Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Adv Exp Med Biol ; 1444: 207-217, 2024.
Article in English | MEDLINE | ID: mdl-38467982

ABSTRACT

In the field of cancer immunotherapy, the effectiveness of a method in which patient-derived T cells are genetically modified ex vivo and administered to patients has been demonstrated. However, problems remain with this method, such as (1) time-consuming, (2) costly, and (3) difficult to guarantee the quality. To overcome these barriers, strategies to regenerate T cells using iPSC technology are being pursued by several groups in the last decade. The authors have been developing a method by which specific TCR genes are introduced into iPSCs and T cells are generated from those iPSCs (TCR-iPSC method). At present, our group is preparing this approach for clinical trial, where iPSCs provided from the iPSC project are transduced with WT1 antigen-specific TCR that had been already clinically tested, and killer T cells are generated from such TCR-iPSCs, to be administered to acute myeloid leukemia patients. While the adoptive T cell therapies have been mainly directed to be used in cancer immunotherapy, it is possible to apply these approaches to viral infections. Strategies by other groups to regenerate various types of T cells from iPSCs will also be introduced.


Subject(s)
Induced Pluripotent Stem Cells , Leukemia, Myeloid, Acute , Humans , T-Lymphocytes, Cytotoxic , Leukemia, Myeloid, Acute/metabolism , Receptors, Antigen, T-Cell , Cell- and Tissue-Based Therapy , Immunotherapy, Adoptive
2.
Rinsho Ketsueki ; 65(9): 1075-1086, 2024.
Article in Japanese | MEDLINE | ID: mdl-39358263

ABSTRACT

Methods in which patient-derived T cells are genetically modified in vitro and administered to patients have been demonstrated effective in the area of cancer immunotherapy. However, these methods have some unresolved issues such as cost, time, and unstable quality. Several groups have developed strategies to overcome these barriers by regenerating T cells from iPSCs. We have been developing a method in which specific TCR genes are introduced into iPSCs and T cells are regenerated from these iPSCs (TCR-iPSC method). We are now using starting iPSCs from the iPSC stock lines provided by CiRA-F, as the iPSC stock cells are less likely to be rejected. A study aimed at application to solid tumors demonstrated the therapeutic effect of regenerated T cells in a patient tissue xenograft model of WT1 antigen-positive renal cell carcinoma. This article will also discuss strategies by other groups to regenerate various types of T cells from iPSCs.


Subject(s)
Induced Pluripotent Stem Cells , Neoplasms , T-Lymphocytes , Humans , Induced Pluripotent Stem Cells/cytology , Animals , Neoplasms/therapy , Neoplasms/immunology , T-Lymphocytes/immunology , Embryonic Stem Cells/cytology , Virus Diseases/therapy , Virus Diseases/immunology
3.
Gan To Kagaku Ryoho ; 50(5): 561-566, 2023 May.
Article in Japanese | MEDLINE | ID: mdl-37218312

ABSTRACT

In the area of cancer immunotherapy, the efficacy of strategies in which patient derived T cells are genetically modified ex vivo and administered to patients has been demonstrated. However, some issues have remained to be addressed; the method using autologous T cells is costly and time consuming, and their quality is unstable. The time consuming problem can be solved by preparing allogeneic T cells in advance. Peripheral blood is being considered as a source of allogeneic T cells, and methods are being explored to avoid the risk of rejection or GVHD, but even so the issues of cost and quality stability still remain. On the other hand, use of pluripotent stem cells such as iPS cells or ES cells as material of T cells may solve the cost issue and achieve homogeneity of products. The authors group has been developing a method to generate T cells from iPS cells transduced with a certain T cell receptor gene, and is currently preparing for clinical trials. We believe that, when this strategy is realized, it becomes possible to deliver a universal and homogeneous T cell preparation immediately when needed.


Subject(s)
Induced Pluripotent Stem Cells , Neoplasms , Humans , T-Lymphocytes , Immunotherapy , Genes, T-Cell Receptor , Neoplasms/therapy , Immunotherapy, Adoptive
4.
Int Immunol ; 33(12): 827-833, 2021 11 25.
Article in English | MEDLINE | ID: mdl-34661676

ABSTRACT

In currently ongoing adoptive T-cell therapies, T cells collected from the patient are given back to the patient after ex vivo cell activation and expansion. In some cases, T cells are transduced with chimeric antigen receptor (CAR) or T-cell receptor (TCR) genes during the ex vivo culture period. Although such strategies have been shown to be effective in some types of cancer, there remain issues to be solved; these methods (i) are time-consuming, (ii) are costly and (iii) it is difficult to guarantee the quality because the products depend on patient-derived T cells. To address these issues, several groups including ours have developed methods in which cytotoxic cells are mass-produced by using induced pluripotent stem cell (iPSC) technology. For the regeneration of T cells, the basic idea is as follows: iPSCs produced from T cells inherit rearranged TCR genes, and thus all regenerated T cells should express the same TCR. Based on this idea, various types of T cells have been regenerated, including conventional cytotoxic T lymphocytes (CTLs), γδT cells, NKT cells and mucosal-associated invariant T (MAIT) cells. On the other hand, any cytotoxic cells can be used as the base cells into which CAR is introduced, and thus iPSC-derived NK cells have been developed. To apply the iPSC-based cell therapy in an allogeneic setting, the authors' group developed a method in which non-T-cell-derived iPSCs are transduced with exogenous TCR genes (TCR-iPSC method). This approach is being prepared for a clinical trial to be realized in Kyoto University Hospital, in which acute myeloid leukemia patients will be treated by the regenerated WT1 antigen-specific CTLs.


Subject(s)
Antigens/immunology , Induced Pluripotent Stem Cells/immunology , T-Lymphocytes/immunology , Animals , Humans
5.
Nature ; 534(7607): 402-6, 2016 06 16.
Article in English | MEDLINE | ID: mdl-27281199

ABSTRACT

Successful treatment of many patients with advanced cancer using antibodies against programmed cell death 1 (PD-1; also known as PDCD1) and its ligand (PD-L1; also known as CD274) has highlighted the critical importance of PD-1/PD-L1-mediated immune escape in cancer development. However, the genetic basis for the immune escape has not been fully elucidated, with the exception of elevated PD-L1 expression by gene amplification and utilization of an ectopic promoter by translocation, as reported in Hodgkin and other B-cell lymphomas, as well as stomach adenocarcinoma. Here we show a unique genetic mechanism of immune escape caused by structural variations (SVs) commonly disrupting the 3' region of the PD-L1 gene. Widely affecting multiple common human cancer types, including adult T-cell leukaemia/lymphoma (27%), diffuse large B-cell lymphoma (8%), and stomach adenocarcinoma (2%), these SVs invariably lead to a marked elevation of aberrant PD-L1 transcripts that are stabilized by truncation of the 3'-untranslated region (UTR). Disruption of the Pd-l1 3'-UTR in mice enables immune evasion of EG7-OVA tumour cells with elevated Pd-l1 expression in vivo, which is effectively inhibited by Pd-1/Pd-l1 blockade, supporting the role of relevant SVs in clonal selection through immune evasion. Our findings not only unmask a novel regulatory mechanism of PD-L1 expression, but also suggest that PD-L1 3'-UTR disruption could serve as a genetic marker to identify cancers that actively evade anti-tumour immunity through PD-L1 overexpression.


Subject(s)
3' Untranslated Regions/genetics , Gene Expression Regulation, Neoplastic , Neoplasms/genetics , Programmed Cell Death 1 Receptor/genetics , Tumor Escape/genetics , Up-Regulation , Adenocarcinoma/genetics , Animals , Antibodies/pharmacology , Antibodies/therapeutic use , CRISPR-Cas Systems , Cell Line, Tumor , Clonal Selection, Antigen-Mediated , Female , Genetic Markers/genetics , Humans , Leukemia-Lymphoma, Adult T-Cell/genetics , Lymphoma, Large B-Cell, Diffuse/genetics , Mice , Neoplasms/pathology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/biosynthesis , RNA Stability , RNA, Messenger/genetics , RNA, Messenger/metabolism , Stomach Neoplasms/genetics
6.
Gan To Kagaku Ryoho ; 46(11): 1677-1682, 2019 Nov.
Article in Japanese | MEDLINE | ID: mdl-31748471

ABSTRACT

In the tumor immunotherapy field, the adoptive immunotherapies, in which patient-derived T cells are strengthened ex vivo by activation or genetic modification, have been shown to be effective. However, some issues still remain to be solved. For example, it is not easy to efficiently expand tumor antigen-specific T cells, since they easily get exhausted during ex vivo culture. Moreover, these strategies are costly and time-consuming, because they are mainly conducted in autologous settings. To address these issues, we have been utilizing the induced pluripotent stem(iPS)cell technology. When iPS cells are established from tumor antigen-specific T cells, T cells regenerated from these iPS cells are expected to express the same T cell receptor(TCR)as the original T cells. In line with this concept, we succeeded in regenerating tumor antigen-specific cytotoxic T cells in 2013. We subsequently succeeded in developing a method by which very potent cytotoxic T lymphocytes are regenerated. We are now developing a strategy where non-T derived iPSCs are transduced with exogenous TCR gene efficacy and safety of which have been clinically tested. We plan to apply this approach to HLA haplotype-homozygous iPS cell stock lines, expecting that it will become possible to establish "off-the-shelf T cell" bank against various types of tumors.


Subject(s)
Induced Pluripotent Stem Cells , Neoplasms , Antigens, Neoplasm , Hematopoietic Stem Cell Transplantation , Humans , Immunotherapy, Adoptive , T-Lymphocytes, Cytotoxic
7.
Ann Hematol ; 93(3): 393-401, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23975214

ABSTRACT

Lymphoma-associated hemophagocytic syndrome (LAHS) is a serious disorder, and its early diagnosis and treatment with appropriate chemotherapy are very important. However, reliable markers for early diagnosis of LAHS have not been identified. We screened serum cytokines using a newly introduced assay system, cytometric bead array (CBA), and identified interferon-inducible protein 10 (IP-10)/CXCL10 and monokine induced by interferon gamma (MIG)/CXCL9 as useful markers. Serum concentrations of IP-10 and MIG at the time of LAHS diagnosis were greater than 500 and 5,000 pg/ml, respectively. The sensitivity and specificity for LAHS diagnosis were 100 and 95 %, respectively, when we set the above values as the cut-off levels. Serum levels of these two chemokines were already elevated at the time of admission and significantly decreased after successful treatment, indicating their usefulness for both the diagnosis and therapeutic outcomes for LAHS. IP-10 and MIG were also useful in distinguishing severe from moderate/mild LAHS, and B-cell-type LAHS from T-cell/natural killer cell-type LAHS. Furthermore, IP-10 and MIG were of use to distinguish LAHS from sepsis in patients with hematologic malignancies. Rapid measurement of IP-10 and MIG by CBA appeared to be important for early diagnosis and treatment of LAHS.


Subject(s)
Biomarkers, Tumor/blood , Chemokine CXCL10/blood , Chemokine CXCL9/blood , Lymphohistiocytosis, Hemophagocytic/diagnosis , Lymphoma, Non-Hodgkin/physiopathology , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/therapeutic use , Diagnosis, Differential , Follow-Up Studies , Hospitals, Urban , Humans , Lymphohistiocytosis, Hemophagocytic/blood , Lymphohistiocytosis, Hemophagocytic/etiology , Lymphohistiocytosis, Hemophagocytic/prevention & control , Lymphoma, Non-Hodgkin/drug therapy , Lymphoma, Non-Hodgkin/pathology , Middle Aged , Neoplasm Staging , Reagent Kits, Diagnostic , Retrospective Studies , Sensitivity and Specificity , Severity of Illness Index , Young Adult
8.
Acta Haematol ; 128(3): 139-43, 2012.
Article in English | MEDLINE | ID: mdl-22890122

ABSTRACT

An 84-year-old Japanese man was admitted because of pancytopenia. The bone marrow was hypoplastic with a predominance of abnormal small lymphocytes and grape cells, which were positive for CD19 and CD20, and partially for the surface ĸ-light chain. Systemic CT scanning showed neither lymph node swelling nor hepatosplenomegaly. Serum immunoelectrophoresis and rocket immunoselection assays showed the presence of monoclonal IgG protein without a corresponding light chain and faint IgMĸ monoclonal protein. Histologic analysis of the clot preparation of the bone marrow aspirate facilitated a diagnosis of lymphoplasmacytic lymphoma (LPL). PCR analysis of the marrow cells demonstrated a clonal rearrangement of the immunoglobulin heavy-chain gene. From these results, we made a final diagnosis of γ-heavy-chain disease (γ-HCD) with underlying LPL localized in the bone marrow. We performed only a single course of immunochemotherapy (rituximab and fludarabine) in view of severely impaired hematopoiesis, which resulted in marked reduction of lymphoma cells and improvement of hematopoiesis. This report suggests the efficacy of rituximab plus fludarabine therapy for LPL-associated γ-HCD.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/therapeutic use , Lymphoma, B-Cell/drug therapy , Vidarabine/analogs & derivatives , Waldenstrom Macroglobulinemia/drug therapy , Aged, 80 and over , Heavy Chain Disease , Humans , Male , Rituximab , Vidarabine/therapeutic use
9.
Rinsho Ketsueki ; 53(11): 1906-10, 2012 Nov.
Article in Japanese | MEDLINE | ID: mdl-23257671

ABSTRACT

Although about 10 to 15% of patients with multiple myeloma (MM) develop AL amyloidosis, liver-restricted fatal amyloidosis is rare. We encountered such an MM patient. A 73-year-old female without a history of carpal tunnel syndrome was diagnosed with IgG-κ MM (Stage I by Durie & Salmon) in January, 2005. Because MM was exacerbated to Stage III in May, 2007, VAD (vincristine, adriamycin, dexamethasone) chemotherapy was performed with minor response, despite 3 courses of this regimen. Three courses of salvage chemotherapy (cyclophosphamide+melphalan; CM) were then performed with near partial response. In March, 2008, just before the 4th cycle of CM chemotherapy, she was slightly icteric with elevated biliary tract enzymes; therefore, treatment was switched to oral cyclophosphamide and prednisolone. At this time, she did not have macroglossia, skin eruption, gastrointestinal dysfunction, or bleeding. Echocardiography was also non-specific. One month later, she developed a marked bleeding tendency and leg edema. Laboratory tests showed a severe deterioration in liver function. In the middle of May, 2008, she progressed to hepatic coma and died of intracranial hemorrhage several days later. Autopsy showed that the liver was almost substituted by AL amyloid substance.


Subject(s)
Amyloidosis/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Liver Failure/etiology , Multiple Myeloma/drug therapy , Aged , Amyloidosis/complications , Amyloidosis/diagnosis , Dexamethasone/therapeutic use , Fatal Outcome , Female , Hemorrhage/complications , Humans , Immunoglobulin Light-chain Amyloidosis , Melphalan/therapeutic use , Multiple Myeloma/complications , Multiple Myeloma/diagnosis , Vincristine/therapeutic use
10.
Mol Ther Oncolytics ; 24: 77-86, 2022 Mar 17.
Article in English | MEDLINE | ID: mdl-35024435

ABSTRACT

To develop effective adoptive cell transfer therapy using T cell receptor (TCR)-engineered T cells, it is critical to isolate tumor-reactive TCRs that have potent anti-tumor activity. In humans, tumor-infiltrating lymphocytes (TILs) have been reported to contain CD8+PD-1+ T cells that express tumor-reactive TCRs. Characterization of tumor reactivity of TILs from non-human primate tumors could improve anti-tumor activity of TCR-engineered T cells in preclinical research. In this study, we sought to isolate TCR genes from CD8+PD-1+ T cells among TILs in a cynomolgus macaque model of tumor transplantation in which the tumors were infiltrated with CD8+ T cells and were eventually rejected. We analyzed the repertoire of TCRα and ß pairs obtained from single CD8+PD-1+ T cells in TILs and circulating lymphocytes and identified multiple TCR pairs with high frequency, suggesting that T cells expressing these recurrent TCRs were clonally expanded in response to tumor cells. We further showed that the recurrent TCRs exhibited cytotoxic activity to tumor cells in vitro and potent anti-tumor activity in mice transplanted with tumor cells. These results imply that this tumor transplantation macaque model recapitulates key features of human TILs and can serve as a platform toward preclinical studies of non-human primate tumor models.

11.
Acta Haematol ; 126(2): 122-8, 2011.
Article in English | MEDLINE | ID: mdl-21701157

ABSTRACT

Blastic plasmacytoid dendritic cell neoplasm (BPDCN), currently considered to originate from immature plasmacytoid dendritic cells (DC), is a rare and aggressive CD4+CD56+ neoplasm that frequently involves the skin and bone marrow. We present a case of an 80-year-old man with a CD4+CD56+ BPDCN that affected the orbital cavity and bone marrow. Although BPDCN has not been reported to express any lineage-specific markers, the neoplastic cells strongly expressed the CD13 antigen. Therefore, in addition to pathological examination, we attempted to induce in vitro morphological and surface marker changes with IL-3 and CD40 ligand. After treatment with these cytokines, the tumor cells enlarged markedly, acquired many fine dendrites, similar to mature DC, and showed enhanced expression of antigens specific to DC or antigen-presenting cells, such as CD40, CD80, CD83 and CD86. To the best of our knowledge, this is the first report of BPDCN expressing a myeloid antigen, CD13, although CD33 expression has been described in some cases. The present patient received 2 courses of combination chemotherapy consisting of cytarabine and etoposide, which resulted in complete remission. Given that the cellular origin of plasmacytoid DC is still controversial, myeloid antigen expression involving CD13 may not exclude a diagnosis of BPDCN.


Subject(s)
Anemia/etiology , Bone Marrow Neoplasms/metabolism , CD13 Antigens/metabolism , Dendritic Cells/pathology , Neoplasms, Plasma Cell/metabolism , Orbital Neoplasms/metabolism , Aged, 80 and over , Bone Marrow Neoplasms/blood , Bone Marrow Neoplasms/pathology , Bone Marrow Neoplasms/physiopathology , Humans , Male , Neoplasms, Plasma Cell/blood , Neoplasms, Plasma Cell/pathology , Neoplasms, Plasma Cell/physiopathology , Orbital Neoplasms/blood , Orbital Neoplasms/pathology , Orbital Neoplasms/physiopathology
12.
Rinsho Ketsueki ; 51(5): 332-8, 2010 May.
Article in Japanese | MEDLINE | ID: mdl-20534954

ABSTRACT

The prognosis of angioimmunoblastic T cell lymphoma (AITL) is poor because of chemotherapy-resistance and the short duration of remission. In recent years, cyclosporin A (CyA) has been employed as a alternative treatment for AITL in some patients with the rationale that CyA inhibits the activity and proliferation of neoplastic T cells. We herein report 4 chemotherapy-resistant AITL patients who were treated with oral CyA. The dosage of CyA was individually determined in each patient in order to achieve a blood CyA concentration of around 200 ng/ml at the trough level. A patient in whom AITL had relapsed 3 months after high dose chemotherapy with autologous hematopoietic stem cell transplantation (HSCT) achieved a sustained complete remission (CR) with CyA and underwent allogeneic HSCT. In 2 patients who had failed to respond to conventional chemotherapies, the circulating lymphoma cells rapidly disappeared after the initiation of CyA, and one of these patients demonstrated a durable CR. The other patient showed a good response to CyA, but the agent was discontinued because of infection. The remaining one patient with advanced AITL did not respond to CyA and died of disease progression. To our knowledge, the efficacy of CyA for chemotherapy-resistant AITL, even in a leukemic state, has not previously been reported.


Subject(s)
Cyclosporine/administration & dosage , Immunoblastic Lymphadenopathy/drug therapy , Immunosuppressive Agents/administration & dosage , Lymphoma, T-Cell/drug therapy , Aged , Fatal Outcome , Female , Humans , Male , Middle Aged , Peripheral Blood Stem Cell Transplantation , Remission Induction , Transplantation, Autologous , Treatment Outcome
13.
Mol Ther Methods Clin Dev ; 16: 126-135, 2020 Mar 13.
Article in English | MEDLINE | ID: mdl-31970197

ABSTRACT

Current adoptive T cell therapies conducted in an autologous setting are costly, time-consuming, and depend on the quality of the patient's T cells, and thus it would be highly beneficial to develop an allogeneic strategy. To this aim, we have developed a method by which cytotoxic T lymphocytes (CTLs) are regenerated from induced pluripotent stem cells that are originally derived from T cells (T-iPSCs). In order to assess the feasibility of this strategy, we investigated the frequency of usable T-iPSC clones in terms of their T cell-generating capability and T cell receptor (TCR) affinity. We first established eight clones of T-iPSCs bearing different MART-1-specific TCRs from a healthy volunteer. Whereas all clones were able to give rise to mature CTLs, cell yield varied greatly, and five clones were considered to be usable. TCR affinity in the regenerated CTLs showed a large variance among the eight clones, but functional avidities measured by cytotoxic activity were almost equivalent among three selected clones representing high, medium, and low TCR affinity. In a total of 50 alloreactivity tests using five CTL clones versus ten target cells, alloreactivity was seen in only three cases. These findings collectively support the feasibility of this T-iPSC strategy.

14.
Mol Ther Methods Clin Dev ; 19: 250-260, 2020 Dec 11.
Article in English | MEDLINE | ID: mdl-33102617

ABSTRACT

In the current adoptive T cell therapy, T cells from a patient are given back to that patient after ex vivo activation, expansion, or genetic manipulation. However, such strategy depends on the quality of the patient's T cells, sometimes leading to treatment failure. It would therefore be ideal to use allogeneic T cells as "off-the-shelf" T cells. To this aim, we have been developing a strategy where potent tumor-antigen-specific cytotoxic T lymphocytes (CTLs) are regenerated from T-cell-derived induced pluripotent stem cells (T-iPSCs). However, certain issues still remain that make it difficult to establish highly potent T-iPSCs: poor reprogramming efficiency of T cells into iPSCs and high variability in the differentiation capability of each T-iPSC clone. To expand the versatility of this approach, we thought of a method to produce iPSCs equivalent to T-iPSCs, namely, iPSCs transduced with exogenous T cell receptor (TCR) genes (TCR-iPSCs). To test this idea, we first cloned TCR genes from WT1-specific CTLs regenerated from T-iPSCs and then established WT1-TCR-iPSCs. We show that the regenerated CTLs from TCR-iPSCs exerted cytotoxic activity comparable to those from T-iPSCs against WT1 peptide-loaded cell line in in vitro model. These results collectively demonstrate the feasibility of the TCR-iPSC strategy.

15.
iScience ; 23(4): 100998, 2020 Apr 24.
Article in English | MEDLINE | ID: mdl-32259478

ABSTRACT

Current adoptive T cell therapies conducted in an autologous setting are costly, time consuming, and depend on the quality of the patient's T cells. To address these issues, we developed a strategy in which cytotoxic T lymphocytes (CTLs) are regenerated from iPSCs that were originally derived from T cells and succeeded in regenerating CTLs specific for the WT1 antigen, which exhibited therapeutic efficacy in a xenograft model of leukemia. In this study, we extended our strategy to solid tumors. The regenerated WT1-specific CTLs had a strong therapeutic effect in orthotopic xenograft model using a renal cell carcinoma (RCC) cell line. To make our method more generally applicable, we developed an allogeneic approach by transducing HLA-haplotype homozygous iPSCs with WT1-specific TCR α/ß genes that had been tested clinically. The regenerated CTLs antigen-specifically suppressed tumor growth in a patient-derived xenograft model of RCC, demonstrating the feasibility of our strategy against solid tumors.

16.
J Exp Med ; 216(4): 982-1000, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30872362

ABSTRACT

Immune checkpoint blockade against programmed cell death 1 (PD-1) and its ligand PD-L1 often induces durable tumor responses in various cancers, including non-small cell lung cancer (NSCLC). However, therapeutic resistance is increasingly observed, and the mechanisms underlying anti-PD-L1 (aPD-L1) antibody treatment have not been clarified yet. Here, we identified two unique secreted PD-L1 splicing variants, which lacked the transmembrane domain, from aPD-L1-resistant NSCLC patients. These secreted PD-L1 variants worked as "decoys" of aPD-L1 antibody in the HLA-matched coculture system of iPSC-derived CD8 T cells and cancer cells. Importantly, mixing only 1% MC38 cells with secreted PD-L1 variants and 99% of cells that expressed wild-type PD-L1 induced resistance to PD-L1 blockade in the MC38 syngeneic xenograft model. Moreover, anti-PD-1 (aPD-1) antibody treatment overcame the resistance mediated by the secreted PD-L1 variants. Collectively, our results elucidated a novel resistant mechanism of PD-L1 blockade antibody mediated by secreted PD-L1 variants.


Subject(s)
Antibodies, Neutralizing/pharmacology , Antibodies, Neutralizing/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , Carcinoma, Non-Small-Cell Lung/drug therapy , Drug Resistance, Neoplasm , Lung Neoplasms/drug therapy , Protein Splicing , Animals , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , CHO Cells , Carcinoma, Non-Small-Cell Lung/pathology , Cricetulus , Female , Humans , Lung Neoplasms/pathology , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/metabolism , THP-1 Cells , Transfection , Xenograft Model Antitumor Assays
17.
Int J Hematol ; 107(3): 271-277, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29388165

ABSTRACT

Recent advances in adoptive immunotherapy using cytotoxic T lymphocytes (CTLs) have led to moderate therapeutic anti-cancer effects in clinical trials. However, a critical issue, namely that CTLs collected from patients are easily exhausted during expansion culture, has yet to be solved. To address this issue, we have been developing a strategy which utilizes induced pluripotent stem cell (iPSC) technology. This strategy is based on the idea that when iPSCs are produced from antigen-specific CTLs, CTLs regenerated from such iPSCs should show the same antigen specificity as the original CTLs. Pursuing this idea, we previously succeeded in regenerating melanoma antigen MART1-specific CTLs, and more recently in producing potent CTLs expressing CD8αß heterodimer. We are now developing a novel method by which non-T derived iPSCs are transduced with exogenous T cell receptor genes. If this method is applied to Human Leukocyte Antigen (HLA) haplotype-homozygous iPSC stock, it will be possible to prepare "off-the-shelf" T cells. As a first-in-human trial, we are planning to apply our strategy to relapsed acute myeloid leukemia patients by targeting the WT1 antigen.


Subject(s)
Cell Engineering/methods , Immunotherapy, Adoptive/methods , Induced Pluripotent Stem Cells , Leukemia, Myeloid, Acute/therapy , T-Lymphocytes, Cytotoxic , Allografts , CD8 Antigens , Genes, T-Cell Receptor , HLA Antigens , Humans , Leukemia, Myeloid, Acute/genetics , Molecular Targeted Therapy , WT1 Proteins
18.
Stem Cell Reports ; 9(3): 853-867, 2017 09 12.
Article in English | MEDLINE | ID: mdl-28867344

ABSTRACT

HLA haplotype-homozygous (HLA-homo) induced pluripotent stem cells (iPSCs) are being prepared to be used for allogeneic transplantation of regenerated tissue into recipients carrying an identical haplotype in one of the alleles (HLA-hetero). However, it remains unaddressed whether natural killer (NK) cells respond to these regenerated cells. HLA-C allotypes, known to serve as major ligands for inhibitory receptors of NK cells, can be classified into group 1 (C1) and group 2 (C2), based on their binding specificities. We found that the T cells and vascular endothelial cells regenerated from HLA-homo-C1/C1 iPSCs were killed by specific NK cell subsets from a putative HLA-hetero-C1/C2 recipient. Such cytotoxicity was canceled when target cells were regenerated from iPSCs transduced with the C2 gene identical to the recipient. These results clarify that NK cells can kill regenerated cells by sensing the lack of HLA-C expression and further provide the basis for an approach to prevent such NK cell-mediated rejection responses.


Subject(s)
HLA Antigens/metabolism , Haplotypes/genetics , Induced Pluripotent Stem Cells/metabolism , Killer Cells, Natural/metabolism , Receptors, KIR/metabolism , Asian People , Cytotoxicity, Immunologic , Homozygote , Humans , Immune Tolerance , Ligands , Lymphocyte Subsets/metabolism , Regeneration , T-Lymphocytes/metabolism , Tissue Donors , Transplantation, Homologous
19.
Cancer Res ; 76(23): 6839-6850, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27872100

ABSTRACT

Although adoptive transfer of cytotoxic T lymphocytes (CTL) offer a promising cancer therapeutic direction, the generation of antigen-specific CTL from patients has faced difficulty in efficient expansion in ex vivo culture. To resolve this issue, several groups have proposed that induced pluripotent stem cell technology be applied for the expansion of antigen-specific CTL, which retain expression of the same T-cell receptor as original CTL. However, in these previous studies, the regenerated CTL are mostly of the CD8αα+ innate type and have less antigen-specific cytotoxic activity than primary CTL. Here we report that, by stimulating purified iPSC-derived CD4/CD8 double-positive cells with anti-CD3 antibody, T cells expressing CD8αß were generated and exhibited improved antigen-specific cytotoxicity compared with CD8αα+ CTL. Failure of CD8αß T-cell production using the previous method was found to be due to killing of double-positive cells by the double-negative cells in the mixed cultures. We found that WT1 tumor antigen-specific CTL regenerated by this method prolonged the survival of mice bearing WT1-expressing leukemic cells. Implementation of our methods may offer a useful clinical tool. Cancer Res; 76(23); 6839-50. ©2016 AACR.


Subject(s)
CD8 Antigens/genetics , T-Lymphocytes, Cytotoxic/immunology , Transcriptome/genetics , Animals , Humans , Mice
20.
AJP Rep ; 6(1): e62-7, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26929873

ABSTRACT

Introduction New-onset systemic lupus erythematosus (SLE) during pregnancy is rare and difficult to diagnose, especially in cases that manifest as preeclampsia. We report a patient with new-onset SLE that manifested as preeclampsia during pregnancy and provide a review of the literature to identify factors for a rapid diagnosis. Case A 32-year-old primigravid Japanese woman was diagnosed with severe preeclampsia and underwent emergent cesarean section at 29 weeks of gestation. Her hypertension and renal disorder gradually improved after the operation, but her thrombocytopenia and anemia worsened. SLE was diagnosed on postoperative day 5 by a comprehensive autoimmune workup. She was discharged on postoperative day 34 with remission. Conclusion Our case and previous reports suggest that distinguishing underlying SLE from preeclampsia in the third trimester is particularly difficult. Helpful factors for diagnosis of suspected SLE in these cases were persistence of symptoms and new atypical symptoms for preeclampsia revealed after delivery (e.g., fever, renal disorder, and thrombocytopenia).

SELECTION OF CITATIONS
SEARCH DETAIL