23 results on '"Némati, Fariba"'
Search Results
2. Multi-omics comparison of malignant and normal uveal melanocytes reveals molecular features of uveal melanoma
- Author
-
Gentien, David, Saberi-Ansari, Elnaz, Servant, Nicolas, Jolly, Ariane, de la Grange, Pierre, Némati, Fariba, Liot, Géraldine, Saule, Simon, Teissandier, Aurélie, Bourc’his, Deborah, Girard, Elodie, Wong, Jennifer, Masliah-Planchon, Julien, Narmanli, Erkan, Liu, Yuanlong, Torun, Emma, Goulancourt, Rebecca, Rodrigues, Manuel, Gaudé, Laure Villoing, Reyes, Cécile, Bazire, Matéo, Chenegros, Thomas, Henry, Emilie, Rapinat, Audrey, Bohec, Mylene, Baulande, Sylvain, M’kacher, Radhia, Jeandidier, Eric, Nicolas, André, Ciriello, Giovanni, Margueron, Raphael, Decaudin, Didier, Cassoux, Nathalie, Piperno-Neumann, Sophie, Stern, Marc-Henri, Gibcus, Johan Harmen, Dekker, Job, Heard, Edith, Roman-Roman, Sergio, and Waterfall, Joshua J.
- Published
- 2023
- Full Text
- View/download PDF
3. Ferrocifen stealth LNCs and conventional chemotherapy: A promising combination against multidrug-resistant ovarian adenocarcinoma
- Author
-
Idlas, Pierre, Ladaycia, Abdallah, Némati, Fariba, Lepeltier, Elise, Pigeon, Pascal, Jaouen, Gerard, Decaudin, Didier, and Passirani, Catherine
- Published
- 2022
- Full Text
- View/download PDF
4. Evaluation of Combined Chemotherapy and Genomic-Driven Targeted Therapy in Patient-Derived Xenografts Identifies New Therapeutic Approaches in Squamous Non-Small-Cell Lung Cancer Patients.
- Author
-
Decaudin, Didier, Némati, Fariba, Masliah Planchon, Julien, Seguin-Givelet, Agathe, Lefevre, Marine, Etienne, Vesnie, Ahnine, Harry, Peretti, Quentin, Sourd, Laura, El-Botty, Rania, Huguet, Lea, Lagha, Sarah, Hegarat, Nadia, Roman-Roman, Sergio, Bièche, Ivan, Girard, Nicolas, and Montaudon, Elodie
- Subjects
- *
THERAPEUTIC use of antineoplastic agents , *CELL metabolism , *ADENOCARCINOMA , *SQUAMOUS cell carcinoma , *GENOMICS , *ENZYME inhibitors , *CANCER patients , *XENOGRAFTS , *CELLULAR signal transduction , *IN vivo studies , *CANCER chemotherapy , *MICE , *DRUG efficacy , *ANIMAL experimentation , *GENETIC mutation , *LUNG cancer , *SEQUENCE analysis - Abstract
Simple Summary: Non-small-cell lung cancer is characterized by high morbidity and mortality. Currently, the precision medicine approach in the adenocarcinoma subtype of non-small-cell lung cancer aims to identify genomic alterations that can be targeted in individual patients and offer them appropriate treatment. Several biomarker-guided therapies have been approved, targeting genes frequently altered in adenocarcinoma, such as EGFR, BRAF, MET, ALK, ROS1, RET and NTRK. To overcome the emergence of resistance and increase the efficacy of these targeted therapies, the combination of chemotherapy and targeted therapy has been studied and validated in adenocarcinoma patients with EGFR mutations. This study proposes to examine whether this type of combined approach, which is difficult to study in clinical trials, could be more widely used by targeting other genetic alterations in the MAPK/PI3K pathways or against alterations in the CDNK2A gene in adenocarcinomas but also in squamous-cell carcinomas. The combination of chemotherapy and targeted therapy has been validated in non-small-cell lung cancer (NSCLC) patients with EGFR mutations. We therefore investigated whether this type of combined approach could be more widely used by targeting other genetic alterations present in NSCLC. PDXs were generated from patients with NSCLC adenocarcinomas (ADCs) and squamous-cell carcinomas (SCCs). Targeted NGS analyses identified various molecular abnormalities in the MAPK and PI3K pathways and in the cell cycle process in our PDX panel. The antitumor efficacy of targeted therapies alone or in combination with chemotherapy was then tested in vivo. We observed that trametinib, BKM120, AZD2014 and palbociclib increased the efficacy of each chemotherapy in SCC PDXs, in contrast to a non-insignificant or slight improvement in ADCs. Furthermore, we observed high efficacy of trametinib in KRAS-, HRAS- and NRAS-mutated tumors (ADCs and SCCs), suggesting that the MEK inhibitor may be useful in a wider population of NSCLC patients, not just those with KRAS-mutated ADCs. Our results suggest that the detection of pathogenic variants by NGS should be performed in all NSCLCs, and particularly in SCCs, to offer patients a more effective combination of chemotherapy and targeted therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
5. A high-risk retinoblastoma subtype with stemness features, dedifferentiated cone states and neuronal/ganglion cell gene expression
- Author
-
Liu, Jing, Ottaviani, Daniela, Sefta, Meriem, Desbrousses, Céline, Chapeaublanc, Elodie, Aschero, Rosario, Sirab, Nanor, Lubieniecki, Fabiana, Lamas, Gabriela, Tonon, Laurie, Dehainault, Catherine, Hua, Clément, Fréneaux, Paul, Reichman, Sacha, Karboul, Narjesse, Biton, Anne, Mirabal-Ortega, Liliana, Larcher, Magalie, Brulard, Céline, Arrufat, Sandrine, Nicolas, André, Elarouci, Nabila, Popova, Tatiana, Némati, Fariba, Decaudin, Didier, Gentien, David, Baulande, Sylvain, Mariani, Odette, Dufour, Florent, Guibert, Sylvain, Vallot, Céline, Rouic, Livia Lumbroso-Le, Matet, Alexandre, Desjardins, Laurence, Pascual-Pasto, Guillem, Suñol, Mariona, Catala-Mora, Jaume, Llano, Genoveva Correa, Couturier, Jérôme, Barillot, Emmanuel, Schaiquevich, Paula, Gauthier-Villars, Marion, Stoppa-Lyonnet, Dominique, Golmard, Lisa, Houdayer, Claude, Brisse, Hervé, Bernard-Pierrot, Isabelle, Letouzé, Eric, Viari, Alain, Saule, Simon, Sastre-Garau, Xavier, Doz, François, Carcaboso, Angel M., Cassoux, Nathalie, Pouponnot, Celio, Goureau, Olivier, Chantada, Guillermo, de Reyniès, Aurélien, Aerts, Isabelle, and Radvanyi, François
- Published
- 2021
- Full Text
- View/download PDF
6. Improvement of the anti-proliferative activity of the peptide ERα17p in MCF-7 breast cancer cells using nanodiamonds
- Author
-
Yip, François, Nemati, Fariba, El Botty, Rania, Belnou, Mathilde, Decaudin, Didier, Mansuy, Christelle, and Jacquot, Yves
- Published
- 2019
- Full Text
- View/download PDF
7. Targeting chronic lymphocytic leukemia with N-methylated thrombospondin-1–derived peptides overcomes drug resistance
- Author
-
Pramil, Elodie, Herbi Bastian, Linda, Denèfle, Thomas, Nemati, Fariba, Xiao, Malina, Lardé, Eva, Maloum, Karim, Roos-Weil, Damien, Chapiro, Elise, Le Garff-Tavernier, Magali, Davi, Frédéric, Decaudin, Didier, Sarfati, Marika, Nguyen-Khac, Florence, Merle-Béral, Hélène, Karoyan, Philippe, and Susin, Santos A.
- Published
- 2019
- Full Text
- View/download PDF
8. PML-Regulated Mitochondrial Metabolism Enhances Chemosensitivity in Human Ovarian Cancers
- Author
-
Gentric, Géraldine, Kieffer, Yann, Mieulet, Virginie, Goundiam, Oumou, Bonneau, Claire, Nemati, Fariba, Hurbain, Ilse, Raposo, Graca, Popova, Tatiana, Stern, Marc-Henri, Lallemand-Breitenbach, Valérie, Müller, Sebastian, Cañeque, Tatiana, Rodriguez, Raphaël, Vincent-Salomon, Anne, de Thé, Hugues, Rossignol, Rodrigue, and Mechta-Grigoriou, Fatima
- Published
- 2019
- Full Text
- View/download PDF
9. Optimization of tumor xenograft dissociation for the profiling of cell surface markers and nutrient transporters
- Author
-
Petit, Vincent, Massonnet, Gérald, Maciorowski, Zofia, Touhami, Jawida, Thuleau, Aurélie, Némati, Fariba, Laval, Julie, Château-Joubert, Sophie, Servely, Jean-Luc, Vallerand, David, Fontaine, Jean-Jacques, Taylor, Naomi, Battini, Jean-Luc, Sitbon, Marc, and Decaudin, Didier
- Published
- 2013
- Full Text
- View/download PDF
10. FAK Inhibitor-Based Combinations with MEK or PKC Inhibitors Trigger Synergistic Antitumor Effects in Uveal Melanoma.
- Author
-
Tarin, Malcy, Némati, Fariba, Decaudin, Didier, Canbezdi, Christine, Marande, Benjamin, Silva, Lisseth, Derrien, Héloïse, Jochemsen, Aart G., Gardrat, Sophie, Piperno-Neumann, Sophie, Rodrigues, Manuel, Mariani, Pascale, Cassoux, Nathalie, Stern, Marc-Henri, Roman-Roman, Sergio, and Alsafadi, Samar
- Subjects
- *
IN vitro studies , *IN vivo studies , *XENOGRAFTS , *GENETIC mutation , *MELANOMA , *PROTEIN kinase inhibitors , *ANIMAL experimentation , *UVEA cancer , *METASTASIS , *ANTINEOPLASTIC agents , *APOPTOSIS , *PROTEIN-tyrosine kinase inhibitors , *CELL survival , *DRUG synergism , *RESEARCH funding , *CELL lines , *MICE , *PHARMACODYNAMICS - Abstract
Simple Summary: Uveal Melanoma (UM) is a rare and malignant intraocular tumor with dismal prognosis. Despite efficient control of the primary tumor by radiation or surgery, up to 50% of patients subsequently develop metastases, mainly in the liver. The treatment of UM metastases is challenging and the patient survival is very poor. Today, most of the tested treatments including inhibitors of protein kinase C (PKC), mitogen-activated protein kinase (MEK) or immune checkpoint blockade have been largely ineffective in patients with metastatic UM. Given that single-agent targeted therapies often activate compensatory mechanisms, combination strategies are relevant to evaluate in UM in preclinical and clinical settings. Our study confirms the previously described synergy of the dual inhibition of focal adhesion kinase (FAK) and MEK, and identifies a novel combination of drugs (FAK and PKC inhibitors) as a promising strategy for therapeutic intervention in metastatic UM. Uveal Melanoma (UM) is a rare and malignant intraocular tumor with dismal prognosis. Even if radiation or surgery permit an efficient control of the primary tumor, up to 50% of patients subsequently develop metastases, mainly in the liver. The treatment of UM metastases is challenging and the patient survival is very poor. The most recurrent event in UM is the activation of Gαq signaling induced by mutations in GNAQ/11. These mutations activate downstream effectors including protein kinase C (PKC) and mitogen-activated protein kinases (MAPK). Clinical trials with inhibitors of these targets have not demonstrated a survival benefit for patients with UM metastasis. Recently, it has been shown that GNAQ promotes YAP activation through the focal adhesion kinase (FAK). Pharmacological inhibition of MEK and FAK showed remarkable synergistic growth-inhibitory effects in UM both in vitro and in vivo. In this study, we have evaluated the synergy of the FAK inhibitor with a series of inhibitors targeting recognized UM deregulated pathways in a panel of cell lines. The combined inhibition of FAK and MEK or PKC had highly synergistic effects by reducing cell viability and inducing apoptosis. Furthermore, we demonstrated that these combinations exert a remarkable in vivo activity in UM patient-derived xenografts. Our study confirms the previously described synergy of the dual inhibition of FAK and MEK and identifies a novel combination of drugs (FAK and PKC inhibitors) as a promising strategy for therapeutic intervention in metastatic UM. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
11. TIPIN depletion leads to apoptosis in breast cancer cells
- Author
-
Baldeyron, Céline, Brisson, Amélie, Tesson, Bruno, Némati, Fariba, Koundrioukoff, Stéphane, Saliba, Elie, De Koning, Leanne, Martel, Elise, Ye, Mengliang, Rigaill, Guillem, Meseure, Didier, Nicolas, André, Gentien, David, Decaudin, Didier, Debatisse, Michelle, Depil, Stéphane, Cruzalegui, Francisco, Pierré, Alain, Roman-Roman, Sergio, Tucker, Gordon C., and Dubois, Thierry
- Published
- 2015
- Full Text
- View/download PDF
12. Patient-derived xenografts recapitulate molecular features of human uveal melanomas
- Author
-
Laurent, Cécile, Gentien, David, Piperno-Neumann, Sophie, Némati, Fariba, Nicolas, André, Tesson, Bruno, Desjardins, Laurence, Mariani, Pascale, Rapinat, Audrey, Sastre-Garau, Xavier, Couturier, Jérôme, Hupé, Philippe, de Koning, Leanne, Dubois, Thierry, Roman-Roman, Sergio, Stern, Marc-Henri, Barillot, Emmanuel, Harbour, William J., Saule, Simon, and Decaudin, Didier
- Published
- 2013
- Full Text
- View/download PDF
13. Dramatic In Vivo Efficacy of the EZH2-Inhibitor Tazemetostat in PBRM1 -Mutated Human Chordoma Xenograft.
- Author
-
Passeri, Thibault, Dahmani, Ahmed, Masliah-Planchon, Julien, Naguez, Adnan, Michou, Marine, El Botty, Rania, Vacher, Sophie, Bouarich, Rachida, Nicolas, André, Polivka, Marc, Franck, Coralie, Schnitzler, Anne, Némati, Fariba, Roman-Roman, Sergio, Bourdeaut, Franck, Adle-Biassette, Homa, Mammar, Hamid, Froelich, Sébastien, Bièche, Ivan, and Decaudin, Didier
- Subjects
GERM cell tumors ,DRUG efficacy ,GENETIC mutation ,ANTINEOPLASTIC agents ,GENOTYPES ,GENOMICS ,PROGRESSION-free survival ,EPIGENOMICS - Abstract
Simple Summary: Chordomas are rare bone tumors characterized by a high recurrence rate. Presently, no medical treatment is available for advanced diseases due to the lack of molecular data and preclinical models. The current study showed the establishment and characterization of the largest panel chordoma xenografts, allowing pharmacological studies. In one PBRM1-mutated model, we demonstrated a strong therapeutic efficacy of the EZH2-inhibitor tazemetostat, encouraging further research on EZH2-inhibitors in chordomas. Chordomas are rare neoplasms characterized by a high recurrence rate and a poor long-term prognosis. Considering their chemo-/radio-resistance, alternative treatment strategies are strongly required, but their development is limited by the paucity of relevant preclinical models. Mutations affecting genes of the SWI/SNF complexes are frequently found in chordomas, suggesting a potential therapeutic effect of epigenetic regulators in this pathology. Twelve PDX models were established and characterized on histological and biomolecular features. Patients whose tumors were able to grow into mice had a statistically significant lower progression-free survival than those whose tumors did not grow after in vivo transplantation (p = 0.007). All PDXs maintained the same histopathological features as patients' tumors. Homozygous deletions of CDKN2A/2B (58.3%) and PBRM1 (25%) variants were the most common genomic alterations found. In the tazemetostat treated PDX model harboring a PBRM1 variant, an overall survival of 100% was observed. Our panel of chordoma PDXs represents a useful preclinical tool for both pharmacologic and biological assessments. The first demonstration of a high antitumor activity of tazemetostat in a PDX model harboring a PBRM1 variant supports further evaluation for EZH2-inhibitors in this subgroup of chordomas. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
14. Protein arginine methyltransferase 5: A novel therapeutic target for triple‐negative breast cancers.
- Author
-
Vinet, Mathilde, Suresh, Samyuktha, Maire, Virginie, Monchecourt, Clarisse, Némati, Fariba, Lesage, Laetitia, Pierre, Fabienne, Ye, Mengliang, Lescure, Auriane, Brisson, Amélie, Meseure, Didier, Nicolas, André, Rigaill, Guillem, Marangoni, Elisabetta, Del Nery, Elaine, Roman‐Roman, Sergio, and Dubois, Thierry
- Abstract
TNBC is a highly heterogeneous and aggressive breast cancer subtype associated with high relapse rates, and for which no targeted therapy yet exists. Protein arginine methyltransferase 5 (PRMT5), an enzyme which catalyzes the methylation of arginines on histone and non‐histone proteins, has recently emerged as a putative target for cancer therapy. Potent and specific PRMT5 inhibitors have been developed, but the therapeutic efficacy of PRMT5 targeting in TNBC has not yet been demonstrated. Here, we examine the expression of PRMT5 in a human breast cancer cohort obtained from the Institut Curie, and evaluate the therapeutic potential of pharmacological inhibition of PRMT5 in TNBC. We find that PRMT5 mRNA and protein are expressed at comparable levels in TNBC, luminal breast tumors, and healthy mammary tissues. However, immunohistochemistry analyses reveal that PRMT5 is differentially localized in TNBC compared to other breast cancer subtypes and to normal breast tissues. PRMT5 is heterogeneously expressed in TNBC and high PRMT5 expression correlates with poor prognosis within this breast cancer subtype. Using the small‐molecule inhibitor EPZ015666, we show that PRMT5 inhibition impairs cell proliferation in a subset of TNBC cell lines. PRMT5 inhibition triggers apoptosis, regulates cell cycle progression and decreases mammosphere formation. Furthermore, EPZ015666 administration to a patient‐derived xenograft model of TNBC significantly deters tumor progression. Finally, we reveal potentiation between EGFR and PRMT5 targeting, suggestive of a beneficial combination therapy. Our findings highlight a distinctive subcellular localization of PRMT5 in TNBC, and uphold PRMT5 targeting, alone or in combination, as a relevant treatment strategy for a subset of TNBC.Alternatives to current treatments are direly needed for patients with triple‐negative breast cancers (TNBC). The present study endorses PRMT5 as a novel therapeutic target in TNBC. PRMT5 pharmacological inhibition decreases viability in a subset of TNBC cell lines, mitigates early‐stage tumor progression in vivo, and displays additive/synergistic effects in combination with EGFR inhibition. Importantly, this study shows distinctive PRMT5 subcellular localization in TNBC, underlining the importance of site‐specific PRMT5 activity in the context of breast cancers. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
15. LRP8 is overexpressed in estrogen‐negative breast cancers and a potential target for these tumors.
- Author
-
Maire, Virginie, Mahmood, Faisal, Rigaill, Guillem, Ye, Mengliang, Brisson, Amélie, Némati, Fariba, Gentien, David, Tucker, Gordon C., Roman‐Roman, Sergio, and Dubois, Thierry
- Subjects
TRIPLE-negative breast cancer ,CELL survival ,CELL cycle ,BREAST cancer ,CELL death ,ESTROGEN - Abstract
Triple‐negative breast cancer (TNBC) is the breast cancer subtype with the worst prognosis. New treatments improving the survival of TNBC patients are, therefore, urgently required. We performed a transcriptome microarray analysis to identify new treatment targets for TNBC. We found that low‐density lipoprotein receptor‐related protein 8 (LRP8) was more strongly expressed in estrogen receptor‐negative breast tumors, including TNBCs and those overexpressing HER2, than in luminal breast tumors and normal breast tissues. LRP8 depletion decreased cell proliferation more efficiently in estrogen receptor‐negative breast cancer cell lines: TNBC and HER2 overexpressing cell lines. We next focused on TNBC cells for which targeted therapies are not available. LRP8 depletion induced an arrest of the cell cycle progression in G1 phase and programmed cell death. We also found that LRP8 is required for anchorage‐independent growth in vitro, and that its depletion in vivo slowed tumor growth in a xenograft model. Our findings suggest that new approaches targeting LRP8 may constitute promising treatments for hormone‐negative breast cancers, those overexpressing HER2 and TNBCs. LRP8 is a transmembrane receptor that has been extensively studied in the field of neuroscience, but rarely in the context of cancer. We show here that LRP8 is highly expressed in estrogen receptor‐negative breast tumors: ER‐/HER2+ and TNBC tumors. We show that LRP8 is essential for cell viability, due to its role in controlling cell cycle progression and apoptosis, and required for cell tumorigenesis both in vitro and in vivo. Our results identify LRP8 as a new potential candidate target for the treatment of estrogen receptor‐negative breast tumors. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
16. Nanobodies against surface biomarkers enable the analysis of tumor genetic heterogeneity in uveal melanoma patient-derived xenografts.
- Author
-
Crépin, Ronan, Gentien, David, Duché, Angeline, Rapinat, Audrey, Reyes, Cecile, Némati, Fariba, Massonnet, Gérald, Decaudin, Didier, Djender, Selma, Moutel, Sandrine, Desrumeaux, Klervi, Cassoux, Nathalie, Piperno ‐ Neumann, Sophie, Amigorena, Sebastian, Perez, Franck, Roman ‐ Roman, Sergio, and Marco, Ario
- Subjects
EYE cancer ,MONOCLONAL antibodies ,XENOGRAFTS ,TUMOR markers ,TUMOR antigens ,CANCER cells ,FLOW cytometry - Abstract
Monoclonal antibodies specific for biomarkers expressed on the surface of uveal melanoma ( UM) cells would simplify the immune capture and genomic characterization of heterogeneous tumor cells originated from patient-derived xenografts ( PDXs). Antibodies against four independent tumor antigens were isolated by panning a nanobody synthetic library. Such antibodies enabled flow cytometry-based sorting of distinct cell subpopulations from UM PDXs and to analyze their genomic features. The complexity and specificity of the biochemical and genomic biomarker combinations mirrored the UM tumor polyclonality. The data showed that MUC18 is highly and universally displayed on the surface of UM cells with different genetic background and consequently represents a reliable pan-biomarker for their identification and purification. In contrast, the other three biomarkers were detected in very variable combinations in UM PDX cells. The availability of the identified nanobodies will be instrumental in developing clone-specific drug evaluation and rational clinical strategies based on accurate genomic profiling. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
17. Upregulation of HLA Expression in Primary Uveal Melanoma by Infiltrating Leukocytes.
- Author
-
van Essen, T. Huibertus, van Pelt, Sake I., Bronkhorst, Inge H. G., Versluis, Mieke, Némati, Fariba, Laurent, Cécile, Luyten, Gregorius P. M., van Hall, Thorbald, van den Elsen, Peter J., van der Velden, Pieter A., Decaudin, Didier, and Jager, Martine J.
- Subjects
HLA histocompatibility antigens ,UVEA cancer ,MELANOMA treatment ,LEUCOCYTES ,GENE expression ,CANCER-related mortality ,IMMUNOHISTOCHEMISTRY ,POLYMERASE chain reaction - Abstract
Introduction: Uveal melanoma (UM) with an inflammatory phenotype, characterized by infiltrating leukocytes and increased human leukocyte antigen (HLA) expression, carry an increased risk of death due to metastases. These tumors should be ideal for T-cell based therapies, yet it is not clear why prognostically-infaust tumors have a high HLA expression. We set out to determine whether the level of HLA molecules in UM is associated with other genetic factors, HLA transcriptional regulators, or microenvironmental factors. Methods: 28 enucleated UM were used to study HLA class I and II expression, and several regulators of HLA by immunohistochemistry, PCR microarray, qPCR and chromosome SNP-array. Fresh tumor samples of eight primary UM and four metastases were compared to their corresponding xenograft in SCID mice, using a PCR microarray and SNP array. Results: Increased expression levels of HLA class I and II showed no dosage effect of chromosome 6p, but, as expected, were associated with monosomy of chromosome 3. Increased HLA class I and II protein levels were positively associated with their gene expression and with raised levels of the peptide-loading gene TAP1, and HLA transcriptional regulators IRF1, IRF8, CIITA, and NLRC5, revealing a higher transcriptional activity in prognostically-bad tumors. Implantation of fresh human tumor samples into SCID mice led to a loss of infiltrating leukocytes, and to a decreased expression of HLA class I and II genes, and their regulators. Conclusion: Our data provides evidence for a proper functioning HLA regulatory system in UM, offering a target for T-cell based therapies. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
18. Vasculature analysis of patient derived tumor xenografts using species-specific PCR assays: evidence of tumor endothelial cells and atypical VEGFA-VEGFR1/2 signalings.
- Author
-
Bieche, Ivan, Vacher, Sophie, Vallerand, David, Richon, Sophie, Hatem, Rana, De Plater, Ludmilla, Dahmani, Ahmed, Némati, Fariba, Angevin, Eric, Marangoni, Elisabetta, Roman-Roman, Sergio, Decaudin, Didier, and Dangles-Marie, Virginie
- Subjects
VASCULAR endothelial growth factor receptors ,XENOGRAFTS ,TUMOR blood vessels ,CELLULAR signal transduction ,GLIOMAS ,POLYMERASE chain reaction ,GENE expression - Abstract
Background: Tumor endothelial transdifferentiation and VEGFR1/2 expression by cancer cells have been reported in glioblastoma but remain poorly documented for many other cancer types. Methods: To characterize vasculature of patient-derived tumor xenografts (PDXs), largely used in preclinical anti-angiogenic assays, we designed here species-specific real-time quantitative RT-PCR assays. Human and mouse PECAM1/CD31, ENG/CD105, FLT1/VEGFR1, KDR/VEGFR2 and VEGFA transcripts were analyzed in a large series of 150 PDXs established from 8 different tumor types (53 colorectal, 14 ovarian, 39 breast and 15 renal cell cancers, 6 small cell and 5 non small cell lung carcinomas, 13 cutaneous melanomas and 5 glioblastomas) and in two bevacizumab-treated non small cell lung carcinomas xenografts. Results: As expected, mouse cell proportion in PDXs -evaluated by quantifying expression of the housekeeping gene TBP- correlated with all mouse endothelial markers and human VEGFA RNA levels. More interestingly, we observed human PECAM1/CD31 and ENG/CD105 expression in all tumor types, with higher rate in glioblastoma and renal cancer xenografts. Human VEGFR expression profile varied widely depending on tumor types with particularly high levels of human FLT1/VEGFR1 transcripts in colon cancers and non small cell lung carcinomas, and upper levels of human KDR/ VEGFR2 transcripts in non small cell lung carcinomas. Bevacizumab treatment induced significant low expression of mouse Pecam1/Cd31, Eng/Cd105, Flt1/Vegfr1 and Kdr/Vefr2 while the human PECAM1/CD31 and VEGFA were upregulated. Conclusions: Taken together, our results strongly suggest existence of human tumor endothelial cells in all tumor types tested and of both stromal and tumoral autocrine VEGFA-VEGFR1/2 signalings. These findings should be considered when evaluating molecular mechanisms of preclinical response and resistance to tumor anti-angiogenic strategies. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
19. Targeting Bcl-2/Bcl-XL Induces Antitumor Activity in Uveal Melanoma Patient-Derived Xenografts.
- Author
-
Némati, Fariba, de Montrion, Catherine, Lang, Guillaume, Kraus-Berthier, Laurence, Carita, Guillaume, Sastre-Garau, Xavier, Berniard, Aurélie, Vallerand, David, Geneste, Olivier, de Plater, Ludmilla, Pierré, Alain, Lockhart, Brian, Desjardins, Laurence, Piperno-Neumann, Sophie, Depil, Stéphane, and Decaudin, Didier
- Subjects
- *
GENE targeting , *BCL genes , *ANTINEOPLASTIC agents , *UVEA cancer , *MELANOMA , *XENOGRAFTS , *CANCER risk factors - Abstract
Purpose: Uveal melanoma (UM) is associated with a high risk of metastases and lack of efficient therapies. Reduced capacity for apoptosis induction by chemotherapies is one obstacle to efficient treatments. Human UM is characterized by high expression of the anti-apoptotic protein Bcl-2. Consequently, regulators of apoptosis such as Bcl-2 family inhibitors may constitute an attractive approach to UM therapeutics. In this aim, we have investigated the efficacy of the Bcl-2/Bcl-XL inhibitor S44563 on 4 UM Patient-Derived Xenografts (PDXs) and derived-cell lines. Experimental Design: Four well characterized UM PDXs were used for in vivo experiments. S44563 was administered alone or combined with fotemustine either concomitantly or after the alkylating agent. Bcl-2, Bcl-XL, and Mcl-1 expressions after S44563 administration were evaluated by immunohistochemistry (IHC). Results: S44563 administered alone by at 50 and 100 mg/kg i.p. induced a significant tumour growth inhibition in only one xenograft model with a clear dose effect. However, when S44563 was concomitantly administered with fotemustine, we observed a synergistic activity in 3 out of the 4 tested models. In addition, S44563 administered after fotemustine induced a tumour growth delay in 2 out of 3 tested xenografts. Finally, IHC analyses showed that Bcl-2, Bcl-XL, and Mcl-1 expression were not modified after S44563 administration. Conclusion: The novel anti-apoptotic experimental compound S44563, despite a relative low efficacy when administered alone, increased the efficacy of fotemustine in either concomitant or sequential combinations or indeed subsequent to fotemustine. These data support further exploration of potential therapeutic effect of Bcl-2/Bcl-xl inhibition in human UM. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
20. Polo-like Kinase 1: A Potential Therapeutic Option in Combination with Conventional Chemotherapy for the Management of Patients with Triple-Negative Breast Cancer.
- Author
-
Maire, Virginie, Némati, Fariba, Richardson, Marion, Vincent-Salomon, Anne, Tesson, Bruno, Rigaill, Guillem, Gravier, Eléonore, Marty-Prouvost, Bérengère, De Koning, Leanne, Lang, Guillaume, Gentien, David, Dumont, Aurélie, Barillot, Emmanuel, Marangoni, Elisabetta, Decaudin, Didier, Roman-Roman, Sergio, Pierré, Alain, Cruzalegui, Francisco, Depil, Stéphane, and Tucker, Gordon C.
- Subjects
- *
BREAST cancer treatment , *CANCER prognosis , *CANCER genetics , *GENE expression , *CYCLOPHOSPHAMIDE - Abstract
Breast cancers are composed of molecularly distinct subtypes with different clinical outcomes and responses to therapy. To discover potential therapeutic targets for the poor prognosis-associated triple-negative breast cancer (TNBC), gene expression profiling was carried out on a cohort of 130 breast cancer samples. Polo-like kinase 1 (PLK1) was found to be significantly overexpressed in TNBC compared with the other breast cancer subtypes. High PLK1 expression was confirmed by reverse phase protein and tissue microarrays. In triple-negative cell lines, RNAi-mediated PLK1 depletion or inhibition of PLK1 activity with a small molecule (BI-2536) induced an increase in phosphorylated H2AX, G2--M arrest, and apoptosis. A soft-agar colony assay showed that PLK1 silencing impaired clonogenic potential of TNBC cell lines. When cells were grown in extracellular matrix gels (Matrigel), and exposed to BI-2536, apoptosiswas observed specifically in TNBC cancerous cells, and not in a normal cell line. When administrated as a single agent, the PLK1 inhibitor significantly impaired tumor growth in in vivo two xenografts models established from biopsies of patients with TNBC. Most importantly, the administration of BI-2536, in combination with doxorubicin + cyclophosphamide chemotherapy, led to a faster complete response compared with the chemotherapy treatment alone and prevented relapse, which is the major risk associated with TNBC. Altogether, our observations suggest PLK1 inhibition as an attractive therapeutic approach, in association with conventional chemotherapy, for the management of patients with TNBC. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
21. PRMT1 Regulates EGFR and Wnt Signaling Pathways and Is a Promising Target for Combinatorial Treatment of Breast Cancer.
- Author
-
Suresh, Samyuktha, Huard, Solène, Brisson, Amélie, Némati, Fariba, Dakroub, Rayan, Poulard, Coralie, Ye, Mengliang, Martel, Elise, Reyes, Cécile, Silvestre, David C., Meseure, Didier, Nicolas, André, Gentien, David, Fayyad-Kazan, Hussein, Le Romancer, Muriel, Decaudin, Didier, Roman-Roman, Sergio, and Dubois, Thierry
- Subjects
BREAST tumor treatment ,BREAST cancer prognosis ,XENOGRAFTS ,COMBINATION drug therapy ,CANCER chemotherapy ,EPIDERMAL growth factor receptors ,APOPTOSIS ,CELLULAR signal transduction ,CELL survival ,TRANSFERASES ,GENE expression profiling ,CELL proliferation ,CELL lines - Abstract
Simple Summary: Patients with triple-negative breast cancer (TNBC) respond well to chemotherapy initially but are prone to relapse. Searching for new therapeutic targets, we found that PRMT1 is highly expressed in TNBC tumor samples and is essential for breast cancer cell survival. Furthermore, this study proposes that targeting PRMT1 in combination with chemotherapies could improve the survival outcome of TNBC patients. Identifying new therapeutic strategies for triple-negative breast cancer (TNBC) patients is a priority as these patients are highly prone to relapse after chemotherapy. Here, we found that protein arginine methyltransferase 1 (PRMT1) is highly expressed in all breast cancer subtypes. PRMT1 depletion decreases cell survival by inducing DNA damage and apoptosis in various breast cancer cell lines. Transcriptomic analysis and chromatin immunoprecipitation revealed that PRMT1 regulates the epidermal growth factor receptor (EGFR) and the Wnt signaling pathways, reported to be activated in TNBC. PRMT1 enzymatic activity is also required to stimulate the canonical Wnt pathway. Type I PRMT inhibitors decrease breast cancer cell proliferation and show anti-tumor activity in a TNBC xenograft model. These inhibitors display synergistic interactions with some chemotherapies used to treat TNBC patients as well as erlotinib, an EGFR inhibitor. Therefore, targeting PRMT1 in combination with these chemotherapies may improve existing treatments for TNBC patients. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
22. Conjunctival low-grade non-Hodgkin's lymphoma: a large single-center study of initial characteristics, natural history and prognostic factors.
- Author
-
Meunier, Jérôme, Rouïc, Livia Lumbroso-Le, Dendale, Rémi, Vincent-Salomon, Anne, Asselain, Bernard, Arnaud, Philippe, Némati, Fariba, Fourquet, Alain, Desjardins, Laurence, Plancher, Corine, Levy, Christine, Chaoui, Driss, Validire, Patricia, and Decaudin, Didier
- Subjects
LYMPHOMAS ,PROGNOSIS ,LYMPHOID tissue ,CONJUNCTIVA ,MULTIVARIATE analysis - Abstract
To define the initial characteristics and prognostic factors of patients with conjunctival low-grade malignant lymphoma, all patients treated for low-grade lymphoma with initial conjunctival involvement were reviewed. Forty-nine cases were selected, including 45 cases with exclusive ophthalmologic conjunctival involvement. Pathologic review showed 55% of mucosa-associated lymphoid tissue type lymphoma, and 23% of lymphoplasmocytic lymphoma. Initial characteristics were median age of 62 years, nodal involvement in 17% of cases, and stage IV in 22% of patients with 10% of bone marrow involvement. With a median follow-up of 75 months, the 5-year disease-free survival (DFS) and overall survival were 65% and 83%, respectively. On multivariate analysis, nodal involvement was the only factor with a pejorative impact on DFS. Our patient cohort represents one of the largest published series defining the characteristics and prognostic factors of primary conjunctival low-grade malignant lymphoma. [ABSTRACT FROM AUTHOR]
- Published
- 2006
- Full Text
- View/download PDF
23. High-content drug screening in zebrafish xenografts reveals high efficacy of dual MCL-1/BCL-XL inhibition against Ewing sarcoma.
- Author
-
Grissenberger, Sarah, Sturtzel, Caterina, Wenninger-Weinzierl, Andrea, Radic-Sarikas, Branka, Scheuringer, Eva, Bierbaumer, Lisa, Etienne, Vesnie, Némati, Fariba, Pascoal, Susana, Tötzl, Marcus, Tomazou, Eleni M., Metzelder, Martin, Putz, Eva M., Decaudin, Didier, Delattre, Olivier, Surdez, Didier, Kovar, Heinrich, Halbritter, Florian, Distel, Martin, and Tomazou, Eleni
- Subjects
- *
EWING'S sarcoma , *MEDICAL screening , *XENOGRAFTS , *BRACHYDANIO , *DNA topoisomerase I - Abstract
Ewing sarcoma is a pediatric bone and soft tissue cancer with an urgent need for new therapies to improve disease outcome. To identify effective drugs, phenotypic drug screening has proven to be a powerful method, but achievable throughput in mouse xenografts, the preclinical Ewing sarcoma standard model, is limited. Here, we explored the use of xenografts in zebrafish for high-throughput drug screening to discover new combination therapies for Ewing sarcoma. We subjected xenografts in zebrafish larvae to high-content imaging and subsequent automated tumor size analysis to screen single agents and compound combinations. We identified three drug combinations effective against Ewing sarcoma cells: Irinotecan combined with either an MCL-1 or an BCL-XL inhibitor and in particular dual inhibition of the anti-apoptotic proteins MCL-1 and BCL-XL, which efficiently eradicated tumor cells in zebrafish xenografts. We confirmed enhanced efficacy of dual MCL-1/BCL-XL inhibition compared to single agents in a mouse PDX model. In conclusion, high-content screening of small compounds on Ewing sarcoma zebrafish xenografts identified dual MCL-1/BCL-XL targeting as a specific vulnerability and promising therapeutic strategy for Ewing sarcoma, which warrants further investigation towards clinical application. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.