Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Int J Cancer ; 134(2): 467-77, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-23818211

ABSTRACT

Preclinical studies with recombinant murine interleukin 4 (IL4) in models of cancer have shown potent tumor growth inhibition. However, systemic administration of human IL4 to cancer patients exhibited modest antitumor activity and considerable toxicities. To improve the therapeutic index and reduce side effects of this cytokine, we developed of a novel "immunocytokine" based on sequential fusion of murine IL4 with the antibody fragment F8 (specific to the alternatively spliced extra-domain A of fibronectin, a marker for tumor-angiogenesis) in diabody format. The resulting fusion protein, termed F8-IL4, retained full antigen-binding activity and cytokine bioactivity and was able to selectively localize on solid tumors in vivo. When used as single agent, F8-IL4 inhibited tumor growth in three different immunocompetent murine cancer models (F9 teratocarcinoma, CT26 colon carcinoma and A20 lymphoma). Furthermore, F8-IL4 showed synergistic effects when coadministered with immunocytokines based on IL2 and IL12. Indeed, combination therapy with an IL12-based immunocytokine yielded complete tumor eradication, in spite of the fact that IL4 and IL12 display opposite immunological mechanisms of action in terms of their polarization of T-cell based responses. No weight loss or any signs of toxicity were observed in treated mice, both in monotherapy and in combination, indicating a good tolerability of the immunocytokine treatment. Interestingly, mice cured from CT26 tumors acquired a durable protective antitumor immunity. Depletion experiments indicated that the antitumor activity was mediated by CD8+ T cells and by NK cells.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Colonic Neoplasms/prevention & control , Disease Models, Animal , Fibrosarcoma/prevention & control , Interleukin-12/immunology , Interleukin-4/immunology , Lymphoma/prevention & control , Neovascularization, Pathologic/prevention & control , Teratocarcinoma/prevention & control , Animals , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal, Humanized , Colonic Neoplasms/blood supply , Colonic Neoplasms/immunology , Female , Fibrosarcoma/blood supply , Fibrosarcoma/immunology , Fluorescent Antibody Technique , Humans , Immunoconjugates , Interleukin-12/administration & dosage , Interleukin-4/administration & dosage , Lymphoma/immunology , Lymphoma/pathology , Male , Mice , Mice, Inbred BALB C , Single-Chain Antibodies/administration & dosage , Teratocarcinoma/blood supply , Teratocarcinoma/immunology , Testicular Neoplasms/blood supply , Testicular Neoplasms/immunology , Testicular Neoplasms/prevention & control , Tissue Distribution
2.
Cancer Cell ; 3(1): 75-88, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12559177

ABSTRACT

Inactivation of the von Hippel-Lindau (VHL) gene is associated with the development of highly vascularized tumors. pVHL targets the alpha subunits of hypoxia inducible factor (HIF) for ubiquitin-mediated degradation in an oxygen-dependent manner. Although pVHL-deficient tumor cell lines demonstrate constitutive stabilization and activation of HIF, it has yet to be shown that loss of murine Vhl alone is sufficient to dysregulate HIF. We utilized a genetic approach to demonstrate that loss of Vhl is sufficient not only to stabilize HIF-alpha subunits under normoxia, but also fully activate HIF-mediated responses. These studies have implications for the hierarchy of signaling events leading to HIF stabilization, nuclear translocation, and target gene expression. We further demonstrate that loss of murine Vhl does not promote teratocarcinoma growth, indicating that other genetic changes must occur to facilitate Vhl-mediated tumorigenesis.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Ligases/genetics , Neoplasms, Experimental/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Transcription Factors , Tumor Suppressor Proteins , Ubiquitin-Protein Ligases , von Hippel-Lindau Disease/genetics , Animals , Apoptosis/genetics , Gene Expression Regulation, Neoplastic , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Ligases/deficiency , Mice , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Protein Subunits/genetics , Protein Subunits/metabolism , Teratocarcinoma/blood supply , Teratocarcinoma/metabolism , Teratocarcinoma/pathology , Tumor Cells, Cultured , Von Hippel-Lindau Tumor Suppressor Protein
3.
Arterioscler Thromb Vasc Biol ; 30(7): 1378-88, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20431070

ABSTRACT

OBJECTIVE: Vezf1 encodes an early zinc finger transcription factor that is essential for normal vascular development and functions in a dose-dependent manner. Here, we investigated the role of Vezf1 during processes of endothelial cell differentiation and maturation by studying mutant Vezf1 embryonic stem (ES) cells using the in vitro embryoid body differentiation model and the in vivo teratocarcinoma model. METHODS AND RESULTS: Vezf1-/- ES cell-derived embryoid bodies failed to form a well-organized vascular network and showed dramatic vascular sprouting defects. Our results indicate that the retinol pathway is an important mediator of Vezf1 function and that loss of Vezf1 results in reduced retinol/vitamin A signaling and aberrant extracellular matrix (ECM) formation. Unexpectedly, we also uncovered defects during in vitro differentiation of Vezf1-/- ES cells along hematopoietic cell lineages. Vezf1-/- ES cell-derived teratocarcinomas were able to spontaneously differentiate into cell types of all 3 germ layers. However, histological and immunohistochemical examination of these tumors showed decreased cell proliferation, delayed differentiation, and large foci of cells with extensive deposition of ECM. Embryoid bodies and teratocarcinomas derived from heterozygous ES cells displayed an intermediate phenotype. CONCLUSIONS: Together, these results suggest that Vezf1 is involved in early differentiation processes of the vasculature by regulating cell differentiation, proliferation, and ECM distribution and deposition.


Subject(s)
Cell Differentiation/genetics , Embryonic Stem Cells/metabolism , Endothelial Cells/metabolism , Gene Expression Regulation, Developmental , Kruppel-Like Transcription Factors/genetics , Mutation , Neovascularization, Physiologic/genetics , Animals , Cell Adhesion , Cell Proliferation , Cells, Cultured , DNA-Binding Proteins , Embryonal Carcinoma Stem Cells/metabolism , Embryonic Stem Cells/transplantation , Endothelial Cells/transplantation , Extracellular Matrix/metabolism , Genotype , Hematopoietic Stem Cells/metabolism , Kruppel-Like Transcription Factors/metabolism , Male , Mice , Mice, Nude , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Phenotype , Signal Transduction , Teratocarcinoma/blood supply , Teratocarcinoma/genetics , Teratocarcinoma/metabolism , Time Factors , Transcription Factors , Vitamin A/metabolism
4.
Int J Androl ; 33(6): 765-74, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20497259

ABSTRACT

Testicular germ cell tumours (TGCTs) represent about 2% of male malignancies, being the most common cancer among adolescents and young adults. As in most neoplasias, TGCTs show a chaotic vascular architecture, altered blood supply and over-expression of pro-angiogenic factors, aspects closely related to tumour overgrowth and metastasis. Following this trend, our laboratory has analysed the effect of the hypoxic tumour microenvironment on cancer stem cells, particularly the expression of factors related to vascularization, such as matrix metalloproteinases, adhesion molecules, vascular endothelial growth factors (VEGF) and VEGF receptors. This review also summarizes our present knowledge on vascularization in the normal and neoplastic testis, the potential role of the factors involved in TGCT neovascularization and their importance as possible therapeutic targets.


Subject(s)
Neoplasms, Germ Cell and Embryonal/blood supply , Neovascularization, Pathologic , Teratocarcinoma/blood supply , Testicular Neoplasms/blood supply , Adolescent , Adult , Angiogenesis Inhibitors/therapeutic use , Animals , Biomarkers, Tumor/blood , Cadherins/physiology , Cell Adhesion Molecules/physiology , Embryonic Stem Cells/transplantation , Humans , Integrins/physiology , Male , Matrix Metalloproteinase 14/physiology , Matrix Metalloproteinases/physiology , Neoplasm Metastasis/physiopathology , Testicular Neoplasms/pathology , Testis/blood supply , Vascular Endothelial Growth Factor A/physiology
5.
Br J Cancer ; 101(1): 64-70, 2009 Jul 07.
Article in English | MEDLINE | ID: mdl-19513074

ABSTRACT

BACKGROUND: Carcinoma in situ (CIS) of the testis is considered to be a precancerous germinal cell lesion, but the precise cellular and molecular mechanisms underlying transformation of CIS into invasive pluripotent cancer cells remain to be elucidated. Moreover, a satisfactory animal model for the experimental study of germinal tumours has not been developed to date. METHODS: We have developed a tumour model that involves the microinjection of green fluorescent protein-labelled embryonic stem (ES) cells (which are functionally equivalent to CIS cells) into syngenic mouse seminiferous tubules, a unique cell microenvironment in which germinal cells mature and CIS arise. To characterise the vascularisation of teratocarcinomas, which arise after cell transplant, we used immunohistochemistry, together with a qualitative and quantitative analysis of scanning electron microscopy images of corrosion casting samples. RESULTS: Embryonic stem cells transplanted into seminiferous tubules did not differentiate into germinal cells, but rather they behaved as invasive embryonal carcinoma (EC) stem cells. The vascular pattern of the experimental teratocarcinomas showed a highly disorganised architecture, and some of the neoplastic capillaries were derived, at least in part, from the original transplanted ES cells. CONCLUSION: The transplantation of pluripotent ES cells into seminiferous tubules efficiently recapitulates the early stages of development of teratocarcinomas. Consequently, this method constitutes a novel in vivo model to study the mechanisms of invasion and progression of experimental germinal tumours.


Subject(s)
Embryonal Carcinoma Stem Cells/pathology , Pluripotent Stem Cells/pathology , Seminiferous Tubules/pathology , Teratocarcinoma/blood supply , Teratocarcinoma/pathology , Testicular Neoplasms/blood supply , Testicular Neoplasms/pathology , Animals , Cell Transformation, Neoplastic/pathology , Male , Mice , Neovascularization, Pathologic/pathology , Pluripotent Stem Cells/transplantation , Stem Cell Transplantation
6.
Cancer Res ; 67(10): 4940-8, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17510424

ABSTRACT

Tumor-targeting immunocytokines represent a new class of anticancer pharmaceutical agents, which often display a superior therapeutic index compared with the corresponding unconjugated cytokines. In this article, we have studied the anticancer properties of interleukin-15 (IL-15) and granulocyte macrophage colony-stimulating factor (GM-CSF), fused to the human antibody fragment scFv(L19), specific to the EDB domain of fibronectin, a marker of angiogenesis. The immunocytokines L19-IL-15 and L19-GM-CSF were expressed in mammalian cells and purified to homogeneity, revealing no loss of cytokine activity in in vitro assays. Furthermore, the ability of the two immunocytokines to selectively localize to tumors in vivo was confirmed by biodistribution analysis with radioiodinated protein preparations. L19-IL-15 and L19-GM-CSF displayed a potent antitumor activity both in s.c. and in metastatic F9 and C51 murine models of cancer in immunocompetent mice. This therapeutic action was superior compared with IL-15-based and GM-CSF-based fusion proteins, containing antibodies of irrelevant specificity in the mouse, which were used as non-tumor-targeting controls. For both L19-IL-15 and L19-GM-CSF immunocytokines, CD8(+) T cells seemed to mostly contribute to the therapeutic action as shown by in vivo cell depletion experiments. The results presented in this article are of clinical significance, considering the fact that the sequence of EDB is identical in mouse and man and that the tumor-targeting ability of the L19 antibody has been extensively shown in clinical trials in patients with cancer.


Subject(s)
Adenocarcinoma/therapy , Colonic Neoplasms/therapy , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Immunoconjugates/pharmacology , Interleukin-15/administration & dosage , Teratocarcinoma/therapy , Adenocarcinoma/blood supply , Adenocarcinoma/immunology , Animals , Cell Growth Processes/drug effects , Cell Growth Processes/immunology , Cloning, Molecular , Colonic Neoplasms/blood supply , Colonic Neoplasms/immunology , Female , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacokinetics , Immunoconjugates/genetics , Immunoconjugates/immunology , Immunoconjugates/pharmacokinetics , Immunoglobulin Fragments/genetics , Immunoglobulin Fragments/immunology , Interleukin-15/genetics , Interleukin-15/pharmacokinetics , Mice , Mice, Inbred BALB C , Neoplasm Metastasis , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/therapy , T-Lymphocytes/immunology , Teratocarcinoma/blood supply , Teratocarcinoma/immunology , Tissue Distribution
7.
Int J Cancer ; 122(11): 2405-13, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18271006

ABSTRACT

The alternatively spliced extra-domain B of fibronectin is one of the best characterized markers of tumor angiogenesis. Similarly, the extra-domain A (EDA), which can also be inserted in the fibronectin transcript by a mechanism of alternative splicing, has been shown to preferentially accumulate around new blood vessels in certain tumors, but this antigen has not been investigated so far as a target for antibody-based biomolecular intervention. We here describe the generation of 3 human monoclonal antibodies (named F8, B7 and D5), which recognize the same epitope of EDA, but which differ in terms of their dissociation constant to the human antigen (K(D) = 3.1, 16 and 17 nM, measured for monomeric preparations of the F8, B7 and D5 antibodies, respectively, in recombinant scFv format). When the 3 antibody fragments were cloned and expressed with a 5 amino acid linker, the 3 resulting homodimeric antibody preparations displayed comparable tumor: organ ratios in quantitative biodistribution studies, performed in immunocompetent 129SvEv mice, bearing subcutaneous syngeneic F9 murine tumors. The percent injected dose per gram (%ID/g) values in tumors 24 hr after intravenous injection were 9.3, 10.2 and 13 for F8, B7 and D5, respectively. The F8 antibody may serve as useful building block for the development of antibody-based targeted anti-cancer therapeutics. Preclinical and clinical investigations are facilitated by the fact that F8 recognizes the human and mouse antigen with comparable affinity, and by the observation that EDA over-expression is detectable not only in solid tumors, but also in hematological malignancies.


Subject(s)
Alternative Splicing , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Fibronectins/immunology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/immunology , Teratocarcinoma/blood supply , Animals , Antibodies, Monoclonal/isolation & purification , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Antibody Affinity , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , DNA Primers , Fibronectins/genetics , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic , Humans , Immunoblotting , Immunohistochemistry , Male , Mice , Protein Structure, Tertiary , Teratocarcinoma/immunology , Testicular Neoplasms/blood supply , Tissue Distribution
8.
Nat Biotechnol ; 15(12): 1271-5, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9359110

ABSTRACT

The oncofetal fibronectin (B-FN) isoform is present in vessels of neoplastic tissues during angiogenesis but not in mature vessels. B-FN could therefore provide a target for diagnostic imaging and therapy of cancer. Phage display libraries have been used to isolate human antibody fragments with pan-species recognition of this isoform. We describe the use of these fragments in nude mice to target an aggressive tumor (grafted F9 murine teratocarcinoma). Imaging in real time was done by infrared photodetection of a chemically coupled fluorophore. The targeting was improved by use of affinity-matured fragments with low kinetic dissociation rates (koff = 1.5 x 10(-4) s-1) and also by engineering dimeric fragments via a C-terminal amphipathic helix.


Subject(s)
Fibronectins , Immunoglobulin Fragments , Neovascularization, Pathologic/diagnosis , Amino Acid Sequence , Animals , Base Sequence , DNA Primers , Fibronectins/metabolism , Humans , Immunoglobulin Fragments/metabolism , Mice , Mice, Nude , Molecular Sequence Data , Recombinant Proteins/metabolism , Teratocarcinoma/blood supply , Teratocarcinoma/diagnosis
9.
Structure ; 7(4): 381-90, 1999 Apr 15.
Article in English | MEDLINE | ID: mdl-10196121

ABSTRACT

BACKGROUND: The process of angiogenesis (i.e. the formation of new blood vessels from pre-existing ones) is fundamental to physiological processes such as reproduction, development and repair, as well as to pathological conditions such as tumor progression, rheumathoid arthritis and ocular disorders. The oncofoetal ED-B domain, a specific marker of angiogenesis, consists of 91 amino acid residues that are inserted by alternative splicing into the fibronectin (FN) molecule. RESULTS: The NMR structure of the ED-B domain is reported and reveals important differences from other FN type III domains. A comparison of the ED-B domain with the crystal structure of a four-domain FN fragment shows the novel features of ED-B to be located in loop regions that are buried at interdomain interfaces, and which therefore largely determine the global shape of the FN molecule. The negatively charged amino acids in this highly acidic protein are uniformly distributed over the molecular surface, with the sole exception of a solvent-exposed hydrophobic patch that represents a potential specific recognition site. Epitope mapping with 82 decapeptides that span the ED-B sequence revealed that three ED-B-specific monoclonal antibodies, which selectively target newly forming blood vessels in tumor-bearing mice, bind to adjacent regions on the ED-B surface. CONCLUSIONS: The NMR structure enables the identification of a large surface area of the ED-B domain that appears to be accessible in vivo, opening up new diagnostic and therapeutic opportunities. Furthermore, the mapping of specific monoclonal antibodies to the three-dimensional structure of the ED-B domain, and their use in angiogenesis inhibition experiments, provides a basis for further investigation of the role of the ED-B domain in the formation of new blood vessels.


Subject(s)
Antigens, Neoplasm/chemistry , Fibronectins/chemistry , Magnetic Resonance Spectroscopy , Neovascularization, Pathologic/metabolism , Protein Conformation , Protein Isoforms/chemistry , Protein Structure, Tertiary , Amino Acid Sequence , Animals , Antigens, Neoplasm/immunology , Antigens, Neoplasm/physiology , Binding Sites , Biomarkers , Epitopes/immunology , Fibronectins/immunology , Fibronectins/physiology , Glycation End Products, Advanced , Glycosylation , Humans , Mice , Mice, Nude , Models, Molecular , Molecular Sequence Data , Neoplasm Transplantation , Protein Isoforms/immunology , Protein Isoforms/physiology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/physiology , Teratocarcinoma/blood supply , Teratocarcinoma/metabolism , Teratocarcinoma/pathology
10.
Cancer Res ; 59(2): 347-52, 1999 Jan 15.
Article in English | MEDLINE | ID: mdl-9927045

ABSTRACT

The formation of new blood vessels (angiogenesis) is an important step in tumor progression. Molecules capable of selectively targeting markers of angiogenesis may offer opportunities for the in vivo imaging of aggressive tumors and for the delivery of toxic agents to the tumoral vasculature. Using antibody phage display libraries and combinatorial mutagenesis, we isolated single-chain Fv antibody fragments, which recognize with different affinities the same epitope of the ED-B domain of fibronectin, a marker of angiogenesis. Two single-chain Fv fragments, E1 and L19, with dissociation constants of 41 nM and 0.054 nM, respectively, were investigated for their ability to target F9 murine teratocarcinoma grafted s.c. in nude mice when injected i.v. in either monomeric or homodimeric form (Mr 27,000 and 54,000, respectively). Biodistribution studies, performed at two time points (4 h and 24 h) with radiolabeled samples, showed that the higher affinity antibody targets the tumor significantly better than the lower affinity one, in terms both of tumor:organ ratios and of the amounts of antibody delivered to the tumor. In particular, more than 20% of the injected dose of dimeric L19 accumulated per gram of tumor at 4 h; the tumor:organ ratios at 4 h and 24 h were in the (2.1-8.6):1 and (10.3-29.4):1 range, respectively. This study demonstrates that, although vasculature represents only a small fraction of the total tumor mass, anti-ED-B antibodies can selectively target tumors in vivo and that this process is particularly efficient if very high-affinity binders are used.


Subject(s)
Fibronectins/immunology , Immunoglobulin Fragments , Neovascularization, Pathologic/diagnostic imaging , Teratocarcinoma/blood supply , Animals , Immunoglobulin Fragments/metabolism , Iodine Radioisotopes , Male , Mice , Mice, Nude , Radionuclide Imaging , Recombinant Proteins/pharmacokinetics , Teratocarcinoma/diagnostic imaging , Tissue Distribution
11.
Cancer Res ; 62(10): 2962-70, 2002 May 15.
Article in English | MEDLINE | ID: mdl-12019179

ABSTRACT

In tumors, rapid cell proliferation associated with deficient vascularization leads to areas of hypoxia. Tumor hypoxia has direct consequences on clinical and prognostic parameters and is a potential therapeutic target. The hypoxic response depends critically on hypoxia-inducible factor-1alpha (HIF-1alpha) in pathological (e.g., tumorigenesis) as well as physiological (e.g., development and wound healing) processes. By s.c. injection of HIF-1alpha(-/-) embryonic stem (ES) cells in nude mice, we were able to demonstrate the role of HIF-1alpha in cell differentiation of teratocarcinomas. HIF-1alpha(+/+) tumors grow fast and preferentially form neuronal tissue, whereas HIF-1alpha(-/-) tumors show delayed growth and favorably form mesenchyme-derived tissue. Mixing wild-type and HIF-1alpha(-/-) ES cells in the same tumor at a ratio as low as 1:100, we showed that HIF-1alpha(+/+) cells can rescue the growth of mixed tumors although these tumors are not significantly different phenotypically or genotypically from the original HIF-1alpha(-/-) tumors. Interestingly, these results are not restricted to teratocarcinomas: they were confirmed with mixtures of Hepa1/Hepa1C4 cells (where HIF-1beta is mutated), demonstrating that growth changes are not related to differences in differentiation observed within teratocarcinomas. We also showed that despite lower mRNA expression, vascular endothelial growth factor protein status in HIF-1alpha(-/-) and mixed tumors does not significantly differ from the HIF-1alpha(+/+) tumors. Moreover, we demonstrated that tumor vascularization remains proportional to vascular endothelial growth factor protein levels, but that hypoxic up-regulation of this growth factor is not the decisive factor influencing tumor growth. Differences in levels of apoptosis are not responsible for alteration in growth because poly(ADP-ribose) polymerase cleavage, a hallmark of the apoptotic process, was similar in HIF-1alpha(+/+), HIF-1alpha(-/-), and mixed tumors. Our data demonstrate that the HIF-1alpha-dependent response of a few cells is capable of sustaining the growth of the whole tumor, probably through the secretion of factors up-regulated under low oxygen conditions.


Subject(s)
Cell Transformation, Neoplastic/pathology , Endothelial Growth Factors/physiology , Lymphokines/physiology , Teratocarcinoma/pathology , Transcription Factors/deficiency , Animals , Apoptosis/physiology , Cell Differentiation/physiology , Cell Division/physiology , Cell Hypoxia/physiology , Endothelial Growth Factors/biosynthesis , Endothelial Growth Factors/genetics , Female , Hypoxia-Inducible Factor 1, alpha Subunit , Lymphokines/biosynthesis , Lymphokines/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Stem Cells/cytology , Stem Cells/physiology , Teratocarcinoma/blood supply , Teratocarcinoma/metabolism , Transcription Factors/genetics , Transcription Factors/physiology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
12.
Cancer Res ; 61(13): 5255-61, 2001 Jul 01.
Article in English | MEDLINE | ID: mdl-11431367

ABSTRACT

Embryonic stem (ES) cells-wild-type, heterozygous, or null for alpha5-integrin-were injected ectopically into syngeneic mice to develop teratocarcinomas. alpha5-null-derived teratocarcinomas were significantly smaller than the wild-type or alpha5 heterozygous tumors. Histological analysis revealed the presence of tissues derived from all three germ layers, in all tumors. However, alpha5-null teratocarcinomas displayed less undifferentiated tissue than did the controls. Decreased proliferation and increased apoptosis were observed in the undifferentiated areas of the alpha5-null teratocarcinomas. The expression of extracellular matrix proteins, fibronectin and tenascin-C, and the basement membrane components, laminin, entactin/nidogen, and collagen IV, was similar in the different tumors, although the deposition of these molecules was more disorganized in alpha5-null teratocarcinomas. The absence of alpha5-integrin in the various tissues of the alpha5-null tumors was confirmed by immunohistochemistry. Many vessels, but not all, stained positively for alpha5-integrin, showing that they were host derived. Analysis of the area occupied by vessels revealed, on average, an 8-fold decrease in alpha5-null teratocarcinomas compared with control tumors. Staining for smooth muscle alpha-actin showed that pericytes and smooth muscle cells were recruited around the vessels in all tumors, suggesting similar vessel differentiation. Deposition of EIIIA and EIIIB and fibronectin around the vessels was observed in all tumors. The fact that some, although few, alpha5-integrin-negative vessels existed in alpha5-null tumors indicated that alpha5-/- ES cells could differentiate into endothelial cells. Endothelial cell differentiation and vessel formation were analyzed also in vitro. alpha5-null ES cells were differentiated into embryoid bodies, although they were delayed in growth and attachment. Differentiation into endothelial cells was achieved, but the organization into a complex vasculature was delayed compared with controls. We conclude that alpha5beta1-integrin plays a significant role in vessel formation both in ES cell cultures and in teratocarcinomas. Reduced vascularization likely contributed to the reduced proliferation and increased apoptosis observed in alpha5-null teratocarcinomas.


Subject(s)
Antigens, CD/physiology , Neovascularization, Pathologic/pathology , Stem Cells/physiology , Teratocarcinoma/blood supply , Teratocarcinoma/pathology , Animals , Apoptosis/physiology , Cell Differentiation/physiology , Cell Division/physiology , Cells, Cultured , Embryo, Mammalian/cytology , Endothelium, Vascular/cytology , Extracellular Matrix/physiology , Growth Substances/pharmacology , Humans , Integrin alpha5 , Male , Mice , Neovascularization, Pathologic/metabolism , Recombinant Proteins/pharmacology , Stem Cells/cytology , Stem Cells/metabolism
13.
Oncogene ; 13(3): 577-87, 1996 Aug 01.
Article in English | MEDLINE | ID: mdl-8760299

ABSTRACT

Neoangiogenesis is a prerequisite for tumor growth and metastasis. In germ cell cancer patients with the disease limited to the testicle (stage A), tumor-associated neovascularization is predictive of metastatic disease (stage B). To investigate the molecular mechanisms underlying neovascularization in human germ cell tumors (GCTs), we analysed the expression of two angiogenic growth factors, vascular endothelial growth factor (VEGF) and placenta growth factor (P1GF), and of their receptors (FLT-1) and Flk-1/KDR) in a panel of testicular tumors. In this study we show a marked increase in VEGF expression in 36/44 (81.8%) primary testicular-derived GCTs, as compared to normal testis, that significantly correlates with a high density of intratumor microvessels (r = 0.72461, P < 0.001; n = 24). As determined by RT - PCR and/or Western blot, the predominant VEGF isoforms expressed in GCTs are the VEGF121 and VEGF165, which are more efficiently secreted by the cells, and thus more active in eliciting angiogenesis. Conversely, in the case of PIGF, only a weak correlation with the vascular density of tumors is observed (r = 0.26599, P < 0.05; n = 24). Northern blot analysis also revealed significant up-regulation of VEGF/ PIGF receptors in highly vascularized germ cell tumors, compared to normal testes. These findings suggest that VEGF may act in a paracrine manner to induce neovascularization, oedema extravasation and cyst formation in human germ cell tumors. The correlation between VEGF expression and the vascular density of tumors, suggest that the evaluation of VEGF expression may be of help in predicting patients at risk for metastatic diseases. Finally, we demonstrate that VEGF up-regulation may occur at the RNA level since no gene amplification is observed; conversely, in in vitro models such as the embryonal stem cell line NTERA-2 and the choricarcinoma JEG-3 cell line, VEGF (but not PIGF) mRNA expression is regulated by hypoxic stress.


Subject(s)
Endothelial Growth Factors/biosynthesis , Germinoma/blood supply , Lymphokines/biosynthesis , Neovascularization, Pathologic/metabolism , Pregnancy Proteins/biosynthesis , Testicular Neoplasms/blood supply , Base Sequence , Blotting, Western , Carcinoma, Embryonal/blood supply , Carcinoma, Embryonal/metabolism , Cell Hypoxia , Choriocarcinoma/blood supply , Choriocarcinoma/metabolism , Endothelial Growth Factors/genetics , Germinoma/metabolism , Humans , Isomerism , Lymphokines/genetics , Male , Molecular Sequence Data , Placenta Growth Factor , Polymerase Chain Reaction , Pregnancy Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptor Protein-Tyrosine Kinases/genetics , Teratocarcinoma/blood supply , Teratocarcinoma/metabolism , Testicular Neoplasms/metabolism , Transcription, Genetic , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
14.
Arterioscler Thromb Vasc Biol ; 23(5): 905-10, 2003 May 01.
Article in English | MEDLINE | ID: mdl-12649087

ABSTRACT

OBJECTIVE: This study was performed to evaluate the mechanisms leading to tumor vessel occlusion by tissue factor-based drugs, which are used in vascular targeting approaches for the treatment of malignant tumors. METHODS AND RESULTS: The effects of nontargeted soluble tissue factor were evaluated in vitro and in vivo. Tumor-bearing mice were treated with (1) the extracellular portion of tissue factor (soluble tissue factor), (2) low nontoxic doses of lipopolysaccharides, or (3) a combination thereof. The combination treatment showed the best effects and resulted in selective thrombosis of tumor vessels. On the basis of our data from subsequent in vitro analyses, including surface plasmon resonance measurements and endothelial cell based coagulation assays, we propose a model on how soluble tissue factor, although lacking its membrane anchor, can promote selective tumor vessel occlusion. CONCLUSIONS: To our knowledge, this is the first report to describe the molecular mechanisms of coagulation induction by untargeted soluble tissue factor in vivo. Combination treatments including soluble tissue factor might represent an alternative vascular targeting approach for the treatment of malignant tumors.


Subject(s)
Blood Coagulation/drug effects , Embolization, Therapeutic/methods , Lipopolysaccharides/therapeutic use , Lymphoma, Non-Hodgkin/therapy , Teratocarcinoma/therapy , Thromboplastin/therapeutic use , Animals , Dose-Response Relationship, Drug , Drug Synergism , Drug Therapy, Combination , Enzyme Activation/drug effects , Factor Xa/biosynthesis , Humans , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/pharmacology , Lymphoma, Non-Hodgkin/blood , Mice , Mice, Inbred BALB C , Mice, SCID , Peptide Fragments/administration & dosage , Peptide Fragments/pharmacology , Peptide Fragments/therapeutic use , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Solubility , Surface Plasmon Resonance , Teratocarcinoma/blood supply , Thromboplastin/administration & dosage , Thromboplastin/genetics , Thromboplastin/pharmacology , Tumor Necrosis Factor-alpha/analysis , Xenograft Model Antitumor Assays
15.
PLoS One ; 8(12): e80555, 2013.
Article in English | MEDLINE | ID: mdl-24324608

ABSTRACT

OBJECT: Antiangiogenic treatments are beginning to give promising outcomes in many vascular diseases including tumor angiogenesis. In this current study the antiangiogenic and pro-apoptotic actions of α1(IV)NC1 and its N- and C- peptides α1S1(IV)NC1, α1S2(IV)NC1 were investigated in-vitro and in-vivo. STUDY METHOD: Endothelial cells (ECs) were treated with α1(IV)NC1, α1S1(IV)NC1, α1S2(IV)NC1 and in-vitro proliferation, migration, tube formation and apoptotic assays were executed. FasL, Fas, Caspase-8, -3 and PARP activations were studied using immunoblotting analysis using specific antibodies. Also the in-vivo antiangiogenic and pro-apoptotic effects were tested using α1(IV)NC1 in a mice model. RESULTS: Like α1(IV)NC1, its N- and C- terminal α1S2(IV)NC1 and α1S1(IV)NC1 domains posses anti-proliferative, pro-apoptotic activity and inhibit ECs migration and tube formation in-vitro. Both α1S1(IV)NC1 and α1S2(IV)NC1 domains promote apoptosis by activating FasL and down stream apoptotic events including activation of caspase-8, -3 and PARP cleavage in a dose dependent manner in-vitro in ECs. Tumors in mice showed apoptotic TUNEL positive microvasculature upon α1(IV)NC1 treatment, indicating inhibition of tumor angiogenesis and tumor growth. Further, the antitumor activity of α1(IV)NC1 was abrogated when caspase-3 inhibitor was used. These results conform additional properties of α1(IV)NC1 as an endogenous angioinhibitor that induces apoptosis in-vitro and in-vivo by activating FasL mediated caspase-3. SIGNIFICANCE: α1(IV)NC1 and its N- and C- terminal α1S1(IV)NC1 and α1S2(IV)NC1 domains also posses pro-apoptotic and angioinhibitory activity in-vitro and in-vivo. α1(IV)NC1 regulates tumor angiogenesis by activating FasL mediated apoptosis in-vitro and in-vivo. These results demonstrate that α1(IV)NC1 and its peptides inhibit neo-vascular diseases.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Collagen Type IV/pharmacology , Fas Ligand Protein/genetics , Gene Expression Regulation, Neoplastic , Skin Neoplasms/blood supply , Teratocarcinoma/blood supply , Animals , Apoptosis/drug effects , Caspase 3/genetics , Caspase 3/metabolism , Caspase 8/genetics , Caspase 8/metabolism , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Fas Ligand Protein/agonists , Fas Ligand Protein/metabolism , Humans , Mice , Neovascularization, Pathologic , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Protein Isoforms/pharmacology , Signal Transduction , Skin Neoplasms/drug therapy , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Teratocarcinoma/drug therapy , Teratocarcinoma/genetics , Teratocarcinoma/pathology , fas Receptor/genetics , fas Receptor/metabolism
16.
Eur J Cancer ; 47(8): 1276-84, 2011 May.
Article in English | MEDLINE | ID: mdl-21396810

ABSTRACT

INTRODUCTION: Various strategies using L19-mediated fibronectin targeting have become useful clinical tools in anti-tumour therapy and diagnostics. The aim of our study was to characterise the microvascular biodistribution and binding process during tumour angiogenesis and after anti-angiogenic therapy. MATERIALS AND METHODS: SF126 glioma and F9 teratocarcinoma cells were implanted into dorsal skin fold chambers (SF126: n = 4; F9: n = 6). Using fluorescence and confocal intravital microscopy the biodistribution process was assessed at t = 0 h, t = 4 h and t = 24 h after intravenous application of Cy3-L19-SIP. Sunitinib treatment was applied for six days and microscopy was performed 2 and 6 days after treatment initiation. Analysed parameters included: vascular and interstitial binding, preferential binding sites of L19-SIP, microvascular blood flow rate, microvascular permeability. Histological analysis included CD31 and DAPI. RESULTS: L19-SIP showed a specific and time-dependent neovascular binding with a secondary extravasation process reaching optimal vascular/interstitial binding ratio 4 hours after iv administration (F9: L19-SIP: vascular binding: 74.6 ± 14.5; interstitial binding: 46.8 ± 12.1; control vascular: 22,2 ± 16.6). Angiogenic sprouts were preferred binding sites (F9: L19-SIP: 188 ± 15.5; RTV: 90.6 ± 13.5). Anti-angiogenic therapy increased microvascular hemodynamics (SF126: Su: 106.6 ± 13.3 µl/sec; Untreated: 19.7 ± 9.1 µl/sec) and induced increased L19-SIP accumulation (SF 126: t24; Su: 92.6 ± 2.7; Untreated: 71.9 ± 5.9) in therapy resistant tumour vessels. CONCLUSION: L19-SIP shows a time and blood-flow dependent microvascular biodistribution process with angiogenic sprouts as preferential binding sites followed by secondary extravasation of the antibody. Microvascular biodistribution is enhanced in anti-angiogenic-therapy resistant tumour vessels.


Subject(s)
Antibodies/chemistry , Antineoplastic Agents/pharmacology , Brain Neoplasms/metabolism , Glioma/metabolism , Neovascularization, Pathologic , Recombinant Fusion Proteins/chemistry , Teratocarcinoma/metabolism , Animals , Brain Neoplasms/blood supply , Cell Line, Tumor , Glioma/pathology , Humans , Mice , Mice, Inbred C57BL , Microcirculation , Microscopy, Confocal/methods , Neovascularization, Pathologic/pathology , Teratocarcinoma/blood supply
17.
Integr Biol (Camb) ; 3(4): 468-78, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21225052

ABSTRACT

The antibody-mediated targeted delivery of cytokines, growth factors and immunomodulators offers great potential for the therapy of cancer and other serious conditions. Interferon-alpha has long been used in the clinic for the treatment of patients with certain malignancies or with viral disease. Promising anticancer activity has recently been reported for two fusion proteins consisting of immunoglobulins bearing the interferon-alpha polypeptide at the C-terminal end of the molecule. Here we describe the design, production and characterization of a novel immunocytokine, in which murine interferon-alpha2 was sequentially fused with the tumor-targeting antibody fragment scFv(F8), specific to the alternatively-spliced EDA domain of fibronectin. The resulting fusion protein (F8-IFNa) could be produced to homogeneity and was shown to retain both antigen binding activity and interferon-alpha activity. Biodistribution studies in tumor-bearing mice with radioiodinated protein preparations confirmed the ability of F8-IFNa to selectively localize at the tumor site. However, using two different murine models of cancer (F9 teratocarcinomas and Cloudman S91 melanomas in immunocompetent mice), we could not detect a striking superiority for the therapeutic performance of F8-IFNa as compared to KSF-IFNa, a fusion protein of irrelevant specificity in the mouse which was used as negative control. In the paper, we present hypotheses why the antibody-based pharmacodelivery of interferon-alpha fails to eradicate tumors, in contrast to the situation observed by our group for other immunocytokines, which benefit from a selective localization at the tumor site.


Subject(s)
Immunoconjugates/therapeutic use , Interferon-alpha/administration & dosage , Interferon-alpha/therapeutic use , Neoplasms/blood supply , Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Single-Chain Antibodies/immunology , Animal Structures/metabolism , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Blood Vessels/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Fibronectins/immunology , Humans , Immunoconjugates/genetics , Immunoconjugates/immunology , Immunoconjugates/pharmacokinetics , Immunoconjugates/pharmacology , Interferon-alpha/genetics , Leukocytes, Mononuclear/pathology , Melanoma/blood supply , Melanoma/drug therapy , Melanoma/metabolism , Melanoma/pathology , Mice , Mice, 129 Strain , Muramidase/immunology , Neoplasms/metabolism , Neoplasms/pathology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/pharmacokinetics , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use , Single-Chain Antibodies/genetics , Teratocarcinoma/blood supply , Teratocarcinoma/drug therapy , Teratocarcinoma/metabolism , Teratocarcinoma/pathology , Tissue Distribution , Treatment Outcome
18.
Cancer Res ; 70(15): 6216-24, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20647324

ABSTRACT

Tumor angiogenesis is a key event in cancer progression. Here, we report that tumors can stimulate tumor angiogenesis by secretion of galectin-1. Tumor growth and tumor angiogenesis of different tumor models are hampered in galectin-1-null (gal-1(-/-)) mice. However, tumor angiogenesis is less affected when tumor cells express and secrete high levels of galectin-1. Furthermore, tumor endothelial cells in gal-1(-/-) mice take up galectin-1 that is secreted by tumor cells. Uptake of galectin-1 by cultured endothelial cells specifically promotes H-Ras signaling to the Raf/mitogen-activated protein kinase/extracellular signal-regulated kinase (Erk) kinase (Mek)/Erk cascade and stimulates endothelial cell proliferation and migration. Moreover, the activation can be blocked by galectin-1 inhibition as evidenced by hampered membrane translocation of H-Ras.GTP and impaired Raf/Mek/Erk phosphorylation after treatment with the galectin-1-targeting angiogenesis inhibitor anginex. Altogether, these data identify galectin-1 as a proangiogenic factor. These findings have direct implications for current efforts on galectin-1-targeted cancer therapies.


Subject(s)
Galectin 1/physiology , Melanoma, Experimental/blood supply , Teratocarcinoma/blood supply , Animals , Cells, Cultured , Chick Embryo , Chorioallantoic Membrane/blood supply , Endothelial Cells/cytology , Endothelial Cells/drug effects , Galectin 1/deficiency , Galectin 1/metabolism , Galectin 1/pharmacology , Humans , MAP Kinase Signaling System , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitogen-Activated Protein Kinases/metabolism , Neovascularization, Pathologic/pathology , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/physiology , Recombinant Proteins/pharmacology , Teratocarcinoma/metabolism , ras Proteins/metabolism
19.
J Environ Pathol Toxicol Oncol ; 29(1): 17-30, 2010.
Article in English | MEDLINE | ID: mdl-20528744

ABSTRACT

In our previous study, PAb, a VEGF polyclonal antibody was found to inhibit murine tumor growth significantly. The main objective of this study was to investigate the efficacy of combination therapy of (PAb) and cisplatin on human ovarian cancer xenograft. Effect of VEGF, PAb, PAb-cisplatin combination, and cisplatin alone on cultured human ovarian teratocarci-noma cell line PA-1 were assessed by measuring cell proliferation, matrigel invasion, MMP-9 expression, and MMP-9 secretion. In vivo, effect of PAb was observed in a xenograft model of ovarian cancer. Antitumor efficacy was monitored by assessment of tumor volume, MVD, serum NO, serum VEGF, and p53 expression. VEGF increased proliferation of PA-1 cell in a dose-dependent manner while addition of PAb inhibited cell proliferation, cell invasion as well as MMP-9 secretion in vitro. Tumor burden in PAb and PAb-cisplatin combination group was reduced by 41% (p < 0.05) and 66% (p < 0.01), respectively. A significant decrease in MVD, serum NO, serum VEGF, and p53 expression was also observed after PAb and PAb-cisplatin combination treatment when compared to normal mouse serum IgG-treated control mice. Thus, it was concluded that VEGF immunoneutralization may enhance cisplatin-in-duced apoptosis in human ovarian cancer and thus may be an effective way to reduce tumor growth in ovarian carcinoma.


Subject(s)
Antibodies, Blocking/pharmacology , Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Ovarian Neoplasms/drug therapy , Teratocarcinoma/drug therapy , Vascular Endothelial Growth Factor A/immunology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Humans , Male , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Neovascularization, Pathologic/chemically induced , Nitric Oxide/blood , Ovarian Neoplasms/blood supply , Ovarian Neoplasms/pathology , Teratocarcinoma/blood supply , Teratocarcinoma/pathology , Tumor Suppressor Protein p53/metabolism , Vascular Endothelial Growth Factor A/blood , Xenograft Model Antitumor Assays
20.
Blood ; 110(4): 1168-77, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17426256

ABSTRACT

Human alpha3 chain, a noncollagenous domain of type IV collagen [alpha3(IV)NC1], inhibits angiogenesis and tumor growth. These biologic functions are partly attributed to the binding of alpha3(IV)NC1 to alphaVbeta3 and alpha3beta1 integrins. alpha3(IV)NC1 binds alphaVbeta3 integrin, leading to translation inhibition by inhibiting focal adhesion kinase/phosphatidylinositol 3-kinase/Akt/mTOR/4E-BP1 pathways. In the present study, we evaluated the role of alpha3beta1 and alphaVbeta3 integrins in tube formation and regulation of cyclooxygenase-2 (COX-2) on alpha3(IV)NC1 stimulation. We found that although both integrins were required for the inhibition of tube formation by alpha3(IV)NC1 in endothelial cells, only alpha3beta1 integrin was sufficient to regulate COX-2 in hypoxic endothelial cells. We show that binding of alpha3(IV)NC1 to alpha3beta1 integrin leads to inhibition of COX-2-mediated pro-angiogenic factors, vascular endothelial growth factor, and basic fibroblast growth factor by regulating IkappaBalpha/NFkappaB axis, and is independent of alphaVbeta3 integrin. Furthermore, beta3 integrin-null endothelial cells, when treated with alpha3(IV)NC1, inhibited hypoxia-mediated COX-2 expression, whereas COX-2 inhibition was not observed in alpha3 integrin-null endothelial cells, indicating that regulation of COX-2 by alpha3(IV)NC1 is mediated by integrin alpha3beta1. Our in vitro and in vivo findings demonstrate that alpha3beta1 integrin is critical for alpha3(IV)NC1-mediated inhibition of COX-2-dependent angiogenic signaling and inhibition of tumor progression.


Subject(s)
Collagen Type IV/metabolism , Cyclooxygenase 2/metabolism , Gene Expression Regulation, Neoplastic , Integrin alpha3beta1/physiology , Integrin alphaVbeta3/antagonists & inhibitors , Neovascularization, Pathologic/metabolism , Teratocarcinoma/metabolism , Angiogenesis Inhibitors/genetics , Angiogenesis Inhibitors/pharmacology , Animals , Cell Adhesion , Cell Hypoxia , Cell Proliferation , Cells, Cultured , Collagen Type IV/genetics , Cyclooxygenase 2/chemistry , Cyclooxygenase 2 Inhibitors/pharmacology , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Fibroblast Growth Factor 2/metabolism , Humans , Immunoblotting , Immunoprecipitation , Male , Mice , NF-kappa B/genetics , NF-kappa B/metabolism , Neoplasms , Neovascularization, Pathologic/drug therapy , Pulmonary Artery/cytology , Pulmonary Artery/metabolism , Teratocarcinoma/blood supply , Teratocarcinoma/drug therapy , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL