Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Bioorg Med Chem Lett ; 23(22): 6068-72, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24095096

ABSTRACT

The development of RGD-based antagonist of αvß3 integrin receptor has enhanced the interest in PET probes to image this receptor for the early detection of cancer, to monitor the disease progression and the response to therapy. In this work, a novel prosthetic group (N-(4-fluorophenyl)pent-4-ynamide or FPPA) for the (18)F-labeling of an αvß3 selective RGD-peptide was successfully prepared. [(18)F]FPPA was obtained in three steps with a radiochemical yield of 44% (decay corrected). Conjugation to c(RGDfK(N3)) by the Cu(II) catalyzed Huisgen azido alkyne cycloaddition provided the [(18)F]FPPA-c(RGDfK) with a radiochemical yield of 29% (decay corrected), in an overall synthesis time of 140 min.


Subject(s)
Amides/chemistry , Fluorine Radioisotopes/chemistry , Integrin alphaVbeta3/analysis , Oligopeptides/chemistry , Radiopharmaceuticals/chemistry , Cell Line, Tumor , Click Chemistry , Fluorobenzenes/chemistry , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Integrin alphaVbeta3/chemistry , Isotope Labeling/methods , Models, Molecular , Molecular Imaging/methods , Positron-Emission Tomography/methods , Radiopharmaceuticals/chemical synthesis
2.
Recent Results Cancer Res ; 180: 83-101, 2010.
Article in English | MEDLINE | ID: mdl-20033379

ABSTRACT

Cells, including endothelial cells, continuously sense their surrounding environment and rapidly adapt to changes in order to assure tissues and organs homeostasis. The extracellular matrix (ECM) provides a physical scaffold for cell positioning and represents an instructive interface allowing cells to communicate over short distances. Cell surface receptors of the integrin family emerged through evolution as essential mediators and integrators of ECM-dependent communication. In preclinical studies, pharmacological inhibition of vascular integrins suppressed angiogenesis and inhibited tumor progression. alpha(V)beta(3) and alpha(V)beta(5) were the first integrins targeted to suppress tumor angiogenesis. Subsequently, additional integrins, in particular alpha(1)beta(1), alpha(2)beta(1), alpha(5)beta(1), and alpha(6)beta(4), emerged as potential therapeutic targets. Integrin inhibitors are currently tested in clinical trials for their safety and antiangiogenic/antitumor activity. In this chapter, we review the role of integrins in angiogenesis and present recent advances in the use of integrin antagonists as potential therapeutics in cancer and discuss future perspectives.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Integrins/antagonists & inhibitors , Neoplasms/blood supply , Neovascularization, Pathologic/drug therapy , Animals , Cell Adhesion , Humans , Integrins/physiology , Neovascularization, Pathologic/diagnostic imaging , Neovascularization, Pathologic/etiology , Positron-Emission Tomography
3.
Biochim Biophys Acta ; 1765(2): 155-77, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16263219

ABSTRACT

The formation of a 'tumor-associated vasculature', a process referred to as tumor angiogenesis, is a stromal reaction essential for tumor progression. Inhibition of tumor angiogenesis suppresses tumor growth in many experimental models, thereby indicating that tumor-associated vasculature may be a relevant target to inhibit tumor progression. Among the antiangiogenic molecules reported to date many are peptides and proteins. They include cytokines, chemokines, antibodies to vascular growth factors and growth factor receptors, soluble receptors, fragments derived from extracellular matrix proteins and small synthetic peptides. The polypeptide tumor necrosis factor (TNF, Beromun) was the first drug registered for the regional treatment of human cancer, whose mechanisms of action involved selective disruption of the tumor vasculature. More recently, bevacizumab (Avastin), an antibody against vascular endothelial growth factor (VEGF)-A, was approved as the first systemic antiangiogenic drug that had a significant impact on the survival of patients with advanced colorectal cancer, in combination with chemotherapy. Several additional peptides and antibodies with antiangiogenic activity are currently tested in clinical trials for their therapeutic efficacy. Thus, peptides, polypeptides and antibodies are emerging as leading molecules among the plethora of compounds with antiangiogenic activity. In this article, we will review some of these molecules and discuss their mechanism of action and their potential therapeutic use as anticancer agents in humans.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Neoplasms/blood supply , Neovascularization, Pathologic/drug therapy , Animals , Humans
4.
Endothelium ; 13(2): 113-35, 2006.
Article in English | MEDLINE | ID: mdl-16728329

ABSTRACT

The notion that tumor angiogenesis may have therapeutic implications in the control of tumor growth was introduced by Dr. Judah Folkman in 1971. The approval of Avastin in 2004 as the first antiangiogenic systemic drug to treat cancer patients came as a validation of this visionary concept and opened new perspectives to the treatment of cancer. In addition, this success boosted the field to the quest for new therapeutic targets and antiangiogenic drugs. Preclinical and clinical evidence indicate that vascular integrins may be valid therapeutic targets. In preclinical studies, pharmacological inhibition of integrin function efficiently suppressed angiogenesis and inhibited tumor progression. alphaVbeta3 and alphaVbeta5 were the first vascular integrins targeted to suppress tumor angiogenesis. Subsequent experiments revealed that at least four additional integrins (i.e., alpha1beta1, alpha2beta1, alpha5beta1, and alpha6beta4) might be potential therapeutic targets. In clinical studies low-molecular-weight integrin inhibitors and anti-integrin function-blocking antibodies demonstrated low toxicity and good tolerability and are now being tested in combination with radiotherapy and chemotherapy for anticancer activity in patients. In this article the authors review the role of integrins in angiogenesis, present recent development in the use of alphaVbeta3 and alpha5beta1 integrin antagonists as potential therapeutics in cancer, and discuss future perspectives.


Subject(s)
Integrins/physiology , Neoplasms/blood supply , Neoplasms/therapy , Animals , Clinical Trials as Topic , Endothelial Cells/pathology , Humans , Integrins/antagonists & inhibitors , Neovascularization, Pathologic/therapy
5.
Thromb Haemost ; 94(1): 155-66, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16113799

ABSTRACT

Integrin adhesion receptors consist of non-covalently linked alpha and beta subunits each of which contains a large extracellular domain, a single transmembrane domain and a short cytoplasmic tail. Engaged integrins recruit to focal structures globally termed adhesion complexes. The cytoplasmic domain of the beta subunit is essential for this clustering. beta1 and beta3 integrins can recruit at distinct cellular locations (i.e. fibrillar adhesions vs focal adhesions, respectively) but it is not clear whether individual beta subunit cytoplasmic and transmembrane domains are by themselves sufficient to drive orthotopic targeting to the cognate adhesion complex. To address this question, we expressed full-length beta3 transmembrane anchored cytoplasmic domains and truncated beta3 cytoplasmic domains as GFP-fusion constructs and monitored their localization in endothelial cells. Membrane-anchored full-length beta3 cytoplasmic domain and a beta3 mutant lacking the NXXY motif recruited to adhesion complexes, while beta3 mutants lacking the NPXY and NXXY motifs or the transmembrane domain did not. Replacing the natural beta subunit transmembrane domain with an unrelated (i.e. HLA-A2 alpha chain) transmembrane domain significantly reduced recruitment to adhesion complexes. Transmembrane anchored beta3 and cytoplasmic domain constructs, however, recruited without discrimination to beta1- and beta3-rich adhesions complexes. These findings demonstrate that membrane anchorage and the NPXY (but not the NXXY) motif are necessary for beta3 cytoplasmic domain recruitment to adhesion complexes and that the natural transmembrane domain actively contributes to this recruitment. The beta3 transmembrane and cytoplasmic domains alone are insufficient for orthotopic recruitment to cognate adhesion complexes.


Subject(s)
Cytoplasm/chemistry , Integrin beta3/chemistry , Amino Acid Motifs , Animals , Blotting, Western , Calcium Phosphates/pharmacology , Cattle , Cell Adhesion , Cell Membrane/metabolism , Cytoplasm/metabolism , Electroporation , Endothelium, Vascular/cytology , Fibroblasts/metabolism , Flow Cytometry , Green Fluorescent Proteins/metabolism , Humans , Integrins/chemistry , Microscopy, Confocal , Microscopy, Fluorescence , Mutation , Polymerase Chain Reaction , Protein Binding , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Transfection
6.
Thromb Haemost ; 94(5): 1060-70, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16363250

ABSTRACT

Expression of isolated beta integrin cytoplasmic domains in cultured endothelial cells was reported to induce cell detachment and death. To test whether cell death was the cause or the consequence of cell detachment, we expressed isolated integrin beta1 cytoplasmic and transmembrane domains (CH1) in cultured human umbilical vein endothelial cells (HUVEC), and monitored detachment, viability, caspase activation and signaling. CH1 expression induced dose-dependent cell detachment. At 24 h over 90% of CH1-expressing HUVEC were detached but largely viable (>85%). No evidence of pro-caspase-8,-3, and PARP cleavage or suppression of phosphorylation of ERK, PKB and Ikappa-B was observed. The caspase inhibitor z-VAD did not prevent cell detachment. At 48 h, however, CH1-expressing cells were over 50% dead. As a comparison trypsin-mediated detachment resulted in a time-dependent cell death, paralleled by caspase-3 activation and suppression of ERK, PKB and Ikappa-B phosphoyrylation at 24 h or later after detachment. HUVEC stimulation with agents that strengthen integrin-mediated adhesion (i.e. PMA, the Src inhibitor PP2 and COMP-Ang1) did not prevent CH1-induced detachment. Expression of CH1 in rat carotid artery endothelial cells in vivo caused endothelial cell detachment and increased nuclear DNA fragmentation among detached cells. A construct lacking the integrin cytoplasmic domain (CH2) had no effect on adhesion and cell viability in vitro and in vivo. These results demonstrate that isolated beta1 cytoplasmic domain expression induces caspase-independent detachment of viable endothelial cells and that death is secondary to detachment (i.e. anoikis). They also reveal an essential role for integrins in the adhesion and survival of quiescent endothelial cells in vivo.


Subject(s)
Anoikis/physiology , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Integrin beta1/genetics , Animals , Carotid Arteries/cytology , Caspase 3 , Caspase 8 , Caspases/metabolism , Cell Adhesion/physiology , Cell Survival/physiology , Cells, Cultured , DNA Fragmentation/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression/physiology , Humans , I-kappa B Proteins/metabolism , Integrin beta1/chemistry , MAP Kinase Signaling System/physiology , Protein Structure, Tertiary , Protein Subunits/chemistry , Protein Subunits/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Umbilical Veins/cytology
7.
Clin Cancer Res ; 18(16): 4365-74, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22711708

ABSTRACT

PURPOSE: Local breast cancer relapse after breast-saving surgery and radiotherapy is associated with increased risk of distant metastasis formation. The mechanisms involved remain largely elusive. We used the well-characterized 4T1 syngeneic, orthotopic breast cancer model to identify novel mechanisms of postradiation metastasis. EXPERIMENTAL DESIGN: 4T1 cells were injected in 20 Gy preirradiated mammary tissue to mimic postradiation relapses, or in nonirradiated mammary tissue, as control, of immunocompetent BALB/c mice. Molecular, biochemical, cellular, histologic analyses, adoptive cell transfer, genetic, and pharmacologic interventions were carried out. RESULTS: Tumors growing in preirradiated mammary tissue had reduced angiogenesis and were more hypoxic, invasive, and metastatic to lung and lymph nodes compared with control tumors. Increased metastasis involved the mobilization of CD11b(+)c-Kit(+)Ly6G(high)Ly6C(low)(Gr1(+)) myeloid cells through the HIF1-dependent expression of Kit ligand (KitL) by hypoxic tumor cells. KitL-mobilized myeloid cells homed to primary tumors and premetastatic lungs, to give rise to CD11b(+)c-Kit(-) cells. Pharmacologic inhibition of HIF1, silencing of KitL expression in tumor cells, and inhibition of c-Kit with an anti-c-Kit-blocking antibody or with a tyrosine kinase inhibitor prevented the mobilization of CD11b(+)c-Kit(+) cells and attenuated metastasis. C-Kit inhibition was also effective in reducing mobilization of CD11b(+)c-Kit(+) cells and inhibiting lung metastasis after irradiation of established tumors. CONCLUSIONS: Our work defines KitL/c-Kit as a previously unidentified axis critically involved in promoting metastasis of 4T1 tumors growing in preirradiated mammary tissue. Pharmacologic inhibition of this axis represents a potential therapeutic strategy to prevent metastasis in breast cancer patients with local relapses after radiotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Proto-Oncogene Proteins c-kit/metabolism , Signal Transduction/drug effects , Stem Cell Factor/metabolism , Animals , Antineoplastic Agents/administration & dosage , CD11b Antigen/metabolism , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Hypoxia , Hypoxia-Inducible Factor 1/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/radiotherapy , Mice , Mice, Inbred C57BL , Neoplasm Metastasis , Neoplasm Recurrence, Local , Neovascularization, Pathologic/drug therapy , Tumor Burden
8.
Cancer Res ; 71(11): 3781-91, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21507936

ABSTRACT

Tumor-mobilized bone marrow-derived CD11b(+) myeloid cells promote tumor angiogenesis, but how and when these cells acquire proangiogenic properties is not fully elucidated. Here, we show that CD11b(+) myelomonocytic cells develop proangiogenic properties during their differentiation from CD34(+) hematopoietic progenitors and that placenta growth factor (PlGF) is critical in promoting this education. Cultures of human CD34(+) progenitors supplemented with conditioned medium from breast cancer cell lines or PlGF, but not from nontumorigenic breast epithelial lines, generate CD11b(+) cells capable of inducing endothelial cell sprouting in vitro and angiogenesis in vivo. An anti-Flt-1 mAb or soluble Flt-1 abolished the generation of proangiogenic activity during differentiation from progenitor cells. Moreover, inhibition of metalloproteinase activity, but not VEGF, during the endothelial sprouting assay blocked sprouting induced by these proangiogenic CD11b(+) myelomonocytes. In a mouse model of breast cancer, circulating CD11b(+) cells were proangiogenic in the sprouting assays. Silencing of PlGF in tumor cells prevented the generation of proangiogenic activity in circulating CD11b(+) cells, inhibited tumor blood flow, and slowed tumor growth. Peripheral blood of breast cancer patients at diagnosis, but not of healthy individuals, contained elevated levels of PlGF and circulating proangiogenic CD11b(+) myelomonocytes. Taken together, our results show that cancer cells can program proangiogenic activity in CD11b(+) myelomonocytes during differentiation of their progenitor cells in a PlGF-dependent manner. These findings impact breast cancer biology, detection, and treatment.


Subject(s)
Breast Neoplasms/blood supply , CD11b Antigen/biosynthesis , Hematopoietic Stem Cells/pathology , Monocytes/pathology , Pregnancy Proteins/immunology , Adult , Aged , Animals , Antigens, CD34/biosynthesis , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Differentiation/physiology , Cell Line, Tumor , Female , Fetal Blood/cytology , Hematopoietic Stem Cells/immunology , Humans , Mice , Mice, Inbred BALB C , Middle Aged , Monocytes/immunology , Placenta Growth Factor , Pregnancy , Pregnancy Proteins/pharmacology , Recombinant Proteins/pharmacology
9.
PLoS One ; 4(2): e4449, 2009.
Article in English | MEDLINE | ID: mdl-19212436

ABSTRACT

Cilengitide is a high-affinity cyclic pentapeptdic alphaV integrin antagonist previously reported to suppress angiogenesis by inducing anoikis of endothelial cells adhering through alphaVbeta3/alphaVbeta5 integrins. Angiogenic endothelial cells express multiple integrins, in particular those of the beta1 family, and little is known on the effect of cilengitide on endothelial cells expressing alphaVbeta3 but adhering through beta1 integrins. Through morphological, biochemical, pharmacological and functional approaches we investigated the effect of cilengitide on alphaVbeta3-expressing human umbilical vein endothelial cells (HUVEC) cultured on the beta1 ligands fibronectin and collagen I. We show that cilengitide activated cell surface alphaVbeta3, stimulated phosphorylation of FAK (Y(397) and Y(576/577)), Src (S(418)) and VE-cadherin (Y(658) and Y(731)), redistributed alphaVbeta3 at the cell periphery, caused disappearance of VE-cadherin from cellular junctions, increased the permeability of HUVEC monolayers and detached HUVEC adhering on low-density beta1 integrin ligands. Pharmacological inhibition of Src kinase activity fully prevented cilengitide-induced phosphorylation of Src, FAK and VE-cadherin, and redistribution of alphaVbeta3 and VE-cadherin and partially prevented increased permeability, but did not prevent HUVEC detachment from low-density matrices. Taken together, these observations reveal a previously unreported effect of cilengitide on endothelial cells namely its ability to elicit signaling events disrupting VE-cadherin localization at cellular contacts and to increase endothelial monolayer permeability. These effects are potentially relevant to the clinical use of cilengitide as anticancer agent.


Subject(s)
Antigens, CD/metabolism , Cadherins/metabolism , Endothelial Cells/drug effects , Integrin alphaVbeta3/antagonists & inhibitors , Intercellular Junctions/drug effects , Snake Venoms/pharmacology , Animals , Antigens, CD/genetics , Cadherins/genetics , Cell Adhesion/drug effects , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Enzyme Induction , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , Integrin alphaVbeta3/metabolism , Integrin beta1/metabolism , Intercellular Junctions/metabolism , Permeability , Phosphorylation , Pyrimidines/metabolism , Pyrroles/metabolism , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
10.
Cancer Res ; 68(18): 7323-31, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18794119

ABSTRACT

Radiotherapy is widely used to treat human cancer. Patients locally recurring after radiotherapy, however, have increased risk of metastatic progression and poor prognosis. The clinical management of postradiation recurrences remains an unresolved issue. Tumors growing in preirradiated tissues have an increased fraction of hypoxic cells and are more metastatic, a condition known as tumor bed effect. The transcription factor hypoxia inducible factor (HIF)-1 promotes invasion and metastasis of hypoxic tumors, but its role in the tumor bed effect has not been reported. Here, we show that tumor cells derived from SCCVII and HCT116 tumors growing in a preirradiated bed, or selected in vitro through repeated cycles of severe hypoxia, retain invasive and metastatic capacities when returned to normoxia. HIF activity, although facilitating metastatic spreading of tumors growing in a preirradiated bed, is not essential. Through gene expression profiling and gain- and loss-of-function experiments, we identified the matricellular protein CYR61 and alphaVbeta5 integrin as proteins cooperating to mediate these effects. The anti-alphaV integrin monoclonal antibody 17E6 and the small molecular alphaVbeta3/alphaVbeta5 integrin inhibitor EMD121974 suppressed invasion and metastasis induced by CYR61 and attenuated metastasis of tumors growing within a preirradiated field. These results represent a conceptual advance to the understanding of the tumor bed effect and identify CYR61 and alphaVbeta5 integrin as proteins that cooperate to mediate metastasis. They also identify alphaV integrin inhibition as a potential therapeutic approach for preventing metastasis in patients at risk for postradiation recurrences.


Subject(s)
Carcinoma, Squamous Cell/pathology , Colorectal Neoplasms/pathology , Immediate-Early Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Radiation Injuries, Experimental/pathology , Receptors, Vitronectin/metabolism , Animals , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Hypoxia/radiation effects , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Cysteine-Rich Protein 61 , Gene Expression Profiling , HCT116 Cells , Humans , Immediate-Early Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/biosynthesis , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Mice , Neoplasm Invasiveness , Radiation Injuries, Experimental/metabolism , Receptors, Vitronectin/antagonists & inhibitors , Receptors, Vitronectin/biosynthesis , Stromal Cells/pathology , Stromal Cells/radiation effects
SELECTION OF CITATIONS
SEARCH DETAIL