Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
J Immunother Cancer ; 11(8)2023 08.
Article in English | MEDLINE | ID: mdl-37607770

ABSTRACT

Cisplatin-based chemotherapy has been associated with durable disease control in a small subset of patients with metastatic urothelial cancer. However, the mechanistic basis for this phenomenon has remained elusive. Antitumor immunity may underlie these exceptional responders. In a phase II trial evaluating a phased schedule of gemcitabine and cisplatin followed by gemcitabine and cisplatin with ipilimumab for metastatic urothelial cancer, 4 of 36 patients achieved durable disease-free treatment-free survival (DDFTFS) and remain in remission over 5 years after enrolment on the study. We sought to identify the genomic and immunological mechanisms associated with functional cures of such patients. Whole exome sequencing was performed on pretreatment archival tumor tissue. Neoantigen prediction and ranking were performed using a novel pipeline. For a subset of patients with available biospecimens, selected peptides were tested for neoantigen-specific T cell reactivity in peripheral blood CD4+ and CD8+ T cells cultured with autologous antigen-presenting cells at baseline, postchemotherapy, and postchemotherapy and ipilimumab timepoints. Multiplex assays of serum protein analytes were also assessed at each time point. Serum proteomic analysis revealed that pretreatment, patients achieving DDFTFS demonstrated an immune activated phenotype with elevations in TH1 adaptive immunity, costimulatory molecules, and immune checkpoint markers. After combination cisplatin-based chemotherapy and ipilimumab treatment, DDFTFS patients again displayed enrichment for markers of adaptive immunity, as well as T cell cytotoxicity. CD27 was uniquely enriched in DDFTFS patients at all timepoints. Neoantigen reactivity was not detected in any patient at baseline or post two cycles of chemotherapy. Both CD4+ and CD8+ neoantigen-specific T cell reactivity was detected in two of two DDFTFS patients in comparison to zero of five non-DDFTFS patients after combination cisplatin-based chemotherapy and ipilimumab treatment. Antitumor immunity may underlie functional cures achieved in patients with metastatic urothelial cancer treated with cisplatin-based chemotherapy and immune checkpoint blockade. Probing the mechanistic basis for DDFTFS may facilitate the identification of biomarkers, therapeutic components, and optimal treatment sequences necessary to extend this ultimate goal to a larger subset of patients.


Subject(s)
CD8-Positive T-Lymphocytes , Carcinoma, Transitional Cell , Humans , Cisplatin/therapeutic use , Ipilimumab/therapeutic use , Proteomics , Disease-Free Survival , Carcinoma, Transitional Cell/drug therapy
2.
Front Immunol ; 13: 840457, 2022.
Article in English | MEDLINE | ID: mdl-35273617

ABSTRACT

Costimulation pathways play an essential role in T cell activation, differentiation, and regulation. CD155 expressed on antigen-presenting cells (APCs) interacts with TIGIT, an inhibitory costimulatory molecule, and CD226, an activating costimulatory molecule, on T cells. TIGIT and CD226 are expressed at varying levels depending on the T cell subset and activation state. T follicular helper cells in germinal centers (GC-Tfh) in human tonsils express high TIGIT and low CD226. However, the biological role of the CD155/TIGIT/CD226 axis in human Tfh cell biology has not been elucidated. To address this, we analyzed tonsillar CD4+ T cell subsets cultured with artificial APCs constitutively expressing CD155. Here we show that CD226 signals promote the early phase of Tfh cell differentiation in humans. CD155 signals promoted the proliferation of naïve CD4+ T cells and Tfh precursors (pre-Tfh) isolated from human tonsils and upregulated multiple Tfh molecules and decreased IL-2, a cytokine detrimental for Tfh cell differentiation. Blocking CD226 potently inhibited their proliferation and expression of Tfh markers. By contrast, while CD155 signals promoted the proliferation of tonsillar GC-Tfh cells, their proliferation required only weak CD226 signals. Furthermore, attenuating CD226 signals rather increased the expression of CXCR5, ICOS, and IL-21 by CD155-stimulated GC-Tfh cells. Thus, the importance of CD226 signals changes according to the differentiation stage of human Tfh cells and wanes in mature GC-Tfh cells. High TIGIT expression on GC-Tfh may play a role in attenuating the detrimental CD226 signals post GC-Tfh cell maturation.


Subject(s)
Antigens, Differentiation, T-Lymphocyte , Receptors, Immunologic , T Follicular Helper Cells , Antigens, Differentiation, T-Lymphocyte/metabolism , Cell Differentiation , Humans , Lymphocyte Activation , Receptors, Immunologic/metabolism , T-Lymphocyte Subsets
3.
Cancer Sci ; 111(6): 1958-1968, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32304127

ABSTRACT

Polyfunctionality/multifunctionality of effector T cells at the single cell level has been shown as an important parameter to predict the quality of T cell response and immunological control of infectious disease and malignancy. However, the fate of polyfunctional CD8+ CTLs and the factors that control the polyfunctionality of T cells remain largely unknown. Here we show that the acquisition of polyfunctionality on the initial stimulation is a sensitive immune correlate of CTL survival and memory formation. CD8+ T cells with high polyfunctionality, assessed with γ-interferon and tumor necrosis factor-α production and surface mobilization of the degranulation marker CD107a, showed enhanced Bcl-2 expression, low apoptosis, and increased CD127high KLRG1low memory precursor phenotype. Consistent with these observations, CD8+ T cells were found to acquire high frequency of cells with polyfunctionality when stimulated in conditions known to enhance memory formation, such as the presence of CD4+ T cells, interleukin (IL)-2, or IL-21. Utilizing T-cell receptor (TCR) transgenic mouse-derived CD8+ T cells that express a TCR specific for a tumor-derived neoantigen, we showed that polyfunctional tumor-specific CTLs generated in the presence of CD4+ T cells showed long persistence in vivo and induced enhanced tumor regression when adoptively transferred into mice with progressing tumor. Acquisition of polyfunctionality thus impacts CTL survival and memory formation associated with immunological control of tumor.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Neoplasms/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Mice
4.
Clin Cancer Res ; 26(2): 450-464, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31857430

ABSTRACT

PURPOSE: Somatic mutations in cancer cells can give rise to novel protein sequences that can be presented by antigen-presenting cells as neoantigens to the host immune system. Tumor neoantigens represent excellent targets for immunotherapy, due to their specific expression in cancer tissue. Despite the widespread use of immunomodulatory drugs and immunotherapies that recharge T and NK cells, there has been no direct evidence that neoantigen-specific T-cell responses are elicited in multiple myeloma. EXPERIMENTAL DESIGN: Using next-generation sequencing data we describe the landscape of neo-antigens in 184 patients with multiple myeloma and successfully validate neoantigen-specific T cells in patients with multiple myeloma and support the feasibility of neoantigen-based therapeutic vaccines for use in cancers with intermediate mutational loads such as multiple myeloma. RESULTS: In this study, we demonstrate an increase in neoantigen load in relapsed patients with multiple myeloma as compared with newly diagnosed patients with multiple myeloma. Moreover, we identify shared neoantigens across multiple patients in three multiple myeloma oncogenic driver genes (KRAS, NRAS, and IRF4). Next, we validate neoantigen T-cell response and clonal expansion in correlation with clinical response in relapsed patients with multiple myeloma. This is the first study to experimentally validate the immunogenicity of predicted neoantigens from next-generation sequencing in relapsed patients with multiple myeloma. CONCLUSIONS: Our findings demonstrate that somatic mutations in multiple myeloma can be immunogenic and induce neoantigen-specific T-cell activation that is associated with antitumor activity in vitro and clinical response in vivo. Our results provide the foundation for using neoantigen targeting strategies such as peptide vaccines in future trials for patients with multiple myeloma.


Subject(s)
Antigens, Neoplasm/genetics , Cancer Vaccines/immunology , Multiple Myeloma/genetics , Multiple Myeloma/immunology , Mutation , Peptides/immunology , T-Lymphocytes/immunology , Adult , Aged , Aged, 80 and over , Cancer Vaccines/therapeutic use , Disease-Free Survival , Drug Resistance, Neoplasm , Female , Humans , Immunotherapy/methods , Male , Middle Aged , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/pathology , Survival Rate
5.
Cancer Immunol Res ; 7(4): 658-669, 2019 04.
Article in English | MEDLINE | ID: mdl-30745365

ABSTRACT

Autologous stem cell transplant (autoSCT), the standard consolidation therapy for multiple myeloma, improves disease-free survival, but is not curative. This could be an ideal setting for immunologic therapy. However, the immune milieu is impaired after autoSCT. We hypothesized that autologous lymphocyte infusion would restore immune competence, allowing immunotherapies such as cancer vaccines to elicit tumor antigen-specific immunity in the setting of autoSCT. In this pilot study (NCT01380145), we investigated safety, immunologic, and clinical outcomes of autologous lymphocyte infusion combined with peri-autoSCT immunotherapy with recombinant MAGE-A3 (a multiple myeloma-associated antigen) and adjuvant. Thirteen patients with multiple myeloma undergoing autoSCT were enrolled. Autologous lymphocyte infusion and MAGE vaccination were well tolerated. Combination immunotherapy resulted in high-titer humoral immunity and robust, antigen-specific CD4+ T-cell responses in all subjects, and the responses persisted at least one year post-autoSCT. CD4+ T cells were polyfunctional and Th1-biased. CD8+ T-cell responses were elicited in 3 of 13 subjects. These cells recognized naturally processed MAGE-A3 antigen. Median progression-free survival was 27 months, and median overall survival was not reached, suggesting no differences from standard-of-care. In 4 of 8 subjects tested, MAGE-A protein expression was not detected by IHC in multiple myeloma cells at relapse, suggesting therapy-induced immunologic selection against antigen-expressing clones. These results demonstrated that autologous lymphocyte infusion augmentation of autoSCT confers a favorable milieu for immunotherapies such as tumor vaccines. This strategy does not require ex vivo manipulation of autologous lymphocyte products and is an applicable platform for further investigation into combination immunotherapies to treat multiple myeloma.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines/therapeutic use , Lymphocyte Transfusion , Multiple Myeloma/therapy , Neoplasm Proteins/immunology , Stem Cell Transplantation , Adult , Aged , Female , Humans , Lymphocytes/immunology , Male , Middle Aged , Multiple Myeloma/immunology , Transplantation, Autologous
6.
Appl Immunohistochem Mol Morphol ; 27(8): 581-583, 2019 09.
Article in English | MEDLINE | ID: mdl-29517505

ABSTRACT

T lymphocytes can be distinguished based on the composition of the T-cell receptor (TCR) chain in α/ß T cells and γ/δ T cells. Correspondingly, α/ß lymphomas can be distinguished from γ/δ lymphomas. The latter are rare neoplasms, which are usually confined to particular organs and tissues and carry a dismal prognosis. Until recently, monoclonal antibody (mAb) clone g3.20 to the TCR γ-chain was the reagent of choice for the immunohistochemical detection of γ/δ T cells and lymphomas in standard formalin-fixed paraffin-embedded tissues. However, due to technical problems, mAb g3.20 became recently unavailable. Our attempts to identify another commercially available clone to the TCR γ-chain were unsuccessful. However, we were able to identify a mAb (clone H-41, SC-100289; Santa Cruz, Dallas, TX) to the TCR δ-chain. H-41 works well in immunohistochemistry on paraffin-embedded tissue and comparison with previously stained cases, shows superior immunolabeling to mAb g3.20. H-41 to the TCR δ-chain appears to be a suitable reagent for the replacement of mAb g3.20.


Subject(s)
Antibodies, Monoclonal/immunology , Lymphoma/diagnosis , Receptors, Antigen, T-Cell, gamma-delta/metabolism , T-Lymphocytes/immunology , Formaldehyde , Humans , Immunohistochemistry , Lymphoma/immunology , Paraffin Embedding
7.
Nat Med ; 24(12): 1845-1851, 2018 12.
Article in English | MEDLINE | ID: mdl-30397353

ABSTRACT

Focal radiation therapy enhances systemic responses to anti-CTLA-4 antibodies in preclinical studies and in some patients with melanoma1-3, but its efficacy in inducing systemic responses (abscopal responses) against tumors unresponsive to CTLA-4 blockade remained uncertain. Radiation therapy promotes the activation of anti-tumor T cells, an effect dependent on type I interferon induction in the irradiated tumor4-6. The latter is essential for achieving abscopal responses in murine cancers6. The mechanisms underlying abscopal responses in patients treated with radiation therapy and CTLA-4 blockade remain unclear. Here we report that radiation therapy and CTLA-4 blockade induced systemic anti-tumor T cells in chemo-refractory metastatic non-small-cell lung cancer (NSCLC), where anti-CTLA-4 antibodies had failed to demonstrate significant efficacy alone or in combination with chemotherapy7,8. Objective responses were observed in 18% of enrolled patients, and 31% had disease control. Increased serum interferon-ß after radiation and early dynamic changes of blood T cell clones were the strongest response predictors, confirming preclinical mechanistic data. Functional analysis in one responding patient showed the rapid in vivo expansion of CD8 T cells recognizing a neoantigen encoded in a gene upregulated by radiation, supporting the hypothesis that one explanation for the abscopal response is radiation-induced exposure of immunogenic mutations to the immune system.


Subject(s)
CD8-Positive T-Lymphocytes/radiation effects , CTLA-4 Antigen/antagonists & inhibitors , Ipilimumab/administration & dosage , Lung Neoplasms/therapy , Aged , Aged, 80 and over , Antibodies, Monoclonal/administration & dosage , CD8-Positive T-Lymphocytes/immunology , CTLA-4 Antigen/immunology , Cell Line, Tumor , Combined Modality Therapy , Drug Resistance, Neoplasm/radiation effects , Female , Humans , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung Neoplasms/radiotherapy , Male , Middle Aged , Radiotherapy
8.
Minim Invasive Ther Allied Technol ; 26(6): 331-337, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28417678

ABSTRACT

PURPOSE: To evaluate the changes in T-cell balance in peripheral blood following percutaneous tumor ablation. MATERIAL AND METHODS: Patients underwent thermal ablation including radiofrequency (n = 9) and microwave ablation (n = 5), or cryoablation (n = 5). Target tumors were located in the lung (n = 7), soft tissue (n = 5), liver (n = 4), and bone (n = 3). Patient peripheral blood samples were collected before and within 14 days after ablation. Peripheral blood populations of cytotoxic T-cells (CTL), type-1 (Th1) and type-2 helper T-cells (Th2), and regulatory T-cells (Treg) were measured using flow cytometry. Changes in CTL/Treg and Th1/Th2 ratios before and after ablation therapy were compared using paired t-tests. RESULTS: Peripheral blood CTL population (27.5 ± 2.1% to 30.2 ± 2.5%, p < .03) and CTL/Treg ratios (18.8 ± 3.7% to 21.6 ± 3.6%, p < .05) increased significantly after ablation. Although a significant increase in CTL/Treg ratios was found after heat-based ablation (18.0 ± 4.4% to 21.6 ± 4.7%, p < .02), it remained unchanged after cryoablation (21.0 ± 7.0% to 21.5 ± 4.3%, p = .92). Th1/Th2 ratio (13.7 ± 3.0% to 17.2 ± 3.5%, p = .12) remained unchanged after ablation. CONCLUSION: Ablation therapy alters the T-cell balance by increasing the systemic CTL/Treg, ratio. Heat-based ablation might be a more effective approach than cryoablation to enhance systemic anti-tumor immunity.


Subject(s)
Ablation Techniques , Neoplasms/surgery , T-Lymphocyte Subsets/immunology , Adult , Aged , Aged, 80 and over , Catheter Ablation , Cryosurgery , Female , Humans , Leukocytes/immunology , Male , Microwaves/therapeutic use , Middle Aged , Prospective Studies , Young Adult
9.
Nat Commun ; 8: 14607, 2017 02 24.
Article in English | MEDLINE | ID: mdl-28233863

ABSTRACT

Genetic evolution that occurs during cancer progression enables tumour heterogeneity, thereby fostering tumour adaptation, therapeutic resistance and metastatic potential. Immune responses are known to select (immunoedit) tumour cells displaying immunoevasive properties. Here we address the role of IFN-γ in mediating the immunoediting process. We observe that, in several mouse tumour models such as HA-expressing 4T1 mammary carcinoma cells, OVA-expressing EG7 lymphoma cells and CMS5 MCA-induced fibrosarcoma cells naturally expressing mutated extracellular signal-regulated kinase (ERK) antigen, the action of antigen-specific cytotoxic T cell (CTL) in vivo results in the emergence of resistant cancer cell clones only in the presence of IFN-γ within the tumour microenvironment. Moreover, we show that exposure of tumours to IFN-γ-producing antigen-specific CTLs in vivo results in copy-number alterations (CNAs) associated with DNA damage response and modulation of DNA editing/repair gene expression. These results suggest that enhanced genetic instability might be one of the mechanisms by which CTLs and IFN-γ immunoedits tumours, altering their immune resistance as a result of genetic evolution.


Subject(s)
Antigens, Neoplasm/immunology , Immune Tolerance/genetics , Interferon-gamma/immunology , Neoplasms/immunology , T-Lymphocytes, Cytotoxic/immunology , Tumor Microenvironment/immunology , Animals , Antigens, Neoplasm/genetics , DNA Damage/immunology , DNA Repair/immunology , Disease Progression , Evolution, Molecular , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/immunology , Female , Gene Expression Regulation, Neoplastic/immunology , Genomic Instability/immunology , Humans , Interferon-gamma/genetics , Interferon-gamma/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mutation , Neoplasms/genetics , Neoplasms/pathology , T-Lymphocytes, Cytotoxic/metabolism , Tumor Microenvironment/genetics , Xenograft Model Antitumor Assays
10.
Int J Oncol ; 50(1): 129-140, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27922671

ABSTRACT

Wilms tumor 1 (WT1) is considered to be a promising target of cancer treatment because it has been reported to be frequently expressed at high levels in various malignancies. Although WT1-targeted cancer treatment has been initiated, conclusive detection methods for WT1 are not established. The present study aimed to consolidate immunohistochemistry for WT1 with statistical basis. Transfected cells with forced WT1 expression yielded specific western blot bands and nuclear immunostaining; cytoplasmic immunostaining was not specifically recognized. Immunohistochemistry, western blotting, and quantitative reverse transcriptase-polymerase chain reaction were performed in 35 human cell lines using multiple WT1 antibodies and their results were quantified. Relationships among the quantified results were statistically analyzed; the nuclear immunostaining positively correlated with western blot bands and mRNA expression levels, whereas cytoplasmic immunostaining did not. These results indicate that nuclear immunostaining reflects WT1 expression but cytoplasmic immunostaining does not. The nuclear immunostaining was barely (3/541) observed in primary cancer of esophagus, bile duct, pancreas and lung. Although the present study has some limitations, the results indicate that the cytoplasmic immunostaining does not correlate with actual WT1 expression and prompts researchers to carefully evaluate target molecule expression in treatment of cancer.


Subject(s)
Biomarkers, Tumor/isolation & purification , Neoplasms/genetics , RNA, Messenger/isolation & purification , WT1 Proteins/isolation & purification , Biomarkers, Tumor/biosynthesis , Cell Line, Tumor , Cytoplasm/genetics , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Neoplasms/pathology , WT1 Proteins/biosynthesis
11.
J Vasc Interv Radiol ; 27(10): 1561-8, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27084711

ABSTRACT

PURPOSE: To evaluate changes in T-cell populations in peripheral blood after bland hepatic artery embolization (HAE). MATERIALS AND METHODS: Bland HAE was performed in 12 patients to treat primary (n = 5) or metastatic (n = 7) liver tumors, using microspheres and polyvinyl alcohol (n = 8) or microspheres alone (n = 4). Patient peripheral blood samples were collected within 1 month before HAE, within 1 week after HAE (early period after HAE), and 2-8 weeks after HAE (follow-up period). Peripheral blood populations of cytotoxic T lymphocytes, CD4(+) T cells, type 1 helper T cells (Th1) and type 2 helper T cells (Th2), and regulatory T cells (Treg) were evaluated using flow cytometry. Changes in T-cell populations before and after bland HAE were compared using paired t tests. RESULTS: Peripheral blood CD4(+) T-cell populations decreased significantly in the early period after HAE (44.0% ± 2.2 to 34.4% ± 3.6, P < .01) and in the follow-up period (44.0% ± 2.2 to 36.3% ± 3.0, P < .01). Among the individual CD4(+) T-cell subtypes, Treg (2.5% ± 0.3 to 1.7% ± 0.2, P < .02) and Th1 (8.1% ± 1.8 to 5.6% ± 1.6, P < .02) decreased significantly in the early period after HAE only. The presence of extrahepatic disease was associated with decreasing Treg (P < .04). CONCLUSIONS: After HAE, the peripheral blood T-cell environment is changed with decreases in Treg and Th1.


Subject(s)
Acrylic Resins/administration & dosage , Embolization, Therapeutic/methods , Gelatin/administration & dosage , Hepatic Artery , Liver Neoplasms/therapy , Polyvinyl Alcohol/administration & dosage , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Acrylic Resins/adverse effects , Adult , Aged , Biomarkers, Tumor/blood , CD4 Lymphocyte Count , Embolization, Therapeutic/adverse effects , Female , Flow Cytometry , Gelatin/adverse effects , Hepatic Artery/diagnostic imaging , Humans , Immunophenotyping/methods , Liver Neoplasms/blood , Liver Neoplasms/blood supply , Liver Neoplasms/immunology , Male , Middle Aged , New York City , Phenotype , Polyvinyl Alcohol/adverse effects , Prospective Studies , Time Factors , Treatment Outcome
12.
Oncol Lett ; 12(6): 4493-4504, 2016 Dec.
Article in English | MEDLINE | ID: mdl-28105158

ABSTRACT

A phase I+II clinical trial of vaccination with MAGE-A4 protein complexed with cholesteryl pullulan melanoma antigen gene-A4 nanogel (CHP-MAGE-A4) is currently underway in patients with MAGE-A4-expressing cancer. In the present study, the primary phase I endpoint was to test the safety of the administration of 300 µg CHP-MAGE-A4 with and without OK-432. Another aim of the study was to clarify the details of the specific humoral immune response to vaccination. The 9 patients enrolled for phase I were vaccinated 6 times, once every 2 weeks: 3 patients with 100 µg and 3 patients with 300 µg CHP-MAGE-A4, and 3 patients with 300 µg CHP-MAGE-A4 plus 0.5 clinical units of OK-432. Toxicities were assessed using Common Terminology Criteria for Adverse Events v3.0. Clinical response was evaluated by modified Response Evaluation Criteria in Solid Tumours. Immunological monitoring of anti-MAGE-A4-specific antibodies was performed by ELISA of pre- and post-vaccination patient sera. The 6 vaccinations produced no severe adverse events. Stable disease was assessed in 4/9 patients. Anti-MAGE-A4 total immunoglobulin (Ig)G titers increased in 7/9 patients. Efficacious anti-MAGE-A4 IgG1, 2 and 3 antibody responses were observed in 7/9 patients. Among them, positive conversions to T helper 2 (Th2)-type antibody responses (IgG4 and IgE) were observed after frequent vaccination in 4/7 patients. The Th2 conversion was possibly associated with undesirable clinical observations, including progressive disease and the appearance of a new relapse lesion. The present study suggested that frequent vaccinations activated a Th2-dominant status in the cancer patients. The identification of a time-dependent IgG subclass and IgE antibody production during vaccination protocols may be a useful surrogate marker indicating a potentially undesirable change of the immunological environment for an effective antitumor immune response in cancer patients.

13.
Clin Cancer Res ; 21(10): 2268-77, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25855804

ABSTRACT

PURPOSE: Preparative lymphodepletion, the temporal ablation of the immune system, has been reported to promote persistence of transferred cells along with increased rates of tumor regression in patients treated with adoptive T-cell therapy. However, it remains unclear whether lymphodepletion is indispensable for immunotherapy with T-cell receptor (TCR) gene-engineered T cells. EXPERIMENTAL DESIGN: We conducted a first-in-man clinical trial of TCR gene-transduced T-cell transfer in patients with recurrent MAGE-A4-expressing esophageal cancer. The patients were given sequential MAGE-A4 peptide vaccinations. The regimen included neither lymphocyte-depleting conditioning nor administration of IL2. Ten patients, divided into 3 dose cohorts, received T-cell transfer. RESULTS: TCR-transduced cells were detected in the peripheral blood for 1 month at levels proportional to the dose administered, and in 5 patients they persisted for more than 5 months. The persisting cells maintained ex vivo antigen-specific tumor reactivity. Despite the long persistence of the transferred T cells, 7 patients exhibited tumor progression within 2 months after the treatment. Three patients who had minimal tumor lesions at baseline survived for more than 27 months. CONCLUSIONS: These results suggest that TCR-engineered T cells created by relatively short-duration in vitro culture of polyclonal lymphocytes in peripheral blood retained the capacity to survive in a host. The discordance between T-cell survival and tumor regression suggests that multiple mechanisms underlie the benefits of preparative lymphodepletion in adoptive T-cell therapy.


Subject(s)
Antigens, Neoplasm/genetics , Carcinoma, Squamous Cell/therapy , Esophageal Neoplasms/therapy , Neoplasm Proteins/genetics , Neoplasm Recurrence, Local/therapy , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/immunology , Adoptive Transfer , Adult , Aged , Carcinoma, Squamous Cell/immunology , Cell Survival , Cells, Cultured , Esophageal Neoplasms/immunology , Female , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/immunology , T-Lymphocytes/transplantation , Transduction, Genetic , Treatment Outcome
14.
Cancer Res ; 74(24): 7205-16, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25320012

ABSTRACT

While viral antigens in human papillomavirus (HPV)-related oropharyngeal cancer (HPVOPC) are attractive targets for immunotherapy, the effects of existing standard-of-care therapies on immune responses to HPV are poorly understood. We serially sampled blood from patients with stage III-IV oropharyngeal cancer undergoing concomitant chemoradiotherapy with or without induction chemotherapy. Circulating immunocytes including CD4(+) and CD8(+) T cells, regulatory T cells (Treg), and myeloid-derived suppressor cells (MDSC) were profiled by flow cytometry. Antigen-specific T-cell responses were measured in response to HPV16 E6 and E7 peptide pools. The role of PD-1 signaling in treatment-related immunosuppression was functionally defined by performing HPV-specific T-cell assays in the presence of blocking antibody. While HPV-specific T-cell responses were present in 13 of 18 patients before treatment, 10 of 13 patients lost these responses within 3 months after chemoradiotherapy. Chemoradiotherapy decreased circulating T cells and markedly elevated MDSCs. PD-1 expression on CD4(+) T cells increased by nearly 2.5-fold after chemoradiotherapy, and ex vivo culture with PD-1-blocking antibody enhanced HPV-specific T-cell responses in 8 of 18 samples tested. Chemoradiotherapy suppresses circulating immune responses in patients with HPVOPC by unfavorably altering effector:suppressor immunocyte ratios and upregulating PD-1 expression on CD4(+) T cells. These data strongly support testing of PD-1-blocking agents in combination with standard-of-care chemoradiotherapy for HPVOPC.


Subject(s)
Immunotherapy , Oropharyngeal Neoplasms/genetics , Oropharyngeal Neoplasms/immunology , Programmed Cell Death 1 Receptor/biosynthesis , CD8-Positive T-Lymphocytes/immunology , Chemoradiotherapy , Female , Human papillomavirus 16/immunology , Human papillomavirus 16/metabolism , Humans , Induction Chemotherapy , Neoplasm Staging , Oropharyngeal Neoplasms/pathology , Papillomavirus E7 Proteins/biosynthesis , Papillomavirus E7 Proteins/metabolism , Papillomavirus Infections/genetics , Papillomavirus Infections/pathology , Programmed Cell Death 1 Receptor/immunology
15.
Eur J Immunol ; 44(6): 1747-58, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24723437

ABSTRACT

T cells express multiple integrin molecules. The significance of signaling through these molecules on acquisition of T-cell effector functions and memory formation capacity remains largely unknown. Moreover, the impact of stimulation through these signals on the generation of T cells for adoptive immunotherapy has not been elucidated. In this study, using a recombinant fragment of fibronectin, CH-296, we demonstrated that stimulation via very late Ag (VLA)-4 and VLA-5 in human and BALB/c mouse CD8(+) T cells, in combination with TCR stimulation, enhances effector multifunctionality and in vivo memory formation. Using TCR-transgenic mouse-derived CD8(+) T cells expressing TCR specific for the syngeneic CMS5 fibrosarcoma-derived tumor Ag, we showed that stimulation by CH-296 improved the ability of tumor-specific CD8(+) T cells to inhibit CMS5 tumor growth when adoptively transferred into hosts with progressing tumors. Improved antitumor effects were associated with decreased infiltration of Foxp3(+) CD4(+) Treg cells in tumors. These results suggest that stimulation via VLA-4 and VLA-5 modulates the qualities of effector T cells and could potentially increase the efficacy of adoptive therapy against cancer.


Subject(s)
Antigens, Neoplasm/immunology , Fibrosarcoma/immunology , Immunologic Memory , Integrin alpha4beta1/immunology , Integrin alpha5beta1/immunology , Adoptive Transfer , Animals , Antigens, Neoplasm/genetics , CD8-Positive T-Lymphocytes , Cell Line, Tumor , Female , Fibrosarcoma/genetics , Fibrosarcoma/pathology , Fibrosarcoma/therapy , Humans , Integrin alpha4beta1/genetics , Integrin alpha5beta1/genetics , Mice , Mice, Inbred BALB C , Mice, Transgenic , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology
16.
J Transl Med ; 11: 246, 2013 Oct 05.
Article in English | MEDLINE | ID: mdl-24093426

ABSTRACT

BACKGROUND: Cholesteryl pullulan (CHP) is a novel antigen delivery system for cancer vaccines. This study evaluated the safety, immune responses and clinical outcomes of patients who received the CHP-NY-ESO-1 complex vaccine, Drug code: IMF-001. METHODS: Patients with advanced/metastatic esophageal cancer were enrolled and subcutaneously vaccinated with either 100 µg or 200 µg of NY-ESO-1 protein complexed with CHP. The primary endpoints were safety and humoral immune responses, and the secondary endpoint was clinical efficacy. RESULTS: A total of 25 patients were enrolled. Thirteen and twelve patients were repeatedly vaccinated with 100 µg or 200 µg of CHP-NY-ESO-1 with a median of 8 or 9.5 doses, respectively. No serious adverse events related to the vaccine were observed. Three out of 13 patients in the 100-µg cohort and 7 out of 12 patients in the 200-µg cohort were positive for anti-NY-ESO-1 antibodies at baseline. In the 100-µg cohort, an antibody response was observed in 5 out of 10 pre-antibody-negatives patients, and the antibody levels were augmented in 2 pre-antibody-positive patients after vaccination. In the 200-µg cohort, all 5 pre-antibody-negative patients became seropositive, and the antibody level was amplified in all 7 pre-antibody-positive patients. No tumor shrinkage was observed. The patients who received 200 µg of CHP-NY-ESO-1 survived longer than patients receiving 100 µg of CHP-NY-ESO-1, even those who exhibited unresponsiveness to previous therapies or had higher tumor burdens. CONCLUSIONS: The safety and immunogenicity of CHP-NY-ESO-1 vaccine were confirmed. The 200 µg dose more efficiently induced immune responses and suggested better survival benefits. (Clinical trial registration number NCT01003808).


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines/therapeutic use , Cholesterol/immunology , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/immunology , Glucans/immunology , Immunity , Kaplan-Meier Estimate , Membrane Proteins/immunology , Aged , Antibodies, Neoplasm/immunology , Antibody Formation/immunology , Cancer Vaccines/adverse effects , Cancer Vaccines/immunology , Demography , Dose-Response Relationship, Immunologic , Enzyme-Linked Immunosorbent Assay , Esophageal Neoplasms/prevention & control , Female , Follow-Up Studies , Humans , Male , Middle Aged , Recombinant Proteins/immunology , Treatment Outcome , Vaccination/adverse effects
17.
Mol Clin Oncol ; 1(2): 326-330, 2013 Mar.
Article in English | MEDLINE | ID: mdl-24649170

ABSTRACT

The aim of this study was to evaluate the frequency of expression of the cancer-testis antigens (CTAs) NY-ESO-1, MAGE-A4 and SAGE, in renal cell carcinoma (RCC) patients compared to that in head and neck cancer (HNC) patients, which represent a positive control with a high incidence of CTA expression, to identify novel target antigens for immunotherapy. We prospectively examined frozen tissue samples collected from surgery or biopsy from 35 RCC and 40 HNC patients. Total RNA was extracted, and real-time reverse transcription-polymerase chain reaction (RT)-PCR was performed to determine the expression of MAGE-A4, NY-ESO-1 and SAGE. MAGE-A4 was not detected in any of the RCC samples, although a low incidence of NY-ESO-1 (5.7%; 2/35) and SAGE (2.9%; 1/35) expression was observed. No samples demonstrated co-expression of the three CTAs. By contrast, a comparatively high incidence of CTA expression was detected in squamous cell carcinoma (SCC) specimens of HNC patients. The actual incidence was 42.5% (17/40) for MAGE-A4, 20% (8/40) for NY-ESO-1 and 15% (6/40) for SAGE. The incidence of co-expression was 7.5% (3/40) for MAGE-A4 and NY-ESO-1, 7.5% (3/40) for MAGE-A4 and SAGE, 7.5% (3/40) for NY-ESO-1 and SAGE, and 2.5% (1/40) for the CTAs. The number of HNC samples positive for MAGE-A4 was significantly higher compared to that of RCC samples. The remaining two antigens, NY-ESO-1 and SAGE, were expressed at high levels in HNC compared to RCC samples. Limited frequency of CTA (NY-ESO-1, MAGE-A4 and SAGE) expression was demonstrated in RCC compared to HNC samples.

18.
Nihon Rinsho ; 70(12): 2125-9, 2012 Dec.
Article in Japanese | MEDLINE | ID: mdl-23259384

ABSTRACT

Cancer/testis (CT) antigen is a group of antigens that are expressed in a wide variety of malignant tumors but not in normal adult tissues except for testis. Since CT antigens are immunogenic and highly restricted to tumors, they are considered as ideal targets for cancer immunotherapy. Many clinical studies targeting CT antigens have been tested. Here we review the history and the recent progress of clinical studies targeting MAGE family and NY-ESO-1 including our trials.


Subject(s)
Antigens, Neoplasm/immunology , Cell- and Tissue-Based Therapy/methods , Immunotherapy/methods , Membrane Proteins/immunology , Neoplasm Proteins/immunology , Neoplasms/therapy , Testis/immunology , Humans , Male , Neoplasms/immunology
19.
Breed Sci ; 61(5): 631-8, 2012 Jan.
Article in English | MEDLINE | ID: mdl-23136502

ABSTRACT

Palmitic acid is the most abundant (approx. 11% of total fatty acids) saturated fatty acid in conventional soybean seed oil. Increasing the saturated acid content of soybean oil improves its oxidative stability and plasticity. We have developed three soybean mutants with high palmitic acid content by X-ray irradiation. In this study, we successfully identified the mutated sites of two of these high-palmitic-acid mutants, J10 and M22. PCR-based mutant analysis revealed that J10 has a 206,203-bp-long deletion that includes the GmKASIIA gene and 16 other predicted genes, and M22 has a 26-bp-long deletion in the sixth intron of GmKASIIB. The small deletion in M22 causes mis-splicing of GmKASIIB transcripts, which should result in nonfunctional products. In addition, we designed co-dominant marker sets for these mutant alleles and confirmed the association of genotypes and palmitic acid contents in F(2) seeds of J10 X M22. This information will be useful in breeding programs to develop novel soybean cultivars with improved palmitic acid content. However, in the third mutant, KK7, we found no polymorphism in either GmKASIIA or GmKASIIB, which suggests that several unknown genes in addition to GmKASIIA and GmKASIIB may be involved in elevating the palmitic acid content of soybean seed oil.

20.
Cancer Sci ; 103(1): 17-25, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21951605

ABSTRACT

Adoptive cell therapy with lymphocytes that have been genetically engineered to express tumor-reactive T-cell receptors (TCR) is a promising approach for cancer immunotherapy. We have been exploring the development of TCR gene therapy targeting cancer/testis antigens, including melanoma-associated antigen (MAGE) family antigens, that are ideal targets for adoptive T-cell therapy. The efficacy of TCR gene therapy targeting MAGE family antigens, however, has not yet been evaluated in vivo. Here, we demonstrate the in vivo antitumor activity in immunodeficient non-obese diabetic/SCID/γc(null) (NOG) mice of human lymphocytes genetically engineered to express TCR specific for the MAGE-A4 antigen. Polyclonal T cells derived from human peripheral blood mononuclear cells were transduced with the αß TCR genes specific for MAGE-A4, then adoptively transferred into NOG mice inoculated with MAGE-A4 expressing human tumor cell lines. The transferred T cells maintained their effector function in vivo, infiltrated into tumors, and inhibited tumor growth in an antigen-specific manner. The combination of adoptive cell therapy with antigen peptide vaccination enhanced antitumor activity, with improved multifunctionality of the transferred cells. These data suggest that TCR gene therapy with MAGE-A4-specific TCR is a promising strategy to treat patients with MAGE-A4-expressing tumors; in addition, the acquisition of multifunctionality in vivo is an important factor to predict the quality of the T-cell response during adoptive therapy with human lymphocytes.


Subject(s)
Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Esophageal Neoplasms/therapy , Genetic Therapy , Immunotherapy, Adoptive , Lung Neoplasms/therapy , Neoplasm Proteins/immunology , Receptors, Antigen, T-Cell, alpha-beta/genetics , Animals , Antigens, Neoplasm/genetics , Combined Modality Therapy , Cytotoxicity, Immunologic/immunology , Esophageal Neoplasms/immunology , Female , Flow Cytometry , Genetic Vectors/therapeutic use , HLA-A Antigens/genetics , HLA-A Antigens/immunology , Humans , Immunoenzyme Techniques , Leukocytes, Mononuclear/immunology , Lung Neoplasms/immunology , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Proteins/genetics , Retroviridae , Transduction, Genetic , Vaccines, Subunit/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...