Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Int J Mol Sci ; 22(6)2021 Mar 16.
Article in English | MEDLINE | ID: mdl-33809722

ABSTRACT

Medullary thyroid carcinoma (MTC) is a tumor deriving from the thyroid C cells. Vandetanib (VAN) and cabozantinib (CAB) are two tyrosine kinase inhibitors targeting REarranged during Transfection (RET) and other kinase receptors and are approved for the treatment of advanced MTC. We aim to compare the in vitro and in vivo anti-tumor activity of VAN and CAB in MTC. The effects of VAN and CAB on viability, cell cycle, and apoptosis of TT and MZ-CRC-1 cells are evaluated in vitro using an MTT assay, DNA flow cytometry with propidium iodide, and Annexin V-FITC/propidium iodide staining, respectively. In vivo, the anti-angiogenic potential of VAN and CAB is evaluated in Tg(fli1a:EGFP)y1 transgenic fluorescent zebrafish embryos by analyzing the effects on the physiological development of the sub-intestinal vein plexus and the tumor-induced angiogenesis after TT and MZ-CRC-1 xenotransplantation. VAN and CAB exert comparable effects on TT and MZ-CRC-1 viability inhibition and cell cycle perturbation, and stimulated apoptosis with a prominent effect by VAN in MZ-CRC-1 and CAB in TT cells. Regarding zebrafish, both drugs inhibit angiogenesis in a dose-dependent manner, in particular CAB shows a more potent anti-angiogenic activity than VAN. To conclude, although VAN and CAB show comparable antiproliferative effects in MTC, the anti-angiogenic activity of CAB appears to be more relevant.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Anilides/therapeutic use , Carcinoma, Neuroendocrine/drug therapy , Piperidines/therapeutic use , Pyridines/therapeutic use , Quinazolines/therapeutic use , Thyroid Neoplasms/drug therapy , Zebrafish/physiology , Anilides/pharmacology , Animals , Apoptosis/drug effects , Carcinoma, Neuroendocrine/blood supply , Carcinoma, Neuroendocrine/pathology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , Embryo, Nonmammalian/blood supply , Embryo, Nonmammalian/drug effects , Humans , Neovascularization, Pathologic/drug therapy , Neovascularization, Physiologic/drug effects , Pyridines/pharmacology , Thyroid Neoplasms/blood supply , Thyroid Neoplasms/pathology , Zebrafish/embryology
2.
Int J Mol Sci ; 22(3)2021 Jan 28.
Article in English | MEDLINE | ID: mdl-33525332

ABSTRACT

Serotonin (5-hydroxytryptamine, 5-HT) is a biogenic monoamine produced from the essential amino acid tryptophan. Serotonin's role as a neurotransmitter in the central nervous system and a motility mediator in the gastrointestinal tract has been well defined, and its function in tumorigenesis in various cancers (gliomas, carcinoids, and carcinomas) is being studied. Many studies have shown a potential stimulatory effect of serotonin on cancer cell proliferation, invasion, dissemination, and tumor angiogenesis. Although the underlying mechanism is complex, it is proposed that serotonin levels in the tumor and its interaction with specific receptor subtypes are associated with disease progression. This review article describes serotonin's role in cancer pathogenesis and the utility of the serotonin pathway as a potential therapeutic target in cancer treatment. Octreotide, an inhibitor of serotonin release, is used in well-differentiated neuroendocrine cancers, and the tryptophan hydroxylase (TPH) inhibitor, telotristat, is currently being investigated in clinical trials to treat patients with metastatic neuroendocrine tumors and advanced cholangiocarcinoma. Several in vitro studies have shown the anticancer effect of 5-HT receptor antagonists in various cancers such as prostate cancer, breast cancer, urinary bladder, colorectal cancer, carcinoid, and small-cell lung cancer. More in vivo studies are needed to assess serotonin's role in cancer and its potential use as an anticancer therapeutic target. Serotonin is also being evaluated for its immunoregulatory properties, and studies have shown its potential anti-inflammatory effect. Therefore, it would be of interest to explore the combination of serotonin antagonists with immunotherapy in the future.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Carcinoma, Neuroendocrine/drug therapy , Cholangiocarcinoma/drug therapy , Neovascularization, Pathologic/drug therapy , Serotonin/metabolism , Signal Transduction/drug effects , Carcinoma, Neuroendocrine/blood supply , Carcinoma, Neuroendocrine/metabolism , Carcinoma, Neuroendocrine/pathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cholangiocarcinoma/blood supply , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/pathology , Gene Expression Regulation, Neoplastic , Humans , Molecular Targeted Therapy/methods , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Octreotide/therapeutic use , Phenylalanine/analogs & derivatives , Phenylalanine/therapeutic use , Pyrimidines/therapeutic use , Receptors, Serotonin/genetics , Receptors, Serotonin/metabolism , Serotonin Antagonists/therapeutic use , Signal Transduction/genetics , Tryptophan Hydroxylase/genetics , Tryptophan Hydroxylase/metabolism , Tumor Cells, Cultured
3.
PLoS One ; 16(1): e0245602, 2021.
Article in English | MEDLINE | ID: mdl-33471819

ABSTRACT

With the advent of potent second-line anti-androgen therapy, we and others have observed an increased incidence of androgen receptor (AR)-null small cell or neuroendocrine prostate cancer (SCNPC) in metastatic castration-resistant prostate cancer (mCRPC). Our study was designed to determine the effect of cabozantinib, a multi-targeted tyrosine kinase inhibitor that inhibits VEGFR2, MET and RET on SCNPC. Transcriptome analysis of the University of Washington rapid autopsy and SU2C mCRPC datasets revealed upregulated MET and RET expression in SCNPCs relative to adenocarcinomas. Additionally, increased MET expression correlated with attenuated AR expression and activity. In vitro treatment of SCNPC patient-derived xenograft (PDX) cells with the MET inhibitor AMG-337 had no impact on cell viability in LuCaP 93 (MET+/RET+) and LuCaP 173.1 (MET-/RET-), whereas cabozantinib decreased cell viability of LuCaP 93, but not LuCaP 173.1. Notably, MET+/RET+ LuCaP 93 and MET-/RET- LuCaP 173.1 tumor volumes were significantly decreased with cabozantinib treatment in vivo, and this activity was independent of MET or RET expression in LuCaP 173.1. Tissue analysis indicated that cabozantinib did not inhibit tumor cell proliferation (Ki67), but significantly decreased microvessel density (CD31) and increased hypoxic stress and glycolysis (HK2) in LuCaP 93 and LuCaP 173.1 tumors. RNA-Seq and gene set enrichment analysis revealed that hypoxia and glycolysis pathways were increased in cabozantinib-treated tumors relative to control tumors. Our data suggest that the most likely mechanism of cabozantinib-mediated tumor growth suppression in SCNPC PDX models is through disruption of the tumor vasculature. Thus, cabozantinib may represent a potential therapy for patients with metastatic disease in tumor phenotypes that have a significant dependence on the tumor vasculature for survival and proliferation.


Subject(s)
Anilides/pharmacology , Carcinoma, Neuroendocrine , Neovascularization, Pathologic , Prostatic Neoplasms , Pyridines/pharmacology , Animals , Carcinoma, Neuroendocrine/blood supply , Carcinoma, Neuroendocrine/drug therapy , Carcinoma, Neuroendocrine/metabolism , Carcinoma, Neuroendocrine/pathology , Cell Line, Tumor , Humans , Male , Mice , Mice, SCID , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-met/metabolism , Proto-Oncogene Proteins c-ret/metabolism , Xenograft Model Antitumor Assays
4.
Semin Cancer Biol ; 71: 109-121, 2021 06.
Article in English | MEDLINE | ID: mdl-32428715

ABSTRACT

Metastatic melanoma is a fatal disease with a rapid systemic dissemination. The most frequent target sites are the liver, bone, and brain. Melanoma metastases represent a heterogeneous cell population, which associates with genomic instability and resistance to therapy. Interaction of melanoma cells with the hepatic sinusoidal endothelium initiates a signaling cascade involving cytokines, growth factors, bioactive lipids, and reactive oxygen and nitrogen species produced by the cancer cell, the endothelium, and also by different immune cells. Endothelial cell-derived NO and H2O2 and the action of immune cells cause the death of most melanoma cells that reach the hepatic microvascularization. Surviving melanoma cells attached to the endothelium of pre-capillary arterioles or sinusoids may follow two mechanisms of extravasation: a) migration through vessel fenestrae or b) intravascular proliferation followed by vessel rupture and microinflammation. Invading melanoma cells first form micrometastases within the normal lobular hepatic architecture via a mechanism regulated by cross-talk with the stroma and multiple microenvironment-related molecular signals. In this review special emphasis is placed on neuroendocrine (systemic) mechanisms as potential promoters of liver metastatic growth. Growing metastatic cells undergo functional and metabolic changes that increase their capacity to withstand oxidative/nitrosative stress, which favors their survival. This adaptive process also involves upregulation of Bcl-2-related antideath mechanisms, which seems to lead to the generation of more resistant cell subclones.


Subject(s)
Carcinoma, Neuroendocrine/secondary , Endothelium/pathology , Liver Neoplasms/secondary , Melanoma/pathology , Oxidative Stress , Tumor Microenvironment , Animals , Carcinoma, Neuroendocrine/blood supply , Cell Survival , Humans , Liver Neoplasms/blood supply , Oxidation-Reduction
5.
BMC Med Imaging ; 20(1): 46, 2020 05 03.
Article in English | MEDLINE | ID: mdl-32362278

ABSTRACT

BACKGROUND: Large cell neuroendocrine carcinoma (LCNEC) of the urinary bladder is an uncommon malignant bladder tumor, and the overall prognosis is poor. Contrast-enhanced ultrasound (CEUS) provides a new effective modality for tumor detection and diagnosis. CASE PRESENTATION: A 30-year-old man complained of repeated painless gross haematuria for half a month. Conventional ultrasound demonstrated a hypoechoic solitary lesion with hyperechoic margins measuring 3.4 × 3.1 cm in the anterior wall of the bladder. Superb microvascular imaging (SMI) showed a strong flow signal in the mass. CEUS revealed that the lesion was characterized by hyper-enhancement in the early phase and hypo-enhancement in the late phase. The entire bladder wall was disrupted by homogeneous hyper-enhanced tumor tissue on CEUS. Time-intensity curves (TICs) showed a rapid wash-in with a high maximum signal intensity (SI) and quick wash-out. Finally, partial cystectomy was performed and the pathological examination confirmed the diagnosis of LCNEC with invasion into the whole layer of the bladder wall. CONCLUSION: This case suggested that CEUS was a valuable imaging method to detect and diagnose LCNEC in the bladder, and that CEUS can provide information related to the depth of wall invasion and the microvasculature.


Subject(s)
Carcinoma, Large Cell/diagnostic imaging , Carcinoma, Neuroendocrine/diagnostic imaging , Urinary Bladder Neoplasms/diagnostic imaging , Adult , Carcinoma, Large Cell/blood supply , Carcinoma, Large Cell/complications , Carcinoma, Large Cell/surgery , Carcinoma, Neuroendocrine/blood supply , Carcinoma, Neuroendocrine/complications , Carcinoma, Neuroendocrine/surgery , Contrast Media/administration & dosage , Cystectomy , Hematuria/etiology , Humans , Male , Microvessels/diagnostic imaging , Microvessels/pathology , Ultrasonography , Urinary Bladder Neoplasms/blood supply , Urinary Bladder Neoplasms/complications , Urinary Bladder Neoplasms/surgery
6.
Acta Radiol ; 60(4): 425-432, 2019 Apr.
Article in English | MEDLINE | ID: mdl-29950111

ABSTRACT

BACKGROUND: Virtual monochromatic images (VMI) generated using spectral computed tomography (CT) are promising recently available tools to improve diagnostic performance in oncologic patients. PURPOSE: To investigate if virtual monochromatic datasets are suitable for clinical routine use in patients with hypervascularized abdominal tumors. MATERIAL AND METHODS: A total of 41 patients with hypervascularized hepatocellular carcinoma (HCC), renal cell carcinoma (RCC), or neuroendocrine tumors (NET) were enrolled in the study; 451 CT series were analyzed. In an intra-individual study design, virtual monochromatic datasets of the arterial phase of each scan were computed. Image quality was assessed objectively by determining signal-to-noise ratio (SNR) and contrast-to-noise ratios (CNR) and subjectively by using five-point Likert-scales. The volume CT dose index (CTDIvol) was taken from each radiation dose report. The increase in reading time was estimated from the increase in the number of images. RESULTS: Intra-individual comparison of the spectral mode in the arterial phase with the portal venous phase revealed no significant increase in the applied dose. SNR, CNRtumor-to-liver , and CNRtumor-to-muscle were significantly increased by lowering virtual monochromatic energy. Subjective image quality scores revealed an increase of contrast in low energy datasets, resulting in significantly higher diagnostic confidence, but an increased image noise at low energies. While diagnostic confidence improved, taking all datasets into account resulted in a significantly longer estimated reading time. CONCLUSION: In clinical practice, the use of low energy VMI improved diagnostic confidence without a significant increase in dose. The main disadvantage is a decrease in efficiency due to longer reading times.


Subject(s)
Abdominal Neoplasms/diagnostic imaging , Carcinoma, Hepatocellular/diagnostic imaging , Carcinoma, Neuroendocrine/diagnostic imaging , Carcinoma, Renal Cell/diagnostic imaging , Liver Neoplasms/diagnostic imaging , Tomography, X-Ray Computed/methods , Abdominal Neoplasms/blood supply , Aged , Carcinoma, Hepatocellular/blood supply , Carcinoma, Neuroendocrine/blood supply , Carcinoma, Renal Cell/blood supply , Female , Humans , Liver/blood supply , Liver/diagnostic imaging , Liver Neoplasms/blood supply , Male , Middle Aged , Neovascularization, Pathologic/diagnostic imaging , Reproducibility of Results , Sensitivity and Specificity , Signal-To-Noise Ratio , Stomach/blood supply
7.
A A Case Rep ; 8(11): 300-303, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28328586

ABSTRACT

The percutaneous isolated hepatic perfusion utilizes a venovenous bypass to administer high-dose chemotherapy exclusively in the liver, getting depurated through a hemofilter before returning to the systemic circulation. The hepatic perfusion is managed under general anesthesia and invasive monitoring as a result of very abrupt changes in venous return and vascular resistances because of the isolation of the hepatic territory and absorption of circulating catecholamines by the hemofilter. We report a case in which we describe the technique, physiologic implications, anesthetic, and goal-directed hemodynamic management for this procedure.


Subject(s)
Anesthesia, General/methods , Carcinoma, Neuroendocrine/drug therapy , Catheterization, Central Venous , Chemotherapy, Cancer, Regional Perfusion/methods , Hemodynamics , Hemofiltration , Liver Circulation , Liver Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Carcinoma, Neuroendocrine/blood supply , Carcinoma, Neuroendocrine/diagnostic imaging , Carcinoma, Neuroendocrine/secondary , Humans , Liver Neoplasms/blood supply , Liver Neoplasms/diagnostic imaging , Liver Neoplasms/secondary , Middle Aged , Treatment Outcome
8.
Methods Mol Biol ; 1464: 151-161, 2016.
Article in English | MEDLINE | ID: mdl-27858364

ABSTRACT

The Rip1Tag2 transgenic mouse model of ß-cell carcinogenesis has been instrumental in studying various aspects of tumor angiogenesis and in investigating the response to anti-angiogenic therapeutics. Thereby, the in-depth assessment of blood and lymphatic vessel phenotypes and functionality represents key experimental analyses. In this chapter, we describe basic protocols to assess tumor blood vessel morphology (pericyte coverage), functionality (perfusion, leakiness, and hypoxia), lymphatic tumor coverage, and tumor cell proliferation and apoptosis based on immunofluorescence microscopy analysis.


Subject(s)
Carcinoma, Neuroendocrine/genetics , Insulinoma/genetics , Neovascularization, Pathologic/pathology , Pancreatic Neoplasms/genetics , Animals , Antigens, Viral, Tumor/genetics , Apoptosis , Carcinoma, Neuroendocrine/blood supply , Carcinoma, Neuroendocrine/pathology , Cell Proliferation , Insulin/genetics , Insulinoma/blood supply , Insulinoma/pathology , Mice , Mice, Transgenic , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/genetics , Neovascularization, Pathologic/genetics , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/pathology , Promoter Regions, Genetic
9.
J Cancer Res Clin Oncol ; 142(5): 905-12, 2016 May.
Article in English | MEDLINE | ID: mdl-26696594

ABSTRACT

PURPOSE: Histological vascular invasion (VI) is major predictor of recurrence after surgery for several cancer types. We previously reported that VI with abundant stromal cell infiltrates was a negative prognostic factor in lung adenocarcinoma. This study examined whether stromal cell infiltrates within VI are associated with prognosis in high-grade neuroendocrine carcinoma (HGNEC) of the lung. METHODS: We investigated the relationship between the frequency and density of VI and recurrence-free survival (RFS) of 60 resected HGNEC patients without adjuvant chemotherapy. We examined prognostic effects of CD204 (+) macrophages, CD34 (+) microvessels and α-smooth muscle actin (+) myofibroblasts within VI. Correlation between expressions of stem cell-related molecules (ALDH-1, Notch 1, CD44) and epithelial-mesenchymal transition-related molecules (E-cadherin, S100A4) in cancer cells within VI and RFS was also analyzed. RESULTS: Among 60 cases, 51 cases showed VI. Although the presence of VI was a significant predictor for worse RFS (P = 0.04), the frequency and density of VI were not correlated with RFS. The number of CD204 (+) macrophage, CD34 (+) microvessels and α-smooth muscle actin (+) myofibroblasts within VI did not influence on the RFS. On the contrary, cases with higher Notch 1 expression in cancer cells in the VI displayed significantly shorter RFS than lower expression group (7.6 vs. 29.4 months, P = 0.02). CONCLUSIONS: The current study revealed the presence of stromal cells within VI was not significant predictor for recurrence in HGNEC. This suggests that in HGNEC, unlike adenocarcinoma, intravascular stromal cells are not major contributors to metastasis.


Subject(s)
Adenocarcinoma/pathology , Carcinoma, Neuroendocrine/pathology , Lung Neoplasms/pathology , Neovascularization, Pathologic/pathology , Actins/metabolism , Adenocarcinoma/blood supply , Adenocarcinoma/drug therapy , Aged , Antigens, CD34/metabolism , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/metabolism , Cadherins/metabolism , Carcinoma, Neuroendocrine/blood supply , Carcinoma, Neuroendocrine/drug therapy , Chemotherapy, Adjuvant , Epithelial-Mesenchymal Transition/drug effects , Female , Follow-Up Studies , Humans , Immunoenzyme Techniques , Lung Neoplasms/blood supply , Lung Neoplasms/drug therapy , Macrophages/pathology , Male , Myofibroblasts/pathology , Neoplasm Grading , Neoplasm Staging , Neovascularization, Pathologic/metabolism , Prognosis , Scavenger Receptors, Class A/metabolism , Stromal Cells/metabolism , Stromal Cells/pathology , Survival Rate
11.
Target Oncol ; 7(2): 99-106, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22585431

ABSTRACT

Neuroendocrine carcinomas are rare neoplasms although of increasing incidence and concern. While traditionally considered of indolent nature, once they progress beyond surgical resectability, the outcome is ultimately fatal for the majority of patients. Somatostatin analogs are useful to control symptoms in functioning tumors and may slow tumor progression in certain disease settings, but sensitivity to conventional cytotoxic chemotherapy is rather limited. In this context, results of the recently published randomized trials with sunitinib and everolimus have demonstrated for the first time that there are agents able to positively impact on the natural history of this complex disease. In this review, we will discuss available data on angiogenesis and mammalian target of rapamycin inhibitors for the treatment of advanced well-differentiated gastroenteropancreatic neuroendocrine tumors.


Subject(s)
Carcinoma, Neuroendocrine/drug therapy , Gastrointestinal Neoplasms/drug therapy , Pancreatic Neoplasms/drug therapy , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Neuroendocrine/blood supply , Clinical Trials as Topic , Everolimus , Gastrointestinal Neoplasms/blood supply , Humans , Indoles/administration & dosage , Indoles/therapeutic use , Molecular Targeted Therapy , Neovascularization, Pathologic/drug therapy , Pancreatic Neoplasms/blood supply , Pyrroles/administration & dosage , Pyrroles/therapeutic use , Randomized Controlled Trials as Topic , Sirolimus/administration & dosage , Sirolimus/analogs & derivatives , Sirolimus/therapeutic use , Sunitinib , TOR Serine-Threonine Kinases/antagonists & inhibitors
13.
J Surg Res ; 154(1): 68-77, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-18708196

ABSTRACT

BACKGROUND: Clinical observations suggest that in neuroendocrine digestive tumors a high intratumoral microvascular density is associated with good prognosis. We used an experimental orthotopic xenograft model to analyze the relations between angiogenic activity and tumor progression in this tumor subset. MATERIAL AND METHODS: We compared 2 endocrine cell lines: STC-1, a low vascular endothelial growth factor (VEGF)-producing cell line, and INS-r3, a high VEGF-producing cell line. Tumor cells were grafted in the adventitial layer of the caecal wall of nude mice, sacrificed after 8 wk. RESULTS: At 8 wk, "primary" tumors were present in all animals. STC-1 derived tumors were morphologically moderately differentiated, with high proliferative and apoptotic activities; in contrast, INS-r3 derived tumors were well differentiated, with low proliferative and apoptotic activities. VEGF was expressed in <50% grafted STC-1 cells but in >90% of grafted INS-r3 cells. Microvascular density was significantly higher in INS-r3 derived tumors than in STC-1 derived tumors. All STC-1 derived tumors (n = 8) have invaded the mucosa, in contrast to none of the INS-r3 derived tumors (n = 8); liver metastases were detected in 7/8 animals bearing STC-1 derived tumors and in 0/8 animals with INS-r3 derived tumors, despite the presence of lymph node metastases. CONCLUSIONS: Our experimental data concur with clinical findings to suggest that in well differentiated digestive neuroendocrine tumors angiogenesis is disconnected from tumor progression: the development of a highly vascular tumor microenvironment is correlated with VEGF secretion but is not associated with invasive and metastatic properties; it must therefore be regarded as an indirect marker of differentiation.


Subject(s)
Carcinoma, Neuroendocrine/pathology , Digestive System Neoplasms/pathology , Intestinal Neoplasms/pathology , Neovascularization, Pathologic/pathology , Animals , Antigens, Viral, Tumor/genetics , Carcinoma, Neuroendocrine/blood supply , Cell Line, Tumor , Digestive System Neoplasms/blood supply , Disease Progression , Glucagon/genetics , Insulin/genetics , Intestinal Neoplasms/blood supply , Lymphatic Metastasis/pathology , Mice , Mice, Nude , Microcirculation , Promoter Regions, Genetic , Rats , Simian virus 40/immunology , Transplantation, Heterologous , Vascular Endothelial Growth Factor A/genetics
14.
J Vasc Interv Radiol ; 19(1): 145-51, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18192482

ABSTRACT

Hepatic artery radioembolization was performed in a cohort of patients with unresectable neuroendocrine hepatic metastases who exhibited hepatic progression or toxicity despite technically adequate embolization procedures without other reasonable therapeutic options. Eight patients (five men) with a median age of 55.5 years met the study criteria. Infusions of yttrium-90 resin microspheres were performed in a lobar fashion. Standard clinical, laboratory, and imaging follow-up was performed. Median hepatic parenchyma replacement by tumor was 55% (range, 25%-60%). Twelve (90)Y resin microsphere infusions were performed, and the median delivered activity was 33.25 mCi (range, 23-55 mCi). One partial response, four cases of disease stabilization, and three cases of progressive disease were noted. No cases of radiation-induced liver disease occurred. Median survival times were 14 months (range, 3-15 months) from the time of (90)Y microsphere treatment and 36.5 months (range, 16-105 months) from the time of diagnosis of hepatic metastases. In this cohort, (90)Y microsphere radioembolization of neuroendocrine hepatic metastases was not precluded by previous nonradioactive embolization procedures, but the effectiveness in this population requires further investigation.


Subject(s)
Brachytherapy/methods , Carcinoma, Neuroendocrine/therapy , Chemoembolization, Therapeutic/methods , Liver Neoplasms/therapy , Microspheres , Yttrium Radioisotopes/administration & dosage , Aged , Angiography , Brachytherapy/adverse effects , Carcinoma, Neuroendocrine/blood supply , Carcinoma, Neuroendocrine/mortality , Carcinoma, Neuroendocrine/radiotherapy , Carcinoma, Neuroendocrine/secondary , Chemoembolization, Therapeutic/adverse effects , Female , Follow-Up Studies , Humans , Liver Neoplasms/blood supply , Liver Neoplasms/mortality , Liver Neoplasms/radiotherapy , Liver Neoplasms/secondary , Male , Middle Aged , Retrospective Studies , Time Factors , Tomography, Emission-Computed, Single-Photon , Tomography, X-Ray Computed , Treatment Outcome
15.
Cancer ; 109(8): 1478-86, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17340592

ABSTRACT

BACKGROUND: Vascular endothelial growth factor (VEGF) is a critical proangiogenic factor in solid tumors. However, its expression and role in human neuroendocrine tumor development and progression remains unclear. METHODS: Using immunohistochemistry, VEGF and Sp1 expression patterns were investigated in 50 cases of human gastrointestinal neuroendocrine tumor having various clinicopathologic characteristics. RESULTS: It was found that strong VEGF expression was detected in tumor cells, whereas no or very weak VEGF expression was detected in stromal cells surrounding or within the tumors. The levels of VEGF expression directly correlated with the expression levels of Sp1 and microvessel density. Strong, weak, and negative VEGF expression was observed in 32%, 54%, and 14% of cases, respectively. Compared with the group with negative VEGF expression, VEGF (weak/strong) expression was associated with metastasis (14% versus 58%; P = .03). The median progression-free survival (PFS) durations of patients with strong and weak VEGF expression were 29 months and 81 months, respectively. With a median follow-up duration of 50 months, the median PFS duration for the group with negative VEGF expression has not been reached. Compared with the log-rank test, VEGF expression was associated with poor PFS (P = .02). Using in vitro and in vivo models, human carcinoid cell lines were treated with bevacizumab, a monoclonal antibody targeting VEGF. Bevacizumab did not inhibit the growth of carcinoid cells in vitro but significantly reduced tumor angiogenesis and impaired tumor growth in animals. CONCLUSIONS: The data suggest that overexpression of VEGF promotes the growth of human neuroendocrine tumors in part through up-regulation of angiogenesis.


Subject(s)
Carcinoma, Neuroendocrine/blood supply , Carcinoma, Neuroendocrine/metabolism , Neovascularization, Pathologic/metabolism , Vascular Endothelial Growth Factor A/biosynthesis , Angiogenesis Inhibitors/pharmacology , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Bevacizumab , Biomarkers, Tumor/analysis , Blotting, Western , Carcinoma, Neuroendocrine/mortality , Cell Line, Tumor , Cell Proliferation/drug effects , Disease-Free Survival , Female , Humans , Immunohistochemistry , Mice , Mice, Nude , Prognosis
16.
Am J Pathol ; 168(3): 991-1003, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16507913

ABSTRACT

Capillaries expressing the laminin alpha2 chain in basement membranes may be considered early developing vessels in normal and neoplastic human tissues. Therefore, we investigated whether up-regulation of this extracellular matrix protein favors transendothelial migration of neoplastic cells and then metastasis. In lung small and large cell neuroendocrine carcinomas, which exhibit a stronger metastatic tendency among carcinomas, laminin alpha2 chain-positive vessels were more numerous than in carcinoid tumors and supraglottis, breast, and lung non-small cell carcinomas, suggesting a direct relationship between these vessels and metastasis. In vitro studies showed that epidermal growth factor (EGF) induced a more efficient migration of the AE-2 lung neuroendocrine carcinoma cell line through the purified laminin alpha2 chain rather than through the laminin beta1 chain and fibronectin. AE-2 cells constitutively expressed all EGF receptors and the alpha6beta1 integrin, which is one of the laminin alpha2 chain receptors. EGF up-regulated alpha6beta1 expression in several tumors. In this regard, we show that EGF increased the chemo-kinetic migration of AE-2 cells through EAHY endothelial monolayers, which was inhibited by the anti-alpha6 integrin chain monoclonal antibody. These data indicate that laminin alpha2 chain and alpha6beta1 may be mutually involved in EGF-dependent migration of AE-2 cells and that laminin alpha2 chain-positive vessels may favor metastasis of EGF-dependent tumors.


Subject(s)
Carcinoma, Neuroendocrine/pathology , Epidermal Growth Factor/metabolism , Laminin/metabolism , Lung Neoplasms/pathology , Antibodies, Monoclonal/pharmacology , Capillaries/chemistry , Carcinoid Tumor/blood supply , Carcinoid Tumor/metabolism , Carcinoid Tumor/pathology , Carcinoma, Neuroendocrine/blood supply , Carcinoma, Neuroendocrine/metabolism , Cell Line, Tumor , Cell Movement , Epidermal Growth Factor/genetics , Gene Expression , Humans , Laminin/analysis , Laminin/antagonists & inhibitors , Lung Neoplasms/blood supply , Lung Neoplasms/metabolism , Models, Biological , Neoplasm Invasiveness , Neoplasm Metastasis , Up-Regulation
17.
Am J Surg Pathol ; 29(3): 324-8, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15725800

ABSTRACT

Few data on the influence of vessel invasion on the progression of neuroendocrine lung tumors are available. Because of the lack of specific markers, previous studies could not reliably discriminate lymphatic and blood vessels. By immunostaining for podoplanin, specific for lymphatic endothelium, and CD34 antigen, we assessed lymphatic and blood vessel invasion in 120 tissue specimens of patients with neuroendocrine lung tumors. Lymphovascular invasion was correlated with clinicopathologic parameters, and its prognostic relevance was evaluated. Lymphatic vessels were identified exclusively at the tumor invasion front, whereas blood capillaries were also seen within tumors. Lymphatic vessel as well as lymphatic and blood vessel invasion was prevalent in patients with high-grade neuroendocrine tumors and advanced tumor stages, closely associated with lymph node metastases (P < 0.0001). In univariate analysis, these two invasion types correlated with decreased disease-free survival (both P < 0.0001), whereas blood vessel invasion alone did not. In multivariate analysis, only tumor grade and lymph node status remained statistically significant factors for prognosis (P = 0.016 and P < 0.0001). Our results suggest that evaluation of lymphatic vessel invasion is important in neuroendocrine lung tumors serving as a prognostic parameter for disease-free survival.


Subject(s)
Blood Vessels/metabolism , Carcinoma, Neuroendocrine/blood supply , Lung Neoplasms/blood supply , Lymphatic Vessels/metabolism , Adolescent , Adult , Aged , Biomarkers/analysis , Blood Vessels/pathology , Carcinoma, Neuroendocrine/secondary , Carcinoma, Neuroendocrine/surgery , Child , Disease-Free Survival , Endothelium, Lymphatic/metabolism , Endothelium, Lymphatic/pathology , Female , Humans , Immunohistochemistry , Lung Neoplasms/pathology , Lung Neoplasms/surgery , Lymph Nodes/metabolism , Lymph Nodes/pathology , Lymphatic Metastasis/pathology , Lymphatic Vessels/pathology , Male , Membrane Glycoproteins/metabolism , Middle Aged , Neoplasm Invasiveness/pathology
18.
Exp Clin Endocrinol Diabetes ; 109(7): 378-85, 2001.
Article in English | MEDLINE | ID: mdl-11573150

ABSTRACT

We report the case of a 34 year old male presenting with symptomatic hypercalcemia due to excessive PTHrP secretion from a pancreatic neuroendocrine carcinoma with extensive hypervascularization and without any evidence for metastatic disease. In the early phase of the disease conventional chemotherapy with streptozocin and doxorubicin was able to control functional activity as well as tumor growth. However, after 2 years tumor escape was indicated by severe therapy-resistant hypercalcemia. Therapeutic options were reduced due to the excessive tumor vascularization and the patient died from his disease after a short period of intensified therapy. The role of PTHrP in hypercalcemia of malignancy (HHM) and its association with neuroendocrine pancreatic tumors as well as possible therapeutic options are reviewed.


Subject(s)
Carcinoma, Neuroendocrine/metabolism , Hypercalcemia/etiology , Neovascularization, Pathologic/physiopathology , Pancreatic Neoplasms/metabolism , Proteins/metabolism , Adult , Carcinoma, Neuroendocrine/blood supply , Carcinoma, Neuroendocrine/therapy , Fatal Outcome , Humans , Male , Neovascularization, Pathologic/therapy , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/therapy , Parathyroid Hormone-Related Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...