Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Semin Cancer Biol ; 86(Pt 3): 199-206, 2022 11.
Article in English | MEDLINE | ID: mdl-35248730

ABSTRACT

Antiangiogenic therapies are considered a promising strategy against solid tumors. Their aim is to inhibit the formation of new blood vasculature, thereby reducing the oxygen and nutrient supply to prevent further tumor growth and spreading. However, the strategy has seen limitations, as survival benefits are modest and often accompanied with increased tumor aggressiveness in form of invasion and metastasis. Antiangiogenic induced changes in the tumor microenvironment, such as hypoxia, mechanical stress or extracellular acidification can activate different receptors of tumoral and stromal cells and induce an extensive remodeling of the entire tumor microenvironment, with the overall goal to invade nearby tissues and regain access to the vasculature. In this regard, receptor tyrosine kinases have been studied intensively and especially the inhibition of c-Met has given promising results, characterized by a reduction in invasiveness and prolonged survival. Receptors that sense changes in the extracellular matrix like integrins or proteoglycans can also induce downstream signaling that stimulates the expression of remodeling factors such as new matrix components, enzymes or chemoattractants. Targeting multiple receptors and sensors of cancer cells simultaneously might represent an effective second line treatment that prevents the formation of malignant side effects.


Subject(s)
Neoplasms , Tumor Microenvironment , Humans , Tumor Microenvironment/physiology , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Extracellular Matrix/metabolism , Neoplasms/drug therapy , Neoplasms/etiology , Neoplasms/metabolism , Signal Transduction
2.
Clin Cancer Res ; 28(1): 137-149, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34593528

ABSTRACT

PURPOSE: FGFR1 amplification (FGFR1amp) is recurrent in metastatic breast cancer (MBC) and is associated with resistance to endocrine therapy and CDK4/6 inhibitors (CDK4/6is). Multi-tyrosine kinase inhibitors (MTKIs) and selective pan-FGFR inhibitors (FGFRis) are being developed for FGFR1amp breast cancer. High-level FGFR amplification and protein expression by IHC have identified breast cancer responders to FGFRis or MTKIs, respectively. EXPERIMENTAL DESIGN: Here, we used preclinical models and patient samples to identify predictive biomarkers to these drugs. We evaluated the antitumor activity of an FGFRi and an MTKI in a collection of 17 breast cancer patient-derived xenografts (PDXs) harboring amplification in FGFR1/2/3/4 and in 10 patients receiving either an FGFRi/MTKI. mRNA levels were measured on FFPE tumor samples using two commercial strategies. Proliferation and angiogenesis were evaluated by detecting Ki-67 and CD31 in viable areas by immunofluorescence. RESULTS: High FGFR1-4 mRNA levels but not copy-number alteration (CNA) is associated with FGFRi response. Treatment with MTKIs showed higher response rates than with FGFRis (86% vs. 53%), regardless of the FGFR1-4 mRNA levels. FGFR-addicted PDXs exhibited an antiproliferative response to either FGFRis or MTKIs, and PDXs exclusively sensitive to MTKI exhibited an additional antiangiogenic response. Consistently, the clinical benefit of MTKIs was not associated with high FGFR1-4 mRNA levels and was observed in patients previously treated with antiangiogenic drugs. CONCLUSIONS: Tailored therapy with FGFRis in molecularly selected MBC based on high FGFR1-4 mRNA levels warrants prospective validation in patients with CDK4/6i-resistant luminal breast cancer and in patients with TNBC without targeted therapeutic options.


Subject(s)
Breast Neoplasms , RNA, Messenger , Receptor Protein-Tyrosine Kinases , Female , Humans , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction
3.
EMBO Mol Med ; 12(12): e11889, 2020 12 07.
Article in English | MEDLINE | ID: mdl-33151035

ABSTRACT

An open debate in antiangiogenic therapies is about their consequence on tumor invasiveness and metastasis, which is undoubtedly relevant for patients currently treated with antiangiogenics, such as renal cell carcinoma patients. To address, this we developed an extensive series of 27 patient biopsy-derived orthotopic xenograft models (Ren-PDOX) that represent inter-patient heterogeneity. In specific tumors, antiangiogenics produced increased invasiveness and metastatic dissemination, while in others aggressiveness remained unchanged. Mechanistically, species-discriminative RNA sequencing identified a tumor cell-specific differential expression profile associated with tumor progression and aggressivity in TCGA RCC patients. Gene filtering using an invasion-annotated patient series pinpointed two candidate genes, of which ALDH1A3 differentiated the pro-invasive subtype of Ren-PDOXs. Validation in an independent series of 15 antiangiogenic-treated patients confirmed that pre-treatment ALDH1A3 can significantly discriminate patients with pro-aggressive response upon treatment. Overall, results confirm that effects of antiangiogenic drugs on tumor invasion and metastasis are heterogeneous and may profoundly affect the natural progression of tumors and promote malignancy. Furthermore, we identify a specific molecular biomarker that could be used to select patients that better benefit from treatment.


Subject(s)
Angiogenesis Inhibitors/adverse effects , Carcinoma, Renal Cell/pathology , Kidney Neoplasms/pathology , Models, Biological , Neoplasm Invasiveness/pathology , Neoplasm Metastasis/pathology , Animals , Cell Line, Tumor , Humans , Male , Mice , Mice, Nude , Precision Medicine , Xenograft Model Antitumor Assays
4.
Cancers (Basel) ; 12(2)2020 Jan 28.
Article in English | MEDLINE | ID: mdl-32012804

ABSTRACT

The transcription factor EVI1 plays an oncogenic role in several types of neoplasms by promoting aggressive cancer features. EVI1 contributes to epigenetic regulation and transcriptional control, and its overexpression has been associated with enhanced PI3K-AKT-mTOR signaling in some settings. These observations raise the possibility that EVI1 influences the prognosis and everolimus-based therapy outcome of clear cell renal cell carcinoma (ccRCC). Here, gene expression and protein immunohistochemical studies of ccRCC show that EVI1 overexpression is associated with advanced disease features and with poorer outcome-particularly in the CC-e.3 subtype defined by The Cancer Genome Atlas. Overexpression of an oncogenic EVI1 isoform in RCC cell lines confers substantial resistance to everolimus. The EVI1 rs1344555 genetic variant is associated with poorer survival and greater progression of metastatic ccRCC patients treated with everolimus. This study leads us to propose that evaluation of EVI1 protein or gene expression, and of EVI1 genetic variants may help improve estimates of prognosis and the benefit of everolimus-based therapy in ccRCC.

5.
Cancers (Basel) ; 11(6)2019 Jun 17.
Article in English | MEDLINE | ID: mdl-31212986

ABSTRACT

Uveal melanoma is considered a rare disease but it is the most common intraocular malignancy in adults. Local treatments are effective, but the systemic recurrence rate is unacceptably high. Moreover, once metastasis have developed the prognosis is poor, with a 5-year survival rate of less than 5%, and systemic therapies, including immunotherapy, have rendered poor results. The tumour biology is complex, but angiogenesis is a highly important pathway in these tumours. Vasculogenic mimicry, the ability of melanomas to generate vascular channels independently of endothelial cells, could play an important role, but no effective therapy targeting this process has been developed so far. Angiogenesis modulates the tumour microenvironment of melanomas, and a close interplay is established between them. Therefore, combining immune strategies with drugs targeting angiogenesis offers a new therapeutic paradigm. In preclinical studies, these approaches effectively target these tumours, and a phase I clinical study has shown encouraging results in cutaneous melanomas. In this review, we will discuss the importance of angiogenesis in uveal melanoma, with a special focus on vasculogenic mimicry, and describe the interplay between angiogenesis and the tumour microenvironment. In addition, we will suggest future therapeutic approaches based on these observations and mention ways in which to potentially enhance current treatments.

6.
Cancer Res ; 79(20): 5328-5341, 2019 Oct 15.
Article in English | MEDLINE | ID: mdl-31239269

ABSTRACT

One of the main consequences of inhibition of neovessel growth and vessel pruning produced by angiogenesis inhibitors is increased intratumor hypoxia. Growing evidence indicates that tumor cells escape from this hypoxic environment to better nourished locations, presenting hypoxia as a positive stimulus for invasion. In particular, anti-VEGF/R therapies produce hypoxia-induced invasion and metastasis in a spontaneous mouse model of pancreatic neuroendocrine cancer (PanNET), RIP1-Tag2. Here, a novel vascular-targeting agent targeting semaphorin 4D (Sema4D) demonstrated impaired tumor growth and extended survival in the RIP1-Tag2 model. Surprisingly, although there was no induction of intratumor hypoxia by anti-Sema4D therapy, the increase in local invasion and distant metastases was comparable with the one produced by VEGFR inhibition. Mechanistically, the antitumor effect was due to an alteration in vascular function by modification of pericyte coverage involving platelet-derived growth factor B. On the other hand, the aggressive phenotype involved a macrophage-derived Sema4D signaling engagement, which induced their recruitment to the tumor invasive fronts and secretion of stromal cell-derived factor 1 (SDF1) that triggered tumor cell invasive behavior via CXCR4. A comprehensive clinical validation of the targets in different stages of PanNETs demonstrated the implication of both Sema4D and CXCR4 in tumor progression. Taken together, we demonstrate beneficial antitumor and prosurvival effects of anti-Sema4D antibody but also unravel a novel mechanism of tumor aggressivity. This mechanism implicates recruitment of Sema4D-positive macrophages to invasive fronts and their secretion of proinvasive molecules that ultimately induce local tumor invasion and distant metastasis in PanNETs. SIGNIFICANCE: An anti-semaphorin-4D vascular targeting agent demonstrates antitumor and prosurvival effects but also unravels a novel promalignant effect involving macrophage-derived SDF1 that promotes tumor invasion and metastasis, both in animal models and patients.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/79/20/5328/F1.large.jpg.See related commentary by Tamagnone and Franzolin, p. 5146.


Subject(s)
Neoplasms , Semaphorins , Animals , Antigens, CD , Humans , Mice , Signal Transduction
7.
Nanoscale ; 11(23): 11331-11339, 2019 Jun 13.
Article in English | MEDLINE | ID: mdl-31166337

ABSTRACT

Nanomedicine has emerged as a promising strategy to address some of the limitations of traditional biomedical sensing, imaging and therapy modalities. Its applicability and efficacy are, in part, hindered by the difficulty in both controllably delivering nanoparticles to specific regions and accurately monitoring them in tissue. Gold nanoparticles are among the most extensively used inorganic nanoparticles which benefit from high biocompatibility, flexible functionalization, strong and tunable resonant absorption, and production scalability. Moreover, their capability to enhance optical fields at their plasmon resonance enables local boosting of non-linear optical processes, which are otherwise very inefficient. In particular, two-photon induced luminescence (TPL) in gold offers high signal specificity for monitoring gold nanoparticles in a biological environment. In this article, we demonstrate that TPL microscopy provides a robust sub-micron-resolution technique able to quantify accumulated gold nanorods (GNRs) both in cells and in tissues. First, the temporal accumulation of GNRs with two different surface chemistries was measured in 786-O cells during the first 24 hours of incubation, and at different nanoparticle concentrations. Subsequently, GNR accumulation in mice, 6 h and 24 hours after tail vein injection, was quantified by TPL microscopy in biopsied tissue from kidney, spleen, liver and clear cell renal cell carcinoma (ccRCC) tumors, in good agreement with inductively coupled mass spectroscopy. Our data suggest that TPL microscopy stands as a powerful tool to understand and quantify the delivery mechanisms of gold nanoparticles, highly relevant to the development of future theranostic medicines.


Subject(s)
Adenocarcinoma, Clear Cell , Gold , Kidney Neoplasms , Metal Nanoparticles , Neoplasms, Experimental , Adenocarcinoma, Clear Cell/diagnostic imaging , Adenocarcinoma, Clear Cell/metabolism , Adenocarcinoma, Clear Cell/pathology , Animals , Cell Line , Gold/chemistry , Gold/pharmacokinetics , Gold/pharmacology , Humans , Kidney Neoplasms/diagnostic imaging , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use , Mice , Microscopy, Fluorescence, Multiphoton , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Surface Plasmon Resonance , Theranostic Nanomedicine
8.
Nanoscale ; 11(12): 5595-5606, 2019 Mar 21.
Article in English | MEDLINE | ID: mdl-30860518

ABSTRACT

Owing to their unique combination of chemical and physical properties, inorganic nanoparticles show a great deal of potential as suitable agents for early diagnostics and less invasive therapies. Yet, their translation to the clinic has been hindered, in part, by the lack of non-invasive methods to quantify their concentration in vivo while also assessing their effect on the tissue physiology. In this work, we demonstrate that diffuse optical techniques, employing near-infrared light, have the potential to address this need in the case of gold nanoparticles which support localized surface plasmons. An orthoxenograft mouse model of clear cell renal cell carcinoma was non-invasively assessed by diffuse reflectance and correlation spectroscopies before and over several days following a single intravenous tail vein injection of polyethylene glycol-coated gold nanorods (AuNRs-PEG). Our platform enables to resolve the kinetics of the AuNR-PEG uptake by the tumor in quantitative agreement with ex vivo inductively coupled plasma mass spectroscopy. Furthermore, it allows for the simultaneous monitoring of local tissue hemodynamics, enabling us to conclude that AuNRs-PEG do not significantly alter the animal physiology. We note that the penetration depth of this current probe was a few millimeters but can readily be extended to centimeters, hence gaining clinical relevance. This study and the methodology presented here complement the nanomedicine toolbox by providing a flexible platform, extendable to other absorbing agents that can potentially be translated to human trials.


Subject(s)
Gold/chemistry , Hemodynamics , Metal Nanoparticles/chemistry , Animals , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/therapy , Cell Line, Tumor , Humans , Hyperthermia, Induced , Infrared Rays , Kidney Neoplasms/pathology , Kidney Neoplasms/therapy , Male , Mass Spectrometry , Mice , Mice, Nude , Phototherapy , Polyethylene Glycols/chemistry , Transplantation, Heterologous
9.
Oncologist ; 24(1): 38-46, 2019 01.
Article in English | MEDLINE | ID: mdl-29794066

ABSTRACT

BACKGROUND: Antitumor activity of the combination of somatostatin analogues (SSAs) and the mammalian target of rapamycin (mTOR) inhibitor everolimus in patients with neuroendocrine tumors (NETs) has been reported but not confirmed in prospective trials. MATERIALS AND METHODS: This prospective, multicenter, single-arm phase II EVERLAR study evaluated everolimus 10 mg/day and the SSA octreotide 30 mg every 28 days in patients with advanced nonfunctioning well-differentiated gastrointestinal NETs (GI-NETs) that progressed in the last 12 months (ClinicalTrials.gov NCT01567488). Prior treatment with SSAs and any systemic or locoregional therapy was allowed except for mTOR inhibitors. Patients continued treatment until disease progression or unacceptable adverse events (AEs). The primary endpoint was progression-free survival (PFS) at 12 months; secondary endpoints included early biochemical response, objective response rate (ORR) by RECIST v1.0, overall survival (OS), AEs, activation of mTOR pathway (insulin-like growth factor 1 receptor [IGF1R] and phosphoS6 [pS6] expression). RESULTS: Forty-three patients were included in the intent-to-treat analyses. After 12 months of treatment, 62.3% (95% confidence interval [CI] 48%-77%) of patients had not progressed or died. The 24-month PFS rate was 43.6% (95% CI 29%-58%). The confirmed ORR was 2.3%, and stable disease was 58.1%. Median OS was not reached after 24 months of median follow-up. Dose reductions and temporary interruptions due to AEs were required in 14 (33%) and 33 (77%) patients, respectively. The most frequent AEs were diarrhea, asthenia, mucositis, rash, and hyperglycemia. No correlation was observed between IGFR1 and pS6 expression and PFS/OS. CONCLUSION: The everolimus-octreotide combination provided clinically relevant efficacy in nonfunctioning GI-NETs, similar to the results of RADIANT-2 in functioning setting. IMPLICATIONS FOR PRACTICE: The EVERLAR study reports prospective data of somatostatin analogue in combination with everolimus in nonfunctioning gastrointestinal neuroendocrine tumors suggesting meaningful activity and favorable toxicity profile that supports drug combination in this setting.


Subject(s)
Everolimus/therapeutic use , Intestinal Neoplasms/drug therapy , Neuroendocrine Tumors/drug therapy , Octreotide/therapeutic use , Pancreatic Neoplasms/drug therapy , Stomach Neoplasms/drug therapy , Aged , Everolimus/pharmacology , Female , Humans , Intestinal Neoplasms/pathology , Male , Neuroendocrine Tumors/pathology , Octreotide/pharmacology , Pancreatic Neoplasms/pathology , Prospective Studies , Stomach Neoplasms/pathology
11.
Nat Commun ; 9(1): 4826, 2018 11 16.
Article in English | MEDLINE | ID: mdl-30446640

ABSTRACT

Angiogenesis is a dynamic process relying on endothelial cell rearrangements within vascular tubes, yet the underlying mechanisms and functional relevance are poorly understood. Here we show that PI3Kα regulates endothelial cell rearrangements using a combination of a PI3Kα-selective inhibitor and endothelial-specific genetic deletion to abrogate PI3Kα activity during vessel development. Quantitative phosphoproteomics together with detailed cell biology analyses in vivo and in vitro reveal that PI3K signalling prevents NUAK1-dependent phosphorylation of the myosin phosphatase targeting-1 (MYPT1) protein, thereby allowing myosin light chain phosphatase (MLCP) activity and ultimately downregulating actomyosin contractility. Decreased PI3K activity enhances actomyosin contractility and impairs junctional remodelling and stabilization. This leads to overstretched endothelial cells that fail to anastomose properly and form aberrant superimposed layers within the vasculature. Our findings define the PI3K/NUAK1/MYPT1/MLCP axis as a critical pathway to regulate actomyosin contractility in endothelial cells, supporting vascular patterning and expansion through the control of cell rearrangement.


Subject(s)
Actomyosin/genetics , Gene Expression Regulation, Developmental , Myosin-Light-Chain Phosphatase/genetics , Neovascularization, Physiologic/genetics , Phosphatidylinositol 3-Kinases/genetics , Protein Kinases/genetics , Repressor Proteins/genetics , Actomyosin/metabolism , Animals , Body Patterning/genetics , Embryo, Mammalian , Embryo, Nonmammalian , Gene Expression Profiling , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Intercellular Junctions/metabolism , Intercellular Junctions/ultrastructure , Lung/blood supply , Lung/cytology , Lung/growth & development , Lung/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myosin-Light-Chain Phosphatase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Kinases/metabolism , Repressor Proteins/metabolism , Retina/cytology , Retina/growth & development , Retina/metabolism , Signal Transduction , Zebrafish
12.
Front Oncol ; 8: 248, 2018.
Article in English | MEDLINE | ID: mdl-30013950

ABSTRACT

Activation of the tumor and stromal cell-driven angiogenic program is one of the first requirements in the tumor ecosystem for growth and dissemination. The understanding of the dynamic angiogenic tumor ecosystem has rapidly evolved over the last decades. Beginning with the canonical sprouting angiogenesis, followed by vasculogenesis and intussusception, and finishing with vasculogenic mimicry, the need for different neovascularization mechanisms is further explored. In addition, an overview of the orchestration of angiogenesis within the tumor ecosystem cellular and molecular components is provided. Clinical evidence has demonstrated the effectiveness of traditional vessel-directed antiangiogenics, stressing on the important role of angiogenesis in tumor establishment, dissemination, and growth. Particular focus is placed on the interaction between tumor cells and their surrounding ecosystem, which is now regarded as a promising target for the development of new antiangiogenics.

13.
J Clin Invest ; 128(9): 3887-3905, 2018 08 31.
Article in English | MEDLINE | ID: mdl-29944140

ABSTRACT

Dormant or slow-cycling tumor cells can form a residual chemoresistant reservoir responsible for relapse in patients, years after curative surgery and adjuvant therapy. We have adapted the pulse-chase expression of H2BeGFP for labeling and isolating slow-cycling cancer cells (SCCCs). SCCCs showed cancer initiation potential and enhanced chemoresistance. Cells at this slow-cycling status presented a distinctive nongenetic and cell-autonomous gene expression profile shared across different tumor types. We identified TET2 epigenetic enzyme as a key factor controlling SCCC numbers, survival, and tumor recurrence. 5-Hydroxymethylcytosine (5hmC), generated by TET2 enzymatic activity, labeled the SCCC genome in carcinomas and was a predictive biomarker of relapse and survival in cancer patients. We have shown the enhanced chemoresistance of SCCCs and revealed 5hmC as a biomarker for their clinical identification and TET2 as a potential drug target for SCCC elimination that could extend patients' survival.


Subject(s)
DNA-Binding Proteins/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Proto-Oncogene Proteins/metabolism , 5-Methylcytosine/analogs & derivatives , 5-Methylcytosine/metabolism , Animals , Biomarkers, Tumor/metabolism , Cell Cycle , Cell Line, Tumor , Cell Survival/drug effects , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Dioxygenases , Drug Resistance, Neoplasm/genetics , Epigenesis, Genetic , Female , Gene Knockdown Techniques , Humans , Mice , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Neoplasms/genetics , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Recurrence , Xenograft Model Antitumor Assays
14.
Arterioscler Thromb Vasc Biol ; 38(5): 1216-1229, 2018 05.
Article in English | MEDLINE | ID: mdl-29449337

ABSTRACT

OBJECTIVE: ALK1 (activin-receptor like kinase 1) is an endothelial cell-restricted receptor with high affinity for BMP (bone morphogenetic protein) 9 TGF-ß (transforming growth factor-ß) family member. Loss-of-function mutations in ALK1 cause a subtype of hereditary hemorrhagic telangiectasia-a rare disease characterized by vasculature malformations. Therapeutic strategies are aimed at reducing potential complications because of vascular malformations, but currently, there is no curative treatment for hereditary hemorrhagic telangiectasia. APPROACH AND RESULTS: In this work, we report that a reduction in ALK1 gene dosage (heterozygous ALK1+/- mice) results in enhanced retinal endothelial cell proliferation and vascular hyperplasia at the sprouting front. We found that BMP9/ALK1 represses VEGF (vascular endothelial growth factor)-mediated PI3K (phosphatidylinositol 3-kinase) by promoting the activity of the PTEN (phosphatase and tensin homolog). Consequently, loss of ALK1 function in endothelial cells results in increased activity of the PI3K pathway. These results were confirmed in cutaneous telangiectasia biopsies of patients with hereditary hemorrhagic telangiectasia 2, in which we also detected an increase in endothelial cell proliferation linked to an increase on the PI3K pathway. In mice, genetic and pharmacological inhibition of PI3K is sufficient to abolish the vascular hyperplasia of ALK1+/- retinas and in turn normalize the vasculature. CONCLUSIONS: Overall, our results indicate that the BMP9/ALK1 hub critically mediates vascular quiescence by limiting PI3K signaling and suggest that PI3K inhibitors could be used as novel therapeutic agents to treat hereditary hemorrhagic telangiectasia.


Subject(s)
Activin Receptors, Type II/genetics , Activin Receptors, Type I/genetics , Endothelial Cells/enzymology , Mutation , Neovascularization, Pathologic , Phosphatidylinositol 3-Kinase/metabolism , Retinal Telangiectasis/genetics , Telangiectasia, Hereditary Hemorrhagic/genetics , Activin Receptors, Type I/deficiency , Angiogenesis Inhibitors/pharmacology , Animals , Case-Control Studies , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/pathology , Enzyme Activation , Gene Deletion , Genetic Predisposition to Disease , Growth Differentiation Factor 2/pharmacology , Human Umbilical Vein Endothelial Cells/enzymology , Human Umbilical Vein Endothelial Cells/pathology , Humans , Hyperplasia , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Retinal Telangiectasis/drug therapy , Retinal Telangiectasis/enzymology , Retinal Telangiectasis/pathology , Signal Transduction , Telangiectasia, Hereditary Hemorrhagic/drug therapy , Telangiectasia, Hereditary Hemorrhagic/enzymology , Telangiectasia, Hereditary Hemorrhagic/pathology , Vascular Endothelial Growth Factor A/pharmacology
15.
Mol Cancer Ther ; 17(2): 532-543, 2018 02.
Article in English | MEDLINE | ID: mdl-29146630

ABSTRACT

Epithelial ovarian cancer is characterized by a low recovery rate because the disease is typically diagnosed at an advanced stage, by which time most patients (80%) already exhibit disseminated neoplasia. The cytokine receptor CXCR4 has been implicated in the development of metastasis in various tumor types. Using a patient-derived tissue macroarray and mRNA expression analysis, we observed high CXCR4 levels in high-grade serous epithelial ovarian carcinomas, the most metastatic tumor, compared with those in endometrioid carcinomas. CXCR4 inhibition by treatment with the CXCR4 antagonist AMD3100 or by expression of shRNA anti-CXCR4 similarly inhibited angiogenesis in several models of ovarian carcinomas orthotopically grown in nude mice, but the effect on tumor growth was correlated with the levels of CXCR4 expression. Moreover, CXCR4 inhibition completely blocked dissemination and metastasis. This effect was associated with reduced levels of active Src, active ERKs, the inhibition of EMT transition, and block of hematogenous ovarian cancer dissemination decreasing circulating human tumoral cells (CTC). In tumors, CXCR4-expressing cells also had more mesenchymal characteristics. In conclusion, our results indicate that CXCR4 expression confers a proinvasive phenotype to ovarian carcinoma cells. Thus, anti-CXCR4 therapy is a possible agent for a complementary treatment of advanced disseminated epithelial high-grade serous ovarian cancer patients. Mol Cancer Ther; 17(2); 532-43. ©2017 AACR.


Subject(s)
Ovarian Neoplasms/genetics , Receptors, CXCR4/genetics , Aged , Animals , Cell Line, Tumor , Cell Proliferation/physiology , Female , Humans , Mice , Mice, Nude , Middle Aged , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Receptors, CXCR4/metabolism , Retrospective Studies , Signal Transduction
16.
Biomed Opt Express ; 8(5): 2563-2582, 2017 May 01.
Article in English | MEDLINE | ID: mdl-28663891

ABSTRACT

The longitudinal effect of an anti-vascular endothelial growth factor receptor 2 (VEGFR-2) antibody (DC 101) therapy on a xenografted renal cell carcinoma (RCC) mouse model was monitored using hybrid diffuse optics. Two groups of immunosuppressed male nude mice (seven treated, seven controls) were measured. Tumor microvascular blood flow, total hemoglobin concentration and blood oxygenation were investigated as potential biomarkers for the monitoring of the therapy effect twice a week and were related to the final treatment outcome. These hemodynamic biomarkers have shown a clear differentiation between two groups by day four. Moreover, we have observed that pre-treatment values and early changes in hemodynamics are highly correlated with the therapeutic outcome demonstrating the potential of diffuse optics to predict the therapy response at an early time point.

17.
Trends Cancer ; 3(1): 10-18, 2017 01.
Article in English | MEDLINE | ID: mdl-28718423

ABSTRACT

Angiogenesis and metabolism are entwined processes that permit tumor growth and progression. Blood vessel supply is necessary for tumor survival not only by providing oxygen and nutrients for anabolism but also by removing waste products from cellular metabolism. On the other hand, blocking angiogenesis with antiangiogenic therapies shows clinical benefits in treating several tumor types. Nevertheless, resistance to therapy emerges over time. In this review we discuss a novel mechanism of adaptive resistance involving metabolic adaptation of tumor cells, and we also provide examples of tumor adaptation to therapy, which may represent a new mechanism of resistance in several types of cancer. Thus, targeting this metabolic tumor adaptation could be a way to avoid resistance in cancer patients.


Subject(s)
Drug Resistance, Neoplasm , Neoplasms , Neovascularization, Pathologic , Angiogenesis Inhibitors/therapeutic use , Animals , Humans , Hypoxia/metabolism , Hypoxia/pathology , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology
18.
J Mol Endocrinol ; 59(1): R77-R91, 2017 07.
Article in English | MEDLINE | ID: mdl-28469004

ABSTRACT

Neuroendocrine tumors (NETs) are a heterogeneous group of neoplasms that arise from cells of the neuroendocrine system. NETs are characterized by being highly vascularized tumors that produce large amounts of proangiogenic factors. Due to their complexity and heterogeneity, progress in the development of successful therapeutic approaches has been limited. For instance, standard chemotherapy-based therapies have proven to be poorly selective for tumor cells and toxic for normal tissues. Considering the urge to develop an efficient therapy to treat NET patients, vascular targeting has been proposed as a new approach to block tumor growth. This review provides an update of the mechanisms regulating different components of vessels and their contribution to tumor progression in order to develop new therapeutic drugs. Following the description of classical anti-angiogenic therapies that target VEGF pathway, new angiogenic targets such as PDGFs, EGFs, FGFs and semaphorins are further explored. Based on recent research in the field, the combination of therapies that target multiple and different components of vessel formation would be the best approach to specifically target NETs and inhibit tumor growth.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Gene Expression Regulation, Neoplastic , Neovascularization, Pathologic/prevention & control , Neuroendocrine Tumors/drug therapy , Epidermal Growth Factor/antagonists & inhibitors , Epidermal Growth Factor/genetics , Epidermal Growth Factor/metabolism , Fibroblast Growth Factors/antagonists & inhibitors , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Genetic Heterogeneity , Humans , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Neuroendocrine Tumors/genetics , Neuroendocrine Tumors/metabolism , Neuroendocrine Tumors/pathology , Neurosecretory Systems/blood supply , Neurosecretory Systems/drug effects , Neurosecretory Systems/metabolism , Neurosecretory Systems/pathology , Platelet-Derived Growth Factor/antagonists & inhibitors , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/metabolism , Semaphorins/antagonists & inhibitors , Semaphorins/genetics , Semaphorins/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
19.
Mol Cell Oncol ; 3(5): e1211979, 2016.
Article in English | MEDLINE | ID: mdl-27857974

ABSTRACT

Several types of tumor are currently treated with antiangiogenic drugs. Unfortunately, most of these patients develop therapy resistance and succumb to the disease. Recently, a novel mechanism of resistance to antiangiogenics involving metabolic symbiosis of tumor cells has been described. Strategies to block resistance are emerging as a promising therapeutic approach.

20.
Oncotarget ; 7(37): 60110-60122, 2016 Sep 13.
Article in English | MEDLINE | ID: mdl-27507050

ABSTRACT

The truncated somatostatin receptor sst5TMD4 is associated with poor prognosis in breast cancer and increases breast cancer cell malignancy. Here, we examined the cellular/molecular mechanisms underlying this association, aiming to identify new molecular tools to improve diagnosis, prognosis or therapy. A gene expression array comparing sst5TMD4 stably-transfected MCF-7 cells and their controls (empty-plasmid) revealed the existence of profound alterations in the expression of genes involved in key tumoral processes, such as cell survival or angiogenesis. Moreover, sst5TMD4-overexpressing MCF-7 and MDA-MB-231 cells demonstrated increased expression/production of pro-angiogenic factors and enhanced capacity to form mammospheres. Consistently, sst5TMD4-expressing MCF-7 cells induced xenografted tumors with higher VEGF levels and elevated number of blood vessels. Importantly, sst5TMD4 was expressed in a subset of breast cancers, where it correlated with angiogenic markers, lymphatic metastasis, and reduced disease-free survival. These results, coupled to our previous data, support a relevant role of sst5TMD4 in the angiogenic process and reinforce the role of sst5TMD4 in breast cancer malignancy and metastatic potential, supporting its possible utility to develop new molecular biomarkers and drug therapies for these tumors.


Subject(s)
Breast Neoplasms/genetics , Mutation , Neovascularization, Pathologic/genetics , Receptors, Somatostatin/genetics , Adult , Aged , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/blood supply , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Lymphatic Metastasis , MCF-7 Cells , Mice, Nude , Middle Aged , Neovascularization, Pathologic/metabolism , Receptors, Somatostatin/metabolism , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...