Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 137
Filter
1.
Nat Immunol ; 21(11): 1346-1358, 2020 11.
Article in English | MEDLINE | ID: mdl-32868929

ABSTRACT

Immune checkpoint blockade has provided a paradigm shift in cancer therapy, but the success of this approach is very variable; therefore, biomarkers predictive of clinical efficacy are urgently required. Here, we show that the frequency of PD-1+CD8+ T cells relative to that of PD-1+ regulatory T (Treg) cells in the tumor microenvironment can predict the clinical efficacy of programmed cell death protein 1 (PD-1) blockade therapies and is superior to other predictors, including PD ligand 1 (PD-L1) expression or tumor mutational burden. PD-1 expression by CD8+ T cells and Treg cells negatively impacts effector and immunosuppressive functions, respectively. PD-1 blockade induces both recovery of dysfunctional PD-1+CD8+ T cells and enhanced PD-1+ Treg cell-mediated immunosuppression. A profound reactivation of effector PD-1+CD8+ T cells rather than PD-1+ Treg cells by PD-1 blockade is necessary for tumor regression. These findings provide a promising predictive biomarker for PD-1 blockade therapies.


Subject(s)
Gene Expression Regulation/drug effects , Immune Checkpoint Inhibitors/pharmacology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/genetics , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Antigens/chemistry , Antigens/immunology , Biomarkers, Tumor , CD28 Antigens/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Humans , Immune Checkpoint Inhibitors/therapeutic use , Immunomodulation , Lymphocyte Activation/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Molecular Targeted Therapy , Neoplasm Metastasis , Neoplasm Staging , Neoplasms/drug therapy , Neoplasms/etiology , Neoplasms/metabolism , Neoplasms/mortality , Peptides/chemistry , Peptides/immunology , Prognosis , Programmed Cell Death 1 Receptor/metabolism , Reactive Oxygen Species/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/drug effects , Treatment Outcome , Tumor Microenvironment/immunology
2.
Immunity ; 53(1): 187-203.e8, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32640259

ABSTRACT

Only a small percentage of patients afflicted with gastric cancer (GC) respond to immune checkpoint blockade (ICB). To study the mechanisms underlying this resistance, we examined the immune landscape of GC. A subset of these tumors was characterized by high frequencies of regulatory T (Treg) cells and low numbers of effector T cells. Genomic analyses revealed that these tumors bore mutations in RHOA that are known to drive tumor progression. RHOA mutations in cancer cells activated the PI3K-AKT-mTOR signaling pathway, increasing production of free fatty acids that are more effectively consumed by Treg cells than effector T cells. RHOA mutant tumors were resistant to PD-1 blockade but responded to combination of PD-1 blockade with inhibitors of the PI3K pathway or therapies targeting Treg cells. We propose that the metabolic advantage conferred by RHOA mutations enables Treg cell accumulation within GC tumors, generating an immunosuppressive TME that underlies resistance to ICB.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immune Checkpoint Inhibitors/pharmacology , Stomach Neoplasms/genetics , T-Lymphocytes, Regulatory/metabolism , rhoA GTP-Binding Protein/genetics , Animals , CD4 Lymphocyte Count , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Chemokine CXCL10/biosynthesis , Chemokine CXCL11/biosynthesis , Fatty Acids, Nonesterified/biosynthesis , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Programmed Cell Death 1 Receptor/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/immunology , Stomach Neoplasms/immunology , Stomach Neoplasms/pathology , T-Lymphocytes, Regulatory/immunology , TOR Serine-Threonine Kinases/metabolism , Tumor Microenvironment/immunology
3.
Proc Natl Acad Sci U S A ; 121(35): e2320189121, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39167601

ABSTRACT

Immune checkpoint inhibitors (ICIs) exert clinical efficacy against various types of cancers by reinvigorating exhausted CD8+ T cells that can expand and directly attack cancer cells (cancer-specific T cells) among tumor-infiltrating lymphocytes (TILs). Although some reports have identified somatic mutations in TILs, their effect on antitumor immunity remains unclear. In this study, we successfully established 18 cancer-specific T cell clones, which have an exhaustion phenotype, from the TILs of four patients with melanoma. We conducted whole-genome sequencing for these T cell clones and identified various somatic mutations in them with high clonality. Among the somatic mutations, an SH2D2A loss-of-function frameshift mutation and TNFAIP3 deletion could activate T cell effector functions in vitro. Furthermore, we generated CD8+ T cell-specific Tnfaip3 knockout mice and showed that Tnfaip3 function loss in CD8+ T cell increased antitumor immunity, leading to remarkable response to PD-1 blockade in vivo. In addition, we analyzed bulk CD3+ T cells from TILs in additional 12 patients and identified an SH2D2A mutation in one patient through amplicon sequencing. These findings suggest that somatic mutations in TILs can affect antitumor immunity and suggest unique biomarkers and therapeutic targets.


Subject(s)
CD8-Positive T-Lymphocytes , Lymphocytes, Tumor-Infiltrating , Tumor Necrosis Factor alpha-Induced Protein 3 , Lymphocytes, Tumor-Infiltrating/immunology , Humans , CD8-Positive T-Lymphocytes/immunology , Animals , Mice , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Melanoma/immunology , Melanoma/genetics , Mutation , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Mice, Knockout , Female
4.
Cancer Sci ; 115(8): 2839-2845, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38898727

ABSTRACT

By taking advantage of forward genetic analysis in mice, we have demonstrated that Pak1 plays a crucial role during DMBA/TPA skin carcinogenesis. Although Pak1 has been considered to promote cancer development, its overall function remains poorly understood. To clarify the functional significance of Pak1 in detail, we sought to evaluate the possible effect of an allosteric inhibitor against PAK1 (NVS-PAK1-1) on a syngeneic mouse model. To this end, we established two cell lines, 9AS1 and 19AS1, derived from DMBA/TPA-induced squamous cell carcinoma (SCC) that engrafted in FVB mice. Based on our present results, NVS-PAK1-1 treatment significantly inhibited the growth of tumors derived from 9AS1 and 19AS1 cells in vitro and in vivo. RNA-sequencing analysis on the engrafted tumors indicates that NVS-PAK1-1 markedly potentiates the epidermal cell differentiation and enhances the immune response in the engrafted tumors. Consistent with these observations, we found an expansion of Pan-keratin-positive regions and potentially elevated infiltration of CD8-positive immune cells in NVS-PAK1-1-treated tumors as examined by immunohistochemical analyses. Together, our present findings strongly suggest that PAK1 is tightly linked to the development of SCC, and that its inhibition is a promising therapeutic strategy against SCC.


Subject(s)
Carcinoma, Squamous Cell , Disease Models, Animal , Skin Neoplasms , p21-Activated Kinases , Animals , p21-Activated Kinases/metabolism , p21-Activated Kinases/genetics , p21-Activated Kinases/antagonists & inhibitors , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/genetics , Mice , Cell Line, Tumor , 9,10-Dimethyl-1,2-benzanthracene/toxicity , Female , Cell Differentiation/drug effects , Tetradecanoylphorbol Acetate , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation/drug effects
5.
Cancer Sci ; 115(10): 3231-3247, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39119813

ABSTRACT

Immune checkpoint inhibitors (ICIs) are effective against many advanced malignancies. However, many patients are nonresponders to immunotherapy, and overcoming this resistance to treatment is important. Boron neutron capture therapy (BNCT) is a local chemoradiation therapy with the combination of boron drugs that accumulate selectively in cancer and the neutron irradiation of the cancer site. Here, we report the first boron neutron immunotherapy (B-NIT), combining BNCT and ICI immunotherapy, which was performed on a radioresistant and immunotherapy-resistant advanced-stage B16F10 melanoma mouse model. The BNCT group showed localized tumor suppression, but the anti-PD-1 antibody immunotherapy group did not show tumor suppression. Only the B-NIT group showed strong tumor growth inhibition at both BNCT-treated and shielded distant sites. Intratumoral CD8+ T-cell infiltration and serum high mobility group box 1 (HMGB1) levels were higher in the B-NIT group. Analysis of CD8+ T cells in tumor-infiltrating lymphocytes (TILs) showed that CD62L- CD44+ effector memory T cells and CD69+ early-activated T cells were predominantly increased in the B-NIT group. Administration of CD8-depleting mAb to the B-NIT group completely suppressed the augmented therapeutic effects. This indicated that B-NIT has a potent immune-induced abscopal effect, directly destroying tumors with BNCT, inducing antigen-spreading effects, and protecting normal tissue. B-NIT, immunotherapy combined with BNCT, is the first treatment to overcome immunotherapy resistance in malignant melanoma. In the future, as its therapeutic efficacy is demonstrated not only in melanoma but also in other immunotherapy-resistant malignancies, B-NIT can become a new treatment candidate for advanced-stage cancers.


Subject(s)
Boron Neutron Capture Therapy , CD8-Positive T-Lymphocytes , Immune Checkpoint Inhibitors , Immunotherapy , Melanoma, Experimental , Animals , Mice , Boron Neutron Capture Therapy/methods , Melanoma, Experimental/therapy , Melanoma, Experimental/immunology , CD8-Positive T-Lymphocytes/immunology , Immunotherapy/methods , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Female , Mice, Inbred C57BL , Lymphocytes, Tumor-Infiltrating/immunology , Cell Line, Tumor , Drug Resistance, Neoplasm , Combined Modality Therapy , HMGB1 Protein/metabolism
6.
Br J Cancer ; 131(2): 283-289, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38834744

ABSTRACT

BACKGROUND: Preoperative chemoradiotherapy (CRT) followed by surgery is the standard treatment for locally advanced rectal cancer (LARC). We reported the short-term outcomes of the VOLTAGE trial that investigated the safety and efficacy of preoperative CRT followed by nivolumab and surgery. Here, we present the 3-year outcomes of this trial. METHODS: Thirty-nine patients with microsatellite stable (MSS) LARC and five patients with microsatellite instability-high (MSI-H) LARC underwent CRT (50.4 Gy) followed by five doses of nivolumab (240 mg) and surgery. The 3-year relapse-free survival (RFS), overall survival (OS), and associations with biomarkers were evaluated. RESULTS: The 3-year RFS rates in patients with MSS and MSI-H were 79.5% and 100%, respectively, and the 3-year OS rates were 97.4% and 100%, respectively. Of the MSS patients, those with pre-CRT PD-L1 positivity, pre-CRT high CD8 + T cell/effector regulatory T cell (eTreg) ratio, pre-CRT high expression of Ki-67, CTLA-4, and PD-1 had a trend toward better 3-year RFS than those without. CONCLUSIONS: Three-year outcomes of patients with MSI-H were better than those of patients with MSS. PD-L1 positivity, elevated CD8/eTreg ratio, and high expression of Ki-67, CTLA-4, and PD-1 could be positive predictors of prognosis in patients with MSS. CLINICAL TRIAL REGISTRATION: ClinicalTrials.gov Identifier: NCT02948348.


Subject(s)
Chemoradiotherapy , Microsatellite Instability , Nivolumab , Rectal Neoplasms , Adult , Aged , Female , Humans , Male , Middle Aged , Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Agents, Immunological/administration & dosage , B7-H1 Antigen/genetics , Chemoradiotherapy/methods , CTLA-4 Antigen , Nivolumab/administration & dosage , Nivolumab/therapeutic use , Rectal Neoplasms/therapy , Rectal Neoplasms/genetics , Rectal Neoplasms/pathology , Rectal Neoplasms/mortality , Treatment Outcome
7.
N Engl J Med ; 384(1): 42-50, 2021 01 07.
Article in English | MEDLINE | ID: mdl-33406329

ABSTRACT

Two cases of pediatric lung cancer (in 23-month-old and 6-year-old boys) resulting from mother-to-infant transmission of uterine cervical tumors were incidentally detected during routine next-generation sequencing of paired samples of tumor and normal tissue. Spontaneous regression of some lesions in the first child and slow growth of the tumor mass in the second child suggested the existence of alloimmune responses against the transmitted tumors. Immune checkpoint inhibitor therapy with nivolumab led to a strong regression of all remaining tumors in the first child. (Funded by the Japan Agency for Medical Research and Development and others; TOP-GEAR UMIN Clinical Trials Registry number, UMIN000011141.).


Subject(s)
Adenocarcinoma, Mucinous/etiology , Carcinoma, Neuroendocrine/etiology , Lung Neoplasms/etiology , Pregnancy Complications, Neoplastic , Uterine Cervical Neoplasms , Adenocarcinoma, Mucinous/diagnostic imaging , Adenocarcinoma, Mucinous/genetics , Adult , Carcinoma, Neuroendocrine/diagnostic imaging , Carcinoma, Neuroendocrine/genetics , Carcinoma, Squamous Cell/pathology , Child , Fatal Outcome , Female , High-Throughput Nucleotide Sequencing , Humans , Infant , Lung/diagnostic imaging , Lung/pathology , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Mothers , Pregnancy , Vagina , Exome Sequencing
8.
Nat Immunol ; 18(12): 1285-1286, 2017 11 16.
Article in English | MEDLINE | ID: mdl-29144496
9.
Cancer Sci ; 114(10): 3848-3856, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37485636

ABSTRACT

Hydrogen peroxide (H2 O2 ) induces oxidative stress and cytotoxicity, and can be used for treating cancers in combination with radiotherapy. A product comprising H2 O2 and sodium hyaluronate has been developed as a radiosensitizer. However, the effects of H2 O2 on antitumor immunity remain unclear. To investigate the effects of H2 O2 , especially the abscopal effect when combined with radiotherapy (RT), we implanted murine tumor cells simultaneously in two locations in mouse models: the hind limb and back. H2 O2 mixed with sodium hyaluronate was injected intratumorally, followed by irradiation only at the hind limb lesion. No treatment was administered to the back lesion. The H2 O2 /RT combination significantly reduced tumor growth at the noninjected/nonirradiated site in the back lesion, whereas H2 O2 or RT individually did not reduce tumor growth. Flow cytometric analyses of the tumor-draining lymph nodes in the injected/irradiated areas showed that the number of dendritic cells increased significantly with maturation in the H2 O2 /RT combination group. In addition, analyses of tumor-infiltrating lymphocytes showed that the number of CD8+ (cluster of differentiation 8) T cells and the frequency of IFN-γ+ (interferon gamma) CD8+ T cells were higher in the noninjected/nonirradiated tumors in the H2 O2 /RT group compared to those in the other groups. PD-1 (programmed death receptor 1) blockade further increased the antitumor effect against noninjected/nonirradiated tumors in the H2 O2 /RT group. Intratumoral injection of H2 O2 combined with RT therefore induces an abscopal effect by activating antitumor immunity, which can be further enhanced by PD-1 blockade. These findings promote the development of H2 O2 /RT therapy combined with cancer immunotherapies, even for advanced cancers.

10.
Cancer Sci ; 114(5): 1859-1870, 2023 May.
Article in English | MEDLINE | ID: mdl-36762794

ABSTRACT

Combination therapy with anti-cytotoxic T lymphocyte-associated protein 4 (CTLA-4) and anti-programmed death-1 (PD-1) monoclonal antibodies (mAbs) has dramatically improved the prognosis of patients with multiple types of cancer, including renal cell carcinoma (RCC). However, more than half of RCC patients fail to respond to this therapy. Regulatory T cells (Treg cells) are a subset of highly immunosuppressive CD4+ T cells that promote the immune escape of tumors by suppressing effector T cells in the tumor microenvironment (TME) through various mechanisms. CTLA-4 is constitutively expressed in Treg cells and is regarded as a key molecule for Treg-cell-mediated immunosuppressive functions, suppressing antigen-presenting cells by binding to CD80/CD86. Reducing Treg cells in the TME with an anti-CTLA-4 mAb with antibody-dependent cellular cytotoxicity (ADCC) activity is considered an essential mechanism to achieve tumor regression. In contrast, we demonstrated that CTLA-4 blockade without ADCC activity enhanced CD28 costimulatory signaling pathways in Treg cells and promoted Treg-cell proliferation in mouse models. CTLA-4 blockade also augmented CTLA-4-independent immunosuppressive functions, including cytokine production, leading to insufficient antitumor effects. Similar results were also observed in human peripheral blood lymphocytes and tumor-infiltrating lymphocytes from patients with RCC. Our findings highlight the importance of Treg-cell depletion to achieve tumor regression in response to CTLA-4 blockade therapies.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Animals , Mice , Humans , T-Lymphocytes, Regulatory , CD8-Positive T-Lymphocytes , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/metabolism , Immunosuppression Therapy , CTLA-4 Antigen/metabolism , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal/metabolism , Kidney Neoplasms/drug therapy , Kidney Neoplasms/metabolism , Tumor Microenvironment
11.
Br J Cancer ; 128(6): 1166-1175, 2023 04.
Article in English | MEDLINE | ID: mdl-36732592

ABSTRACT

BACKGROUND: Identifying biomarkers to predict immune checkpoint inhibitor (ICI) efficacy is warranted. Considering that somatic mutation-derived neoantigens induce strong immune responses, patients with a high tumour mutational burden reportedly tend to respond to ICIs. However, there are several conflicting data. Therefore, we focused on the original function of neoantigenic mutations and their impact on the tumour microenvironment (TME). METHODS: We evaluated 88 high-frequency microsatellite instability (MSI-H) colorectal cancers and analysed the function of the identified neoantigenic mutations and their influence on programmed cell death 1 (PD-1) blockade efficacy. The results were validated using The Cancer Genome Atlas (TCGA) datasets. RESULTS: We identified frameshift mutations in RNF43 as a common neoantigenic gene mutation in MSI-H tumours. However, loss-of-function RNF43 mutations induced noninflamed TME by activating the WNT/ß-catenin signalling pathway. In addition, loss of RNF43 function induced resistance to PD-1 blockade even in neoantigen-rich tumours. TCGA dataset analyses demonstrated that passenger rather than driver gene mutations were related to the inflamed TME in diverse cancer types. CONCLUSIONS: We propose a novel concept of "paradoxical neoantigenic mutations" that can induce noninflamed TME through their original gene functions, despite deriving neoantigens, suggesting the significance of qualities as well as quantities in neoantigenic mutations.


Subject(s)
Colorectal Neoplasms , Neoplasms , Humans , Programmed Cell Death 1 Receptor , Tumor Microenvironment , Neoplasms/genetics , Mutation , Microsatellite Instability , Colorectal Neoplasms/pathology
12.
Gastroenterology ; 162(3): 799-812, 2022 03.
Article in English | MEDLINE | ID: mdl-34687740

ABSTRACT

BACKGROUND & AIMS: A detailed understanding of antitumor immunity is essential for optimal cancer immune therapy. Although defective mutations in the B2M and HLA-ABC genes, which encode molecules essential for antigen presentation, have been reported in several studies, the effects of these defects on tumor immunity have not been quantitatively evaluated. METHODS: Mutations in HLA-ABC genes were analyzed in 114 microsatellite instability-high colorectal cancers using a long-read sequencer. The data were further analyzed in combination with whole-exome sequencing, transcriptome sequencing, DNA methylation array, and immunohistochemistry data. RESULTS: We detected 101 truncating mutations in 57 tumors (50%) and loss of 61 alleles in 21 tumors (18%). Based on the integrated analysis that enabled the immunologic subclassification of microsatellite instability-high colorectal cancers, we identified a subtype of tumors in which lymphocyte infiltration was reduced, partly due to reduced expression of HLA-ABC genes in the absence of apparent genetic alterations. Survival time of patients with such tumors was shorter than in patients with other tumor types. Paradoxically, tumor mutation burden was highest in the subtype, suggesting that the immunogenic effect of accumulating mutations was counterbalanced by mutations that weakened immunoreactivity. Various genetic and epigenetic alterations, including frameshift mutations in RFX5 and promoter methylation of PSMB8 and HLA-A, converged on reduced expression of HLA-ABC genes. CONCLUSIONS: Our detailed immunogenomic analysis provides information that will facilitate the improvement and development of cancer immunotherapy.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Genes, MHC Class I/genetics , Tumor Escape/genetics , Tumor Escape/immunology , beta 2-Microglobulin/genetics , Alleles , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , DNA Methylation , Epigenesis, Genetic , Gene Expression , HLA-A Antigens/genetics , HLA-A Antigens/metabolism , Humans , Immunogenetics , Lymphocytes, Tumor-Infiltrating , Microsatellite Instability , Proteasome Endopeptidase Complex/genetics , Regulatory Factor X Transcription Factors/genetics , Survival Rate , beta 2-Microglobulin/metabolism
13.
Exp Dermatol ; 32(3): 240-249, 2023 03.
Article in English | MEDLINE | ID: mdl-36437644

ABSTRACT

Immune checkpoint inhibitors (ICIs) have contributed significantly to the treatment of various types of cancer, including skin cancer. However, not all patients respond; some patients do not respond at all (primary resistance), while others experience recurrence after the initial response (acquired resistance). Therefore, overcoming ICI resistance is an urgent priority. Numerous ICI resistance mechanisms have been reported. They are seemingly quite complex, varying from patient to patient. However, most involve T-cell activation processes, especially in the tumor microenvironment (TME). ICIs exert their effects in the TME by reactivating suppressed T cells through inhibition of immune checkpoint molecules, such as cytotoxic T-lymphocyte antigen-4 (CTLA-4) and programmed cell death protein 1 (PD-1). Thus, this review focuses on the resistance mechanisms based on the T-cell activation process. Here, we classify the main mechanisms of ICI resistance into three categories based on (1) antigen recognition, (2) T-cell migration and infiltration, and (3) effector functions of T cells. By identifying and understanding these resistance mechanisms individually, including unknown mechanisms, we seek to contribute to the development of novel treatments to overcome ICI resistance.


Subject(s)
Neoplasms , Skin Neoplasms , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Tumor Microenvironment , Neoplasms/drug therapy , Neoplasms/pathology , Skin Neoplasms/drug therapy , Immunotherapy
14.
Int Immunol ; 34(11): 563-570, 2022 10 05.
Article in English | MEDLINE | ID: mdl-35460561

ABSTRACT

In T-cell biology, 'exhaustion' was initially described as a hyporesponsive state in CD8+ T cells during chronic infections. Recently, exhaustion has been recognized as a T-cell dysfunctional state in the tumor microenvironment (TME). The term 'exhaustion' is used mainly to refer to effector T cells with a reduced capacity to secrete cytokines and an increased expression of inhibitory receptors. The up-regulation of exhaustion-related inhibitory receptors, including programmed cell death protein 1 (PD-1), in such T cells has been associated with the development of tumors, prompting the development of immune checkpoint inhibitors. In addition to CD8+ T cells, CD4+ T cells, including the regulatory T (Treg) cell subset, perform a wide variety of functions within the adaptive immune system. Up-regulation of the same inhibitory receptors that are associated with CD8+ T-cell exhaustion has also been identified in CD4+ T cells in chronic infections and cancers, suggesting a similar CD4+ T-cell exhaustion phenotype. For instance, high expression of PD-1 has been observed in Treg cells in the TME, and such Treg cells can play an important role in the resistance to PD-1 blockade therapies. Furthermore, recent progress in single-cell RNA sequencing has shown that CD4+ T cells with cytotoxic activity are also vulnerable to exhaustion. In this review, we will discuss novel insights into various exhausted T-cell subsets, which could reveal novel therapeutic targets and strategies to induce a robust anti-tumor immune response.


Subject(s)
Neoplasms , Programmed Cell Death 1 Receptor , CD8-Positive T-Lymphocytes , Cytokines/metabolism , Humans , Immune Checkpoint Inhibitors , Tumor Microenvironment
15.
Cancer Sci ; 113(10): 3303-3312, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35848888

ABSTRACT

Immune checkpoint inhibitors (ICIs) are effective for various types of cancer, and their application has led to paradigm shifts in cancer treatment. While many patients can obtain clinical benefits from ICI treatment, a large number of patients are primarily resistant to such treatment or acquire resistance after an initial response. Thus, elucidating the resistance mechanisms is warranted to improve the clinical outcomes of ICI treatment. ICIs exert their antitumor effects by activating T cells in the tumor microenvironment. There are various resistance mechanisms, such as insufficient antigen recognition by T cells, impaired T-cell migration and/or infiltration, and reduced T-cell cytotoxicity, most of which are related to the T-cell activation process. Thus, we classify them into three main mechanisms: resistance mechanisms related to antigen recognition, T-cell migration and/or infiltration, and effector functions of T cells. In this review, we summarize these mechanisms of resistance to ICIs related to the T-cell activation process and progress in the development of novel therapies that can overcome resistance.


Subject(s)
Immune Checkpoint Inhibitors , Neoplasms , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy , Neoplasms/pathology , T-Lymphocytes/pathology , Tumor Microenvironment
16.
Int J Clin Oncol ; 27(2): 316-322, 2022 Feb.
Article in English | MEDLINE | ID: mdl-33783658

ABSTRACT

Aging leads to numerous changes that affect many components of the immune system, called "immunosenescence". Indeed, elderly individuals exhibit dysregulated immune responses against pathogens, poor responses to vaccination, and increased susceptibility to many diseases including cancer, autoimmune disorders, and other chronic inflammatory diseases. Despite progressed understanding of immunosenescence, its detailed mechanisms are still not fully understood. With advances in medicine, the population of older cancer patients is expected to rapidly increase in the coming years. Cancer immunotherapies, including immune checkpoint inhibitors (ICIs), have been shown to be effective for multiple cancer types, whereas to date, few specific data for elderly individuals have been published. Some systemic reviews have demonstrated that ICIs exhibit similar efficacy in older cancer patients, but they seem to be less effective in very old patients. In addition, toxicities might be more frequently observed in such patients. Here, we provide a summary to better understand immunosenescence and an overview of its relationship with cancer and antitumor immunity, including the efficacy and toxicity of ICIs.


Subject(s)
Immunosenescence , Neoplasms , Aged , Aging , Humans , Immunotherapy , Neoplasms/therapy , Vaccination
17.
Proc Natl Acad Sci U S A ; 116(20): 9999-10008, 2019 05 14.
Article in English | MEDLINE | ID: mdl-31028147

ABSTRACT

PD-1 blockade is a cancer immunotherapy effective in various types of cancer. In a fraction of treated patients, however, it causes rapid cancer progression called hyperprogressive disease (HPD). With our observation of HPD in ∼10% of anti-PD-1 monoclonal antibody (mAb)-treated advanced gastric cancer (GC) patients, we explored how anti-PD-1 mAb caused HPD in these patients and how HPD could be treated and prevented. In the majority of GC patients, tumor-infiltrating FoxP3highCD45RA-CD4+ T cells [effector Treg (eTreg) cells], which were abundant and highly suppressive in tumors, expressed PD-1 at equivalent levels as tumor-infiltrating CD4+ or CD8+ effector/memory T cells and at much higher levels than circulating eTreg cells. Comparison of GC tissue samples before and after anti-PD-1 mAb therapy revealed that the treatment markedly increased tumor-infiltrating proliferative (Ki67+) eTreg cells in HPD patients, contrasting with their reduction in non-HPD patients. Functionally, circulating and tumor-infiltrating PD-1+ eTreg cells were highly activated, showing higher expression of CTLA-4 than PD-1- eTreg cells. PD-1 blockade significantly enhanced in vitro Treg cell suppressive activity. Similarly, in mice, genetic ablation or antibody-mediated blockade of PD-1 in Treg cells increased their proliferation and suppression of antitumor immune responses. Taken together, PD-1 blockade may facilitate the proliferation of highly suppressive PD-1+ eTreg cells in HPDs, resulting in inhibition of antitumor immunity. The presence of actively proliferating PD-1+ eTreg cells in tumors is therefore a reliable marker for HPD. Depletion of eTreg cells in tumor tissues would be effective in treating and preventing HPD in PD-1 blockade cancer immunotherapy.


Subject(s)
Antineoplastic Agents, Immunological/adverse effects , Nivolumab/adverse effects , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Stomach Neoplasms/immunology , T-Lymphocytes, Regulatory/drug effects , Aged , Animals , CTLA-4 Antigen/metabolism , Disease Progression , Humans , Lymphocytes, Tumor-Infiltrating/drug effects , Male , Mice , Stomach Neoplasms/drug therapy , T-Lymphocytes, Regulatory/metabolism
18.
Gan To Kagaku Ryoho ; 49(9): 907-913, 2022 Sep.
Article in Japanese | MEDLINE | ID: mdl-36156003

ABSTRACT

Cancer immunotherapy has shown efficacy in many types of cancer. However, there are many challenges, such as the difficulty in predicting therapeutic efficacy. In the tumor microenvironment, tumor cells evolve to escape the anti-tumor immune response and have capacity of proliferation, whereas immune cells also evolve along with tumor cells. Elucidating the detailed clonal evolution is helpful for the development of biomarkers for prediction of therapeutic effects and novel therapies. To elucidate clonal evolution in the tumor microenvironment, analyses at the single-cell level, rather than in bulks, is necessary for heterogeneous and highly diverse cell populations. Recently, single-cell sequencing can be used to analyze comprehensive gene expression, or it is possible to focus on specific regions, such as T-cell or B-cell receptor sequences. In addition, technologies have been developed that allow spatial analyses by a single-cell level while preserving tissue location information. Recently, new findings have been clarified using pre- and post-treatment samples from same patients to analyze the clonal progression of the tumor cells themselves and immune cells based on sequential changes in the tumor microenvironment.


Subject(s)
Neoplasms , Tumor Microenvironment , Biomarkers, Tumor , Clonal Evolution/genetics , Humans , Immunotherapy , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/therapy , Receptors, Antigen, B-Cell
19.
Int Immunol ; 32(4): 273-281, 2020 04 12.
Article in English | MEDLINE | ID: mdl-31867666

ABSTRACT

Biliary tract cancer (BTC) is an aggressive cancer with a poor prognosis partially due to the limited success in developing novel therapies, including molecularly targeted therapies and immunotherapies. Programmed cell death-1 (PD-1) blockade therapy is less effective against BTCs, necessitating further studies to understand the detailed immunological status of the tumor microenvironment (TME) in BTC. Here, we examined the immunological status of the TME in 37 BTCs with early- to late-stage disease, especially focusing on PD-1+CD8+ T cells. PD-1+CD8+ T cells, which are reportedly associated with the clinical response to PD-1 blockade therapy, were frequently observed in early-stage BTC and decreased with disease progression. Imaging mass cytometry for representative PD-1+CD8+TIL-high and -low patients demonstrated that tumor-infiltrating PD-1+CD8+ T cells were localized adjacent to tumor cells, whereas PD-1-CD8+ T cells were detected mainly in the stroma of the TME. In a mouse model, PD-1 expression by tumor-infiltrating CD8+ T cells was higher in smaller tumors and decreased with tumor growth. Consequently, large tumors became resistant to PD-1 blockade, while small tumors containing higher numbers of PD-1+CD8+ T cells were sensitive. We propose the important role of tumor-infiltrating PD-1+CD8+ T cells in anti-tumor immunity and the potential application of PD-1 blockade therapy for early-stage BTC.


Subject(s)
Biliary Tract Neoplasms/immunology , Biliary Tract Neoplasms/therapy , CD8-Positive T-Lymphocytes/immunology , Immunotherapy , Neoplasms, Experimental/immunology , Programmed Cell Death 1 Receptor/immunology , Aged , Aged, 80 and over , Animals , Biliary Tract Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Female , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Tumor Microenvironment/immunology
20.
BMC Cancer ; 20(1): 336, 2020 Apr 20.
Article in English | MEDLINE | ID: mdl-32312286

ABSTRACT

BACKGROUND: The standard treatment for patients with unresectable locally advanced esophageal squamous cell carcinoma (ESCC) is definitive chemoradiotherapy (CRT) using 5-FU plus cisplatin. However, complete response (CR) rates are low at 11-25%, resulting in 9-10 months of median overall survival (OS). An improved therapeutic efficacy by combining immunotherapy with radiation has been reported in patients with locally advanced non-small cell lung cancer. The results using ESCC cell lines suggest sequential treatment with anti-PD-L1 agents soon after completion of CRT is the most effective combination. METHODS: TENERGY trial is a multicenter, phase II, proof-of-concept study to assess the efficacy and safety of atezolizumab following definitive CRT in patients with locally advanced ESCC. The main inclusion criteria are unresectable locally advanced ESCC without distant metastasis, completion of 60 Gy of radiation plus two concomitant cycles of chemotherapy (cisplatin 70 mg/m2 on day 1 and 5-FU 700 mg/m2 on days 1-4, every 28 days), and adequate organ function. Within 6 weeks after CRT, participants will start taking 1200 mg of atezolizumab every three weeks and continue until 12 months or disease progression. The primary endpoint is the confirmed CR rate by the investigator's assessment. Secondary endpoints include overall response rate, progression-free survival (PFS), OS, adverse events, and confirmed CR rate by central assessment. We will enroll 50 patients (40 with primary locally advanced ESCC and 10 with postoperative locoregionally recurrent ESCC). We will obtain biopsies from the primary site and will collect blood at 3 time points (before CRT, after CRT, and four weeks after the start of atezolizumab) for an exploratory biomarker study. We will analyze the phenotype of immune-competent cells, neoantigens, tumor mutational burden, PD-L1 status, and Human Leukocyte Antigen haplotyping. DISCUSSION: The synergistic efficacies of the sequential combination of CRT and atezolizumab should improve the CR rate, resulting in survival improvement for patients with unresectable locally advanced ESCC. Because CRT is a standard treatment option for patients with early stage to locally advanced ESCC, the sequential combination of CRT and atezolizumab has the potential to change the standard ESCC treatments. TRIAL REGISTRATION: UMIN000034373, 10/04/2018 and EPOC1802.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Chemoradiotherapy/mortality , Esophageal Neoplasms/therapy , Esophageal Squamous Cell Carcinoma/therapy , Neoadjuvant Therapy/mortality , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized/administration & dosage , Cisplatin/administration & dosage , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/pathology , Female , Fluorouracil/administration & dosage , Follow-Up Studies , Humans , Male , Middle Aged , Prognosis , Survival Rate , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL