Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Ryoichi Katsube is active.

Publication


Featured researches published by Ryoichi Katsube.


Journal of Vascular Access | 2016

Ultrasound-guided block of selective branches of the brachial plexus for vascular access surgery in the forearm: a preliminary report

Hiroaki Matsuda; Yoshinari Oka; Shigeko Takatsu; Ryoichi Katsube; Ryuichi Yoshida; Takanori Oyama; Yoshimasa Takeda; Masashi Miyazaki

Purpose The operative field for vascular access (VA) surgery in the forearm is on the volar surface, and motor nerve block is not necessary for regional anesthesia. Therefore, selective block of branches of the brachial plexus may be a more efficient anesthesia technique. Methods Individual nerve blocks in the axillary brachial plexus and selective blocks of the musculocutaneous and medial antebrachial cutaneous nerves in the upper arm were performed using low doses and concentrations of a local anesthetic mixture of lidocaine and ropivacaine under ultrasound (US) guidance in patients undergoing VA surgery in the forearm. The targeted nerves were identified by continuous US tracing along the upper arm to the axilla in a short-axis view. We performed three VA surgeries in the forearm using an axillary brachial plexus block and four using a selective two-nerve bock in the upper arm. We recorded any additional anesthetic requirement and evaluated intraoperative pain using the Wong-Baker Faces Pain Rating Scale (WBFRS; 0 = no pain; 10 = worst pain). Results All of the target nerve branches were clearly identified by US tracing. All patients had satisfactory intraoperative pain control (0 or 2 score on WBFRS). Four patients required small additional doses of local anesthetic. Conclusions US-guided block of individual branches of the brachial plexus at the axilla achieved effective anesthesia using small amounts of local anesthetic. An advanced selective nerve block in the upper arm allows minimum necessary anesthesia and provides safe and efficient analgesia for VA surgery in the forearm.


Cancer Biology & Therapy | 2016

Iron depletion enhances the effect of sorafenib in hepatocarcinoma

Shinichi Urano; Toshiaki Ohara; Kazuhiro Noma; Ryoichi Katsube; Takayuki Ninomiya; Yasuko Tomono; Hiroshi Tazawa; Shunsuke Kagawa; Yasuhiro Shirakawa; Fumiaki Kimura; Kazuhiro Nouso; Akihiro Matsukawa; Kazuhide Yamamoto; Toshiyoshi Fujiwara

ABSTACT Human hepatocellular carcinoma (HCC) is known to have a poor prognosis. Sorafenib, a molecular targeted drug, is most commonly used for HCC treatment. However, its effect on HCC is limited in clinical use and therefore new strategies regarding sorafenib treatment are required. Iron overload is known to be associated with progression of chronic hepatitis and increased risk of HCC. We previously reported that iron depletion inhibited cancer cell proliferation and conversely induced angiogenesis. Indeed iron depletion therapy including iron chelator needs to be combined with anti-angiogenic drug for its anti-cancer effect. Since sorafenib has an anti-angiogenic effect by its inhibitory targeting VEGFR, we hypothesized that sorafenib could complement the anti-cancer effect of iron depletion. We retrospectively analyzed the relationship between the efficacy of sorafenib and serum iron-related markers in clinical HCC patients. In clinical cases, overall survival was prolonged in total iron binding capacity (TIBC) high- and ferritin low-patients. This result suggested that the low iron-pooled patients, who could have a potential of more angiogenic properties in/around HCC tumors, could be adequate for sorafenib treatment. We determined the effect of sorafenib (Nexavar®) and/or deferasirox (EXJADE®) on cancer cell viability, and on cell signaling of human hepatocarcinoma HepG2 and HLE cells. Both iron depletion by deferasirox and sorafenib revealed insufficient cytotoxic effect by each monotherapy, however, on the basis of increased angiogenesis by iron depletion, the addition of deferasirox enhanced anti-proliferative effect of sorafenib. Deferasirox was confirmed to increase vascular endothelial growth factor (VEGF) secretion into cellular supernatants by ELISA analysis. In in vivo study sorafenib combined with deferasirox also enhanced sorafenib-induced apoptosis. These results suggested that sorafenib combined with deferasirox could be a novel combination chemotherapy for HCC.


Clinical Cancer Research | 2018

Cancer-Associated Fibroblasts Affect Intratumoral CD8+ and FoxP3+ T Cells Via IL6 in the Tumor Microenvironment

Takuya Kato; Kazuhiro Noma; Toshiaki Ohara; Hajime Kashima; Yuki Katsura; Hiroaki Sato; Satoshi Komoto; Ryoichi Katsube; Takayuki Ninomiya; Hiroshi Tazawa; Yasuhiro Shirakawa; Toshiyoshi Fujiwara

Purpose: Cancer-associated fibroblasts (CAFs) in the tumor microenvironment (TME) play a central role in tumor progression. We investigated whether CAFs can regulate tumor-infiltrating lymphocytes (TILs) and their role in tumor immunosuppression. Experimental Design: A total of 140 cases of esophageal cancer were analyzed for CAFs and CD8+ or forkhead box protein 3 (FoxP3+) TILs by IHC. We analyzed cytokines using murine or human fibroblasts and cancer cells. Murine-derived fibroblasts and cancer cells were also inoculated into BALB/c or BALB/c-nu/nu mice and the tumors treated with recombinant IL6 or anti-IL6 antibody. Results: CD8+ TILs and CAFs were negatively correlated in intratumoral tissues (P < 0.001), whereas FoxP3+ TILs were positively correlated (P < 0.001) in esophageal cancers. Cocultured Colon26 cancer cells and fibroblasts resulted in accelerated tumor growth in BALB/c mice, along with decreased CD8+ and increased FoxP3+ TILs, compared with cancer cells alone. In vitro, IL6 was highly secreted in both murine and human cancer cell/fibroblast cocultures. IL6 significantly increased Colon26 tumor growth in immune-competent BALB/c (P < 0.001) with fewer CD8+ TILs than untreated tumors (P < 0.001), whereas no difference in BALB/c-nu/nu mice. In contrast, FoxP3+ TILs increased in IL6-treated tumors (P < 0.001). IL6 antibody blockade of tumors cocultured with fibroblasts resulted not only in regression of tumor growth but also in the accumulation of CD8+ TILs in intratumoral tissues. Conclusions: CAFs regulate immunosuppressive TIL populations in the TME via IL6. IL6 blockade, or targeting CAFs, may improve preexisting tumor immunity and enhance the efficacy of conventional immunotherapies. Clin Cancer Res; 24(19); 4820–33. ©2018 AACR.


Oncotarget | 2017

Iron depletion is a novel therapeutic strategy to target cancer stem cells

Takayuki Ninomiya; Toshiaki Ohara; Kazuhiro Noma; Yuki Katsura; Ryoichi Katsube; Hajime Kashima; Takuya Kato; Yasuko Tomono; Hiroshi Tazawa; Shunsuke Kagawa; Yasuhiro Shirakawa; Fumiaki Kimura; Ling Chen; Tomonari Kasai; Masaharu Seno; Akihiro Matsukawa; Toshiyoshi Fujiwara

Adequate iron levels are essential for human health. However, iron overload can act as catalyst for the formation of free radicals, which may cause cancer. Cancer stem cells (CSCs), which maintain the hallmark stem cell characteristics of self-renewal and differentiation capacity, have been proposed as a driving force of tumorigenesis and metastases. In the present study, we investigated the role of iron in the proliferation and stemness of CSCs, using the miPS-LLCcm cell model. Although the anti-cancer agents fluorouracil and cisplatin suppressed the proliferation of miPS-LLCcm cells, these drugs did not alter the expression of stemness markers, including Nanog, SOX2, c-Myc, Oct3/4 and Klf4. In contrast, iron depletion by the iron chelators deferasirox and deferoxamine suppressed the proliferation of miPS-LLCcm cells and the expression of stemness markers. In an allograft model, deferasirox inhibited the growth of miPS-LLCcm implants, which was associated with decreased expression of Nanog and Sox2. Altogether, iron appears to be crucial for the proliferation and maintenance of stemness of CSCs, and iron depletion may be a novel therapeutic strategy to target CSCs.


International Journal of Cancer | 2018

Cancer-associated fibroblasts (CAFs) promote the lymph node metastasis of esophageal squamous cell carcinoma: Cancer-associated fibroblasts (CAFs) promote the lymph node metastasis of esophageal squamous cell carcinoma

Hajime Kashima; Kazuhiro Noma; Toshiaki Ohara; Takuya Kato; Yuki Katsura; Satoshi Komoto; Hiroaki Sato; Ryoichi Katsube; Takayuki Ninomiya; Hiroshi Tazawa; Yasuhiro Shirakawa; Toshiyoshi Fujiwara

Lymph node metastasis is a pathognomonic feature of spreading tumors, and overcoming metastasis is a challenge in attaining more favorable clinical outcomes. Esophageal cancer is an aggressive tumor for which lymph node metastasis is a strong poor prognostic factor, and the tumor microenvironment (TME), and cancer‐associated fibroblasts (CAFs) in particular, has been implicated in esophageal cancer progression. CAFs play a central role in the TME and have been reported to provide suitable conditions for the progression of esophageal cancer, similar to their role in other malignancies. However, little is known concerning the relevance of CAFs to the lymph node metastasis of esophageal cancer. Here, we used clinical samples of esophageal cancer to reveal that CAFs promote lymph node metastasis and subsequently verified the intercellular relationships in vitro and in vivo using an orthotopic metastatic mouse model. In the analysis of clinical samples, FAP+ CAFs were strongly associated with lymph node metastasis rather than with other prognostic factors. Furthermore, CAFs affected the ability of esophageal cancer cells to acquire metastatic phenotypes in vitro; this finding was confirmed by data from an in vivo orthotopic metastatic mouse model showing that the number of lymph node metastases increased upon injection of cocultured cancer cells and CAFs. In summary, we verified in vitro and in vivo that the accumulation of CAFs enhances the lymph node metastasis of ESCC. Our data suggest that CAF targeted therapy can reduce lymph node metastasis and improve the prognosis of patients with esophageal cancer in the future.


Cancer Research | 2016

Abstract 1560: Correlation of FAP(fibroblast activation protein)-expressing cancer associated fibroblasts (CAFs) and tumor metastasis in esophageal carcinoma

Hajime Kashima; Kazuhiro Noma; Yuki Katsura; Takuya Kato; Ryoichi Katsube; Takayuki Ninomiya; Toshiaki Ohara; Hiroshi Tazawa; Shunsuke Kagawa; Yasuhiro Shirakawa; Toshiyoshi Fujiwara

Background: The tumor and its microenvironment have dynamic interactions and signaling that strongly affect tumor progression. Cancer associated fibroblasts (CAFs) are activated fibroblasts and thought to be an important player in the tumor microenvironment. Although there are several indicators of CAF, fibroblast activation protein (FAP) is unique in its selectivity for CAFs in comparison with other markers. The aim of this study is to investigate CAFs expressing FAP and its relationship to cancer metastasis in esophageal cancer. Methods: Sections of paraffin-embedded resected primary human esophageal cancer specimens from 2008 through 2010 in Okayama University hospital were stained with antibody directed against FAP. Overall percentage of stromal FAP staining of the primary tumor was assessed semi-quantitatively (0, 1, 2, 3, 4, 5) and staining intensity was also graded (none, weak, intermediate, strong). Survival time and time to recurrence data were analyzed using Kaplan-Meier plots, log-rank tests, and Cox proportional hazards models. Results: 49 patients with resected specimens were available for study with 26 (53.1%) stage I, 8 (16.3%) stage II, 14 (28.6%) stage III, and 1 (2.0%) stage IV patients. All patients were divided to 2 groups, FAP high group and FAP low group, which are 25 patients (51%) and 24 (49%) respectively. The cutoff for subgroups was defined at the median value. FAP (immune) expression at tumoral stroma was a significant predictive factor for lymph node metastasis (p Conclusions: Our clinicopathological analysis indicates that patients whose esophageal carcinoma have high levels of stromal FAP expression are more likely to have an aggressive disease progression and a potential development of metastases and recurrence. Although therapeutic strategies targeting the tumor cells have been generally inadequate in esophageal carcinomas yet, here we announce that a stroma-targeted therapy should be considered. Now we are ongoing to evaluate metastatic potential of CAFs in vitro and vivo, in order to elucidate the function of FAP expressing CAFs and, in future to establish a novel therapeutic strategy. Citation Format: Hajime Kashima, Kazuhiro Noma, Yuki Katsura, Takuya Kato, Ryoichi Katsube, Takayuki Ninomiya, Toshiaki Ohara, Hiroshi Tazawa, Shunsuke Kagawa, Yasuhiro Shirakawa, Toshiyoshi Fujiwara. Correlation of FAP(fibroblast activation protein)-expressing cancer associated fibroblasts (CAFs) and tumor metastasis in esophageal carcinoma. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 1560.


Cancer Research | 2015

Abstract 4243: Iron control is a novel therapeutic target of cancer stem cells

Takayuki Ninomiya; Toshiaki Ohara; Hajime Kashima; Ryoichi Katsube; Kazuhiro Noma; Yasuko Tomono; Akifumi Mizutani; Tomonari Kasai; Masaharu Seno; Shinji Kuroda; Hiroyuki Kishimoto; Hiroshi Tazawa; Yasuhiro Shirakawa; Shunsuke Kagawa; Toshiyoshi Fujiwara

Proceedings: AACR 106th Annual Meeting 2015; April 18-22, 2015; Philadelphia, PA Background: Iron overload is known to cause cancer in animal models. Several studies have shown that iron deprivation treatment has a strong anti-cancer effect. However, it is unclear whether iron deprivation treatment suppresses cancer stem cells or not. A cancer stem cell model, miPS-LLCcm, was recently epigenetically established from murine induced pluripotent stem cells (miPS cells) in Okayama University. In this model, the green fluorescent protein (GFP) and the puromycin resistant gene were inserted into the 59-bp untranslated region of the Nanog gene of miPS cells. By this procedure, undifferentiated cancer progenitor cells are recognized as GFP positive cells. We then examined the iron dependency of these cancer stem cells both in in vitro and in vivo studies and we used this model to examine the possibility of cancer stem cells by iron deprivation. Materials and Methods: In in vitro studies, the miPS-LLCcm cells were used as cancer stem cells and colon26 and 4T1 cells were used as differentiated murine cancer cells. Puromycin was used to purify the cancer stem cells before seeding of the miPS-LLCcm cells. The dependency on iron for cell proliferation was examined following transferrin addition. Transferrin (Holo) was used to simulate an iron rich condition. Iron free medium and the iron chelators, Deferasirox and Deferoxamine, plus 1% fetal bovine serum (FBS) were used to simulate iron depletion conditions. Cell proliferation assays and flow cytometric analyses were performed 48 hours after adjustment of the iron concentration level. In in vivo studies, an iron depleted diet was used to simulate iron depleted conditions. Nude mice were divided into normal diet and iron depleted diet groups. The mice were fed with these diets for three weeks and then a suspension of miPS-LLCcm cells was injected into the backs of the nude mice. Tumor size was measured and the tumors were immunohistologically examined. Results: In the in vitro studies, transferrin strongly promoted the proliferation of cancer stem cells under iron depletion conditions compared to no transferrin(p<0.001). However, transferrin did not promote the proliferation of the differentiated 4T1 and colon26 cancer cells. The number of GFP-expressing cancer stem cells decreased as the iron concentration was decreased. In the vivo studies, iron depletion significantly suppressed the tumor growth of the cancer stem cells (p<0.05). Immunohistological analysis indicated that Nanog expression was also decreased in the tumors of the iron depletion diet group. Conclusions: Iron is a key element for the proliferation and differentiation of cancer stem cells. Iron controlling therapy including iron chelators is a novel therapeutic target of cancer stem cells. Citation Format: Takayuki Ninomiya, Toshiaki Ohara, Hajime Kashima, Ryoichi Katsube, Kazuhiro Noma, Yasuko Tomono, Akifumi Mizutani, Tomonari Kasai, Masaharu Seno, Shinji Kuroda, Hiroyuki Kishimoto, Hiroshi Tazawa, Yasuhiro Shirakawa, Shunsuke Kagawa, Toshiyoshi Fujiwara. Iron control is a novel therapeutic target of cancer stem cells. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 4243. doi:10.1158/1538-7445.AM2015-4243


Cancer Research | 2015

Abstract 401: A novel photoimmunotherapy targeting cancer-associated fibroblasts (CAFs) overcomes therapeutic resistance in human esophageal cancer

Ryoichi Katsube; Kazuhiro Noma; Shinichiro Watanabe; Shinichi Urano; Takayuki Ninomiya; Toshiaki Ohara; Hiroshi Tazawa; Shunsuke Kagawa; Hisataka Kobayashi; Toshiyoshi Fujiwara

Cancer associated fibroblasts (CAFs) are thought to play an essential role in cancer invasion, migration, metastasis and resistance to anticancer drugs. CAFs were originally defined as fibroblasts expressing α-SMA in cancer tissue; however, fibroblast activation protein (FAP) has been recently identified as a more functional and specific surface protein of CAFs. The photoimmunotherapy (PIT) is a new molecular targeted therapy, which is based on a near-infrared (NIR) photosensitizer IR700, conjugated to monoclonal antibody (mAb) targeting particular molecules. PIT induces the selective destruction of targeted cells. The aim of this study is to analyze the influence of CAFs on human esophageal cancer and to develop a novel therapeutic strategy to eliminate CAFs by using FAP-targeting PIT. We produced the IR700-FAP antibody for this study. Human esophageal cancer cell lines (TE-4 and OE-19) and FEF3 human fibroblasts were used in this study. TE-4 and OE-19 cells stimulated with conditioned medium (CM) of CAFs exhibited malignant phenotypes confirmed by invasion assay, migration assay, and colony formation assay. Tumor cells were significantly more malignant when directly co-cultured with CAFs. We further examined whether tumor cells stimulated by CM (CAFs) could acquire the resistance to chemotherapy or radiotherapy. Cell viability assay showed that tumor cells stimulated with CAFs was more resistant to conventional chemotherapy or radiation than unstimulated tumor cells. Western blot analysis demonstrated that E-cadherin expression was decreased and that of vimentin was increased in CAF-stimulated tumor cells, indicating the epithelial mesenchymal transition (EMT) induction. Moreover, OE-19 cells stimulated with CAFs contained more CD133-positive cancer stem-like cells. In vivo experiments demonstrated that subcutaneous TE4 tumors were more refractory to chemotherapy in the presence of co-inoculated CAFs as observed in vitro. Finally, we investigated whether elimination of CAFs by FAP-targeting PIT could affect the resistance to chemotherapy in vitro and in vivo. In vivo PIT following intratumoral anti-FAP mAb-conjugated IR700 suppressed the growth of subcutaneous tumors co-inoculated with CAFs. These results demonstrated that human tumor cells became more malignant and resistant in the presence of CAFs, which could be reversed by using CAF-specific PIT. Targeting fibroblast itself is a unique strategy and can be clinically promising as combination targeting cancer cells and their fundamental microenvironment. Citation Format: Ryoichi Katsube, Kazuhiro Noma, Shinichiro Watanabe, Shinichi Urano, Takayuki Ninomiya, Toshiaki Ohara, Hiroshi Tazawa, Shunsuke Kagawa, Hisataka Kobayashi, Toshiyoshi Fujiwara. A novel photoimmunotherapy targeting cancer-associated fibroblasts (CAFs) overcomes therapeutic resistance in human esophageal cancer. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 401. doi:10.1158/1538-7445.AM2015-401


Cancer Research | 2014

Abstract 1681: Iron chelation therapy increased the anticancer effect of sorafenib in hepatocarcinoma

Shinichi Urano; Toshiaki Ohara; Ryoichi Katsube; Shinichiro Watanabe; Kazuhiro Noma; Yasuko Tomono; Hiroshi Tazawa; Kazuhiro Nouso; Yasuhiro Shirakawa; Toshiyoshi Fujiwara

Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA [Purpose] Iron overload is known to associate with the progression of chronic hepatitis and the increase risk of hepatocellular carcinoma (HCC). We have reported that iron depletion inhibited the cancer cell proliferation and reciprocally induced angiogenesis. Anti-angiogenic drug synergistically enhanced the anti-tumor effect in iron depletion condition. Sorafenib is molecular-targeting drug with anti-angiogenic effect in HCC. We hypothesized that iron depletion also enhanced the effect of sorafenib and investigated it. [Experimental Design] HepG2 and HLE (Human HCC cell line) were used in this study. We used sorafenib (NexavarTM) and an oral iron chilator, deferasirox (EXJADETM). The anti-cancer effect and mechanism were determined using cell viability assay, VEGF ELISA, Flow Cytometry, Western blot analysis and in vivo subcutaneous tumor model. In clinical cases, we retrospectively analyzed the relationship between sorafenib response and serum iron-related makers(n=58). [Results] In HepG2 and HLE cells, deferasirox suppressed the cancer cell proliferation by itself and increased the suppression of sorafenib. Deferasirox increased VEGF secretion of supernatant on dose dependent manner. In the analysis of combination therapy, cell cycle arrest and apoptosis was enhanced in the combination therapy by down-regulation of cyclinD1 and cyclinE and up-regulation of cleaved PARP. The suppression of HepG2 subcutaneous tumor was enhanced in the combination therapy. In clinical cases, sorafenib prolonged overall survival in TIBC high and Ferritin low group. [Conclusion] Iron depletion by deferasirox increased the anti-cancer effect of sorafenib in vitro and in vivo experiments. In clinical cases, sorafenib was also effective in iron deficient patients. These results suggested that iron depletion therapy can be a novel combination chemotherapy with sorafenib for hepatocarcinoma patients. Citation Format: Shinichi Urano, Toshiaki Ohara, Ryoichi Katsube, Shinichiro Watanabe, Kazuhiro Noma, Yasuko Tomono, Hiroshi Tazawa, Kazuhiro Nouso, Yasuhiro Shirakawa, Toshiyoshi Fujiwara. Iron chelation therapy increased the anticancer effect of sorafenib in hepatocarcinoma. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 1681. doi:10.1158/1538-7445.AM2014-1681


Acta Medica Okayama | 2013

Successfully treated pneumatosis cystoides intestinalis with pneumoperitoneum onset in a patient administered α-glucosidase inhibitor.

Shunsuke Tanabe; Yasuhiro Shirakawa; Yuko Takehara; Naoaki Maeda; Ryoichi Katsube; Toshiaki Ohara; Kazufumi Sakurama; Kazuhiro Noma; Toshiyoshi Fujiwara

Collaboration


Dive into the Ryoichi Katsube's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge