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)  (19)
  • 1
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 15_Supplement ( 2015-08-01), p. 1673-1673
    Abstract: Introduction: The TGF-β pathway that has been associated with hepatocellular carcinoma (HCC) progression, can be targeted by galunisertib, a selective ATP-mimetic TGF-β receptor (TβR)-I inhibitor currently under clinical investigation in HCC patients after sorafenib failure or ineligible for sorafenib treatment. Our study aimed to investigate the tumor effects of galunisertib with or without sorafenib in different models of HCC and in freshly grown HCC specimens from patients. Material and methods: Transgenic mice developing stage-defined HCC were treated for 12 weeks (W) with either vehicle, sorafenib (30mg/kg), galunisertib (100mg/kg) or sorafenib plus galunisertib. Human hepatocarcinoma HUH7 cells were subcutaneously injected in nude mice and treated daily with either vehicle, sorafenib (60mg/kg) or sorafenib combined with galunisertib (100mg/kg). Ex vivo experiments were realized in surgical specimens from HCC patients that were cut into 300μM and grown in specific media. Slices were randomly exposed to galunisertib (10μM), sorafenib (5μM) and sorafenib plus galunisertib for 48h. At the end of treatments, tumor samples were analyzed by IF or IHC. Results: In transgenic mice, the number of liver tumor macronodules was significantly lower in all treatment arms compared to control placebo (p3 fold decrease in proliferation (Ki67 expression) and 3-5 fold increase in apoptosis induction (caspase-3 expression). Conclusion: In vivo and ex vivo, TGF-β signalling inhibition using galunisertib in combination with sorafenib in HCC models showed promising data on downstream signaling pathway, angiogenesis and tumor growth inhibition and may prevent sorafenib-induced EMT. Citation Format: Annemilai Tijeras-Raballand, Maria Serova, Cindy Neuzillet, Miguel Albuquerque, Nathalie Colnot, Matthias Barral, Anthony Dohan, Matthias Barral, Philippe Bonnin, Marc Pocard, Valérie Paradis, Eric Raymond, Sandrine Faivre, Armand de Gramont. Galunisertib combined with sorafenib affects proliferation, EMT and delays tumor growth in mice and in hepatocellular carcinoma (HCC) patient samples ex vivo. [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 1673. doi:10.1158/1538-7445.AM2015-1673
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 13, No. 18 ( 2007-09-15), p. 5331-5340
    Abstract: Purposes: Intestinal complications after radiotherapy are caused by transmural fibrosis and impair the quality of life of cancer survivors. Radiation fibrosis was considered permanent and irreversible, but recently, its dynamic nature was shown, providing new opportunities for the development of antifibrotic therapies. Among these new targets, we identified the Rho/ROCK pathway and thought to investigate whether pravastatin treatment inhibits Rho pathway activation and elicits an antifibrotic action. Experimental Design: Rho and ROCK activities were monitored in human explants presenting radiation fibrosis remodeling after incubation with pravastatin. Subsequent modulation of CCN2, type I collagen, and fibronectin expression were assessed ex vivo and in intestinal smooth muscle cells derived from radiation enteropathy. Then, the therapeutic relevance of the antifibrotic action of pravastatin was explored in vivo in a rat model of chronic radiation fibrosis (19 Gy X-rays) treated with 30 mg/kg/d pravastatin in the drinking water. Results: The results obtained with human explants show that pravastatin specifically inhibits Rho activity in submucosal mesenchymal cells. Pravastatin also elicits ROCK inhibition, and subsequent CCN2 production in human explants and smooth muscle cells isolated from radiation enteropathy. Inhibition of type I collagen and fibronectin does occur, showing that pravastatin modulates the secretory phenotype of mesenchymal cells. Lastly, curative pravastatin administration improves radiation enteropathy in rats. This structural improvement is associated with decreased deposition of CCN2 and subsequent decreased extracellular matrix deposition. Conclusion: Targeting established fibrosis with pravastatin is an efficient and safe antifibrotic strategy in radiation-induced enteropathy, and is easily transferable into the clinic.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2007
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 13_Supplement ( 2017-07-01), p. 4044-4044
    Abstract: Introduction: Notch pathway has been involved in cell fate determination, cell differentiation, proliferation and death. Notch promotes cell survival, angiogenesis and treatment resistance in numerous cancers, making it a promising target for cancer therapy. The role of the Notch pathways in HCC tumorigenesis has shown some controversies over time and across studies, being endowed with both oncogenic and tumor suppressive properties. Our study aimed at investigating the anti-tumoral effects of combining LY3039478, a novel Notch inhibitor, to sorafenib in an in vivo transgenic model of HCC. Methods: Transgenic mice developing stage-defined HCC were treated for 8 weeks (W) from W8 to W16 with either vehicle, LY3039478 (8mg/kg, thrice weekly, oral gavage), sorafenib (30mg/kg, daily, oral gavage) or LY3039478 plus sorafenib. Tumor growth was evaluated by ultrasound (liver size), by the number of macronodules, and the number of micronodules on HPS sections at sacrifice. Angiogenesis was evaluated by doppler (blood flow in the coeliac trunk) and by CD31 staining. Results: Liver size and the number of liver tumor macronodules were significantly lower in all treatment arms compared to control placebo at both the W12 intermediary sacrifice and W16 final sacrifice; the combination of LY3039478 and sorafenib showing increased tumor control at W16 (4,39±0,82 in mean liver volumes (in mm3) in the combination arm vs 5,41±1,0, 5,84±0,47, 7,09±1,5 in the sorafenib, LY3039478 and placebo arms respectively). Angiogenesis assessed by measuring the mean blood flow in the coeliac trunk (TCm), decreased in all treatment arms, compared to placebo. At W16, LY3039478 potentiated the effect of sorafenib, with a TCm decrease of 36% compared to 23% and18% the sorafenib and LY3039478 arms, respectively. These results will be confirmed by the assessment of micronodules number on HPS section for tumor growth and CD31 staining for angiogenesis analysis (number of vessels and the vessel lumen area). The effects of the combination versus monotherapies will be evaluated on the immune landscape. Conclusion: The combination of LY3039478 and sorafenib showed promising anti-tumor activities that were associated with decreased angiogenesis. Citation Format: Annemilai Tijeras-Raballand, Christian Hobeika, Elise Payen, Matthieu Martinet, Philippe Bonnin, Karim A. Benhadji, Bharvin R. Patel, Clarisse Eveno, Marc Pocard, Sandrine Faivre, Eric Raymond, Armand de Gramont. LY3039579, a novel Notch inhibit, potentiates the anti-tumoral effects of sorafenib in hepatocellular carcinoma (HCC) [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 4044. doi:10.1158/1538-7445.AM2017-4044
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 4096-4096
    Abstract: Background: Hepatocellular carcinoma (HCC) is a complex multistep malignancy in need of new therapeutic options often arising on underlying chronic liver disease. HCC animal models relevant to clinical situations are crucial to investigate new anticancer drugs, alone or in combination. Mice bearing xenografts and transgenic mice are the two main models used for preclinical drug development, but most of them fail to mimic the different step of HCC observed in patients. In this study, we aim to describe a stage-defined, transgenic immunocompetent HCC mouse model. Methods: ASV-B is a transgenic mouse model that spontaneously develops, upon SV40 T-Ag oncogene expression in hepatocytes, a reproducible stage-defined HCC. Hyperplasia at week(W)8 is followed by nodular stage at W12 (multiple well-delimited tumor nodules of about 1 mm, growing progressively up to 1 cm), then diffuse carcinoma stage at W16-20. HCC is restricted to male, backcrossed with C57BL/6J mice, the female littermates being used as controls. Results: Liver volume assessed by ultrasound at each step of carcinogenesis showed a 2.7-, 2.7-, 3.9-, and 5.8-fold increase (p & lt;0.001) in transgenic mice compared to controls (CTRL) at W8, 12, 16, and 20, respectively. ASV-B model displays marked arterialization with intense arterial flow in liver tumor similar to human HCC. Sinusoids in tumor nodules are tortuous and dilated, surrounded by activated hepatic stellate cells (HSCs) expressing smooth muscle actin (SMA) while HSCs surrounding normal sinusoids only express desmin. In addition, ASV-B livers develop along carcinogenesis a mesenchymal phenotype (increased vimentin and decreased e-cadherin staining), mimicking certain clinical situations of acquired resistance to VEGFR inhibitors. Angiogenesis, monitored using Doppler assessing blood flow velocity (BFV) in the hepatic artery, is enhanced by 29%, 51% and 156% at W8, W12 and W16, respectively, in transgenic liver animals as compared to CTRL. Increased angiogenesis was confirmed by CD31 staining in tumor specimen showing increased number of vessels per field. ASV-B model has been used to investigate the toxicity and efficacy of new drugs (galunisertib, MET/AXL inhibitor), including antiangiogenic agents (sorafenib and ramucirumab) alone or in combination, and may be relevant to explore the effects of immunotherapy agents. At the conference, we will display further characterization of liver tumors regarding vascularization, and immune cells localization using IHC. Conclusion: ASV-B transgenic mouse model mimics several characteristics of human HCC developing on healthy liver including multinodular disease, vessel abnormalities, and dedifferentiation toward mesenchymal phenotype. Further steps will consist of characterizing the adjacent nontumor liver, and developing nonalcoolic steatohepatitis (NASH) on ASV-B model using specific diets. Citation Format: Annemilaï Tijeras-Raballand, Patricia Hainaud, Christian Hobeika, Clarisse Eveno, Marc Pocard, Philippe Bonnin, Valérie Paradis, Mohamed Bouattour, Eric Raymond, Armand de Gramont, Evelyne Dupuy, Sandrine Faivre. Stage-defined, transgenic immunocompetent mouse model (ASV-B) to investigate new drugs for hepatocellular carcinoma [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 4096.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 15_Supplement ( 2015-08-01), p. 3564-3564
    Abstract: Background: Hepatocellular carcinoma (HCC), is characterized by severe vessel anomalies with an intense arterial blood supply, and acquisition of a basal membrane rich in laminin by sinusoids. A role of the VEGF-A/Dll4-Notch4/ephrin B2 cascade in tumor vessel anomalies has been demonstrated, raising the question of the nature of the VEGF receptor pathway implicated in triggering this cascade. This study was undertaken to determinate the role of PlGF and its receptor VEGFR-1 in the development of vascular anomalies in HCC and to evaluate the role of the downstream Notch4 pathway. Material and Methods: In vitro, we used primary culture of HUVECs, isolated in our laboratory from human umbilical cords. HUVECs were cultured in EBM2 medium, with 20% of FBS and 2ng/ml of FGF-2. The expression of the active form of Notch4 and Dll4 in HUVECs were assessed by Western Blot. In vivo, the differential expression of VEGF-A, PlGF and their receptors were evaluated by qRT-PCR and immunostaining in transgenic mice developing stage-defined HCC. Results: In vitro, to determine which growth factor/receptor combination was predominant in activating the Notch4 pathway, we compared the effects of VEGF-A, VEGF-E and PlGF on Dll4 expression and Notch4 activation in HUVECs. Treatment with all 3 growth factors increased Dll4 and active Notch4 expressions whose levels were significantly greater in PlGF than VEGF-A and VEGF-E treated HUVECs. Moreover, downregulation of VEGFR-1 by si RNA, but not VEGFR-2, abrogated the effects of VEGF-A and PlGF stimulation on Dll4 expression and Notch4 activation. PlGF silencing significantly reduced Dll4 dependent expression and Notch4 activation in response to VEGF-E and VEGF-A. To confirm in vivo the prevalence of the PlGF/VEGFR-1 pathway in induction of vessel anomalies, we evaluated the differential expression of PlGF, VEGF-A, VEGFR-1 and VEGFR-2 in our transgenic HCC model at different stages. VEGFR-1 expression is increased with HCC progression and restricted in macrophages and sinusoidal endothelial cells. Moreover, PlGF levels were maximal at the stage that coincides with the beginning of liver vessels anomalies in HCC. Silencing PlGF or blocking Notch4 activation, using a γ-secretase inhibitor (DAPT), in vivo delayed tumor growth, reduced arterial vessel length, and sinusoids anomalies with a decrease in Dll4 and active Notch4 expression levels without affecting microvascular density or normal liver. Conclusions: Together, these data suggest that the activation of PlGF-VEGFR-1/ Dll4- Notch4/ephrin B2 cascades played a role in vessel remodeling and tumor growth. Inhibitor of γ-secretase or silencing PlGF in vivo reduced activation of Notch4, prevent tumor vessels remodelling and arterialization, leading to a delay in tumor growth. These results would argue that targeting PlGF or active Notch4 may be valuable new anti-angiogenic strategies in the treatment of HCC. Citation Format: Annemilai Tijeras-Raballand, Jean-Olivier Contreres, Patricia Hainaud-Hakim, Carole Le Hénaff, Marc Pocard, Evelyne Dupuy, Armand de Gramont. PlGF/VEGFR-1-dependent activation of the Dll4-Notch4/Ephrin B2 cascade contributes to liver vessel anomalies in hepatocellular carcinoma. [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 3564. doi:10.1158/1538-7445.AM2015-3564
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 17, No. 1_Supplement ( 2018-01-01), p. B145-B145
    Abstract: Introduction: Galunisertib, a selective ATP-mimetic TGF-β receptor (TGFβR)-I inhibitor, displays a very safe toxicity profile in clinical trials, making it suitable for combinations. Galunisertib showed activity as second-line treatment in advanced HCC patients with high baseline AFP & gt; 200 ng/mL. Patients experiencing & gt;20% reduction in AFP level during treatment with galunisertib presented an outstanding median overall survival of 21.4 months (Faivre et al., ASCO GI 2014). Poor-prognosis HCC patients require innovative combinations to improve outcomes observed with antiangiogenic therapies. DC101 is a rat antagonist antibody to mouse VEGFR-2, and is used to model VEGFR-2 blockade in murine model. Both therapies have been shown to be independently beneficial to some subpopulation of HCC patients. Our study aimed at exploring the antitumoral potency of a combination of galunisertib and DC101 in an in vivo transgenic mouse model of HCC. Materials and Methods: Transgenic mice developing stage-defined HCC were treated for 8 weeks (W) from W8 to W16 with either vehicle, DC101 (40mg/kg, twice weekly, IP), galunisertib (100mg/kg, daily, oral gavage), or DC101 plus galunisertib. Tumor growth was evaluated by ultrasound (liver size) and by the number of macronodules at sacrifice. Angiogenesis was evaluated by Doppler (blood flow in the coeliac trunk) and by CD31 staining. Results: First, we confirmed the mode of action of galunisertib by assessing pSMAD2 expression, the pharmacodynamics target of galunisertib, before and after treatment. An 87% decrease in pSMAD2 expression was observed in liver tumors treated by galunisertib compared to placebo or DC101. Liver size and the number of liver tumor macronodules were significantly lower in all treatment arms compared to placebo control at both the W12 intermediary sacrifice and W16 final sacrifice; the combination of galunisertib and DC101 improved tumor control at W16 in the combination arm (4.43 mm3±0,55 mean liver volume) vs in the DC101, galunisertib, and placebo arms (5.22±0,86, 5.68±0,78, 7.09±1,54 respectively). Tumor growth inhibition was confirmed at W16 by a 31% and 52% decrease in the number of macronodules compared to placebo, in the galunisertib- and DC101-treated mice, respectively. This inhibition reached 62% in the combination group compared to placebo. Moreover, on hematoxylin phloxine saffron (HPS) section, no decrease in the number of micronodules was observed, whereas the size of these micronodules was dramatically reduced, especially in the combination arm with a 72% decrease compared to placebo. Angiogenesis, assessed by measuring the mean blood flow in the coeliac trunk (TCm), decreased in all treatment arms compared to placebo. At W16, galunisertib potentiated the effect of DC101 with a TCm decrease of 66% compared to 59% and 10% in the DC101 and galunisertib arms, respectively. CD31 confirmed the antiangiogenic effects of galunisertib and DC101, with a 25% and 51% decrease in vessel number, respectively, compared to placebo. These effects on angiogenesis were potentiated in the combination arm with an 80% decrease compared to placebo. Conclusion: The combination of galunisertib and DC101 showed promising antitumor activities that were associated with decreased angiogenesis. Citation Format: Annemilai Tijeras-Raballand, Christian Hobeika, Matthieu Martinet, Elise Paven, Philippe Bonin, Clarisse Eveno, Marc Pocard, Mohamed Bouattour, Sandrine Faivre, Eric Raymond, Armand de Gramont. Addition of galunisertib to DC101 improved angiogenesis inhibition and tumor growth control in hepatocellular carcinoma (HCC) [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2017 Oct 26-30; Philadelphia, PA. Philadelphia (PA): AACR; Mol Cancer Ther 2018;17(1 Suppl):Abstract nr B145.
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. 19-19
    Abstract: Introduction: Hepatocellular carcinoma (HCC), is characterized by severe vessel anomalies with an intense arterial blood supply (arterialisation), and acquisition ofa basal membrane rich in laminin by sinusoids(sinusoid capillarisation). A role of the VEGF-A/Dll4-Notch4/ephrin B2 cascade in tumor vessel anomalies has been demonstrated, raising the question of the nature of theVEGF receptor pathway implicated in triggering this cascade. This study was aimed at determining whether PlGF/VEGFR-1 or VEGF-A/VEGFR-2 pathways were implicated in the vessel anomalies observed during HCC development. Material and Methods:In vitro, we used primary culture of HUVECs, isolated in our laboratory from human umbilical cords. HUVECs were cultures in EBM2 medium, complemented with 20% of FBS and 2ng/ml of FGF-2. To determine which pathway was predominant in the activation of the Dll4/Notch4 cascade, the effects of PlGF (a specific ligand of VEGFR-1), VEGF-E (a specific ligand of VEGFR-2) and VEGF-A (a ligand of both receptors) on the expression of the active form of Notch4 and Dll4 in HUVECs were compared by Western Blot. In vivo, () the differential expression of VEGF-A, PlGF and their receptors were evaluated by qRT-PCR and immunostainings in transgenic mice developing stage-defined HCC. Results: In vitro, HUVECs stimulation by either VEGF-A, VEGF-E or PlGF increased Dll4 expression and Notch 4 activation. However, PlGF had the most important effect with a 5-fold increase of Dll4 expression level and a 4-fold increase in Notch 4 activation, compared to control HUVECs. Moreover, downregulation of VEGFR-1 by small interfering RNA, but not VEGFR-2, abrogated the effects of VEGF-A and PlGF stimulation on Dll4 expression and Notch4 activation. To confirm in vivothe prevalence of the PlGF/VEGFR-1 pathway in induction of vessel anomalies, we first evaluated the differential expression of PlGF, VEGF-A, VEGFR-1 and VEGFR-2 in our transgenic HCC model at different stages of the disease. We showed that VEGFR-1 increased with HCC progression and was expressed by macrophages and sinusoidal endothelial cells. Moreover, PlGF levels were maximal at the stage that coincides with the beginning of liver vessels anomalies in HCC. Silencing PlGF in vivo delayed tumor growth (p & lt;0.05), reduced arterial vessel length by 84% (p & lt;0.05), and sinusoids anomalies with a decrease in Dll4 and active Notch4 expression levels without affecting microvascular density. Conclusions: We demonstrated that PlGF and VEGFR-1 played a key role in the development of vessel anomalies in HCC and that silencing PlGF prevents liver vessel anomalies and delays tumor growth. Our results bring a new insight in the mechanism responsible for vessel anomalies observed in HCC, giving a strong rationale for the development of new antiangiogenic strategies based on PlGF inhibition in HCC. Citation Format: Annemilai Tijeras-Raballand, Armand de Gramont, Patricia Hainaud, Jean-Olivier Contreres, Carole Le Hénaff, Marc Pocard, Evelyne Dupuy. PlGF/VEGFR-1 dependent activation of the Dll4-Notch4/Ephrin B2 cascade contributes to liver vessel anomalies in hepatocellular carcinoma. [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 19. doi:10.1158/1538-7445.AM2014-19
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. 1636-1636
    Abstract: Background: Non-alcoholic fatty liver disease (NAFLD), especially non-alcoholic steatohepatitis (NASH) is a chronic liver disease commonly associated with hepatic fibrosis. NASH patients have an increased risk for hepatocellular carcinoma (HCC). Due to western way of life, NASH incidence is rising and predicted to become the leading cause of liver transplant in 2020 and of HCC in the next decades. There is an urgent need for robust animal models fully recapitulating the NASH-related HCC carcinogenesis due to this changing in HCC etiology. In this study, we develop and characterize specific diet-induced variants from our transgenic HCC mouse model, focusing on immune landscape. Methods: We used in the whole study ASV-B mice: a transgenic mouse model (C57BL/6J) spontaneously developing a reproducible stage-defined HCC (hyperplasia at week(W)8, nodular stage at W12, and diffuse carcinoma at W16-20). Livers were characterized for angiogenesis and immune populations using immunostaining (IHC and IF), and qRT-PCR (LightCycler, Roche). To mimic NASH, ASV-B mice were exposed to 5 different diets. Ten ASV-B and 5 control mice were fed as follows: classic diet as control (yellow), or a high-fat diet (blue), a diet enriched with saturated fatty acids + 1.25% cholesterol (green), a diet containing 22% of vegetal oil + 0.2% cholesterol (orange), and a 1.25% cholesterol diet containing 21% of milkfat (red). All mice fed with special diets also received 30% fructose in the drink water. A second experiment was performed on ASV-B and C57BL/6J wild type mice, using control and Nash-inducing regimen, to confirm our first results. Results: ASV-B model showed an increase in liver volume and angiogenesis, HCC livers harboring marked arterialization and capillarization as compared to control. Assessing immune markers on 7 evaluable tumor specimens, we observed an increase in CD8, Foxp3, INOS, CD11b, PD-1, PD-L1, IL1β, IFN-γ, TNF-α, IL17A and IL17F mRNA expressions, as frequently observed in human inflammatory HCC. In addition, IHC staining showed intratumoral infiltration of lymphocytes (CD8+) and macrophages (F4/80+, a well-characterized and extensively referenced mouse macrophage marker). ASV-B mice receiving yellow, blue, and green regimens showed similar liver volumes and weights. By macroscopic analysis, we observed increased liver steatosis, and fibrosis in the red and orange regimen compared to others. Moreover, we observed a 40% mortality rate in the orange regimen, and a 20% mortality rate in the blue and green regimens. Interestingly, by microscopic analysis, we observed liver steatosis and inflammation, in 100%, and 75% of the mice, respectively. These results indicate that the green regimen is the most suitable to induce NASH-underlyning disease in our transgenic HCC model. At the conference, we will show in the diet-variants, the immune landscape of the livers and the results of our ongoing second experiment. Conclusion: ASV-B transgenic mouse model mimics several characteristics of human HCC developing on healthy liver including inflammatory reaction and immune cell infiltration. In the ASV-B model, we have been able to develop specific-diets variants mimicking NASH characteristics that could be used for drug testing. Citation Format: Annemilaï Tijeras-Raballand, Christian Hobeika, Philippe Bonnin, Benoit Rousseau, Aurélie Rodrigues, Fouad Ladfil, Marc Pocard, Armand de Gramont, Eric Raymond, Sandrine Faivre, Valérie Paradis, Clarisse Eveno. Diet-variants and immune characterization of a stage-defined, transgenic immunocompetent mouse model of HCC (ASV-B) [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 1636.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 11, No. 17 ( 2005-09-01), p. 6212-6217
    Abstract: Purpose: The aim of our study was to assess whether the polymorphism of the nucleotide excision repair enzyme, excision repair cross-complementing rodent repair deficiency, complementation group 1 (ERCC1), had an effect on the tumor response in patients treated with standard chemotherapy regimens for a metastatic colorectal cancer. We have studied the synonymous polymorphism that causes a single nucleotide change C to T at codon 118 converting a codon of common usage (AAC) to a less used codon (AAT), both coding asparagine. This change results in a decreased ERCC1 gene expression, which impairs repair activity. Experimental Design: Ninety-one patients with a median age of 55.1 years treated for a metastatic colorectal cancer were included in our retrospective study. The ERCC1 polymorphism was analyzed in the normal tissue of all patients. Results: Twenty (22%) were homozygous for AAC codon (C/C genotype), 30 were (33%) homozygous for AAT codon (T/T genotype), and 41 (45%) were heterozygous (C/T genotype). The objective response rate to oxaliplatin in combination with 5-fluorouracil (5-FU) was significantly higher in the T/T genotype group compared with the C/T and the C/C genotype groups (61.9%, 42.3%, and 21.4%, respectively; P = 0.018). By contrast, no significant difference was observed when patients were treated with either 5-FU alone (45%, 29.2%, and 33.3%, respectively; P = 0.407) or in combination with irinotecan (46.1%, 25.0%, and 27.3%, respectively; P = 0.305). Conclusions: Our observations allowed us to define the first useful predictive criterion for oxaliplatin/5-FU response in patients with metastatic colorectal cancer.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2005
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 67, No. 1 ( 2007-01-01), p. 398-407
    Abstract: Obtaining representative human colon cancer cell lines from fresh tumors is technically difficult. Using 32 tumor fragments from patients with colon cancer, the present study shows that prior xenograft leads to more efficient cell line establishment compared with direct establishment from fresh tumors (P & lt; 0.05). From 26 tumor specimens, we successfully established 20 tumor xenografts in nude mice (77%); among 19 of these xenografts, 9 (47%) led to cell lines, including four from liver metastases. Only 3 of 31 tumor specimens (9.7%) grew immediately in vitro, and all were derived from primary tumors. To compare major phenotypic and genotypic characteristics of human colon cancer cell lines derived from the same tumor fragment using two protocols, the two pairs of cell lines obtained from 2 of 32 tumor fragments were extensively studied. They displayed similar morphology and were able to form compact spheroids. Chemosensitivity to 5-fluorouracil, CPT11, and L-OHP differed between cell lines obtained from patient tumors and those derived from xenografts. Matched cell lines shared a common core of karyotype alterations and distinctive additional chromosomal aberrations. Expression levels of genes selected for their role in oncogenesis evaluated by real-time quantitative PCR were found to be statistically correlated whatever the in vitro culture model used. In conclusion, xenotransplantation in mice of tumor fragments before establishment of cell lines enables generation of more novel human cancer cell lines for investigation of colon cancer cell biology, opening up the opportunity of reproducing the diversity of this disease. [Cancer Res 2007;67(1):398–407]
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2007
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages