Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Immunity ; 42(5): 929-41, 2015 May 19.
Article in English | MEDLINE | ID: mdl-25992863

ABSTRACT

It has long been thought that clonal deletion efficiently removes almost all self-specific T cells from the peripheral repertoire. We found that self-peptide MHC-specific CD8(+) T cells in the blood of healthy humans were present in frequencies similar to those specific for non-self antigens. For the Y chromosome-encoded SMCY antigen, self-specific T cells exhibited only a 3-fold lower average frequency in males versus females and were anergic with respect to peptide activation, although this inhibition could be overcome by a stronger stimulus. We conclude that clonal deletion prunes but does not eliminate self-specific T cells and suggest that to do so would create holes in the repertoire that pathogens could readily exploit. In support of this hypothesis, we detected T cells specific for all 20 amino acid variants at the p5 position of a hepatitis C virus epitope in a random group of blood donors.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Clonal Deletion , Animals , Antigenic Variation , Female , Flow Cytometry , Humans , Male , Mice , Receptors, Antigen, T-Cell, alpha-beta/genetics , Self Tolerance/immunology
2.
Gan To Kagaku Ryoho ; 50(5): 577-583, 2023 May.
Article in Japanese | MEDLINE | ID: mdl-37218315

ABSTRACT

Traditionally, the 3 major standard treatments for cancers-surgery, chemotherapy, and radiation therapy-have been applied and have saved many lives. However, malignancies have been the leading cause of death in Japan for more than 40 years since 1981, and the trend is still accelerating. According to the statistics report from the Ministry of Health, Labour and Welfare, cancers accounted for 26.5% of all deaths in 2021, which means that about 1 in 3.5 of all deaths in Japan was due to cancers. Additionally, medical expenditure spent on the diagnosis and treatments for cancer patients have significantly increased, contributing to the pressure on the Japanese economy. Therefore, there is a demand to develop the novel technologies concerning diagnostic methods, effective treatment, and recurrence prevention of cancers. Chimeric antigen receptor(CAR)-T cell therapy has attracted much attention as the next generation of cancer immunotherapy following immune checkpoint blockade therapy, which was the subject of the 2018 Nobel Prize in Physiology or Medicine. CAR-T cell therapy was first approved in the United States in 2017, followed by the EU in 2018 and Japan in March 2019, after having demonstrated significant therapeutic efficacies against B-cell malignancies in clinical trials. However, current CAR-T cell therapies are not yet complete, and there still remain challenges to be overcome. In particular, it is one of the most important issues that current CAR-T cell therapies do not work effectively against solid cancers, which make up the majority of malignant tumors. This review provides an overview of the development toward establishing the next generation CAR-T cell therapy with therapeutic potential against solid cancers.


Subject(s)
Neoplasms , Receptors, Chimeric Antigen , Humans , Neoplasms/therapy , Immunotherapy, Adoptive , Immunotherapy , T-Lymphocytes , Tumor Microenvironment
3.
Cancer Sci ; 113(12): 4020-4029, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36047968

ABSTRACT

The three major standard therapies, that is, surgery, chemotherapy, and radiation therapy have conventionally been applied to the treatments for cancers and have saved many patients. In addition, for intractable, refractory, or advanced malignancies that cannot be cured by the three standard therapies, immunotherapy is an important subject of basic and clinical researches. Immune checkpoint inhibitor therapy (ICI) has shown significant therapeutic efficacies on some types of tumors in large-scale randomized clinical trials, making a major impact on clinical oncology by scientifically proving and establishing the effectiveness of an immunotherapy. In 2018, ICI was awarded the Nobel Prize in Physiology or Medicine, and immunotherapy is now becoming the "fourth" standard therapy for cancers. Recently, adoptive cell therapies, in which genetically modified T cells with enhanced reactivity against tumors are infused into the patients, have been attracting considerable attention as a hopeful immunotherapy following ICI. Particularly, chimeric antigen receptor (CAR)-T-cell therapies demonstrate marked therapeutic efficacies against some hematologic malignancies, and have been approved in many countries. However, current CAR-T-cell therapy is considered to be little effective against solid tumors, which is one of the challenging issues to be overcome in CAR-T-cell therapy. In this review, we at first introduce CAR and CAR-T cell, and then focus on the recent progress of CAR-T-cell therapy against solid tumors as well as the novel concept on a role of CAR-T cells, aiming to further understandings of the novel cancer immunotherapies.


Subject(s)
Neoplasms , Receptors, Chimeric Antigen , Humans , Receptors, Antigen, T-Cell/genetics , Immunotherapy, Adoptive , Neoplasms/therapy , Cell- and Tissue-Based Therapy , Tumor Microenvironment
4.
Cancer Sci ; 112(4): 1417-1428, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33539630

ABSTRACT

Chimeric antigen receptor (CAR)-T cell therapy has shown salient efficacy in cancer immunotherapy, particularly in the treatment of B cell malignancies. However, the efficacy of CAR-T for solid tumors remains inadequate. In this study, we displayed that c-met is an appropriate therapeutic target for papillary renal cell carcinoma (PRCC) using clinical samples, developed an anti-human c-met CAR-T cells, and investigated the anti-tumor efficacy of the CAR-T cells using an orthotopic mouse model as pre-clinical research. Administration of the anti-c-met CAR-T cells induced marked infiltration of the CAR-T cells into the tumor tissue and unambiguous suppression of tumor growth. Furthermore, in combination with axitinib, the anti-tumor efficacy of the CAR-T cells was synergistically augmented. Taken together, our current study demonstrated the potential for clinical application of anti-c-met CAR-T cells in the treatment of patients with PRCC.


Subject(s)
Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/therapy , Kidney Neoplasms/immunology , Kidney Neoplasms/therapy , Proto-Oncogene Proteins c-met/immunology , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , Aged , Animals , Antibodies/immunology , Antigens, Neoplasm/immunology , Cell Line, Tumor , Cells, Cultured , Disease Models, Animal , Female , Humans , Immunotherapy/methods , Male , Mice , Mice, Inbred NOD , Xenograft Model Antitumor Assays/methods
5.
Cancer Immunol Immunother ; 70(9): 2503-2515, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33559069

ABSTRACT

Chimeric antigen receptor (CAR)-T cell therapy has impressive efficacy in hematological malignancies, but its application in solid tumors remains a challenge. Multiple hurdles associated with the biological and immunological features of solid tumors currently limit the application of CAR-T cells in the treatment of solid tumors. Using syngeneic mouse models, we recently reported that CAR-T cells engineered to concomitantly produce interleukin (IL)-7 and chemokine (C-C motif) ligand 19 (CCL19)-induced potent anti-tumor efficacy against solid tumors through an improved ability of migration and proliferation even in an immunosuppressive tumor microenvironment. In this study, for a preclinical evaluation preceding clinical application, we further explored the potential of IL-7/CCL19-producing human CAR-T cells using models that mimic the clinical features of solid tumors. Human anti-mesothelin CAR-T cells producing human IL-7/CCL19 achieved complete eradication of orthotopic pre-established malignant mesothelioma and prevented a relapse of tumors with downregulated antigen expression. Moreover, mice with patient-derived xenograft of mesothelin-positive pancreatic cancers exhibited significant inhibition of tumor growth and prolonged survival following treatment with IL-7/CCL19-producing CAR-T cells, compared to treatment with conventional CAR-T cells. Transfer of IL-7/CCL19-producing CAR-T cells resulted in an increase in not only CAR-T cells but also non-CAR-T cells within the tumor tissues and downregulated the expression of exhaustion markers, including PD-1 and TIGIT, on the T cells. Taken together, our current study elucidated the exceptional anti-tumor efficacy of IL-7/CCL19-producing human CAR-T cells and their potential for clinical application in the treatment of patients with solid tumors.


Subject(s)
Chemokine CCL19/metabolism , Immunotherapy, Adoptive , Interleukin-7/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Cell Line, Tumor , Chemokine CCL19/genetics , Disease Models, Animal , Female , Humans , Immunophenotyping , Immunotherapy, Adoptive/methods , Interleukin-7/genetics , Mesothelin , Mesothelioma, Malignant/etiology , Mesothelioma, Malignant/pathology , Mesothelioma, Malignant/therapy , Mice , Mice, Knockout , Receptors, Chimeric Antigen , Recurrence , T-Lymphocytes/pathology , Treatment Outcome , Xenograft Model Antitumor Assays
6.
Cancer Sci ; 110(10): 3079-3088, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31432594

ABSTRACT

Chimeric antigen receptor-engineered T (CAR-T)-cell therapy holds significant promise for the treatment of hematological malignancies, especially for B-cell leukemia and lymphoma. However, its efficacy against non-hematological malignancies has been limited as a result of several biological problems characteristic of the tumor microenvironment of solid tumors. One of the main hurdles is the heterogeneous nature of tumor-associated antigens (TAA) expressed in solid tumors. Another hurdle is the inefficient activation and limited persistence of CAR-T cells, mainly as a result of T-cell exhaustion caused by immunosuppressive factors in the tumor microenvironment. In the present study, to address these problems, we engineered CAR-T cells to produce antagonistic anti-programmed cell death protein 1 (PD-1) single-chain variable fragment (scFv), by which PD-1-dependent inhibitory signals in CAR-T cells and adjacent tumor-specific non-CAR-T cells are attenuated. In mouse solid tumor models, PD-1 scFv-producing CAR-T cells induced potent therapeutic effects superior to those of conventional CAR-T cells, along with a significant reduction of apoptotic cell death not only in CAR-T cells themselves but also in TAA-specific T cells in the tumor tissue. In addition, the treatment with anti-PD-1 scFv-producing CAR-T cells resulted in an increased concentration of PD-1 scFv in tumor tissue but not in sera, suggesting an induction of less severe systemic immune-related adverse events. Hence, the present study developed anti-PD-1 scFv-producing CAR-T cell technology and explored its cellular mechanisms underlying potent antitumor efficacy.


Subject(s)
Immunotherapy, Adoptive/methods , Neoplasms/therapy , Programmed Cell Death 1 Receptor/immunology , Receptors, Chimeric Antigen/metabolism , Single-Chain Antibodies/metabolism , Animals , Apoptosis , Cell Line, Tumor , Coculture Techniques , Male , Mice , Neoplasms/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Xenograft Model Antitumor Assays
7.
Cancer Immunol Immunother ; 68(2): 201-211, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30357491

ABSTRACT

Although a role of PD-L1 in the suppression of anti-tumor immunity and its value as a predictive biomarker has been suggested by various preclinical and clinical studies, the precise mechanisms how PD-L1 and PD-L2, another ligand of PD-1, regulate anti-tumor immunity in the tumor microenvironment are yet to be fully explored. Here, we address this issue using PD-L1-deficient tumor cells, PD-L1-knockout (KO) mice, anti-PD-L1 monoclonal antibody (mAb), and anti-PD-L2 mAb. Firstly, PD-L1-deficient or competent tumor cells were inoculated into wild-type or PD-L1-KO mice. Results of tumor growth and mouse survival indicated that both tumor- and host-derived PD-L1 are functional to suppress anti-tumor immunity, while the former contributes predominantly than the latter. Experiments using bone marrow (BM) chimeric mice, generated by transferring PD-L1-KO BM cells into wild-type mice or vice versa, further suggested that PD-L1 expressed on BM-derived hematopoietic cells mediates the suppressive effects on anti-tumor immunity. Secondly, anti-PD-L2 mAb treatment demonstrated a profound synergy with anti-PD-L1 mAb therapy, whereas anti-PD-L2 mAb alone hardly induced any anti-tumor effects, suggesting that PD-L2's function becomes evident when the effects of PD-L1 are abrogated by anti-PD-L1 mAb. Consistent with this notion, PD-L2 expression was upregulated on tumor-associated macrophages (TAM) when mice were treated with anti-PD-L1 mAb. Taken together, our study elucidated the importance of PD-L1 associated with tumor cells and non-tumor host cells, particularly BM-derived hematopoietic cells, as well as PD-L2 inducibly expressed on TAM in the suppression of anti-tumor immunity in the tumor microenvironment.


Subject(s)
B7-H1 Antigen/immunology , Neoplasms, Experimental/immunology , Programmed Cell Death 1 Ligand 2 Protein/immunology , Tumor Microenvironment/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Bone Marrow Cells/immunology , Cell Line, Tumor , Mice, Inbred C57BL , Mice, Knockout , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Programmed Cell Death 1 Ligand 2 Protein/genetics , Programmed Cell Death 1 Ligand 2 Protein/metabolism , Survival Analysis , Tumor Burden/drug effects , Tumor Burden/genetics , Tumor Burden/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics
8.
Cancer Sci ; 107(4): 398-406, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27079438

ABSTRACT

Therapeutic cancer vaccines are designed to treat cancer by boosting the endogenous immune system to fight against the cancer. In the development of clinically effective cancer vaccines, one of the most practical objectives is to identify adjuvants that are capable of optimizing the vaccine effects. In this study, we explored the potential of polyinosinic-polycytidylic acid (poly(I:C)) and LAG-3-Ig (soluble recombinant protein of lymphocyte activation gene-3 [LAG-3] extracellular domain fused with human IgG Fc region) as adjuvants for P1A tumor antigen peptide vaccine in a pre-established P815 mouse tumor model with a transfer of tumor-specific T cells. Whereas the use of poly(I:C) or LAG-3-Ig as a signal adjuvant induced a slight enhancement of P1A vaccine effects compared to incomplete Freund's adjuvant, combined treatment with poly(I:C) plus LAG-3-Ig remarkably potentiated antitumor effects, leading to complete rejection of pre-established tumor and long-term survival of mice. The potent adjuvant effects of poly(I:C) plus LAG-3-Ig were associated with an enhanced infiltration of T cells in the tumor tissues, and an increased proliferation and Th1-type cytokine production of tumor-reactive T cells. Importantly, the combined adjuvant of poly(I:C) plus LAG-3-Ig downregulated expressions of PD-1, LAG-3, and TIGIT on P1A-specific T cells, indicating prevention of T cell exhaustion. Taken together, the results of the current study show that the combined adjuvants of poly(I:C) plus LAG-3-Ig with tumor peptide vaccine induce profound antitumor effects by activating tumor-specific T cells.


Subject(s)
Antigens, CD/immunology , Cancer Vaccines/immunology , Immunoglobulin G/immunology , Poly I-C/administration & dosage , Adjuvants, Immunologic/administration & dosage , Animals , Antigens, CD/administration & dosage , Antigens, CD/genetics , CD8-Positive T-Lymphocytes/drug effects , Humans , Immunoglobulin G/administration & dosage , Lymphocyte Activation/drug effects , Mice , Mice, Inbred C57BL , Poly I-C/immunology , Lymphocyte Activation Gene 3 Protein
9.
Gan To Kagaku Ryoho ; 43(2): 143-50, 2016 Feb.
Article in Japanese | MEDLINE | ID: mdl-27067677

ABSTRACT

Since 1981, cancer has been the leading cause of death in Japan, and its upward trend remains continuing until now. In 2013, approximately 365,000 Japanese died of cancer, and the ratio of the number of cancer-related deaths to the total number of deaths was 28.8%, implying that approximately 1 out of 3.5 deaths in Japan are caused by cancer. In recent years, immunotherapies are rapidly developed and recognized as an important anti-cancer treatment modality in addition to surgery, chemotherapy, and radiotherapy. In particular, the"immune checkpoint blockade therapy"has been attracting tremendous attention because of its potent clinical efficacy in the treatment of advanced cancers. The fundamental mechanism of the therapy is the induction of anti-tumor immune responses by disrupting the immunosuppressive mechanisms in the tumor milieu. Antibodies for the immune checkpoint blockade have been approved as clinical drugs in some countries, including in Japan, and a large number of clinical trials are currently in progress to further expand its application. However, several critical issues remain, one of which is the identification of biomarkers associated with clinical benefits or adverse events. In this review, we discuss recently reported genome-based biomarkers predictive for clinical benefits of immune checkpoint blockade therapies, namely activation of b-catenin signaling pathway and DNA mismatch repair deficiency.


Subject(s)
Genome, Human , Immunotherapy , Neoplasms/genetics , Neoplasms/therapy , DNA Mismatch Repair , Genetic Markers/genetics , Humans , Neoplasms/immunology , Signal Transduction , beta Catenin/metabolism
10.
Cancer Sci ; 106(8): 945-50, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25981182

ABSTRACT

Recent progress in tumor immunology has revealed that tumors generate immunologically restrained milieu during the process of their growth, which facilitates the escape of tumors from host immune systems. Immune checkpoint molecules, which transduce co-inhibitory signals to immuno-competent cells, are one of the most important components conferring the immunosuppressive capacity in the tumor microenvironment. Cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death-1 (PD-1) are typical immune checkpoint molecules intimately involved in the suppression of anti-tumor immunity. Antibodies against those molecules have been developed, such as ipilimumab (anti-CTLA-4 antibody), nivolumab and pembrolizumab (anti-PD-1 antibody), and have been approved by regulatory agencies and used in some countries. Treatment with these antibodies demonstrates previously unobserved clinical efficacies superior to the conventional therapies. In this review, we first discuss the escape mechanisms of cancer from host immune systems, and then focus on the recent advances in immune checkpoint blockade therapy and on the new findings of related immune reactions, aiming to provide a better understanding of the novel cancer immunotherapies.


Subject(s)
Immunotherapy/methods , Neoplasms/drug therapy , Neoplasms/immunology , Tumor Escape/immunology , Animals , Humans
11.
Proc Natl Acad Sci U S A ; 108(4): 1549-54, 2011 Jan 25.
Article in English | MEDLINE | ID: mdl-21205892

ABSTRACT

Naïve T lymphocytes display weaker and slower responses than antigen-experienced cells for reasons that are not well understood. Here we show that T-cell receptor (TCR) stimulation induces distinct ERK and p38 phosphorylation patterns in naïve and antigen-experienced human T cells, and that these contribute to the differential responses shown by these cells. Specifically, TCR ligation triggers the activation of the ERK pathway in naïve cells. This phosphorylation of ERK attenuates subsequent calcium influx and accelerates the degradation of the signalsome. In contrast, anti-CD3 stimulation of experienced cells results in the phosphorylation of p38 via an association with Discs large (Dlg). Thus, there are distinct signaling pathways triggered by TCR ligation that impair signaling in naïve cells and facilitate it in antigen-experienced cells.


Subject(s)
Antigens/immunology , Receptors, Antigen, T-Cell/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Adaptor Proteins, Signal Transducing/metabolism , Blotting, Western , Butadienes/pharmacology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Discs Large Homolog 1 Protein , Enterotoxins/immunology , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Imidazoles/pharmacology , Interferon-gamma/metabolism , Lymphocyte Activation/immunology , Membrane Proteins/metabolism , Nitriles/pharmacology , Phosphorylation/drug effects , Pyridines/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
12.
Cancer Med ; 12(11): 12569-12580, 2023 06.
Article in English | MEDLINE | ID: mdl-37031457

ABSTRACT

BACKGROUND: While chimeric antigen receptor (CAR)-T cell therapy has demonstrated excellent efficacy in hematopoietic malignancies, its clinical application in solid cancers has yet to be achieved. One of the reasons for such hurdle is a lack of suitable CAR targets in solid cancers. METHODS: GM2 is one of the gangliosides, a group of glycosphingolipids with sialic acid in the glycan, and overexpressed in various types of solid cancers. In this study, by using interleukin (IL)-7 and chemokine (C-C motif) ligand 19 (CCL19)-producing human CAR-T system which we previously developed, a possibility of GM2 as a solid tumor target for CAR-T cell therapy was explored in a mouse model with human small-cell lung cancer. RESULTS: Treatment with anti-GM2 IL-7/CCL19-producing CAR-T cells induced complete tumor regression along with an abundant T cell infiltration into the solid tumor tissue and long-term memory responses, without any detectable adverse events. In addition, as measures to control cytokine-release syndrome and neurotoxicity which could occur in association with clinical use of CAR-T cells, we incorporated Herpes simplex virus-thymidine kinase (HSV-TK), a suicide system to trigger apoptosis by administration of ganciclovir (GCV). HSV-TK-expressing anti-GM2 IL-7/CCL19-producing human CAR-T cells were efficiently eliminated by GCV administration in vivo. CONCLUSIONS: Our study revealed the promising therapeutic efficacy of anti-GM2 IL-7/CCL19-producing human CAR-T cells with an enhanced safety for clinical application in the treatment of patients with GM2-positive solid cancers.


Subject(s)
Neoplasms , Receptors, Chimeric Antigen , Mice , Animals , Humans , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , Interleukin-7/metabolism , Heterografts , Neoplasms/metabolism , T-Lymphocytes , Chemokine CCL19/metabolism
13.
Mol Cancer Ther ; 21(1): 138-148, 2022 01.
Article in English | MEDLINE | ID: mdl-34675119

ABSTRACT

Although adoptive transfer of T cells genetically engineered to express chimeric antigen receptor (CAR) or T-cell receptor (TCR) has been actively developed and applied into clinic recently, further improvement of these modalities is highly demanded, especially in terms of its efficacy. Because we previously revealed the profound enhancement of antitumor effects of CAR T cells by concomitant expression of IL7 and CCL19, this study further explored a potential of IL7/CCL19 production technology to augment antitumor effects of TCR T cells. IL7/CCL19-producing P1A tumor antigen-specific TCR T cells (7 × 19 P1A T cells) demonstrated significantly improved antitumor effects, compared with those without IL7/CCL19 production, and generated long-term memory responses. The antitumor effects of 7×19 P1A T cells were further upregulated by combination with anti-PD-1 antibody, in which blockade of PD-1 signal in both 7×19 P1A T cells and endogenous T cells plays an important role. Taken together, our study demonstrated that concomitant production of IL7 and CCL19 by genetically engineered tumor-reactive T cells could synergize with PD-1 blockade therapy to generate potent and long-lasting antitumor immunity.


Subject(s)
Chemokine CCL19/metabolism , Genetic Engineering/methods , Interleukin-7/metabolism , Receptors, Antigen, T-Cell/metabolism , Animals , Cell Line, Tumor , Female , Humans , Male , Mice
14.
J Agric Food Chem ; 68(8): 2547-2553, 2020 Feb 26.
Article in English | MEDLINE | ID: mdl-31995978

ABSTRACT

Anticholinergic organophosphate (OP) agents act on the diverse serine hydrolases, thereby revealing unexpected biological effects. Epidemiological studies indicate a relationship between the OP exposure and development of attention-deficit/hyperactivity disorder (ADHD)-like symptoms, whereas no plausible mechanism for the OP-induced ADHD has been established. The present investigation employs ethyl octylphosphonofluoridate (EOPF) as an OP-probe, which is an extremely potent inhibitor of endocannabinoid (EC, anandamide and 2-arachidonoylglycerol)-hydrolyzing enzymes: that is, fatty acid amide hydrolase (FAAH) and monoacylglycerol lipase (MAGL). An ex vivo experiment shows that EOPF treatment decreases FAAH and MAGL activities and conversely increases EC levels in the rat brain. Subsequently, EOPF (treated intraperitoneally once at 0, 1, 2, or 3 mg/kg) clearly induces ADHD-like behaviors (in elevated plus-maze test) in both Wistar and spontaneously hypertensive rats. The EOPF-induced behaviors are reduced by a concomitant administration of cannabinoid receptor inverse agonist SLV-319. Accordingly, the EC system is a feasible target for OP-caused ADHD-like behaviors in adolescent rats.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Attention Deficit Disorder with Hyperactivity/enzymology , Brain/enzymology , Endocannabinoids/metabolism , Enzyme Inhibitors/adverse effects , Monoacylglycerol Lipases/antagonists & inhibitors , Organophosphorus Compounds/adverse effects , Amidohydrolases/metabolism , Animals , Attention Deficit Disorder with Hyperactivity/chemically induced , Attention Deficit Disorder with Hyperactivity/metabolism , Attention Deficit Disorder with Hyperactivity/psychology , Brain/drug effects , Brain/metabolism , Humans , Male , Monoacylglycerol Lipases/metabolism , Rats , Rats, Wistar
15.
J Gastroenterol ; 53(9): 999-1005, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30003334

ABSTRACT

Three major standard treatments, i.e., surgery, chemotherapy, and radiotherapy, were traditionally applied to the treatment of cancer and saved many patients. Meanwhile, clinical studies as well as basic research of immunotherapy are being actively conducted for intractable or advanced malignancies that cannot be cured by the conventional standard treatments. Remarkable therapeutic efficacies have been recently reported in clinical trials on some cancer types, and immunotherapy is now being recognized as the "fourth" standard therapy against cancer. In particular, immune checkpoint inhibitor therapy (ICI) has demonstrated the effectiveness of immunotherapy through large-scale randomized clinical trials, leading to the paradigm-shift in cancer treatment. Immune checkpoint molecules transduce co-inhibitory signals to immunocompetent cells including T cells, and crucially contribute to the formation of an immunosuppressive microenvironment in tumor tissues, which intrinsically confers the treatment resistance. Programmed death-1 (PD-1, CD279) is one of the typical immune checkpoint molecules. Anti-tumor therapies targeting PD-1 and its ligands had been developed and approved in many countries, and various studies utilizing clinical specimens are currently progressing. In this review, we provide an overview of the biomarkers based on the analysis of enteric microbiota that correlate with the clinical efficacy/inefficacy of PD-1-based therapy.


Subject(s)
Biomarkers, Tumor/immunology , Gastrointestinal Microbiome/immunology , Immunotherapy/methods , Molecular Targeted Therapy/methods , Neoplasms/immunology , Neoplasms/therapy , Animals , Antibodies, Monoclonal/therapeutic use , Fecal Microbiota Transplantation/methods , Humans , Immunologic Factors/therapeutic use , Mice , Programmed Cell Death 1 Receptor/immunology , Treatment Outcome
16.
Nat Biotechnol ; 36(4): 346-351, 2018 04.
Article in English | MEDLINE | ID: mdl-29505028

ABSTRACT

Infiltration, accumulation, and survival of chimeric antigen receptor T (CAR-T) cells in solid tumors is crucial for tumor clearance. We engineered CAR-T cells to express interleukin (IL)-7 and CCL19 (7 × 19 CAR-T cells), as these factors are essential for the maintenance of T-cell zones in lymphoid organs. In mice, 7 × 19 CAR-T cells achieved complete regression of pre-established solid tumors and prolonged mouse survival, with superior anti-tumor activity compared to conventional CAR-T cells. Histopathological analyses showed increased infiltration of dendritic cells (DC) and T cells into tumor tissues following 7 × 19 CAR-T cell therapy. Depletion of recipient T cells before 7 × 19 CAR-T cell administration dampened the therapeutic effects of 7 × 19 CAR-T cell treatment, suggesting that CAR-T cells and recipient immune cells collaborated to exert anti-tumor activity. Following treatment of mice with 7 × 19 CAR-T cells, both recipient conventional T cells and administered CAR-T cells generated memory responses against tumors.


Subject(s)
Cell- and Tissue-Based Therapy , Chemokine CCL19/administration & dosage , Interleukin-7/administration & dosage , Receptors, Antigen, T-Cell/administration & dosage , Receptors, Chimeric Antigen/administration & dosage , Allografts , Animals , Cell Line, Tumor , Cell Survival/drug effects , Chemokine CCL19/genetics , Dendritic Cells/immunology , Dendritic Cells/transplantation , Gene Expression Regulation/drug effects , Interleukin-7/genetics , Mice , Receptors, Antigen, T-Cell/immunology , Receptors, Chimeric Antigen/immunology
17.
PLoS Negl Trop Dis ; 11(9): e0005872, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28863133

ABSTRACT

BACKGROUND: Large-scale schistosomiasis control programs are implemented in regions with diverse social and economic environments. A key epidemiological feature of schistosomiasis is its small-scale heterogeneity. Locally profiling disease dynamics including risk factors associated with its transmission is essential for designing appropriate control programs. To determine spatial distribution of schistosomiasis and its drivers, we examined schoolchildren in Kwale, Kenya. METHODOLOGY/PRINCIPAL FINDINGS: We conducted a cross-sectional study of 368 schoolchildren from six primary schools. Soil-transmitted helminths and Schistosoma mansoni eggs in stool were evaluated by the Kato-Katz method. We measured the intensity of Schistosoma haematobium infection by urine filtration. The geometrical mean intensity of S. haematobium was 3.1 eggs/10 ml urine (school range, 1.4-9.2). The hookworm geometric mean intensity was 3.2 eggs/g feces (school range, 0-17.4). Heterogeneity in the intensity of S. haematobium and hookworm infections was evident in the study area. To identify factors associated with the intensity of helminth infections, we utilized negative binomial generalized linear mixed models. The intensity of S. haematobium infection was associated with religion and socioeconomic status (SES), while that of hookworm infection was related to SES, sex, distance to river and history of anthelmintic treatment. CONCLUSIONS/SIGNIFICANCE: Both S. haematobium and hookworm infections showed micro-geographical heterogeneities in this Kwale community. To confirm and explain our observation of high S. haematobium risk among Muslims, further extensive investigations are necessary. The observed small scale clustering of the S. haematobium and hookworm infections might imply less uniform strategies even at finer scale for efficient utilization of limited resources.


Subject(s)
Ancylostomatoidea/isolation & purification , Hookworm Infections/epidemiology , Schistosoma haematobium/isolation & purification , Schistosomiasis haematobia/epidemiology , Adolescent , Animals , Child , Cross-Sectional Studies , Demography , Feces/parasitology , Female , Humans , Islam , Kenya , Linear Models , Male , Parasite Egg Count , Risk Factors , Schools , Social Class , Soil/parasitology , Students/statistics & numerical data
18.
Parasitol Int ; 65(5 Pt B): 520-525, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27080249

ABSTRACT

Entamoeba histolytica is the third leading parasitic cause of man mortality in the world. Infection occurs via ingestion of food or water contaminated with cysts of E. histolytica. Amoebae primarily colonize the intestine. The majority of amoebic infections are asymptomatic, but under some conditions, approximately 4-10% of infections progress to the invasive form of the disease. To better understand the pathogenesis of amoebiasis and the interaction between amoebae and their hosts, the development of suitable animal models is crucial. Pigs, gerbils, cats and mice are used as animal models for the study of amoebiasis in the laboratory. Among these, the most commonly used model is the mouse. In addition to intestinal amoebiasis, we developed a mouse model of liver abscess by inoculating amoeba through portal vein. However, the frequency of successful infection remains low, which is dependent on the conditions of amoebae in the laboratory. As the maintenance of virulent amoebae in the laboratory is unstable, it needs further refinement. This review summarizes mouse models of amoebiasis and the current state of laboratory culture method of amoebae.

19.
Virchows Arch ; 446(6): 663-73, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15906084

ABSTRACT

Natural killer-T (NKT) cells are rich in the liver. However, their involvement in liver injury is not fully understood. We developed here a new murine model of NKT-cell-activation-associated liver injury, and investigated a role of tumor necrosis factor alpha (TNF-alpha) and Fas in pathogenesis. We injected intraperitoneally alpha-galactosylceramide (alpha-GalCer), an NKT-cell stimulant, into D-galactosamine (GalN)-sensitized mice. Survival rate, pathological changes of the liver, and plasma concentrations of cytokines were studied. Alpha-GalCer/GalN administration gave a lethal effect within 7 h, making pathological changes such as massive parenchymal hemorrhage, hepatocyte apoptosis, sinusoidal endothelial cell injury, and close apposition of lymphocytes to apoptotic hepatocytes. Anti-NK1.1 mAb-pretreated mice and Valpha14NKT knock out (KO) mice did not develop liver injury. Tumor necrosis factor-alpha (TNF-alpha) and interferon-gamma (IFN-gamma) were elevated at 4 h in the plasma. These cytokines were produced by hepatic lymphocytes as demonstrated by in vitro stimulation with alpha-GalCer. The lethal effect was suppressed in TNF-alpha KO mice, TNF receptor-1 KO mice, and lpr/lpr (Fas deficient) mice, whereas it was not in IFN-gamma KO mice. These results indicate that the present liver injury is characterized by parenchymal hemorrhage and hepatocyte apoptosis, and mediated by TNF-alpha secretion and direct cytotoxicity of alpha-GalCer-activated NKT cells.


Subject(s)
Galactosylceramides/toxicity , Killer Cells, Natural/immunology , Liver Diseases/immunology , Liver/immunology , Liver/injuries , Animals , Apoptosis/physiology , Chemical and Drug Induced Liver Injury , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Galactosamine/pharmacology , In Situ Nick-End Labeling , Interferon-gamma/blood , Interferon-gamma/deficiency , Interferon-gamma/genetics , Liver/pathology , Liver Diseases/pathology , Mice , Mice, Knockout , Receptors, Tumor Necrosis Factor/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/metabolism
20.
J Vet Med Sci ; 66(8): 945-9, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15353845

ABSTRACT

The glucose uptake activity in Babesia rodhaini and B. microti - infected red blood cell (IRBC) was investigated in mice using 2-deoxy-D-glucose (2DOG) and L-glucose (L-Glc), a non-metabolizable analogue of D-glucose and non-incorporative glucose to non-infected RBC (NRBC), respectively. The uptake activities of both DOG and L-Glc were higher in IRBCs than those in NRBC. The concentration dependent uptake of 2DOG and L-Glc in both IRBC revealed a linear curve, indicating non-transporter mediated uptake. In addition, B. microti IRBC showed higher 2DOG uptake than B. rodhaini IRBC, whereas no difference was observed in L-Glc uptake. These results indicated that some new glucose uptake system, at least two systems, developed in both IRBC. The new systems were sodium independent, non-competitive to L-Glc, and sensitive to temperature. One of two systems had no kinetical difference between B. rodhaini and B. microti IRBC, however another one might have higher uptake activity in B. microti IRBC compared to that in B. rodhaini IRBC.


Subject(s)
Babesia microti , Erythrocytes/metabolism , Glucose/pharmacokinetics , Mice/metabolism , Animals , Erythrocytes/parasitology , Isomerism , Kinetics , Mice/parasitology , Sodium/metabolism , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL