102 results on '"Sachiko Okamoto"'
Search Results
2. Intradermal administration of DNA vaccine targeting Omicron SARS-CoV-2 via pyro-drive jet injector provides the prolonged neutralizing antibody production via germinal center reaction
- Author
-
Hiroki Hayashi, Jiao Sun, Yuka Yanagida, Takako Otera, Jiayu A. Tai, Tomoyuki Nishikawa, Kunihiko Yamashita, Naoki Sakaguchi, Shota Yoshida, Satoshi Baba, Chin Yang Chang, Munehisa Shimamura, Sachiko Okamoto, Yasunori Amaishi, Hideto Chono, Junichi Mineno, Hiromi Rakugi, Ryuichi Morishita, and Hironori Nakagami
- Subjects
Medicine ,Science - Abstract
Abstract Emerging SARS-CoV-2 Omicron variants are highly contagious with enhanced immune escape mechanisms against the initially approved COVID-19 vaccines. Therefore, we require stable alternative-platform vaccines that confer protection against newer variants of SARS-CoV-2. We designed an Omicron B.1.1.529 specific DNA vaccine using our DNA vaccine platform and evaluated the humoral and cellular immune responses. SD rats intradermally administered with Omicron-specific DNA vaccine via pyro-drive jet injector (PJI) thrice at 2-week intervals elicited high antibody titers against the Omicron subvariants as well as the ancestral strain. Indeed, the Omicron B.1.1.529-specific antibody titer and neutralizing antibody were higher than that of other strains. Longitudinal monitoring indicated that anti-spike (ancestral and Omicron) antibody titers decreased toward 30 weeks after the first vaccination dose. However, neutralization activity remained unaltered. Germinal center formation was histologically detected in lymph nodes in rats immunized with Omicron DNA vaccine. Ancestral spike-specific immune cell response was slightly weaker than Omicron spike-specific response in splenocytes with Omicron-adapted DNA vaccine, evaluated by ELISpot assay. Collectively, our findings suggest that Omicron targeting DNA vaccines via PJI can elicit robust durable antibody production mediated by germinal center reaction against this new variant as well as partially against the spike protein of other SARS-CoV-2 variants.
- Published
- 2023
- Full Text
- View/download PDF
3. Exhaustion, rather than lack of infiltration and persistence, of CAR-T cells hampers the efficacy of CAR-T therapy in an orthotopic PDAC xenograft model
- Author
-
Yuta Takeuchi, Yizheng Wang, Katsunori Sasaki, Osamu Sato, Takahiro Tsuchikawa, Linan Wang, Yasunori Amaishi, Sachiko Okamoto, Junichi Mineno, Yoshifumi Hirokawa, Kanako C. Hatanaka, Yutaka Hatanaka, Takuma Kato, Hiroshi Shiku, and Satoshi Hirano
- Subjects
Chimeric antigen receptor ,Pancreatic cancer orthotopic xenograft model ,Tumor infiltrating lymphocyte ,Metabolic pathway ,Mitochondrial dynamics ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Chimeric antigen receptor T-cell (CAR-T) therapy has demonstrated impressive success in the treatment of patients with hematologic tumors yet achieved very limited efficacy for solid tumors due to hurdles unique to solid tumors. It is also noted that the tumor microenvironment composition varies between tumor type, which again imposes unique set of hurdles in each solid tumor. Therefore, elucidation of individual hurdles is key to achieving successful CAR-T therapy for solid tumors. In the present study, we employed an orthotopic human PDAC xenograft model, in which quantitative, spatial and functional dynamics of CAR-T cells in tumor tissues were analyzed to obtain insights into ways of overcoming PDAC related hurdles. Contrary to previous studies that demonstrated a limited persistency and infiltration of CAR-T cells in many solid tumors, they persist and accumulated in PDAC tumor tissues. Ex vivo analysis revealed that CAR-T cells that had been recovered at different time points from mice bearing an orthotopic PDAC tumor exhibited a gradual loss of tumor reactivity. This loss of tumor reactivity of CAR-T cells was associated with the increased expression of AMP-activated protein kinase and Mitofusin 1/ Dynamin-related protein 1 ratio.
- Published
- 2024
- Full Text
- View/download PDF
4. 434 Concurrent identification of true neoantigens and neoantigen-specific T cells by scRNA-Seq and scTCR-Seq
- Author
-
Yukari Kobayashi, Sachiko Okamoto, Kazuhiro Kakimi, and Koji Nagaoka
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2023
- Full Text
- View/download PDF
5. Modified DNA vaccine confers improved humoral immune response and effective virus protection against SARS-CoV-2 delta variant
- Author
-
Hiroki Hayashi, Jiao Sun, Yuka Yanagida, Takako Otera, Miwa Sasai, Chin Yang Chang, Jiayu A. Tai, Tomoyuki Nishikawa, Kunihiko Yamashita, Naoki Sakaguchi, Shota Yoshida, Satoshi Baba, Munehisa Shimamura, Sachiko Okamoto, Yasunori Amaishi, Hideto Chono, Junichi Mineno, Hiromi Rakugi, Ryuichi Morishita, Masahiro Yamamoto, and Hironori Nakagami
- Subjects
Medicine ,Science - Abstract
Abstract Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has led to a global pandemic. New technologies have been utilized to develop several types of vaccines to prevent the spread of SARS-CoV-2 infection, including mRNA vaccines. Our group previously developed an effective DNA-based vaccine. However, emerging SARS-CoV-2 variants of concern (VOCs), such as the delta variant, have escaped mutations against vaccine-induced neutralizing antibodies. This suggests that modified vaccines accommodating VOCs need to be developed promptly. Here, we first modified the current DNA vaccine to enhance antigenicity. Compared with the parental DNA vaccine, the modified version (GP∆-DNA vaccine) induced rapid antibody production. Next, we updated the GP∆-DNA vaccine to spike glycoprotein of the delta variant (GP∆-delta DNA vaccine) and compared the efficacy of different injection routes, namely intramuscular injection using a needle and syringe and intradermal injection using a pyro-drive jet injector (PJI). We found that the levels of neutralizing antibodies induced by the intradermal PJI injection were higher than intramuscular injection. Furthermore, the PJI-injected GP∆-delta DNA vaccine effectively protected human angiotensin-converting enzyme 2 (hACE2) knock-in mice from delta-variant infection. These results indicate that the improved DNA vaccine was effective against emerging VOCs and was a potential DNA vaccine platform for future VOCs or global pandemics.
- Published
- 2022
- Full Text
- View/download PDF
6. CAR-Modified Vγ9Vδ2 T Cells Propagated Using a Novel Bisphosphonate Prodrug for Allogeneic Adoptive Immunotherapy
- Author
-
Yizheng Wang, Linan Wang, Naohiro Seo, Satoshi Okumura, Tae Hayashi, Yasushi Akahori, Hiroshi Fujiwara, Yasunori Amaishi, Sachiko Okamoto, Junichi Mineno, Yoshimasa Tanaka, Takuma Kato, and Hiroshi Shiku
- Subjects
Vγ9Vδ2 T cell ,chimeric antigen receptor (CAR) ,carcinoembryonic antigen (CEA) ,graft-versus-host disease (GVHD) ,off-the-shelf ,glucocorticoid-induced TNFR-related protein (GITR) ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
The benefits of CAR-T therapy could be expanded to the treatment of solid tumors through the use of derived autologous αβ T cell, but clinical trials of CAR-T therapy for patients with solid tumors have so far been disappointing. CAR-T therapy also faces hurdles due to the time and cost intensive preparation of CAR-T cell products derived from patients as such CAR-T cells are often poor in quality and low in quantity. These inadequacies may be mitigated through the use of third-party donor derived CAR-T cell products which have a potent anti-tumor function but a constrained GVHD property. Vγ9Vδ2 TCR have been shown to exhibit potent antitumor activity but not alloreactivity. Therefore, in this study, CAR-T cells were prepared from Vγ9Vδ2 T (CAR-γδ T) cells which were expanded by using a novel prodrug PTA. CAR-γδ T cells suppressed tumor growth in an antigen specific manner but only during a limited time window. Provision of GITR co-stimulation enhanced anti-tumor function of CAR-γδ T cells. Our present results indicate that, while further optimization of CAR-γδ T cells is necessary, the present results demonstrate that Vγ9Vδ2 T cells are potential source of ‘off-the-shelf’ CAR-T cell products for successful allogeneic adoptive immunotherapy.
- Published
- 2023
- Full Text
- View/download PDF
7. NY-ESO-1-specific redirected T cells with endogenous TCR knockdown mediate tumor response and cytokine release syndrome
- Author
-
Takashi Kojima, Hiroaki Ikeda, Sachiko Okamoto, Junichi Mineno, Hiroshi Shiku, Shigehisa Kitano, Shinichi Kageyama, Yoshihiro Miyahara, Mikiya Ishihara, Daisuke Tomura, Ikuei Nukaya, Tetsuro Sasada, Noboru Yamamoto, Takashi Watanabe, Hideyuki Mishima, Alessandra Nardin, Evan Newell, Hidefumi Kato, Hiroyoshi Hattori, Takeru Funakoshi, Eiichi Sato, Hideto Chono, Muhammad Faris Kairi, Phuong Diem Hoang Nguyen, Yannick Simoni, and Michael Fehlings
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2022
- Full Text
- View/download PDF
8. 103 Quality improvement of anti-CD38-JAK/STAT CAR-T cells by suppressing CD38 expression and inhibition of tyrosine kinase
- Author
-
Sachiko Okamoto, Junichi Mineno, Yasunori Amaishi, Izumi Maki, Maiko Sugizaki, and Kenichiro Mihara
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2021
- Full Text
- View/download PDF
9. 129 A novel CAR conducting antigen-specific JAK-STAT signals demonstrates superior antitumor effects with minimal undesired non-specific activation
- Author
-
Sachiko Okamoto, Junichi Mineno, Naoto Hirano, Yasunori Amaishi, Yu Okubo, Mitsuki Shigeta, and Yota Ohashi
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2020
- Full Text
- View/download PDF
10. A Functionally Superior Second-Generation Vector Expressing an Aurora Kinase-A-Specific T-Cell Receptor for Anti-Leukaemia Adoptive Immunotherapy.
- Author
-
Nicholas Paul Casey, Hiroshi Fujiwara, Kazushi Tanimoto, Sachiko Okamoto, Junichi Mineno, Kiyotaka Kuzushima, Hiroshi Shiku, and Masaki Yasukawa
- Subjects
Medicine ,Science - Abstract
Aurora Kinase A is a cancer-associated protein normally involved in the regulation of mitosis. Being over-expressed in a range of cancers, it is a suitable target for cell-based immunotherapy. Gene transfer of T-cell receptor sequences cognisant of HLA-A*0201-restricted Aurora Kinase A antigen has previously been shown to transfer specific immunoreactivity against the target peptide in a Human Lymphocyte Antigen-restricted manner. While T cell receptor gene-transfer has great potential in overcoming the difficulties of isolating and expanding tumour-reactive lymphocytes from a patient's own cells, one hurdle is potential mispairing and competition between exogenous and endogenous T cell receptor chains. We have used a retroviral vector design bearing a short-interfering RNA that downregulates endogenous T cell receptor chains, without affecting expression of the transgenic T cell receptor sequences. The T cell receptor expression cassette also includes a 2A self-cleaving peptide, resulting in equimolar expression of the T cell receptor alpha and beta chains, further enhancing formation of the desired T cell receptor. Via a simple, modular cloning method, we have cloned the alpha and beta chains of the anti-Aurora Kinase A-reactive T cell receptor into this 'siTCR' vector. We then compared the activity of this vector against the original, 'conventional' vector across a panel of assays. T cell receptors expressed from the siTCR-vector retained the cytotoxic functionality of the original vector, with evidence of reduced off-target reactivity. The rate of expression of correctly-formed T cell receptors was superior using the siTCR design, and this was achieved at lower vector copy numbers. Maintaining T cell receptor efficacy with a reduced vector copy number reduces the risk of genotoxicity. The siTCR design also reduces the risk of mispairing and cross-reactivity, while increasing the functional titre. Such improvements in the safety of T cell receptor gene-transfer will be crucial for clinical applications of this technology.
- Published
- 2016
- Full Text
- View/download PDF
11. Co-introduced functional CCR2 potentiates in vivo anti-lung cancer functionality mediated by T cells double gene-modified to express WT1-specific T-cell receptor.
- Author
-
Hiroaki Asai, Hiroshi Fujiwara, Jun An, Toshiki Ochi, Yukihiro Miyazaki, Kozo Nagai, Sachiko Okamoto, Junichi Mineno, Kiyotaka Kuzushima, Hiroshi Shiku, Hirofumi Inoue, and Masaki Yasukawa
- Subjects
Medicine ,Science - Abstract
BACKGROUND AND PURPOSE: Although gene-modification of T cells to express tumor-related antigen-specific T-cell receptor (TCR) or chimeric antigen receptor (CAR) has clinically proved promise, there still remains room to improve the clinical efficacy of re-directed T-cell based antitumor adoptive therapy. In order to achieve more objective clinical responses using ex vivo-expanded tumor-responsive T cells, the infused T cells need to show adequate localized infiltration into the tumor. METHODOLOGY/PRINCIPAL FINDINGS: Human lung cancer cells variously express a tumor antigen, Wilms' Tumor gene product 1 (WT1), and an inflammatory chemokine, CCL2. However, CCR2, the relevant receptor for CCL2, is rarely expressed on activated T-lymphocytes. A HLA-A2402(+) human lung cancer cell line, LK79, which expresses high amounts of both CCL2 and WT1 mRNA, was employed as a target. Normal CD8(+) T cells were retrovirally gene-modified to express both CCR2 and HLA-A*2402-restricted and WT1(235-243) nonapeptide-specific TCR as an effector. Anti-tumor functionality mediated by these effector cells against LK79 cells was assessed both in vitro and in vivo. Finally the impact of CCL2 on WT1 epitope-responsive TCR signaling mediated by the effector cells was studied. Introduced CCR2 was functionally validated using gene-modified Jurkat cells and human CD3(+) T cells both in vitro and in vivo. Double gene-modified CD3(+) T cells successfully demonstrated both CCL2-tropic tumor trafficking and cytocidal reactivity against LK79 cells in vitro and in vivo. CCL2 augmented the WT1 epitope-responsive TCR signaling shown by relevant luciferase production in double gene-modified Jurkat/MA cells to express luciferase and WT1-specific TCR, and CCL2 also dose-dependently augmented WT1 epitope-responsive IFN-γ production and CD107a expression mediated by these double gene-modified CD3(+) T cells. CONCLUSION/SIGNIFICANCE: Introduction of the CCL2/CCR2 axis successfully potentiated in vivo anti-lung cancer reactivity mediated by CD8(+) T cells double gene-modified to express WT1-specific TCR and CCR2 not only via CCL2-tropic tumor trafficking, but also CCL2-enhanced WT1-responsiveness.
- Published
- 2013
- Full Text
- View/download PDF
12. A Promising Vector for TCR Gene Therapy: Differential Effect of siRNA, 2A Peptide, and Disulfide Bond on the Introduced TCR Expression
- Author
-
Sachiko Okamoto, Yasunori Amaishi, Yumi Goto, Hiroaki Ikeda, Hiroshi Fujiwara, Kiyotaka Kuzushima, Masaki Yasukawa, Hiroshi Shiku, and Junichi Mineno
- Subjects
Therapeutics. Pharmacology ,RM1-950 - Abstract
Adoptive immunotherapy using TCR gene-modified T-lymphocytes is an attractive strategy for targeting malignancies. However, TCR mispairings between endogenous and introduced TCR chains are a major concern, as they may induce mixed TCRs with unknown specificities and may reduce the expression of therapeutic TCRs. To overcome these problems, we have recently established a novel retroviral siTCR vector encoding small-interfering RNAs (siRNAs) to knockdown endogenous TCR genes for the efficient expression of therapeutic TCRs. In this study, to improve the efficacy of siTCR vectors, we developed 2A peptide-based siTCR vectors that could increase the expression levels of transduced TCRs compared with internal promoter-based siTCR vectors. We also evaluated the efficacy of an siTCR strategy and the addition of a new interchain disulfide bond created by cysteine modification. We found that the effect of the cysteine modification depended on TCR variations, while the siTCR strategy improved the expression of all TCRs tested. Furthermore, the combined effect of the siTCR and cysteine modification strategies was highly significant for certain TCRs. Therefore, our novel siTCR technology, in isolation or in combination with another strategy, may open the door to effective immunotherapy for cancer patients.
- Published
- 2012
- Full Text
- View/download PDF
13. CAR-Modified Vγ9Vδ2 T Cells Propagated Using a Novel Bisphosphonate Prodrug for Allogeneic Adoptive Immunotherapy
- Author
-
Shiku, Yizheng Wang, Linan Wang, Naohiro Seo, Satoshi Okumura, Tae Hayashi, Yasushi Akahori, Hiroshi Fujiwara, Yasunori Amaishi, Sachiko Okamoto, Junichi Mineno, Yoshimasa Tanaka, Takuma Kato, and Hiroshi
- Subjects
Vγ9Vδ2 T cell ,chimeric antigen receptor (CAR) ,carcinoembryonic antigen (CEA) ,graft-versus-host disease (GVHD) ,off-the-shelf ,glucocorticoid-induced TNFR-related protein (GITR) ,tetrakis-pivaloyloxymethyl 2-(thiazole-2-ylamino)ethylidene-1,1-bisphosphonate (PTA) - Abstract
The benefits of CAR-T therapy could be expanded to the treatment of solid tumors through the use of derived autologous αβ T cell, but clinical trials of CAR-T therapy for patients with solid tumors have so far been disappointing. CAR-T therapy also faces hurdles due to the time and cost intensive preparation of CAR-T cell products derived from patients as such CAR-T cells are often poor in quality and low in quantity. These inadequacies may be mitigated through the use of third-party donor derived CAR-T cell products which have a potent anti-tumor function but a constrained GVHD property. Vγ9Vδ2 TCR have been shown to exhibit potent antitumor activity but not alloreactivity. Therefore, in this study, CAR-T cells were prepared from Vγ9Vδ2 T (CAR-γδ T) cells which were expanded by using a novel prodrug PTA. CAR-γδ T cells suppressed tumor growth in an antigen specific manner but only during a limited time window. Provision of GITR co-stimulation enhanced anti-tumor function of CAR-γδ T cells. Our present results indicate that, while further optimization of CAR-γδ T cells is necessary, the present results demonstrate that Vγ9Vδ2 T cells are potential source of ‘off-the-shelf’ CAR-T cell products for successful allogeneic adoptive immunotherapy.
- Published
- 2023
- Full Text
- View/download PDF
14. Data from Gene-Modified Human α/β-T Cells Expressing a Chimeric CD16-CD3ζ Receptor as Adoptively Transferable Effector Cells for Anticancer Monoclonal Antibody Therapy
- Author
-
Masaki Yasukawa, Eiichi Ishii, John Barrett, Hiroshi Shiku, Kiyotaka Kuzushima, Junichi Mineno, Sachiko Okamoto, Yukihiro Miyazaki, Hiroaki Asai, Kazushi Tanimoto, Hiroshi Fujiwara, and Fumihiro Ochi
- Abstract
The central tumoricidal activity of anticancer monoclonal antibodies (mAb) is exerted by FcγR IIIa (CD16)–expressing effector cells in vivo via antibody-dependent cell-mediated cytotoxicity (ADCC), as observed for natural killer (NK) cells. In practice, chemotherapy-induced leukopenia and exhaustion of NK cells resulting from ADCC often hamper the clinical efficacy of cancer treatment. To circumvent this drawback, we examined in vivo the feasibility of T cells, gene-modified to express a newly generated affinity-matured (158V/V) chimeric CD16-CD3ζ receptor (cCD16ζ-T cells), as a transferable alternative effector for cancer mAb therapy. cCD16ζ-T cells were readily expandable in ex vivo culture using anti-CD2/CD3/CD28 beads and recombinant human interleukin-2 (rhIL-2), and they successfully displayed ADCC-mediated tumoricidal activity in vitro. During ADCC, ligation of opsonized cancer cells to the introduced cCD16ζ-T cells stimulated the effector cells to produce proinflammatory cytokines and release toxic granules through the activation of the Nuclear factor of activated T cells (NFAT) pathway after phosphorylation of the CD3ζ chain. In parallel, these stimulated cCD16ζ-T cells transiently proliferated and differentiated into effector memory T cells. In contrast, NK cells activated by rhIL-2 displayed similar ADCC activity, but failed to proliferate. Human cCD16ζ-T cells infused concomitantly with anti-CD20 mAb synergistically inhibited the growth of disseminated Raji cells, a CD20+ lymphoma cell line, in immunodeficient mice, whereas similarly infused rhIL-2–treated NK cells survived for a shorter time and displayed less effective tumor suppression. Our findings strongly suggest the clinical feasibility of cCD16ζ-T cells as adoptively transferable ADCC effector cells that could potentially enhance the clinical responses mediated by currently available anticancer mAbs. Cancer Immunol Res; 2(3); 249–62. ©2014 AACR.
- Published
- 2023
15. Supplementary Figure 2 from Gene-Modified Human α/β-T Cells Expressing a Chimeric CD16-CD3ζ Receptor as Adoptively Transferable Effector Cells for Anticancer Monoclonal Antibody Therapy
- Author
-
Masaki Yasukawa, Eiichi Ishii, John Barrett, Hiroshi Shiku, Kiyotaka Kuzushima, Junichi Mineno, Sachiko Okamoto, Yukihiro Miyazaki, Hiroaki Asai, Kazushi Tanimoto, Hiroshi Fujiwara, and Fumihiro Ochi
- Abstract
PDF file - 4938K, A. Representative patterns of tumor growth displayed in mice treated with cCD16ζ-T cells in combination with rituximab in the late phase determined using in vivo bioluminescence assay. In mice treated with cCD16ζ-T cells combined with rituximab, tumor growth at truncal sites appeared to be durably localized and suppressed. However, the brain lesion showed obvious progression, even after the second therapeutic infusion on day 24. On day 32, the cCD16ζ-T-No.8 mouse was euthanatized to examine the persistence of therapeutically infused effector cells in the spleen and BM. B. In cCD16ζ-T-No.8 mouse, all detected human cells in both the spleen and BM were CD3+ T cells, and not CD20+ Raji cells. About 20% of these residual T cells displayed cell-surface CD16, i.e., cCD16ζ-T cells defined as hCD45+CD3+CD16+. C. cCD16ζ-T cells in both the spleen (upper) and BM (lower) retained their responsiveness exclusively against rituximab-opsonized Raji cells, reflected as increased expression of CD107a.
- Published
- 2023
16. Supplementary Figure 1 from Gene-Modified Human α/β-T Cells Expressing a Chimeric CD16-CD3ζ Receptor as Adoptively Transferable Effector Cells for Anticancer Monoclonal Antibody Therapy
- Author
-
Masaki Yasukawa, Eiichi Ishii, John Barrett, Hiroshi Shiku, Kiyotaka Kuzushima, Junichi Mineno, Sachiko Okamoto, Yukihiro Miyazaki, Hiroaki Asai, Kazushi Tanimoto, Hiroshi Fujiwara, and Fumihiro Ochi
- Abstract
PDF file - 5868K, A. Tumor growth in mice treated with activated NK cells combined with rituximab was serially measured using in vivo bioluminescence assay. Two mice (NK-No.1* and NK-No.2**) were euthanized at different time points on day 14 and day 18, respectively. Thereafter, human NK cells and Raji cells persisting in mouse spleen and femoral bone marrow (BM) were measured by flow cytometry. B. Tumor growth in mice similarly treated with cCD16ζ-T cells and rituximab was assessed by in vivo bioluminescence assay. Experiments in Supplementary Fig. S1A and Fig. S1B were conducted in parallel. For comparison with NK-treated mice, a cCD16ζ-T-No.7# mouse was euthanized on day 14, and cCD16ζ-T cells and Raji cells were measured similarly to those in Supplementary Fig. S1A. C. In mice treated with activated NK cells, on day 14, Raji cells were detectable at 8.0% among BM cells, but not in the spleen, as indicated by the bioluminescence assay in Supplementary Fig. S1A. Along with disease progression in Supplementary Fig. S1A from day 14 to day 18, the number of Raji cells increased from 0% to 7.4% in spleen and from 8.0% to 13.7% in BM, respectively. On the other hand, at these two time points, the relative proportion of NK cells remained constant at 4% in the spleen and 0.4% in the BM, respectively. D. Unlike mice treated with NK cells on day 14, higher numbers of effector cells were detectable (9.9% in spleen and 0.8% in BM, respectively), and no Raji cells were detected in either the spleen or BM in the cCD16ζ-T-No.7 mouse. Taken together, the data indicated that the in vivo tumor suppression mediated by cCD16ζ-T cells exploiting ADCC was superior to that mediated by NK cells.
- Published
- 2023
17. Data from Development of Engineered T Cells Expressing a Chimeric CD16-CD3ζ Receptor to Improve the Clinical Efficacy of Mogamulizumab Therapy Against Adult T-Cell Leukemia
- Author
-
Masaki Yasukawa, A. John Barrett, Takashi Sugiyama, Hiroshi Shiku, Kiyotaka Kuzushima, Junichi Mineno, Sachiko Okamoto, Nicholas Casey, Kazushi Tanimoto, Fumihiro Ochi, Hiroshi Fujiwara, and Hiroki Tanaka
- Abstract
Purpose: Mogamulizumab (Mog), a humanized anti-CC chemokine receptor 4 (CCR4) mAb that mediates antibody-dependent cellular cytotoxicity (ADCC) using FcγR IIIa (CD16)-expressing effector cells, has recently been approved for treatment of CCR4-positive adult T-cell leukemia (ATL) in Japan. However, Mog failure has sometimes been observed in patients who have accompanying chemotherapy-associated lymphocytopenia. In this study, we examined whether adoptive transfer of artificial ADCC effector cells combined with Mog would overcome this drawback.Experimental Design: We lentivirally gene-modified peripheral blood T cells from healthy volunteers and ATL patients expressing the affinity-increased chimeric CD16-CD3ζ receptor (cCD16ζ-T cells). Subsequently, we examined the ADCC effect mediated by those cCD16ζ-T cells in the presence of Mog against ATL tumor cells both in vitro and in vivo.Results: cCD16ζ-T cells derived from healthy donors killed in vitro Mog-opsonized ATL cell line cells (n = 7) and primary ATL cells (n = 4) depending on both the number of effector cells and the dose of the antibody. cCD16ζ-T cells generated from ATL patients (n = 3) also exerted cytocidal activity in vitro against Mog-opsonized autologous ATL cells. Using both intravenously disseminated model (n = 5) and subcutaneously inoculated model (n = 4), coadministration of Mog and human cCD16ζ-T cells successfully suppressed tumor growth in xenografted immunodeficient mice, and significantly prolonged their survival (P < 0.01 and P = 0.02, respectively).Conclusions: These data strongly suggest clinical feasibility of the novel combined adoptive immunotherapy using cCD16ζ-T cells and Mog for treatment of aggressive ATL, particularly in patients who are ineligible for allogeneic hematopoietic stem cell transplantation. Clin Cancer Res; 22(17); 4405–16. ©2016 AACR.
- Published
- 2023
18. Supplementary Figure S2 from Development of Engineered T Cells Expressing a Chimeric CD16-CD3ζ Receptor to Improve the Clinical Efficacy of Mogamulizumab Therapy Against Adult T-Cell Leukemia
- Author
-
Masaki Yasukawa, A. John Barrett, Takashi Sugiyama, Hiroshi Shiku, Kiyotaka Kuzushima, Junichi Mineno, Sachiko Okamoto, Nicholas Casey, Kazushi Tanimoto, Fumihiro Ochi, Hiroshi Fujiwara, and Hiroki Tanaka
- Abstract
ADCC activity mediated by cCD16ζ-T cells against Mog-opsonized ATL tumor cells was dependent on both effector cell number and Mog dose. However, at a pharmacological dose of Mog, the decline of ADCC was more obviously dependent on the number of effector cells. (a) At a pharmacological dose of Mog (1μg/mL), ADCC activity mediated by cCD16ζ-T cells against Mog-opsonized ATL tumor cells declined sharply when the number of effector cells was lower. In order to exclude NK cell activity, K562-A24 was employed in this experiment as a negative control. Each experiment at the indicated dose of Mog was conducted in triplicate using three independent sets of cells from three different healthy donors. LCL, EBV-immortalized B cell line; Mog, mogamulizumab. Each experiment was conducted in triplicate (n=3). Error bars depict SD. (b) In the presence of a sufficient number of effector cells (E/T ratio 5:1), ADCC activity mediated by cCD16ζ-T cells against ATL tumor cells was also Mog dose-dependent. However, at pharmacological doses of Mog (>0.1μg/mL), the decline of ADCC activity was limited in comparison to that observed in S2a. LCL, EBV-immortalized B cell line: Mog, mogamulizumab. Each experiment was conducted in triplicate (n=3). Error bars depict SD.
- Published
- 2023
19. Supplementary Table S1 from Development of Engineered T Cells Expressing a Chimeric CD16-CD3ζ Receptor to Improve the Clinical Efficacy of Mogamulizumab Therapy Against Adult T-Cell Leukemia
- Author
-
Masaki Yasukawa, A. John Barrett, Takashi Sugiyama, Hiroshi Shiku, Kiyotaka Kuzushima, Junichi Mineno, Sachiko Okamoto, Nicholas Casey, Kazushi Tanimoto, Fumihiro Ochi, Hiroshi Fujiwara, and Hiroki Tanaka
- Abstract
In our limited experience with Mog monotherapy for relapsed/refractory ATL (including the case shown in Fig.S3), excluding patients who received allo-HSCT and combined therapy with Mog and intensified chemotherapy mSLG15(ref.32) had shown that lymphocyte counts during Mog monotherapy in patients for whom the treatment failed to suppress disease progression (failure n=4) tended to be lower than that when Mog monotherapy successfully controlled disease (successful n=2). Total time points when cell counts were measured were shown, respectively. Data indicated as mean {plus minus} SD, respectively.
- Published
- 2023
20. Supplementary Figure Legends 1-3 from Improved Expression and Reactivity of Transduced Tumor-Specific TCRs in Human Lymphocytes by Specific Silencing of Endogenous TCR
- Author
-
Ikunoshin Kato, Hiroshi Shiku, Masaki Yasukawa, Hiroshi Fujiwara, Hiroaki Ikeda, Junichi Mineno, and Sachiko Okamoto
- Abstract
Supplementary Figure Legends 1-3 from Improved Expression and Reactivity of Transduced Tumor-Specific TCRs in Human Lymphocytes by Specific Silencing of Endogenous TCR
- Published
- 2023
21. Supplementary Figure 2 from Improved Expression and Reactivity of Transduced Tumor-Specific TCRs in Human Lymphocytes by Specific Silencing of Endogenous TCR
- Author
-
Ikunoshin Kato, Hiroshi Shiku, Masaki Yasukawa, Hiroshi Fujiwara, Hiroaki Ikeda, Junichi Mineno, and Sachiko Okamoto
- Abstract
Supplementary Figure 2 from Improved Expression and Reactivity of Transduced Tumor-Specific TCRs in Human Lymphocytes by Specific Silencing of Endogenous TCR
- Published
- 2023
22. Supplementary Figure 3 from Improved Expression and Reactivity of Transduced Tumor-Specific TCRs in Human Lymphocytes by Specific Silencing of Endogenous TCR
- Author
-
Ikunoshin Kato, Hiroshi Shiku, Masaki Yasukawa, Hiroshi Fujiwara, Hiroaki Ikeda, Junichi Mineno, and Sachiko Okamoto
- Abstract
Supplementary Figure 3 from Improved Expression and Reactivity of Transduced Tumor-Specific TCRs in Human Lymphocytes by Specific Silencing of Endogenous TCR
- Published
- 2023
23. Supplementary Figure 1 from Improved Expression and Reactivity of Transduced Tumor-Specific TCRs in Human Lymphocytes by Specific Silencing of Endogenous TCR
- Author
-
Ikunoshin Kato, Hiroshi Shiku, Masaki Yasukawa, Hiroshi Fujiwara, Hiroaki Ikeda, Junichi Mineno, and Sachiko Okamoto
- Abstract
Supplementary Figure 1 from Improved Expression and Reactivity of Transduced Tumor-Specific TCRs in Human Lymphocytes by Specific Silencing of Endogenous TCR
- Published
- 2023
24. Measurement report: Shipborne observations of black carbon aerosols in the western Arctic Ocean during summer and autumn 2016-2020: boreal fire impacts.
- Author
-
Yange Deng, Hiroshi Tanimoto, Kohei Ikeda, Sohiko Kameyama, Sachiko Okamoto, Jinyoung Jung, Young Jun Yoon, Eun Jin Yang, and Sung-Ho Kang
- Abstract
Black carbon (BC) aerosol is considered one of the important contributors to the fast climate warming and snow and sea ice melting in the Arctic. Yet the observations of BC in the Arctic Ocean have been limited due to infrastructural and logistical difficulties. We observed BC mass concentrations (m
BC ) using light absorption methods on board the icebreaker R/V Araon in the Arctic Ocean (166° E-156° W and <80° N) as well as the North Pacific Ocean in summer and early Autumn of 2016 to 2020. The levels, interannual variations and pollution episodes of mBC in the Arctic were examined, and the emission sources responsible for the high-BC episodes were analyzed with global chemistry-transport model simulations. The average mBC in the surface air over the Arctic Ocean (72-80° N) observed in 2019 was over 70 ng m-3 20, which was substantially higher than in other years (approximately 10 ng m-3 ). The much higher mBC observed in 2019 was perhaps due to more frequent wildfires occurred in the Arctic region than in other years. The model suggested that biomass burning composed the largest contribution to the observed BC in the western Arctic Ocean and the marginal seas. For these five years, we identified 10 elevated-BC episodes, including one in 2018 that was associated with co-enhancements of CO and CH4 but not CO2 and O3 . The model analysis indicated that most episodes were attributed to the airmasses transported from boreal fires to the Arctic Ocean, with some near-surface and others in the mid-troposphere. This study provides crucial datasets on BC mass concentrations and the mixing ratios of O3 , CH4 , CO, and CO2 in the western Arctic Ocean regions and highlights the significant impact of boreal fires on the observed Arctic BC during the summer and early autumn months. [ABSTRACT FROM AUTHOR]- Published
- 2023
- Full Text
- View/download PDF
25. Supplementary material to 'Impact of different sources of precursors on a high-ozone event over Europe analysed with IASI+GOME2 multispectral satellite observations and model simulations'
- Author
-
Sachiko Okamoto, Juan Cuesta, Matthias Beekmann, Gaëlle Dufour, Maxim Eremenko, Kazuyuki Miyazaki, Cathy Bonne, Hiroshi Tanimoto, and Hajime Akimoto
- Published
- 2022
26. Impact of different sources of precursors on a high-ozone event over Europe analysed with IASI+GOME2 multispectral satellite observations and model simulations
- Author
-
Sachiko Okamoto, Juan Cuesta, Matthias Beekmann, Gaëlle Dufour, Maxim Eremenko, Kazuyuki Miyazaki, Cathy Bonne, Hiroshi Tanimoto, and Hajime Akimoto
- Abstract
We examine the impact of different sources of ozone precursors on the daily evolution of successive major ozone pollution outbreaks across Europe in July 2017 by using a multispectral satellite approach called IASI+GOME2, and a tropospheric chemistry reanalysis called TCR-2. IASI+GOME2, combining IASI and GOME-2 measurements respectively in the infrared and the ultraviolet, allows the observation of the daily horizontal distribution of ozone in the lowermost troposphere (defined here as the atmospheric layer between the surface and 3 km above sea level). IASI+GOME2 observations show a fair capacity to depict near-surface ozone evolution as compared to surface measurements from 188 European stations for the period 15–27 July 2017. At the beginning of this event (on 16 July), a major ozone outbreak is initially formed over the Iberian Peninsula likely linked with high temperature-induced enhancements of biogenic volatile organic compounds concentrations and collocated anthropogenic emissions. In the following days, the ozone plume splits into two branches, one being transported eastward across the Western Mediterranean and Italy, and the other one over Western and Central Europe. The southern branch encounters ozone precursors emitted over the Balkan Peninsula by wildfires along the coast of the Adriatic Sea and biogenic sources in the inland region of the Peninsula. Ozone concentrations of the northern plume enhance by photochemical production associated with anthropogenic sources of ozone precursors over Central Europe and by mixing with an ozone plume arriving from the North Sea that was originally produced over North America. Finally, both ozone branches are transported eastwards and mix gradually, as they reach the northern coast of the Black Sea. There, emissions from agricultural fires after harvesting clearly favor photochemical production of ozone within the pollution plume, which is advected eastwards in the following days. Based on satellite analysis, this paper shows the interplay of various ozone precursor sources to sustain a two-week long ozone pollution event over different parts of Europe.
- Published
- 2022
27. NY-ESO-1-specific redirected T cells with endogenous TCR knockdown mediate tumor response and cytokine release syndrome
- Author
-
Mikiya Ishihara, Shigehisa Kitano, Shinichi Kageyama, Yoshihiro Miyahara, Noboru Yamamoto, Hidefumi Kato, Hideyuki Mishima, Hiroyoshi Hattori, Takeru Funakoshi, Takashi Kojima, Tetsuro Sasada, Eiichi Sato, Sachiko Okamoto, Daisuke Tomura, Ikuei Nukaya, Hideto Chono, Junichi Mineno, Muhammad Faris Kairi, Phuong Diem Hoang Nguyen, Yannick Simoni, Alessandra Nardin, Evan Newell, Michael Fehlings, Hiroaki Ikeda, Takashi Watanabe, and Hiroshi Shiku
- Subjects
Pharmacology ,Cancer Research ,T-Lymphocytes ,Immunology ,Receptors, Antigen, T-Cell ,Membrane Proteins ,Oncology ,Antigens, Neoplasm ,Neoplasms ,Molecular Medicine ,Immunology and Allergy ,Cytokines ,Humans ,Immunotherapy ,Cytokine Release Syndrome ,Cyclophosphamide - Abstract
BackgroundBecause of the shortage of ideal cell surface antigens, the development of T-cell receptor (TCR)-engineered T cells (TCR-T) that target intracellular antigens such as NY-ESO-1 is a promising approach for treating patients with solid tumors. However, endogenous TCRs in vector-transduced T cells have been suggested to impair cell-surface expression of transduced TCR while generating mispaired TCRs that can become self-reactive.MethodsWe conducted a first-in-human phase I clinical trial with the TCR-transduced T-cell product (TBI-1301) in patients with NY-ESO-1-expressing solid tumors. In manufacturing TCR-T cells, we used a novel affinity-enhanced NY-ESO-1-specific TCR that was transduced by a retroviral vector that enables siRNA (small interfering RNA)-mediated silencing of endogenous TCR. The patients were divided into two cohorts. Cohort 1 was given a dose of 5×108 cells (whole cells including TCR-T cells) preconditioned with 1500 mg/m2 cyclophosphamide. Cohort 2 was given 5× 109 cells preconditioned with 1500 mg/m2 cyclophosphamide.ResultsIn vitro study showed that both the CD8+ and CD4+ T fractions of TCR-T cells exhibited cytotoxic effects against NY-ESO-1-expressing tumor cells. Three patients and six patients were allocated to cohort 1 and cohort 2, respectively. Three of the six patients who received 5×109 cells showed tumor response, while three patients developed early-onset cytokine release syndrome (CRS). One of the patients developed a grade 3 lung injury associated with the infiltration of the TCR-T cells. No siRNA-related adverse events other than CRS were observed. Cytokines including interleukin 6 I and monocyte chemotactic protein-1/chemokine (C-C motif) ligand (CCL2)increased in the sera of patients with CRS. In vitro analysis showed these cytokines were not secreted from the T cells infused. A significant fraction of the manufactured T cells in patients with CRS was found to express either CD244, CD39, or both at high levels.ConclusionsThe trial showed that endogenous TCR-silenced and affinity-enhanced NY-ESO-1 TCR-T cells were safely administered except for grade 3 lung injury. The TCR-T cell infusion exhibited significant tumor response and early-onset CRS in patients with tumors that express NY-ESO-1 at high levels. The differentiation properties of the manufactured T cells may be prognostic for TCR-T-related CRS.Trial registration numberNCT02366546.
- Published
- 2022
28. Color Temperature Estimation of Fluorescent Scene Illumination.
- Author
-
Shoji Tominaga and Sachiko Okamoto
- Published
- 2003
- Full Text
- View/download PDF
29. Reflectance-based material classification for printed circuit boards.
- Author
-
Shoji Tominaga and Sachiko Okamoto
- Published
- 2003
- Full Text
- View/download PDF
30. Tumor Growth Suppression of Pancreatic Cancer Orthotopic Xenograft Model by CEA-Targeting CAR-T Cells
- Author
-
Osamu Sato, Takahiro Tsuchikawa, Takuma Kato, Yasunori Amaishi, Sachiko Okamoto, Junichi Mineno, Yuta Takeuchi, Katsunori Sasaki, Toru Nakamura, Kazufumi Umemoto, Tomohiro Suzuki, Linan Wang, Yizheng Wang, Kanako C. Hatanaka, Tomoko Mitsuhashi, Yutaka Hatanaka, Hiroshi Shiku, and Satoshi Hirano
- Subjects
Cancer Research ,orthotopic xenograft mouse model ,Oncology ,chimeric antigen receptor engineered T cell ,carcinoembryonic antigen ,adoptive cell therapy ,pancreatic ductal carcinoma - Abstract
Simple Summary Pancreatic ductal adenocarcinoma is one of the most lethal malignancies, and there are vast unmet medical needs. In this study, we hypothesized that chimeric antigen receptor engineered T cell (CAR-T) targeting carcinoembryonic antigen (CEA) would be effective in the treatment of pancreatic ductal adenocarcinoma. In vivo experiments in a more clinically similar environment were considered necessary; we examined the antitumor effects of adoptive anti-CEA-CAR-T, using orthotopic xenograft mouse models of pancreatic ductal adenocarcinoma. As result, the therapeutic effect of anti-CEA-CAR-T therapy was related to the CEA expression level. Furthermore, the retrospective analysis of pathological findings from pancreatic ductal adenocarcinoma patients showed a correlation between the intensity of CEA immunostaining and tumor heterogeneity. These findings show that anti-CEA-CAR-T therapy can be useful for pancreatic ductal adenocarcinoma; furthermore, the pathological findings of CEA can be clinically used as biomarkers to select cases for anti-CEA-CAR-T therapy. Chimeric antigen receptor engineered T cell (CAR-T) therapy has high therapeutic efficacy against blood cancers, but it has not shown satisfactory results in solid tumors. Therefore, we examined the therapeutic effect of CAR-T therapy targeting carcinoembryonic antigen (CEA) in pancreatic adenocarcinoma (PDAC). CEA expression levels on the cell membranes of various PDAC cell lines were evaluated using flow cytometry and the cells were divided into high, medium, and low expression groups. The relationship between CEA expression level and the antitumor effect of anti-CEA-CAR-T was evaluated using a functional assay for various PDAC cell lines; a significant correlation was observed between CEA expression level and the antitumor effect. We created orthotopic PDAC xenograft mouse models and injected with anti-CEA-CAR-T; only the cell line with high CEA expression exhibited a significant therapeutic effect. Thus, the therapeutic effect of CAR-T therapy was related to the target antigen expression level, and the further retrospective analysis of pathological findings from PDAC patients showed a correlation between the intensity of CEA immunostaining and tumor heterogeneity. Therefore, CEA expression levels in biopsies or surgical specimens can be clinically used as biomarkers to select PDAC patients for anti-CAR-T therapy.
- Published
- 2023
31. 103 Quality improvement of anti-CD38-JAK/STAT CAR-T cells by suppressing CD38 expression and inhibition of tyrosine kinase
- Author
-
Yasunori Amaishi, Junichi Mineno, Kenichiro Mihara, Maiko Sugizaki, Sachiko Okamoto, and Izumi Maki
- Subjects
Pharmacology ,Cancer Research ,Chemistry ,Immunology ,JAK-STAT signaling pathway ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,CD38 ,Cd38 expression ,Oncology ,hemic and lymphatic diseases ,Cancer research ,Molecular Medicine ,Immunology and Allergy ,Car t cells ,human activities ,Tyrosine kinase ,RC254-282 - Abstract
BackgroundCAR-T cell therapy has shown highly effective clinical results in several diseases, but further improvement is necessary to target a wider range of antigens and tumors. In particular, excessive activation of CAR-T cells leads to cell exhaustion and reduction of naive/memory T cells’ population, which are important for long-term immune response. Therefore, suppressing non-antigen-specific activity is necessary for CAR-T cell production. However, when targeting tumor-related antigens that are also expressed on T cells, CAR-T cells recognize the antigens on the T cells, resulting in fratricide, poor cell growth, differentiation, and exhaustion during cell production process.In this study, we investigated a method for producing CAR-T cells targeting CD38 antigen that is common to T cells and tumor cells. CD38 is a suitable target antigen for CAR-T cell therapy because it is highly expressed in lymphocyte malignant tumors including B-cell non-Hodgkin’s lymphoma and multiple myeloma. However, as it is also intermediately expressed in normal blood cells, unwanted activation of CAR-T cells may be caused.MethodsWe tried to suppress the expression of CD38 in CAR-T cells by co-expressing CD38 siRNAs, and prevent activation during cell production by modifying the signal domain of anti-CD38-CAR to the newly developed JAK/STAT-CAR. JAK/STAT-CAR contains the intracellular domain of the IL-2 receptor β chain and the STAT3 binding motif, which have been shown to improve the proliferation of CAR-T cells and suppresses differentiation compared to conventional second-generation CAR-T cells.For further improvement, CAR-T cells were prepared in the presence of the tyrosine kinase inhibitor Dasatinib to suppress activation during the cell manufacturing process.ResultsCD38 siRNA co-expressing CAR-T cells showed decreased expression of CD38 and exhaustion markers, and the further reduction of exhaustion marker expression was observed in JAK/STAT CAR-T cells. However, compared to CAR-T cells targeting other antigens, CD38-CAR-T cells tended to be more exhausted and differentiated. As Dasatinib treatment maintained a high proportion of naive/memory T cells and was able to suppress exhaustion, combination of these approaches (CD38 siRNA-expressing CD38-JAK/STAT CAR-T cells with Dasatinib treatment) showed long-term persistence of antitumor activity in in vitro re-challenge assay.ConclusionsCD38 siRNA co-expressing CD38-JAK/STAT CAR-T cells produced in the presence of a tyrosine kinase inhibitor are expected to be suppressed excessive activation and maintain long-term antigen-specific activity. This approach is also expected to be applied to other CAR-T cell therapies targeting tumor-related antigens expressed on T cells.
- Published
- 2021
32. Highly efficient genome editing for single-base substitutions using optimized ssODNs with Cas9-RNPs
- Author
-
Tatsuji Enoki, Sachiko Okamoto, Junichi Mineno, Yasunori Amaishi, and Izumi Maki
- Subjects
0301 basic medicine ,Induced Pluripotent Stem Cells ,Cell Culture Techniques ,Oligonucleotides ,DNA, Single-Stranded ,lcsh:Medicine ,Computational biology ,Transfection ,Article ,Homology (biology) ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Genome editing ,Humans ,Gene Knock-In Techniques ,lcsh:Science ,Gene ,Subgenomic mRNA ,Ribonucleoprotein ,Gene Editing ,Multidisciplinary ,Base Sequence ,Chemistry ,Cas9 ,lcsh:R ,HEK293 Cells ,030104 developmental biology ,Ribonucleoproteins ,Feasibility Studies ,lcsh:Q ,CRISPR-Cas Systems ,Homologous recombination ,030217 neurology & neurosurgery ,DNA ,RNA, Guide, Kinetoplastida - Abstract
Target-specific genome editing using engineered nucleases has become widespread in various fields. Long gene knock-in and single-base substitutions can be performed by homologous recombination (HR), but the efficiency is usually very low. To improve the efficiency of knock-in with single-stranded oligo DNA nucleotides (ssODNs), we have investigated optimal design of ssODNs in terms of the blocking mutation, orientation, size, and length of homology arms to explore the optimal parameters of ssODN design using reporter systems for the detection of single-base substitutions. We have also investigated the difference in knock-in efficiency among the delivery forms and methods of Cas9 and sgRNA. The knock-in efficiencies for optimized ssODNs were much higher than those for ssODNs with no blocking mutation. We have also demonstrated that Cas9 protein/sgRNA ribonucleoprotein complexes (Cas9-RNPs) can dramatically reduce the re-cutting of the edited sites.
- Published
- 2019
33. Preclinical Study of DNA Vaccines Targeting SARS-CoV-2
- Author
-
Junichi Mineno, Ritsuko Kubota-Koketsu, Shota Yoshida, Munehisa Shimamura, Akiko Tenma, Jiao Sun, Yoshimi Saito, Hisashi Arase, Makoto Sakaguchi, Hideto Chono, Yasunori Amaishi, Tatsuo Shioda, Ryo Nakamaru, Yoshiharu Matsuura, Hiromi Rakugi, Hiroki Hayashi, Hironori Nakagami, Hideki Tomioka, Sotaro Kawabata, Chikako Ono, Takako Ehara, Yuka Yanagida, Ryoko Ide, Takao Komatsuno, Nan Ju, Takako Ootera, Ryuichi Morishita, and Sachiko Okamoto
- Subjects
History ,Polymers and Plastics ,biology ,business.industry ,medicine.medical_treatment ,Genetic enhancement ,Virology ,Industrial and Manufacturing Engineering ,Viral vector ,DNA vaccination ,law.invention ,Epitope mapping ,law ,medicine ,biology.protein ,Recombinant DNA ,Business and International Management ,Alum adjuvant ,Antibody ,business ,Adjuvant - Abstract
As potential pandemic vaccines, DNA/RNA vaccines, viral vector vaccines and protein-based vaccines have been rapidly developed to prevent pandemic spread worldwide. In this study, we designed plasmid DNA vaccine targeting the SARS-CoV-2 Spike glycoprotein (S protein) as pandemic vaccine, and the humoral, cellular, and functional immune responses were characterized to support proceeding to initial human clinical trials. After intramuscular injection of DNA vaccine encoding S protein with alum adjuvant (three times at 2-week intervals), the humoral immunoreaction, as assessed by anti-S protein or anti-receptor-binding domain (RBD) antibody titers, and the cellular immunoreaction, as assessed by antigen-induced IFNγ expression, were up-regulated. In IgG subclass analysis, IgG2b was induced as the main subclass. Based on these analyses, DNA vaccine with alum adjuvant preferentially induced Th1-type T cell polarization. We confirmed the neutralizing action of DNA vaccine-induced antibodies by a binding assay of RBD recombinant protein with angiotensin-converting enzyme 2 (ACE2), a receptor of SARSCoV-2, and pseudo-virus assay, TCID assay with live SARS-CoV-2. Further B cell epitope mapping analysis using a peptide array showed that most vaccine-induced antibodies recognized the S2 and RBD subunits. Finally, DNA vaccine protected hamsters form SARSCoV-2 infection. In conclusion, DNA vaccine targeting the spike glycoprotein of SARS-CoV-2 might be an effective and safe approach to combat the COVID-19 pandemic. Funding: This study was supported by Project Promoting Support for Drug Discovery grants (JP20nk0101602 and JP21nf0101623h102) from the Japan Agency for Medical Research and Development and Panasonic Co. (Japan). To fight against the worldwide COVID-19 pandemic, the development of an effective and safe The Department of Health Development and Medicine is an endowed department supported by Anges, Daicel, and FunPep. The Department of Clinical Gene Therapy is financially supported by Novartis, AnGes, Shionogi, Boeringher, Fancl, Saisei Mirai Clinics, Rohto and Funpep. Declaration of Interest: R.M. is a stockholder of FunPep and Anges. T.O. T.K. and Y.S. are employees of Anges. R.I, A.T, H.K, S.K, E.T, S.M, and H.T are employees of FunPep. R.M, H.T, and A.T. are FunPep stockholders. All other authors declare no competing interests. Ethical Approval: All experiments were approved by the Ethical Committee for Animal Experiments of the Osaka University Graduate School of Medicine.
- Published
- 2021
34. 129 A novel CAR conducting antigen-specific JAK-STAT signals demonstrates superior antitumor effects with minimal undesired non-specific activation
- Author
-
Naoto Hirano, Yasunori Amaishi, Sachiko Okamoto, Yu Okubo, Junichi Mineno, Yota Ohashi, and Mitsuki Shigeta
- Subjects
biology ,Chemistry ,T cell ,T-cell receptor ,CD28 ,JAK-STAT signaling pathway ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,lcsh:RC254-282 ,CD19 ,Chimeric antigen receptor ,Cell biology ,medicine.anatomical_structure ,Antigen ,biology.protein ,medicine ,Signal transduction - Abstract
Background Despite recent impressive successes in chimeric antigen receptor (CAR)-T cell therapy, there are still considerable clinical challenges. To improve T cell persistence and antitumor effect, which are critical for clinical responses, various efforts have been made to optimize the CAR design such as the inclusion of a costimulatory domain(s). It is known that non-specific activation of CAR-T cells is greatly influenced by the CAR design, and excessive T cell activation leads exhaustion of T cells and depletion of naive/memory subsets important for durable clinical responses. Thus, the CAR construct needs to be optimized so that transduced T cells persist and induce potent antigen-specific response with reduced non-specific activation. For optimal T cell activation and proliferation, three signals including TCR (signal 1), co-stimulatory (signal 2), and cytokine (signal 3) signals, are essential. The conventional second and third generation CARs containing CD3ζ and a co-stimulatory domain such as a signal domain of CD28 and 4-1BB can conduct signal 1 and 2, but not signal 3. Recently, we have developed a new generation JAK-STAT CAR composed of a truncated cytoplasmic domain of the IL-2 receptor β chain and STAT3/5 binding motifs, CD28 co-stimulatory domain, and CD3ζ domain. The novel anti-CD19 JAK-STAT CAR-T cells showed antigen-specific activation of the JAK-STAT signaling pathway, enhanced proliferation, and limited terminal differentiation in vitro compared to second generation 28ζ CAR or 4-1BBζ CAR-transduced T cells. Furthermore, the anti-CD19 JAK-STAT CAR-T cells demonstrated superior in vivo persistence and antitumor effect in mouse models.1 In addition, we previously showed that a hinge region and the composition of a single chain variable fragment (scFv) such as the order of VH and VL regions critically influence not only antigen-dependent activation but also undesired antigen-independent activation known as tonic signaling.2 Methods In this study, we have optimized the scFv design in 28ζ CAR and JAK-STAT CAR constructs to show superior antigen-specific activation and reduced tonic signaling for several targets (CD19, CD20, Mesothelin, and GD2). And we have evaluated the feature of JAK-STAT CAR-T cells compared to 28ζ CAR-T cells. Results JAK-STAT CAR-T cells showed superior antigen-specific proliferation with less differentiated status, whereas 28ζ CAR-T cells showed antigen-independent proliferation and displayed higher exhaustion marker expression after repetitive stimulations. Conclusions These results suggest that our JAK-STAT-CARs with enhanced antigen-specific response with minimized tonic signaling targeting various antigens has the potential to demonstrate improved clinical efficacy. References Kagoya Y, et al. A novel chimeric antigen receptor containing a JAK–STAT signaling domain mediates superior antitumor effects. Nat Med 2018;24:p352–359. Okamoto S, et al. Detail analysis of non-specific activation of CD19 CAR-T cells caused by CAR design. ASGCT ( 2015)
- Published
- 2020
35. 99 Structural optimization of anti-CEA-GITR-CAR to reduce tonic signaling and improve antigen-specific reactivity
- Author
-
Sachiko Okamoto, Yu Okubo, Junichi Mineno, Yizheng Wang, Takuma Kato, Linan Wang, Hiroshi Shiku, and Yasunori Amaishi
- Subjects
LAG3 ,biology ,Chemistry ,T cell ,Cell ,CD28 ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,lcsh:RC254-282 ,Chimeric antigen receptor ,CD19 ,Cell therapy ,medicine.anatomical_structure ,Antigen ,Cancer research ,medicine ,biology.protein ,human activities - Abstract
Background Adoptive immunotherapy using chimeric antigen receptor (CAR) is recently reported as one of the effective cancer therapy. Especially CAR-T cell therapy targeting CD19 antigen in B-cell tumors have shown impressive clinical results and CAR-T cell products targeting CD19 have already approved. However as the high relapse rate is still the problem and the clinical efficacy of CAR-T cell therapy for solid tumors is currently inadequate, further improvement of CAR design is required.It is known that the design of CAR construct affects the function of CAR-T cells. For example co-stimulatory domain such as CD28 and 4-1BB is used in the second generation CARs, CD28z-CAR-T cells show higher anti-tumor activity, whereas 4-1BBz-CAR-T cells demonstrate superior in vivo persistence. To enhance survival of T cells, several attempts had been made to optimize the signaling domains. Recently, we have developed the novel CARs incorporated GITR (glucocorticoid-induced tumor necrosis factor receptor family-related protein) intracellular domain for T cell survival prolongation and inhibition of regulatory T cells’ suppressive activity. It is also reported that the antigen-nonspecific activation of CAR-T cells (tonic signaling) is influenced by the CAR design, and excessive T cell activation leads exhaustion of CAR-T cells. Previously, we have found that the design of CAR, not only single chain variable fragments (scFvs), affect the strength of tonic signaling. Thus, the optimization of CAR construct is essential to induce antigen-specific response with minimal non-specific activation, which results in maximal efficacy. Methods We have optimized the structure of anti-CEA-GITR-CAR targeting CEA antigen expressing solid tumor such as gastric and pancreatic cancer. We have constructed several CARs with the different composition such as hinge region, transmembrane domain, and the order of VL/VH in scFV region, and compared the tonic signaling and antigen-specific activity in CAR-T cells. Results The property of CAR-T cells was largely affected by the CAR constructs, especially the hinge region. The CAR-T cells with CD8α hinge showed strong tonic signaling, the CAR-T cells with short hinge-CAR lost antigen specificity, and elimination of hinge region lowered the CAR expression level and antigen reactivity. Furthermore, GITR-CAR-T cells showed higher proportion of CCR7+CD45RA+ cells and lower expression of exhaustion markers (PD1, Tim3, and LAG3) compared to CD28z-CAR-T cells. Conclusions Our CEA-GITR-CAR with the optimized scFV design and CD28-hinge demonstrated improved antigen-specific response with reduced tonic signaling, potentially indicating that our novel CAR-T cells may show improved clinical efficacy on solid tumor.
- Published
- 2020
36. Preclinical study of DNA vaccines targeting SARS-CoV-2
- Author
-
Takako Otera, Shota Yoshida, Yoshimi Saito, Yasunori Amaishi, Hiroki Hayashi, Ryo Nakamaru, Sotaro Kawabata, Chikako Ono, Takako Ehara, Sachiko Okamoto, Akiko Tenma, Hisashi Arase, Ritsuko Kubota-Kotetsu, Makoto Sakaguchi, Hironori Nakagami, Tatsuo Shioda, Junichi Mineno, Hideki Tomioka, Yuka Yanagida, Yoshiharu Matsuura, Munehisa Shimamura, Jiao Sun, Hideto Chono, Hiromi Rakugi, Ryuichi Morishita, Takao Komatsuno, and Ryoko Ide
- Subjects
Immune system ,Epitope mapping ,biology ,law ,Protein subunit ,biology.protein ,Recombinant DNA ,Antibody ,Alum adjuvant ,Virology ,DNA vaccination ,Viral vector ,law.invention - Abstract
To fight against the worldwide COVID-19 pandemic, the development of an effective and safe vaccine against SARS-CoV-2 is required. As potential pandemic vaccines, DNA/RNA vaccines, viral vector vaccines and protein-based vaccines have been rapidly developed to prevent pandemic spread worldwide. In this study, we designed plasmid DNA vaccine targeting the SARS-CoV-2 Spike glycoprotein (S protein) as pandemic vaccine, and the humoral, cellular, and functional immune responses were characterized to support proceeding to initial human clinical trials. After intramuscular injection of DNA vaccine encoding S protein with alum adjuvant (three times at 2-week intervals), the humoral immunoreaction, as assessed by anti-S protein or anti-receptor-binding domain (RBD) antibody titers, and the cellular immunoreaction, as assessed by antigen-induced IFNγ expression, were up-regulated. In IgG subclass analysis, IgG2b was induced as the main subclass. Based on these analyses, DNA vaccine with alum adjuvant preferentially induced Th1-type T cell polarization. We confirmed the neutralizing action of DNA vaccine-induced antibodies via two different methods, a binding assay of RBD recombinant protein with angiotensin-converting enzyme 2 (ACE2), a receptor of SARS-CoV-2, and pseudovirus assay. Further B cell epitope mapping analysis using a peptide array showed that most vaccine-induced antibodies recognized the S2 and RBD subunits, but not the S1 subunit. In conclusion, DNA vaccine targeting the spike glycoprotein of SARS-CoV-2 might be an effective and safe approach to combat the COVID-19 pandemic.
- Published
- 2020
37. 64 A cloning and expression system of the neoantigen-specific TCRs from tumor-infiltrating lymphocytes by single-cell sequencing of paired TCRα and TCRβ chains
- Author
-
Junichi Mineno, Toshikazu Nishie, Sachiko Okamoto, Koji Nagaoka, Jun Nakajima, Kazuhiro Kakimi, Yukari Kobayashi, Kaori Kubo, Shunji Takahashi, Yasuyoshi Sato, and Tatsuji Enoki
- Subjects
Pharmacology ,Cloning ,Cancer Research ,Oncology ,Single cell sequencing ,Tumor-infiltrating lymphocytes ,Immunology ,Molecular Medicine ,Immunology and Allergy ,hemic and immune systems ,chemical and pharmacologic phenomena ,Biology ,Molecular biology - Abstract
BackgroundT-cells that target tumor neoantigens arising from cancer mutations are the primary mediators of cancer immunotherapies. Identifying neoantigens and T-cells that recognize them is essential for T-cell-based immunotherapy. However, neoantigen-reactive Tumor-infiltrating lymphocytes (TILs) are highly differentiated or exhausted with a limited proliferative capacity; it is challenging to expand them for a sufficient number to probe their specificity. Therefore, we developed a novel cloning and expression system to examine TCRs discovered by single-cell sequencing of TILs for their neoantigen-specificity.MethodsTILs of lung cancer and sarcoma were analyzed. Surgically removed tumors were divided into several pieces. They were enzymatically digested to prepare fresh tumor digest (FTD) and cryopreserved. They were used to generate TIL cultures and perform WES and RNA-Seq to identify tumor-specific mutations. MHCflurry was used to predict the binding affinity of potential epitopes arising from these mutations to HLA class I. Peptides that were predicted to bind to patients‘ own MHC class I molecules strongly were then synthesized. Single TILs isolated with the ICELL8® cx system (Takara Bio) were dispensed into a nanowell TCR chip containing preprinted barcodes. Barcoded cDNAs were PCR-amplified in-chip, pooled off-chip, and used as a template in the TCR-specific PCR or for the whole transcriptome library generation of 5’ ends of all transcripts. Based on single-cell transcriptome data and TCR profiles of TILs, we predict and prioritize neoantigen-specific TCRs and cloned them into siTCR® retrovirus vectors. These TCRs were transduced into SUP-T1-based reporter cells in which ZsGreen fluorescent protein expression is controlled by AP-1 and NFAT binding sites. TCR-expressing reporter cells were cocultured with patient autologous APCs pulsed with a pool of candidate neoantigen peptides. ZsGreen expression indicates that TCRs match their cognate neoantigens.ResultsIn a lung cancer patient, we set up 18 TIL cultures and obtained 12 TILs. TILs were cocultured with FTD; IFN-γ production was measured by ELISA to evaluate their reactivity to the autologous tumor. NGS identified 197 somatic mutations, 4 fusion genes, and 8 highly expressed cancer-testis antigens. Among them, 339 candidate peptides were synthesized and screened. In addition, we cloned 3 pairs of TCRαβ chains from most expanded TIL cultures and 4 TCRs from ex vivo TILs with exhausted phenotype. Two reporter cells that express TCRs from exhausted TILs responded to the same neoantigen peptide.ConclusionsGenerating TCR expressing cell lines facilitated the identifying neoantigens and their cognate TCR sequences from patients.Ethics ApprovalG3545
- Published
- 2021
38. Safety and persistence of WT1-specific T-cell receptor gene−transduced lymphocytes in patients with AML and MDS
- Author
-
Nobuhiko Emi, Naoki Inoue, Sachiko Okamoto, Naoyuki Katayama, Isao Tawara, Tomohide Kidokoro, Ikuei Nukaya, Junichi Mineno, Kazushi Tanimoto, Hiroshi Fujiwara, Yoshiki Akatsuka, Yoshihiro Miyahara, Tetsuya Nishida, Hideto Chono, Tomoki Naoe, Masaki Yasukawa, Seitaro Terakura, Makoto Murata, Yoko Inaguma, Daisuke Tomura, Hiroshi Shiku, Hiroaki Ikeda, Masahiro Masuya, and Shinichi Kageyama
- Subjects
Male ,0301 basic medicine ,Adoptive cell transfer ,Myeloid ,Acute myeloblastic leukemia ,T-Lymphocytes ,Immunology ,Biochemistry ,03 medical and health sciences ,0302 clinical medicine ,Bone Marrow ,Transduction, Genetic ,medicine ,Humans ,WT1 Proteins ,Aged ,Acute leukemia ,business.industry ,Cell Biology ,Hematology ,Middle Aged ,medicine.disease ,Adoptive Transfer ,Genes, T-Cell Receptor ,Kinetics ,Leukemia, Myeloid, Acute ,Leukemia ,Haematopoiesis ,030104 developmental biology ,medicine.anatomical_structure ,Myelodysplastic Syndromes ,030220 oncology & carcinogenesis ,Female ,Bone marrow ,Peptides ,business ,Ex vivo - Abstract
Wilms' tumor 1 (WT1) is constantly expressed in leukemic cells of acute leukemia and myelodysplastic syndrome (MDS). A T-cell receptor (TCR) that specifically reacts with WT1 peptide in the context of HLA-A*24:02 has been identified. We conducted a first-in-human trial of TCR-gene transduced T-cell (TCR-T-cell) transfer in patients with refractory acute myeloblastic leukemia (AML) and high-risk MDS to investigate the safety and cell kinetics of the T cells. The WT1-specific TCR-gene was transduced to T cells using a retroviral vector encoding small interfering RNAs for endogenous TCR genes. The T cells were transferred twice with a 4-week interval in a dose-escalating design. After the second transfer, sequential WT1 peptide vaccines were given. Eight patients, divided into 2 dose cohorts, received cell transfer. No adverse events of normal tissue were seen. The TCR-T cells were detected in peripheral blood for 8 weeks at levels proportional to the dose administered, and in 5 patients, they persisted throughout the study period. The persisting cells maintained ex vivo peptide-specific immune reactivity. Two patients showed transient decreases in blast counts in bone marrow, which was associated with recovery of hematopoiesis. Four of 5 patients who had persistent T cells at the end of the study survived more than 12 months. These results suggest WT1-specific TCR-T cells manipulated by ex vivo culture of polyclonal peripheral lymphocytes survived in vivo and retained the capacity to mount an immune reaction to WT1. This trial was registered at www.umin.ac.jp as #UMIN000011519.
- Published
- 2017
39. Development of Engineered T Cells Expressing a Chimeric CD16-CD3ζ Receptor to Improve the Clinical Efficacy of Mogamulizumab Therapy Against Adult T-Cell Leukemia
- Author
-
Hiroshi Shiku, Kiyotaka Kuzushima, Takashi Sugiyama, Fumihiro Ochi, Nicholas Casey, Hiroshi Fujiwara, A. John Barrett, Hiroki Tanaka, Junichi Mineno, Masaki Yasukawa, Kazushi Tanimoto, and Sachiko Okamoto
- Subjects
0301 basic medicine ,Cancer Research ,Adoptive cell transfer ,Receptors, CCR4 ,CD3 Complex ,Recombinant Fusion Proteins ,T-Lymphocytes ,medicine.medical_treatment ,Genetic Vectors ,T-cell leukemia ,Gene Expression ,T-Cell Antigen Receptor Specificity ,Hematopoietic stem cell transplantation ,Antibodies, Monoclonal, Humanized ,Immunotherapy, Adoptive ,Mice ,03 medical and health sciences ,Antineoplastic Agents, Immunological ,0302 clinical medicine ,Transduction, Genetic ,immune system diseases ,Cell Line, Tumor ,medicine ,Mogamulizumab ,Animals ,Humans ,Leukemia-Lymphoma, Adult T-Cell ,Antibody-dependent cell-mediated cytotoxicity ,business.industry ,Lentivirus ,Receptors, IgG ,Antibody-Dependent Cell Cytotoxicity ,hemic and immune systems ,Immunotherapy ,medicine.disease ,Combined Modality Therapy ,Xenograft Model Antitumor Assays ,Killer Cells, Natural ,Disease Models, Animal ,Leukemia ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Monoclonal ,Immunology ,Female ,business ,medicine.drug - Abstract
Purpose: Mogamulizumab (Mog), a humanized anti-CC chemokine receptor 4 (CCR4) mAb that mediates antibody-dependent cellular cytotoxicity (ADCC) using FcγR IIIa (CD16)-expressing effector cells, has recently been approved for treatment of CCR4-positive adult T-cell leukemia (ATL) in Japan. However, Mog failure has sometimes been observed in patients who have accompanying chemotherapy-associated lymphocytopenia. In this study, we examined whether adoptive transfer of artificial ADCC effector cells combined with Mog would overcome this drawback. Experimental Design: We lentivirally gene-modified peripheral blood T cells from healthy volunteers and ATL patients expressing the affinity-increased chimeric CD16-CD3ζ receptor (cCD16ζ-T cells). Subsequently, we examined the ADCC effect mediated by those cCD16ζ-T cells in the presence of Mog against ATL tumor cells both in vitro and in vivo. Results: cCD16ζ-T cells derived from healthy donors killed in vitro Mog-opsonized ATL cell line cells (n = 7) and primary ATL cells (n = 4) depending on both the number of effector cells and the dose of the antibody. cCD16ζ-T cells generated from ATL patients (n = 3) also exerted cytocidal activity in vitro against Mog-opsonized autologous ATL cells. Using both intravenously disseminated model (n = 5) and subcutaneously inoculated model (n = 4), coadministration of Mog and human cCD16ζ-T cells successfully suppressed tumor growth in xenografted immunodeficient mice, and significantly prolonged their survival (P < 0.01 and P = 0.02, respectively). Conclusions: These data strongly suggest clinical feasibility of the novel combined adoptive immunotherapy using cCD16ζ-T cells and Mog for treatment of aggressive ATL, particularly in patients who are ineligible for allogeneic hematopoietic stem cell transplantation. Clin Cancer Res; 22(17); 4405–16. ©2016 AACR.
- Published
- 2016
40. Antitumor activity of CAR-T cells targeting the intracellular oncoprotein WT1 can be enhanced by vaccination
- Author
-
Motohiro Yoneyama, Satoshi Okumura, Yasushi Akahori, Hiroshi Shiku, Yoshiki Akatsuka, Takehiro Maki, Naohiro Seo, Linan Wang, Junichi Mineno, Hiroshi Fujiwara, Yasunori Amaishi, Hiroaki Ikeda, Sachiko Okamoto, Takuma Kato, and Yoshihiro Miyahara
- Subjects
0301 basic medicine ,Immunobiology and Immunotherapy ,medicine.medical_treatment ,T-Lymphocytes ,Immunology ,chemical and pharmacologic phenomena ,Major histocompatibility complex ,urologic and male genital diseases ,Biochemistry ,Immunotherapy, Adoptive ,Cell therapy ,03 medical and health sciences ,Cancer immunotherapy ,Antigen ,In vivo ,Cell Line, Tumor ,Neoplasms ,medicine ,Animals ,Humans ,WT1 Proteins ,Immunity, Cellular ,Receptors, Chimeric Antigen ,biology ,Chemistry ,urogenital system ,Vaccination ,Cell Biology ,Hematology ,Immunotherapy ,respiratory system ,Xenograft Model Antitumor Assays ,Chimeric antigen receptor ,female genital diseases and pregnancy complications ,030104 developmental biology ,Cell culture ,Cancer research ,biology.protein ,human activities - Abstract
The recent success of chimeric antigen receptor (CAR)-T cell therapy for treatment of hematologic malignancies supports further development of treatments for both liquid and solid tumors. However, expansion of CAR-T cell therapy is limited by the availability of surface antigens specific for the tumor while sparing normal cells. There is a rich diversity of tumor antigens from intracellularly expressed proteins that current and conventional CAR-T cells are unable to target. Furthermore, adoptively transferred T cells often suffer from exhaustion and insufficient expansion, in part, because of the immunosuppressive mechanisms operating in tumor-bearing hosts. Therefore, it is necessary to develop means to further activate and expand those CAR-T cells in vivo. The Wilms tumor 1 (WT1) is an intracellular oncogenic transcription factor that is an attractive target for cancer immunotherapy because of its overexpression in a wide range of leukemias and solid tumors, and a low level of expression in normal adult tissues. In the present study, we developed CAR-T cells consisting of a single chain variable fragment (scFv) specific to the WT1235-243/HLA-A*2402 complex. The therapeutic efficacy of our CAR-T cells was demonstrated in a xenograft model, which was further enhanced by vaccination with dendritic cells (DCs) loaded with the corresponding antigen. This enhanced efficacy was mediated, at least partly, by the expansion and activation of CAR-T cells. CAR-T cells shown in the present study not only demonstrate the potential to expand the range of targets available to CAR-T cells, but also provide a proof of concept that efficacy of CAR-T cells targeting peptide/major histocompatibility complex can be boosted by vaccination.
- Published
- 2017
41. Gene-Modified Human α/β-T Cells Expressing a Chimeric CD16-CD3ζ Receptor as Adoptively Transferable Effector Cells for Anticancer Monoclonal Antibody Therapy
- Author
-
Hiroaki Asai, Junichi Mineno, Hiroshi Fujiwara, Kazushi Tanimoto, Fumihiro Ochi, Sachiko Okamoto, John Barrett, Kiyotaka Kuzushima, Masaki Yasukawa, Eiichi Ishii, Yukihiro Miyazaki, and Hiroshi Shiku
- Subjects
Cancer Research ,Adoptive cell transfer ,CD3 Complex ,Recombinant Fusion Proteins ,T-Lymphocytes ,Immunology ,Antineoplastic Agents ,chemical and pharmacologic phenomena ,Mice, SCID ,Biology ,GPI-Linked Proteins ,Mice ,Interleukin 21 ,NK-92 ,Cell Line, Tumor ,Animals ,Humans ,IL-2 receptor ,CD40 ,Lymphokine-activated killer cell ,Receptors, IgG ,Antibodies, Monoclonal ,Adoptive Transfer ,Molecular biology ,Cancer cell ,biology.protein ,Interleukin 12 ,Female - Abstract
The central tumoricidal activity of anticancer monoclonal antibodies (mAb) is exerted by FcγR IIIa (CD16)–expressing effector cells in vivo via antibody-dependent cell-mediated cytotoxicity (ADCC), as observed for natural killer (NK) cells. In practice, chemotherapy-induced leukopenia and exhaustion of NK cells resulting from ADCC often hamper the clinical efficacy of cancer treatment. To circumvent this drawback, we examined in vivo the feasibility of T cells, gene-modified to express a newly generated affinity-matured (158V/V) chimeric CD16-CD3ζ receptor (cCD16ζ-T cells), as a transferable alternative effector for cancer mAb therapy. cCD16ζ-T cells were readily expandable in ex vivo culture using anti-CD2/CD3/CD28 beads and recombinant human interleukin-2 (rhIL-2), and they successfully displayed ADCC-mediated tumoricidal activity in vitro. During ADCC, ligation of opsonized cancer cells to the introduced cCD16ζ-T cells stimulated the effector cells to produce proinflammatory cytokines and release toxic granules through the activation of the Nuclear factor of activated T cells (NFAT) pathway after phosphorylation of the CD3ζ chain. In parallel, these stimulated cCD16ζ-T cells transiently proliferated and differentiated into effector memory T cells. In contrast, NK cells activated by rhIL-2 displayed similar ADCC activity, but failed to proliferate. Human cCD16ζ-T cells infused concomitantly with anti-CD20 mAb synergistically inhibited the growth of disseminated Raji cells, a CD20+ lymphoma cell line, in immunodeficient mice, whereas similarly infused rhIL-2–treated NK cells survived for a shorter time and displayed less effective tumor suppression. Our findings strongly suggest the clinical feasibility of cCD16ζ-T cells as adoptively transferable ADCC effector cells that could potentially enhance the clinical responses mediated by currently available anticancer mAbs. Cancer Immunol Res; 2(3); 249–62. ©2014 AACR.
- Published
- 2014
42. Tumor responses and early onset cytokine release syndrome in synovial sarcoma patients treated with a novel affinity-enhanced NY-ESO-1-targeting TCR-redirected T cell transfer
- Author
-
Sachiko Okamoto, Eiichi Sato, Junichi Mineno, Hidefumi Kato, Takeru Funakoshi, Noboru Yamamoto, Shinichi Kageyama, Takashi Kojima, Hiroyoshi Hattori, Mikiya Ishihara, Takashi Watanabe, Hiroaki Ikeda, Tetsuro Sasada, Hiroshi Shiku, Hideyuki Mishima, Hideto Chono, Daisuke Tomura, Yoshihiro Miyahara, Ikuei Nukaya, and Shigehisa Kitano
- Subjects
Cancer Research ,Adoptive cell transfer ,business.industry ,medicine.medical_treatment ,T cell ,Melanoma ,T-cell receptor ,medicine.disease ,Synovial sarcoma ,03 medical and health sciences ,Cytokine release syndrome ,0302 clinical medicine ,Cytokine ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,medicine ,Cancer research ,Sarcoma ,business ,030215 immunology - Abstract
2530 Background: Adoptive transfer of TCR-redirected T cells has been reported to exhibit efficacy in some of melanoma and sarcoma patients. However, there have not been well known about cytokine release syndrome (CRS) or its relations to tumor responses. This study evaluates clinical responses in association with the cell kinetics and CRSs after transfer of high-affinity NY-ESO-1 TCR-gene transduced T cells in NY-ESO-1-expressiong cancer patients (NCT02366546). Methods: We developed a novel-type affinity-enhanced NY-ESO-1-specific TCR and an originally-developed retrovirus vector that encodes siRNA to silence endogenous TCR creation. The NY-ESO-1/TCR sequence is mutated for high affinity with replacements of G50A and A51E in CDR2 region. This is a first-in-man clinical trial of the novel NY-ESO-1-specfic TCR-T cell transfer to evaluate the safety, in vivo cell kinetics and clinical responses. It was designed as a cell-dose escalation from 5 x108 to 5 x109 cells. NY-ESO-1-expressing refractory cancer patients were enrolled, with 3+3 cohort design. Cyclophosphamide (1,500mg/m2) were administered prior to the TCR-T cell transfer as pre-conditioning. Results: 9 patients were treated with the NY-ESO-1/TCR-T cell transfer. The TCR-T cells expanded in peripheral blood with a dose-dependent manner, associated with rapid proliferation within 5 days after the cell transfer. 3 patients receiving 5x109 cells developed early-onset CRSs, with elevations of serum IL-6, IFN-γ. The CRSs developed on day1 or 2 after the cell transfer. They were well managed with tocilizumab treatment. 3 synovial sarcoma patients exhibited tumor shrinkages of partial responses, and they all had high-expression of NY-ESO-1 in the tumor samples, namely, 75% or more. Exploratory analysis revealed that multiple chemotactic cytokines including CCL2 and CCL7, and IL-3 increased in the serum from the patients with CRS. The proportions of effector-memory phenotype T cells in the infused cell-product were significantly associated with CRS development. Conclusions: The affinity-enhanced NY-ESO-1/TCR-T cell transfer exhibited early-onset CRS in association with in vivo cell proliferation and sequential tumor responses in the patients with high-NY-ESO-1-expressing synovial sarcoma. Clinical trial information: NCT02366546.
- Published
- 2019
43. Development of a novel redirected T-cell–based adoptive immunotherapy targeting human telomerase reverse transcriptase for adult T-cell leukemia
- Author
-
Sachiko Okamoto, Kiyotaka Kuzushima, Yukihiro Miyazaki, Junichi Mineno, Hiroshi Shiku, Toshiki Ochi, Taichi Azuma, Hiroaki Asai, Masaki Yasukawa, Hiroshi Fujiwara, Fumihiro Ochi, and Takashi Ishida
- Subjects
Male ,Telomerase ,Adoptive cell transfer ,medicine.medical_treatment ,T cell ,Transplantation, Heterologous ,Immunology ,T-cell leukemia ,Receptors, Antigen, T-Cell ,HLA-A24 Antigen ,CD8-Positive T-Lymphocytes ,Biology ,Biochemistry ,Mice ,medicine ,Animals ,Humans ,Leukemia-Lymphoma, Adult T-Cell ,Cytotoxic T cell ,Telomerase reverse transcriptase ,Cell Line, Transformed ,Cell Biology ,Hematology ,Immunotherapy ,Adoptive Transfer ,Molecular biology ,Neoplasm Proteins ,medicine.anatomical_structure ,Female ,K562 Cells ,Peptides ,Neoplasm Transplantation ,CD8 - Abstract
Although adult T-cell leukemia (ATL) has a poor prognosis, successful allogeneic hematopoietic stem cell transplantation (allo-HSCT) in some cases suggests that a cellular immune-mediated strategy can be effective. So far, however, no effective target for anti-ATL immunotherapy has been defined. Here we demonstrated for the first time that human telomerase reverse transcriptase (hTERT) is a promising therapeutic target for ATL, and we developed a novel redirected T-cell-based immunotherapy targeting hTERT. hTERT messenger RNA was produced abundantly in ATL tumor cells but not in steady-state normal cells. Rearranged human leukocyte antigen-A*24:02 (HLA-A*24:02) -restricted and hTERT461-469 nonameric peptide-specific T-cell receptor (TCR) α/β genes were cloned from our previously established cytotoxic T lymphocyte clone (K3-1) and inserted into a novel retroviral TCR expression vector encoding small interfering RNAs for endogenous TCR genes in redirected T cells (hTERT-siTCR vector). Consequently, allogeneic or autologous gene-modified CD8(+) T cells prepared using the hTERT-siTCR vector successfully killed ATL tumor cells, but not normal cells including steady-state hematopoietic progenitors, in an HLA-A*24:02-restricted manner both in vitro and in vivo. Our experimental observations support the development of a novel hTERT-targeting redirected T-cell-based adoptive immunotherapy for ATL patients, especially those for whom suitable allo-HSCT donors are lacking.
- Published
- 2013
44. Efficient tumor regression by adoptively transferred CEA-specific CAR-T cells associated with symptoms of mild cytokine release syndrome
- Author
-
Takuma Kato, Sachiko Okamoto, Naohiro Seo, Hiroshi Shiku, Linan Wang, Kazutoh Takesako, Ning Ma, Yasunori Amaishi, Junichi Mineno, and Eiichi Sato
- Subjects
0301 basic medicine ,Adoptive cell transfer ,medicine.drug_class ,Immunology ,Adoptive T-cell therapy ,Inflammation ,Monoclonal antibody ,03 medical and health sciences ,0302 clinical medicine ,Carcinoembryonic antigen ,Antigen ,preconditioning ,neurotoxicity ,Immunology and Allergy ,Medicine ,Original Research ,biology ,chimeric antigen receptor ,business.industry ,carcinoembryonic antigen ,cytokine release syndrome ,medicine.disease ,Chimeric antigen receptor ,Cytokine release syndrome ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer cell ,biology.protein ,medicine.symptom ,business - Abstract
Carcinoembryonic antigen (CEA) is a cell surface antigen highly expressed in various cancer cell types and in healthy tissues. It has the potential to be a target for chimeric antigen receptor (CAR)-modified T-cell therapy; however, the safety of this approach in terms of on-target/off-tumor effects needs to be determined. To address this issue in a clinically relevant model, we used a mouse model in which the T cells expressing CEA-specific CAR were transferred into tumor-bearing CEA-transgenic (Tg) mice that physiologically expressed CEA as a self-antigen. The adoptive transfer in conjunction with lymphodepleting and myeloablative preconditioning mediated significant tumor regression but caused weight loss in CEA-Tg, but not in wild-type mice. The weight loss was not associated with overt inflammation in the CEA-expressing gastrointestinal tract but was associated with malnutrition, reflected in elevated systemic levels of cytokines linked to anorexia, which could be controlled by the administration of an anti-IL-6 receptor monoclonal antibody without compromising efficacy. The apparent relationship between lymphodepleting and myeloablative preconditioning, efficacy, and off-tumor toxicity of CAR-T cells would necessitate the development of CEA-specific CAR-T cells with improved signaling domains that require less stringent preconditioning for their efficacy. Taken together, these results suggest that CEA-specific CAR-based adoptive T-cell therapy may be effective for patients with CEA+ solid tumors. Distinguishing the fine line between therapeutic efficacy and off-tumor toxicity would involve further modifications of CAR-T cells and preconditioning regimens.
- Published
- 2016
45. Favorable climatic regime for maintaining the present-day geometry of the Gregoriev Glacier, Inner Tien Shan
- Author
-
A. B. Surazakov, Sachiko Okamoto, Koji Fujita, S. A. Nikitin, Nozomu Takeuchi, Vladimir B. Aizen, and Jumpei Kubota
- Subjects
Glacier ice accumulation ,lcsh:GE1-350 ,geography ,geography.geographical_feature_category ,lcsh:QE1-996.5 ,Accumulation zone ,Glacier ,Present day ,Snow ,Current (stream) ,lcsh:Geology ,Glacier mass balance ,Climatology ,Precipitation ,Geology ,lcsh:Environmental sciences ,Earth-Surface Processes ,Water Science and Technology - Abstract
We conducted 2 yr (2005–2007) of in situ meteorological and glaciological observations on the Gregoriev Glacier, a flat-top glacier within the Inner Tien Shan, Kyrgyzstan. Relative carrier-phase GPS surveys reveal a vertical lowering at the summit of the glacier. Based on snow density data and an energy-mass balance model, we estimate that the annual precipitation and summer mean temperature required to maintain the glacier in the current state are 289 mm and −3.8 °C at the glacier summit (4600 m a.s.l.), respectively. The good agreement between dynamically derived precipitation and the long-term observed precipitation at a nearby station in the Tien Shan (296 mm at 3614 m a.s.l. for the period 1930–2002) suggests that the glacier has been in a near steady-state in terms of mass supply. The glacier mass-balance, reconstructed based on meteorological data from the Tien Shan station for the past 80 yr, explains the observed fluctuations in glacier extent, particularly the negative mass balance in the 1990s.
- Published
- 2011
46. Cytokine Release Syndrome and Tumor Responses in a First-in-Man Trial of a Novel Affinity-Enhanced TCR-Gene Transduced T Cell Transfer Targeting NY-ESO-1 Antigen
- Author
-
Hidefumi Kato, Takashi Kojima, Hiroshi Shiku, Shinichi Kageyama, Naoki Inoue, Mikiya Ishihara, Hideto Chono, Junichi Mineno, Yoshihiro Miyahara, Daisuke Tomura, Ikuei Nukaya, Sachiko Okamoto, Tomohide Kidokoro, Shigehisa Kitano, Hiroaki Ikeda, Takeru Funakoshi, Noboru Yamamoto, Hiroaki Iwase, Takashi Watanabe, Hideyuki Mishima, and Hiroyoshi Hattori
- Subjects
business.industry ,Cell growth ,T cell ,Immunology ,Cell ,T-cell receptor ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Cell therapy ,Cytokine release syndrome ,medicine.anatomical_structure ,Antigen ,In vivo ,Cancer research ,Medicine ,business - Abstract
Adoptive cell transfers of receptor gene-engineered T cells include chimeric antigen receptor-gene transduced T (CAR-T) cell therapy and TCR-gene transduced T (TCR-T) cell therapy. In CD19-CAR-T cell therapy, high incidence of cytokine release syndrome (CRS) is associated with in vivo CAR-T cell proliferation and its clinical efficacy. In human TCR-T cell therapies, there have not been well known about CRS and its association with in vivo T cell kinetics or tumor responses. We have been developing a novel-type affinity-enhanced NY-ESO-1-specific TCR, and an original retrovirus vector that encodes siRNA to silence endogenous TCR creation. The NY-ESO-1 TCR is mutated for high affinity with replacements of G50A and A51E in CDR2 region, which is restricted with HLA-A*02:01 and A*02:06. We extensively examined potential cross-reactivities to different antigen-peptides in preclinical studies, and the high-affinity NY-ESO-1 TCR did not recognize analogous peptides. The new generation retroviral TCR-vector provides enhanced expression of transduced tumor-specific TCRs and an inhibition effect of formations of self-reactive TCRs. This is a first-in-man clinical trial of the novel NY-ESO-1-specfic TCR-T cell transfer to evaluate the safety, in vivo cell kinetics and clinical responses. It is designed as a cell-dose escalation from 5 x108 to 5 x109 cells. NY-ESO-1-expressing refractory cancer patients were enrolled, with 3+3 cohort design. Cyclophosphamide with/without fludarabine were administered prior to the TCR-T cell transfer as pre-conditioning. Six patients were treated with the NY-ESO-1 TCR-T cell transfer, and evaluated for the safety and in vivo cell kinetics. The TCR-T cells appeared in peripheral blood with a dose-dependent manner, associated with in vivo proliferation in an early phase. In three patients given 5x108 cells, no toxicities were seen. Two patients receiving 5x109 cells developed early-phase CRS (G2), with elevations of serum IL-6 and IFN-gamma. They were managed the treatment of anti-IL-6 receptor monoclonal antibody, tocilizumab. In a patient who developed CRS, an event of lung injury (G3) occurred, which was associated with marked infiltration of the NY-ESO-1 TCR-T cells. It was successfully treated with steroid. Two synovial sarcoma patients exhibited tumor responses of PRs. In one patient, progression-free survival lasted more than 8 months. In summary, the affinity-enhanced NY-ESO-1 TCR-T cell transfer exhibited CRSs in association with in vivo cell proliferation and sequential tumor responses. Disclosures No relevant conflicts of interest to declare.
- Published
- 2017
47. Rapid αβ TCR-mediated responses in γδ T cells transduced with cancer-specific TCR genes
- Author
-
Hiroshi Shiku, Yoshimasa Tanaka, Sachiko Okamoto, Hiroyoshi Nishikawa, Seung Shin Yu, Junichi Mineno, Nagahiro Minato, I Kato, Hideto Chono, Atsunori Hiasa, Shigehisa Kitano, and Michiko Hirayama
- Subjects
education.field_of_study ,Adoptive cell transfer ,T-cell receptor ,Population ,hemic and immune systems ,chemical and pharmacologic phenomena ,T lymphocyte ,Biology ,Major histocompatibility complex ,Cell biology ,stomatognathic diseases ,Immune system ,Immunology ,Genetics ,biology.protein ,Molecular Medicine ,Cytotoxic T cell ,education ,Molecular Biology ,CD8 - Abstract
Adoptive T-cell transfer of in vitro cultured T cells derived from cancer patients with naturally developed immune responses has met with some success as an immunotherapeutic approach, although only a limited number of patients showed spontaneous immune responses. To find alternative ways, such as cancer-specific T-cell receptor (TCR) gene transfer, in preparation for sufficient numbers of antigen-specific T cells is an important issue in the field of adoptive T-cell therapy. Given the inherent disadvantage of alphabeta TCR transfer to other alphabeta T cells, namely the possible formation of mixed TCR heterodimers with endogenous alpha or beta TCR, we employed gammadelta T cells as a target for retroviral transfer of cancer-specific TCR and examined whether gammadelta T cells were useful as an alternative population for TCR transfer. Although retroviral transduction to gammadelta T cells with TCR alphabeta genes alone, isolated from a MAGE-A4(143-151)-specific alphabeta CD8(+) cytotoxic T lymphocyte (CTL) clone, did not provide sufficient affinity to recognize major histocompatibility (MHC)-peptide complexes due to the lack of CD8 co-receptor, gammadelta T cells co-transduced with TCR alphabeta and CD8 alphabeta genes acquired cytotoxicity against tumor cells and produced cytokines in both alphabeta- and gammadelta-TCR-dependent manners. Furthermore, alphabeta TCR and CD8-transduced gammadelta T cells, stimulated either through alphabeta TCR or gammadelta TCR, rapidly responded to target cells compared with conventional alphabeta T cells, reminiscent of gammadelta T cells. We propose alphabeta TCR-transduced gammadelta T cells as an alternative strategy for adoptive T-cell transfer.
- Published
- 2009
48. Hepatitis delta antigen binds to the clamp of RNA polymerase II and affects transcriptional fidelity
- Author
-
Hiroshi Handa, Sachiko Okamoto, Takashi Mura, Yuki Yamaguchi, and Sittinan Chanarat
- Subjects
Models, Molecular ,Transcription, Genetic ,viruses ,Molecular Sequence Data ,RNA-dependent RNA polymerase ,RNA polymerase II ,Models, Biological ,Substrate Specificity ,Evolution, Molecular ,chemistry.chemical_compound ,Transcription (biology) ,Genetics ,Humans ,Amino Acid Sequence ,Conserved Sequence ,Hepatitis delta Antigens ,Base Sequence ,biology ,RNA ,RNA virus ,Cell Biology ,biology.organism_classification ,Molecular biology ,Protein Structure, Tertiary ,Elongation factor ,chemistry ,biology.protein ,RNA Polymerase II ,Transcription factor II D ,DNA ,HeLa Cells ,Protein Binding - Abstract
Hepatitis delta virus (HDV) is an RNA virus whose replication and transcription are considered to proceed via RNA-dependent RNA synthesis by RNA polymerase II (Pol II), and the viral protein called hepatitis delta antigen (HDAg) is essential for these processes. HDAg was previously shown to stimulate Pol II elongation on both DNA and RNA templates in vitro. Here, the mechanism of elongation control by HDAg was investigated because it serves as a prototype of cellular transcription elongation factors and also plays an interesting role in HDV proliferation. With site-specific photocrosslinking and transcription using reconstituted elongation complexes, evidence is presented that HDAg functionally interacts with the clamp of Pol II, a mobile structure that holds DNA and RNA in place. Strikingly, HDAg not only increases the rate of elongation but also affects the decision of which nucleotide is incorporated. These and our previous findings lead us to propose a model in which HDAg interacts with and loosens the clamp, and thereby accelerates forward translocation of Pol II at the cost of fidelity. By reducing transcriptional fidelity in terms of not only discrimination of incoming nucleotides but also recognition of templates, HDAg may facilitate the unusual RNA-dependent RNA synthesis by Pol II.
- Published
- 2007
49. Antileukemia multifunctionality of CD4(+) T cells genetically engineered by HLA class I-restricted and WT1-specific T-cell receptor gene transfer
- Author
-
Hiroaki Asai, Yukihiro Miyazaki, Hiroshi Shiku, Kiyotaka Kuzushima, Junichi Mineno, Masaki Yasukawa, Hiroshi Fujiwara, Toshiki Ochi, Fumihiro Ochi, Sachiko Okamoto, and Miwako Narita
- Subjects
CD4-Positive T-Lymphocytes ,congenital, hereditary, and neonatal diseases and abnormalities ,Cancer Research ,Biology ,urologic and male genital diseases ,Lymphocyte Activation ,Immunotherapy, Adoptive ,Interleukin 21 ,Mice ,Cell Movement ,Cytotoxic T cell ,Animals ,Humans ,IL-2 receptor ,Antigen-presenting cell ,WT1 Proteins ,Interleukin 3 ,Leukemia ,urogenital system ,ZAP70 ,Histocompatibility Antigens Class I ,Hematology ,Natural killer T cell ,Virology ,Molecular biology ,female genital diseases and pregnancy complications ,Genes, T-Cell Receptor ,Oncology ,Interleukin 12 ,Female ,Genetic Engineering ,T-Lymphocytes, Cytotoxic - Abstract
To develop gene-modified T-cell-based antileukemia adoptive immunotherapy, concomitant administration of CD4(+) and CD8(+) T cells that have been gene modified using identical HLA class I-restricted leukemia antigen-specific T-cell receptor (TCR) gene transfer has not yet been fully investigated. Here, using CD4(+) and CD8(+) T cells that had been gene modified with a retroviral vector expressing HLA-A*24:02-restricted and Wilms' tumor 1 (WT1)-specific TCR-α/β genes and siRNAs for endogenous TCRs (WT1-siTCR/CD4(+) T cells and WT1-siTCR/CD8(+) T cells), we examined the utility of this strategy. WT1-siTCR/CD4(+) T cells sufficiently recognized leukemia cells in an HLA class I-restricted manner and provided target-specific Th1 help for WT1-siTCR/CD8(+) T cells. By using a xenografted mouse model, we found that WT1-siTCR/CD4(+) T cells migrated to leukemia sites and subsequently attracted WT1-siTCR/CD8(+) T cells via chemotaxis. Therapy-oriented experiments revealed effective enhancement of leukemia suppression mediated by concomitant administration of WT1-siTCR/CD4(+) T cells and WT1-siTCR/CD8(+) T cells. Importantly, this augmented efficacy in the presence of WT1-siTCR/CD4(+) T cells was correlated with longer survival and enhanced formation of memory T cells by WT1-siTCR/CD8(+) T cells. Collectively, our experimental findings strongly suggest that this strategy would be clinically advantageous for the treatment of human leukemia.
- Published
- 2015
50. Direct tumor recognition by a human CD4(+) T-cell subset potently mediates tumor growth inhibition and orchestrates anti-tumor immune responses
- Author
-
Sachiko Okamoto, Sacha Gnjatic, Lloyd J. Old, Kunle Odunsi, Hiroshi Shiku, Protul Shrikant, Junko Matsuzaki, Immanuel F. Luescher, Junichi Mineno, and Takemasa Tsuji
- Subjects
CD4-Positive T-Lymphocytes ,Cytotoxicity, Immunologic ,Cell Survival ,Transplantation, Heterologous ,Antigen presentation ,Mice, SCID ,CD8-Positive T-Lymphocytes ,Biology ,Article ,Interferon-gamma ,Jurkat Cells ,Interleukin 21 ,Antigens, Neoplasm ,Cell Line, Tumor ,Animals ,Humans ,Cytotoxic T cell ,IL-2 receptor ,Antigen-presenting cell ,Cells, Cultured ,Cell Proliferation ,Ovarian Neoplasms ,Multidisciplinary ,Tumor Necrosis Factor-alpha ,Lymphokine ,Membrane Proteins ,T-Lymphocytes, Helper-Inducer ,Acquired immune system ,Coculture Techniques ,Tumor antigen ,Tumor Burden ,3. Good health ,Immunology ,Antigens, Neoplasm/immunology ,Antigens, Neoplasm/metabolism ,CD4-Positive T-Lymphocytes/immunology ,CD4-Positive T-Lymphocytes/metabolism ,CD8-Positive T-Lymphocytes/immunology ,CD8-Positive T-Lymphocytes/metabolism ,Cell Proliferation/drug effects ,Cell Survival/drug effects ,Cell Survival/immunology ,Cytotoxicity, Immunologic/immunology ,Female ,Interferon-gamma/immunology ,Interferon-gamma/metabolism ,Membrane Proteins/immunology ,Membrane Proteins/metabolism ,Ovarian Neoplasms/immunology ,Ovarian Neoplasms/pathology ,T-Lymphocytes, Helper-Inducer/immunology ,T-Lymphocytes, Helper-Inducer/metabolism ,Tumor Burden/immunology ,Tumor Necrosis Factor-alpha/immunology ,Tumor Necrosis Factor-alpha/metabolism ,Cancer research - Abstract
Tumor antigen-specific CD4+ T cells generally orchestrate and regulate immune cells to provide immune surveillance against malignancy. However, activation of antigen-specific CD4+ T cells is restricted at local tumor sites where antigen-presenting cells (APCs) are frequently dysfunctional, which can cause rapid exhaustion of anti-tumor immune responses. Herein, we characterize anti-tumor effects of a unique human CD4+ helper T-cell subset that directly recognizes the cytoplasmic tumor antigen, NY-ESO-1, presented by MHC class II on cancer cells. Upon direct recognition of cancer cells, tumor-recognizing CD4+ T cells (TR-CD4) potently induced IFN-γ-dependent growth arrest in cancer cells. In addition, direct recognition of cancer cells triggers TR-CD4 to provide help to NY-ESO-1-specific CD8+ T cells by enhancing cytotoxic activity and improving viability and proliferation in the absence of APCs. Notably, the TR-CD4 either alone or in collaboration with CD8+ T cells significantly inhibited tumor growth in vivo in a xenograft model. Finally, retroviral gene-engineering with T cell receptor (TCR) derived from TR-CD4 produced large numbers of functional TR-CD4. These observations provide mechanistic insights into the role of TR-CD4 in tumor immunity and suggest that approaches to utilize TR-CD4 will augment anti-tumor immune responses for durable therapeutic efficacy in cancer patients.
- Published
- 2015
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.