Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 137
Filter
3.
Neuro Oncol ; 18(4): 486-96, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26995790

ABSTRACT

BACKGROUND: We recently demonstrated that brain endothelial cells and astrocytes protect cancer cells from chemotherapy through an endothelin-dependent signaling mechanism. Here, we evaluated the efficacy of macitentan, a dual endothelin receptor (ETAR and ETBR) antagonist, in the treatment of experimental breast and lung cancer brain metastases. METHODS: The effect of macitentan on astrocyte- and brain endothelial cell-mediated chemoprotective properties was measured in cytotoxic assays. We compared survival of mice bearing established MDA-MB-231 breast cancer or PC-14 non-small cell lung cancer (NSCLC) brain metastases that were treated with vehicle, macitentan, paclitaxel, or macitentan plus paclitaxel. Cell division, apoptosis, tumor vasculature, and expression of survival-related proteins were assessed by immunofluorescent microscopy. RESULTS: Cancer cells and tumor-associated endothelial cells expressed activated forms of AKT and MAPK in vehicle- and paclitaxel-treated groups in both metastasis models, but these proteins were downregulated in metastases of mice that received macitentan. The survival-related proteins Bcl2L1, Gsta5, and Twist1 that localized to cancer cells and tumor-associated endothelial cells in vehicle- and paclitaxel-treated tumors were suppressed by macitentan. Macitentan or paclitaxel alone had no effect on survival. However, when macitentan was combined with paclitaxel, we noted a significant reduction in cancer cell division and marked apoptosis of both cancer cells and tumor-associated endothelial cells. Moreover, macitentan plus paclitaxel therapy significantly increased overall survival by producing complete responses in 35 of 35 mice harboring brain metastases. CONCLUSIONS: Dual antagonism of ETAR and ETBR signaling sensitizes experimental brain metastases to paclitaxel and may represent a new therapeutic option for patients with brain metastases.


Subject(s)
Breast Neoplasms/drug therapy , Lung Neoplasms/drug therapy , Paclitaxel/pharmacology , Pyrimidines/pharmacology , Receptors, Endothelin/chemistry , Sulfonamides/pharmacology , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Drug Therapy, Combination , Female , Humans , Immunoenzyme Techniques , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Mice , Mice, Nude , NIH 3T3 Cells , Receptors, Endothelin/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Cancer Metastasis Rev ; 34(4): 635-41, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26328524

ABSTRACT

Metastases that are resistant to conventional therapy are the major cause of death from cancer. In most patients, metastasis has already occurred by the time of diagnosis. Thus, the prevention of metastasis is unlikely to be of therapeutic benefit. The biological heterogeneity of metastases presents a major obstacle to treatment. However, the growth and survival of metastases depend on interactions between tumor cells and host homeostatic mechanisms. Targeting these interactions, in addition to the tumor cells, can produce synergistic therapeutic effects against existing metastases.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasm Metastasis/drug therapy , Neoplasm Metastasis/pathology , Neoplasms/pathology , Humans , Intercellular Signaling Peptides and Proteins , Neoplasm Metastasis/prevention & control , Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology
6.
Cancer J ; 21(4): 284-93, 2015.
Article in English | MEDLINE | ID: mdl-26222080

ABSTRACT

Many patients with lung cancer, breast cancer, and melanoma develop brain metastases that are resistant to conventional therapy. The median survival for untreated patients is 1 to 2 months, which may be extended to 6 months with surgery, radiotherapy, and chemotherapy. The outcome of metastasis depends on multiple interactions of unique metastatic cells with host homeostatic mechanisms which the tumor cells exploit for their survival and proliferation. The blood-brain barrier is leaky in metastases that are larger than 0.5-mm diameter because of production of vascular endothelial growth factor by metastatic cells. Brain metastases are surrounded and infiltrated by microglia and activated astrocytes. The interaction with astrocytes leads to up-regulation of multiple genes in the metastatic cells, including several survival genes that are responsible for the increased resistance of tumor cells to cytotoxic drugs. These findings substantiate the importance of the "seed and soil" hypothesis and that successful treatment of brain metastases must include targeting of the organ microenvironment.


Subject(s)
Antineoplastic Agents/therapeutic use , Astrocytes , Blood-Brain Barrier/metabolism , Brain Neoplasms/secondary , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Microglia , Neovascularization, Pathologic/drug therapy , Animals , Brain Neoplasms/blood supply , Brain Neoplasms/drug therapy , Humans , Mice , Neoplasm Metastasis , Neoplasms/drug therapy , Neoplasms/pathology , Tumor Microenvironment , Vascular Endothelial Growth Factor A/antagonists & inhibitors
7.
Clin Cancer Res ; 21(20): 4630-41, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26106074

ABSTRACT

PURPOSE: The objective of the study was to determine whether astrocytes and brain endothelial cells protect glioma cells from temozolomide through an endothelin-dependent signaling mechanism and to examine the therapeutic efficacy of the dual endothelin receptor antagonist, macitentan, in orthotopic models of human glioblastoma. EXPERIMENTAL DESIGN: We evaluated several endothelin receptor antagonists for their ability to inhibit astrocyte- and brain endothelial cell-induced protection of glioma cells from temozolomide in chemoprotection assays. We compared survival in nude mice bearing orthotopically implanted LN-229 glioblastomas or temozolomide-resistant (LN-229(Res) and D54(Res)) glioblastomas that were treated with macitentan, temozolomide, or both. Tumor burden was monitored weekly with bioluminescence imaging. The effect of therapy on cell division, apoptosis, tumor-associated vasculature, and pathways associated with cell survival was assessed by immunofluorescent microscopy. RESULTS: Only dual endothelin receptor antagonism abolished astrocyte- and brain endothelial cell-mediated protection of glioma cells from temozolomide. In five independent survival studies, including temozolomide-resistant glioblastomas, 46 of 48 (96%) mice treated with macitentan plus temozolomide had no evidence of disease (P < 0.0001), whereas all mice in other groups died. In another analysis, macitentan plus temozolomide therapy was stopped in 16 mice after other groups had died. Only 3 of 16 mice eventually developed recurrent disease, 2 of which responded to additional cycles of macitentan plus temozolomide. Macitentan downregulated proteins associated with cell division and survival in glioma cells and associated endothelial cells, which enhanced their sensitivity to temozolomide. CONCLUSIONS: Macitentan plus temozolomide are well tolerated, produce durable responses, and warrant clinical evaluation in glioblastoma patients.


Subject(s)
Dacarbazine/analogs & derivatives , Endothelin Receptor Antagonists/pharmacology , Glioblastoma/drug therapy , Pyrimidines/pharmacology , Sulfonamides/pharmacology , Animals , Cell Division/drug effects , Cell Line , Cell Line, Tumor , Dacarbazine/pharmacology , Down-Regulation/drug effects , Endothelial Cells/drug effects , Endothelial Cells/pathology , Female , Glioblastoma/pathology , Glioma/drug therapy , Glioma/pathology , Humans , Mice , Mice, Nude , NIH 3T3 Cells , Temozolomide
8.
Int J Oncol ; 46(5): 2067-75, 2015 May.
Article in English | MEDLINE | ID: mdl-25776572

ABSTRACT

We investigated how varying the composition of cell culture formulations and growing cancer cells at different densities might affect tumor cell genotype. Specifically, we compared gene expression profiles generated by human MDA­MB­231 breast cancer cells cultured in different media [minimum essential medium (MEM), Dulbecco's modified Eagle's medium (DMEM), or Roswell Park Memorial Institute (RPMI)­1640 medium] containing different concentrations of fetal bovine serum (FBS) or different sera (equine or bovine) that were grown at different cell densities. More than 2,000 genes were differentially modulated by at least a 2­fold difference when MDA­MB­231 cancer cells were 90% confluent and compared with cultures that were 50% confluent. Altering the concentration of serum produced an even more pronounced effect on MDA­MB­231 cancer cell gene expression in that 2,981 genes were differentially expressed in a comparison between cells cultured in 0.1% FBS and same cell density cultures that were maintained in 10% FBS. A comparison between MDA­MB­231 cancer cells that were 90% confluent in MEM, DMEM, or RPMI­1640 media, all containing 10% FBS, resulted in 8,925 differentially expressed genes. Moreover, one­quarter (25.6%) of genes from our genome­wide expression analysis were expressed at significantly different levels by cells grown in MEM, DMEM, or RPMI­1640 media. Genes associated with epithelial­mes-enchymal transition (EMT) were among the genes that were differentially modulated by cells grown in different cell culture formulations and these genes were verified at the protein level. Collectively, these results underscore the importance of accurate reporting and maintenance of uniform culture conditions to ensure reproducible results.


Subject(s)
Cell Culture Techniques/methods , Transcriptome , Blotting, Western , Breast Neoplasms/genetics , Cell Count , Cell Line, Tumor , Genotype , Humans , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction
9.
Nat Cell Biol ; 17(3): 311-21, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25686251

ABSTRACT

Although recent studies have shown that adenosine-to-inosine (A-to-I) RNA editing occurs in microRNAs (miRNAs), its effects on tumour growth and metastasis are not well understood. We present evidence of CREB-mediated low expression of ADAR1 in metastatic melanoma cell lines and tumour specimens. Re-expression of ADAR1 resulted in the suppression of melanoma growth and metastasis in vivo. Consequently, we identified three miRNAs undergoing A-to-I editing in the weakly metastatic melanoma but not in strongly metastatic cell lines. One of these miRNAs, miR-455-5p, has two A-to-I RNA-editing sites. The biological function of edited miR-455-5p is different from that of the unedited form, as it recognizes a different set of genes. Indeed, wild-type miR-455-5p promotes melanoma metastasis through inhibition of the tumour suppressor gene CPEB1. Moreover, wild-type miR-455 enhances melanoma growth and metastasis in vivo, whereas the edited form inhibits these features. These results demonstrate a previously unrecognized role for RNA editing in melanoma progression.


Subject(s)
Adenosine/metabolism , Gene Expression Regulation, Neoplastic , Inosine/metabolism , Melanoma/genetics , RNA Editing , Skin Neoplasms/genetics , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Animals , Base Sequence , Cell Line, Tumor , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Disease Progression , Female , Genes, Reporter , Humans , Luciferases/genetics , Luciferases/metabolism , Melanoma/metabolism , Melanoma/pathology , Mice , Mice, Nude , MicroRNAs , Molecular Sequence Data , Neoplasm Metastasis , Neoplasm Transplantation , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Transcription Factors/genetics , Transcription Factors/metabolism , mRNA Cleavage and Polyadenylation Factors/genetics , mRNA Cleavage and Polyadenylation Factors/metabolism
10.
Cancer Res ; 75(3): 554-65, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25511375

ABSTRACT

Breast cancer brain metastasis is resistant to therapy and a particularly poor prognostic feature in patient survival. Altered metabolism is a common feature of cancer cells, but little is known as to what metabolic changes benefit breast cancer brain metastases. We found that brain metastatic breast cancer cells evolved the ability to survive and proliferate independent of glucose due to enhanced gluconeogenesis and oxidations of glutamine and branched chain amino acids, which together sustain the nonoxidative pentose pathway for purine synthesis. Silencing expression of fructose-1,6-bisphosphatases (FBP) in brain metastatic cells reduced their viability and improved the survival of metastasis-bearing immunocompetent hosts. Clinically, we showed that brain metastases from human breast cancer patients expressed higher levels of FBP and glycogen than the corresponding primary tumors. Together, our findings identify a critical metabolic condition required to sustain brain metastasis and suggest that targeting gluconeogenesis may help eradicate this deadly feature in advanced breast cancer patients.


Subject(s)
Brain Neoplasms/secondary , Breast Neoplasms/pathology , Glucose/chemistry , Amino Acids/chemistry , Animals , Brain/pathology , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation , Cell Survival , Female , Fructose-Bisphosphatase/genetics , Gene Silencing , Glycogen/chemistry , Glycolysis , Humans , Mass Spectrometry , Mice , Mice, Inbred BALB C , Oxygen/chemistry , RNA, Small Interfering/metabolism
11.
J Med Invest ; 61(3-4): 241-53, 2014.
Article in English | MEDLINE | ID: mdl-25264041

ABSTRACT

Brain metastases occur in 20-40% of patients with advanced malignancies and lung cancer is one of the most common causes of brain metastases. The occurrence of brain metastases is associated with poor prognosis and high morbidity in patients with advanced lung cancer, even after intensive multimodal therapy. Progress in treating brain metastases has been hampered by a lack of model systems, a lack of human tissue samples, and the exclusion of brain metastatic patients from many clinical trials. While the biology of brain metastasis is still poorly understood, it is encouraging to see more efforts are beginning to be directed toward the study of brain metastasis. During the multi-step process of metastasis, functional significance of gene expressions, changes in brain vasculature, abnormal secretion of soluble factors and activation of autocrine/paracrine signaling are considered to contribute to the brain metastasis development. A better understanding of the mechanism of this disease will help us to identify the appropriate therapeutic strategies, which leads to circumvent brain metastases. Recent findings on the biology of lung cancer brain metastases and translational leads identified by molecular studies are discussed in this review.


Subject(s)
Brain Neoplasms/secondary , Lung Neoplasms/pathology , Astrocytes/physiology , Brain Neoplasms/therapy , Cadherins/physiology , ErbB Receptors/genetics , Humans , Integrins/physiology , Molecular Targeted Therapy , Polymorphism, Single Nucleotide , Vascular Endothelial Growth Factor A/physiology
12.
Cancer Cell ; 26(1): 77-91, 2014 Jul 14.
Article in English | MEDLINE | ID: mdl-25026212

ABSTRACT

Ovarian cancer has a clear predilection for metastasis to the omentum, but the underlying mechanisms involved in ovarian cancer spread are not well understood. Here, we used a parabiosis model that demonstrates preferential hematogenous metastasis of ovarian cancer to the omentum. Our studies revealed that the ErbB3-neuregulin 1 (NRG1) axis is a dominant pathway responsible for hematogenous omental metastasis. Elevated levels of ErbB3 in ovarian cancer cells and NRG1 in the omentum allowed for tumor cell localization and growth in the omentum. Depletion of ErbB3 in ovarian cancer impaired omental metastasis. Our results highlight hematogenous metastasis as an important mode of ovarian cancer metastasis. These findings have implications for designing alternative strategies aimed at preventing and treating ovarian cancer metastasis.


Subject(s)
Neoplasms, Glandular and Epithelial/secondary , Neoplastic Cells, Circulating/pathology , Omentum/pathology , Ovarian Neoplasms/pathology , Peritoneal Neoplasms/pathology , Animals , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Cell Movement , Cell Proliferation , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Nude , Neoplasm Invasiveness , Neoplasms, Glandular and Epithelial/genetics , Neoplasms, Glandular and Epithelial/metabolism , Neoplasms, Glandular and Epithelial/prevention & control , Neoplastic Cells, Circulating/metabolism , Neuregulin-1/genetics , Neuregulin-1/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/therapy , Parabiosis , Peritoneal Neoplasms/genetics , Peritoneal Neoplasms/metabolism , Peritoneal Neoplasms/prevention & control , RNA Interference , Receptor, ErbB-3/genetics , Receptor, ErbB-3/metabolism , Signal Transduction , Time Factors , Transfection , Xenograft Model Antitumor Assays
13.
Neuro Oncol ; 16(12): 1585-98, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25008093

ABSTRACT

BACKGROUND: Recent evidence suggests that astrocytes protect cancer cells from chemotherapy by stimulating upregulation of anti-apoptotic genes in those cells. We investigated the possibility that activation of the endothelin axis orchestrates survival gene expression and chemoprotection in MDA-MB-231 breast cancer cells and H226 lung cancer cells. METHODS: Cancer cells, murine astrocytes, and murine fibroblasts were grown in isolation, and expression of endothelin (ET) peptides and ET receptors (ETAR and ETBR) compared with expression on cancer cells and astrocytes (or cancer cells and fibroblasts) that were co-incubated for 48 hours. Type-specific endothelin receptor antagonists were used to evaluate the contribution of ETAR and ETBR to astrocyte-induced activation of the protein kinase B (AKT)/mitogen-activated protein kinase (MAPK) signal transduction pathways, anti-apoptotic gene expression, and chemoprotection of cancer cells. We also investigated the chemoprotective potential of brain endothelial cells and microglial cells. RESULTS: Gap junction signaling between MDA-MB-231 cancer cells and astrocytes stimulates upregulation of interleukin 6 (IL-6) and IL-8 expression in cancer cells, which increases ET-1 production from astrocytes and ET receptor expression on cancer cells. ET-1 signals for activation of AKT/MAPK and upregulation of survival proteins that protect cancer cells from taxol. Brain endothelial cell-mediated chemoprotection of cancer cells also involves endothelin signaling. Dual antagonism of ETAR and ETBR is required to abolish astrocyte- and endothelial cell-mediated chemoprotection. CONCLUSIONS: Bidirectional signaling between astrocytes and cancer cells involves upregulation and activation of the endothelin axis, which protects cancer cells from cytotoxicity induced by chemotherapeutic drugs.


Subject(s)
Astrocytes/metabolism , Breast Neoplasms/genetics , Endothelins/genetics , Lung Neoplasms/genetics , Receptors, Endothelin/genetics , 3T3 Cells , Animals , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Survival/genetics , Endothelins/metabolism , Female , Gene Expression , Humans , Lung Neoplasms/metabolism , MAP Kinase Signaling System , Mice , Receptors, Endothelin/metabolism , Up-Regulation
14.
Am J Transl Res ; 5(4): 441-9, 2013.
Article in English | MEDLINE | ID: mdl-23724167

ABSTRACT

Sodium/glucose co-transporter 1 (SGLT1), which actively and energy-dependently uptakes glucose, plays critical roles in the development of various diseases including diabetes mellitus and cancer, and has been viewed as a promising therapeutic target for these diseases. Protein-protein interaction with EGFR has been shown to regulate the expression and activity of SGLT1. Exogenous expression of SGLT1 is one of the essential approaches to characterize its functions; however, exogenously expressed SGLT1 is not firmly detectable by Western blot at its calculated molecular weight, which creates a hurdle for further understanding the molecular events by which SGLT1 is regulated. In this study, we demonstrated that exogenous SGLT1 functions in glucose-uptake normally but is consistently detected near the interface between stacking gel and running gel rather than at the calculated molecular weight in Western blot analysis, suggesting that the overexpressed SGLT1 forms SDS-resistant aggregates, which cannot be denatured and effectively separated on SDS-PAGE. Co-expression of EGFR enhances both the glucose-uptake activity and protein level of the SGLT1. However, fusion with Flag or HA tag at its carboxy- but not its amino-terminus abolished the glucose-uptake activity of exogenous SGLT1 without affecting its protein level. Furthermore, the solubility of SGLT1 aggregates was not affected by other detergents but was partially improved by inhibition of o-link glycosylation. These findings suggested exogenous overexpression of SGLT1 can function normally but may not be consistently detectable at its formula weight due to its gel-shift behavior by forming the SDS-resistant aggregates.

15.
Cancer Lett ; 334(2): 319-27, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23000514

ABSTRACT

Pancreatic cancer is a highly fatal disease characterized by a dominant stroma formation. Exploring new biological targets, specifically those overexpressed in stroma cells, holds significant potential for the design of specific nanocarriers to attain homing of therapeutic and imaging agents to the tumor. In clinical specimens of pancreatic cancer, we found increased expression of CD59 in tumor associated endothelial cells as well as infiltrating cells in the stroma as compared to uninvolved pancreas. We explored this dual targeting effect using orthotopic human pancreatic cancer in nude mice. By immunofluorescence analysis, we confirmed the increased expression of Ly6C, mouse homolog of CD59, in tumor associated endothelial cells as well as in macrophages within the stroma. We decorated the surface of porous silicon nanocarriers with Ly6C antibody. Targeted nanocarriers injected intravenously accumulated to tumor associated endothelial cells within 15min. At 4h after administration, 9.8±2.3% of injected dose/g tumor of the Ly6C targeting nanocarriers accumulated in the pancreatic tumors as opposed to 0.5±1.8% with non-targeted nanocarriers. These results suggest that Ly6C (or CD59) can serve as a novel dual target to deliver therapeutic agents to the stroma of pancreatic tumors.


Subject(s)
Antineoplastic Agents/administration & dosage , Endothelial Cells/metabolism , Macrophages/metabolism , Nanostructures/administration & dosage , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Silicon/administration & dosage , Animals , Antigens, Ly/biosynthesis , Antigens, Ly/metabolism , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , CD59 Antigens/biosynthesis , CD59 Antigens/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Delivery Systems/methods , Endothelial Cells/drug effects , Endothelial Cells/pathology , Female , Humans , Macrophages/drug effects , Macrophages/pathology , Male , Mice , Mice, Nude , Pancreatic Neoplasms/pathology , Silicon/pharmacokinetics
16.
Clin Chem ; 59(1): 180-9, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23115057

ABSTRACT

BACKGROUND: It is estimated that at least 200 000 cases of brain metastases occur each year in the US, which is 10 times the number of patients diagnosed with primary brain tumors. Brain metastasis is associated with poor prognosis, neurological deterioration, diminished quality of life, and extremely short survival. Favorable interactions between tumor cells and cerebral microvascular endothelial cells encourage tumor growth in the central nervous system, while tumor cell interactions with astrocytes protect brain metastases from the cytotoxic effects of chemotherapy. CONTENT: We review the pathogenesis of brain metastasis and emphasize the contributions of microvascular endothelial cells and astrocytes to disease progression and therapeutic resistance. Animal models used to study brain metastasis are also discussed. SUMMARY: Brain metastasis has many unmet clinical needs. There are few clinically relevant tumor models and no targeted therapies specific for brain metastases, and the mean survival for untreated patients is 5 weeks. Improved clinical outcomes are dependent on an enhanced understanding of the metastasis-initiating population of cells and the identification of microenvironmental factors that encourage disease progression in the central nervous system.


Subject(s)
Brain Neoplasms/secondary , Brain Neoplasms/blood supply , Brain Neoplasms/pathology , Endothelium, Vascular/pathology , Humans , Neovascularization, Pathologic
17.
Hum Vaccin Immunother ; 8(8): 1141-2, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22854675

ABSTRACT

Despite significant improvements in diagnosis, surgical techniques, and advancements in general patient care, the majority of deaths from cancer are caused by the continuous growth of metastases that are resistant to conventional therapies. In a large number of cancer patients, metastasis may well have occurred by the time of diagnosis. The metastases can be located in different distant organs and in different regions within a single organ. The major obstacle for the eradication of metastases is the biologic heterogeneity of tumor cells that constitute primary cancers and metastases. Specifically, by the time of diagnosis, malignant neoplasms contain multiple cell populations with diverse biological heterogeneity in growth rate, karyotype, cell surface receptors, antigenicity, immunogenicity, maker enzymes, gene expression, sensitivity to different cytotoxic drugs, invasion, and metastasis. This biologic heterogeneity is not restricted to primary lesions. The cellular composition of metastases in the same organ or in different organs is heterogeneous, both within a single metastasis (intralesional heterogeneity) and among different metastases (interlesional heterogeneity). This heterogeneity is due to two major processes: the selective nature of the metastatic process, and the rapid evolution and phenotypic diversification of clonal tumor cell populations during progressive tumor growth resulting from inherent genetic and epigenetic instability of many clonal populations of tumor cells.


Subject(s)
Antigens, Neoplasm/genetics , Genetic Variation , Neoplasms/genetics , Neoplasms/therapy , Cancer Vaccines/immunology , Cancer Vaccines/isolation & purification , Humans , Immunotherapy/methods
18.
Transl Oncol ; 5(1): 39-47, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22348175

ABSTRACT

Endothelin receptors (ETRs) are often overexpressed in ovarian tumors, which can be resistant to conventional therapies. Thus, we investigated whether blockage of the ETR pathways using the dual ETR antagonist macitentan combined with taxol or cisplatinum can produce therapy for orthotopically growing multidrug-resistant (MDR) human ovarian carcinoma. In several studies, nude mice were injected in the peritoneal cavity with HeyA8-MDR human ovarian cancer cells. Ten days later, mice were randomized to receive vehicle (saline), macitentan (oral, daily), taxol (intraperitoneal, weekly), cisplatinum (intraperitoneal, weekly), macitentan plus taxol, or macitentan plus cisplatinum. Moribund mice were killed, and tumors were collected, weighed, and prepared for immunohistochemical analysis. The HeyA8-MDR tumors did not respond to taxol, cisplatinum, or macitentan administered as single agents. In contrast, combination therapy with macitentan and taxol or macitentan and cisplatinum significantly decreased the tumor incidence and weight and significantly increased the survival of mice and their general condition. Multiple immunohistochemical analyses revealed that treatment with macitentan and macitentan plus taxol or cisplatinum inhibited the phosphorylation of ETRs, decreased the levels of pVEGFR2, pAkt, and pMAPK in tumor cells after 2 weeks of treatment and induced a first wave of apoptosis in tumor-associated endothelial cells followed by apoptosis in surrounding tumor cells. Our study shows that ovarian cancer cells, which express the endothelin axis and are multidrug resistant, are exquisitely sensitive to treatment with a dual ET antagonist and can be resensitized to both taxol and cisplatinum. This combined therapy led to a significant reduction in tumor weight.

19.
Proc Natl Acad Sci U S A ; 108(42): 17456-61, 2011 Oct 18.
Article in English | MEDLINE | ID: mdl-21987811

ABSTRACT

Although the importance of the cellular microenvironment (soil) during invasion and metastasis of cancer cells (seed) has been well-recognized, technical challenges have limited the ability to assess the influence of the microenvironment on cancer cells at the molecular level. Here, we show that an experimental strategy, competitive cross-species hybridization of microarray experiments, can characterize the influence of different microenvironments on cancer cells by independently extracting gene expression data of cancer and host cells when human cancer cells were xenografted into different organ sites of immunocompromised mice. Surprisingly, the analysis of gene expression data showed that the brain microenvironment induces complete reprogramming of metastasized cancer cells, resulting in a gain of neuronal cell characteristics and mimicking neurogenesis during development. We also show that epigenetic changes coincide with transcriptional reprogramming in cancer cells. These observations provide proof of principle for competitive cross-species hybridization of microarray experiments to characterize the effect of the microenvironment on tumor cell behavior.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/secondary , Animals , Astrocytes/metabolism , Base Sequence , Brain Neoplasms/metabolism , Cell Line, Tumor , Coculture Techniques , DNA Methylation , DNA Primers/genetics , DNA, Neoplasm/genetics , Epigenesis, Genetic , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Neurons/metabolism , Nucleic Acid Hybridization , Oligonucleotide Array Sequence Analysis , Species Specificity , Transcriptome , Transplantation, Heterologous , Tumor Microenvironment/genetics
20.
Transl Oncol ; 4(2): 83-91, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21461171

ABSTRACT

There is an urgent need to develop novel markers of pancreatic cancer to facilitate early diagnosis. Pancreatic carcinoma is characterized by marked stroma formation with a high number of infiltrating tumor-associated macrophages (TAMs) that originate from circulating mononuclear cells (MNCs). We hypothesized that differential analysis of protein expression and phosphorylation in circulating MNCs from healthy nude mice and nude mice bearing orthotopic human pancreatic cancer would identify a surrogate marker of pancreatic cancer. These differences were analyzed by two-dimensional gel electrophoresis followed by Western blot analysis using antibody against phosphorylated tyrosine proteins (pY). Protein and phosphorylated protein spots of interest were identified by mass spectrometry and validated by Western blot analysis as candidate markers for pancreatic cancer. We found that the expression and phosphorylation of Src family proteins were significantly higher in circulating MNCs from mice bearing pancreatic cancer than in circulating MNCs from healthy mice. TAMs in mice with pancreatic tumors also had higher Src family protein expression and phosphorylation than resident macrophages in the pancreas of healthy mice. The expression and phosphorylation of Src family proteins were correlated with tumor weight; however, increased Src expression and phosphorylation also occurred in MNCs from mice with chronic pancreatitis. This is the first report to explore novel pancreatic tumor markers in circulating MNCs. Although the specificity of the marker for pancreatic cancer was low, it could be used to monitor the disease or to select high-risk patients with chronic pancreatitis.

SELECTION OF CITATIONS
SEARCH DETAIL