Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 110
Filter
1.
JTO Clin Res Rep ; 5(7): 100683, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39091595

ABSTRACT

Introduction: This study validated real-world pharmacokinetic (PK) data using an established population PK (PopPK) model for atezolizumab in Japanese patients with NSCLC and explored the relationship between PK parameters, effectiveness, and adverse events (AEs) for the 1200 mg once every three weeks regimen. Methods: A subgroup of 262 of 1039 patients from J-TAIL consented to this exploratory research for PK evaluation of atezolizumab monotherapy for unresectable advanced/recurrent NSCLC (August 2018 to October 2019; 197 institutions). We evaluated plasma concentrations before the start of the third cycle of atezolizumab infusion classified into quartiles 1 to 4, their association with effectiveness, and the association between atezolizumab maximum plasma concentrations (Cmax) calculated using the existing PopPK model and AEs of special interest (AESIs). Results: Overall, 175 of 262 patients were included; baseline characteristics were similar to those of patients enrolled in J-TAIL (Eastern Cooperative Oncology Group performance status ≥ 2, 12.0%; age ≥ 75 y, 28.9%; atezolizumab as more than or equal to third-line treatment, 57.5%). Atezolizumab plasma concentrations were similar to previously reported data among Japanese/non-Japanese patients. The overall survival was significantly shorter in patients with lower atezolizumab plasma concentrations in Q1 versus Q2 to Q4, although progression-free survival remained the same. The PK data adequately fit the PopPK model, with the frequency of AESIs increasing as the calculated Cmax at cycle 1 increased. Conclusions: In real-world Japanese patients with unresectable advanced/recurrent NSCLC, PKs were similar to previous reports. Certain patient populations had shorter overall survival, and atezolizumab plasma concentrations in cycle 3 were lower in this population. Elevated Cmax at cycle 1 may be associated with an increased frequency of AESIs.

2.
Cancer Sci ; 115(1): 59-69, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37923388

ABSTRACT

Sinus macrophages in draining lymph nodes (DLNs) are involved in anti-tumor immune reactions. CD169 (Sialoadhesin, Siglec-1) is expressed on sinus macrophages and is considered a surrogate marker for the immunostimulatory phenotype of macrophages. In this study, the significance of sinus macrophages in immunotherapy was evaluated using mouse models. Treatment with anti-programmed death-ligand 1 (PD-L1) antibody suppressed the subcutaneous tumor growth of MC38 and E0771 cells but was not effective against MB49 and LLC tumors. Decreased cytotoxic T-lymphocyte (CTL) infiltration in tumor tissues and CD169 expression in sinus macrophages were observed in MB49 and LLC cells compared to corresponding parameters in MC38 and E0771 cells. The anti-tumor effects of the anti-PD-L1 antibody on MC38 and E0771 cells were abolished when sinus macrophages in DLNs were depleted, suggesting that sinus macrophages are involved in the therapeutic effect of the anti-PD-L1 antibody. Naringin activated sinus macrophages. Naringin inhibited tumor growth in MB49- and LLC-bearing mice but did not affect that in MC38- and E0771-bearing mice. The infiltration of CTLs in tumor tissues and their activation were increased by naringin, and this effect was impaired when sinus macrophages were depleted. Combination therapy with naringin and anti-PD-L1 antibody suppressed MB49 tumor growth. In conclusion, CD169-positive sinus macrophages in DLNs are critical for anti-tumor immune responses, and naringin suppresses tumor growth by activating CD169-positive sinus macrophages and anti-tumor CTL responses. The activation status of sinus macrophages has been suggested to differ among tumor models, and this should be investigated in future studies.


Subject(s)
Antineoplastic Agents , Neoplasms , Animals , Mice , Neoplasms/drug therapy , Antineoplastic Agents/pharmacology , T-Lymphocytes, Cytotoxic/metabolism , Antibodies/therapeutic use , Immunotherapy , Macrophages/metabolism , B7-H1 Antigen/metabolism , Cell Line, Tumor
3.
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
4.
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
5.
Hepatol Res ; 53(7): 649-660, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36929310

ABSTRACT

AIM: Developing effective adjuvant therapies is essential for improving the surgical outcomes in patients with hepatocellular carcinoma (HCC). Immunotherapy against HCC has become a promising strategy; however, only approximately 30% of all HCC patients respond to immunotherapy. Previously, we generated the novel therapeutic vaccine comprising multi-human leukocyte antigen-binding heat shock protein 70/glypican-3 peptides with a novel adjuvant combination of hLAG-3Ig and poly-ICLC. We also confirmed the safety of this vaccination therapy, as well as its capacity for the effective induction of immune responses in a previous clinical trial. METHODS: In this phase I study, we administered this vaccine intradermally six times before surgery, and 10 times after surgery to patients with untreated, surgically resectable HCC (stage II to IVa). The primary end-points of this study were the safety and feasibility of this treatment. We also analyzed the resected tumor specimens pathologically using hematoxylin-eosin staining and immunohistochemistry for heat shock protein 70, glypican 3, CD8 and programmed death-1. RESULTS: A total of 20 human leukocyte antigen-matched patients received this vaccination therapy with an acceptable side-effect profile. All patients underwent planned surgery without vaccination-related delay. Immunohistochemical analyses revealed that potent infiltration of CD8+ T cells into tumors with target antigen expression was observed in 12 of 20 (60%) patients. CONCLUSIONS: This novel therapeutic vaccine was safe as perioperative immunotherapy for patients with HCC, and has the potential to strongly induce CD8+ T cells infiltration into tumors.

6.
Nat Biomed Eng ; 7(1): 24-37, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36509913

ABSTRACT

The effectiveness of chimaeric antigen receptor (CAR) T-cell immunotherapies against solid tumours relies on the accumulation, proliferation and persistency of T cells at the tumour site. Here we show that the proliferation of CD8αß cytotoxic CAR T cells in solid tumours can be enhanced by deriving and expanding them from a single human induced-pluripotent-stem-cell clone bearing a CAR selected for efficient differentiation. We also show that the proliferation and persistency of the effector cells in the tumours can be further enhanced by genetically knocking out diacylglycerol kinase, which inhibits antigen-receptor signalling, and by transducing the cells with genes encoding for membrane-bound interleukin-15 (IL-15) and its receptor subunit IL-15Rα. In multiple tumour-bearing animal models, the engineered hiPSC-derived CAR T cells led to therapeutic outcomes similar to those of primary CD8 T cells bearing the same CAR. The optimization of effector CAR T cells derived from pluripotent stem cells may aid the development of long-lasting antigen-specific T-cell immunotherapies for the treatment of solid tumours.


Subject(s)
Induced Pluripotent Stem Cells , Neoplasms , Animals , Humans , Receptors, Antigen, T-Cell/genetics , Induced Pluripotent Stem Cells/pathology , CD8-Positive T-Lymphocytes , Neoplasms/therapy , Cell Proliferation
7.
Int J Hematol ; 117(5): 647-651, 2023 May.
Article in English | MEDLINE | ID: mdl-36309630

ABSTRACT

Chimeric antigen receptor-T (CAR-T) cell therapy has shown significant therapeutic efficacy in the treatment of hematological B-cell malignancies. However, the efficacy of CAR-T cell therapy against solid tumors is limited due to the heterogeneity of tumor antigens and the immunosuppressive tumor microenvironment. Therefore, there is strong demand for novel technologies to improve the efficacy of CAR-T cell therapy. In addition, as CAR-T cells often cause severe side effects, systems to control the activity of CAR-T cells so as to avoid or lessen the occurrence and intensity of these side effects are needed. Here, we describe recently emerging approaches to enhance and/or regulate CAR-T cell functions. These approaches have led to the development of CAR-T therapies with improved efficacy and safety, which are expected to be clinically applied to a variety of cancer types in combination with other therapies, such as immune checkpoint inhibitors, chemotherapy, molecular targeted drugs, and radiation therapy.


Subject(s)
Hematologic Neoplasms , Neoplasms , Receptors, Chimeric Antigen , Humans , Receptors, Antigen, T-Cell , Immunotherapy, Adoptive/adverse effects , Neoplasms/therapy , Hematologic Neoplasms/drug therapy , Tumor Microenvironment , Cell- and Tissue-Based Therapy
8.
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
9.
Sci Rep ; 12(1): 12007, 2022 07 14.
Article in English | MEDLINE | ID: mdl-35835809

ABSTRACT

Ever since immune checkpoint inhibitors have been approved for anti-cancer therapy in several cancers, including triple-negative breast cancer, the significance of programmed death-1 ligand 1 (PD-L1) expression in the tumor immune microenvironment has been a topic of interest. In the present study, we investigated the detailed mechanisms of PD-L1 overexpression on tumor-associated macrophages (TAMs) in breast cancer. In in vitro culture studies using human monocyte-derived macrophages, lymphocytes, and breast cancer cell lines, PD-L1 overexpression on macrophages was induced by the conditioned medium (CM) of activated lymphocytes, but not that of cancer cells. Granulocyte-macrophage colony-stimulating factor (GM-CSF) derived from activated lymphocytes was found to be involved in PD-L1 overexpression, in addition to interferon (IFN)-γ, via STAT3 pathway activation. Macrophages suppressed lymphocyte activation, and this inhibition was impaired by PD-1 blocking. The CM of activated lymphocytes also induced the overexpression of PD-L2, but GM-CSF did not affect PD-L2 expression. In the murine E0771 breast cancer model, anti-GM-CSF therapy did not affect PD-L1 expression on TAMs, and the mechanisms of PD-L1 expression on TAMs might differ between humans and mice. However, not only PD-L1, but also PD-L2 was overexpressed on TAMs in the E0771 tumor model, and their expression levels were significantly lower in the tumors in nude mice than in wild-type mice. Anti-PD-L1 antibody and anti-PD-L2 antibody synergistically inhibited E0771 tumor development. In conclusion, PD-L1 and PD-L2 were overexpressed on TAMs, and they potentially contributed to immunosuppression. The GM-CSF-STAT3 pathway is thought to represent a new mechanism of PD-L1 overexpression on TAMs in human breast cancer microenvironment.


Subject(s)
Breast Neoplasms , Granulocyte-Macrophage Colony-Stimulating Factor , Animals , B7-H1 Antigen , Female , Humans , Ligands , Mice , Mice, Nude , Tumor Microenvironment , Tumor-Associated Macrophages
10.
Cancer Immunol Immunother ; 71(11): 2645-2661, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35352168

ABSTRACT

Programmed cell death-1 (PD-1) and PD-1 ligand 1 (PD-L1) are target molecules for immunotherapy in non-small cell lung cancer. PD-L1 is expressed not only in cancer cells, but also on macrophages, and has been suggested to contribute to macrophage-mediated immune suppression. We examined the clinical significance of PD-L1 expression on macrophages in human lung adenocarcinoma. The mechanism of PD-L1 overexpression on macrophages was investigated by means of cell culture studies and animal studies. The results showed that high PD-L1 expression on macrophages was correlated with the presence of EGFR mutation, a lower cancer grade, and a shorter cancer-specific overall survival. In an in vitro study using lung cancer cell lines and human monocyte-derived macrophages, the conditioned medium from cancer cells was found to up-regulate PD-L1 expression on macrophages via STAT3 activation, and a cytokine array revealed that granulocyte-macrophage colony-stimulating factor (GM-CSF) was a candidate factor that induced PD-L1 expression. Culture studies using recombinant GM-CSF, neutralizing antibody, and inhibitors indicated that PD-L1 overexpression was induced via STAT3 activation by GM-CSF derived from cancer cells. In a murine Lewis lung carcinoma model, anti-GM-CSF therapy inhibited cancer development via the suppression of macrophage infiltration and the promotion of lymphocyte infiltration into cancer tissue; however, the PD-L1 expression on macrophages remained unchanged. PD-L1 overexpression on macrophages via the GM-CSF/STAT3 pathway was suggested to promote cancer progression in lung adenocarcinoma. Cancer cell-derived GM-CSF might be a promising target for anti-lung cancer therapy.


Subject(s)
Adenocarcinoma of Lung , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Adenocarcinoma of Lung/pathology , Animals , Antibodies, Neutralizing , B7-H1 Antigen/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Culture Media, Conditioned/metabolism , Cytokines/metabolism , ErbB Receptors/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Ligands , Macrophages , Mice , Programmed Cell Death 1 Receptor
11.
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
12.
Oncol Lett ; 22(4): 689, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34457044

ABSTRACT

The interaction between prostate cancer cells and osteoblasts is essential for the development of bone metastasis. Previously, novel androgen receptor axis-targeted agents (ARATs) were approved for metastatic castration-naïve and non-metastatic castration-resistant prostate cancer (CRPC); both of which are pivotal for investigating the association between the bone microenvironment and tumors. The present study established a novel in vitro 3D microenvironment model that simulated the bone microenvironment of CRPC, and evaluated the drug susceptibility of ARATs and the efficacy of the combination of abiraterone and dutasteride. Green fluorescent protein-transferred C4-2 cells (a CRPC cell line) and red fluorescent protein-transferred human osteoblasts differentiated from human mesenchymal stem cells were co-cultured in chitosan nanofiber matrix-coated culture plates to simulate the 3D scaffold of the bone microenvironment. The growth of C4-2 was quantified using live-cell imaging and the Cell3 iMager duos analysis system. The growth of C4-2 colonies were quantified for a maximum of 30 days. The expression of TGF-ß increased and promoted EMT in C4-2 cells co-cultured with osteoblasts, indicating resistance to ARATs. The IC50 of each drug and the combination effect of abiraterone and dutasteride were evaluated using this model. Combination treatment with abiraterone and dutasteride synergistically inhibited the growth of C2-4 colonies compared with individual investigational agents. This could be attributed to the reduction of 3-keto-5α-abiraterone, an androgen receptor agonist. The bone microenvironment model of the present study is unique and useful for evaluating new drug susceptibility testing in prostate cancer cells. This model may help to reveal the unknown mechanisms underlying micro- to clinical bone metastasis in prostate cancer.

13.
Cancers (Basel) ; 13(11)2021 Jun 01.
Article in English | MEDLINE | ID: mdl-34206082

ABSTRACT

Unrelated bone marrow transplantation (uBMT) is performed to treat blood disorders, and it uses bone marrow from an unrelated donor as the transplant source. Although the importance of HLA matching in uBMT has been established, that of other genetic factors, such as single-nucleotide polymorphisms (SNPs), remains unclear. The application of immunoinhibitory receptors as anticancer drugs has recently been attracting attention. This prompted us to examine the importance of immunoinhibitory receptor SNPs in uBMT. We retrospectively genotyped five single-nucleotide polymorphisms (SNPs) in the immune checkpoint genes, BTLA, PD-1, LAG3, and CTLA4, and two SNPs in the methylase genes, DNMT1 and EZH2, in 999 uBMT donor-recipient pairs coordinated through the Japan Marrow Donor Program matched at least at HLA-A, -B, and -DRB1. No correlations were observed between these SNPs and post-uBMT outcomes (p > 0.005). This result questions the usefulness of these immune checkpoint gene polymorphisms for predicting post-BMT outcomes. However, the recipient EZH2 histone methyltransferase gene SNP, which encodes the D185H substitution, exhibited a low p-value in regression analysis of grade 2-4 acute graft-versus-host disease (p = 0.010). Due to a low minor allele frequency, this SNP warrants further investigation in a larger-scale study.

14.
Cancer Res ; 81(17): 4545-4559, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34210751

ABSTRACT

Blood levels of acute-phase protein α1-acid glycoprotein (AGP, orosmucoid) increase in patients with cancer. Although AGP is produced from hepatocytes following stimulation by immune cell-derived cytokines under conditions of inflammation and tumorigenesis, the functions of AGP in tumorigenesis and tumor progression remain unknown. In the present study, we revealed that AGP contributes directly to tumor development by induction of programmed death ligand 1 (PD-L1) expression and IL6 production in macrophages. Stimulation of AGP induced PD-L1 expression in both human monocyte-derived macrophages through STAT1 activation, whereas AGP had no direct effect on PD-L1 expression in tumor cells. AGP also induced IL6 production from macrophages, which stimulated proliferation in tumor cells by IL6R-mediated activation of STAT3. Furthermore, administration of AGP to AGP KO mice phenocopied effects of tumor-associated macrophages (TAM) on tumor progression. AGP decreased IFNγ secretion from T cells and enhanced STAT3 activation in subcutaneous tumor tissues. In addition, AGP regulated PD-L1 expression and IL6 production in macrophages by binding with CD14, a coreceptor for Toll-like receptor 4 (TLR4), and inducing TLR4 signaling. These results provide the first evidence that AGP is directly involved in tumorigenesis by interacting with TAMs and that AGP might be a target molecule for anticancer therapy. SIGNIFICANCE: AGP-mediated suppression of antitumor immunity contributes to tumor progression by inducing PD-L1 expression and IL6 production in TAMs.


Subject(s)
B7-H1 Antigen/metabolism , Macrophages/metabolism , Orosomucoid/metabolism , Tumor-Associated Macrophages/metabolism , Animals , Carcinogenesis , Cell Proliferation , Disease Progression , Enhancer Elements, Genetic , Hepatocytes/metabolism , Immunosuppression Therapy , Interferon-gamma/metabolism , Macrophages/cytology , Membrane Proteins , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Monocytes/cytology , Orosomucoid/genetics , Signal Transduction , Toll-Like Receptor 4/metabolism
15.
J Allergy Clin Immunol ; 148(2): 563-573.e7, 2021 08.
Article in English | MEDLINE | ID: mdl-33581199

ABSTRACT

BACKGROUND: The programmed cell death-1 (PD-1)/programmed death ligand 1 (PD-L1) pathway is known to inhibit the activation of effector CD8+ T cells. However, just how this regulatory pathway is involved in the pathophysiology of CD8+ T-cell-mediated inflammatory skin diseases remains unclear. OBJECTIVE: Our aim was to elucidate the mechanisms by which the PD-1/PD-L1 pathway exerts its regulatory roles in CD8+ T-cell-mediated cutaneous immune responses. METHODS: PD-L1-deficient (Pdl1-/-) mice were used for the murine contact hypersensitivity model. Inflammatory responses such as IFN-γ production from CD8+ T cells in the skin was evaluated by flow cytometry. RESULTS: Compared with wild-type mice, Pdl1-/- mice exhibited exacerbated ear swelling and increased numbers of IFN-γ+ CD8+ T cells in the skin. Adoptive T-cell transfer experiments revealed the involvement of the PD-1/PD-L1 pathway in the elicitation phase of contact hypersensitivity. Bone marrow chimera experiments showed that PD-L1 on radioresistant cells was responsible for this regulatory pathway. Flow cytometric analysis revealed that among the radioresistant cells in the skin, PD-L1 was most highly expressed on mast cells (MCs) before and after elicitation. Administration of anti-PD-L1 blocking antibody during the elicitation phase significantly enhanced ear swelling responses and increased the number of IFN-γ+CD8+ T cells in the skin of wild-type mice, whereas no significant effects were observed in MC-deficient (WBB6F1/J-KitW/KitW-v/J and C57BL/6-KitW-sh/W-sh) mice. The high level of expression of PD-L1 on human skin MCs was confirmed by database analysis and immunohistochemical analysis. CONCLUSION: PD-L1 on MCs negatively regulates CD8+ T-cell activation in the skin.


Subject(s)
B7-H1 Antigen/immunology , CD8-Positive T-Lymphocytes/immunology , Dermatitis, Contact/immunology , Lymphocyte Activation , Skin/immunology , Animals , B7-H1 Antigen/genetics , CD8-Positive T-Lymphocytes/pathology , Dermatitis, Contact/genetics , Dermatitis, Contact/pathology , Mice , Mice, Knockout , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Skin/pathology
16.
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
17.
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
18.
Cancer Sci ; 111(5): 1478-1490, 2020 May.
Article in English | MEDLINE | ID: mdl-32133731

ABSTRACT

The use of allogeneic, pluripotent stem-cell-derived immune cells for cancer immunotherapy has been the subject of recent clinical trials. In Japan, investigator-initiated clinical trials will soon begin for ovarian cancer treatment using human leukocyte antigen (HLA)-homozygous-induced pluripotent stem cell (iPSC)-derived anti-glypican-3 (GPC3) chimeric antigen receptor (CAR)-expressing natural killer/innate lymphoid cells (NK/ILC). Using pluripotent stem cells as the source for allogeneic immune cells facilitates stringent quality control of the final product, in terms of efficacy, safety and producibility. In this paper, we describe our methods for the stable, feeder-free production of CAR-expressing NK/ILC cells from CAR-transduced iPSC with clinically relevant scale and materials. The average number of cells that could be differentiated from 1.8-3.6 × 106 iPSC within 7 weeks was 1.8-4.0 × 109 . These cells showed stable CD45/CD7/CAR expression, effector functions of cytotoxicity and interferon gamma (IFN-γ) production against GPC3-expressing tumor cells. When the CAR-NK/ILC cells were injected into a GPC3-positive, ovarian-tumor-bearing, immunodeficient mouse model, we observed a significant therapeutic effect that prolonged the survival of the animals. When the cells were injected into immunodeficient mice during non-clinical safety tests, no acute systemic toxicity or tumorigenicity of the final product or residual iPSC was observed. In addition, our test results for the CAR-NK/ILC cells generated with clinical manufacturing standards are encouraging, and these methods should accelerate the development of allogeneic pluripotent stem cell-based immune cell cancer therapies.


Subject(s)
Glypicans/immunology , Induced Pluripotent Stem Cells/immunology , Killer Cells, Natural/immunology , Lymphocytes/immunology , Receptors, Chimeric Antigen/immunology , Animals , Cell Differentiation , Cell Survival , Cytotoxicity, Immunologic , Disease Models, Animal , Female , Glypicans/genetics , Glypicans/metabolism , Humans , Immunity, Innate , Immunotherapy, Adoptive , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Interferon-gamma/immunology , Killer Cells, Natural/cytology , Killer Cells, Natural/transplantation , Lymphocyte Transfusion , Lymphocytes/cytology , Mice , Mice, SCID , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Ovarian Neoplasms/therapy , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism
19.
Cancer Immunol Immunother ; 69(8): 1651-1662, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32219501

ABSTRACT

BACKGROUND: This phase I study aimed to evaluate the safety, peptide-specific immune responses, and anti-tumor effects of a novel vaccination therapy comprising multi-HLA-binding heat shock protein (HSP) 70/glypican-3 (GPC3) peptides and a novel adjuvant combination of hLAG-3Ig and Poly-ICLC against metastatic gastrointestinal cancers. METHODS: HSP70/GPC3 peptides with high binding affinities for three HLA types (A*24:02, A*02:01, and A*02:06) were identified with our peptide prediction system. The peptides were intradermally administered with combined adjuvants on a weekly basis. This study was a phase I dose escalation clinical trial, which was carried out in a three patients' cohort; in total, 11 patients were enrolled for the recommended dose. RESULTS: Seventeen patients received this vaccination therapy without dose-limiting toxicity. All treatment-related adverse events were of grades 1 to 2. Peptide-specific CTL induction by HSP70 and GPC3 proteins was observed in 11 (64.7%) and 13 (76.5%) cases, respectively, regardless of the HLA type. Serum tumor marker levels were decreased in 10 cases (58.8%). Immunological analysis using PBMCs indicated that patients receiving dose level 3 presented with significantly reduced T cell immunoglobulin and mucin-domain containing-3 (TIM3)-expressing CD4 + T cells after one course of treatment. PD-1 or TIM3-expressing CD4 + T cells and T cell immunoreceptor with immunoglobulin and ITIM domains (TIGIT)-expressing CD8 + T cells in PBMCs before vaccination were negative predictive factors for survival. CONCLUSIONS: This novel peptide vaccination therapy was safe for patients with metastatic gastrointestinal cancers.


Subject(s)
Carboxymethylcellulose Sodium/analogs & derivatives , Gastrointestinal Neoplasms/therapy , Glypicans/immunology , HLA-A Antigens/immunology , HLA-G Antigens/administration & dosage , HSP70 Heat-Shock Proteins/immunology , Peptide Fragments/administration & dosage , Poly I-C/administration & dosage , Polylysine/analogs & derivatives , Adjuvants, Immunologic/administration & dosage , Adult , Aged , Aged, 80 and over , Carboxymethylcellulose Sodium/administration & dosage , Cohort Studies , Female , Follow-Up Studies , Gastrointestinal Neoplasms/immunology , Gastrointestinal Neoplasms/metabolism , Gastrointestinal Neoplasms/pathology , Glypicans/metabolism , HLA-A Antigens/metabolism , HSP70 Heat-Shock Proteins/metabolism , Humans , Male , Middle Aged , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/therapy , Peptide Fragments/immunology , Peptide Fragments/metabolism , Polylysine/administration & dosage , Prognosis , Survival Rate
20.
Br J Cancer ; 122(8): 1185-1193, 2020 04.
Article in English | MEDLINE | ID: mdl-32066911

ABSTRACT

BACKGROUND: Cancer peptide vaccines show only marginal effects against cancers. Immune checkpoint inhibitors (ICIs) show significant curative effects in certain types of cancers, but the response rate is still limited. In this study, we aim to improve cancer peptide vaccination by targeting Ag peptides selectively to a dendritic cell (DC) subset, XCR1-expressing DCs (XCR1+ DCs), with high ability to support CD8+ T-cell responses. METHODS: We have generated a fusion protein, consisting of an Ag peptide presented with MHC class I, and an XCR1 ligand, XCL1, and examined its effects on antitumour immunity in mice. RESULTS: The fusion protein was delivered to XCR1+ DCs in an XCR1-dependent manner. Immunisation with the fusion protein plus an immune adjuvant, polyinosinic:polycytidylic acids (poly(I:C)), more potently induced Ag-specific CD8+ T-cell responses through XCR1 than the Ag peptide plus poly(I:C) or the Ag protein plus poly(I:C). The fusion protein plus poly(I:C) inhibited the tumour growth efficiently in the prophylactic and therapeutic tumour models. Furthermore, the fusion protein plus poly(I:C) showed suppressive effects on tumour growth in synergy with anti-PD-1 Ab. CONCLUSIONS: Cancer Ag targeting to XCR1+ DCs should be a promising procedure as a combination anticancer therapy with immune checkpoint blockade.


Subject(s)
Antigens/immunology , Cancer Vaccines/immunology , Chemokines, C/immunology , Cross-Priming/immunology , Dendritic Cells/immunology , Immune Checkpoint Inhibitors/therapeutic use , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Mice, Inbred C57BL , Neoplasms, Experimental/therapy , Poly I-C/pharmacology , Vaccines, Subunit/immunology
SELECTION OF CITATIONS
SEARCH DETAIL