC); positively correlation in testicular germ cell tumors (TGCT). The different phosphorylation sites of ACE2 were analyzed in CPTAC dataset, and the DNA methylation of ACE2 in colon adenocarcinoma (COAD), kidney renal papillary cell carcinoma (KIRP), and rectum adenocarcinoma (READ) was lower than that of normal control by using SMART database. Moreover, we summarized the interaction proteins and targeted miRNAs of ACE2 through bioinformatics. Then we found the endocrine process and the regulation of systemic arterial blood pressure were involved in the functional mechanisms of ACE2 by using KEGG and GO analysis. Our study offers a relatively comprehensive understanding of ACE2.
Subject(s)
Neoplasms , Ovarian Diseases , Lymphoma, B-Cell , Stomach Neoplasms , Rectal Neoplasms , Esophagitis , Glioma , Carcinoma, Renal Cell , Cardiovascular Diseases , Carcinosarcoma , Heart Failure , Coronavirus Infections , Colorectal Neoplasms , Endometrial NeoplasmsABSTRACT
CAR T-cell recipients experience profound B-cell aplasia and hypogammaglobulinemia, being unable to mount any humoral response and at higher risk for severe COVID-19. Tixagevimab/cilgavimab has been approved for COVID-19 pre-exposure prophylaxis (PrEP) in immunocompromised people. 150/150 mg of tixagevimab/cilgavimab does not adequately neutralize against Omicron BA.5 and these results support recommendations on dose increase to 300/300 mg for prophylaxis in order to enhance effectiveness probability, until the European regulatory agency makes a decision on the usability of this compound as the FDA has already done
Subject(s)
COVID-19 , Lymphoma, B-Cell , AgammaglobulinemiaABSTRACT
Targeted therapy is a powerful treatment option in chronic lymphocytic leukemia (CLL) that has outperformed conventional chemoimmunotherapy in most clinical settings. Except for selected young, fit patients with a mutated immunoglobulin heavy chain variable region gene, most patients benefit from targeted therapy with either a continuous BTK inhibitor or 1-year fixed-duration venetoclax-obinutuzumab as first-line treatment of CLL. Treatment selection is driven by patient-, treatment-, and disease-related factors, encompassing patient preference, concomitant medications, comorbidities, safety profile of the regimen, and TP53 aberration. Clinical trials are actively investigating the simultaneous inhibition of Bruton's tyrosine kinase (BTK) and B-cell lymphoma 2 (BCL-2) proteins with or without a CD20 monoclonal antibody, which can achieve deep response in most patients (52%-89% undetectable minimal residual disease in bone marrow).
Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell , Lymphoma, B-Cell , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Agammaglobulinaemia Tyrosine Kinase , Immunotherapy , Lymphoma, B-Cell/drug therapyABSTRACT
Individuals with hematologic malignancies are at increased risk for severe coronavirus disease 2019 (COVID-19), yet profound analyses of COVID-19 vaccine-induced immunity are scarce. Here we present an observational study with expanded methodological analysis of a longitudinal, primarily BNT162b2 mRNA-vaccinated cohort of 60 infection-naive individuals with B cell lymphomas and multiple myeloma. We show that many of these individuals, despite markedly lower anti-spike IgG titers, rapidly develop potent infection neutralization capacities against several severe acute respiratory syndrome coronavirus 2 variants of concern (VoCs). The observed increased neutralization capacity per anti-spike antibody unit was paralleled by an early step increase in antibody avidity between the second and third vaccination. All individuals with hematologic malignancies, including those depleted of B cells and individuals with multiple myeloma, exhibited a robust T cell response to peptides derived from the spike protein of VoCs Delta and Omicron (BA.1). Consistently, breakthrough infections were mainly of mild to moderate severity. We conclude that COVID-19 vaccination can induce broad antiviral immunity including ultrapotent neutralizing antibodies with high avidity in different hematologic malignancies.
Subject(s)
COVID-19 , Hematologic Neoplasms , Lymphoma, B-Cell , Multiple Myeloma , Humans , COVID-19 Vaccines , BNT162 Vaccine , COVID-19/prevention & control , SARS-CoV-2 , T-Lymphocytes , Antibodies, Neutralizing , VaccinationABSTRACT
How infection by a viral variant showing antigenic drift impacts a preformed mature human memory B cell (MBC) repertoire remains an open question. Here, we studied the MBC response up to 6 months after Omicron BA.1 breakthrough infection in individuals previously vaccinated with three doses of mRNA vaccine. Longitudinal analysis, using single-cell multi-omics and functional analysis of monoclonal antibodies from RBD-specific MBCs, revealed that a BA.1 breakthrough infection mostly recruited pre-existing cross-reactive MBCs with limited de novo response against BA.1-restricted epitopes. Reorganization of clonal hierarchy and new rounds of germinal center reaction, however, combined to maintain diversity and induce progressive maturation of the MBC repertoire against common Hu-1 and BA.1, but not BA.5-restricted, SARS-CoV-2 Spike RBD epitopes. Such remodeling was further associated with marked improvement in overall neutralizing breadth and potency. These findings have fundamental implications for the design of future vaccination booster strategies.
Subject(s)
Lymphoma, B-Cell , Severe Acute Respiratory Syndrome , Breakthrough PainABSTRACT
In patients with common variable immune deficiencies, primary vaccination followed by two booster doses is recommended for protection against COVID-19. Seroconversion has been shown in 60% of patients. We have no information on whether serum antibodies reflect the generation of durable immune memory. In a longitudinal study on 47 common variable immune deficiencies patients who received the third and fourth vaccine dose, we show that the measurement of specific antibodies is not sufficient to predict the establishment of immune memory and the ability to respond to antigen re-exposure. Our results indicate that the combination of antibodies and memory B cells responses represents a more reliable read-out of vaccine immune efficacy in vulnerable patients. This analysis may not only identify individuals remaining unprotected after vaccination and unable to respond to additional booster doses, but also address the search for the underlying immune defect and suggest patient-tailored management strategies.
Subject(s)
COVID-19 , Immunologic Deficiency Syndromes , Lymphoma, B-CellABSTRACT
People living with HIV (PLWH) on suppressive antiretroviral therapy (ART) can have residual immune dysfunction and often display poorer responses to vaccination. We assessed in a cohort of PLWH (n=110) and HIV negative controls (n=64) the humoral and spike-specific B-cell responses following 1, 2 or 3 SARS-CoV-2 vaccine doses. PLWH had significantly lower neutralizing antibody (nAb) titers than HIV-negative controls at all studied timepoints. Moreover, their neutralization breadth was reduced with fewer individuals developing a neutralizing response against the Omicron variant (BA.1) relative to controls. We also observed a delayed development of neutralization in PLWH that was underpinned by a reduced frequency of spike-specific memory B cells (MBCs) and pronounced B cell dysfunction. Improved neutralization breadth was seen after the third vaccine dose in PLWH but lower nAb responses persisted and were associated with global, but not spike-specific, MBC dysfunction. In contrast to the inferior antibody responses, SARS-CoV-2 vaccination induced robust T cell responses that cross-recognized variants in PLWH. Strikingly, a subset of PLWH with low or absent neutralization had detectable functional T cell responses. These individuals had reduced numbers of circulating T follicular helper cells and an enriched population of CXCR3+CD127+CD8+ T cells after two doses of SARS-CoV-2 vaccination, which may compensate for sub-optimal serological responses in the event of infection. Therefore, normalisation of B cell homeostasis could improve serological responses to vaccines in PLWH and evaluating T cell immunity could provide a more comprehensive immune status profile in these individuals and others with B cell imbalances.
Subject(s)
HIV Infections , Lymphoma, B-Cell , Sexual Dysfunction, PhysiologicalABSTRACT
Oral drugs such as ibrutinib play an important role in the treatment of mature B-cell lymphoma (BCL) due to their reliable efficacy, manageable safety, high accessibility, and convenience for use. Still, no guidelines or consensus focusing on oral drug therapies for BCL is available. To provide a reference of oral agent-based treatment for mature BCL, a panel of experts from the Lymphocyte Disease Group, Chinese Society of Hematology, Chinese Medical Association conducted an extensive discussion and reached a consensus on oral drugs for Chinese BCL patients on the basis of the current application status of oral drugs in China, combined with the latest authoritative guidelines in the world and current research reports. This consensus reviewed the application of oral drugs in the treatment of BCL and the latest research and provided appropriate recommendations on the use of oral drugs for indolent or aggressive BCL patients. With the deepening of research and the development of standardized clinical applications, oral medications will bring better treatment to BCL patients, enabling more patients to benefit from them.
Subject(s)
Lymphoma, B-Cell , Humans , Consensus , Lymphoma, B-Cell/drug therapy , ChinaABSTRACT
B cell responses to different pathogens recruit tailored effector mechanisms, resulting in functionally specialized subsets. For human memory B cells (MBCs), these include CD21+ resting, CD21-CD27+ activated, and CD21-CD27- atypical cells. Whether these subsets follow deterministic or interconnected fates is unknown. We demonstrate in COVID-19 patients that single clones of SARS-CoV-2-specific MBCs followed multiple fates with distinctive phenotypic and functional characteristics. 6-12 months after infection, most circulating MBCs were CD21+ resting cells, which also accumulated in peripheral lymphoid organs where they acquired markers of tissue residency. Conversely, at acute infection and following SARS-CoV-2-specific immunization, CD21- MBCs became the predominant subsets, with atypical MBCs expressing high T-bet, inhibitory molecules, and distinct chemokine receptors. B cell receptor sequencing allowed tracking of individual MBC clones differentiating into CD21+, CD21-CD27+, and CD21-CD27- cell fates. Collectively, single MBC clones can adopt functionally different trajectories, thus contributing to immunity to infection.
Subject(s)
COVID-19 , Lymphoma, B-CellABSTRACT
Memory B cells (MBCs) generate rapid antibody responses upon secondary encounter with a pathogen. Here, we investigated the kinetics, avidity and cross-reactivity of serum antibodies and MBCs in 155 SARS-CoV-2 infected and vaccinated individuals over a 16-month timeframe. SARS-CoV-2-specific MBCs and serum antibodies reached steady-state titers with comparable kinetics in infected and vaccinated individuals. Whereas MBCs of infected individuals targeted both pre- and postfusion Spike (S), most vaccine-elicited MBCs were specific for prefusion S, consistent with the use of prefusion-stabilized S in mRNA vaccines. Furthermore, a large fraction of MBCs recognizing postfusion S cross-reacted with human betacoronaviruses. The avidity of MBC-derived and serum antibodies increased over time resulting in enhanced resilience to viral escape by SARS-CoV-2 variants, including Omicron BA.1 and BA.2 sub-lineages, albeit only partially for BA.4 and BA.5 sublineages. Overall, the maturation of high-affinity and broadly-reactive MBCs provides the basis for effective recall responses to future SARS-CoV-2 variants.
Subject(s)
Lymphoma, B-Cell , Infections , Severe Acute Respiratory SyndromeABSTRACT
Understanding the evolution of antibody immunity following heterologous SAR-CoV-2 breakthrough infection will inform the development of next-generation vaccines. Here, we tracked SARS-CoV-2 receptor binding domain (RBD)-specific antibody responses up to six months following Omicron BA.1 breakthrough infection in mRNA-vaccinated individuals. Cross-reactive serum neutralizing antibody and memory B cell (MBC) responses declined by two- to four-fold through the study period. Breakthrough infection elicited minimal de novo Omicron-specific B cell responses but drove affinity maturation of pre-existing cross-reactive MBCs toward BA.1. Public clones dominated the neutralizing antibody response at both early and late time points, and their escape mutation profiles predicted newly emergent Omicron sublineages. The results demonstrate that heterologous SARS-CoV-2 variant exposure drives the evolution of B cell memory and suggest that convergent neutralizing antibody responses continue to shape viral evolution.
Subject(s)
Lymphoma, B-Cell , Breakthrough PainABSTRACT
The primary two-dose SARS-CoV-2 mRNA vaccine series are strongly immunogenic in humans, but the emergence of highly infectious variants necessitated additional doses of these vaccines and the development of new variant-derived ones. SARS-CoV-2 booster immunizations in humans primarily recruit pre-existing memory B cells (MBCs). It remains unclear, however, whether the additional doses induce germinal centre (GC) reactions where reengaged B cells can further mature and whether variant-derived vaccines can elicit responses to novel epitopes specific to such variants. Here, we show that boosting with the original SARS-CoV-2 spike vaccine (mRNA-1273) or a B.1.351/B.1.617.2 (Beta/Delta) bivalent vaccine (mRNA-1273.213) induces robust spike-specific GC B cell responses in humans. The GC response persisted for at least eight weeks, leading to significantly more mutated antigen-specific MBC and bone marrow plasma cell compartments. Interrogation of MBC-derived spike-binding monoclonal antibodies (mAbs) isolated from individuals boosted with either mRNA-1273, mRNA-1273.213, or a monovalent Omicron BA.1-based vaccine (mRNA-1273.529) revealed a striking imprinting effect by the primary vaccination series, with all mAbs (n=769) recognizing the original SARS-CoV-2 spike protein. Nonetheless, using a more targeted approach, we isolated mAbs that recognized the spike protein of the SARS-CoV-2 Omicron (BA.1) but not the original SARS-CoV-2 spike from the mRNA-1273.529 boosted individuals. The latter mAbs were less mutated and recognized novel epitopes within the spike protein, suggesting a naive B cell origin. Thus, SARS-CoV-2 boosting in humans induce robust GC B cell responses, and immunization with an antigenically distant spike can overcome the antigenic imprinting by the primary vaccination series.
Subject(s)
Lymphoma, B-Cell , Breast NeoplasmsABSTRACT
Mass vaccination schemes have been launched for COVID-19 worldwide. However, recent studies have revealed that SARS-CoV-2 Omicron and its sub-lineages efficiently evade humoral immunity from vaccination or previous infection. Therefore, it is of great importance to investigate the contribution of cellular immunity against infection of emerging variants of SARS-CoV-2 in the context of vaccine-induced immunity. By using C57BL/6J and K18-hACE2 mouse models, we demonstrated that BNT162b2 induces robust protective immunity in B-cell deficient (μMT) mice. We further demonstrated that this protection is attributed to the cellular immunity mediated by robust IFN-γ production. In addition, we revealed that SARS-CoV-2 Omicron BA.1 could also induce strong cellular responses in vaccinated μMT mice upon viral challenge, which highlights the significance of cellular immunity against the ever-emerging SARS-CoV-2 variants that evade antibody-mediated immunity. Overall, our study provides evidence that BNT162b2 can induce significant protective immunity in mice that are unable to produce antibodies.
Subject(s)
COVID-19 , Lymphoma, B-CellABSTRACT
Background: and objective Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), continues to ravage the world. Despite many prevention and control measures, morbidity and mortality have not decreased due to SARS-CoV-2-induced organ damage, which occur via unknown mechanisms. Methods: Primary Human small intestinal mucosa epithelial cells (HSIMECs), human colonic epithelial cells (HCoEpiCs), and human type II alveolar epithelial cells (hTIIAECs) were treated with recombinant SARS-CoV-2 S protein for 48 h. Cell morphology, permeability, and viability were detected. The expression of cysteinyl aspartate specific proteinase 3 (caspase 3) and B-cell lymphoma-2 (Bcl-2) was examined using Western blotting. Enzyme-linked immunosorbent assay (ELISA) was performed to detect the levels of inflammatory cytokines in the supernatant. Apoptosis was observed using a Hoechst 33258 Staining Kit. SB431542 and BAY11-7082 were used to inhibit transforming growth factor-β (TGF-β)/suppressor of mothers against decapentaplegics (Smads) and the inhibitor of kappa B kinase (IKK)/nuclear factor-κB (NF-κB) pathways, respectively. Results: S protein produced no obvious changes in morphology, but decreased cell viability and increased permeability were observed in a concentration- and time-dependent manner compared to the control ( P <0.05). Apoptosis occurred with increased caspase 3 and decreased Bcl-2 ( P <0.05). S protein stimulated a disordered secretion of cytokines, including interleukin (IL)-6, IL-10, tumour necrosis factor α (TNF-α), and IL-13 ( P <0.05). Suppression of TGF-β/Smad3, but not the IKK/NF-κB, pathway relieved the damage to colon cells caused by the S protein in HSIMECs and HCoEpiCs and inhibited apoptosis mediated by TNF-α and granulocyte-macrophage colony-stimulating factor (GM-CSF) in hTIIAECs. Conclusion: The SARS-CoV-2 S protein damaged intestine and lung cells, which was associated with cytokine production and the induction of apoptosis mediated by the TGF-β/Smad3, but not the NF-κB, pathway.
Subject(s)
COVID-19 , Neoplasms , Lymphoma, B-Cell , Coronavirus InfectionsABSTRACT
Background The use of variant-based severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccine as a booster is being evaluated to overcome reduced neutralisation of variants induced by the original SARS-CoV-2 vaccine and waning protection over time. Methods This is a phase one, prospective, randomized, and open-labeled trial to study the safety and immunogenicity of a booster dose consisting of a subunit vaccine based on the stabilized prefusion SARS-CoV-2 spike protein, MVC-COV1901 or its Beta version, MVC-COV1901-Beta. One-hundred and seven participants aged [≥]18 and <55 years, who received two or three prior doses of MVC-COV1901 vaccines, were enrolled and were to receive a booster dose of either 15 mcg of MVC-COV1901, 15 mcg or 25 mcg of MVC-COV1901-Beta in 1:1:1 ratio. The primary endpoints were the incidences of adverse events and immunogenicity of the booster dose from Visit 2 (the day of the booster) to Visit 5 (four weeks after the booster). Cellular immunity was also investigated with memory B cell (MBC) and T cell assays. Findings Adverse reactions after either MVC-COV1901 or MVC-COV1901-Beta booster doses after two or three doses of MVC-COV1901 were comparable and mostly mild and transient. At four weeks after the booster dose, participants with two prior doses of MVC-COV1901 exhibited numerically higher levels of neutralising antibodies against SARS-CoV-2 or Beta variant than participants with three prior doses of MVC-COV1901 regardless of the type of booster used. However, compared to 15 mcg of MVC-COV1901, 25 mcg of MVC-COV1901-Beta significantly improved neutralising antibody titre against Beta variant and BA.4/BA.5 Omicron variant pseudoviruses. The booster dose also significantly increased the proportion of spike-specific MBCs, including those of Beta and Omicron variants. Interpretation MVC-COV1901-Beta can be effectively used as a booster dose against SARS-CoV-2, including the circulating BA.4/BA.5 Omicron variant. Funding Medigen Vaccine Biologics Corporation
Subject(s)
Coronavirus Infections , Lymphoma, B-CellABSTRACT
Feedback inhibition of humoral immunity by antibodies was initially documented in guinea pigs by Theobald Smith in 1909, who showed that passive administration of excess anti-Diphtheria toxin inhibited immune responses1. Subsequent work documented that antibodies can enhance or inhibit immune responses depending on antibody isotype, affinity, the physical nature of the antigen, and engagement of immunoglobulin (Fc) and complement (C) receptors2, 3. However, little is known about how pre-existing antibodies might influence the subsequent development of memory B cells. Here we examined the memory B cell response in individuals who received two high-affinity IgG1 anti-SARS-CoV-2 receptor binding domain (RBD)-specific monoclonal antibodies, C144-LS and C135-LS, and subsequently two doses of a SARS-CoV-2 mRNA vaccine. The two antibodies target Class 2 and 3 epitopes that dominate the initial immune response to SARS-CoV-2 infection and mRNA vaccination4-8. Antibody responses to the vaccine in C144-LS and C135-LS recipients produced plasma antigen binding and neutralizing titers that were fractionally lower but not statistically different to controls. In contrast, memory B cells enumerated by flow cytometry after the second vaccine dose were present in higher numbers than in controls. However, the memory B cells that developed in antibody recipients differed from controls in that they were not enriched in VH3-53, VH1-46 and VH3-66 genes and predominantly expressed low-affinity IgM antibodies that carried small numbers of somatic mutations. These antibodies showed altered RBD target specificity consistent with epitope masking, and only 1 out of 77 anti-RBD memory antibodies tested neutralized the virus. The results indicate that pre-existing high-affinity antibodies bias memory B cell selection and have a profound effect on the development of immunological memory in humans that may in part explain the shifting target profile of memory antibodies elicited by the 3rd mRNA vaccine dose.
Subject(s)
COVID-19 , Lymphoma, B-Cell , Immune System Diseases , Masked HypertensionABSTRACT
T-cell dysregulation in chronic lymphocytic leukemia (CLL) associates with low response rates to autologous T cell-based therapies. How CLL affects antigen-specific T-cell responses remains largely unknown. We investigated (epi)genetic and functional consequences of antigen-specific T-cell responses in presence of CLL in vitro and in an adoptive-transfer murine model. Already at steady-state, antigen-experienced patient-derived T cells were skewed towards short-lived effector cells (SLEC) at the expense of memory-precursor effector cells (MPEC). Stimulation of these T cells in vitro showed rapid induction of effector genes and suppression of key memory transcription factors only in presence of CLL cells, indicating epigenetic regulation. This was investigated in vivo by following antigen-specific responses of naïve OT-I CD8 + cells to mCMV-OVA in presence/absence of TCL1 B-cell leukemia. Presence of leukemia resulted in increased SLEC formation, with disturbed inflammatory cytokine production. Chromatin and transcriptome profiling revealed strong epigenetic modifications, leading to activation of an effector and silencing of a memory profile through presence of CLL cells. Secondary challenge in vivo confirmed dysfunctional memory responses by antigen-experienced OT-I cells generated in presence of CLL. Altogether, we show that presence of CLL induces a short-lived effector phenotype and impaired memory responses by epigenetic reprogramming during primary responses.