96 research outputs found

    Limitation and challenges in using pancreatic cancer-derived organoids as a preclinical tool

    Get PDF
    Pancreatic ductal adenocarcinoma (PDAC) is a dismaldisease with a fast evolution and unpredictable treatmentresponse. Nowadays, FOLFIRINOX and gemcitabine are the preferred treatments with a response rateof 33% and 11%, respectively. This poor patient responsehas been associated with an inefficient/non-personalizedtreatment allocation. Consequently, developing a rapidand efficient preclinical tool to test tumor drug sensitivityfor each patient is hugely needed. Biopsy patient-derivedorganoid (PDO) appears to be a promising tool for devel-oping individualized treatments for patients with PDAC.Several PDO-based platforms are in development world-wide as a guide to optimize therapy by directing tailored treatments.Fil: Fraunhoffer Navarro, Nicolas Alejandro. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Houssay. Centro de Estudios Farmacológicos y Botánicos. Universidad de Buenos Aires. Facultad de Medicina. Centro de Estudios Farmacológicos y Botánicos; Argentina. Aix-Marseille University; FranciaFil: Abuelafia, Analía Meilerman. Aix-Marseille University; FranciaFil: Dusetti, Nelson. Aix-Marseille University; FranciaFil: Iovanna, Juan Lucio. Aix-Marseille University; Franci

    Pancreatic Cancer Heterogeneity Can Be Explained Beyond the Genome

    Get PDF
    Pancreatic ductal adenocarcinoma (PDAC) remains a major health problem because it induces almost systematic mortality. Carcinogenesis begins with genetic aberrations which trigger epigenetic modifications. While genetic mutations initiate tumorigenesis, they are unable to explain the vast heterogeneity observed among PDAC patients. Instead, epigenetic changes drive transcriptomic alterations that can regulate the malignant phenotype. The contribution of factors from the environment and tumor microenvironment defines different epigenetic landscapes that outline two clinical subtypes: basal, with the worst prognosis, and classical. The epigenetic nature of PDAC, as a reversible phenomenon, encouraged several studies to test epidrugs. However, these drugs lack specificity and although there are epigenetic patterns shared by all PDAC tumors, there are others that are specific to each subtype. Molecular characterization of the epigenetic mechanisms underlying PDAC heterogeneity could be an invaluable tool to predict personalized therapies, stratify patients and search for novel therapies with more specific phenotype-based targets. Novel therapeutic strategies using current anticancer compounds or existing drugs used in other pathologies, alone or in combination, could be used to kill tumor cells or convert aggressive tumors into a more benign phenotype

    Inhibition of glucuronidation in pancreatic cancer improves gemcitabine anticancer activity

    Get PDF
    Pancreatic ductal adenocarcinoma (PDAC) treatmentis focused on two regimens. The polychemotherapy, FOLFIRINOX (folinic acid, fluorouracil, irinotecan, oxali-platin), is used in patients with good health conditions, while gemcitabine, as monotherapy, in patients withpoor health conditions. Gemcitabine resistance-associated pathways have been targeted to sensitize cancercells, but the results were disappointing. Using a transcrip-tomic bioinformatics analysis combined with biologicalvalidation, we showed that glucuronidation was associated with the gemcitabine resistance in PDAC, and its inhibition could switch tumors from resistant to sensitive.To unravel the biological drivers of gemcitabineresponse in PDAC, we determined the transcriptomic dissimilarity between two preclinical models with definedgemcitabine sensitivity.Fil: Fraunhoffer Navarro, Nicolas Alejandro. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Houssay. Centro de Estudios Farmacológicos y Botánicos. Universidad de Buenos Aires. Facultad de Medicina. Centro de Estudios Farmacológicos y Botánicos; ArgentinaFil: Meilerman Abuelafia, Analía. Inserm; FranciaFil: Chanez, Brice. Inserm; FranciaFil: Bigonnet, Martin. Inserm; FranciaFil: Gayet, Odile. Inserm; FranciaFil: Roques, Julie. Inserm; FranciaFil: Chuluyan, Hector Eduardo. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Houssay. Centro de Estudios Farmacológicos y Botánicos. Universidad de Buenos Aires. Facultad de Medicina. Centro de Estudios Farmacológicos y Botánicos; ArgentinaFil: Dusetti, Nelson. Inserm; FranciaFil: Iovanna, Juan Lucio. Inserm; Franci

    Novel role of VMP1 as modifier of the pancreatic tumor cell response to chemotherapeutic drugs

    Get PDF
    We hypothesized that inhibiting molecules that mediate the adaptation response to cellular stress can antagonize the resistance of pancreatic cancer cells to chemotherapeutic drugs. Toward this end, here, we investigated how VMP1, a stress-induced autophagy-associated protein, modulate stress responses triggered by chemotherapeutic agents in PDAC. We find that VMP1 is particularly over-expressed in poorly differentiated human pancreatic cancer. Pharmacological studies show that drugs that work, in part, via the endoplasmic reticulum stress response, induce VMP1 expression. Similarly, VMP1 is induced by known endoplasmic reticulum stress activators. Genetic inactivation of VMP1 using RNAi-based antagonize the pancreatic cancer stress response to antitumoral agents. Functionally, we find that VMP1 regulates both autophagy and chemotherapeutic resistance even in the presence of chloroquin, ATG5 or Beclin 1 siRNAs, or a Beclin 1-binding VMP1 mutant. In addition, VMP1 modulates endoplasmic reticulum stress independently of its coupling to the molecular and cellular autophagy machinery. Preclinical studies demonstrate that xenografts expressing an inducible and tractable form of VMP1 show increased resistance to the gemcitabine treatment. These results underscore a novel role for VMP1 as a potential therapeutic target for combinatorial therapies aimed at sensitizing pancreatic cancer cells to chemotherapeutic agents as well as provide novel molecular mechanisms to better understand this phenomenon.Fil: Gilabert, Mariana. Cancer Research Center of Marseille; Francia. Aix-Marseille University; Francia. Centre National de la Recherche Scientifique; FranciaFil: Vaccaro, Maria Ines. Universidad de Buenos Aires. Facultad de Farmacia y Bioquímica; Argentina. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Houssay. Instituto de Bioquímica y Medicina Molecular; ArgentinaFil: Fernandez Zapico, Martín E.. Mayo Clinic Cancer Center; Estados UnidosFil: Calvo, Ezequiel L.. Molecular Endocrinology and Oncology Research Center; CanadáFil: Turrini, Olivier. Cancer Research Center of Marseille; Francia. Aix-Marseille University; Francia. Centre National de la Recherche Scientifique; FranciaFil: Secq, Véronique. Cancer Research Center of Marseille; Francia. Aix-Marseille University; Francia. Centre National de la Recherche Scientifique; FranciaFil: Garcia, Stéphanie. Cancer Research Center of Marseille; Francia. Aix-Marseille University; Francia. Centre National de la Recherche Scientifique; FranciaFil: Moutardier, Vincent. Cancer Research Center of Marseille; Francia. Aix-Marseille University; Francia. Centre National de la Recherche Scientifique; FranciaFil: Lomberk, Gwen. Mayo Clinic; Estados UnidosFil: Dusetti, Nelson. Cancer Research Center of Marseille; Francia. Aix-Marseille University; Francia. Centre National de la Recherche Scientifique; FranciaFil: Urrutia, Raul. Mayo Clinic; Estados UnidosFil: Iovanna, Juan L.. Cancer Research Center of Marseille; Francia. Aix-Marseille University; Francia. Centre National de la Recherche Scientifique; Franci

    Evidencing a pancreatic ductal adenocarcinoma subpopulation sensitive to the proteasome inhibitor Carfilzomib

    Get PDF
    Purpose: Pancreatic ductal adenocarcinoma (PDAC) is a lethal cancer with a survival rate less than 5%. Multiple chemotherapeutic drugs have been tested to improve patient prognosis; however, the clinical efficacy of these treatments is low. One of the most controversial family of drugs are the proteasome inhibitors, which have displayed promising effects in preclinical studies, but low clinical performance. Here, we unravel a specific transcriptomic signature that discriminates a subgroup of patients sensitive to the proteasome inhibitor carfilzomib. Experimental Design: First, we identified a subpopulation of PDAC-derived primary cells cultures (PDPCC) sensitive to the proteasome inhibitor carfilzomib. Then, we selected a transcriptomic signature that predicts carfilzomib chemosensitivity using independent component analysis on the transcriptome of PDPCC. Finally, we validated the signature in an independent cohort of PDAC biopsy-derived pancreatic organoids. Results: Sensitive phenotype was characterized by a high expression of genes related with a cornified/squamous pathway and a downregulation of epithelial-mesenchymal transition genes. Interestingly, carfilzomib-sensitive transcriptomic profile did not show any association with the proteasome activity but strongly correlates with ATF4 and CHOP expression, which are key markers of the unfolded protein response and critical to trigger the cell death program. Concordantly, sensitive phenotype showed a high level of the de novo RNA and protein synthesis compared with the resistant one and, most important, cell death induced by carfilzomib is dependent of the translational activity. Conclusions: We demonstrate the existence of a carfilzomib-sensitive PDAC subgroup with a specific transcriptomic phenotype that could explain the biological reason for this responsiveness.Fil: Fraunhoffer Navarro, Nicolas Alejandro. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Houssay. Centro de Estudios Farmacológicos y Botánicos. Universidad de Buenos Aires. Facultad de Medicina. Centro de Estudios Farmacológicos y Botánicos; Argentina. Inserm; FranciaFil: Meilerman Abuelafia, Miriam Analia. Inserm; FranciaFil: Bigonnet, Martin. Inserm; FranciaFil: Gayet, Odile. Inserm; FranciaFil: Roque, Julie. Inserm; FranciaFil: Telle, Emmanuel. Inserm; FranciaFil: Santofimia-Castaño, Patricia. Inserm; FranciaFil: Borrello, Maria Teresa. Inserm; FranciaFil: Chuluyan, Hector Eduardo. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Houssay. Centro de Estudios Farmacológicos y Botánicos. Universidad de Buenos Aires. Facultad de Medicina. Centro de Estudios Farmacológicos y Botánicos; ArgentinaFil: Dusetti, Nelson. Inserm; FranciaFil: Iovanna, Juan Lucio. Inserm; Franci

    Exploring the complementarity of pancreatic ductal adenocarcinoma preclinical models

    Get PDF
    Purpose: Compare pancreatic ductal adenocarcinoma (PDAC), preclinical models, by their transcriptome and drug response landscapes to evaluate their complementarity. Experimental De-sign: Three paired PDAC preclinical models—patient‐derived xenografts (PDX), xenograft‐derived pancreatic organoids (XDPO) and xenograft‐derived primary cell cultures (XDPCC)—were derived from 20 patients and analyzed at the transcriptomic and chemosensitivity level. Transcriptomic characterization was performed using the basal‐like/classical subtyping and the PDAC molecular gradient (PAMG). Chemosensitivity for gemcitabine, irinotecan, 5‐fluorouracil and oxaliplatin was established and the associated biological pathways were determined using independent component analysis (ICA) on the transcriptome of each model. The selection criteria used to identify the different components was the chemosensitivity score (CSS) found for each drug in each model. Results: PDX was the most dispersed model whereas XDPO and XDPCC were mainly classical and basal-like, respectively. Chemosensitivity scoring determines that PDX and XDPO display a positive correlation for three out of four drugs tested, whereas PDX and XDPCC did not correlate. No match was observed for each tumor chemosensitivity in the different models. Finally, pathway analysis shows a significant association between PDX and XDPO for the chemosensitivity‐associated pathways and PDX and XDPCC for the chemoresistance‐associated pathways. Conclusions: Each PDAC preclinical model possesses a unique basal‐like/classical transcriptomic phenotype that strongly in-fluences their global chemosensitivity. Each preclinical model is imperfect but complementary, sug-gesting that a more representative approach of the clinical reality could be obtained by combining them. Translational Relevance: The identification of molecular signatures that underpin drug sensitivity to chemotherapy in PDAC remains clinically challenging. Importantly, the vast majority of studies using preclinical in vivo and in vitro models fail when transferred to patients in a clinical setting despite initially promising results. This study presents for the first time a comparison between three preclinical models directly derived from the same patients. We show that their applica-bility to preclinical studies should be considered with a complementary focus, avoiding tumor-based direct extrapolations, which might generate misleading conclusions and consequently the overlook of clinically relevant features.Fil: Hoare, Owen. Centre National de la Recherche Scientifique; FranciaFil: Fraunhoffer Navarro, Nicolas Alejandro. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Houssay. Centro de Estudios Farmacológicos y Botánicos. Universidad de Buenos Aires. Facultad de Medicina. Centro de Estudios Farmacológicos y Botánicos; ArgentinaFil: Elkaoutari, Abdessamad. Centre National de la Recherche Scientifique; FranciaFil: Gayet, Odile. Centre National de la Recherche Scientifique; FranciaFil: Bigonnet, Martin. Centre National de la Recherche Scientifique; FranciaFil: Roques, Julie. Centre National de la Recherche Scientifique; FranciaFil: Nicolle, Rémy. No especifíca;Fil: McGuckin, Colin. Cell Therapy Research Institute; FranciaFil: Forraz, Nico. Cell Therapy Research Institute; FranciaFil: Sohier, Emilie. Le Centre Régional de Lutte Contre Le Cancer Léon Bérard; FranciaFil: Tonon, Laurie. Le Centre Régional de Lutte Contre Le Cancer Léon Bérard; FranciaFil: Wajda, Pauline. Le Centre Régional de Lutte Contre Le Cancer Léon Bérard; FranciaFil: Boyault, Sandrine. Le Centre Régional de Lutte Contre Le Cancer Léon Bérard; FranciaFil: Attignon, Valéry. Le Centre Régional de Lutte Contre Le Cancer Léon Bérard; FranciaFil: Tabone, Luciana Belen. Le Centre Régional de Lutte Contre Le Cancer Léon Bérard; FranciaFil: Barbier, Sandrine. No especifíca;Fil: Mignard, Caroline. No especifíca;Fil: Duchamp, Olivier. No especifíca;Fil: Iovanna, Juan. Centre National de la Recherche Scientifique; FranciaFil: Dusetti, Nelson J.. Centre National de la Recherche Scientifique; Franci

    Gene expression profiling of patient‐derived pancreatic cancer xenografts predicts sensitivity to the BET bromodomain inhibitor JQ1: implications for individualized medicine efforts

    Get PDF
    Abstract c‐MYC controls more than 15% of genes responsible for proliferation, differentiation, and cellular metabolism in pancreatic as well as other cancers making this transcription factor a prime target for treating patients. The transcriptome of 55 patient‐derived xenografts show that 30% of them share an exacerbated expression profile of MYC transcriptional targets (MYC‐high). This cohort is characterized by a high level of Ki67 staining, a lower differentiation state, and a shorter survival time compared to the MYC‐low subgroup. To define classifier expression signature, we selected a group of 10 MYC target transcripts which expression is increased in the MYC‐high group and six transcripts increased in the MYC‐low group. We validated the ability of these markers panel to identify MYC‐high patient‐derived xenografts from both: discovery and validation cohorts as well as primary cell cultures from the same patients. We then showed that cells from MYC‐high patients are more sensitive to JQ1 treatment compared to MYC‐low cells, in monolayer, 3D cultured spheroids and in vivo xenografted tumors, due to cell cycle arrest followed by apoptosis. Therefore, these results provide new markers and potentially novel therapeutic modalities for distinct subgroups of pancreatic tumors and may find application to the future management of these patients within the setting of individualized medicine clinics

    CDK2 and PKA Mediated-Sequential Phosphorylation Is Critical for p19INK4d Function in the DNA Damage Response

    Get PDF
    DNA damage triggers a phosphorylation-based signaling cascade known as the DNA damage response. p19INK4d, a member of the INK4 family of CDK4/6 inhibitors, has been reported to participate in the DNA damage response promoting DNA repair and cell survival. Here, we provide mechanistic insight into the activation mechanism of p19INK4d linked to the response to DNA damage. Results showed that p19INK4d becomes phosphorylated following UV radiation, β-amyloid peptide and cisplatin treatments. ATM-Chk2/ATR-Chk1 signaling pathways were found to be differentially involved in p19INK4d phosphorylation depending on the type of DNA damage. Two sequential phosphorylation events at serine 76 and threonine 141 were identified using p19INK4d single-point mutants in metabolic labeling assays with 32P-orthophosphate. CDK2 and PKA were found to participate in p19INK4d phosphorylation process and that they would mediate serine 76 and threonine 141 modifications respectively. Nuclear translocation of p19INK4d induced by DNA damage was shown to be dependent on serine 76 phosphorylation. Most importantly, both phosphorylation sites were found to be crucial for p19INK4d function in DNA repair and cell survival. In contrast, serine 76 and threonine 141 were dispensable for CDK4/6 inhibition highlighting the independence of p19INK4d functions, in agreement with our previous findings. These results constitute the first description of the activation mechanism of p19INK4d in response to genotoxic stress and demonstrate the functional relevance of this activation following DNA damage

    Puesta en evidencia y caracterización de una nueva familia multigénica de proteínas asociadas a la pancreatitis : análisis de su expresión y relación evolutiva

    No full text
    La "Pancreatitis Associated Protein" (PAP) es una proteína pancreática secretoria, estructuralmente relacionada a las lectinas y secretada durante la pancreatitis aguda por las células acinares pancreáticas. Su expresión muy débil en el páncreas sano, aumenta fuertemente (más de 400 veces) durante la fase aguda de la pancreatitis. En este trabajo de tesis, se puso en evidencia la expresión constitutiva de un transcripto idéntico en el intestino. Se mostró tambien que la PAP proveniente del intestino posee las mismas modificaciones post traduccionales que su homólogo pancreático. Finalmente se estudió su expresión en diferentes situaciones fisiológicas. En el curso de estos trabajos aparecieron una serie de indicios que sugirieron la presencia de otras moléculas estructuralmente emparentadas. La búsqueda de éstas, finalizó con el clonado de los ADNc correspondientes a las PAP II y III. El estudio de la expresión, reveló que la PAP III es indistinguible de la PAP I tanto en su expresión intestinal como pancreática, mientras que la PAP II es específica del páncreas. En busca de las secuencias promotoras responsables de la expresión de estas moléculas, clonamos y secuenciamos los genes que codifican para las 3 PAP y para la Lithostathina en la rata, y la PAP I humana. El análisis de su organización, mostró que todos estos genes, pertenecen a un nuevo grupo de lectinas de tipo C. Esta observación nos llevó a postular un camino evolutivo alternativo para el CRD "Carbohydrate Recognition Domain" característico de este grupo de lectinas. Por otra parte, los genes de rata mostraron una organización intrón-exón muy similar y una localización cromosómica idéntica, sugiriendo un origen común por duplicación génica, a partir de un mismo gen ancestral. Los trabajos realizados sobre el promotor de la PAP I, muestran que la región responsable de la especificidad de tejido se encuentra en las primeras 180 pares de bases. En esta región se encontró un represor capaz de "silenciar" la expresión de este gen en las líneas celulares Rat2 e IEC6 (no pancreáticas). En esta misma región se encuentra un elemento de regulación positiva que sólo es activo en la línea celular pancreática AR-42J. Los primeros indicios de la caracterización de los factores nucleares responsables de estas actividades, muestran la existencia de proteínas expresadas especificamente en cada línea celular, las que serian responsables de otorgar la especificidad de tejido al gen de la PAP I. Finalmente, los trabajos realizados sobre la inducción de la expresión del ARNm de la PAP I en la línea celular pancreática AR-42J, muestran la presencia de factores capaces de inducir específicamente este gen, en el suero de ratas afectadas de pancreatitis aguda. El ARNm de la PAP I se reveló también inducible por algunos de los mediadores generales de las proteínas de estrés celular (IL6, dexametasona, alcohol y agua oxigenada).Fil:Dusetti, Nelson. Universidad de Buenos Aires. Facultad de Ciencias Exactas y Naturales; Argentina
    corecore