Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Exp Cell Res ; 426(2): 113568, 2023 05 15.
Article in English | MEDLINE | ID: mdl-36967104

ABSTRACT

l-Asparaginase is a cornerstone of acute lymphoblastic leukemia (ALL) therapy since lymphoblasts lack asparagine synthetase (ASNS) and rely on extracellular asparagine availability for survival. Resistance mechanisms are associated with increased ASNS expression in ALL. However, the association between ASNS and l-Asparaginase efficacy in solid tumors remains unclear, thus limiting clinical development. Interestingly, l-Asparaginase also has a glutaminase co-activity that is crucial in pancreatic cancer where KRAS mutations activate glutamine metabolism. By developing l-Asparaginase-resistant pancreatic cancer cells and using OMICS approaches, we identified glutamine synthetase (GS) as a marker of resistance to l-Asparaginase. GS is the only enzyme able to synthesize glutamine, and its expression also correlates with l-Asparaginase efficacy in 27 human cell lines from 11 cancer indications. Finally, we further demonstrated that GS inhibition prevents cancer cell adaptation to l-Asparaginase-induced glutamine starvation. These findings could pave the way to the development of promising drug combinations to overcome l-Asparaginase resistance.


Subject(s)
Pancreatic Neoplasms , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Humans , Asparaginase/pharmacology , Glutamate-Ammonia Ligase/genetics , Glutaminase/genetics , Glutamine/metabolism , Pancreatic Neoplasms/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Pancreatic Neoplasms
2.
iScience ; 24(12): 103423, 2021 Dec 17.
Article in English | MEDLINE | ID: mdl-34849474

ABSTRACT

Lack of preclinical patient-derived xenograft cancer models in which to conduct large-scale molecular studies seriously impairs the development of effective personalized therapies. We report here an in vivo concept consisting of implanting human tumor cells in targeted tissues of an avian embryo, delivering therapeutics, evaluating their efficacy by measuring tumors using light sheet confocal microscopy, and conducting large-scale RNA-seq analysis to characterize therapeutic-induced changes in gene expression. The model was established to recapitulate triple-negative breast cancer (TNBC) and validated using TNBC standards of care and an investigational therapeutic agent.

3.
Cancer Med ; 6(6): 1437-1452, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28544589

ABSTRACT

Erymet is a new therapy resulting from the encapsulation of a methionine gamma-lyase (MGL; EC number 4.4.1.11) in red blood cells (RBC). The aim of this study was to evaluate erymet potential efficacy in methionine (Met)-dependent cancers. We produced a highly purified MGL using a cGMP process, determined the pharmacokinetics/pharmacodynamics (PK/PD) properties of erymet in mice, and assessed its efficacy on tumor growth prevention. Cytotoxicity of purified MGL was tested in six cancer cell lines. CD1 mice were injected with single erymet product supplemented or not with vitamin B6 vitamer pyridoxine (PN; a precursor of PLP cofactor). NMRI nude mice were xenografted in the flank with U-87 MG-luc2 glioblastoma cells for tumor growth study following five intravenous (IV) injections of erymet with daily PN oral administration. Endpoints included efficacy and event-free survival (EFS). Finally, a repeated dose toxicity study of erymet combined with PN cofactor was conducted in CD1 mice. Recombinant MGL was cytotoxic on 4/6 cell lines tested. MGL half-life was increased from <24 h to 9-12 days when encapsulated in RBC. Conversion of PN into PLP by RBC was demonstrated. Combined erymet + PN treatment led to a sustained Met depletion in plasma for several days with a 85% reduction of tumor volume after 45 days following cells implantation, and a significant EFS prolongation for treated mice. Repeated injections in mice exhibited a very good tolerability with only minor impact on clinical state (piloerection, lean aspect) and a slight decrease in hemoglobin and triglyceride concentrations. This study demonstrated that encapsulation of methioninase inside erythrocyte greatly enhanced pharmacokinetics properties of the enzyme and is efficacy against tumor growth. The perspective on these results is the clinical evaluation of the erymet product in patients with Met starvation-sensitive tumors.


Subject(s)
Antineoplastic Agents/administration & dosage , Carbon-Sulfur Lyases/administration & dosage , Drug Delivery Systems , Erythrocytes , Neoplasms/drug therapy , Pyridoxine/administration & dosage , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/toxicity , Carbon-Sulfur Lyases/pharmacokinetics , Carbon-Sulfur Lyases/therapeutic use , Carbon-Sulfur Lyases/toxicity , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , Male , Methionine/blood , Methionine/metabolism , Mice, Nude , Neoplasms/blood , Neoplasms/metabolism , Neoplasms/pathology , Pyridoxal Phosphate/blood , Pyridoxine/pharmacokinetics , Pyridoxine/therapeutic use , Pyridoxine/toxicity , Recombinant Proteins/administration & dosage , Recombinant Proteins/therapeutic use , Recombinant Proteins/toxicity , Tumor Burden/drug effects
4.
Pancreas ; 44(7): 1141-7, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26355551

ABSTRACT

OBJECTIVES: Asparaginase encapsulated in erythrocytes (ERY-ASP) is a potentially effective drug in patients with pancreatic adenocarcinoma (PAC) with null/low asparagine synthetase (ASNS) expression. Our aims were to assess ASNS expression in PAC from a large cohort and its prognostic and/or predictive value and to conduct a phase I trial with ERY-ASP in patients with metastatic PAC. METHODS: Asparagine synthetase expression was evaluated using immunohistochemistry in resected PAC (471 patients) and in pairs of primary tumor and metastases (55 patients). Twelve patients were included in the phase I trial and received a single administration of ERY-ASP (25-150 IU/kg). RESULTS: Null/low ASNS expression was found in 79.4% of the resected PAC with a high concordance between primary tumor and metastases. Asparagine synthetase expression was significantly correlated with sex and CXCR4 expression. In the phase I trial, ERY-ASP was well tolerated by patients with metastatic PAC. No patient had DLTs, and 6 patients had at least 1 ERY-ASP causally related adverse event out of the 12 adverse events reported. CONCLUSIONS: Given the high rate of PAC with null/low ASNS expression and the good tolerability profile of ERY-ASP, ERY-ASP should be evaluated in further clinical studies in metastatic PAC.


Subject(s)
Adenocarcinoma/drug therapy , Asparaginase/therapeutic use , Erythrocytes/enzymology , Pancreatic Neoplasms/drug therapy , Adenocarcinoma/enzymology , Administration, Intravenous , Adult , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Asparaginase/adverse effects , Asparaginase/biosynthesis , Chemical and Drug Induced Liver Injury/etiology , Cohort Studies , Drug Hypersensitivity/etiology , Erythrocyte Transfusion/adverse effects , Erythrocyte Transfusion/methods , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Middle Aged , Outcome Assessment, Health Care/methods , Outcome Assessment, Health Care/statistics & numerical data , Pancreatic Neoplasms/enzymology , Prognosis , Proportional Hazards Models , Pancreatic Neoplasms
5.
Pancreas ; 41(6): 940-8, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22513289

ABSTRACT

OBJECTIVES: In this study, our aim was to test whether asparagine synthetase (ASNS) deficiency in pancreatic malignant cells can lead to sensitivity to asparagine starvation. We also investigated, in tumor-bearing mice, the efficacy of L-asparaginase entrapped in red blood cells (RBCs), a safe formulation, to induce asparagine depletion. METHODS: First, ASNS expression was evaluated by immunohistochemistry in sporadic pancreatic ductal adenocarcinoma. Then, 4 pancreatic carcinoma cell lines were examined by Western blot, immunocytochemistry, and cytotoxicity assay to L-asparaginase and in asparagine-free or reduced-asparagine media. Finally, mice bearing the most in vitro sensitive cell line received RBC-entrapped L-asparaginase to investigate the anticancer efficacy of serum asparagine depletion in vivo. RESULTS: Approximately 52% of pancreatic adenocarcinomas expressed no or low ASNS. The highest in vitro cytotoxicity to L-asparaginase or to reduced asparagine medium was observed with SW1990 line when ASNS expression was the lowest. In vivo sensitivity was confirmed for this cell line. CONCLUSIONS: Plasma asparagine depletion by RBC-entrapped L-asparaginase in selected patients having no low or no ASNS may be a promising therapeutic approach for pancreatic cancer.


Subject(s)
Antineoplastic Agents/administration & dosage , Asparaginase/administration & dosage , Asparagine/deficiency , Aspartate-Ammonia Ligase/metabolism , Carcinoma, Pancreatic Ductal/drug therapy , Erythrocytes/enzymology , Pancreatic Neoplasms/drug therapy , Animals , Antineoplastic Agents/blood , Asparaginase/blood , Asparagine/blood , Blotting, Western , Carcinoma, Pancreatic Ductal/blood , Carcinoma, Pancreatic Ductal/enzymology , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation , Cell Survival , Down-Regulation , Humans , Immunohistochemistry , Injections, Intravenous , Mice , Mice, Inbred BALB C , Mice, Nude , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/pathology , Time Factors , Tumor Burden , Xenograft Model Antitumor Assays
6.
PLoS Genet ; 5(7): e1000575, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19629168

ABSTRACT

Inactivation of the Transforming Growth Factor Beta (TGFbeta) tumor suppressor pathway contributes to the progression of Pancreatic Ductal AdenoCarcinoma (PDAC) since it is inactivated in virtually all cases of this malignancy. Genetic lesions inactivating this pathway contribute to pancreatic tumor progression in mouse models. Transcriptional Intermediary Factor 1 gamma (TIF1gamma) has recently been proposed to be involved in TGFbeta signaling, functioning as either a positive or negative regulator of the pathway. Here, we addressed the role of TIF1gamma in pancreatic carcinogenesis. Using conditional Tif1gamma knockout mice (Tif1gamma(lox/lox)), we selectively abrogated Tif1gamma expression in the pancreas of Pdx1-Cre;Tif1gamma(lox/lox) mice. We also generated Pdx1-Cre;LSL-Kras(G12D);Tif1gamma(lox/lox) mice to address the effect of Tif1gamma loss-of-function in precancerous lesions induced by oncogenic Kras(G12D). Finally, we analyzed TIF1gamma expression in human pancreatic tumors. In our mouse model, we showed that Tif1gamma was dispensable for normal pancreatic development but cooperated with Kras activation to induce pancreatic tumors reminiscent of human Intraductal Papillary Mucinous Neoplasms (IPMNs). Interestingly, these cystic lesions resemble those observed in Pdx1-Cre;LSL-Kras(G12D);Smad4(lox/lox) mice described by others. However, distinctive characteristics, such as the systematic presence of endocrine pseudo-islets within the papillary projections, suggest that SMAD4 and TIF1gamma don't have strictly redundant functions. Finally, we report that TIF1gamma expression is markedly down-regulated in human pancreatic tumors by quantitative RT-PCR and immunohistochemistry supporting the relevance of these findings to human malignancy. This study suggests that TIF1gamma is critical for tumor suppression in the pancreas, brings new insight into the genetics of pancreatic cancer, and constitutes a promising model to decipher the respective roles of SMAD4 and TIF1gamma in the multifaceted functions of TGFbeta in carcinogenesis and development.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Genes, Tumor Suppressor , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Transcription Factors/metabolism , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Humans , Mice , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Transcription Factors/genetics
7.
Hum Pathol ; 39(10): 1420-30, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18656242

ABSTRACT

The actin-binding protein gelsolin is involved in cell motility via the regulation of actin cytoskeleton, and its expression is modified in several human cancers. However, the potential implication of this protein in colorectal carcinogenesis is debated. By using immunohistochemistry, we studied gelsolin expression in 69 cases of colon adenocarcinomas and in 72 lesions representative of the different stages of colonic tumorigenesis. In addition, we performed Northern blot analysis of gelsolin messenger RNA in 12 paired samples of human colon cancer and normal corresponding mucosa. Gelsolin protein and messenger RNA expressions were severely down-regulated in all adenocarcinomas tested. Moreover, gelsolin protein was down-regulated in a large proportion of high-grade adenomas (14/16) before the acquisition of invasive properties but in only a small proportion of low grade adenomas and serrated adenomas (2/30) and in none of the 9 cases of nonneoplastic hyperplastic polyps tested. Our results therefore demonstrate that gelsolin down-regulation is an early and almost constant event in colon carcinogenesis and is associated with the transition from adenoma to carcinoma.


Subject(s)
Adenocarcinoma/metabolism , Adenoma/metabolism , Biomarkers, Tumor/metabolism , Cell Transformation, Neoplastic/metabolism , Colonic Neoplasms/metabolism , Gelsolin/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/secondary , Adenoma/genetics , Adenoma/pathology , Aged , Cell Count , Colectomy , Colon/anatomy & histology , Colon/metabolism , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Down-Regulation , Female , Gelsolin/genetics , Humans , Immunoenzyme Techniques , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Lymph Nodes/metabolism , Lymph Nodes/pathology , Male , Microsatellite Repeats , Middle Aged , Neoplasm Staging , RNA, Messenger/metabolism , RNA, Neoplasm/analysis
8.
Int J Cancer ; 121(10): 2162-71, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17583572

ABSTRACT

Actin depolymerizing factor (ADF)/cofilin family proteins are key regulators of actin filament turnover and cytoskeleton reorganization. The role of cofilin-1 in cell motility has been demonstrated in several cell types but remained poorly documented in the case of colon cancer. In addition, the putative function of destrin (also known as ADF) had not been explored in this context despite the fact that it is expressed in all colon cancer cell lines examined. We were therefore prompted to evaluate the respective contributions of these proteins to the invasive properties of the human colon cancer Isreco1 cell line, which expresses a comparatively high destrin/cofilin ratio. Reduction of cofilin-1 or destrin expression in Isreco1 cells using RNA interference led to an increase of the number of multinucleated cells and altered polarized lamellipodium protrusion and distribution of paxillin-containing adhesions. Both cofilin-1 and destrin silencing enhanced cell adhesion to extracellular matrix components. However, only destrin appeared to be required for cell migration on collagen I and for cell invasion through Matrigel in response to the proinvasive neuroendocrine peptide bombesin. This differential functional involvement was supported by a destrin-dependent, cofilin-independent phosphorylation of p130Crk-associated substrate (p130Cas) upon cell adhesion to collagen I or Matrigel. Taken together, our results suggest that destrin is a significant regulator of various processes important for invasive phenotype of human colon cancer Isreco1 cells whereas cofilin-1 may be involved in only a subset of them.


Subject(s)
Cofilin 1/metabolism , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Destrin/metabolism , Bombesin/pharmacology , Cell Adhesion , Cell Line, Tumor , Cell Movement , Cofilin 1/genetics , Colonic Neoplasms/genetics , Crk-Associated Substrate Protein/metabolism , Destrin/genetics , Extracellular Matrix/metabolism , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Invasiveness/pathology , Phosphorylation/drug effects , RNA, Small Interfering/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...