Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 990
Filter
1.
HLA ; 104(1): e15612, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39041339

ABSTRACT

The novel HLA-A*02:1144 allele differs from HLA-A*02:03:01:01 by 3 nucleotides in exon 7.


Subject(s)
Exons , HLA-A2 Antigen , Humans , Alleles , Blood Donors , East Asian People/genetics , HLA-A2 Antigen/genetics , Sequence Analysis, DNA
2.
Front Immunol ; 15: 1415102, 2024.
Article in English | MEDLINE | ID: mdl-39007132

ABSTRACT

Human regulatory T cells (Treg) suppress other immune cells. Their dysfunction contributes to the pathophysiology of autoimmune diseases, including type 1 diabetes (T1D). Infusion of Tregs is being clinically evaluated as a novel way to prevent or treat T1D. Genetic modification of Tregs, most notably through the introduction of a chimeric antigen receptor (CAR) targeting Tregs to pancreatic islets, may improve their efficacy. We evaluated CAR targeting of human Tregs to monocytes, a human ß cell line and human islet ß cells in vitro. Targeting of HLA-A2-CAR (A2-CAR) bulk Tregs to HLA-A2+ cells resulted in dichotomous cytotoxic killing of human monocytes and islet ß cells. In exploring subsets and mechanisms that may explain this pattern, we found that CD39 expression segregated CAR Treg cytotoxicity. CAR Tregs from individuals with more CD39low/- Tregs and from individuals with genetic polymorphism associated with lower CD39 expression (rs10748643) had more cytotoxicity. Isolated CD39- CAR Tregs had elevated granzyme B expression and cytotoxicity compared to the CD39+ CAR Treg subset. Genetic overexpression of CD39 in CD39low CAR Tregs reduced their cytotoxicity. Importantly, ß cells upregulated protein surface expression of PD-L1 and PD-L2 in response to A2-CAR Tregs. Blockade of PD-L1/PD-L2 increased ß cell death in A2-CAR Treg co-cultures suggesting that the PD-1/PD-L1 pathway is important in protecting islet ß cells in the setting of CAR immunotherapy. In summary, introduction of CAR can enhance biological differences in subsets of Tregs. CD39+ Tregs represent a safer choice for CAR Treg therapies targeting tissues for tolerance induction.


Subject(s)
Apyrase , Receptors, Chimeric Antigen , T-Lymphocytes, Regulatory , Humans , Apyrase/immunology , Apyrase/metabolism , T-Lymphocytes, Regulatory/immunology , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , Cytotoxicity, Immunologic , Islets of Langerhans/immunology , Islets of Langerhans/metabolism , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/therapy , HLA-A2 Antigen/immunology , HLA-A2 Antigen/genetics , HLA-A2 Antigen/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/metabolism , Antigens, CD
3.
J Clin Invest ; 134(11)2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38828721

ABSTRACT

The adoptive transfer of T cell receptor-engineered (TCR-engineered) T cells (ACT) targeting the HLA-A2-restricted cancer-testis epitope NY-ESO-1157-165 (A2/NY) has yielded favorable clinical responses against several cancers. Two approaches to improve ACT are TCR affinity optimization and T cell coengineering to express immunomodulatory molecules that can exploit endogenous immunity. By computational design we previously developed a panel of binding-enhanced A2/NY-TCRs including A97L, which augmented the in vitro function of gene-modified T cells as compared with WT. Here, we demonstrated higher persistence and improved tumor control by A97L-T cells. In order to harness macrophages in tumors, we further coengineered A97L-T cells to secrete a high-affinity signal regulatory protein α (SiRPα) decoy (CV1) that blocks CD47. While CV1-Fc-coengineered A97L-T cells mediated significantly better control of tumor outgrowth and survival in Winn assays, in subcutaneous xenograft models the T cells, coated by CV1-Fc, were depleted. Importantly, there was no phagocytosis of CV1 monomer-coengineered T cells by human macrophages. Moreover, avelumab and cetuximab enhanced macrophage-mediated phagocytosis of tumor cells in vitro in the presence of CV1 and improved tumor control upon coadministration with A97L-T cells. Taken together, our study indicates important clinical promise for harnessing macrophages by combining CV1-coengineered TCR-T cells with targeted antibodies to direct phagocytosis against tumor cells.


Subject(s)
Macrophages , Phagocytosis , Receptors, Immunologic , Animals , Humans , Mice , Antigens, Differentiation/immunology , Antigens, Neoplasm/immunology , CD47 Antigen/immunology , Cell Line, Tumor , HLA-A2 Antigen/immunology , HLA-A2 Antigen/genetics , Immunotherapy, Adoptive , Macrophages/immunology , Macrophages/metabolism , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism , Receptors, Immunologic/genetics , T-Lymphocytes/immunology , Xenograft Model Antitumor Assays , Male , Female
5.
Cancer Genomics Proteomics ; 21(4): 414-420, 2024.
Article in English | MEDLINE | ID: mdl-38944423

ABSTRACT

BACKGROUND/AIM: Clear cell carcinoma is a prevalent histological type of ovarian cancer in East Asia, particularly in Japan, known for its resistance to chemotherapeutic agents and poor prognosis. ARID1A gene mutations, commonly found in ovarian clear cell carcinoma (OCCC), contribute to its pathogenesis. Recent data revealed that the ARID1A mutation is related to better outcomes of cancer immunotherapy. Thus, this study aimed to investigate the immunotherapy treatment susceptibility of OCCC bearing ARID1A mutations. MATERIALS AND METHODS: Expression of ARID1A was analyzed using western blotting in ovarian cancer cell lines. OCCC cell lines JHOC-9 and RMG-V were engineered to overexpress NY-ESO-1, HLA-A*02:01, and ARID1A. Sensitivity to chemotherapy and T cell receptor-transduced T (TCR-T) cells specific for NY-ESO-1 was assessed in ARID1A-restored cells compared to ARID1A-deficient wild-type cells. RESULTS: JHOC-9 cells and RMG-V cells showed no expression of ARID1A protein. Overexpression of ARID1A in JHOC-9 and RMG-V cells did not impact sensitivity to gemcitabine. While ARID1A overexpression decreased sensitivity to cisplatin in RMG-V cells, it had no such effect in JHOC-9 cells. ARID1A overexpression reduced the reactivity of NY-ESO-1-specific TCR-T cells, as observed by the IFNγ ESLIPOT assay. CONCLUSION: Cancer immunotherapy is an effective approach to target ARID1A-deficient clear cell carcinoma of the ovary.


Subject(s)
Adenocarcinoma, Clear Cell , DNA-Binding Proteins , Ovarian Neoplasms , T-Lymphocytes, Cytotoxic , Transcription Factors , Humans , Female , Ovarian Neoplasms/immunology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Ovarian Neoplasms/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Adenocarcinoma, Clear Cell/pathology , Adenocarcinoma, Clear Cell/genetics , Adenocarcinoma, Clear Cell/immunology , Adenocarcinoma, Clear Cell/metabolism , T-Lymphocytes, Cytotoxic/immunology , Cell Line, Tumor , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Antigens, Neoplasm/immunology , HLA-A2 Antigen/genetics , HLA-A2 Antigen/immunology , HLA-A2 Antigen/metabolism , Membrane Proteins
6.
HLA ; 103(5): e15505, 2024 May.
Article in English | MEDLINE | ID: mdl-38797560

ABSTRACT

The novel HLA-A*02:829 allele is likely generated from the recombination of both HLA-A*02:05:01:01 and HLA-A*32:01:01:01 alleles.


Subject(s)
Alleles , Base Sequence , Exons , HLA-A2 Antigen , Histocompatibility Testing , Sequence Analysis, DNA , Humans , Sequence Analysis, DNA/methods , HLA-A2 Antigen/genetics , HLA-A2 Antigen/immunology , Sequence Alignment , Recombination, Genetic , Codon
7.
Viruses ; 16(5)2024 04 25.
Article in English | MEDLINE | ID: mdl-38793551

ABSTRACT

Epstein-Barr Virus (EBV) is closely linked to nasopharyngeal carcinoma (NPC), notably prevalent in southern China. Although type II latency of EBV plays a crucial role in the development of NPC, some lytic genes and intermittent reactivation are also critical for viral propagation and tumor progression. Since T cell-mediated immunity is effective in targeted killing of EBV-positive cells, it is important to identify EBV-derived peptides presented by highly prevalent human leukocyte antigen class I (HLA-I) molecules throughout the EBV life cycle. Here, we constructed an EBV-positive NPC cell model to evaluate the presentation of EBV lytic phase peptides on streptavidin-tagged specific HLA-I molecules. Utilizing a mass spectrometry (LC-MS/MS)-based immunopeptidomic approach, we characterized eleven novel EBV peptides as well as two previously identified peptides. Furthermore, we determined these peptides were immunogenic and could stimulate PBMCs from EBV VCA/NA-IgA positive donors in an NPC endemic southern Chinese population. Overall, this work demonstrates that highly prevalent HLA-I-specific EBV peptides can be captured and functionally presented to elicit immune responses in an in vitro model, which provides insight into the epitopes presented during EBV lytic cycle and reactivation. It expands the range of viral targets for potential NPC early diagnosis and treatment.


Subject(s)
Epstein-Barr Virus Infections , HLA-A2 Antigen , Herpesvirus 4, Human , Nasopharyngeal Carcinoma , Peptides , Humans , Herpesvirus 4, Human/immunology , Herpesvirus 4, Human/genetics , Peptides/immunology , Peptides/chemistry , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/virology , HLA-A2 Antigen/immunology , HLA-A2 Antigen/genetics , Nasopharyngeal Carcinoma/immunology , Nasopharyngeal Carcinoma/virology , HLA-A11 Antigen/immunology , HLA-A11 Antigen/genetics , Proteomics/methods , Nasopharyngeal Neoplasms/immunology , Nasopharyngeal Neoplasms/virology , China , Tandem Mass Spectrometry , Epitopes, T-Lymphocyte/immunology , Cell Line, Tumor
9.
Acta Biochim Biophys Sin (Shanghai) ; 56(7): 986-996, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38655616

ABSTRACT

Vaccines play essential roles in the fight against the COVID-19 pandemic. The development and assessment of COVID-19 vaccines have generally focused on the induction and boosting of neutralizing antibodies targeting the SARS-CoV-2 spike (S) protein. Due to rapid and continuous variation in the S protein, such vaccines need to be regularly updated to match newly emerged dominant variants. T-cell vaccines that target MHC I- or II-restricted epitopes in both structural and non-structural viral proteins have the potential to induce broadly cross-protective and long-lasting responses. In this work, the entire proteome encoded by SARS-CoV-2 (Wuhan-hu-1) is subjected to immunoinformatics-based prediction of HLA-A*02:01-restricted epitopes. The immunogenicity of the predicted epitopes is evaluated using peripheral blood mononuclear cells from convalescent Wuhan-hu-1-infected patients. Furthermore, predicted epitopes that are conserved across major SARS-CoV-2 lineages and variants are used to construct DNA vaccines expressing multi-epitope polypeptides. Most importantly, two DNA vaccine constructs induce epitope-specific CD8 + T-cell responses in a mouse model of HLA-A*02:01 restriction and protect immunized mice from challenge with Wuhan-hu-1 virus after hACE2 transduction. These data provide candidate T-cell epitopes useful for the development of T-cell vaccines against SARS-CoV-2 and demonstrate a strategy for quick T-cell vaccine candidate development applicable to other emerging pathogens.


Subject(s)
COVID-19 Vaccines , COVID-19 , Computational Biology , Epitopes, T-Lymphocyte , HLA-A2 Antigen , SARS-CoV-2 , Vaccines, DNA , Epitopes, T-Lymphocyte/immunology , Humans , SARS-CoV-2/immunology , SARS-CoV-2/genetics , Animals , Vaccines, DNA/immunology , Vaccines, DNA/genetics , HLA-A2 Antigen/immunology , HLA-A2 Antigen/genetics , Mice , COVID-19/immunology , COVID-19/prevention & control , COVID-19/virology , COVID-19 Vaccines/immunology , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Female , Mice, Inbred BALB C , Immunoinformatics
10.
Int J Mol Sci ; 25(3)2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38338657

ABSTRACT

Sensitization to HLA can result in allograft loss for kidney transplantation (KT) patients. Therefore, it is required to develop an appropriate desensitization (DSZ) technique to remove HLA-donor-specific anti-HLA antibody (DSA) before KT. The aim of this research was to investigate whether combined use of the IL-6 receptor-blocking antibody, tocilizumab (TCZ), and bone-marrow-derived mesenchymal stem cells (BM-MSCs) could attenuate humoral immune responses in an allo-sensitized mouse model developed using HLA.A2 transgenic mice. Wild-type C57BL/6 mice were sensitized with skin allografts from C57BL/6-Tg (HLA-A2.1)1Enge/J mice and treated with TCZ, BM-MSC, or both TCZ and BM-MSC. We compared HLA.A2-specific IgG levels and subsets of T cells and B cells using flow cytometry among groups. HLA.A2-specific IgG level was decreased in all treated groups in comparison with that in the allo-sensitized control (Allo-CONT) group. Its decrease was the most significant in the TCZ + BM-MSC group. Regarding the B cell subset, combined use of TCZ and BM-MSC increased proportions of pre-pro B cells but decreased proportions of mature B cells in BM (p < 0.05 vs. control). In the spleen, an increase in transitional memory was observed with a significant decrease in marginal, follicular, and long-lived plasma B cells (p < 0.05 vs. control) in the TCZ + BM-MSC group. In T cell subsets, Th2 and Th17 cells were significantly decreased, but Treg cells were significantly increased in the TCZ+BM-MSC group compared to those in the Allo-CONT group in the spleen. Regarding RNA levels, IL-10 and Foxp3 showed increased expression, whereas IL-23 and IFN-γ showed decreased expression in the TCZ + BM-MSC group. In conclusion, combined use of TCZ and BM-MSC can inhibit B cell maturation and up-regulate Treg cells, finally resulting in the reduction of HLA.A2-specific IgG in a highly sensitized mouse model. This study suggests that the combined use of TCZ and BM-MSC can be proposed as a novel strategy in a desensitization protocol for highly sensitized patients.


Subject(s)
Antibodies, Monoclonal, Humanized , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Humans , Mice , Animals , Mice, Inbred C57BL , B-Lymphocytes , Mice, Transgenic , HLA-A2 Antigen/genetics , HLA Antigens/metabolism , Immunoglobulin G/metabolism , Mesenchymal Stem Cells/metabolism
11.
J Neurol Neurosurg Psychiatry ; 95(6): 554-560, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38212058

ABSTRACT

BACKGROUND: Large register-based studies have reported an association between head trauma and increased risk of multiple sclerosis (MS). We aimed to investigate possible interactions between head trauma and MS-associated HLA genes in relation to MS risk. METHODS: We used a Swedish population-based case-control study (2807 incident cases, 5950 matched controls with HLA genotypes available for 2057 cases, 2887 controls). Subjects with and without a history of self-reported head trauma were compared regarding MS risk, by calculating ORs with 95% CIs using logistic regression models. Additive interaction between head trauma, HLA-DRB1*1501 and absence of HLA-A*0201, was assessed by calculating the attributable proportion (AP) due to interaction. RESULTS: A history of head trauma was associated with a 30% increased risk of subsequently developing MS (OR 1.34, 95% CI 1.17 to 1.53), with a trend showing increased risk of MS with increasing number of head impacts (p=0.03). We observed synergistic effects between recent head trauma and HLA-DRB1*15:01 as well as absence of HLA*02:01 in relation to MS risk (each AP 0.40, 95% CI 0.1 to 0.7). Recent head trauma in individuals with both genetic risk factors rendered an 18-fold increased risk of MS, compared with those with neither the genetic risk factors nor a history of head trauma (OR 17.7, 95% CI 7.13 to 44.1). CONCLUSIONS: Our findings align with previous observations of a dose-dependent association between head trauma and increased risk of MS and add a novel aspect of this association by revealing synergistic effects between recent head trauma and MS-associated HLA genes.


Subject(s)
Craniocerebral Trauma , Genetic Predisposition to Disease , HLA-DRB1 Chains , Multiple Sclerosis , Humans , Multiple Sclerosis/genetics , Multiple Sclerosis/epidemiology , Female , Male , Case-Control Studies , HLA-DRB1 Chains/genetics , Genetic Predisposition to Disease/genetics , Craniocerebral Trauma/epidemiology , Adult , Sweden/epidemiology , Middle Aged , Genotype , Risk Factors , HLA-A2 Antigen/genetics , Young Adult , Aged
12.
HLA ; 102(4): 449-463, 2023 10.
Article in English | MEDLINE | ID: mdl-37503860

ABSTRACT

The presence of donor-specific antibodies (DSA), mainly against HLA, increases the risk of allograft rejection. Moreover, antibody-mediated rejection (ABMR) remains an important barrier to optimal long-term outcomes after solid organ transplantation. The development of chimeric autoantibody receptor T lymphocytes has been postulated for targeted therapy of autoimmune diseases. We aimed to develop a targeted therapy for DSA desensitization and ABMR, generating T cells with a chimeric HLA antibody receptor (CHAR) that specifically eliminates DSA-producing B cells. We have genetically engineered an HLA-A2-specific CHAR (A2-CHAR) and transduced it into human T cells. Then, we have performed in vitro experiments such as cytokine measurement, effector cell activation, and cytotoxicity against anti-HLA-A2 antibody-expressing target cells. In addition, we have performed A2-CHAR-Tc cytotoxic assays in an immunodeficient mouse model. A2-CHAR expressing T cells could selectively eliminate HLA-A2 antibody-producing B cells in vitro. The cytotoxic capacity of A2-CHAR expressing T cells mainly depended on Granzyme B release. In the NSG mouse model, A2-CHAR-T cells could identify and eradicate HLA-A2 antibody-producing B cells even when those cells are localized in the bone marrow. This ability is effector:target ratio dependent. CHAR technology generates potent and functional human cytotoxic T cells to target alloreactive HLA class I antibody-producing B cells. Thus, we consider that CHAR technology may be used as a selective desensitization protocol or an ABMR therapy in transplantation.


Subject(s)
Graft Rejection , HLA Antigens , Mice , Animals , Humans , HLA Antigens/genetics , Alleles , Antibodies , HLA-A2 Antigen/genetics , Receptors, Antigen, T-Cell , Isoantibodies
13.
Microbes Infect ; 25(8): 105182, 2023.
Article in English | MEDLINE | ID: mdl-37423326

ABSTRACT

CD8+ T cells from HLA-A2.1-transgenic mice, but not wild-type mice, immunized with the amino-terminus region (aa 41-152) of dense granule protein 6 (GRA6Nt) of Toxoplasma gondii secreted large amounts of perforin and granzyme B in response to GRA6Nt through antigen presentation by HLA-A2.1 in vitro. When those CD8+ T cells were transferred into chronically infected HLA-A2.1-expressing NSG mice deficient in T cells, cerebral cyst burden of the recipients of HLA-A2.1-transgenic T cells, but not of WT T cells, became significantly less than that of control mice with no cell transfer. Furthermore, the significant reduction of the cyst burden by a transfer of the HLA-A2.1-transgenic CD8+ immune T cells required an expression of HLA-A2.1 in the recipient NSG mice. Thus, antigen presentation of GRA6Nt by human HLA-A2.1is able to activate anti-cyst CD8+ T cells that eliminate T. gondii cysts through antigen presentation by human HLA-A2.1.


Subject(s)
Parasites , Toxoplasma , Humans , Mice , Animals , Toxoplasma/genetics , CD8-Positive T-Lymphocytes , T-Lymphocytes, Cytotoxic , HLA-A2 Antigen/genetics , HLA-A2 Antigen/metabolism , Antigen Presentation , Immunization , Mice, Transgenic
14.
Front Immunol ; 14: 1114770, 2023.
Article in English | MEDLINE | ID: mdl-37215108

ABSTRACT

Background: The overall 5-year survival rate of hepatocellular carcinoma (HCC), a major form of liver cancer, is merely 20%, underscoring the need for more effective therapies. We recently identified T cell receptors (TCR) specific for the HLA-A2/alpha fetoprotein amino acids 158-166 (AFP158) and showed that these TCR engineered T cells could control HCC xenografts in NSG mice. However, their efficacy was limited by poor expansion, loss of function, and short persistence of the TCR T cells. Here, we studied whether overexpression of c-Jun, a transcription factor required for T cell activation, in the TCR T cells could enhance their expansion, function, and persistence in HCC tumor models. Methods: Recombinant lentiviral vectors (lv), expressing either the HLA-A2/AFP158-specific TCR or both the TCR and c-Jun (TCR-JUN), were constructed and used to transduce primary human T cells to generate the TCR or TCR-JUN T cells, respectively. We compared the expansion, effector function, and exhaustion status of the TCR and TCR-JUN T cells in vitro after HCC tumor stimulation. Additionally, we studied the persistence and antitumor effects of the TCR and TCR-JUN T cells using the HCC xenografts in NSG mice. Results: We could effectively transduce primary human T cells to express both TCR and c-Jun. Compared to the HLA-A2/AFP158 TCR T cells, the TCR-JUN T cells have better expansion potential in culture, with enhanced functional capacity against HCC tumor cells. In addition, the TCR-JUN T cells were less apoptotic and more resistant to exhaustion after HepG2 tumor stimulation. In the HCC xenograft tumor model, c-Jun overexpression enhanced the TCR T cell expansion and increased the overall survival rate of the treated mice. Importantly, the TCR-JUN T cells were less exhausted in the tumor lesions and demonstrated enhanced tumor infiltration, functionality, and persistence. Conclusion: c-Jun overexpression can enhance the expansion, function, and persistence of the A2/AFP158 TCR engineered T cells. The c-Jun gene co-delivery has the potential to enhance the antitumor efficacy of AFP specific TCR T cells when treating patients with HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Mice , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/therapy , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/therapy , Liver Neoplasms/metabolism , alpha-Fetoproteins/genetics , HLA-A2 Antigen/genetics , HLA-A2 Antigen/metabolism , Genes, jun , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes
17.
Eur J Immunol ; 53(5): e2250054, 2023 05.
Article in English | MEDLINE | ID: mdl-36794567

ABSTRACT

High-frequency mutation of the TP53 tumor suppressor gene is observed in multiple human cancers, which promotes cancer progression. However, the mutated gene-encoded protein may serve as a tumor antigen to elicit tumor-specific immune responses. In this study, we detected widespread expression of shared TP53-Y220C neoantigen in hepatocellular carcinoma with low affinity and low stability of binding to HLA-A0201 molecules. We substituted the amino acid sequences VVPCEPPEV with VLPCEPPEV in the TP53-Y220C neoantigen to yield a TP53-Y220C (L2) neoantigen. This altered neoantigen was found to increase affinity and stability and induce more cytotoxic T lymphocytes (CTLs), indicating improvements in immunogenicity. In vitro assays showed the cytotoxicity of CTLs stimulated by both TP53-Y220C and TP53-Y220C (L2) neoantigens against multiple HLA-A0201-positive cancer cells expressing TP53-Y220C neoantigens; however, the TP53-Y220C (L2) neoantigen showed higher cytotoxicity than the TP53-Y220C neoantigen against cancer cells. More importantly, in vivo assays demonstrated greater inhibition of hepatocellular carcinoma cell proliferation by TP53-Y220C (L2) neoantigen-specific CTLs relative to TP53-Y220C neoantigen in zebrafish and nonobese diabetic/severe combined immune deficiency mouse models. The results of this study demonstrate enhanced immunogenicity of the shared TP53-Y220C (L2) neoantigen, which has the potential as dendritic cells or peptide vaccines for multiple cancers.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Mice , Humans , T-Lymphocytes, Cytotoxic , HLA-A2 Antigen/genetics , Epitopes , Zebrafish , Antigens, Neoplasm , Cytotoxicity, Immunologic , Tumor Suppressor Protein p53/genetics
18.
Nat Commun ; 13(1): 7189, 2022 11 23.
Article in English | MEDLINE | ID: mdl-36424374

ABSTRACT

MHC restriction, which describes the binding of TCRs from CD4+ T cells to class II MHC proteins and TCRs from CD8+ T cells to class I MHC proteins, is a hallmark of immunology. Seemingly rare TCRs that break this paradigm exist, but mechanistic insight into their behavior is lacking. TIL1383I is a prototypical class-mismatched TCR, cloned from a CD4+ T cell but recognizing the tyrosinase tumor antigen presented by the class I MHC HLA-A2 in a fully functional manner. Here we find that TIL1383I binds this class I target with a highly atypical geometry. Despite unorthodox binding, TCR signaling, antigen specificity, and the ability to use CD8 are maintained. Structurally, a key feature of TIL1383I is an exceptionally long CDR3ß loop that mediates functions that are traditionally performed separately by hypervariable and germline loops in canonical TCR structures. Our findings thus expand the range of known TCR binding geometries compatible with normal function and specificity, provide insight into the determinants of MHC restriction, and may help guide TCR selection and engineering for immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes , Receptors, Antigen, T-Cell , Cell Membrane , Engineering , HLA-A2 Antigen/genetics
19.
J Biomed Sci ; 29(1): 80, 2022 Oct 12.
Article in English | MEDLINE | ID: mdl-36224625

ABSTRACT

BACKGROUND: Human Papillomavirus type 18 (HPV18) is a high-risk HPV that is commonly associated with cervical cancer. HPV18 oncogenes E6 and E7 are associated with the malignant transformation of cells, thus the identification of human leukocyte antigen (HLA)-restricted E6/E7 peptide-specific CD8 + T cell epitopes and the creation of a HPV18 E6/E7 expressing cervicovaginal tumor in HLA-A2 transgenic mice will be significant for vaccine development. METHODS: In the below study, we characterized various human HLA class I-restricted HPV18 E6 and E7-specific CD8 + T cells mediated immune responses in HLA class I transgenic mice using DNA vaccines encoding HPV18E6 and HPV18E7. We then confirmed HLA-restricted E6/E7 specific CD8 + T cell epitopes using splenocytes from vaccinated mice stimulated with HPV18E6/E7 peptides. Furthermore, we used oncogenic DNA plasmids encoding HPV18E7E6(delD70), luciferase, cMyc, and AKT to create a spontaneous cervicovaginal carcinoma model in HLA-A2 transgenic mice. RESULTS: Therapeutic HPV18 E7 DNA vaccination did not elicit any significant CD8 + T cell response in HLA-A1, HLA-24, HLA-B7, HLA-B44 transgenic or wild type C57BL/6 mice, but it did generate a strong HLA-A2 and HLA-A11 restricted HPV18E7-specific CD8 + T cell immune response. We found that a single deletion of aspartic acid (D) at location 70 in HPV18E6 DNA abolishes the presentation of HPV18 E6 peptide (aa67-75) by murine MHC class I. We found that the DNA vaccine with this mutant HPV18 E6 generated E6-specific CD8 + T cells in HLA-A2. HLA-A11, HLA-A24 and HLA-b40 transgenic mice. Of note, HLA-A2 restricted, HPV18 E7 peptide (aa7-15)- and HPV18 E6 peptide (aa97-105)-specific epitopes are endogenously processed by HPV18 positive Hela-AAD (HLA-A*0201/Dd) cells. Finally, we found that injection of DNA plasmids encoding HPV18E7E6(delD70), AKT, cMyc, and SB100 can result in the development of adenosquamous carcinoma in the cervicovaginal tract of HLA-A2 transgenic mice. CONCLUSIONS: We characterized various human HLA class I-restricted HPV18 E6/E7 peptide specific CD8 + T cell epitopes in human HLA class I transgenic mice. We demonstrated that HPV18 positive Hela cells expressing chimeric HLA-A2 (AAD) do present both HLA-A2-restricted HPV18 E7 (aa7-15)- and HPV18 E6 (aa97-105)-specific CD8 + T cell epitopes. A mutant HPV18E6 that had a single deletion at location 70 obliterates the E6 presentation by murine MHC class I and remains oncogenic. The identification of these human MHC restricted HPV antigen specific epitopes as well as the HPV18E6/E7 expressing adenosquamous cell carcinoma model may have significant future translational potential.


Subject(s)
Carcinoma, Adenosquamous , Oncogene Proteins, Viral , Papillomavirus Infections , Vaccines, DNA , Animals , Aspartic Acid , CD8-Positive T-Lymphocytes , Carcinoma, Adenosquamous/complications , Epitopes, T-Lymphocyte/genetics , Female , HLA-A Antigens , HLA-A1 Antigen , HLA-A11 Antigen , HLA-A2 Antigen/genetics , HLA-A24 Antigen , HLA-B40 Antigen , HLA-B44 Antigen , HLA-B7 Antigen , HeLa Cells , Human papillomavirus 18 , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oncogene Proteins, Viral/genetics , Papillomavirus Infections/complications , Peptides , Proto-Oncogene Proteins c-akt , T-Lymphocytes, Cytotoxic , Vaccines, DNA/genetics
20.
J Immunother Cancer ; 10(10)2022 Oct.
Article in English | MEDLINE | ID: mdl-36302563

ABSTRACT

Diffuse midline glioma is the leading cause of solid cancer-related deaths in children with very limited treatment options. A majority of the tumors carry a point mutation in the histone 3 variant (H3.3) creating a potential HLA-A*02:01 binding epitope (H3.3K27M26-35). Here, we isolated an H3.3K27M-specific T cell receptor (TCR) from transgenic mice expressing a diverse human TCR repertoire. Despite a high functional avidity of H3.3K27M-specific T cells, we were not able to achieve recognition of cells naturally expressing the H3.3K27M mutation, even when overexpressed as a transgene. Similar results were obtained with T cells expressing the published TCR 1H5 against the same epitope. CRISPR/Cas9 editing was used to exclude interference by endogenous TCRs in donor T cells. Overall, our data provide strong evidence that the H3.3K27M mutation is not a suitable target for cancer immunotherapy, most likely due to insufficient epitope processing and/or amount to be recognized by HLA-A*02:01 restricted CD8+ T cells.


Subject(s)
Glioma , HLA-A2 Antigen , Animals , Humans , Mice , CD8-Positive T-Lymphocytes , Epitopes , Glioma/genetics , Glioma/therapy , Glioma/metabolism , Histones/genetics , Histones/metabolism , HLA-A2 Antigen/genetics , HLA-A2 Antigen/metabolism , Immunotherapy , Mice, Transgenic , Mutation , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL