Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Molecules ; 24(9)2019 May 06.
Article in English | MEDLINE | ID: mdl-31064153

ABSTRACT

Inhibiting the interaction of neuropilin-1 (NRP-1) with vascular endothelial growth factor (VEGF) has become an interesting mechanism for potential anticancer therapies. In our previous works, we have obtained several submicromolar inhibitors of this interaction, including branched pentapeptides of general structure Lys(Har)-Xxx-Xxx-Arg. With the intent to improve the proteolytic stability of our inhibitors, we turned our attention to 1,4-disubstituted 1,2,3-triazoles as peptide bond isosteres. In the present contribution, we report the synthesis of 23 novel triazolopeptides along with their inhibitory activity. The compounds were synthesized using typical peptide chemistry methods, but with a conversion of amine into azide completely on solid support. The inhibitory activity of the synthesized derivatives spans from 9.2% to 58.1% at 10 µM concentration (the best compound Lys(Har)-GlyΨ[Trl]GlyΨ[Trl]Arg, 3, IC50 = 8.39 µM). Synthesized peptidotriazoles were tested for stability in human plasma and showed remarkable resistance toward proteolysis, with half-life times far exceeding 48 h. In vitro cell survival test resulted in no significant impact on bone marrow derived murine cells 32D viability. By means of molecular dynamics, we were able to propose a binding mode for compound 3 and discuss the observed structure-activity relationships.


Subject(s)
Angiogenesis Inhibitors/chemistry , Neuropilin-1/antagonists & inhibitors , Peptides/chemistry , Triazoles/chemistry , Vascular Endothelial Growth Factors/antagonists & inhibitors , Amino Acid Sequence , Amino Acids/chemistry , Angiogenesis Inhibitors/pharmacology , Animals , Bone Marrow Cells , Cell Survival/drug effects , Chromatography, High Pressure Liquid/methods , Click Chemistry/methods , Humans , Mice , Molecular Dynamics Simulation , Molecular Structure , Peptides/pharmacology , Protein Binding , Proteolysis , Solid-Phase Synthesis Techniques/methods , Structure-Activity Relationship , Tandem Mass Spectrometry/methods , Triazoles/pharmacology
2.
Eur J Med Chem ; 158: 453-462, 2018 Oct 05.
Article in English | MEDLINE | ID: mdl-30241012

ABSTRACT

The demonstrated involvement of VEGF165/NRP-1 complex in pathological angiogenesis has catalyzed interest in blocking this interaction to combat angiogenesis dependent diseases. It was shown before that Lys-Pro-Pro-Arg is a fairly strong inhibitor of the VEGF165/NRP-1 interaction. Our current findings suggest that the side chain elongation of the Lys1 by branching it with additional homoarginine (Har) residue, to obtain Lys(Har)-Pro-Pro-Arg, allows more effective inhibition. Moreover, increasing the flexibility of the middle part of molecule, in particular with simultaneous introduction of additional interacting elements at the second or third position, produced compounds up to 30-fold more active (IC50 = 0.2 µM) than the heptapeptide ATWLPPR (A7R) which is one of the first peptide known as an effective antagonist of the VEGF165 binding to NRP-1 and in vivo decreases breast cancer angiogenesis and growth. Herein, we present also the structure-activity study of Lys(Har)-Pro-Pro-Arg, discussing the design, synthesis, inhibitory activity, proteolytic stability and molecular modeling of the prepared derivatives. For two of the most active analogs the high proteolytic stability was also observed. These studies provide the next step for elucidating the optimal structure of the small peptidic inhibitors of VEGF165/NRP-1 interaction that could serve as research tools or be prospective drug candidates.


Subject(s)
Drug Design , Neuropilin-1/metabolism , Oligopeptides/chemistry , Oligopeptides/pharmacology , Protein Interaction Maps/drug effects , Vascular Endothelial Growth Factor A/metabolism , Amino Acid Sequence , Angiogenesis Inhibitors/blood , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/metabolism , Angiogenesis Inhibitors/pharmacology , Humans , Molecular Docking Simulation , Oligopeptides/blood , Oligopeptides/metabolism , Protein Binding/drug effects
3.
Peptides ; 94: 25-32, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28627371

ABSTRACT

Neuropilin-1 is considered as one of the key receptors responsible for signaling pathways involved in pathological angiogenesis necessary for tumor progression, therefore targeting of VEGF165 binding to NRP-1 could be a relevant strategy for antiangiogenic treatment. It was shown before that the VEGF165/NRP-1 interaction can be inhibited by short tetrapeptides with K/RXXR sequence. Here, we present a structure-activity relationship study of the systematic optimization of amino acid residues in positions 1-3 in the above tetrapeptides. All the 13 synthesized analogs possessed C-terminal arginine that is a necessary element for interaction with NRP-1. The obtained results of the inhibitory activity and modeling by molecular dynamics indicate that simultaneous interactions of the basic amino acid residues in position 1 and 4 (Arg) with Neuropilin-1 are crucial and their cooperation strongly affects the inhibitory activity. In addition, the binding strength is modulated by the flexibility of the peptide backbone (in the central part of the peptide), and the nature of the side chain of the amino acids at the second or third position. A dramatic decrease in the activity to the receptor is observed in flexible derivatives that are missing proline residues. The results described in this paper should prove useful for future studies aimed at establishing the best pharmacophore for inhibitors of VEGF165 binding to NRP-1.


Subject(s)
Neuropilin-1/agonists , Oligopeptides/pharmacology , Vascular Endothelial Growth Factor A/metabolism , Angiogenesis Inhibitors/pharmacology , Animals , Humans , Molecular Dynamics Simulation , Neuropilin-1/metabolism , Rats , Structure-Activity Relationship
4.
J Pept Sci ; 23(6): 445-454, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28466607

ABSTRACT

Neuropilin-1 has been found to be overexpressed in several kinds of malignant tumors, and it is postulated that its interaction with the vascular endothelial growth factor 165 leads to progression of tumor vascularization and growth. Several analogues (KxxR) with various conformational latitudes have been synthesized and found as inhibitors of NRP-1. Detailed insight provided by molecular dynamics simulation allowed forming a clear relationship between flexibility of xx part of the molecule and its inhibitory activity.


Subject(s)
Neuropilin-1/antagonists & inhibitors , Oligopeptides/pharmacology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Binding Sites/drug effects , Dose-Response Relationship, Drug , Humans , Models, Molecular , Neuropilin-1/chemistry , Oligopeptides/chemical synthesis , Oligopeptides/chemistry , Protein Conformation , Structure-Activity Relationship , Vascular Endothelial Growth Factor A/chemistry
5.
J Nanopart Res ; 19(5): 160, 2017.
Article in English | MEDLINE | ID: mdl-28503085

ABSTRACT

Superparamagnetic iron oxide-based nanoparticles (SPIONs) are promising carriers as targeted drug delivery vehicles, because they can be guided to their target with the help of an external magnetic field. Functionalization of nanoparticles' surface with molecules, which bind with high affinity to receptors on target tissue significantly facilitates delivery of coated nanoparticles to their targeted site. Here, we demonstrate conjugation of an antiangiogenic and antitumor peptide ATWLPPR (A7R) to SPIONs modified with sebacic acid (SPIONs-SA). Successful conjugation was confirmed by various analytical techniques (FTIR, SERS, SEM-EDS, TEM, TGA). Cell cytotoxicity studies, against two cell lines (HUVEC and MDA-MB-231) indicated that SPIONs modified with A7R reduced HUVEC cell viability at concentrations higher than 0.01 mg Fe/mL, in comparison to cells that were exposed to either the nanoparticles modified with sebacic acid or A7R peptide solely, what might be partially caused by a process of internalization.

6.
Bioorg Med Chem ; 25(2): 597-602, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27889287

ABSTRACT

Inhibition of angiogenesis is one of the most promising approaches in anticancer therapy. It was recently suggested that Neuropilin-1 (NRP-1) in tumour cells may serve as a separate receptor for Vascular Endothelial Growth Factor-165 (VEGF165) which is one of the main pro-angiogenic agents in the organism. Therefore molecules inhibiting VEGF165 binding to NRP-1 could be potential candidates for new antiangiogenic and anticancer drugs. Here we present a structure-activity relationship study of the peptide H-c[Lys-Pro-Glu]-Arg-OH which showed high inhibitory effect on VEGF165/NRP-1 binding (IC50=0.18µM) in our previous study. We report the design, synthesis, in vitro assays and docking analysis of four small cyclic peptides (14-,15-membered ring) and one bigger cyclic compound (30-membered ring). Our study shows that both the ring size and configuration of amino acid residues present in the structure are crucial for high inhibitory effect.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Neovascularization, Pathologic/drug therapy , Neuropilin-1/antagonists & inhibitors , Peptides, Cyclic/pharmacology , Vascular Endothelial Growth Factors/antagonists & inhibitors , Angiogenesis Inhibitors/chemical synthesis , Angiogenesis Inhibitors/chemistry , Dose-Response Relationship, Drug , Humans , Molecular Docking Simulation , Molecular Structure , Neuropilin-1/metabolism , Peptides, Cyclic/chemical synthesis , Peptides, Cyclic/chemistry , Structure-Activity Relationship , Tumor Cells, Cultured , Vascular Endothelial Growth Factors/metabolism
7.
Bioorg Med Chem ; 22(15): 4042-8, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24961874

ABSTRACT

Neuropilin-1 (NRP-1), one of the most important co-receptors of vascular endothelial growth factor-A (VEGF-A), increases its angiogenic action in several chronic diseases including cancer by increasing the activity of associated tyrosine kinase receptors, VEGFR1 and VEGFR2. Binding of VEGF-A to NRP-1 plays a critical role in pathological angiogenesis and tumor progression. Today, targeting this interaction is a validated approach to fight against angiogenesis-dependent diseases. Only anti-NRP-1 antibodies, peptide and peptidomimetic drug-candidates or hits have been developed thus far. In order to identify potent orally active small organic molecules various experimental and in silico approaches can be used. Here we report, novel promising small drug-like molecules disrupting the binding of VEGF-A165 to NRP-1. We carried out structure-based virtual screening experiments using the ChemBridge compound collection on the VEGF-A165 binding pocket of NRP-1. After docking and two rounds of similarity search computations, we identified 4 compounds that inhibit the biotinylated VEGF-A165 binding to recombinant NRP-1 with Ki of about 10 µM. These compounds contain a common chlorobenzyloxy alkyloxy halogenobenzyl amine scaffold that can serve as a base for further development of new NRP-1 inhibitors.


Subject(s)
Neuropilin-1/metabolism , Vascular Endothelial Growth Factor A/metabolism , Amines/chemistry , Amines/metabolism , Binding Sites , Drug Evaluation, Preclinical , Molecular Docking Simulation , Neuropilin-1/antagonists & inhibitors , Neuropilin-1/genetics , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Tertiary , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Structure-Activity Relationship , Vascular Endothelial Growth Factor A/antagonists & inhibitors
8.
Peptides ; 28(12): 2397-402, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17983687

ABSTRACT

Heptapeptide ATWLPPR (A7R), identified in our laboratory by screening a mutated phage library, was shown to bind specifically to neuropilin-1 (NRP-1) and then to selectively inhibit VEGF(165) binding to this receptor. In vivo, treatment with A7R resulted in decreasing breast cancer angiogenesis and growth. The present work is focused on structural characterization of A7R. Analogs of the peptide, obtained by substitution of each amino acid with alanine (alanine-scanning) or by amino acid deletion, have been systematically assayed to determine the relative importance of the side chains of each residue with respect to the inhibitory effect of A7R on VEGF(165) binding to NRP-1. We show here the importance of the C-terminal sequence LPPR and particularly the key role of C-terminal arginine. In solution, A7R displays significant secondary structure of the backbone adopting an extended conformation. However, the functional groups of arginine are very flexible in the absence of NRP-1 pointing to an induced fit upon binding to the receptor. A MD trajectory of the A7R/NRP-1 complex in explicit water, based on the recent tuftsin/NRP-1 crystal structure, has revealed the hydrogen-bonding network that contributes to A7R's binding activity.


Subject(s)
Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacology , Neuropilin-1/metabolism , Oligopeptides/chemistry , Oligopeptides/pharmacology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Amino Acid Sequence , Animals , In Vitro Techniques , Models, Molecular , Multiprotein Complexes , Protein Binding , Protein Conformation , Rats , Recombinant Proteins/metabolism , Structure-Activity Relationship , Thermodynamics , Vascular Endothelial Growth Factor A/metabolism
9.
J Biol Chem ; 281(49): 37844-52, 2006 Dec 08.
Article in English | MEDLINE | ID: mdl-17028197

ABSTRACT

Therapeutic induction of angiogenesis is a potential treatment for chronic ischemia. Heparan sulfate proteoglycans are known to play an important role by their interactions with proangiogenic growth factors such as vascular endothelial growth factor (VEGF). Low molecular weight fucoidan (LMWF), a sulfated polysaccharide from brown seaweeds that mimic some biological activities of heparin, has been shown recently to promote revascularization in rat critical hindlimb ischemia. In this report, we first used cultured human endothelial cells (ECs) to investigate the possible ability of LMWF to enhance the actions of VEGF(165). Data showed that LMWF greatly enhances EC tube formation in growth factor reduced matrigel. LMWF is a strong enhancer of VEGF(165)-induced EC chemotaxis, but not proliferation. In addition, LMWF has no effect on VEGF(121)-induced EC migration, a VEGF isoform that does not bind to heparan sulfate proteoglycans. Then, with binding studies using (125)I-VEGF(165), we observed that LMWF enhances the binding of VEGF(165) to recombinant VEGFR-2 and Neuropilin-1 (NRP1), but not to VEGFR-1. Surface plasmon resonance analysis showed that LMWF binds with high affinity to VEGF(165) (1.2 nm) and its receptors (5-20 nm), but not to VEGF(121). Pre-injection of LMWF on immobilized receptors shows that VEGF(165) has the highest affinity for VEGFR-2 and NRP1, as compared with VEGFR-1. Overall, the effects of LMWF were much more pronounced than those of LMW heparin. These findings suggested an efficient mechanism of action of LMWF by promoting VEGF(165) binding to VEGFR-2 and NRP1 on ECs that could help in stimulating therapeutic revascularization.


Subject(s)
Endothelial Cells/drug effects , Endothelial Cells/physiology , Neuropilin-1/metabolism , Polysaccharides/pharmacology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Cell Movement/drug effects , Cell Movement/physiology , Cells, Cultured , Humans , Molecular Weight , Neovascularization, Physiologic/drug effects , Neuropilin-1/genetics , Polysaccharides/chemistry , Protein Binding , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics
10.
Life Sci ; 79(25): 2370-81, 2006 Nov 17.
Article in English | MEDLINE | ID: mdl-16959272

ABSTRACT

Neuropilin-1 (NRP-1), a non-tyrosine kinase receptor of vascular endothelial growth factor-165 (VEGF165), was found expressed on endothelial and some tumor cells. Since its overexpression is correlated with tumor angiogenesis and progression, the targeting of NRP-1 could be a potential anti-cancer strategy. To explore this hypothesis, we identified a peptide inhibiting the VEGF165 binding to NRP-1 and we tested whether it was able to inhibit tumor growth and angiogenesis. To prove the target of peptide action, we assessed its effects on binding of radiolabeled VEGF165 to recombinant receptors and to cultured cells expressing only VEGFR-2 (KDR) or NRP-1. Antiangiogenic activity of the peptide was tested in vitro in tubulogenesis assays and in vivo in nude mice xenotransplanted in fat-pad with breast cancer MDA-MB-231 cells. Tumor volumes, vascularity and proliferation indices were determined. The selected peptide, ATWLPPR, inhibited the VEGF165 binding to NRP-1 but not to tyrosine kinase receptors, VEGFR-1 (flt-1) and KDR; nor did it bind to heparin. It diminished the VEGF-induced human umbilical vein endothelial cell proliferation and tubular formation on Matrigel and in co-culture with fibroblasts. Administration of ATWLPPR to nude mice inhibited the growth of MDA-MB-231 xenografts, and reduced blood vessel density and endothelial cell area but did not alter the proliferation indices of the tumor. In conclusion, ATWLPPR, a previously identified KDR-interacting peptide, was shown to inhibit the VEGF165 interactions with NRP-1 but not with KDR and to decrease the tumor angiogenesis and growth, thus validating, in vivo, NRP-1 as a possible target for antiangiogenic and antitumor agents.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antineoplastic Agents/therapeutic use , Mammary Neoplasms, Experimental/drug therapy , Neuropilin-1/metabolism , Peptide Fragments/therapeutic use , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Blotting, Western , CHO Cells , Cells, Cultured , Coculture Techniques , Cricetinae , Cross-Linking Reagents , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Immunoenzyme Techniques , Iodine Radioisotopes , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Nude , Neovascularization, Pathologic/drug therapy , Protein Binding , Umbilical Veins , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/metabolism , Xenograft Model Antitumor Assays
11.
Int J Oncol ; 28(2): 457-62, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16391801

ABSTRACT

A current target of cancer gene therapy is tumour vasculature. We present a gene-directed enzyme prodrug therapy (GDEPT) approach to target tumours in vivo by modifying endothelial cells (ECs) with the Escherichia coli nitroreductase (ntr) gene. Firstly, we isolated two ntr-transfected clones of the human umbilical vein endothelial cell line (HUV-EC-C/ntr+) that showed a differential sensitivity in vitro to the prodrug, dinitroaziridinylbenzamide (CB1954), with respect to untransfected HUV-EC-C cells (HUV-EC-C/ntr-). Then, these cells were injected subcutaneously into nude mice, either in association with the murine melanoma cell line, B16-F10 ('co-injected' groups), or into tumour-bearing animals ('post-injected' groups). After intratumoural injection, we demonstrated, using PCR analysis, that human ECs resided in the site of the injection without spreading to other organs, such as the liver or lung. After the treatment of mice with CB1954, we observed a prolonged survival of animals carrying the HUV-EC-C/ntr+ clones with respect to control animals injected with HUV-EC-C/ntr- cells. Significant differences in tumour growth were also observed and, after immuno-histological analysis, tumours carrying HUV-EC-C/ntr+ clones showed large areas of tumour necrosis, probably due to tumour ischemia, as well as the presence of major histocompatibility complex class-II (MHC-II) positive cells. Collectively, our data indicate that targeting of the tumour vasculature by this GDEPT strategy may be an efficient approach for cancer treatment in vivo, depending on two possible bystander mechanisms based on tumour ischemia and immune cell activation.


Subject(s)
Drug Delivery Systems , Endothelial Cells/metabolism , Melanoma, Experimental/blood supply , Nitroreductases/genetics , Animals , Antineoplastic Agents/pharmacology , Aziridines/pharmacology , Bystander Effect , Cell Survival , Cells, Cultured , Dose-Response Relationship, Drug , Endothelium, Vascular/drug effects , Escherichia coli/genetics , Genetic Therapy , Humans , Melanoma, Experimental/prevention & control , Mice , Mice, Nude , Neoplasm Transplantation , Neovascularization, Pathologic/prevention & control , Nitroreductases/metabolism , Prodrugs/pharmacology , Transfection
12.
Nucl Med Biol ; 31(5): 575-81, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15219275

ABSTRACT

Neuropilins (NRP) are receptors of angiogenic vascular endothelial growth factor (VEGF). Their overexpression was correlated with tumor angiogenesis and growth suggesting that their specific targeting could provide a new marker of tumor progression. Here, we observed in vitro that new (99m)Tc-labeled derivative of anti-VEGF heptapeptide, ATWLPPR, binds to NRP1 but not to NRP2. Our radiotracer is stable up to 24 h in human serum and in cysteine challenge. But, its too low affinity and too fast extraction indicate further improvement to give a successful imaging of tumor in vivo.


Subject(s)
Breast Neoplasms/diagnostic imaging , Breast Neoplasms/metabolism , Neuropilin-1/metabolism , Peptides/pharmacokinetics , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism , Animals , Cell Line, Tumor , Female , Humans , Isotope Labeling/methods , Male , Metabolic Clearance Rate , Mice , Mice, Nude , Neuropilin-1/chemistry , Organ Specificity , Peptides/chemical synthesis , Peptides/chemistry , Protein Binding , Radionuclide Imaging , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacokinetics , Rats , Reproducibility of Results , Sensitivity and Specificity , Technetium/chemistry , Technetium/pharmacology , Tissue Distribution
13.
Biochem Biophys Res Commun ; 311(4): 822-8, 2003 Nov 28.
Article in English | MEDLINE | ID: mdl-14623255

ABSTRACT

Tumor cells are elusive targets for standard anticancer chemotherapy due to their heterogeneity and genetic instability. On the other hand, proliferating host endothelial cells (ECs) are genetically stable and have a low mutational rate. Thus, antiangiogenic therapy directed against tumor's ECs should, in principle, improve the efficacy of antitumor therapy by inducing little or no drug resistance. Here we present a gene-directed enzyme prodrug therapy (GDEPT) strategy for targeting the tumor vasculature, using the Escherichia coli nitroreductase (ntr) gene delivery associated with the treatment with the prodrug CB1954. In a first time we demonstrated the ability of the ntr/CB1954 system to induce an apoptotic-mediated cell death on monolayer cultures of human umbilical vein ECs (HUV-EC-C). Then, when ntr-transfected HUV-EC-C cells (HUV-EC-C/ntr(+)) were associated in a three-dimensional (3-D) multicellular nodule model with untransfected B16-F10 murine melanoma cell line, we observed a CB1954-mediated bystander cell killing effect from endothelial to neighboring melanoma cells. To our knowledge, this is the first report indicating that GDEPT-based antiangiogenic targeting may be an effective approach for cancer treatment relied on the spreading of the bystander effect from endothelial to tumor cells.


Subject(s)
Aziridines , Bystander Effect , Endothelium, Vascular , Genetic Therapy , Melanoma , Nitroreductases , Animals , Humans , Mice , 3T3 Cells , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Aziridines/administration & dosage , Bystander Effect/genetics , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Coculture Techniques/methods , Dose-Response Relationship, Drug , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Escherichia coli/enzymology , Escherichia coli/genetics , Genetic Therapy/methods , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Nitroreductases/genetics , Nitroreductases/metabolism , Prodrugs/administration & dosage , Umbilical Veins/drug effects , Umbilical Veins/metabolism
14.
Br J Pharmacol ; 135(8): 1859-71, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11959788

ABSTRACT

1. Since the sodium phenylacetate (NaPa) was reported to enhance the inhibitory effect of carboxymethyl benzylamide dextran (CMDB) on the breast cancer growth, we performed the esterification of CMDB with NaPa to obtain a new drug carrying the characteristics of these two components. A new molecule, phenylacetate carboxymethyl benzylamide dextran, was named NaPaC. 2. We investigated in vitro and in vivo the effects of NaPaC on MCF-7ras cell growth as well as its apoptotic and antiangiogenic effects in comparison to NaPa and CMDB. In addition, we assessed in vitro the antiproliferative effects of these drugs on other breast cancer cells, including MDA-MB-231, MDA-MB-435 and MCF-7. 3. In vitro, NaPaC inhibited MCF-7ras cell proliferation by 40% at concentration lower than that of CMDB and NaPa (12 microM vs 73 microM and 10 mM). IC(50)s were 6 and 28 microM for NaPaC and CMDB, respectively. The similar results were obtained for three other breast cancer cell lines. NaPaC reduced the DNA replication and induced cell recruitment in G(0)/G(1) phase more efficiently than its components. Moreover, it induced a cell death at concentration 1000-fold lower than NaPa. 4. In vivo, CMDB (150 mg kg(-1)) and NaPa (40 mg kg(-1)) inhibited the MCF-7ras tumour growth by 37 and 57%, respectively, whereas NaPaC (15 mg kg(-1)) decreased tumour growth by 66% without toxicity. 5. NaPa or CMDB reduced the microvessel number in tumour by 50% after 7 weeks of treatment. NaPaC had the same effect after only 2 weeks. After 7 weeks, it generated a large necrosis area without detectable microvessels. In vitro, NaPaC inhibited human endothelial cell proliferation more efficiently than CMDB or NaPa. NaPaC interacts with vascular endothelial growth factor as observed by affinity electrophoresis. 6. NaPaC acts like NaPa and CMDB but in more potent manner than components used separately. Its antiproliferative, aponecrotic and anti-angiogenic actions make it a good candidate for a new anti-cancer drug.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Dextrans/pharmacology , Growth Inhibitors/pharmacology , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/pathology , 3T3 Cells , Angiogenesis Inhibitors/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Culture Media, Conditioned/pharmacology , Dextrans/chemistry , Dextrans/therapeutic use , Dose-Response Relationship, Drug , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Female , Growth Inhibitors/therapeutic use , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Necrosis , Phenylacetates/pharmacology , Phenylacetates/therapeutic use , Xenograft Model Antitumor Assays/statistics & numerical data
15.
Anticancer Res ; 22(6C): 3925-31, 2002.
Article in English | MEDLINE | ID: mdl-12553014

ABSTRACT

Sodium phenylacetate (NaPa) and some bisphosphonates demonstrated antiproliferative and proapoptotic properties against cancer. We have previously shown that NaPa inhibited cell proliferation of MCF7-ras tumor breast cells both in vitro and in vivo. On the other hand, bisphosphonate activities have only been demonstrated in vitro. Here we evaluated the antitumor effects of a new bisphosphonate, the phenylacetate-bisphosphonate (PaBp), on human breast cancer MCF7 and MCF7-ras cell lines, both in vitro and in vivo. To our knowledge, this is the first report indicating the use of a bisphosphonate derivative as a powerful cytostatic and cytotoxic agent, with proapoptotic and antiangiogenic properties on human breast cancer cells lines, with no animal toxicity.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Diphosphonates/pharmacology , Animals , Breast Neoplasms/pathology , Cell Division/drug effects , Female , Humans , Mice , Mice, Nude , Phenylacetates/pharmacology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...