Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Publication year range
1.
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
2.
Biol Aujourdhui ; 206(2): 111-23, 2012.
Article in French | MEDLINE | ID: mdl-22748049

ABSTRACT

Tumor microenvironment is a complex system composed of a largely altered extracellular matrix (ECM) with different cell types that determine the angiogenic response. Upon the influence of hypoxia, tumor cells secrete cytokines that activate stromal cells to produce proteases and angiogenic factors. The proteases degrade the stromal ECM and participate in the release of various ECM fragments, named matrikines or matricryptins, capable to control tumor invasion and metastasis dissemination. We will focus on the matrikines derived from the NC1 domains of the different constitutive chains of basement membrane-associated collagens and mainly collagen IV. The putative targets of the matrikine action are the proliferation and invasive properties of tumor or inflammatory cells, and the angiogenic and lymphangiogenic responses. For example, canstatin, tumstatin and tetrastatin, respectively derived from the NC1 domains of α2, α3 and α4 chains of collagen IV, inhibit in vivo tumor growth in various experimental cancer models. Their anti-cancer activity comprises an anti-proliferative effect on tumor cells and on endothelial cells by induction of cell apoptosis or cell cycle blockade and the induction of a loss of their migratory phenotype. Matrikines constitute a new family of potent anticancer agents that could be used under various therapeutic strategies: i) induction of their overexpression by cancer cells or by the host cells, ii) use of recombinant proteins or synthetic peptides or structural analogues designed from the structure of the active sequences. These matrikines could be used in combination with conventional chemotherapy or radiotherapy to limit tumor progression.


Subject(s)
Antineoplastic Agents/therapeutic use , Cytokines/physiology , Cytokines/therapeutic use , Extracellular Matrix/metabolism , Neoplasms/drug therapy , Animals , Basement Membrane/drug effects , Basement Membrane/metabolism , Basement Membrane/physiology , Clinical Trials as Topic , Collagen/chemistry , Collagen/metabolism , Collagen/physiology , Cytokines/chemistry , Cytokines/metabolism , Humans , Models, Biological , Neoplasms/metabolism
3.
PLoS One ; 7(4): e29587, 2012.
Article in English | MEDLINE | ID: mdl-22539938

ABSTRACT

BACKGROUND: NC1 domains from α1, α2, α3 and α6(IV) collagen chains were shown to exert anti-tumor or anti-angiogenic activities, whereas the NC1 domain of the α4(IV) chain did not show such activities so far. METHODOLOGY/PRINCIPAL FINDINGS: We demonstrate in the present paper that the NC1 α4(IV) domain exerts a potent anti-tumor activity both in vitro and in an experimental human melanoma model in vivo. The overexpression of NC1 α4(IV) in human UACC-903 melanoma cells strongly inhibited their in vitro proliferative (-38%) and invasive (-52%) properties. MT1-MMP activation was largely decreased and its cellular distribution was modified, resulting in a loss of expression at the migration front associated with a loss of migratory phenotype. In an in vivo xenograft model in athymic nude mice, the subcutaneous injection of NC1 α4(IV)-overexpressing melanoma cells induced significantly smaller tumors (-80% tumor volume) than the Mock cells, due to a strong inhibition of tumor growth. Exogenously added recombinant human NC1 α4(IV) reproduced the inhibitory effects of NC1 α4(IV) overexpression in UACC-903 cells but not in dermal fibroblasts. An anti-αvß3 integrin blocking antibody inhibited cell adhesion on recombinant human NC1 α4(IV) substratum. The involvement of αvß3 integrin in mediating NC1 α4(IV) effect was confirmed by surface plasmon resonance (SPR) binding assays showing that recombinant human NC1 α4(IV) binds to αvß3 integrin (K(D) = 148 ± 9.54 nM). CONCLUSION/SIGNIFICANCE: Collectively, our results demonstrate that the NC1 α4(IV) domain, named tetrastatin, is a new endogenous anti-tumor matrikine.


Subject(s)
Antineoplastic Agents/therapeutic use , Collagen Type IV/chemistry , Melanoma, Experimental/drug therapy , Animals , Antibodies/immunology , Antineoplastic Agents/pharmacology , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Collagen Type IV/genetics , Collagen Type IV/metabolism , Humans , Integrin alphaVbeta3/immunology , Integrin alphaVbeta3/metabolism , Matrix Metalloproteinases/metabolism , Mice , Mice, Nude , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Surface Plasmon Resonance , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...