GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
Filter
  • American Association for Cancer Research (AACR)  (12)
  • Shirakawa, Yasuhiro  (12)
Material
Publisher
  • American Association for Cancer Research (AACR)  (12)
Language
Years
Subjects(RVK)
  • 1
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 24, No. 19 ( 2018-10-01), p. 4820-4833
    Abstract: 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 & lt; 0.001), whereas FoxP3+ TILs were positively correlated (P & lt; 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 & lt; 0.001) with fewer CD8+ TILs than untreated tumors (P & lt; 0.001), whereas no difference in BALB/c-nu/nu mice. In contrast, FoxP3+ TILs increased in IL6-treated tumors (P & lt; 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.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 14_Supplement ( 2016-07-15), p. 4160-4160
    Abstract: Backgrounds: Cancer associated fibroblasts (CAFs) are thought to play an essential role in cancer invasion, migration, metastasis, and tumor immunosuppression. However, there has been still a little evidence of the correlation of tumor immunosuppression and CAFs in human esophageal carcinoma. The other hand, as like rising of PD-1 antibody targeting therapy to tumor immunosuppression have been shown dramatic cure responses in melanoma and now ongoing to other malignancies. Tumor-infiltrating lymphocytes (TILs) are considered typically to represent a host immune response against carcinoma. In many types of tumors TILs have been shown their strong correlation to patient's clinicopathological features. In this study, we evaluated the prognostic correlation of CAFs and TILs, which are classified respectively in tumor-associated CD8+ cytotoxic T lymphocytes (CTL) and FoxP3+ regulatory T cells (Treg) in surgically resected esophageal carcinoma. Materials and methods: Total 58 cases with esophageal carcinoma in our institution were evaluated for the presence of CAFs and TILs by immunohistochemistry. TILs of CTL and Treg were calculated each in the intratumoral and the peripheral tissues, and the cutoff for subgroups was defined at the median value. CAFs were defined as fibroblasts expressing alpha smooth muscle actin (α-SMA), and evaluated with the α-SMA scoring by using “Area Index”, which is calculated by imageJ. TILs and CAFs were assessed for the associations with pathological invasion depth (pT), lymph node metastases (pN), histological types, and disease-free survival (DFS) or overall survival (OS). Result: In intratumoral tissues, Treg was significantly associated with advanced T stages, and Treg and a CTL/Treg ratio were associated with lymph node metastasis. Higher CTL, higher CTL/Treg ratio and lower Treg were significantly associated with improved DFS and OS in univariate analysis. Furthermore, multivariate analysis demonstrated selected higher CTL as an independent prognostic factor (P = 0.010). On the other hand, in peripheral tissues, CTL, Treg, and CTL/Treg ratio were not correlated with clinicopathological factors or the any prognosis. Additionally the overexpression of CAFs was strongly associated with poor prognosis (P = 0.002) and the “Area Index” of α-SMA was inversely correlated with the CTL and CTL/Treg ratio in intratumoral tissues (P = 0.029, 0.017). It suggests that CAFs accumulation in tumor tissue is correlated to status of intratumoral immunesuppression. Conclusion: The CTL in intratumoral tissues are independent prognostic factors, and are considered to play an essential role in tumor immunity. Our results demonstrate that CAFs are significant correlation of immunosuppression in esophageal carcinoma. In the future, targeting CAFs therapy itself might improve tumor immunosuppression, and there is possibility to prolong a prognosis. Citation Format: Takuya Kato, Kazuhiro Noma, Yuki Katsura, Hajime Kashima, Takayuki Ninomiya, Toshiaki Ohara, Hiroshi Tazawa, Shunsuke Kagawa, Yasuhiro Shirakawa, Toshiyoshi Fujiwara. Prognostic correlation of tumor-infiltrating lymphocytes (TILs) and cancer associated fibroblasts (CAFs) in patients with human 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 4160.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 1741-1741
    Abstract: Cancer-associated fibroblasts (CAFs) in the tumor microenvironment (TME) are the most abundant cell population and play a central role in tumor progression. It is currently considered that the TME may strongly affect tumor immunosuppression. We investigated whether CAFs can regulate tumor-infiltrating lymphocytes (TILs) and their role in tumor immunosuppression. 140 cases of esophageal cancer were analyzed for CAFs, and CD8+ or forkhead box protein 3 (FoxP3)+ TILs by immunohistochemistry (IHC). We performed cytokine assays in a co-culture model 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 interleukin 6 (IL-6) or anti-IL-6 antibody. In clinical samples, patients with high CD8+ TIL numbers in intra-tumoral sites had significantly longer OS than those with low numbers (HR = 0.42, 95% CI = 0.25-0.70; P = 0.001). On the other hand, the high FoxP3+ TIL group for intra-tumoral tissues had significantly a shorter OS (HR = 2.82, 95% CI = 1.66-4.78; P & lt; 0.001). However, in peri-tumoral tissues, no significant correlations between CD8+ or FoxP3+ TIL numbers and prognosis were identified. Furthermore, CD8+ TILs and CAFs were negatively correlated in intra-tumoral tissues (P & lt; 0.001), while FoxP3+ TILs and CAFs, were positively correlated (P & lt; 0.001). In vivo, Co-cultured Colon26 cancer cells and NIH/3T3 fibroblasts resulted in subcutaneous tumors with accelerated growth in BALB/c mice, along with decreased CD8+ and increased FoxP3+ TILs, compared with cancer cells alone. Furthermore, the tumor progression ratio of co-cultured group to cancer cells alone group was demonstrated more strongly in BALB/c rather than BALB/c-nu/nu mice, which are immunodeficient mice. In vitro, IL-6 was secreted at high levels in both murine and human cancer cell/fibroblast co-cultures rather than cancer cell/fibroblast alone. Treatment with IL-6 significantly increased growth of Colon26 subcutaneous tumors in immune-competent BALB/c mice (P & lt; 0.001), whereas no difference was observed in BALB/c-nu/nu mice. IHC demonstrated fewer CD8+ TILs in Colon26+IL-6 subcutaneous tumors than in untreated tumors (P & lt; 0.001). In contrast, FoxP3+ TILs increased in IL-6-treated tumors (P & lt; 0.001). IL-6 antibody blockade of tumors co-cultured with fibroblasts resulted not only in regression of tumor growth but also in the accumulation of CD8+ TILs in intra-tumoral tissues. In conclusion, CAFs regulate immunosuppressive TIL populations in the TME via IL-6. IL-6 blockade, or targeting CAFs, may improve pre-existing tumor immunity and enhance the efficacy of conventional immunotherapies. Citation Format: Takuya Kato, Kazuhiro Noma, Yuki Katsura, Hiroaki Sato, Satoshi Kohmoto, Toshiaki Ohara, Hiroshi Tazawa, Yasuhiro Shirakawa, Masaru Inagaki, Toshiyoshi Fujiwara. Cancer-associated fibroblasts regulate intratumoral CD8+/FoxP3+ T cells via interleukin 6 in the tumor immune microenvironment [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 1741.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 15_Supplement ( 2015-08-01), p. CT123-CT123
    Abstract: Background: Telomerase activation is considered to be a critical step in carcinogenesis and its activity is closely correlated with human telomerase reverse transcriptase (hTERT) expression. We constructed an adenovirus 5 vector OBP-301 (Telomelysin), in which the hTERT promoter drives expression of E1A and E1B genes. OBP-301 causes selective replication and lysis of a variety of human cancer cells, and also inhibits the repair of radiation-induced DNA double-strand breaks, leading to radiosensitization. A phase I study has confirmed the safety and biological activity of intratumoral administration of OBP-301 alone in patients with advanced solid tumors in the United States. To further determine the feasibility, efficacy, and pharmacokinetics of OBP-301 in combination with radiotherapy, a phase I/II study was designed in elderly patients with esophageal cancer. Methods: Patients with histologically confirmed esophageal cancer who were not eligible for standard treatments such as surgery and chemotherapy were enrolled into this study (UMIN000010158). Study treatment consisted of intratumoral needle injections of OBP-301 on days 1, 18, and 32 of treatment. Radiation therapy was administered concurrently over 6 weeks, beginning on day 4, to a total of 60 Gy. Virus administration was performed by intratumoral injection of the primary or metastatic tumor through a flexible endoscope. OBP-301 doses will be escalated initially in cohorts of two for the first 9 patients (1 × 10e10 and 1 × 10e11 virus particles [vp]). Six subsequent patients will receive the highest dose (1 × 10e12 vp). Virus shedding will be monitored in the saliva, sputum, urine, and plasma by a quantitative DNA-PCR assay. Results: Six patients were enrolled and treated in the cohort with 1 × 10e10 vp of OBP-301. The patients comprised 4 males and 2 females, with median age of 83.5 years (range, 68 to 92 years). Only two patients had prior platinum-based chemotherapy. By November 2014, 3 patients completed treatment. All patients developed a transient, self-limited lymphopenia. A 92-year-old female showed a grade 4 lymphopenia classified as being possibly related to the treatment, although it recovered by the interruption of radiation. No other virus-related toxicities were noted. Objective responses were complete response (CR) in 2 patients and partial response (PR) with tumor regression, resulting in reopening of the esophagus, in 1 patient. Pathological analysis in biopsy specimens obtained from completely responded patients demonstrated no viable malignant cells for 3 to 5 months after the treatment completion. Conclusions: Multiple courses of endoscopic OBP-301 injection in combination with locoregional radiotherapy were feasible and well tolerated in elderly patients with esophageal cancer, and appeared to provide clinical benefit. Citation Format: Shunsuke Tanabe, Hiroshi Tazawa, Shunsuke Kagawa, Kazuhiro Noma, Kiyoto Takehara, Takeshi Koujima, Hajime Kashima, Takuya Kato, Shinji Kuroda, Satoru Kikuchi, Yasuhiro Shirakawa, Toshiyoshi Fujiwara. Phase I/II trial of endoscopic intratumoral administration of OBP-301, a novel telomerase-specific oncolytic virus, with radiation in elderly esophageal cancer patients. [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 CT123. doi:10.1158/1538-7445.AM2015-CT123
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 13_Supplement ( 2017-07-01), p. 5905-5905
    Abstract: Background: Cancer associated fibroblasts (CAFs) are activated fibroblasts and an important player in the tumor microenvironment. Their activity promotes cancer cell proliferation, migration, and invasion, and metastasis. The most prognostic factor in tumor progression is metastasis. Cancer metastasis is a multi-step process that tumor cells detach from primary site, survive in the bloodstream, and seed in target organ. Although previous studies have focused on the interaction between CAFs and cancer cells in primary tumor site, the roles of CAFs in blood circulation remain largely unknown. We investigated the effect of CAFs coexisting with cancer cells in bloodstream on tumor metastasis in vivo mouse model, and also examined the effect of CAFs in subcutaneous tumor. Methods: We used female BALB/c-nu/nu mice and BALB/c mice in the experiments. Cancer cells were mouse mammary carcinoma cell lines 4T1 transfected with luciferase, colon cancer cell lines Colon 26 transfected with luciferase. Fibroblast cell lines were mouse embryonic fibroblast MEF and NIH-3T3. In venous injection mouse model, we injected 1 × 106 cancer cells alone or the same number of cancer and fibroblast co-cultured together. When we injected cancer cells transfected with luciferase, the mice were subjected to in vivo imaging system (IVIS) and measured luminescence intensity of metastatic sites. We have also harvested lung and compared its weight and metastatic nodule under microscopy. Furthermore, in subcutaneous tumor metastatic mouse model, cancer cells alone or mixed with cancer cells and CAFs were subcutaneously inoculated into the mice. Results: In the group of mice injected with cancer cells and fibroblasts, luminescent intensity of each lungs were higher than cancer cell alone. Harvested lung weight and the number of metastatic nodules were also higher in the group with cancer cells and fibroblasts.In subcutaneous model, mice inoculated with cancer cells and fibroblast had much more metastatic sites than cancer cells alone. Conclusions: Our data indicate that CAFs promote tumor metastasis by stimulating cancer cells in blood circulation, as well as in primary tumor site.These findings suggest that CAFs in both bloodstream and primary site could be a promising therapeutic target. CAF-targeted therapy could reduce tumor metastasis and improve the prognosis of cancer patients. Citation Format: Hajime Kashima, Kazuhiro Noma, Hiroaki Sato, Yuki Katsura, Takuya Kato, Toshiaki Ohara, Hiroshi Tazawa, Yasuhiro Shirakawa, Toshiyoshi Fujiwara. Cancer associated fibroblasts promote tumor metastasis coexisting with cancer cells in blood circulation [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 5905. doi:10.1158/1538-7445.AM2017-5905
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2017
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 13_Supplement ( 2019-07-01), p. LB-310-LB-310
    Abstract: Esophageal cancer is known to one of the most aggressive malignant tumors, however, the effect of conventional chemotherapy and radiotherapy is insufficiently. Cancer-associated fibroblasts (CAFs) are thought to play an essential role in cancer invasion, migration, metastasis, immunosuppression, and resistance to anticancer drugs. Previously, we have developed a novel near-infrared photoimmunotherapy (NIR-PIT) targeting CAFs, which induces the selective destruction by targeting specific surface protein of CAFs; fibroblast activation protein (FAP). The aim of this study is to analyze the influence of CAFs for human esophageal cancer and to evaluate the improvement of conventional therapeutic resistance using NIR-PIT targeting CAFs.We produced the IR700-FAP antibody for this study. Human esophageal cancer cell lines (TE4 and OE19) and FEF3 human fibroblasts were used. At first, we demonstrated that cancer cells stimulated by CAFs acquired resistance to conventional therapies by cell viability assay. Furthermore, invasion, migration, and colony-formation assays demonstrated the CAF-stimulated cancer cells had a more malignant phenotype. Western blotting analysis showed that E-cadherin expression was decreased and vimentin expression was increased in CAF-stimulated cancer cells, indicating the epithelial mesenchymal transition induction. In vivo experiments demonstrated that subcutaneous tumors co-injected with CAFs were more refractory to chemotherapy than the tumors without CAFs as same as observed in vitro. Secondary, we found that acquired therapeutic resistance in stimulated cancer cells were restored by excluding continuous stimulation of CAFs. Finally, we investigated whether depleting CAFs by NIR-PIT could affect the chemo-resistance of cancer cells in vitro and in vivo. To evaluate the combination effect of conventional chemotherapy and NIR-PIT in vivo, xenograft models co-injected TE4 cells with CAFs were treated with 5-FU plus NIR-PIT, 5-FU alone, or PBS as control. 5-FU alone therapy could not suppress tumor growth, however combination therapy as 5-FU plus NIR-PIT could suppress tumor growth compared to control significantly. These results demonstrated that CAFs gave human esophageal cancer cells higher malignant and resistant phenotype, which could be overcome by using NIR-PIT targeting CAFs. Targeting fibroblast itself is a unique strategy and can be clinically promising as combination targeting cancer cells and their fundamental microenvironment, CAFs. Citation Format: Satoshi Komoto, Kazuhiro Noma, Ryoichi Katsube, Takuya Kato, Toshiaki Ohara, Hiroaki Sato, Toru Narusaka, Noriyuki Nisiwaki, Hiroshi Tazawa, Yasuhiro Shirakawa, Toshiyoshi Fujiwara. Overcoming resistance of conventional therapies by targeting cancer-associated fibroblasts (CAFs) with near-infrared photoimmunotherapy (NIR-PIT) [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr LB-310.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 13_Supplement ( 2017-07-01), p. 5934-5934
    Abstract: Background: As like rising of PD-1 antibody, targeting therapy to tumor immunosuppression have been shown dramatic cure responses in melanoma and other malignancies. However there are still problems that the responders of those checkpoint inhibitors are limited. Recently, it is considered that tumor immunosuppression may be caused by not only cancer cells but also tumor microenvironments (TME). Cancer-associated fibroblasts (CAFs) are one of the major components and have a central role for tumor progression in TME. It has been hypothesized that they can also play an essential role in tumor immunosuppression. In this study, we evaluated the correlation between CAFs and tumor immunity system, especially tumor-infiltrating lymphocytes (TILs) in clinical samples and further in vivo study. Materials and methods: Total 140 cases of esophageal cancer were evaluated for the presence of CAFs and TILs by immunohistochemistry (IHC). CAFs were defined as fibroblasts expressing alpha smooth muscle actin. CD8+ cytotoxic T lymphocytes and FoxP3+ regulatory T cells were investigated as TILs in each specimen’s intratumoral and peritumoral tissues. The outcome was set as the correlation between CAFs and TILs, and Overall survival. In vivo experiments, we used a mouse-derived cancer cell (colon26-Luc) and mouse embryonic fibroblasts (NIH/3T3). BALB/c or BALB/c-nu/nu mice were inoculated with only cancer cells (control) or cancer cells with CAFs (co-CAFs) into right flank to examine the immunosuppression affected by CAFs. The proliferation was examined by in vivo imaging system. Result: In clinical analysis, CD8+ TILs and CAFs demonstrated strong relations, which were significantly “negative” correlation and showed moderate correlation coefficient in intratumoral tissues (P & lt;0.001, r=-0.416). In FoxP3+ TILs, “positive” correlation was detected significantly (P & lt;0.001, r=0.484). In terms of prognosis, high group of CD8+ TILs had a good prognosis significantly (P & lt;0.001), whereas high group of FoxP3+ TILs had a poor prognosis significantly (P & lt;0.001) in intratumoral tissues. However, there was no difference of the correlation and the prognosis in peritumoral tissues. In vivo experiments demonstrated that promoted ratio of tumor progression by co-CAFs was more strongly demonstrated in BALB/c mice rather than BALB/c-nu/nu mice, suggesting that the group co-CAFs showed inducing immunosuppression in TME. IHC analysis of those tumors showed that accumulation of CD8+ TILs was decreased in tumors of co-CAFs as compared with tumors of control. On the other hand, accumulation of FoxP3+ TIL was increased, showing the same trends as clinical analysis. Conclusion: Our results suggest that Both CD8+ and FoxP3+ TILs in intratumoral tissues are independent prognostic factors, and CAFs have a significant correlation to those TILs. We consider that CAFs may affect tumor immunosuppression by regulating the migration of TILs. Citation Format: Takuya Kato, Kazuhiro Noma, Hajime Kashima, Yuki Katsura, Hiroaki Sato, Takayuki Ninomiya, Toshiaki Ohara, Yasuhiro Shirakawa, Toshiyoshi Fujiwara. Cancer-associated fibroblasts contribute to tumor immunosuppression by regulating tumor-infiltrating lymphocytes [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 5934. doi:10.1158/1538-7445.AM2017-5934
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2017
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 5066-5066
    Abstract: Background: Esophageal cancer has been reported to express HER family, especially EGFR and HER2, of which expressed cases have been reported to show poor prognosis. However, although there are some clinical trials that target those markers, using cetuximab (Cet), panitumumab (Pan), and trasutuzumab (Tra), the result is not sufficient. On the other hand, cancer-associated fibroblasts (CAFs) are one of the major components of tumor microenvironment and suggested to be associated with those chemoprevention, leading to poor prognosis. Thus, to target both cancer cells and CAFs, we focused on near-infrared photoimmunotherapy (NIR-PIT). NIR-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. We developed the CAFs-targeting PIT targeted on fibroblast activated protein (FAP) and have confirmed the efficacy to FAP-expressed CAFs. In this study, we hypothesized that the effect of cancer-targeting PIT is decreased under CAF-rich condition as like other antibody-based therapy. Here we firstly evaluated the effect of cancer-targeting PIT in esophageal cancer cells and furthermore the possibility of additional effect by “dual-targeting PIT” for CAFs and cancer cells. Materials and Methods: Human esophageal cancer cell lines and human esophageal fibroblasts (FEF3) were used in this study. FEF3 stimulated with conditioned medium (CM) of cancer cells defined as CAFs. We used cetuximab and panitumumab for anti-EGFR antibody and trastuzumab for anti-HER2 antibody. The expression of EGFR, HER2 in esophageal cancer cells was confirmed by Western blot (WB). Cell viability for quantitative evaluation was determined using an XTT Cell Proliferation Kit II. Result: WB demonstrated that HER2 expression was increased in TE4 and OE19, and EGFR was increased in TE8. FEF3 stimulated with CM was increased FAP expression compared with control. XTT assay demonstrated that the effect of cancer-targeting PIT using Pan-IR700, Tra-IR700 is efficient. Additionally, we confirmed that CAFs-targeting PIT worked in co-culture condition with cancer cells as well as monoculture condition. Furthermore, “dual-targeting PIT” showed rapid cell death of both in co-culture condition, and XTT assay showed the additive effects in vitro, as expected. Discussion: In this study, dual-targeting therapy has shown cellular specific cell death by targeting to cell-specific antigen and induced both cells death simultaneously. NIR-PIT can be novel effective therapy for esophageal cancer, and also for CAFs in tumor microenvironment. Thus, “dual-targeting PIT," which is combined with both mAb-IR700 for cancer cells and CAFs, can regulate tumor stroma as well as cancer cells simultaneously and can be highly expected to have strong antitumor effect by remodeling tumor microenvironment. Citation Format: Hiroaki Sato, Kazuhiro Noma, Toru Narusaka, Satoshi Komoto, Yuki Katsura, Takuya Kato, Takayuki Ninomiya, Toshiaki Ohara, Hiroshi Tazawa, Yasuhiro Shirakawa, Hisataka Kobayashi, Toshiyoshi Fujiwara. Dual-targeting photoimmunotherapy (NIR-PIT) for esophageal cancer cells and cancer-associated fibroblasts (CAFs) [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 5066.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. LB-045-LB-045
    Abstract: Background: Cancer stem cells (CSCs) have been proposed to be responsible for tumor initiating, drug resistance, metastasis, and recurrence. Although CSCs are considered to be important targets therapeutically, clinically effective CSC targeting strategies have not yet been established. Iron plays an important role in critical cellular processes, such as energy production and DNA synthesis. Although adequate iron levels are essential for human health, iron overload is known to cause some types of cancer. CSCs are considered to be involved in the tumor regardless of early stage. Thus we have focused on the relationship between iron and CSCs. We previously reported that the expression of stemness markers was suppressed by iron chelator in miPS and miPS inducible CSC model cells (miPS-LLCcm). Here, we hypothesized that the expression of stemness markers and function were also suppressed by iron chelator in high malignant cancer cells with stemness potential. Method: We used 4 stemness markers expressing cancer cell lines (HSC-2: Oral squamous cancer, TE-4 and OE33: Esophageal cancer, and NT-2: Embryonal cell carcinoma). Deferasirox (DFX), an oral iron chelator, was used. The stemness markers expression including Nanog, Sox2, Oct3/4, Klf-4 and c-Myc was examined by western blot analysis. Self-renewal ability was examined by sphere formation assay. Anti-proliferative effect of DFX was assessed combined with CDDP using HSC-2 cells. Result: DFX suppressed the expression of stemness markers and sphere formation ability in 4 cancer cell lines. In contrast, CDDP could not suppressed them. In HSC-2 cells, combination therapy with CDDP and DFX synergistically suppressed the proliferation with suppression of stemness markers. Conclusion: Our results indicated that iron depletion generally suppressed the stemness markers and functions of high malignant cells same as miPS and miPS-LLCcm. Thus iron depletion can be a novel therapy for CSCs via suppression of CSCs function. Citation Format: Toshiaki Ohara, Yuki Katsura, Kazuhiro Noma, Toru Narusaka, Takuya Kato, Hiroaki Sato, Satoshi Komoto, Yasuko Tomono, Takayuki Ninomiya, Hiroshi Tazawa, Shunsuke Kagawa, Yasuhiro Shirakawa, Toshiyoshi Fujiwara. Iron depletion suppress the stemness markers and functions of cancer stem cells [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr LB-045.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 13_Supplement ( 2017-07-01), p. 925-925
    Abstract: Background: Iron plays a crucial role in the various metabolic pathways and it is essential for life. However excess of iron is known to cause cancer. There are many reports that iron depletion treatment indicates antitumor effect. Previously, we have confirmed that iron depletion treatment indicates antitumor effect against Cancer stem cell (CSC) model, miPS-LLCcm, which was converted from murine induced pluripotent stem cell (miPS cell) in Okayama university (Chen at el. PLOS ONE 2012), and also suppresses CSC markers. Many tumors contain phenotypically and functionally heterogenous cancer cells due to pluripotency of CSC. Although there are various theories about the origin of CSC, miPS has a pluripotency and regarded as the top of the hierarchy, it can be the candidate of the origin of CSC. miPS cell is known to form teratoma when inoculated into nude mouse because of its undifferentiated status. We hypothesized if we could suppress the stemness and tumorigenicity of miPS cell by iron depletion treatment, it can be applied to the therapy for CSC and undifferentiated status cancer cell. In this study, we investigated the stemness and tumorigenecity of miPS cell with iron depletion treatment. Methods: We used miPS cell(cell name : iPS-MEF-Ng-20D-17) purchased from Riken Cell Bank(Japan). Deferasirox (DFX), an popular commercial available oral iron chelator was used for iron depletion treatment. Western blot analysis was done to examine the expression of the stemness markers including Nanog, Oct3/4, Sox2, Klf4, c-Myc. Subcutaneous model of miPS cell and miPS cell treated with DFX (50μM) were used in vivo study. Tumors were harvested for immunohistochemistry. Results: Stemness markers of miPS cell were suppressed strongly by DFX in western blot analysis. DFX also suppressed the subcutaneous tumor growth. The average tumor volume of the control group was 665.3±430.8mm3 while that of the DFX treated group was 204.3±76.2 mm3(p & lt;0.05) on day 20. Immunohistochemistry of the tumor revealed the suppression of the stemness markers in the DFX treated group compared with the control group. Citation Format: Yuki Katsura, Toshiaki Ohara, Hajime Kashima, Hiroaki Sato, Takuya Kato, Takayuki Ninomiya, Kazuhiro Noma, Yasuko Tomono, Hiroshi Tazawa, Shunsuke Kagawa, Yasuhiro Shirakawa, Toshiyoshi Fujiwara. Tumorigenesis of murine iPS cell is prevented by iron depletion with downregulation of stemness markers [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 925. doi:10.1158/1538-7445.AM2017-925
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2017
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...