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)  (54)
Material
Publisher
  • American Association for Cancer Research (AACR)  (54)
Language
Years
Subjects(RVK)
  • 1
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2014
    In:  Cancer Research Vol. 74, No. 19_Supplement ( 2014-10-01), p. 167-167
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. 167-167
    Abstract: The tumor microenvironment consists of a number of distinct cell types including tumor cells, fibroblasts, endothelial cells, immune cells, and adipocytes, and produces a wide variety of factors, which contribute to tumor progression. We previously observed that high-fat diet (HFD) feeding stimulates solid tumor growth and lymph node (LN) metastasis in mice subcutaneously injected with B16F10 melanoma cells. We also observed that HFD feeding increases the number of lipid vacuoles and M2-macrophage (M2-Mϕ)s as well as the expression of various cytokines/growth factors in tumor tissues. In the present study, we noted that HFD increased the expression of CCR7 in the tumor. The concentrations of CCL19 and CCL21, the CCR7 ligands, were significantly higher in the LN than those in tumor, and HFD feeding increased the concentrations of these ligands in the LN with negligible changes in the tumor tissue creating higher concentration gradients of these two ligands between the LN and tumor. In order to explore the interactions between tumor cells, adipocytes, lymphatic endothelial cell (LEC)s, and/or M2-Mϕs, we established in vitro co-culture models involving 3 distinct cell types. The mRNA levels of cytokines and growth factors were increased in B16F10 cells and M2-Mϕs when B16F10 cells, M2-Mϕs, and/or mature adipocyte (MA)s were co-cultured in a transwell system. Among these cytokines/growth factors, the mRNA levels of MCP-1 were synergistically increased in B16F10 cells when co-cultured with M2-Mϕs and MAs. The migration of monocytes was tremendously elevated when media conditioned by B16F10 cells/M2-Mϕs/MAs co-cultures was used as a chemoattractant. Furthermore, MAs increased the expression of CCR7 mRNA in B16F10 cells and the expression of CCL19 and CCL21 mRNAs in LECs. These results indicate that in HFD-fed mice, adipocytes induce MCP-1 expression and monocyte infiltration which play important roles in solid tumor growth and LN metastasis. Additionally, adipocyte-stimulated activation of CCL19, CCL21/CCR7 axis in lymphatic endothelial cells and tumor cells helps tumor cells to enter the lymphatic vessels and increased ligand concentration gradients between the LN and tumor facilitate tumor cell migration to the LN. Citation Format: Han Jin Cho, Jae In Jung, Yoo Jin Jung, Ki Won Lee, Mi-Kyung Sung, Jung Han Yoon Park. Roles of adipocytes and M2-macrophages in high-fat diet-stimulated lymph node metastasis of B16F10 melanoma. [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 167. doi:10.1158/1538-7445.AM2014-167
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2014
    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 ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 15_Supplement ( 2015-08-01), p. 911-911
    Abstract: Epidemiological studies indicate that obesity-associated metabolic stress increases the risk of developing several cancers. We previously reported that a high-fat diet (HFD)-induced obesity stimulates tumor growth and metastasis of B16F10 melanoma cancer cells in C57BL/6 mice. OL, the most abundant phenolic compound in olives, was reported to exert several physiological activities, including antioxidative, anticancer, and antimicrobial activities. In the present study, we examined whether OL inhibits HFD-induced melanoma progression in the B16F10 allograft model. Four-week old, male C57BL/6N mice were fed a diet [control diet (10 kcal% fat), HFD (60 kcal% fat), HFD + 0.02% OL, or HFD + 0.04% OL]. Twelve weeks after the initiation of feeding, fasting blood glucose, plasma insulin, and HOMA-IR were increased in HFD-fed mice, which was suppressed by dietary OL. Dual-energy X-ray absorptiometry results revealed that the proportion of lean body mass was decreased, whereas that of fat mass was increased in HFD-fed mice, and these changes were suppressed by dietary OL. After 16 weeks of feeding, B16F10-luc cells were subcutaneously injected into the animals’ left flank; bioluminescence imaging was conducted; and the primary tumor was resected 3 weeks later. OL suppressed HFD-induced solid tumor growth; HFD-induced increases in the expression of Ki67, cyclin D1, VEGF-A, VEGF-C, VEGF-D, CD31 (platelet endothelial cell adhesion molecule-1) and LYVE (lymphatic vessel endothelial receptor)-1; and HFD-induced decreases in TUNEL-positive apoptotic cells and cleaved PARP levels in the tumor tissues. OL also suppressed HFD-induced changes in the levels of many cytokines/chemokines in tumor tissues. All animals were killed 3 weeks after the tumor resection. OL suppressed HFD-induced increases in the size of LN; LN metastasis; and expression of VEGF-A and VEGF-C in the LN. OL suppresses HFD-induced increases in the size of adipocytes and accumulation of F4/80+ macrophages in adipose tissues surrounding the LN. In vitro culture results revealed that OL inhibited lipid accumulation in 3T3-L1 preadipocytes and the migration and viability of B16F10 cells. The present results demonstrated that dietary OL suppresses tumor growth and LN metastasis in HFD-fed mice. These effects are probably mediated, at least in part, via the inhibition of lipid accumulation, thereby reducing obesity-associated metabolic stress. Additionally, in vitro results indicate that OL may directly inhibit melanoma cell growth and migration, which may be one of the mechanisms by which dietary OL suppress melanoma progression in these mice. Citation Format: Hyerim Song, Jae In Jung, Gyoo Taik Kwon, So Young Park, Han Jin Cho, Jung Han Yoon Park. Dietary oleuropein (OL) improves high-fat diet-induced metabolic stress and suppresses solid tumor growth and lymph node (LN) metastasis in the B16F10 melanoma allograft model. [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 911. doi:10.1158/1538-7445.AM2015-911
    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 ...
  • 3
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2013
    In:  Cancer Research Vol. 73, No. 8_Supplement ( 2013-04-15), p. 1424-1424
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 8_Supplement ( 2013-04-15), p. 1424-1424
    Abstract: Tumor-associated macrophages, which are derived from the infiltration of circulating bone marrow-derived monocytes, consist primarily of a polarized M2 macrophage population and are associated with poor prognosis in various cancers. We previously observed that solid tumor growth and lung metastasis were enhanced when 4T1 mammary carcinoma cells were injected with M2 macrophages into the mammary fat pad of BALB/c mice. Cancer cell proliferation and macrophage infiltration were markedly increased when M2 macrophages were co-injected with 4T1 cells. In the present study, we explored the mechanisms underlying the crosstalk between 4T1 cells and M2 macrophages in tumor microenvironment. The results of in vitro experiments revealed that the migration of monocytes and the secretion of many cytokines including macrophage-colony stimulating factor (M-CSF) and monocyte chemotactic protein-1 (MCP-1) were increased when 4T1 cells were co-cultured with M2 macrophages, as compared to when the 4T1 cells were cultured alone. The mRNA expression of M-CSF and MCP-1 increased in 4T1 cells when 4T1 cells were co-cultured with M2 macrophages. To investigate the potential roles of M-CSF and MCP-1 in tumor growth and metastasis, 4T1 cells were infected with M-CSF, MCP-1, or control shRNA lentivirus particles and selected with puromycin. 4T1 cells infected with M-CSF shRNA did not grow or metastasize to the lung when injected into the mammary fat pads of BALB/c mice. Inhibition of MCP-1 expression in tumor cells also suppressed 4T1 tumor growth and metastasis, however, the degree of inhibition was much smaller than that of M-CSF inhibition. M-CSF knockdown decreased M2 macrophage (mouse mannose receptor+) population, whereas MCP-1 knockdown decreased the number of both M1 and M2 macrophages in tumor tissues. Taken together, these results indicate that M-CSF and MCP-1 are produced by the crosstalk between 4T1 cells and M2 macrophages, and these cytokines play important roles in mammary tumor progression. Citation Format: Han Jin Cho, Jung Han Yoon Park. Inhibition of M-CSF or MCP-1 expression in tumor cells reduces tumor growth and lung metastasis in a murine mammary cancer model. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 1424. doi:10.1158/1538-7445.AM2013-1424
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2013
    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
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2010
    In:  Cancer Research Vol. 70, No. 8_Supplement ( 2010-04-15), p. 2876-2876
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 70, No. 8_Supplement ( 2010-04-15), p. 2876-2876
    Abstract: 3,3’-Diindolymethane (DIM) is a major in vivo derivative of indole-3-carbinol, which is present in cruciferous vegetables and has been reported to evidence anti-carcinogenic properties. In the present study, we attempted to determine whether DIM inhibits the development of prostate cancer using the transgenic adenocarcinoma mouse prostate (TRAMP) model. DIM feeding inhibited prostate carcinogenesis in TRAMP mice, reduced the number of cells expressing the SV40 large tumor antigen and proliferating cell nuclear antigen, and increased the number of terminal dUTP nick-end labeling (TUNEL)-positive cells in the dorsolateral lobes of the prostate. Additionally, DIM feeding reduced the expression of cyclin A, cyclin-dependent kinase (CDK)2, and CDK4, and increased Bax expression. To assess the mechanisms by which DIM induces apoptosis, LNCaP and DU145 human prostate cancer cells were cultured with various concentrations (0 – 30 μmol/L) of DIM. DIM induced a substantial reduction in the numbers of viable cells and induced apoptosis in LNCaP and DU145 cells. DIM increased the cleavage of caspase-9, −7, −3, and poly (ADP-ribose) polymerase (PARP). DIM increased mitochondrial membrane permeability and the translocation of cytochrome c and Smac/Diablo from the mitochondria. Additionally, DIM induced increases in the levels of cleaved caspase-8, truncated Bid, Fas, and Fas ligand, and the caspase-8 inhibitor z-IETD-FMK was shown to mitigate DIM-induced apoptosis and the cleavage of caspase-3, PARP, and Bid. These results indicate that DIM inhibits prostate carcinogenesis via induction of apoptosis and inhibition of cell cycle progression. DIM induces apoptosis in prostate cancer cells via the mitochondria- and death receptor-mediated pathways. Note: This abstract was not presented at the AACR 101st Annual Meeting 2010 because the presenter was unable to attend. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 2876.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2010
    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
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2012
    In:  Cancer Research Vol. 72, No. 8_Supplement ( 2012-04-15), p. 609-609
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 72, No. 8_Supplement ( 2012-04-15), p. 609-609
    Abstract: Benzyl isothiocyanate (BITC) is a hydrolysis product of glucotropaeolin, a compound found in cruciferous vegetables, and has also been shown to have anti-tumor properties. In the present study, we attempted to determine whether BITC inhibits the development of prostate cancer using the transgenic adenocarcinoma mouse prostate (TRAMP) model. Male TRAMP mice and their nontransgenic (normal) littermates at 5 wk of age were randomly divided into control and BITC-treatment groups and gavage-fed with 0 (vehicle), 5, or 10 mg/kg of BITC every other day. At the time of sacrifice (24 wk of age), BITC did not affect body weight of normal or transgenic animals. The genitourinary tract weight of TRAMP mice was increased markedly as compared to normal mice, and this increase was suppressed significantly via the oral administration of 5 or 10 mg/kg of BITC. H & E staining of the dorsolateral lobes of the prostate demonstrated that well-differentiated carcinoma (WDC) was a predominant feature in the 24 week old vehicle-fed TRAMP mice, whereas number of lobes with WDC was reduced and the number of lobes with prostatic intraepithelial neoplasia was increased by feeding with 5 or 10 mg/kg of BITC. BITC feeding reduced the number of cells expressing the Ki67 (a proliferation marker) in the prostatic tissue. Additionally, BITC feeding reduced the expression of cyclin E, cyclin D1, cyclin A, and cyclin-dependent kinase (CDK)2. Our in vitro cell culture results revealed that BITC decreased DNA synthesis and CDK2 activity in TRAMP-C2 mouse prostate cancer cells. However, BITC did not affect apoptosis either in transgenic mice in vivo or in TRAMP-C2 cells in vitro. These results indicate that inhibition of cell cycle progression contributes to the inhibition of prostate cancer development in animals treated with BITC. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 609. doi:1538-7445.AM2012-609
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2012
    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
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2011
    In:  Cancer Research Vol. 71, No. 8_Supplement ( 2011-04-15), p. 465-465
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 8_Supplement ( 2011-04-15), p. 465-465
    Abstract: Tumor microenvironment consists of a number of components including malignant cells, stromal cells, endothelial cells, infiltrating immune cells, and extracellular matrix. Because inflammation plays an important role in cancer development and progression, tumor-associated macrophages (TAMs) can be one of key regulators of tumor microenvironment. Recent evidence has shown that TAM is associated with poor prognosis in a variety of cancers, including breast, lung, and pancreatic cancer. TAMs account for up to 50% of the total tumor mass, resemble M2-macrophages, and exhibit a number of pro-tumoral functions, including the expression of growth factors and matrix proteases, the promotion of angiogenesis, and the suppression of adaptive immunity. In the present study, we examined the effect of M2 macrophages on the development of and metastasis of mammary cancer using a 4T1-orthotopic tumor model. Monocytes were isolated from mouse bone marrow and treated with IL-4 to induce differentiation into an M2 phenotype. In order to determine whether or not M2 microphages stimulate mammary cancer growth and progression, 4T1 mammary carcinoma cells were injected into the inguinal mammary fat pad of syngeneic BALB/C mice, either alone or coupled with bone marrow-derived alternatively-activated macrophages (BMDAM). Co-injection of 4T1 cells with BMDAM increased solid tumor volumes (at 2 weeks after inoculation) and lung metastasis (at 3 weeks after inoculation). We prepared 4T1-luc cells that stably express firefly luciferase which allows for direct examination of tumor growth in animals by estimating the luciferase activity of 4T1 cancer cells via an in vivo imaging system. Luminescence was quantified, and the results demonstrated that the accumulation of 4T1-luc cells was significantly more pronounced when 4T1 cells were co-injected with BMDAM. Immunohistochemical staining of tumor tissues revealed that the proportion of Ki67 (proliferation index)-positive cells was increased in tumor tissues when 4T1 cells were co-injected with BMDAM. We also observed that co-injection of BMDAM with 4T1 cells increased the expression of hypoxia-inducible factor-1α and CD31 (a glycoprotein expressed on endothelial cells and in platelets)-positive cells. These results indicate that TAM increases the proliferation of mammary tumor cells, which may lead to the stimulation of lung metastasis in the 4T1 mammary carcinoma model. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 465. doi:10.1158/1538-7445.AM2011-465
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2011
    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. 83, No. 7_Supplement ( 2023-04-04), p. 5107-5107
    Abstract: Background: The Aryl hydrocarbon receptor (AhR) is one of the most predominant regulators of cancer metabolism. The AhR exerts important immunosuppressive functions by activating Treg cells and myeloid-derived suppressor cells and repressing CD8+ effector T cells. Here, we propose that a best-in-class AhR inhibitor, DA-4505, improves anti-tumor efficacy via modulation of tumor immune surveillance compared to BAY2416964, an AHR antagonist drug candidate being studied in the clinical phase. Methods: To evaluate anti-tumor effects of DA-4505 and BAY2416964, the two AhR inhibitors were dosed at 10 mg/kg once daily alone or in combination with aPD-1 (10 mg/kg) in surgical and chemotherapy models, and a PDX model (YHIM2004). Tumor volume, relapse, and survival were evaluated, and immune profiles were analyzed with IHC, flow cytometry, and scRNAseq. Results: A significant tumor reduction appeared in the CT26 and 4T1 tumor models after the DA-4505 treatment compared to vehicle group (P & lt;0.05). In contrast, DA-4505 treatment did not induce significant tumor regression compared to vehicle group in tumor-bearing NOG mice, suggesting that anti-tumor effects of DA-4505 were driven by immunologic mechanisms. To evaluate the role of DA-4505 in conjunction with surgery, DA-4505 alone or in combination with anti-PD-1 was given prior to and following resection of the tumors in 4T1 tumor-bearing mice. Survival of mice treated with DA-4505 alone or DA-4505 combined with anti-PD-1 was significantly prolonged after resection compared to aPD-1 treatment group (P & lt;0.05). In addition, there were four mice that did not have a relapse by treating DA-4505 with or without aPD-1 after surgery (4/5). A tumor regression also appeared in the YHIM2004-engrafted humanized mouse study. A tumor reduction was shown by treating DA-4505 alone or in combination with pembrolizumab compared to vehicle group (P & lt;0.05). Next, we co-administered an AhR inhibitor and aPD-1 as a partner to improve the antitumor effects of chemotherapy. The DA-4505 add-on group showed tumor regression when compared with the combination therapy group treated with aPD-1 and chemotherapy (P & lt;0.0001). In addition, a significant increase in survival rate was shown in the group treated with a DA-4505 add-on compared to vehicle group (P & lt;0.001). Analysis of scRNAseq showed that M1 macrophage expressing CCL7 and CCL8 were increased in DA-4505 treated group compared to the vehicle and aPD-1 groups. This suggests that immune modulatory effect of DA-4505 may be due to enhanced recruitment of immune cells into the tumor site by macrophages with high chemotactic activity. Conclusion: The AhR inhibitor DA-4505 demonstrated an improvement in anti-tumor efficacy. In addition, it has shown a synergistic effect when combined with aPD-1. Discoveries from this study provide a preclinical rationale for future clinical implications in solid tumor. Citation Format: DongKwon Kim, Sujeong Baek, Seung Min Yang, Yu Jin Han, Seong-san Kang, Chun-Bong Synn, Mi Hyun Kim, Heekyung Han, Kwangmin Na, Young Taek Kim, Sungwoo Lee, Taedong Han, Hyounmie Doh, Jongho Cho, Dajeong Kim, Daewon Cha, Jae Hwan Kim, Youngseon Byeon, Young Seob Kim, Mi Ran Yun, Ji Yun Lee, Jii Bum Lee, Chang Gon Kim, Min Hee Hong, Sun Min Lim, Byoung Chul Cho, Kyoung-Ho Pyo. A novel AhR inhibitor ‘DA-4505’ improved the anti-cancer efficacy of surgical and chemotherapy via synergistic anti-tumor effects of aPD-1. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 5107.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    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. 83, No. 7_Supplement ( 2023-04-04), p. 3234-3234
    Abstract: Prostaglandin E2 (PGE2) is widely recognized as one of the major bioactive lipids that, with the striking regenerative potential, promote drug-resistance in cancer cells as well as immune evasion in the tumor microenvironment (TME). Primarily driven by apoptotic cell death, PGE2 is thought to elicit wound-healing responses to help provide an immunosuppressive and proliferative niche that supports cancer stem cell repopulation and thereby therapy-resistance. While COX1/2 inhibitors that attenuate PGE2 production have shown promising anti-cancer effects in various (pre-)clinical settings, the gastrointestinal- and cardiotoxicities precluded their development as anti-cancer agents. It is anticipated that specific targeting of PGE2 signaling via its cognate receptors constitutes a safer and potentially more effective approach. Of the receptor subtypes EP1-4, Gα,s-coupled EP2 and EP4 are believed to be directly involved in immunosuppressive effects of PGE2.OCT-598 is a novel, highly potent and selective EP2/EP4 dual antagonist with Ki values of 23 nM and 0.2 nM vs EP2 and EP4, respectively. PGE2 inhibited normal differentiation of human monocytes into CD1a+CD16- dendritic cells under the presence of GM-CSF and IL-4 and promoted differentiation towards CD1a-CD16+ macrophages in vitro. However, EP2/EP4 dual inhibition by OCT-598 reversed this phenomenon to a greater extent than either EP2- or EP4-specific inhibitor alone. In vivo, OCT-598 effected tumor growth inhibition in multiple syngeneic mouse models as a single agent as well as in combination with an immune checkpoint blocker (ICB). Furthermore, the addition of OCT-598 to the lung cancer standard-of-care regimen (anti-PD-1 plus chemotherapy) in TC-1 mouse lung adenocarcinoma model gave rise to complete tumor regression. In conclusion, dual blockade of EP2 and EP4 by OCT-598 is shown to be a compelling strategy to reinforce antitumor effects by thwarting PGE2-mediated therapy resistance and immune evasion.Findings from this study provide a rationale for clinical development of OCT-598 as a therapeutic option for human malignant cancers. Citation Format: Youngrae Lee, Sujeong Baek, Dong Kwon Kim, Yeri Lee, Donggeon Kim, Seongin Jo, Sang Kyun Lim, Young Sook Shin, Soonsang Kwon, Seung Min Yang, Young Taek Kim, Seong-San Kang, Chun-Bong Synn, Kwangmin Na, Mi Hyun Kim, Heekyung Han, Yu Jin Han, Sungwoo Lee, Jae Hwan Kim, Mi Ran Yun, Youngseon Byeon, Young Seob Kim, Ji Yun Lee, Jii Bum Lee, Chang Gon Kim, Min Hee Hong, Sun Min Lim, Kyoung-Ho Pyo, Byoung Chul Cho, Taeyoung Yoon. OCT-598, a novel EP2/EP4 dual antagonist, promotes anti-tumor immune responses in syngeneic mouse tumor models in combination with standard-of-care chemo- and immunotherapies [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 3234.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    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. 83, No. 7_Supplement ( 2023-04-04), p. 6780-6780
    Abstract: Heterogeneity in resistant to immunotherapies of tumor microenvironment (TME) has been implicated in immunotherapies to cause immune evasion or drug resistance. This study was conducted to explore the heterogeneity of TME through multiplex IHC, spatial and RNA sequencing analysis. We selected a sample from a lung adenocarcinoma patient without EGFR-activating mutation and expressing 30% of PD-L1. For quantitative analysis by multiplex IHC, various markers including CD4, CD8, FoxP3, granzyme B, CD20 and pan-cytokeratin were stained with 7 different fluorescence dyes, which was imaged with Vectra Polaris (Akoya). For scRNAseq and spatial RNAseq, we used 5’ chromium library kit (10X Genomics) to make library construction. Integrated raw data was generated using Cell ranger, Seurat pipeline and Azimuth package. The tumor area was divided into 16 clusters in which we selected 2 clusters based on CD3/45 expression. There was a noticeable distinction between the two clusters which were defined as the ‘High’ region (CD45highCD3high cluster) and the ‘Low’ region (CD45lowCD3low cluster). By multiplex IHC, percentage of CD8+T cells was higher in the ‘High’ region than in the ‘Low’ region (8.5% vs. 0.8%, respectively). Subsequent analysis of two clusters using spatial and single cell RNA seq, the ‘Low’ region was characterized by increased hypoxia-associated gene expressions including HIF1A, HIF3A and VEGFA. Various immune cells were abundant in the ‘High’ region and CD45 expression level was 11-fold higher in the ‘High’ region compared to the ‘Low’ region. Cytokine/chemokine network analysis via spatial RNAseq revealed that gene expression of tumor necrosis factor (TNF) family-associated factors increased in the 'High' region compared to the ‘Low’ region (TNF, FAS, TRAIL, RANKL and CD40), which is well-known to promotes apoptosis, programmed cell death, or necrosis of certain cancer. Additionally, the ‘High’ region also had elevated levels of the PD-1/PD-L1, CD155, CD122/TIGIT, Siglec10/CD24, LAG3/LAGLS3, and CD47/CD172a axes, suggesting active immune responses. Intriguingly, combined analyses showed that ‘High’ region showed enhanced level of CD44 expression as the leading-edged gene, which suggests the metastatic potential of tumor cells. Furthermore, scRNA analysis confirmed that CD44 expression was mainly higher in macrophages, suggesting that tumor-associated macrophages partially affected tumor cell metastasis in the ‘High’ region. Our finding suggests that understanding the intratumoral immunological heterogeneity of lung adenocarcinoma can help to study the mechanism of tumor heterogeneity by integrated spatial RNAseq and scRNAseq analyses. This type of technique could be applied to understand complex networks of anti-tumor immune activities, drug resistance mechanisms and immunotherapeutic response of cancer. Citation Format: Seul Lee, Jae-Hwan Kim, Kwangmin Na, Seung Min Yang, Dong Kwon Kim, Sujeong Baek, Seong-san Kang, Yu Jin Han, Chun-Bong Synn, Mi hyun Kim, Heekyung Han, Young Taek Kim, Sungwoo Lee, Youngseon Byeon, Young Seob Kim, Ji Yun Lee, Jii Bum Lee, Chang Gon Kim, Min Hee Hong, Sun Min Lim, Kyoung-Ho Pyo, Byoung Chul Cho. Characterization of immunological heterogeneity in the tumor microenvironment by integrated analyses using single cell RNAseq, spatial RNAseq and multiplex IHC [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 6780.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    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. 83, No. 7_Supplement ( 2023-04-04), p. 6433-6433
    Abstract: Backgrounds: Live biotherapeutic products (LBPs) emerged as potential therapeutics to overcome the limitation of ICIs. This research shows that CJRB-101, a novel bacterial strain, can improve anti-tumor effects in synergy with pembrolizumab in non-small cell lung cancer (NSCLC). Objectives and Methods: Tumors from NSCLC patients (anti-PD-1 refractory and resistant) were transplanted into Hu-CD34-NSG to establish humanized patient-derived xenograft (PDX) mice models. Five models (YHIM-2003, 2004, 2009, 2010 and 2014) were treated with CJRB-101 at low (5 × 107 CFU) or high (109 CFU) doses, or with pembrolizumab (10 mg/kg, i.p., Q5D) or in combination. Tumor growth inhibition (TGI) rate was measured. Tumor microenvironment (TME) was analyzed using multiplex IHC, flow cytometry and single cell RNA sequencing. Ex-vivo assays were performed to validate in silico findings. Results: Tumor in PDX models was unresponsive to pembrolizumab alone, however, in combination with CJRB-101 effectively suppressed tumor growth. The synergy was highlighted in YHIM-2009 where TGI was 10-fold higher (56%) than pembrolizumab group (5%). Immune profiling revealed that macrophages may be responsible for the anti-tumor effects of CJRB-101. IHC showed significantly increased antigen presenting specialized DCs (CD16+CD68−CD11c+) and granzyme B+ CD8+ T cells in the tumor by CJRB-101 compared to pembrolizumab (p & lt;0.01). This suggested that CJRB-101 induced infiltration of cytotoxic CD8 T cells into the tumor nest by enhancing antigen presenting machinery. Trajectory analysis showed that CJRB-101 induced repolarization of M2 to M1 macrophages, characterized by high expression of CXCL9/10. CXCL9+/10+ M1 macrophages were comparatively more abundant in the combination group (23.11%) than the pembrolizumab group (0.91%). CXCL9/CXCL10 expression in macrophages was higher in the CJRB-101 group compared to the pembrolizumab group (p & lt;0.0001). The combination group (10.84%) had a higher relative abundance of CD8+ T cells compared to the pembrolizumab group (1.58%) and higher IFNγ expression in CD8+ T cells compared to the pembrolizumab group (p=0.0152), suggesting that CJRB-101 repolarized macrophages and recruited active CD8+ T cells. Co-culture assays using bone marrow-derived macrophages validated that CJRB-101 drove differentiation towards F4/80+ or MHC II+ expressing M1 macrophage (p & lt;0.0001) and repolarized existing M2 (CD206+) to M1 (p=0.0002). Conclusion: Combination treatment of CJRB-101 with anti-PD-1 showed synergistic anti-tumor effects via repolarization of M2 to M1 macrophages, leading to activation of CD8+ T cells in TME. Especially, CXCL9+/10+ M1 macrophage playing a key role in TGI induced by CJRB-101 in NSCLC models. Findings from this study provided rationale for clinical investigation of CJRB-101. Citation Format: Arim Min, Chun-bong Synn, Seong-san Kang, Bo-eun Kwon, Junwon Yang, Hyunkyung Park, Jieun Im, Hyunjeong Kim, Sujeong Beak, Dong Kwon Kim, Jii Bum Lee, Hyeonseok Oh, Seung Min Yang, Yu Jin Han, Mi hyun Kim, Heekyung Han, Kwangmin Na, Young Taek Kim, Sungwoo Lee, Mi Ran Yun, Jae Hwan Kim, Youngseon Byeon, Young Seob Kim, Ji Yun Lee, Chang Gon Kim, Min Hee Hong, Sun Min Lim, Kyoung-Ho Pyo, Byoung Chul Cho. A novel bacterial strain, CJRB-101, induces anti-cancer effects by repolarization of M2 to CXCL9 and CXCL10 dual expressing M1 macrophages in humanized non-small cell lung cancer mice models [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 6433.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    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...