351 results on '"Toshio, Imai"'
Search Results
2. Methylthioacetic acid, a derivative of aroma compounds from Cucumis melo var. conomon dose-dependently triggers differentiation and apoptosis of RCM-1 human colorectal cancer cells
- Author
-
Miyu Kamimura, Azusa Sasaki, Yui Otani, Yasushi Nakamura, Takako Nakamura, Kouji Kuramochi, Toshio Imai, Nakao Kubo, and Shigehisa Okamoto
- Subjects
Toxicology - Published
- 2023
3. ER-851, a Novel Selective Inhibitor of AXL, Overcomes Resistance to Antimitotic Drugs
- Author
-
Shuntaro Tsukamoto, Naoko Hata Sugi, Kyoko Nishibata, Youya Nakazawa, Daisuke Ito, Sayo Fukushima, Takayuki Nakagawa, Kenji Ichikawa, Yu Kato, Dai Kakiuchi, Aya Goto, Machiko Itoh-Yagi, Tomoki Aota, Satoshi Inoue, Yoshinobu Yamane, Norio Murai, Hiroshi Azuma, Satoshi Nagao, Ken Sasai, Tsuyoshi Akagi, Toshio Imai, Junji Matsui, and Tomohiro Matsushima
- Subjects
Cancer Research ,Oncology - Abstract
Innate and adaptive resistance to cancer therapies, such as chemotherapies, molecularly targeted therapies, and immune-modulating therapies, is a major issue in clinical practice. Subpopulations of tumor cells expressing the receptor tyrosine kinase AXL become enriched after treatment with antimitotic drugs, causing tumor relapse. Elevated AXL expression is closely associated with drug resistance in clinical samples, suggesting that AXL plays a pivotal role in drug resistance. Although several molecules with AXL inhibitory activity have been developed, none have sufficient activity and selectivity to be clinically effective when administered in combination with a cancer therapy. Here, we report a novel small molecule, ER-851, which is a potent and highly selective AXL inhibitor. To investigate resistance mechanisms and identify driving molecules, we conducted a comprehensive gene expression analysis of chemoresistant tumor cells in mouse xenograft models of genetically engineered human lung cancer and human triple-negative breast cancer. Consistent with the effect of AXL knockdown, cotreatment of ER-851 and antimitotic drugs produced an antitumor effect and prolonged relapse-free survival in the mouse xenograft model of human triple-negative breast cancer. Importantly, when orally administered to BALB/c mice, this compound did not induce retinal toxicity, a known side effect of chronic MER inhibition. Together, these data strongly suggest that AXL is a therapeutic target for overcoming drug resistance and that ER-851 is a promising candidate therapeutic agent for use against AXL-expressing antimitotic-resistant tumors.
- Published
- 2022
4. Feeding a High-Fat Diet for a Limited Duration Increases Cancer Incidence in a Breast Cancer Model
- Author
-
Toshio, Imai, Mie, Naruse, Yukino, Machida, Gen, Fujii, Michihiro, Mutoh, Masako, Ochiai, Mami, Takahashi, and Hitoshi, Nakagama
- Subjects
Cancer Research ,Nutrition and Dietetics ,Oncology ,Medicine (miscellaneous) - Abstract
High-fat intake by young Asian women impacts the risk of breast cancer. Understanding the underlying molecular mechanisms may be essential for disease prevention in Asia as well as globally. We aimed to examine the effects of corn oil- and animal fat-based high-fat diets (32.9 and 31.4%, respectively, of fat energy ratio as compared to 12.3% in the standard diet) on mammary carcinogenesis and alterations in gene expression and epigenetic statuses in the mammary gland during the growth stages in a rat model. An increased incidence of carcinomas was observed after the cessation of high-fat feeding. In addition, rapid tumor growth and elevations in
- Published
- 2022
5. Alteration in molecular properties during establishment and passaging of endometrial carcinoma patient-derived xenografts
- Author
-
Toshio Imai, Hiroshi Yoshida, Yukino Machida, Mizuki Kuramochi, Hitoshi Ichikawa, Takashi Kubo, Mami Takahashi, and Tomoyasu Kato
- Subjects
Multidisciplinary - Abstract
Patient-derived xenograft (PDX) tumor models are known to maintain the genomic and phenotypic profiles, including the histopathological structures, of the parental tumors. On the other hand, unique enrichment of single-nucleotide variants or copy number aberrations has been reported in several types of tumors. However, an understanding of endometrial carcinoma PDXs is limited. The purpose of the present study was to clarify the presence or absence of the molecular properties of endometrial carcinomas in PDXs passaged up to eight times. Established PDXs of endometrioid carcinomas maintained their histopathological characteristics, but those of carcinosarcomas predominantly consisted of sarcomatous components when compared to the parental tumors. Alterations in the proportion of cells with positive/negative immunohistochemical staining for estrogen receptor, PTEN, PAX8, and PAX2 were observed, whereas the proportions of cells with AE1/AE3, TP53, ARID1A, PMS2, and MSH6 staining were unchanged. Variants of cancer-associated genes were compared between PDXs and parental tumors. Mutations in POLE and a frameshift deletion in BRCA1 were observed in the parental tumor tissue in each of the six cases, and additional genomic alterations, which were not apparently related to histopathological and immunohistochemical alterations, were found in the PDXs of these cases. The genomic and phenotypic alterations observed between endometrial carcinoma PDXs and parental tumors were partly associated with endometrial cancer-specific characteristics related to cellular differentiation and gene mutations.
- Published
- 2023
6. Supplementary Figures S1-S7 from ER-851, a Novel Selective Inhibitor of AXL, Overcomes Resistance to Antimitotic Drugs
- Author
-
Tomohiro Matsushima, Junji Matsui, Toshio Imai, Tsuyoshi Akagi, Ken Sasai, Satoshi Nagao, Hiroshi Azuma, Norio Murai, Yoshinobu Yamane, Satoshi Inoue, Tomoki Aota, Machiko Itoh-Yagi, Aya Goto, Dai Kakiuchi, Yu Kato, Kenji Ichikawa, Takayuki Nakagawa, Sayo Fukushima, Daisuke Ito, Youya Nakazawa, Kyoko Nishibata, Naoko Hata Sugi, and Shuntaro Tsukamoto
- Abstract
Supplementary Data Figure S1-S7
- Published
- 2023
7. Supplementary Table S1 from ER-851, a Novel Selective Inhibitor of AXL, Overcomes Resistance to Antimitotic Drugs
- Author
-
Tomohiro Matsushima, Junji Matsui, Toshio Imai, Tsuyoshi Akagi, Ken Sasai, Satoshi Nagao, Hiroshi Azuma, Norio Murai, Yoshinobu Yamane, Satoshi Inoue, Tomoki Aota, Machiko Itoh-Yagi, Aya Goto, Dai Kakiuchi, Yu Kato, Kenji Ichikawa, Takayuki Nakagawa, Sayo Fukushima, Daisuke Ito, Youya Nakazawa, Kyoko Nishibata, Naoko Hata Sugi, and Shuntaro Tsukamoto
- Abstract
Supplementary Table S1
- Published
- 2023
8. Supplementary Table S5 from ER-851, a Novel Selective Inhibitor of AXL, Overcomes Resistance to Antimitotic Drugs
- Author
-
Tomohiro Matsushima, Junji Matsui, Toshio Imai, Tsuyoshi Akagi, Ken Sasai, Satoshi Nagao, Hiroshi Azuma, Norio Murai, Yoshinobu Yamane, Satoshi Inoue, Tomoki Aota, Machiko Itoh-Yagi, Aya Goto, Dai Kakiuchi, Yu Kato, Kenji Ichikawa, Takayuki Nakagawa, Sayo Fukushima, Daisuke Ito, Youya Nakazawa, Kyoko Nishibata, Naoko Hata Sugi, and Shuntaro Tsukamoto
- Abstract
Supplementary Data Table S5
- Published
- 2023
9. Supplementary Table S3 from ER-851, a Novel Selective Inhibitor of AXL, Overcomes Resistance to Antimitotic Drugs
- Author
-
Tomohiro Matsushima, Junji Matsui, Toshio Imai, Tsuyoshi Akagi, Ken Sasai, Satoshi Nagao, Hiroshi Azuma, Norio Murai, Yoshinobu Yamane, Satoshi Inoue, Tomoki Aota, Machiko Itoh-Yagi, Aya Goto, Dai Kakiuchi, Yu Kato, Kenji Ichikawa, Takayuki Nakagawa, Sayo Fukushima, Daisuke Ito, Youya Nakazawa, Kyoko Nishibata, Naoko Hata Sugi, and Shuntaro Tsukamoto
- Abstract
Supplementary Table S3
- Published
- 2023
10. Supplementary Table S4 from ER-851, a Novel Selective Inhibitor of AXL, Overcomes Resistance to Antimitotic Drugs
- Author
-
Tomohiro Matsushima, Junji Matsui, Toshio Imai, Tsuyoshi Akagi, Ken Sasai, Satoshi Nagao, Hiroshi Azuma, Norio Murai, Yoshinobu Yamane, Satoshi Inoue, Tomoki Aota, Machiko Itoh-Yagi, Aya Goto, Dai Kakiuchi, Yu Kato, Kenji Ichikawa, Takayuki Nakagawa, Sayo Fukushima, Daisuke Ito, Youya Nakazawa, Kyoko Nishibata, Naoko Hata Sugi, and Shuntaro Tsukamoto
- Abstract
Supplementary Table S4
- Published
- 2023
11. Supplementary Table S2 from ER-851, a Novel Selective Inhibitor of AXL, Overcomes Resistance to Antimitotic Drugs
- Author
-
Tomohiro Matsushima, Junji Matsui, Toshio Imai, Tsuyoshi Akagi, Ken Sasai, Satoshi Nagao, Hiroshi Azuma, Norio Murai, Yoshinobu Yamane, Satoshi Inoue, Tomoki Aota, Machiko Itoh-Yagi, Aya Goto, Dai Kakiuchi, Yu Kato, Kenji Ichikawa, Takayuki Nakagawa, Sayo Fukushima, Daisuke Ito, Youya Nakazawa, Kyoko Nishibata, Naoko Hata Sugi, and Shuntaro Tsukamoto
- Abstract
Supplementary Table S2
- Published
- 2023
12. Long-term safety and efficacy of E6011, an anti-fractalkine monoclonal antibody, in patients with rheumatoid arthritis inadequately responding to methotrexate
- Author
-
Yoshiya Tanaka, Tsutomu Takeuchi, Hisashi Yamanaka, Toshihiro Nanki, Hisanori Umehara, Nobuyuki Yasuda, Fumitoshi Tago, Yasumi Kitahara, Makoto Kawakubo, Kentaro Torii, Seiichiro Hojo, Tetsu Kawano, and Toshio Imai
- Subjects
Rheumatology - Abstract
Objectives To evaluate the long-term safety and efficacy of E6011, a humanized anti-fractalkine monoclonal antibody, in patients with rheumatoid arthritis (RA) with an inadequate response to methotrexate (MTX). Methods Active RA patients with an inadequate response to MTX were randomly assigned to the E6011 or placebo group and received the study drug subcutaneously every 2 weeks during a 24-week double-blind study period. Subjects who completed evaluations at Week 24 were rolled over into the extension phase and received open-label E6011 (200 or 400 mg) every 2 weeks until Week 102. The safety analysis was conducted up to Week 104, and the efficacy analysis was conducted up to Week 84. Results A total of 169 subjects completed the double-blind treatment phase and were rolled over into the extension phase. In total, 167 (98.8%) subjects experienced any adverse events, and the incidence of treatment-related adverse events was 56.2%. The American College of Rheumatology 20 response rates were observed between 40 and 70% during the extension phase. Conclusions E6011 was safe and well tolerated with no notable safety concerns up to 102 weeks in RA patients with an inadequate response to MTX.
- Published
- 2023
13. Long-term evaluation of E6011, an anti-fractalkine monoclonal antibody, in patients with rheumatoid arthritis inadequately responding to biological disease-modifying antirheumatic drugs
- Author
-
Yoshiya Tanaka, Tsutomu Takeuchi, Hisashi Yamanaka, Toshihiro Nanki, Hisanori Umehara, Nobuyuki Yasuda, Fumitoshi Tago, Yasumi Kitahara, Makoto Kawakubo, Kentaro Torii, Seiichiro Hojo, Tetsu Kawano, and Toshio Imai
- Subjects
Rheumatology - Abstract
Objectives The objective of the study is to evaluate the long-term safety and efficacy of E6011, a humanized anti-fractalkine monoclonal antibody, in patients with rheumatoid arthritis with an inadequate response to biological disease-modifying antirheumatic drugs. Methods In the double-blind treatment phase (24 weeks), placebo or E6011 400 mg was administered until Week 10. Thereafter, E6011 200 mg or 400 mg was administered to Week 22. Subjects who completed the evaluation at Week 24 of the treatment phase were rolled over into the extension phase. The extension phase lasted until Week 104, and all subjects received E6011 400 mg or 200 mg every 2 weeks in an open-label manner until Week 102. Results A total of 47 subjects completed the double-blind treatment phase and were rolled over into the extension phase. In total, 46 (97.9%) subjects experienced any adverse events, and the incidence of treatment-related adverse events was 57.4%. No clear efficacy trend in the American College of Rheumatology 20% response rates was observed. Conclusions E6011 was well tolerated in active rheumatoid arthritis patients who had shown an inadequate response to biologic disease-modifying antirheumatic drugs, but no clear benefit in the American College of Rheumatology 20% response rates was observed. Further studies are needed to clarify the clinical benefit of E6011.
- Published
- 2023
14. Methylthioacetic acid, a derivative of aroma compounds from Cucumis melo var. conomon dose-dependently triggers differentiation and apoptosis of RCM-1 human colorectal cancer cells
- Author
-
Miyu, Kamimura, Azusa, Sasaki, Yui, Otani, Yasushi, Nakamura, Takako, Nakamura, Kouji, Kuramochi, Toshio, Imai, Nakao, Kubo, and Shigehisa, Okamoto
- Abstract
Methylthioacetic acid (MTA) is an acid-hydrolyzed derivative of a natural aroma compound, methylthioacetic acid ethyl ester isolated from Cucumis melo var. conomon (Katsura-uri, Japanese Picking Melon), and induces a villiform-like structure dome in RCM-1 human colorectal cancer cell culture. Thus far, the physiological and molecular properties of MTA-mediated dome formation remain unknown. Herein, MTA (not more than 2 mM) was demonstrated to differentiate the unorganized cell mass into the dome in RCM-1 cell culture by disclosing the correlation between dome formation and several intestinal differentiation markers such as alkaline phosphatase activity and the protein levels of dipeptidyl peptidase 4, villin, and Krüppel-like factor 4. Dome formation in RCM-1 cell culture was additively enhanced by the simultaneous administration of MTA and butyric acid (BA), suggesting that MTA directs the differentiation of RCM-1 cells, potentially through the same or similar pathway(s) shared with BA. Notably, a high dose of MTA (2 mM or more) elevated several apoptosis markers, such as DNA fragmentation, caspase-3/7 activity, and cleavage of poly(ADP-ribose) polymerase. Altogether, in addition to RCM-1 cell differentiation, MTA triggers apoptosis. These results indicate that MTA is a potential anticarcinogenic agent applicable in differentiation therapy and traditional chemotherapy against colorectal cancers.
- Published
- 2023
15. ER-001259851-000, a novel selective inhibitor of AXL, overcomes resistance to antimitotic drugs
- Author
-
Shuntaro, Tsukamoto, Naoko Hata, Sugi, Kyoko, Nishibata, Youya, Nakazawa, Daisuke, Ito, Sayo, Fukushima, Takayuki, Nakagawa, Kenji, Ichikawa, Yu, Kato, Dai, Kakiuchi, Aya, Goto, Machiko, Itoh-Yagi, Tomoki, Aota, Satoshi, Inoue, Yoshinobu, Yamane, Norio, Murai, Hiroshi, Azuma, Satoshi, Nagao, Ken, Sasai, Tsuyoshi, Akagi, Toshio, Imai, Junji, Matsui, and Tomohiro, Matsushima
- Abstract
Innate and adaptive resistance to cancer therapies, such as chemotherapies, molecularly targeted therapies, and immune-modulating therapies, is a major issue in clinical practice. Subpopulations of tumor cells expressing the receptor tyrosine kinase AXL become enriched after treatment with anti-mitotic drugs, causing tumor relapse. Elevated AXL expression is closely associated with drug resistance in clinical samples, suggesting that AXL plays a pivotal role in drug resistance. Although several molecules with AXL inhibitory activity have been developed, none have sufficient activity and selectivity to be clinically effective when administered in combination with a cancer therapy. Here, we report a novel small molecule, ER-001259851-000, which is a potent and highly selective AXL inhibitor. To investigate resistance mechanisms and identify driving molecules, we conducted a comprehensive gene expression analysis of chemo-resistant tumor cells in mouse xenograft models of genetically engineered human lung cancer and human triple-negative breast cancer. Consistent with the effect of AXL knockdown, co-treatment of ER-001259851-000 and antimitotic drugs produced an anti-tumor effect and prolonged relapse-free survival in the mouse xenograft model of human triple-negative breast cancer. Importantly, when orally administered to BALB/c mice, this compound did not induce retinal toxicity, a known side effect of chronic MER inhibition. Together, these data strongly suggest that AXL is a therapeutic target for overcoming drug resistance and that ER-001259851-000 is a promising candidate therapeutic agent for use against AXL-expressing anti-mitotic-resistant tumors.
- Published
- 2022
16. The <scp>Fractalkine‐CX3CR1</scp> Axis Regulates Non‐inflammatory Osteoclastogenesis by Enhancing Precursor Cell Survival
- Author
-
Yoshikazu Kuboi, Yukiko Kuroda, Masayoshi Ohkuro, Sotaro Motoi, Yoshiya Tomimori, Hisataka Yasuda, Nobuyuki Yasuda, Toshio Imai, and Koichi Matsuo
- Subjects
Endocrinology, Diabetes and Metabolism ,Orthopedics and Sports Medicine - Abstract
The chemokine fractalkine (FKN) is produced by various cell types, including osteoblasts and endothelial cells in bone tissue, and signals through a sole receptor, CX3CR1, which is expressed on monocytes/macrophages, including osteoclast precursors (OCPs). However, the direct effects of FKN signaling on osteoclast lineage cells under homeostatic noninflammatory conditions remain unclear. Here, we report that FKN regulates mouse OCP survival and primes OCPs for subsequent osteoclast differentiation. Wild-type but not CX3CR1-deficient OCPs grown on immobilized FKN showed enhanced osteoclast formation following receptor activator of NF-κB ligand (RANKL) stimulation, with increased expression of osteoclast differentiation markers. Interestingly, the growth of OCPs on immobilized FKN increased the expression of
- Published
- 2022
17. Author response for 'The <scp>Fractalkine‐CX3CR1</scp> axis regulates non‐inflammatory osteoclastogenesis by enhancing precursor cell survival'
- Author
-
null Yoshikazu Kuboi, null Yukiko Kuroda, null Masayoshi Ohkuro, null Sotaro Motoi, null Yoshiya Tomimori, null Hisataka Yasuda, null Nobuyuki Yasuda, null Toshio Imai, and null Koichi Matsuo
- Published
- 2022
18. Novel Complement C5 Small-interfering RNA Lipid Nanoparticle Prolongs Graft Survival in a Hypersensitized Rat Kidney Transplant Model
- Author
-
Hidetoshi Ishigooka, Haruki Katsumata, Kan Saiga, Daisuke Tokita, Sotaro Motoi, Chiyuki Matsui, Yuta Suzuki, Ayaka Tomimatsu, Tomoya Nakatani, Yoshikazu Kuboi, Takafumi Yamakawa, Takashi Ikeda, Rumi Ishii, Toshio Imai, Toshio Takagi, and Kazunari Tanabe
- Subjects
Graft Rejection ,Transplantation ,Rats, Inbred Lew ,Graft Survival ,Animals ,Complement C5 ,RNA, Small Interfering ,Kidney Transplantation ,Antibodies ,Immunosuppressive Agents ,Rats - Abstract
Prophylaxis of antibody-mediated rejection (AMR) caused by donor-specific antibodies remains challenging. Given the critical roles of complement activity in antibody-mediated graft injury, we developed a lipid nanoparticle (LNP) formulation of small-interfering RNA against complement C5 (C5 siRNA-LNP) and investigated whether C5 siRNA-LNP could downregulate the complement activity and act as an effective treatment for AMR.Lewis recipient rats were sensitized by skin grafting from Brown Norway donor rats. Kidney transplantation was performed at 4 wk post-skin grafting.C5 siRNA- or control siRNA-LNP was administered intravenously, and the weekly injections were continued until the study's conclusion. Cyclosporin (CsA) and/or deoxyspergualin (DSG) were used as adjunctive immunosuppressants. Complement activity was evaluated using hemolysis assays. The deposition of C5b9 in the grafts was evaluated using immunohistochemical analysis on day 7 posttransplantation.C5 siRNA-LNP completely suppressed C5 expression and complement activity (hemolytic activity ≤ 20%) 7 d postadministration. C5 siRNA-LNP in combination with CsA and DSG (median survival time: 56.0 d) prolonged graft survival compared with control siRNA-LNP in combination with CsA and DSG (median survival time: 21.0 d; P = 0.0012; log-rank test). Immunohistochemical analysis of the grafts revealed that downregulation of C5 expression was associated with a reduction in C5b9-positive area ( P = 0.0141, Steel-Dwass test).C5 siRNA-LNP combined with immunosuppressants CsA and DSG downregulated C5 activity and significantly prolonged graft survival compared with control siRNA-LNP with CsA and DSG. Downregulation of C5 expression using C5 siRNA-LNP may be an effective therapeutic approach for AMR.
- Published
- 2022
19. A phase 2 study of E6011, an anti-Fractalkine monoclonal antibody, in patients with rheumatoid arthritis inadequately responding to biological disease-modifying antirheumatic drugs
- Author
-
Fumitoshi Tago, Hisanori Umehara, Seiichiro Hojo, Kentaro Torii, Hisashi Yamanaka, Makoto Kawakubo, Nobuyuki Yasuda, Toshio Imai, Toshihiro Nanki, Tsutomu Takeuchi, Tetsu Kawano, Yasumi Kitahara, and Yoshiya Tanaka
- Subjects
Adult ,Male ,medicine.drug_class ,Phases of clinical research ,Disease ,Immunologic Tests ,Antibodies, Monoclonal, Humanized ,Monoclonal antibody ,Arthritis, Rheumatoid ,03 medical and health sciences ,0302 clinical medicine ,Double-Blind Method ,Rheumatology ,medicine ,Humans ,In patient ,030212 general & internal medicine ,030203 arthritis & rheumatology ,Chemokine CX3CL1 ,business.industry ,Middle Aged ,medicine.disease ,Treatment Outcome ,Antirheumatic Agents ,Rheumatoid arthritis ,Immunology ,Female ,Antirheumatic drugs ,business - Abstract
To evaluate the safety and efficacy of E6011, a novel humanized anti-fractalkine monoclonal antibody, in patients with active rheumatoid arthritis (RA) with an inadequate response to biological disease-modifying antirheumatic drugs (DMARDs).Active RA patients inadequately responding to biological DMARDs were randomly assigned to placebo or E6011 400-mg group at a 1:1 ratio, and administered E6011 at weeks 0, 1, 2, and subsequently every 2 weeks. Primary endpoint was American College of Rheumatology (ACR)20 response at week 12.Of 64, 33 received placebo, 31 received E6011 400-mg. The ACR20 response rate at week 12 (non-responder imputation) was 27.3% and 22.6% in the placebo and E6011 groups, respectively. ACR50, ACR70 response rates at week 12 were 3.0%, 0% in the placebo and 9.7%, 3.2% in the E6011 group. Exploratory PK exposure analysis revealed that the effect of E6011 tended to be clearer in patients with higher serum trough E6011 concentration. E6011 was well tolerated with no notable safety concerns.E6011 400-mg was well tolerated but had no clear efficacy at week 12 in RA patients with inadequate response to biologics. Further investigations are warranted to determine the optimal clinical dose and evaluation period for E6011.
- Published
- 2021
20. Emerging Role of Fractalkine in the Treatment of Rheumatic Diseases
- Author
-
Nobuyuki Yasuda, Kana Hoshino-Negishi, Toshio Imai, Yoshiya Tanaka, Yoshikazu Kuboi, and Fumitoshi Tago
- Subjects
Chemokine ,biology ,business.industry ,Immunology ,Inflammation ,medicine.disease ,Proinflammatory cytokine ,Chemokine receptor ,Immune system ,Rheumatoid arthritis ,CX3CR1 ,medicine ,biology.protein ,Immunology and Allergy ,medicine.symptom ,CX3CL1 ,business - Abstract
Rheumatoid arthritis (RA) is an autoimmune disorder that affects joints and is characterized by synovial hyperplasia and bone erosion associated with neovascularization and infiltration of proinflammatory cells. The introduction of biological disease-modifying anti-rheumatic drugs has dramatically changed the treatment of RA over the last 20 years. However, fewer than 50% of RA patients enter remission, and 10-15% are treatment refractory. There is currently no cure for RA. Fractalkine (FKN, also known as CX3CL1) is a cell membrane-bound chemokine that can be induced on activated vascular endothelial cells. FKN has dual functions as a cell adhesion molecule and a chemoattractant. FKN binds specifically to the chemokine receptor CX3CR1, which is selectively expressed on subsets of immune cells such as patrolling monocytes and killer lymphocytes. The FKN-CX3CR1 axis is thought to play important roles in the initiation of the inflammatory cascade and can contribute to exacerbation of tissue injury in inflammatory diseases. Accordingly, studies in animal models have shown that inhibition of the FKN-CX3CR1 axis not only improves rheumatic diseases but also reduces associated complications, such as pulmonary fibrosis and cardiovascular disease. Recently, a humanized anti-FKN monoclonal antibody, E6011, showed promising efficacy with a dose-dependent clinical response and favorable safety profile in a Phase 2 clinical trial in patients with RA (NCT02960438). Taken together, the preclinical and clinical results suggest that E6011 may represent a new therapeutic approach for rheumatic diseases by suppressing a major contributor to inflammation and mitigating concomitant cardiovascular and fibrotic diseases. In this review, we describe the role of the FKN-CX3CR1 axis in rheumatic diseases and the therapeutic potential of anti-FKN monoclonal antibodies to fulfill unmet clinical needs.
- Published
- 2020
21. Monoclonal antibodies specific for podocalyxin expressed on human induced pluripotent stem cells
- Author
-
Hiroaki Tateno, Tomoko Watanabe, Kayo Hasehira, Toshio Imai, Jungo Kakuta, Kayo Kiyoi, and Sayoko Saito
- Subjects
0301 basic medicine ,Fucosyltransferase ,medicine.drug_class ,Sialoglycoproteins ,Induced Pluripotent Stem Cells ,Biophysics ,Transfection ,Monoclonal antibody ,Biochemistry ,Fluorescence ,Cell Line ,Flow cytometry ,Cell therapy ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Polysaccharides ,medicine ,Animals ,Humans ,Molecular Biology ,Cells, Cultured ,biology ,medicine.diagnostic_test ,HEK 293 cells ,Mesenchymal stem cell ,Antibodies, Monoclonal ,Cell Biology ,Flow Cytometry ,Fucosyltransferases ,Cell biology ,HEK293 Cells ,030104 developmental biology ,Podocalyxin ,chemistry ,030220 oncology & carcinogenesis ,biology.protein - Abstract
Human induced pluripotent stem cells (hiPSCs) are useful starting materials for the generation of cell therapy products, due to their pluripotency and ability to self-renew. Quality control of hiPSCs is extremely important in creating a stable supply of hPSC-derived products. Previously we identified an hiPSC-specific lectin probe, rBC2LCN, which binds specifically to α1,2-fucosylated glycan and recognizes podocalyxin (PODXL) as a glycoprotein ligand. In this study, we produced monoclonal antibodies (mAbs) specific for α1,2-fucosylated PODXL expressed on hiPSCs. PODXL was recombinantly expressed in fucosyltransferase 1 (FUT1)-transfected HEK293, followed by immunization into mice. Monoclonal antibodies, which bind to PODXL/FUT1-transfected cells, but not to cells transfected with only one of PODXL or FUT1, were screened by flow cytometry. The two mAbs generated (179-6B8C9 and 179-7E12E10), termed α1,2-fucosylated PODXL-specific mAbs (FpMabs), showed binding specificity to PODXL/FUT1-transfected cells. The FpMabs bound to hiPSCs but never to human adipose-derived mesenchymal stem cells, human dermal fibroblasts, or hiPSC-derived mesoderm. Altogether, FpMabs are highly specific probes for hiPSCs, which might be a powerful tool for the characterization of hiPSCs used in regenerative medicine.
- Published
- 2020
22. Rationale for and clinical development of anti-fractalkine antibody in rheumatic diseases
- Author
-
Sei Muraoka, Yoshikazu Kuboi, Toshio Imai, Toshihiro Nanki, and Junko Nishio
- Subjects
0301 basic medicine ,Clinical Biochemistry ,Adhesion (medicine) ,Arthritis, Rheumatoid ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Drug Development ,Rheumatic Diseases ,Drug Discovery ,CX3CR1 ,medicine ,Humans ,Lupus Erythematosus, Systemic ,Pharmacology ,Clinical Trials as Topic ,biology ,Chemokine CX3CL1 ,business.industry ,Immunization, Passive ,Antibodies, Monoclonal ,Chemotaxis ,medicine.disease ,Sjogren's Syndrome ,030104 developmental biology ,030220 oncology & carcinogenesis ,Rheumatoid arthritis ,Immunology ,biology.protein ,Antibody ,business ,Target organ ,Systemic vasculitis - Abstract
Introduction: Rheumatic diseases are inflammatory diseases that damage target organs via multiple subsets of immune cells. Fractalkine (FKN) acts as chemoattractant as well as adhesion molecule. It...
- Published
- 2020
23. A yolk sac tumor of the pancreas and derived xenograft model effectively responded to VIP chemotherapy
- Author
-
Rikako Ishigamori, Mami Takahashi, Junpei Yonemaru, Nobuyoshi Hiraoka, Hitoshi Ichikawa, Toshio Imai, and Satoshi Nara
- Subjects
Liver tumor ,Vindesine ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Adenocarcinoma ,Diagnosis, Differential ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Animals ,Humans ,Ifosfamide ,Yolk sac ,Etoposide ,Aged ,Cisplatin ,Chemotherapy ,Hepatology ,business.industry ,Liver Neoplasms ,Endodermal Sinus Tumor ,Gastroenterology ,medicine.disease ,Xenograft Model Antitumor Assays ,Gemcitabine ,Pancreatic Neoplasms ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cancer research ,Female ,030211 gastroenterology & hepatology ,business ,Pancreas ,medicine.drug - Abstract
Background Yolk sac tumors (YSTs) of the pancreas are extremely rare, and no drug responsiveness data are available regarding YSTs. Methods We report a pancreatic YST in a 70-year-old woman, and its chemotherapeutic responsiveness based on clinical records and evaluation of a patient-derived xenograft (PDX) line of the YST. Results The YST was an 11-cm, solid mass located in the pancreatic tail. Histologically, the tumor showed medullary proliferation of tumor cells, with a variety of growth patterns including microcystic/reticular, endodermal sinus, and hepatoid patterns. Immunohistochemically, the tumor cells were positive for Sall4, glypican-3, and alpha-fetoprotein. We administered VIP (etoposide, ifosfamide, cisplatin) chemotherapy for a recurrent liver tumor, and obtained complete pathological remission. A drug-response assay using the PDX line from this YST revealed that both VIP and gemcitabine effectively inhibit tumor growth. Conclusions These results suggest that differential diagnosis of YST from adenocarcinoma is important for selecting appropriate chemotherapy.
- Published
- 2020
24. The potential of organoids in toxicologic pathology: Histopathological and immunohistochemical evaluation of a mouse normal tissue-derived organoid-based carcinogenesis model
- Author
-
Rikako Ishigamori, Mie Naruse, Akihiro Hirata, Yoshiaki Maru, Yoshitaka Hippo, and Toshio Imai
- Subjects
Toxicology ,Pathology and Forensic Medicine - Abstract
Recently, we introduced an organoid-based chemical carcinogenesis model using mouse normal tissue-derived organoids. In the present review article, the histopathological and immunohistochemical characteristics of mouse normal tissue-derived organoids and tumors derived from these organoids after their
- Published
- 2022
25. Different types of reactions to E7386 among colorectal cancer patient-derived organoids and corresponding CAFs
- Author
-
Toshio Imai, Mie Naruse, Masako Ochiai, Kenji Matsumoto, Satsuki Ikeda, Manami Kani, Yuyu Kato, Akiyoshi Hirayama, Tomoyoshi Soga, Yusaku Hori, Akira Yokoi, and Atsushi Ochiai
- Subjects
Cancer Research ,Oncology - Abstract
Colorectal cancer (CRC) harbors genetic alterations in a component of the Wnt signaling pathway in ~90% of cases. In addition, the Wnt signaling pathway has been previously suggested to serve a notable role in the pathophysiology of CRC cells and cancer-associated fibroblasts (CAFs). In the present study, the possible effects of E7386, a selective inhibitor of the interaction between β-catenin and the cAMP response element-binding protein-binding protein, were evaluated using organoids and the corresponding CAFs derived from patients with CRC. E7386 at 100 nM was revealed to decrease the viability of CRC organoids and CAFs. Analysis of the gene expression profiles revealed marked changes in the expression levels of different types of cancer-associated genes associated with E7386 concentrations in the organoids and/or CAFs, such as those regulating glucose and amino acid metabolism [phosphoenolpyruvate carboxykinase 2, asparagine synthetase (glutamine-hydrolyzing), phosphoserine aminotransferase 1 and phosphoglycerate dehydrogenase], stimulation of natural killer cell-mediated cytotoxicity (UL16-binding protein 1) and modification of the Wnt/β-catenin signaling pathway (indicated by very low density lipoprotein receptor). Results of the hydrophilic metabolome analysis in the organoids were consistent with those of the transcriptomic analysis.
- Published
- 2022
26. The Unique Genetic and Histological Characteristics of DMBA-Induced Mammary Tumors in an Organoid-Based Carcinogenesis Model
- Author
-
Mie Naruse, Rikako Ishigamori, and Toshio Imai
- Subjects
squamous cell carcinoma ,adenocarcinoma ,DMBA ,mammary tissue ,Biology ,QH426-470 ,medicine.disease_cause ,mutations ,Cancer research ,Organoid ,medicine ,Genetics ,Molecular Medicine ,Carcinogenesis ,carcinogenesis ,nude mouse ,Genetics (clinical) ,organoids ,Original Research - Abstract
Here, we report a model system using in vitro 7,12-dimethylbenz[a]anthracene (DMBA; 0.6 μM)-treated mammary tissue-derived organoids generated from heterozygous BALB/c-Trp53 knockout mice to induce tumors after injection into the nude mouse subcutis. In parallel, a single oral dose of DMBA (50 mg/kg bodyweight) to the same murine strain induced mammary adenocarcinomas, characterized by biphasic structures differentiated into luminal and myoepithelial lineages and frequent Hras mutations at codon 61. In the present study, the genetic and histological characteristics of DMBA-induced tumors in the organoid-based model were evaluated to validate its similarities to the in vivo study. The organoid-derived tumors were low-grade adenocarcinomas composed of luminal and basal/myoepithelial cells. When the organoid-derived carcinomas were passaged to other nude mice, they partly progressed to squamous cell carcinomas (SCCs). Whole exome sequencing revealed no mutations at Hras codon 61 in the organoid-derived tumors. However, various mutations were detected in other genes such as Tusc3 and Tgfbr2, which have been reported as cancer-associated or homeostatic squamous cell genes. The most common mutational pattern observed in these genes were the G:C to T:A transversions and G:C to A:T transitions, which are not typical of the mutations caused by DMBA treatment. In conclusion, DMBA exhibited carcinogenicity in the both the ex vivo and in vivo mammary carcinogenesis models, albeit with distinct histological and genetical alterations. Further studies are needed to clarify whether organoid-based carcinogenesis models generated following chemical treatment in vitro could be applied to the clarification of the novel mode of action of chemical carcinogenesis.
- Published
- 2021
27. Integrin α5 mediates cancer cell-fibroblast adhesion and peritoneal dissemination of diffuse-type gastric carcinoma
- Author
-
Yoshiko Nagano, Toshio Imai, Yuko Nagamura, Masato Tanaka, Masakazu Yashiro, Rieko Ohki, Kazuyoshi Yanagihara, Shingo Miyamoto, Ryuichi Sakai, Kazuki Sasaki, Takeshi Kawamura, Makoto Miyazaki, and Hideki Yamaguchi
- Subjects
Cancer Research ,Immunoprecipitation ,medicine.drug_class ,Integrin ,Mice, Nude ,Integrin alpha5 ,Monoclonal antibody ,Transfection ,Mice ,Stomach Neoplasms ,medicine ,Animals ,Humans ,Fibroblast ,biology ,Chemistry ,Cancer ,Fibroblasts ,medicine.disease ,In vitro ,Rats ,Fibronectin ,medicine.anatomical_structure ,Oncology ,Cancer cell ,Cancer research ,biology.protein ,Female - Abstract
Diffuse-type gastric carcinoma (DGC) has a poor prognosis due to its rapid diffusive infiltration and frequent peritoneal dissemination. DGC is associated with massive fibrosis caused by aberrant proliferation of cancer-associated fibroblasts (CAFs). Previously, we reported that direct heterocellular interactions between cancer cells and CAFs is important for the peritoneal dissemination of DGC. In this study, we aimed to identify and target the molecules that mediate such heterocellular interactions. Monoclonal antibodies (mAbs) against intact DGC cells were generated and subjected to high-throughput screening to obtain several mAbs that inhibit the adhesion of DGC cells to CAFs. Immunoprecipitation and mass spectrometry revealed that all mAbs recognized integrin α5 complexed with integrin β1. Blocking integrin α5 in DGC cells or fibronectin, a ligand of integrin α5β1, deposited on CAFs abrogated the heterocellular interaction. Administration of mAbs or knockout of integrin α5 in DGC cells suppressed their invasion led by CAFs in vitro and peritoneal dissemination in a mouse xenograft model. Altogether, these findings demonstrate that integrin α5 mediates the heterotypic cancer cell-fibroblast interaction during peritoneal dissemination of DGC and may thus be a therapeutic target.
- Published
- 2021
28. Single-cell DNA and RNA sequencing reveals the dynamics of intra-tumor heterogeneity in a colorectal cancer model
- Author
-
Hanako Ono, Momoko Nagai, Toshio Imai, Arai Yasuhito, Tatsuhiro Shibata, Hiromi Nakamura, Koji Okamoto, Koshi Mimori, Eisaku Furukawa, Mamoru Kato, Daichi Narushima, Yoshitaka Hippo, Tetsuya Matsuura, and Daisuke Shiokawa
- Subjects
Tumor suppressor gene ,Colorectal cancer ,QH301-705.5 ,Physiology ,Intra-tumor heterogeneity ,Plant Science ,Biology ,medicine.disease_cause ,General Biochemistry, Genetics and Molecular Biology ,Metastasis ,Transcriptome ,Genetic Heterogeneity ,Mice ,Structural Biology ,Cancer genomics ,medicine ,Tumor Microenvironment ,Animals ,Exome ,Biology (General) ,Gene ,Ecology, Evolution, Behavior and Systematics ,Sequence Analysis, RNA ,Cell Biology ,DNA ,medicine.disease ,Transplantation ,Single-cell sequencing ,Tumor progression ,Tumorigenesis ,Cancer cell ,Cancer research ,RNA ,General Agricultural and Biological Sciences ,Carcinogenesis ,Colorectal Neoplasms ,Research Article ,Developmental Biology ,Biotechnology - Abstract
Background Intra-tumor heterogeneity (ITH) encompasses cellular differences in tumors and is related to clinical outcomes such as drug resistance. However, little is known about the dynamics of ITH, owing to the lack of time-series analysis at the single-cell level. Mouse models that recapitulate cancer development are useful for controlled serial time sampling. Results We performed single-cell exome and transcriptome sequencing of 200 cells to investigate how ITH is generated in a mouse colorectal cancer model. In the model, a single normal intestinal cell is grown into organoids that mimic the intestinal crypt structure. Upon RNAi-mediated downregulation of a tumor suppressor gene APC, the transduced organoids were serially transplanted into mice to allow exposure to in vivo microenvironments, which play relevant roles in cancer development. The ITH of the transcriptome increased after the transplantation, while that of the exome decreased. Mutations generated during organoid culture did not greatly change at the bulk-cell level upon the transplantation. The RNA ITH increase was due to the emergence of new transcriptional subpopulations. In contrast to the initial cells expressing mesenchymal-marker genes, new subpopulations repressed these genes after the transplantation. Analyses of colorectal cancer data from The Cancer Genome Atlas revealed a high proportion of metastatic cases in human subjects with expression patterns similar to the new cell subpopulations in mouse. These results suggest that the birth of transcriptional subpopulations may be a key for adaptation to drastic micro-environmental changes when cancer cells have sufficient genetic alterations at later tumor stages. Conclusions This study revealed an evolutionary dynamics of single-cell RNA and DNA heterogeneity in tumor progression, giving insights into the mesenchymal-epithelial transformation of tumor cells at metastasis in colorectal cancer.
- Published
- 2021
29. Epigenetic priming sensitizes gastric cancer cells to irinotecan and cisplatin by restoring multiple pathways
- Author
-
Toshikazu Ushijima, Hiroshi Moro, Kana Kimura, Masahiro Maeda, Masakazu Yashiro, Naoko Hattori, Toshio Imai, and Yoshiaki Nakamura
- Subjects
Cancer Research ,Mice, Nude ,Decitabine ,Irinotecan ,Epigenesis, Genetic ,03 medical and health sciences ,0302 clinical medicine ,Stomach Neoplasms ,Cell Line, Tumor ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Animals ,Humans ,Cytotoxic T cell ,Epigenetics ,neoplasms ,Cisplatin ,business.industry ,Gastroenterology ,PYCARD ,General Medicine ,DNA Methylation ,Xenograft Model Antitumor Assays ,Gene Expression Regulation, Neoplastic ,DNA demethylation ,Oncology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,DNA methylation ,Cancer cell ,Cancer research ,Female ,030211 gastroenterology & hepatology ,business ,medicine.drug - Abstract
Gastric cancer is heavily influenced by aberrant DNA methylation that alters multiple cancer-related pathways, and may respond to DNA demethylating agents, such as 5-aza-2′-deoxycytidine (5-aza-dC). Here, we aimed to analyze whether 5-aza-dC can sensitize gastric cancer cells to clinically used cytotoxic drugs. Ten gastric cancer cell lines were treated with 5-aza-dC for 72 h and their growth was analyzed by conducting WST assay. In vivo effect of the drugs was analyzed using xenografts of OCUM-2 M/SN38 cells. Genome-wide expression and DNA methylation analyses were conducted using microarrays, and biological functions were identified through ingenuity pathway analysis. The cell lines most resistant to SN38 (an active metabolite of irinotecan), CDDP, PTX, and 5-FU, were identified. 5-Aza-dC pre-treatment of the resistant cell lines decreased the IC50 values for SN38 (TMK1, 226.4 nM to 32.91 nM; 44As3, 128.2 nM to 19.32 nM; OCUM2 M/SN38, 74.43 nM to 16.47 nM) and CDDP (TMK1, 5.05 µM to 2.26 µM; OCUM2 M, 10.79 µM to 2.77 µM), but not PTX and 5-FU. The reactivation of apoptosis-related genes, such as RUNX3, PYCARD, TNF, FAS, and FASLG, was induced by pre-treatment with 5-aza-dC, and the DNA demethylation of promoter CpG islands of RUNX3 and PYCARD was confirmed. In a xenograft model with OCUM2 M/SN38, treatment with 5-aza-dC before irinotecan showed markedly enhanced tumor suppression. Epigenetic priming with 5-aza-dC can improve the sensitivity of gastric cancer cells to SN38 and CDDP.
- Published
- 2019
30. Inhibition of the Progression of Skin Inflammation, Fibrosis, and Vascular Injury by Blockade of the <scp>CX</scp> 3 <scp>CL</scp> 1/ <scp>CX</scp> 3 <scp>CR</scp> 1 Pathway in Experimental Mouse Models of Systemic Sclerosis
- Author
-
Tetsu Kawano, Naoto Ishii, Akihito Machinaga, Takashi Matsushita, Takashi Obara, Akira Utsunomiya, Hideaki Ogasawara, Wataru Ikeda, Toshio Imai, Takenao Chino, Minoru Hasegawa, Vu H. Luong, Yoshikazu Kuboi, Noritaka Oyama, and Le Huu Doanh
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Immunology ,Connective tissue ,Inflammation ,Bleomycin ,03 medical and health sciences ,Subcutaneous injection ,chemistry.chemical_compound ,0302 clinical medicine ,Rheumatology ,Fibrosis ,medicine ,Immunology and Allergy ,030203 arthritis & rheumatology ,integumentary system ,business.industry ,Growth factor ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,Cancer research ,medicine.symptom ,business ,Type I collagen ,Transforming growth factor - Abstract
OBJECTIVE To assess the preclinical efficacy and mechanism of action of an anti-CX3 CL1 monoclonal antibody (mAb) in systemic sclerosis (SSc). METHODS Cultured human dermal fibroblasts were used to evaluate the direct effect of anti-CX3 CL1 mAb on fibroblasts. In addition, bleomycin-induced and growth factor-induced models of SSc were used to investigate the effect of anti-CX3 CL1 mAb on leukocyte infiltration, collagen deposition, and vascular damage in the skin. RESULTS Anti-CX3 CL1 mAb treatment significantly inhibited Smad3 phosphorylation (P < 0.05) and expression of type I collagen and fibronectin 1 (P < 0.01) in dermal fibroblasts stimulated with transforming growth factor β1 (TGFβ1). In the bleomycin model, daily subcutaneous bleomycin injection increased serum CX3 CL1 levels (P < 0.05) and augmented lesional CX3 CL1 expression. Simultaneous administration of anti-CX3 CL1 mAb or CX3 CR1 deficiency significantly suppressed the dermal thickness, collagen content, and capillary loss caused by bleomycin (P < 0.05). Injection of bleomycin induced expression of pSmad3 and TGFβ1 in the skin, which was inhibited by anti-CX3 CL1 mAb. Further, the dermal infiltration of CX3 CR1+ cells, macrophages (inflammatory and alternatively activated [M2-like] subsets), and CD3+ cells significantly decreased following anti-CX3 CL1 mAb therapy (P < 0.05), as did the enhanced skin expression of fibrogenic molecules, such as thymic stromal lymphopoietin and secreted phosphoprotein 1 (P < 0.05). However, the treatment did not significantly reduce established skin fibrosis. In the second model, simultaneous anti-mCX3 CL1 mAb therapy significantly diminished the skin fibrosis induced by serial subcutaneous injection of TGFβ and connective tissue growth factor (P < 0.01). CONCLUSION Anti-CX3 CL1 mAb therapy may be a novel approach for treating early skin fibrosis in inflammation-driven fibrotic skin disorders such as SSc.
- Published
- 2019
31. Organoid-based ex vivo reconstitution of Kras-driven pancreatic ductal carcinogenesis
- Author
-
Masashi Izumiya, Mika Hori, Kenji Tatsuno, Yoshiaki Maru, Masako Ochiai, Tetsuya Matsuura, Shingo Kato, Hiroyuki Aburatani, Shogo Yamamoto, Yoshitaka Hippo, Toshio Imai, Atsushi Nakajima, and Daisuke Hoshi
- Subjects
0301 basic medicine ,Cancer Research ,Pancreatic Intraepithelial Neoplasia ,Mice, Nude ,Apoptosis ,Biology ,medicine.disease_cause ,Proto-Oncogene Proteins p21(ras) ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Tumor Cells, Cultured ,medicine ,Organoid ,Animals ,Humans ,Cell Proliferation ,Mice, Inbred BALB C ,Pancreatic Ducts ,General Medicine ,medicine.disease ,Xenograft Model Antitumor Assays ,Mice, Inbred C57BL ,Organoids ,Pancreatic Neoplasms ,Cell Transformation, Neoplastic ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,Adenocarcinoma ,Syngenic ,KRAS ,Tumor Suppressor Protein p53 ,Carcinogenesis ,Pancreas ,Ex vivo ,Carcinoma, Pancreatic Ductal - Abstract
The organoid culture technique has been recently applied to modeling carcinogenesis in several organs. To further explore its potential and gain novel insights into tumorigenesis, we here investigated whether pancreatic ductal adenocarcinoma (PDA) could be generated as subcutaneous tumors in immunocompromised nude mice, by genetic engineering of normal organoids. As expected, acute induction of KrasG12Din vitro occasionally led to development of tiny nodules compatible with early lesions known as pancreatic intraepithelial neoplasia (PanIN). KrasG12D-expressing cells were enriched after inoculation in the subcutis, yet proved rather declined during culture, suggesting that its advantage might depend on surrounding environments. Depletion of growth factors or concurrent Trp53 deletion resulted in its robust enrichment, invariably leading to development of PanIN or large high-grade adenocarcinoma, respectively, consistent with in vivo mouse studies for the same genotype. Progression from PanIN was also recapitulated by subsequent knockdown of common tumor suppressors, whereas the impact of Tgfbr2 deletion was only partially recapitulated, illustrating genotype-dependent requirement of the pancreatic niche for tumorigenesis. Intriguingly, analysis of tumor-derived organoids revealed that KrasG12D-expressing cells with spontaneous deletion of wild-type Kras were positively selected and exhibited an aging-related mutation signature in nude mice, mirroring the pathogenesis of human PDA, and that the sphere-forming potential and orthotopic tumorigenicity in syngenic mice were significantly augmented. These observations highlighted the relevance of the subcutis of nude mice in promoting PDA development despite its ectopic nature. Taken together, pancreatic carcinogenesis could be considerably recapitulated with organoids, which would probably serve as a novel disease model.
- Published
- 2019
32. Role of Anti‐Fractalkine Antibody in Suppression of Joint Destruction by Inhibiting Migration of Osteoclast Precursors to the Synovium in Experimental Arthritis
- Author
-
Miyuki Nishimura, Masayoshi Ohkuro, Tsutomu Kamisako, Akiko Hamaguchi, Kana Hoshino-Negishi, Fumihiro Sugiyama, Minoru Kumai, Jungo Kakuta, Toshio Imai, Nobuyuki Yasuda, Wataru Ikeda, Yoko Ida, Tomoya Nakatani, Naoto Ishii, and Yoshikazu Kuboi
- Subjects
Cartilage, Articular ,musculoskeletal diseases ,0301 basic medicine ,Immunology ,CX3C Chemokine Receptor 1 ,Type II collagen ,Osteoclasts ,Arthritis ,Cartilage Oligomeric Matrix Protein ,Arthritis, Rheumatoid ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Rheumatology ,Cell Movement ,Osteoclast ,Synovitis ,medicine ,Animals ,Immunology and Allergy ,030203 arthritis & rheumatology ,Cartilage oligomeric matrix protein ,biology ,Chemokine CX3CL1 ,Tartrate-Resistant Acid Phosphatase ,Chemistry ,Stem Cells ,Cartilage ,Synovial Membrane ,Antibodies, Monoclonal ,medicine.disease ,Arthritis, Experimental ,030104 developmental biology ,medicine.anatomical_structure ,Mice, Inbred DBA ,Rheumatoid arthritis ,biology.protein ,Cancer research ,Matrix Metalloproteinase 3 ,Antibody - Abstract
Objective To elucidate the role of the fractalkine (FKN)/CX3 CR1 pathway in joint destruction in rheumatoid arthritis. Methods We examined the effect of treatment with anti-mouse FKN (anti-mFKN) monoclonal antibody (mAb) on joint destruction and the migration of osteoclast precursors (OCPs) into the joint, using the collagen-induced arthritis (CIA) model. DBA/1 mice were immunized with bovine type II collagen to induce arthritis, and then treated with anti-mFKN mAb. Disease severity was monitored by arthritis score, and joint destruction was evaluated by soft x-ray and histologic analyses. Plasma levels of joint destruction markers were assessed by enzyme-linked immunosorbent assay. FKN expression on endothelial cells was detected by immunohistochemistry. Bone marrow-derived OCPs were labeled with fluorescein and transferred to mice with CIA, and the migration of the OCPs to the joints was then analyzed. Results Both prophylactic and therapeutic treatment with anti-mFKN mAb significantly decreased the arthritis and soft x-ray scores. Plasma levels of cartilage oligomeric matrix protein and matrix metalloproteinase 3 decreased after treatment with anti-mFKN mAb. Histologic analysis revealed that anti-mFKN mAb inhibited synovitis, pannus formation, and cartilage destruction, as well as suppressed bone damage, with a marked reduction in the number of tartrate-resistant acid phosphatase-positive osteoclasts. Anti-mFKN mAb strongly inhibited the migration of bone marrow-derived OCPs into the affected synovium. Conclusion Anti-mFKN mAb notably ameliorates arthritis and joint destruction in the CIA model, as well as inhibits migration of OCPs into the synovium. These results suggest that inhibition of the FKN/CX3 CR1 pathway could be a novel strategy for treatment of both synovitis and joint destruction in rheumatoid arthritis.
- Published
- 2019
33. GPR31-dependent dendrite protrusion of intestinal CX3CR1+ cells by bacterial metabolites
- Author
-
Hisako Kayama, Junichi Kikuta, Naoki Morita, Junken Aoki, Asuka Inoue, Takeshi Haneda, Kiyoshi Takeda, Masaru Ishii, Ikuo Kimura, Ryu Okumura, Ryusuke Nabeshima, setsuko fujita, Akio Hayashi, Yuichi Maeda, Nobuhiko Okada, Hitomi Mimuro, Toshiyuki Kida, Toshio Imai, and Eiji Umemoto
- Subjects
0301 basic medicine ,Multidisciplinary ,Chemistry ,Dendrite ,Small intestine ,Cell biology ,Lactic acid ,03 medical and health sciences ,GPR31 ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Immune system ,Antigen ,030220 oncology & carcinogenesis ,medicine ,Dendrite extension ,Pyruvic acid - Abstract
Small intestinal mononuclear cells that express CX3CR1 (CX3CR1+ cells) regulate immune responses1-5. CX3CR1+ cells take up luminal antigens by protruding their dendrites into the lumen1-4,6. However, it remains unclear how dendrite protrusion by CX3CR1+ cells is induced in the intestine. Here we show in mice that the bacterial metabolites pyruvic acid and lactic acid induce dendrite protrusion via GPR31 in CX3CR1+ cells. Mice that lack GPR31, which was highly and selectively expressed in intestinal CX3CR1+ cells, showed defective dendrite protrusions of CX3CR1+ cells in the small intestine. A methanol-soluble fraction of the small intestinal contents of specific-pathogen-free mice, but not germ-free mice, induced dendrite extension of intestinal CX3CR1+ cells in vitro. We purified a GPR31-activating fraction, and identified lactic acid. Both lactic acid and pyruvic acid induced dendrite extension of CX3CR1+ cells of wild-type mice, but not of Gpr31b-/- mice. Oral administration of lactate and pyruvate enhanced dendrite protrusion of CX3CR1+ cells in the small intestine of wild-type mice, but not in that of Gpr31b-/- mice. Furthermore, wild-type mice treated with lactate or pyruvate showed an enhanced immune response and high resistance to intestinal Salmonella infection. These findings demonstrate that lactate and pyruvate, which are produced in the intestinal lumen in a bacteria-dependent manner, contribute to enhanced immune responses by inducing GPR31-mediated dendrite protrusion of intestinal CX3CR1+ cells.
- Published
- 2019
34. Blockade of the fractalkine–CX3CR1 axis ameliorates experimental colitis by dislodging venous crawling monocytes
- Author
-
Yoshikazu Kuboi, Miyuki Nishimura, Wataru Ikeda, Tomoya Nakatani, Yukie Seki, Yui Yamaura, Kana Ogawa, Akiko Hamaguchi, Kenzo Muramoto, Keiko Mizuno, Hideaki Ogasawara, Toshihiko Yamauchi, Nobuyuki Yasuda, Hiroshi Onodera, and Toshio Imai
- Subjects
Male ,0301 basic medicine ,Mice, Inbred BALB C ,Chemokine CX3CL1 ,Immunology ,CX3C Chemokine Receptor 1 ,Antibodies, Monoclonal ,General Medicine ,Colitis ,Monocytes ,Mice ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Administration, Rectal ,Animals ,Humans ,Immunology and Allergy ,Female ,Oxazoles ,030215 immunology - Abstract
Chemokine systems modulate inflammatory and immune responses in inflammatory bowel disease (IBD). The colons of IBD patients show increased levels of fractalkine (FKN) and high numbers of FKN receptor-positive (CX3CR1+) cells; however, the FKN–CX3CR1 axis’s role in intestinal inflammation, especially in intravascular leukocyte behaviors, still remains unclear. Here, we show that interruption of the FKN–CX3CR1 axis by anti-FKN monoclonal antibody (mAb) ameliorates murine colitis through regulation of intravascular monocyte behaviors in murine colitis models. FKN expression was detectable in vascular endothelium and CX3CR1+ macrophages accumulated in the mucosal lamina propria and submucosa of the inflamed colons. CD115+ monocytes tethered to the venous endothelium and expressed pro-inflammatory mediators. The anti-FKN mAb improved colitis symptoms, markedly reduced pro-inflammatory factors in the colon, maintained blood vessel integrity and reduced tethered monocytes in the inflamed veins. Intravital imaging revealed that CD115+Gr-1low/− monocytes crawled on the apical surfaces of venous endothelium, and anti-FKN mAb rapidly dislodged the crawling monocytes and inhibited their patrolling behavior. These findings suggest that the FKN–CX3CR1 axis triggers the patrolling behavior of crawling monocytes on the venous endothelium of inflamed colons, and accelerates the subsequent leukocyte activation and infiltration by locally producing inflammatory cytokines and chemokines. The mAb also ameliorated symptoms in another IBD model, T-cell-transferred colitis. Blocking the FKN–CX3CR1 axis with an anti-FKN mAb considerably inhibits the colitis-triggered inflammatory cascades, which may be an alternative strategy to treat IBD.
- Published
- 2019
35. Increased susceptibility to mammary carcinogenesis and an opposite trend in endometrium in Trp53 heterozygous knockout female mice by backcrossing the BALB/c strain onto the background C3H strain
- Author
-
Yukiko Sudo, Naoaki Uchiya, Yukino Machida, and Toshio Imai
- Subjects
Trp53 ,040301 veterinary sciences ,Short Communication ,Toxicology ,medicine.disease_cause ,Endometrium ,Pathology and Forensic Medicine ,BALB/c ,0403 veterinary science ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,medicine ,backcrossing ,mammary carcinoma ,Mutation ,biology ,Strain (biology) ,Mesenchymal stem cell ,04 agricultural and veterinary sciences ,C3H ,biology.organism_classification ,medicine.disease ,uterine carcinoma ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Backcrossing ,Cancer research ,Mammary carcinogenesis - Abstract
Patients with dominantly inherited Li-Fraumeni syndrome have a loss-of-function mutation in TP53 and develop diverse mesenchymal and epithelial neoplasms at multiple sites. Trp53 +/- female mice with the BALB/c background provide unique characteristics for the study of breast cancer in Li-Fraumeni syndrome; however, we previously found that female C3H-Trp53+/ - mice did not spontaneously develop mammary tumors. Therefore, we obtained F1 and N2-N4 female mice by backcrossing the BALB/c strain and examined the incidence of mammary and other tumors in lifetime studies. Malignant lymphomas, osteosarcomas, and uterine adenocarcinomas spontaneously developed in approximately 20% or more of Trp53+/ - mice with the C3H background. In contrast, the incidence of uterine adenocarcinomas showed a tendency to decrease, while that of mammary adenocarcinomas gradually increased in mice with the BALB/c strain backcross. Wild-type BALB/c female mice are predisposed to a wide spectrum of neoplasms, including mammary tumors, partly due to genetic factors, whereas uterine tumors are uncommon not only in BALB/c mice but also C3H mice. Thus, genetic factors appear to contribute to a strain-specific predisposition to malignant neoplasms in Trp53+/- mice, and further studies are needed to clarify the detailed mechanisms.
- Published
- 2019
36. Expression of nischarin, an imidazoline 1 receptor candidate protein, in the ventrolateral medulla of newborn rats
- Author
-
Yukari Nagakura, Ryoji Ide, Toshio Imai, Chikako Saiki, and Nana Sato Hashizume
- Subjects
Male ,Neurons ,medicine.medical_specialty ,Medulla Oblongata ,General Neuroscience ,Solitary nucleus ,Spinal trigeminal nucleus ,Intracellular Signaling Peptides and Proteins ,Imidazoline receptor ,Rostral ventrolateral medulla ,Biology ,Rats ,medicine.anatomical_structure ,Endocrinology ,Vestibular nuclei ,Internal medicine ,medicine ,Animals ,Female ,Imidazoline Receptors ,Brainstem ,Cuneate nucleus ,Rats, Wistar ,Medulla - Abstract
The activation of imidazoline 1 (I1) receptors is suggested to stimulate the respiratory drive in newborn rats. Here, we immunohistochemically examined whether nischarin, an I1 receptor candidate protein, is expressed in the ventrolateral medulla, where cardiorespiratory centers are located. Newborn rats (age, 3–5 days) were deeply anesthetized with isoflurane; the brainstem was dissected, sectioned sagittally, and labeled with nischarin. Nischarin-associated signals were observed broadly throughout the newborn rat brainstem, including at motor nuclei (motor trigeminal nucleus and facial nucleus), sensory nuclei (lateral superior olive, medial and spinal vestibular nuclei, cuneate nucleus, spinal trigeminal nucleus, and solitary nucleus), and the rostral and caudal ventrolateral medullar regions. In particular, the rostral ventrolateral medulla included a layer of aggregated nischarin expression along the ventral surface, and the layer was in close contact with GFAP-positive processes. In addition, some Phox2b-positive neurons were positive for nischarin in the region. Our results reveal nischarin expression in the newborn rat brainstem and suggest that I1 receptor activation at the level of the ventrolateral medulla contributes to central chemoreception and respiratory control in newborn rats.
- Published
- 2021
37. Characterization of the large-scale Japanese patient-derived xenograft (J-PDX) library
- Author
-
Ken Kato, Yasushi Yatabe, Akinobu Hamada, Takeshi Kuwata, Atsushi Ohtsu, Toshio Imai, Mikiko Suzuki, Kazuaki Shimada, Mami Takahashi, Hiroyuki Mano, Shigehiro Yagishita, Toshirou Nishida, and Atsushi Ochiai
- Subjects
0301 basic medicine ,Oncology ,Adult ,Male ,Patient-Specific Modeling ,Cancer Research ,medicine.medical_specialty ,patient‐derived xenograft ,Adolescent ,Colorectal cancer ,Tumor heterogeneity ,03 medical and health sciences ,Mice ,Young Adult ,0302 clinical medicine ,Japan ,Internal medicine ,Neoplasms ,medicine ,Animals ,Humans ,Clinical efficacy ,Stage (cooking) ,Lung cancer ,Child ,Tumor xenograft ,Aged ,Neoplasm Staging ,Aged, 80 and over ,J‐PDX ,business.industry ,Cancer type ,Cancer ,General Medicine ,Original Articles ,Middle Aged ,medicine.disease ,030104 developmental biology ,Drug Discovery and Delivery ,Organ Specificity ,030220 oncology & carcinogenesis ,Child, Preschool ,coclinical trial ,Female ,Original Article ,business ,Neoplasm Transplantation - Abstract
The use of patient‐derived xenografts (PDXs) has recently attracted attention as a drug discovery platform with a high predictive clinical efficacy and a preserved tumor heterogeneity. Given the racial differences in genetic variations, it would be desirable to establish a PDX library from Japanese cancer patients on a large scale. We thus tried to construct the Japanese PDX (J‐PDX) library with a detailed clinical information for further clinical utilization. Between August 2018 and May 2020, a total of 1126 cancer specimens from 1079 patients were obtained at the National Cancer Center Hospital and National Cancer Center Hospital East, Japan, and were immediately transplanted to immunodeficient mice at the National Cancer Center Research Institute. A total of 298 cross‐cancer PDXs were successfully established. The time to engraftment varied greatly by cancer subtypes, especially in the first passage. The engraftment rate was strongly affected by the clinical stage and survival time of the original patients. Approximately 1 year was needed from tumor collection to the time when coclinical trials were conducted to test the clinical utility. The 1‐year survival rates of the patients who were involved in establishing the PDX differed significantly, from 95.6% for colorectal cancer to 56.3% for lung cancer. The J‐PDX library consisting of a wide range of cancer subtypes has been successfully established as a platform for drug discovery and development in Japan. When conducting coclinical trials, it is necessary to consider the target cancer type, stage, and engraftment rate in light of this report., We have successfully established a PDX library derived from Japanese cancer patients. In less than 2 years, we have registered more than 1000 specimens and established nearly 300 PDXs. We will further enrich the library and use it as a platform to accelerate drug discovery in Japan.
- Published
- 2021
38. Downregulation of Immune Response- and External Stimulus-Related Genes in CRC Organoids, and Their Re-Expression by Co-Culturing with CAFs
- Author
-
Takashi Kubo, Masako Ochiai, Atsushi Ochiai, Mie Naruse, Teruhiko Yoshida, Hiromi Sakamoto, Hirokazu Taniguchi, Kenji Matsumoto, Hitoshi Ichikawa, Shigeki Sekine, and Toshio Imai
- Subjects
Text mining ,Immune system ,Downregulation and upregulation ,business.industry ,Organoid ,Biology ,Stimulus (physiology) ,business ,Gene ,Cell biology ,Co culturing - Abstract
Organoids derived from epithelial tumors have recently been utilized as a preclinical model in basic and translational studies. This model is considered to reproduce the features of cell-cell contacted and differentiated original tumor cells, but not the tumor microenvironment. In this study, we established organoids and paired cancer-associated fibroblasts (CAFs) from surgical specimens of colorectal carcinomas (CRCs), and evaluated gene expression profiles in organoids with and without co-culture with CAFs to assess interactions between tumor cells and CAFs in tumor tissues. We found that the expression levels of several genes, which are highly expressed in original CRC tissues, were downregulated in organoids but re-expressed by co-culturing with CAFs. They comprised immune response- and external stimulus-related genes, e.g., REG family and dual oxidases (DUOXs), which are known to have malignant functions, e.g., cell-proliferation and/or reducing apoptosis of epithelia and drug resistance for anti-cancer drugs in tumors. In addition, the degree of re-production of REG1 and DUOX2 in the co-culture system varied depending on CAFs from each CRC case. In conclusion, the co-culture system of CRC organoids with paired CAFs was able to partially reproduce the tumor microenvironment.
- Published
- 2020
39. Emerging Role of Fractalkine in the Treatment of Rheumatic Diseases
- Author
-
Yoshiya, Tanaka, Kana, Hoshino-Negishi, Yoshikazu, Kuboi, Fumitoshi, Tago, Nobuyuki, Yasuda, and Toshio, Imai
- Subjects
CX3CR1 ,fractalkine ,rheumatic diseases ,Review ,humanized anti-fractalkine monoclonal antibody (E6011) ,CD16+ monocyte - Abstract
Rheumatoid arthritis (RA) is an autoimmune disorder that affects joints and is characterized by synovial hyperplasia and bone erosion associated with neovascularization and infiltration of proinflammatory cells. The introduction of biological disease-modifying anti-rheumatic drugs has dramatically changed the treatment of RA over the last 20 years. However, fewer than 50% of RA patients enter remission, and 10–15% are treatment refractory. There is currently no cure for RA. Fractalkine (FKN, also known as CX3CL1) is a cell membrane-bound chemokine that can be induced on activated vascular endothelial cells. FKN has dual functions as a cell adhesion molecule and a chemoattractant. FKN binds specifically to the chemokine receptor CX3CR1, which is selectively expressed on subsets of immune cells such as patrolling monocytes and killer lymphocytes. The FKN–CX3CR1 axis is thought to play important roles in the initiation of the inflammatory cascade and can contribute to exacerbation of tissue injury in inflammatory diseases. Accordingly, studies in animal models have shown that inhibition of the FKN–CX3CR1 axis not only improves rheumatic diseases but also reduces associated complications, such as pulmonary fibrosis and cardiovascular disease. Recently, a humanized anti-FKN monoclonal antibody, E6011, showed promising efficacy with a dose-dependent clinical response and favorable safety profile in a Phase 2 clinical trial in patients with RA (NCT02960438). Taken together, the preclinical and clinical results suggest that E6011 may represent a new therapeutic approach for rheumatic diseases by suppressing a major contributor to inflammation and mitigating concomitant cardiovascular and fibrotic diseases. In this review, we describe the role of the FKN–CX3CR1 axis in rheumatic diseases and the therapeutic potential of anti-FKN monoclonal antibodies to fulfill unmet clinical needs.
- Published
- 2020
40. Efficacy and Safety of E6011, an Anti-Fractalkine Monoclonal Antibody, in Patients With Active Rheumatoid Arthritis With Inadequate Response to Methotrexate: Results of a Randomized, Double-Blind, Placebo-Controlled Phase II Study
- Author
-
Seiichiro Hojo, Hisanori Umehara, Nobuyuki Yasuda, Kentaro Torii, Hisashi Yamanaka, Toshio Imai, Tetsu Kawano, Yasumi Kitahara, Fumitoshi Tago, Makoto Kawakubo, Yoshiya Tanaka, Tsutomu Takeuchi, and Toshihiro Nanki
- Subjects
0301 basic medicine ,Male ,medicine.medical_specialty ,Immunology ,Full Length ,Arthritis ,Phases of clinical research ,Rheumatoid Arthritis ,Placebo ,Antibodies, Monoclonal, Humanized ,Gastroenterology ,law.invention ,Arthritis, Rheumatoid ,03 medical and health sciences ,0302 clinical medicine ,Rheumatology ,Randomized controlled trial ,Double-Blind Method ,law ,Internal medicine ,Statistical significance ,medicine ,Immunology and Allergy ,Humans ,Aged ,030203 arthritis & rheumatology ,business.industry ,Middle Aged ,medicine.disease ,030104 developmental biology ,Methotrexate ,Treatment Outcome ,Rheumatoid arthritis ,Antirheumatic Agents ,Retreatment ,Female ,business ,medicine.drug - Abstract
Objective To evaluate the efficacy and safety of E6011, a humanized IgG2 monoclonal antibody against human fractalkine (FKN), in a phase II, double-blind, placebo-controlled study in rheumatoid arthritis (RA) patients. Methods Patients with moderate-to-severe RA who had an inadequate response to methotrexate were randomly assigned to a placebo group or to E6011 100-mg, 200-mg, or 400/200-mg groups at a 2:1:2:2 ratio. During the 24-week period, patients received the study drug subcutaneously at weeks 0, 1, and 2 and then once every 2 weeks. The primary end point was the American College of Rheumatology 20% improvement criteria (ACR20) response rate at week 12. Results Study drugs were administered to 190 patients (placebo, n = 54; E6011 100 mg, n = 28; E6011 200 mg, n = 54; E6011 400/200 mg, n = 54), and 169 patients completed treatment. A significant difference from placebo was not found in ACR20 response rates at week 12 (37.0% [placebo], 39.3% [100 mg], 48.1% [200 mg], and 46.3% [400/200 mg], using nonresponder imputation). As a secondary end point, ACR20 response rate in the 200-mg and 400/200-mg groups attained statistical significance at week 24 (35.2% [placebo], 39.3% [100 mg], 53.7% [200 mg], and 57.4% [400/200 mg]). Subsequent exploratory subgroup analysis revealed greater efficacy of E6011, particularly in patients with a higher baseline proportion of CD16+ monocytes; ACR20 response rates in this patient subgroup at week 24 were 30.0% (placebo), 46.7% (100 mg), 57.7% (200 mg), and 69.6% (400/200 mg). E6011 administered for 24 weeks was well tolerated. Conclusion This is the first evidence that E6011, a novel cell trafficking inhibitor targeting the FKN-CX3 CR1 interaction, is modestly effective with 24 weeks of treatment in RA patients, although the primary end point was not met.
- Published
- 2020
41. Efficacy and Safety of E6011, an Anti-Fractalkine Monoclonal Antibody, in Active Rheumatoid Arthritis Patients with Inadequate Response to Methotrexate: Results of a Randomized, Double-Blind, Placebo-Controlled Phase 2 Study
- Author
-
Yoshiya Tanaka, Tsutomu Takeuchi, Hisashi Yamanaka, Toshihiro Nanki, Hisanori Umehara, Nobuyuki Yasuda, Fumitoshi Tago, Yasumi Kitahara, Makoto Kawakubo, Kentaro Torii, Seiichiro Hojo, Tetsu Kawano, and Toshio Imai
- Published
- 2020
42. E6130, a Novel CX3C Chemokine Receptor 1 (CX3CR1) Modulator, Attenuates Mucosal Inflammation and Reduces CX3CR1+ Leukocyte Trafficking in Mice with Colitis
- Author
-
Yoshikazu Kuboi, Toshio Imai, Hisashi Wakita, and Tatsuya Yanagawa
- Subjects
0301 basic medicine ,Pharmacology ,Chemokine ,biology ,Chemistry ,medicine.disease ,Inflammatory bowel disease ,Ulcerative colitis ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Immune system ,030220 oncology & carcinogenesis ,Immunology ,CX3CR1 ,medicine ,biology.protein ,Molecular Medicine ,Colitis ,Receptor ,CX3CL1 - Abstract
The chemokine fractalkine (CX3C chemokine ligand 1; CX3CL1) and its receptor CX3CR1 are involved in the pathogenesis of several diseases, including inflammatory bowel diseases such as Crohn's disease and ulcerative colitis, rheumatoid arthritis, hepatitis, myositis, multiple sclerosis, renal ischemia, and atherosclerosis. There are no orally available agents that modulate the fractalkine/CX3CR1 axis. [(3S,4R)-1-[2-Chloro-6-(trifluoromethyl)benzyl]-3-{[1-(cyclohex-1-en-1-ylmethyl)piperidin-4-yl]carbamoyl}-4-methylpyrrolidin-3-yl]acetic acid (2S)-hydroxy(phenyl)acetate (E6130) is an orally available highly selective modulator of CX3CR1 that may be effective for treatment of inflammatory bowel disease. We found that E6130 inhibited the fractalkine-induced chemotaxis of human peripheral blood natural killer cells (IC50 4.9 nM), most likely via E6130-induced down-regulation of CX3CR1 on the cell surface. E6130 had agonistic activity via CX3CR1 with respect to guanosine 5'-3-O-(thio)triphosphate binding in CX3CR1-expressing Chinese hamster ovary K1 (CHO-K1) membrane and had no antagonistic activity. Orally administered E6130 ameliorated several inflammatory bowel disease-related parameters in a murine CD4+CD45RBhigh T-cell-transfer colitis model and a murine oxazolone-induced colitis model. In the CD4+CD45RBhigh T-cell transfer model, E6130 inhibited the migration of CX3CR1+ immune cells and decreased the number of these cells in the gut mucosal membrane. These results suggest that E6130 is a promising therapeutic agent for treatment of inflammatory bowel disease.
- Published
- 2017
43. Safety, pharmacokinetics, and efficacy of E6011, an antifractalkine monoclonal antibody, in a first-in-patient phase 1/2 study on rheumatoid arthritis
- Author
-
Toshihiro Nanki, Hisanori Umehara, Yoshiya Tanaka, Fumitoshi Tago, Makoto Kawakubo, Toshio Imai, Tetsu Kawano, Yasumi Kitahara, Seiichiro Hojo, Tsutomu Takeuchi, and Nobuyuki Yasuda
- Subjects
Adult ,Male ,0301 basic medicine ,medicine.medical_specialty ,Chemokine ,Antibodies, Monoclonal, Humanized ,Gastroenterology ,Arthritis, Rheumatoid ,03 medical and health sciences ,0302 clinical medicine ,Rheumatology ,Internal medicine ,CX3CR1 ,medicine ,Humans ,Adverse effect ,CX3CL1 ,030203 arthritis & rheumatology ,biology ,Chemokine CX3CL1 ,business.industry ,Antibodies, Monoclonal ,Middle Aged ,medicine.disease ,030104 developmental biology ,Antirheumatic Agents ,Rheumatoid arthritis ,Immunology ,biology.protein ,Female ,Methotrexate ,Tumor necrosis factor alpha ,business ,medicine.drug - Abstract
Fractalkine (CX3CL1/FKN) is a chemokine that regulates chemotaxis and adhesion of CX3C chemokine receptor 1 (CX3CR1)-expressing inflammatory cells. We conducted the first phase 1/2, open-label, multiple ascending dose study of E6011, a humanized anti-FKN monoclonal antibody, in Japanese rheumatoid arthritis (RA) patients (clinicaltrial.gov identifier: NCT02196558).Active RA patients with an inadequate response or intolerance to methotrexate or tumor necrosis factor (TNF) inhibitor received E6011 at week 0, 1, 2, and thereafter every 2 weeks for 12 weeks.Twelve, 15, and 10 subjects were enrolled in the 100, 200, and 400 mg cohorts, respectively. No severe adverse events (AEs) or deaths occurred, and no major differences were observed in the incidence or severity of AEs across the cohorts. Serum E6011 concentrations increased dose dependently. American College of Rheumatology (ACR) 20, 50, and 70 responses at week 12 were 75.0%, 33.3%, and 8.3% in the 100 mg cohort; 66.7%, 20.0%, and 13.3% in the 200 mg cohort; and 60.0%, 30.0%, and 20.0% in the 400 mg cohort, respectively.E6011 appeared to be safe and well tolerated in RA patients during this 12-week treatment period, suggesting that E6011 has an effective clinical response in active RA patients.
- Published
- 2017
44. Horseradish extract promotes urinary bladder carcinogenesis when administered to F344 rats in drinking water
- Author
-
Akiyoshi Nishikawa, Young-Man Cho, Mai Hasumura, Kumiko Ogawa, Shigeaki Takami, and Toshio Imai
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Urology ,Toxicology ,medicine.disease_cause ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Internal medicine ,medicine ,Urothelium ,Urinary bladder ,business.industry ,Cell growth ,Hyperplasia ,medicine.disease ,Allyl isothiocyanate ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,chemistry ,Nitrosamine ,030220 oncology & carcinogenesis ,Isothiocyanate ,business ,Carcinogenesis - Abstract
Horseradish extract (HRE), consisting mainly of a mixture of allyl isothiocyanate and other isothiocyanates, has been used as a food additive. To evaluate the potential hazards of HRE, a 104-week chronic study, a 2-week analysis of cell proliferation in the urinary bladder and a medium-term promotion bioassay of HRE were conducted with administration at concentrations of up to 0.04% HRE in the drinking water to male F344 rats. In the 104-week chronic study with 32 male rats per group, no treatment-related increases in the incidences of neoplastic lesions in any organ, including urinary bladder, were observed, except for simple hyperplasia in the urinary bladder in rats treated with HRE at concentrations of more than 0.01% (5.0 mg kg-1 body weight day-1 ). In the promotion study, HRE treatment after N-butyl-N-(4-hydroxybutyl)nitrosamine initiation caused a clear increase in papillary or nodular hyperplasia, papilloma, and urothelial carcinoma of the urinary bladder in the groups given HRE for 13 weeks at doses higher than 0.005%, 0.01%, and 0.04% (2.7, 5.4 and 20.5 mg kg-1 body weight day-1 ), respectively. In the 2-week cell proliferation analysis, treatment with HRE at concentrations greater than 0.005% (3.9 mg kg-1 body weight day-1 ) caused transient increases in 5-bromo-2'-deoxyuridine labeling indices in the urothelium. Although clear tumor induction was not observed, administration of relatively low-dose HRE increased cell proliferation in the urothelium and exerted obvious promoting effects on rat urinary bladder carcinogenesis. Further studies are needed to elucidate the mode of action of HRE in the rat urinary bladder to facilitate data extrapolation from the present study and provide insights into risk assessment. Copyright © 2017 John Wiley & Sons, Ltd.
- Published
- 2017
45. An organoid-based carcinogenesis model induced by in vitro chemical treatment
- Author
-
Yoshiaki Maru, Ryoichi Masui, Toshio Imai, Yoshitaka Hippo, Masako Ochiai, and Mie Naruse
- Subjects
0301 basic medicine ,Cancer Research ,Carcinogenesis ,9,10-Dimethyl-1,2-benzanthracene ,DMBA ,Mice, Nude ,medicine.disease_cause ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Nude mouse ,Organoid ,medicine ,Animals ,Diethylnitrosamine ,Mode of action ,Lung ,Carcinogen ,Mice, Knockout ,Acrylamide ,biology ,Chemistry ,General Medicine ,Neoplasms, Experimental ,biology.organism_classification ,In vitro ,Organoids ,030104 developmental biology ,Liver ,030220 oncology & carcinogenesis ,Ethyl Methanesulfonate ,Cancer research ,Carcinogens ,Tumor Suppressor Protein p53 ,Ex vivo - Abstract
Animal carcinogenesis models induced by environmental chemicals have been widely used for basic and applied cancer research. However, establishment of in vitro or ex vivo models is essential for molecular mechanistic elucidation of early events in carcinogenesis, leading to clarification of the total mode of action. In the present study, to establish an organoid-based chemical carcinogenesis model, mouse organoids were treated in vitro with 4 genotoxic chemicals, e.g. ethyl methanesulfonate (EMS), acrylamide (AA), diethylnitrosamine (DEN) and 7,12-dimethylbenz[a]anthracene (DMBA) to examine their tumorigenicity after injection to nude mice. The four chemicals were reported to induce lung, liver or mammary carcinomas in mouse models. DMBA-treated mammary tissue-derived organoids with Trp53 heterozygous knockout exhibited tumorigenicity, but not those with wild-type Trp53, reflecting previous reports of corresponding animal models. Treatment of lung organoids with or without Trp53 knockout with EMS or AA resulted in carcinogenic histopathological characteristics, and the activation of oncogenic kinases was demonstrated in the nodules from the nude mouse subcutis. DEN-treated liver (biliary tract) organoids also had an increased number of similar changes. In conclusion, an ex vivo model for chemical carcinogenesis was established using normal mouse tissue-derived organoids. This model will be applied to detect early molecular events, leading to clarification of the mode of action of chemical carcinogenesis.
- Published
- 2019
46. THU0056 EFFECTS OF CX3CL1 INHIBITION ON MURINE BLEOMYCIN-INDUCED INTERSTITIAL PNEUMONIA
- Author
-
Soichi Yamada, Hiroshi Sato, Shinichi Kawai, Natsuko Kusunoki, Toshio Imai, Naoto Ishii, Tetsuo Mikami, Kana Hoshino-Negishi, Hiroyasu Nakano, Yoshikazu Kuboi, Shion Miyoshi, and Toshihiro Nanki
- Subjects
Chemokine ,biology ,business.industry ,medicine.drug_class ,Arthritis ,Pharmacology ,medicine.disease ,Monoclonal antibody ,Bleomycin ,chemistry.chemical_compound ,chemistry ,Rheumatoid arthritis ,CX3CR1 ,biology.protein ,Medicine ,business ,CX3CL1 ,Receptor - Abstract
Background Pathological findings of interstitial pneumonia (IP) reveal the accumulation of inflammatory cells and proliferation of fibroblasts in lung tissue. Although a treatment has not yet been established for IP, particularly for IP with collagen diseases, chemokines may play a role in the pathogenesis of IP for inflammatory cell infiltration. We previously reported that chemokine (C-X3-C motif) ligand 1 (CX3CL1, also known as fractalkine) has potential as a therapeutic target for rheumatoid arthritis (RA).1,2,3Humanized anti-human CX3CL1 monoclonal antibody (mAb) is currently undergoing clinical trials for RA.4 Objectives In the present study, we examined the therapeutic effects of CX3CL1 blockade in a murine model of IP. Methods Bleomycin (BLM)-induced IP was developed by the intratracheal administration of BLM to C57BL/6 mice. The murine lung was stained with hematoxylin and eosin, and the expression of CX3CL1 and CX3CR1, a receptor for CX3CL1, was analyzed by immunohistochemistry. Mice were treated with anti-CX3CL1 mAb for 2 weeks. Collagen eluted from the lung was quantified using the SircolTM Collagen Assay. The expression of CX3CL1 and CX3CR1 by mouse lung fibroblasts (MLFs) was examined with quantitative RT-PCR and Western blotting, respectively. Cell movement was investigated using the scrape motility assay. Results The expression of CX3CL1 and CX3CR1 was upregulated in BLM-induced IP. The treatment with anti-CX3CL1 mAb did not significantly alter inflammatory cell infiltration. However, collagen in the lung was decreased by the treatment with anti-CX3CL1 mAb. Stimulation with CX3CL1 did not alter the in vitro production of collagen by MLFs, but significantly enhanced cell movement. Conclusion CX3CL1 may be involved in increasing collagen in IP and the cell movement of MLFs. The present results suggest that CX3CL1 plays an important role in fibrosis in IP. References [1] Nanki T, et al. Inhibition of fractalkine ameliorates murine collagen-induced arthritis. J Immunol. 2004;173(11):7010-6. [2] Nanki T, et al. Migration of CX3CR1-positive T cells producing type 1 cytokines and cytotoxic molecules into the synovium of patients with rheumatoid arthritis. Arthritis Rheum. 2002;46(11):2878-83. [3] Nanki T, et al. Fractalkine/CX3CL1 in rheumatoid arthritis. Mod Rheumatol. 2017 May;27(3):392-397. [4] Tanaka Y, et al. Safety, pharmacokinetics, and efficacy of E6011, an antifractalkine monoclonal antibody, in a first-in-patient phase 1/2 study on rheumatoid arthritis. Modern rheumatology. 2018;28(1):58-65. Disclosure of Interests Soichi Yamada: None declared, Shion Miyoshi: None declared, Natsuko Kusunoki: None declared, Hiroshi Sato: None declared, Yoshikazu Kuboi Shareholder of: Eisai Co., Ltd., Employee of: KAN Research Institute, Inc. (subsidiary of Eisai Co., Ltd.), Kana Hoshino-Negishi Shareholder of: Eisai Co., Ltd., Employee of: KAN Research Institute, Inc. (subsidiary of Eisai Co., Ltd.), NAOTO ISHII Shareholder of: Eisai Co., Ltd., Employee of: KAN Research Institute, Inc. (subsidiary of Eisai Co., Ltd.), Toshio Imai Shareholder of: Eisai Co., Ltd., Employee of: KAN Research Institute, Inc. (subsidiary of Eisai Co., Ltd.), Tetsuo Mikami Speakers bureau: EA Pharma, Hiroyasu Nakano: None declared, Shinichi Kawai Grant/research support from: Nippon Zoki, Chugai, Ono, Ayumi, Hisamitu, Eisai, Japan Tobacco, Nippon Kayaku, Daiichi Sankyo, Astellas, Yutoku, and Mitsubishi Tanabe, Consultant for: Santen, Japan Tobacco, Speakers bureau: Ayumi, Chugai, Ono, Astellas, Pfizer, Celltrion, Toshihiro Nanki Grant/research support from: Chugai, Eisai, Takeda, Teijin, Eli Lilly, Bristol-Myers, AbbVie, Ono, Novartis, Asahikasei, Mitsubishi-Tanabe, Astellas, Ayumi, Pfizer, Daiichi Sankyo, Shionogi, Sanofi, Nippon Kayaku, Yutoku, Actelion, UCB, Bayer, Nihon Pharmaceutical., Consultant for: UCB, Eisai, Chugai, Ono, Gilead., Speakers bureau: Mitsubishi-Tanabe, Chugai, Eisai, Takeda, Astellas, Janssen, Eli Lilly, Ayumi, Pfizer, Asahikasei, Sanofi, Daiichi Sankyo, Otsuka, AbbVie, Ono, Teijin, Nippon Kayaku, UCB.
- Published
- 2019
47. SAT0048 ANTI-FRACTALKINE MONOCLONAL ANTIBODY AMELIORATE JOINT DESTRUCTION AND SYNOVIUM THROUGH SUPPRESSION OF OSTEOCLAST PRECURSOR MIGRATION AND INDUCTION OF SYNOVIAL CELL DEATH IN COLLAGEN-INDUCED ARTHRITIS MODEL
- Author
-
Nobuyuki Yasuda, Wataru Ikeda, Naoto Ishii, Tomoya Nakatani, Kana Hoshino-Negishi, Masayoshi Ohkuro, Yoshikazu Kuboi, and Toshio Imai
- Subjects
musculoskeletal diseases ,biology ,business.industry ,Type II collagen ,Arthritis ,medicine.disease ,medicine.anatomical_structure ,Synovial Cell ,Osteoclast ,RANKL ,Synovitis ,CX3CR1 ,biology.protein ,Cancer research ,Medicine ,Tumor necrosis factor alpha ,business - Abstract
Background In the Phase 1/2 clinical study, E6011, a novel humanized anti-fractalkine (FKN) mAb demonstrated a promising efficacy in active RA patients who were inadequately controlled by MTX and/or TNF-α inhibitors (NCT 02196558)1.In RA joint tissue, increased expression of fractalkine (FKN) and abundant infiltration of CX3CR1-positive cells were observed2. FKN is expressed on endothelial cells and fibroblast-like synoviocytes in synovium and also expressed on osteoblasts. CX3CR1 is expressed on monocytes/macrophages and osteoclast precursors (OCPs). Therefore, FKN-CX3CR1 interaction could play pivotal roles in migration, differentiation and activation of those cells. However, the precise mechanism(s) of FKN-CX3CR1 axis in RA, especially on joint destruction remains to be elucidated. Objectives To elucidate the roles of FKN-CX3CR1 axis in cartilage destruction, bone damage and proliferated synovial cells by using anti-mouse FKN mAb (anti-mFKN mAb). Methods For the induction of CIA, mice were immunized with bovine type II collagen. Anti-mFKN mAb was injected twice a week. The clinical arthritis score was monitored, and joint destruction was evaluated by soft x-ray and histology. The mRNA expression levels were assessed by quantitative RT-PCR. Blood parameters were measured using ELISA. In in vitro, effect of immobilized FKN on RANK ligand (RANKL)-induced osteoclast differentiation was examined. In in vivo, bone marrow-derived OCPs were fluorescein-labeled and transferred to CIA mice to evaluate the migration of OCPs into synovium. Inhibitory effect of anti-mFKN mAb, etanercept or tofacitinib against OCP migration was assessed. To examine the effect of anti-mFKN mAb against proliferated synovial cells, propidium iodide (PI) was injected to anti-mFKN mAb-treated CIA mice to detect the synovial cell death. Results Anti-mFKN mAb significantly reduced the arthritis and soft x-ray scores in both prophylactic and therapeutic treatment. Anti-mFKN mAb histologically improved synovitis, cartilage destruction and bone damage, with marked reduction of osteoclast numbers. Plasma levels of COMP and MMP-3 were also decreased. Interestingly, anti-mFKN mAb significantly suppressed Tnf and Il6 mRNA expression in the affected joints. In in vitro, RANKL-induced osteoclast differentiation was enhanced by immobilized FKN, and anti-mFKN mAb suppressed FKN-dependent osteoclast formation. In in vivo, anti-mFKN mAb strongly inhibited the migration of OCPs into the proliferated synovial cells of mice with CIA, whereas etanercept or tofacitinib had no significantly effect. Importantly, synovial cell death was abundantly found in proliferated synovial cells of mice with CIA after the anti-mFKN mAb treatment3. Conclusion Anti-mFKN mAb remarkably ameliorated the joint destruction with the marked reduction of osteoclasts by the inhibition of both OCP survival and OCP migration in inflamed joint tissues. In addition, anti-mFKN mAb immediately induced synovial cell death in the proliferated synovial cells, suggesting the direct inhibitory effect in the synovitis. These results strongly indicate that inhibition of FKN-CX3CR1 axis by a humanized anti-FKN mAb, E6011, is an attractive and affected joints-selective therapeutic strategy for the treatment of both inflammatory synovitis, cartilage destruction and bone damage in RA patients. References [1] Tanaka Y, et al., Mod Rheumatol (2018) 28, 58-65 [2] Nanki T, et al., Arthritis Rheum (2002) 46, 2878-83 [3] Hoshino K, et al., Arthritis Rheumatol (2018) Aug 6 [Epub ahead of print] Disclosure of Interests NAOTO ISHII Shareholder of: Eisai Co., Ltd., Employee of: KAN Research Institute, Inc. (subsidiary of Eisai Co., Ltd.), Kana Hoshino-Negishi Shareholder of: Eisai Co., Ltd., Employee of: KAN Research Institute, Inc. (subsidiary of Eisai Co., Ltd.), Masayoshi Ohkuro Employee of: EA Pharma Co., Ltd., Tomoya Nakatani Employee of: KAN Research Institute, Inc. (subsidiary of Eisai Co., Ltd.), Wataru Ikeda Shareholder of: Eisai Co., Ltd., Employee of: KAN Research Institute, Inc. (subsidiary of Eisai Co., Ltd.), Yoshikazu Kuboi Shareholder of: Eisai Co., Ltd., Employee of: KAN Research Institute, Inc. (subsidiary of Eisai Co., Ltd.), Nobuyuki Yasuda Shareholder of: Eisai Co., Ltd., Employee of: KAN Research Institute, Inc. (subsidiary of Eisai Co., Ltd.), Toshio Imai Shareholder of: Eisai Co., Ltd., Employee of: KAN Research Institute, Inc. (subsidiary of Eisai Co., Ltd.)
- Published
- 2019
48. OP0223 EFFICACY AND SAFETY OF E6011, AN ANTI-FRACTALKINE MONOCLONAL ANTIBODY, IN MTX-IR PATIENTS WITH RHEUMATOID ARTHRITIS
- Author
-
Toshihiro Nanki, Hisanori Umehara, Tetsu Kawano, Seiichiro Hojo, Makoto Kawakubo, Tsutomu Takeuchi, Yoshiya Tanaka, Fumitoshi Tago, Yasumi Kitahara, Nobuyuki Yasuda, Kentaro Torii, Hisashi Yamanaka, and Toshio Imai
- Subjects
030203 arthritis & rheumatology ,0301 basic medicine ,medicine.medical_specialty ,Study drug ,business.industry ,Significant difference ,Placebo group ,Acr20 response ,03 medical and health sciences ,Safety profile ,030104 developmental biology ,0302 clinical medicine ,Internal medicine ,Comparison study ,medicine ,In patient ,business ,Bristol-Myers - Abstract
Background Fractalkine (CX3CL1, designated as FKN hereafter) is the sole member of the CX3C-chemokine which leads to dual actions, chemotaxis and cell adhesion for leukocytes expressing the cognate receptor, CX3CR1, during their migration. We have conducted clinical trials of E6011, a novel humanized anti-FKN monoclonal antibody, for patients with rheumatoid arthritis (RA) in Japan1. This is the first report of efficacy and safety results of E6011 from a Phase 2, multicenter, randomized, double-blind, placebo-controlled, parallel-group comparison study in RA patients inadequately responding to methotrexate (NCT02960438). Objectives To evaluate efficacy and safety of three dose of E6011 compared with placebo. Methods During the 24-week double-blind period, patients with moderately to severely active RA of inadequate response to MTX were randomly assigned to E6011 100 mg, 200 mg, 400/200 mg, or placebo groups at a 1:2:2:2 ratio. In the E6011 100 mg, 200 mg, and placebo groups, subjects received 100 mg, 200 mg, or placebo at Weeks 0, 1, 2, and every 2 weeks subsequently. In the E6011 400/200 mg group, subjects received 400 mg at Weeks 0, 1, 2, 4, 6, 8, 10 and then 200 mg every 2 weeks subsequently. Results A total of 190 subjects (54 in the placebo group, 28 in the 100 mg group, 54 in the 200 mg group, 54 in the 400/200 mg group) received study drug. Of the 190 subjects, 169 completed and 21 discontinued study treatment prematurely during the 24-week double-blind period. The ACR20 response rate at Week 12 (non-responder imputation), the primary endpoint, was 37.0% in the placebo group, 39.3% in the 100 mg group, 48.1% in the 200 mg group, and 46.3% in the 400/200 mg group (not statistically significant), and statistically significant difference from placebo in ACR20 response rate was found in the 200 mg and 400/200 mg groups at Week 24 (35.2% for placebo, 39.3% for 100 mg, 53.7% for 200 mg, 57.4% for 400/200 mg). In addition, we focused on CD16+ monocytes which highly expressing FKN receptor/CX3CR1 as a blood biomarker that linked to the clinical response to E6011. The whole patient population was divided into 2 groups according to the median value of baseline CD16+ monocyte percentage (Median: 10.35%). Much clearer ACR20 response was observed in a dose dependent manner in the subjects who showed higher baseline CD16+ monocytes over the median at Week 24 (NRI) (30.0% for placebo, 46.7% for 100 mg, 57.7% for 200 mg, and 69.6% for 400/200 mg) although such fashion was obscure in the subjects below the median value. Adverse events that occurred in ≥5% of subjects in any E6011 group were nasopharyngitis, upper respiratory tract infection, stomatitis, bronchitis, back pain, pharyngitis, and dental caries. As a results, E6011 was well tolerated with no notable safety concerns at doses of 100, 200, and 400/200 mg when administered subcutaneously for 24 weeks. Conclusion E6011 provided clinical improvements with a good safety profile in RA patients with inadequately responding to MTX. Especially, a higher efficacy of E6011 was suggested in patients with higher baseline CD16+ monocytes (%). This is the world-first evidence suggesting that a novel approach to target FKN/CX3CR1 interaction could be beneficial for RA. References [1] Tanaka Y, et al., Mod Rheumatol (2018) 28, 58-65 Acknowledgement The authors wish to thank the study investigators. Disclosure of Interests Yoshiya Tanaka Grant/research support from: Abbvie, Astellas, Bristol-Myers Squibb, Chugai, Daiichi-Sankyo, Eisai, Mitsubishi-Tanabe, MSD, Ono, Taisho-Toyama, Takeda, Speakers bureau: Abbvie, Asahi-kasei, Astellas, Bristol-Myers Squibb, Chugai, Daiichi-Sankyo, Eli Lilly, Eisai, Glaxo-Smithkline, Janssen, Mitsubishi-Tanabe, Novartis, Pfizer Japan Inc, Sanofi, Takeda, UCB, YL Biologics, Tsutomu Takeuchi Grant/research support from: Astellas Pharma Inc, Chugai Pharmaceutical Co, Ltd., Daiichi Sankyo Co., Ltd., Takeda Pharmaceutical Co., Ltd., AbbVie GK, Asahikasei Pharma Corp., Mitsubishi Tanabe Pharma Co., Pfizer Japan Inc., Eisai Co., Ltd., AYUMI Pharmaceutical Corporation, Nipponkayaku Co. Ltd., Novartis Pharma K.K., Grant/research support from: AbbVie, Asahi Kasei, Astellas, AstraZeneca, AYUMI, Bristol-Myers Squibb, Chugai, Daiichi Sankyo, Eisai, Eli Lilly Japan, Janssen, Mitsubishi Tanabe, Nippon Kayaku, Novartis, Pfizer Japan Inc, Taiho, Taisho Toyama, Takeda, Teijin, Grant/research support from: Astellas Pharma Inc., Bristol Myers Squibb, Chugai Pharmaceutical Co., Ltd., Mitsubishi Tanabe Pharma Co., Pfizer Japan Inc., Santen Pharmaceutical Co., Ltd., Takeda Pharmaceutical Co., Ltd., Teijin Pharma Ltd., AbbVie GK, Asahi Kasei Pharma Corp., Taisho Toyama Pharmaceutical Co., Ltd., SymBio Pharmaceuticals Ltd., Janssen Pharmaceutical K.K., Celltrion Inc., Nipponkayaku Co. Ltd., and UCB Japan, Consultant for: Astra Zeneca K.K., Eli Lilly Japan K.K., Novartis Pharma K.K., Mitsubishi Tanabe Pharma Co., Abbivie GK, Nipponkayaku Co.Ltd, Janssen Pharmaceutical K.K., Astellas Pharma Inc., Taiho Pharmaceutical Co. Ltd., Chugai Pharmaceutical Co. Ltd., Taisho Toyama Pharmaceutical Co. Ltd., GlaxoSmithKline K.K., UCB Japan Co. Ltd., Consultant for: AbbVie, Asahi Kasei, Astellas, AstraZeneca, AYUMI, Bristol-Myers Squibb, Chugai, Daiichi Sankyo, Eisai, Eli Lilly Japan, Janssen, Mitsubishi Tanabe, Nippon Kayaku, Novartis, Pfizer Japan Inc, Taiho, Taisho Toyama, Takeda, Teijin, Consultant for: Astra Zeneca K.K., Eli Lilly Japan K.K., Novartis Pharma K.K., Mitsubishi Tanabe Pharma Co., Asahi Kasei Medical K.K., AbbVie GK, Daiichi Sankyo Co., Ltd., Bristol Myers Squibb, and Nipponkayaku Co. Ltd., Speakers bureau: Astellas Pharma Inc., Bristol Myers Squibb, Chugai Pharmaceutical Co., Ltd., Mitsubishi Tanabe Pharma Co., Pfizer Japan Inc., Santen Pharmaceutical Co., Ltd., Takeda Pharmaceutical Co., Ltd., Teijin Pharma Ltd., AbbVie GK, Asahi Kasei Pharma Corp., Taisho Toyama Pharmaceutical Co., Ltd., SymBio Pharmaceuticals Ltd., Janssen Pharmaceutical K.K., Celltrion Inc., Nipponkayaku Co. Ltd., and UCB Japan, Speakers bureau: AbbVie, Asahi Kasei, Astellas, AstraZeneca, AYUMI, Bristol-Myers Squibb, Chugai, Daiichi Sankyo, Eisai, Eli Lilly Japan, Janssen, Mitsubishi Tanabe, Nippon Kayaku, Novartis, Pfizer Japan Inc, Taiho, Taisho Toyama, Takeda, Teijin, Speakers bureau: AbbVie GK., Bristol–Myers K.K., Chugai Pharmaceutical Co. Ltd., Mitsubishi Tanabe Pharma Co., Pfizer Japan Inc., Astellas Pharma Inc, Diaichi Sankyo Co. Ltd., Eisai Co. Ltd., Sanofi K.K., Teijin Pharma Ltd., Takeda Pharmaceutical Co. Ltd., Novartis Pharma K.K., Hisashi Yamanaka Grant/research support from: AbbVie, Eisai, Bristol-Meyers, Novartis, Behringer, Astellas, Kaken, Nippon-Shinyaku, Pfizer, UCB, Ayumi, Ono, Daiichi-Sankyo, Taisyo-Toyama, Takeda, Tanabe-Mitsubishi, Chugai, Teijin Pharma, Torii, YLbio, Speakers bureau: Bristol-Meyers, Astellas, Pfizer, Daiichi-Sankyo, Takeda, Tanabe-Mitsubishi, Chugai, Teijin Pharma, YLbio, Toshihiro Nanki Grant/research support from: Chugai, Eisai, Takeda, Teijin, Eli Lilly, Bristol-Myers, AbbVie, Ono, Novartis, Asahikasei, Mitsubishi-Tanabe, Astellas, Ayumi, Pfizer, Daiichi Sankyo, Shionogi, Sanofi, Nippon Kayaku, Yutoku, Actelion, UCB, Bayer, Nihon Pharmaceutical., Consultant for: UCB, Eisai, Chugai, Ono, Gilead., Speakers bureau: Mitsubishi-Tanabe, Chugai, Eisai, Takeda, Astellas, Janssen, Eli Lilly, Ayumi, Pfizer, Asahikasei, Sanofi, Daiichi Sankyo, Otsuka, AbbVie, Ono, Teijin, Nippon Kayaku, UCB., Hisanori Umehara: None declared, Nobuyuki Yasuda Shareholder of: Eisai Co., Ltd., Employee of: KAN Research Institute, Inc. (subsidiary of Eisai Co., Ltd.), Fumitoshi Tago Employee of: Eisai Co., Ltd., Yasumi Kitahara Shareholder of: Eisai Co., Ltd., Employee of: Eisai Co., Ltd., Makoto Kawakubo Shareholder of: Eisai Co., Ltd., Employee of: Eisai Co., Ltd., Kentaro Torii Shareholder of: Eisai Co., Ltd., Employee of: Eisai Co., Ltd., Seiichiro Hojo Employee of: Eisai Co., Ltd., Tetsu Kawano Employee of: KAN Research Institute, Inc. (subsidiary of Eisai Co., Ltd.), Toshio Imai Shareholder of: Eisai Co., Ltd., Employee of: KAN Research Institute, Inc. (subsidiary of Eisai Co., Ltd.)
- Published
- 2019
49. SAT0126 A PHASE 2 STUDY OF E6011, AN ANTI-FRACTALKINE MONOCLONAL ANTIBODY, IN PATIENTS WITH RHEUMATOID ARTHRITIS INADEQUATELY RESPONDING TO BIOLOGICS
- Author
-
Makoto Kawakubo, Hisanori Umehara, Yasumi Kitahara, Fumitoshi Tago, Tetsu Kawano, Nobuyuki Yasuda, Tsutomu Takeuchi, Seiichiro Hojo, Toshihiro Nanki, Hisashi Yamanaka, Hayato Hisaki, Yoshiya Tanaka, and Toshio Imai
- Subjects
030203 arthritis & rheumatology ,0301 basic medicine ,medicine.medical_specialty ,Study drug ,business.industry ,Phases of clinical research ,Placebo group ,Acr20 response ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Internal medicine ,Injection site ,medicine ,Comparison study ,In patient ,business ,Bristol-Myers - Abstract
Background Fractalkine (CX3CL1, designated as FKN hereafter) is the sole member of the CX3C-chemokine which leads to dual actions, chemotaxis and cell adhesion for leukocytes expressing the cognate receptor, CX3CR1, during their migration. We have conducted clinical trials of E6011, a novel humanized anti-FKN monoclonal antibody, for patients with rheumatoid arthritis (RA) in Japan1. This is the first report of a Phase 2, multicenter, randomized, double-blind, placebo-controlled, parallel-group comparison study of E6011 in RA patients inadequately responding to biologics (NCT02960490). Objectives To evaluate efficacy and safety of E6011 compared with placebo. Methods During the 24-week double-blind period, patients with moderately to severely active RA of inadequate response to biologics were randomly assigned to E6011 400 mg or placebo groups at a 1:1 ratio. Patients who continued the study beyond Week 12 were further allocated to E6011 200 mg or 400 mg at a 1:1 ratio within the initially assigned group. Patients received either E6011 400 mg or placebo at Weeks 0, 1, 2, and every 2 weeks subsequently until Week 10 and then E6011 200 mg or 400 mg every 2 weeks between Weeks 12 and 22 in a double-blind manner. This abstract reports the results from the first 12-week placebo controlled period. Results A total of 64 subjects (33 in the placebo group and 31 in the E6011 400 mg group) received study drug. Of the 64 subjects, 55 completed and 9 discontinued study treatment prematurely during the 12-week placebo controlled double-blind period. The ACR20 response rate at Week 12 (non-responder imputation), the primary endpoint, was 27.3% (9/33 subjects) in the placebo group and 22.6% (7/31 subjects) in the E6011 group. ACR50 and ACR70 response rate at Week 12 were 3.0%, 0% in the placebo group and 9.7%, 3.2% in the E6011 group, respectively. Numerically greater improvement from baseline was found for some parameters of the ACR components in the E6011 group, however, no statistically significant differences were found in any of the ACR components between the placebo and E6011 groups. From the exploratory PK exposure analysis, there was a tendency that the effect of E6011 became clearer in the subjects who achieved higher serum trough E6011 concentration. Adverse events that occurred in at least 2 subjects in the E6011 group were stomatitis, injection site erythema, nasopharyngitis, and blood creatine phosphokinase increased. As a results, E6011 was well tolerated with no notable safety concerns at doses of 400 mg when administered subcutaneously for 12 weeks. Conclusion E6011 400 mg was well tolerated but did not show clear efficacy at Week 12 compared with placebo in RA patients with inadequately responding to biologics. Further investigation to seek an optimal clinical dose and evaluation period of E6011 are warranted. Reference [1] Tanaka Y, et al., Mod Rheumatol (2018) 28, 58-65 Acknowledgement The authors wish to thank the study investigators. Disclosure of Interests Yoshiya Tanaka Grant/research support from: Abbvie, Astellas, Bristol-Myers Squibb, Chugai, Daiichi-Sankyo, Eisai, Mitsubishi-Tanabe, MSD, Ono, Taisho-Toyama, Takeda, Speakers bureau: Abbvie, Asahi-kasei, Astellas, Bristol-Myers Squibb, Chugai, Daiichi-Sankyo, Eli Lilly, Eisai, Glaxo-Smithkline, Janssen, Mitsubishi-Tanabe, Novartis, Pfizer Japan Inc, Sanofi, Takeda, UCB, YL Biologics, Tsutomu Takeuchi Grant/research support from: Astellas Pharma Inc, Chugai Pharmaceutical Co, Ltd., Daiichi Sankyo Co., Ltd., Takeda Pharmaceutical Co., Ltd., AbbVie GK, Asahikasei Pharma Corp., Mitsubishi Tanabe Pharma Co., Pfizer Japan Inc., Eisai Co., Ltd., AYUMI Pharmaceutical Corporation, Nipponkayaku Co. Ltd., Novartis Pharma K.K., Grant/research support from: AbbVie, Asahi Kasei, Astellas, AstraZeneca, AYUMI, Bristol-Myers Squibb, Chugai, Daiichi Sankyo, Eisai, Eli Lilly Japan, Janssen, Mitsubishi Tanabe, Nippon Kayaku, Novartis, Pfizer Japan Inc, Taiho, Taisho Toyama, Takeda, Teijin, Grant/research support from: Astellas Pharma Inc., Bristol Myers Squibb, Chugai Pharmaceutical Co., Ltd., Mitsubishi Tanabe Pharma Co., Pfizer Japan Inc., Santen Pharmaceutical Co., Ltd., Takeda Pharmaceutical Co., Ltd., Teijin Pharma Ltd., AbbVie GK, Asahi Kasei Pharma Corp., Taisho Toyama Pharmaceutical Co., Ltd., SymBio Pharmaceuticals Ltd., Janssen Pharmaceutical K.K., Celltrion Inc., Nipponkayaku Co. Ltd., and UCB Japan, Consultant for: Astra Zeneca K.K., Eli Lilly Japan K.K., Novartis Pharma K.K., Mitsubishi Tanabe Pharma Co., Abbivie GK, Nipponkayaku Co.Ltd, Janssen Pharmaceutical K.K., Astellas Pharma Inc., Taiho Pharmaceutical Co. Ltd., Chugai Pharmaceutical Co. Ltd., Taisho Toyama Pharmaceutical Co. Ltd., GlaxoSmithKline K.K., UCB Japan Co. Ltd., Consultant for: AbbVie, Asahi Kasei, Astellas, AstraZeneca, AYUMI, Bristol-Myers Squibb, Chugai, Daiichi Sankyo, Eisai, Eli Lilly Japan, Janssen, Mitsubishi Tanabe, Nippon Kayaku, Novartis, Pfizer Japan Inc, Taiho, Taisho Toyama, Takeda, Teijin, Consultant for: Astra Zeneca K.K., Eli Lilly Japan K.K., Novartis Pharma K.K., Mitsubishi Tanabe Pharma Co., Asahi Kasei Medical K.K., AbbVie GK, Daiichi Sankyo Co., Ltd., Bristol Myers Squibb, and Nipponkayaku Co. Ltd., Speakers bureau: Astellas Pharma Inc., Bristol Myers Squibb, Chugai Pharmaceutical Co., Ltd., Mitsubishi Tanabe Pharma Co., Pfizer Japan Inc., Santen Pharmaceutical Co., Ltd., Takeda Pharmaceutical Co., Ltd., Teijin Pharma Ltd., AbbVie GK, Asahi Kasei Pharma Corp., Taisho Toyama Pharmaceutical Co., Ltd., SymBio Pharmaceuticals Ltd., Janssen Pharmaceutical K.K., Celltrion Inc., Nipponkayaku Co. Ltd., and UCB Japan, Speakers bureau: AbbVie, Asahi Kasei, Astellas, AstraZeneca, AYUMI, Bristol-Myers Squibb, Chugai, Daiichi Sankyo, Eisai, Eli Lilly Japan, Janssen, Mitsubishi Tanabe, Nippon Kayaku, Novartis, Pfizer Japan Inc, Taiho, Taisho Toyama, Takeda, Teijin, Speakers bureau: AbbVie GK., Bristol–Myers K.K., Chugai Pharmaceutical Co. Ltd., Mitsubishi Tanabe Pharma Co., Pfizer Japan Inc., Astellas Pharma Inc, Diaichi Sankyo Co. Ltd., Eisai Co. Ltd., Sanofi K.K., Teijin Pharma Ltd., Takeda Pharmaceutical Co. Ltd., Novartis Pharma K.K., Hisashi Yamanaka Grant/research support from: AbbVie, Eisai, Bristol-Meyers, Novartis, Behringer, Astellas, Kaken, Nippon-Shinyaku, Pfizer, UCB, Ayumi, Ono, Daiichi-Sankyo, Taisyo-Toyama, Takeda, Tanabe-Mitsubishi, Chugai, Teijin Pharma, Torii, YLbio, Speakers bureau: Bristol-Meyers, Astellas, Pfizer, Daiichi-Sankyo, Takeda, Tanabe-Mitsubishi, Chugai, Teijin Pharma, YLbio, Toshihiro Nanki Grant/research support from: Chugai, Eisai, Takeda, Teijin, Eli Lilly, Bristol-Myers, AbbVie, Ono, Novartis, Asahikasei, Mitsubishi-Tanabe, Astellas, Ayumi, Pfizer, Daiichi Sankyo, Shionogi, Sanofi, Nippon Kayaku, Yutoku, Actelion, UCB, Bayer, Nihon Pharmaceutical., Consultant for: UCB, Eisai, Chugai, Ono, Gilead., Speakers bureau: Mitsubishi-Tanabe, Chugai, Eisai, Takeda, Astellas, Janssen, Eli Lilly, Ayumi, Pfizer, Asahikasei, Sanofi, Daiichi Sankyo, Otsuka, AbbVie, Ono, Teijin, Nippon Kayaku, UCB., Hisanori Umehara: None declared, Nobuyuki Yasuda Shareholder of: Eisai Co., Ltd., Employee of: KAN Research Institute, Inc. (subsidiary of Eisai Co., Ltd.), Fumitoshi Tago Employee of: Eisai Co., Ltd., Yasumi Kitahara Shareholder of: Eisai Co., Ltd., Employee of: Eisai Co., Ltd., Makoto Kawakubo Shareholder of: Eisai Co., Ltd., Employee of: Eisai Co., Ltd., Hayato Hisaki Shareholder of: Eisai Co., Ltd., Employee of: Eisai Co., Ltd., Seiichiro Hojo Employee of: Eisai Co., Ltd., Tetsu Kawano Employee of: KAN Research Institute, Inc. (subsidiary of Eisai Co., Ltd.), Toshio Imai Shareholder of: Eisai Co., Ltd., Employee of: KAN Research Institute, Inc. (subsidiary of Eisai Co., Ltd.)
- Published
- 2019
50. Anti-Apoptotic Effects of Recombinant Human Hepatocyte Growth Factor on Hepatocytes Were Associated with Intrahepatic Hemorrhage Suppression Indicated by the Preservation of Prothrombin Time
- Author
-
Katsuhiro Moriya, Akio Ido, Kana Negishi, Yoshikazu Kuboi, Yoshihisa Arita, Tetsu Kawano, Motohiro Soejima, Hiroko Toyoda, Takashi Obara, Etsuko Ohta, Sotaro Motoi, Hirohito Tsubouchi, and Toshio Imai
- Subjects
Male ,medicine.medical_treatment ,Apoptosis ,lcsh:Chemistry ,cytokeratin-18 ,Mice ,0302 clinical medicine ,lcsh:QH301-705.5 ,Spectroscopy ,Cells, Cultured ,medicine.diagnostic_test ,Chemistry ,Hepatocyte Growth Factor ,Liver Diseases ,General Medicine ,Recombinant Proteins ,Computer Science Applications ,030220 oncology & carcinogenesis ,030211 gastroenterology & hepatology ,Hepatocyte growth factor ,Intracellular ,medicine.drug ,Hemorrhage ,Catalysis ,Article ,Inorganic Chemistry ,03 medical and health sciences ,Cytokeratin ,Inhibitory Concentration 50 ,Parenchyma ,medicine ,Animals ,Humans ,fas Receptor ,Physical and Theoretical Chemistry ,Molecular Biology ,Blood Coagulation ,Prothrombin time ,Growth factor ,Organic Chemistry ,Fas antigen ,acute liver failure ,Liver Failure, Acute ,prothrombin time ,In vitro ,Disease Models, Animal ,lcsh:Biology (General) ,lcsh:QD1-999 ,Cancer research ,Hepatocytes - Abstract
Hepatocyte growth factor (HGF) is an endogenously expressed bioactive substance that has a strong anti-apoptotic effect. In this study, we biochemically and histologically characterized the effects of rh-HGF on in vitro human hepatocyte injury and mouse acute liver failure (ALF) models, both of which were induced by antibody-mediated Fas signaling. rh-HGF inhibited intracellular caspase-3/7 activation and cytokeratin 18 (CK-18) fragment release in both models. Histologically, rh-HGF dramatically suppressed parenchymal damage and intrahepatic hemorrhage. Among the laboratory parameters, prothrombin time (PT) was strongly preserved by rh-HGF, and PT was well correlated with the degree of intrahepatic hemorrhage. These results showed that the anti-apoptotic effect of rh-HGF on hepatocytes coincided strikingly with the suppression of intrahepatic hemorrhage. PT was considered to be the best parameter that correlated with the intrahepatic hemorrhages associated with hepatocellular damage. The action of rh-HGF might derive not only from its anti-apoptosis effects on liver parenchymal cells but also from its stabilization of structural and vasculature integrity.
- Published
- 2019
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.