Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Cancer Cell ; 41(2): 340-355.e6, 2023 02 13.
Article in English | MEDLINE | ID: mdl-36787696

ABSTRACT

Effective therapeutic strategies are needed for non-small cell lung cancer (NSCLC) patients with epidermal growth factor receptor (EGFR) mutations that acquire resistance to EGFR tyrosine kinase inhibitors (TKIs) mediated by epithelial-to-mesenchymal transition (EMT). We investigate cell surface proteins that could be targeted by antibody-based or adoptive cell therapy approaches and identify CD70 as being highly upregulated in EMT-associated resistance. Moreover, CD70 upregulation is an early event in the evolution of resistance and occurs in drug-tolerant persister cells (DTPCs). CD70 promotes cell survival and invasiveness, and stimulation of CD70 triggers signal transduction pathways known to be re-activated with acquired TKI resistance. Anti-CD70 antibody drug conjugates (ADCs) and CD70-targeting chimeric antigen receptor (CAR) T cell and CAR NK cells show potent activity against EGFR TKI-resistant cells and DTPCs. These results identify CD70 as a therapeutic target for EGFR mutant tumors with acquired EGFR TKI resistance that merits clinical investigation.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , CD27 Ligand/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Epithelial-Mesenchymal Transition/genetics , ErbB Receptors/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Mutation , /therapeutic use
2.
Pancreatology ; 21(1): 115-123, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33341341

ABSTRACT

Glutathione-S-transferases (GSTs) not only show cytoprotective role and their involvement in the development of anticancer drug resistance, but also transmit signals that control cell proliferation and apoptosis. However, the role of GST isoforms in chemotherapy resistance remains elusive in pancreatic cancer. Here, we demonstrated that gemcitabine treatment increased the GSTM2 expression in pancreatic cancer cell lines. Knockdown of GSTM2 by siRNA elevated apoptosis and decreased viability of pancreatic cancer cells treated with gemcitabine. Moreover, in vivo experiments further showed that shRNA induced GSTM2 downregulation enhanced drug sensitivity of gemcitabine in orthotopic pancreatic tumor mice. We also found that GSTM2 levels were lower in tumor tissues than in non-tumor tissues and higher GSTM2 expression was significantly associated with longer overall survival. In conclusion, our findings indicate that GSTM2 expression is essential for the survival of pancreatic cancer cells undergoing gemcitabine treatment and leads to chemo resistance. Downregulation of GSTM2 in pancreatic cancer may benefit gemcitabine treatment. GSTM2 expression in patients also shows significant correlation with overall survival. Thus, our study suggests that GSTM2 is a potential target for chemotherapy optimization and prognostic biomarker of pancreatic cancer.


Subject(s)
Deoxycytidine/analogs & derivatives , Down-Regulation , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Glutathione Transferase/metabolism , Pancreatic Neoplasms/drug therapy , Antineoplastic Agents/pharmacology , Biomarkers, Tumor , Deoxycytidine/pharmacology , Drug Resistance, Neoplasm/genetics , Genetic Markers , Glutathione Transferase/genetics , Humans , RNA Interference , Gemcitabine
3.
Sci Transl Med ; 10(464)2018 10 24.
Article in English | MEDLINE | ID: mdl-30355799

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) has a dismal prognosis, and it is unclear whether its stromal infiltrate contributes to its aggressiveness. Here, we demonstrate that Dickkopf-3 (DKK3) is produced by pancreatic stellate cells and is present in most human PDAC. DKK3 stimulates PDAC growth, metastasis, and resistance to chemotherapy with both paracrine and autocrine mechanisms through NF-κB activation. Genetic ablation of DKK3 in an autochthonous model of PDAC inhibited tumor growth, induced a peritumoral infiltration of CD8+ T cells, and more than doubled survival. Treatment with a DKK3-blocking monoclonal antibody inhibited PDAC progression and chemoresistance and prolonged survival. The combination of DKK3 inhibition with immune checkpoint inhibition was more effective in reducing tumor growth than either treatment alone and resulted in a durable improvement in survival, suggesting that DKK3 neutralization may be effective as a single targeted agent or in combination with chemotherapy or immunotherapy for PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Disease Progression , Intercellular Signaling Peptides and Proteins/metabolism , Adaptor Proteins, Signal Transducing , Animals , Antibodies, Neutralizing/pharmacology , Antibodies, Neutralizing/therapeutic use , Autocrine Communication/drug effects , Carcinoma, Pancreatic Ductal/drug therapy , Cell Line, Tumor , Chemokines , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Disease Models, Animal , Drug Resistance, Neoplasm/drug effects , Gene Silencing , Humans , Immunotherapy , Mice, Inbred C57BL , Mice, Nude , NF-kappa B/metabolism , Neutralization Tests , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/drug effects , Pancreatic Stellate Cells/metabolism , Pancreatic Stellate Cells/pathology , Paracrine Communication/drug effects , Survival Analysis , Gemcitabine , Pancreatic Neoplasms
4.
Cell Physiol Biochem ; 39(2): 740-50, 2016.
Article in English | MEDLINE | ID: mdl-27459514

ABSTRACT

BACKGROUND/AIMS: The cell surface protein transmembrane 4 L6 family member 1 (TM4SF1) has been detected in various tumors and plays a major role in the development of cancer. We aimed to investigate the effects of TM4SF1 on the migration and invasion of pancreatic cancer in vitro and in vivo and explore its related molecular mechanisms. METHODS: qRT-PCR and immunohistochemical analyses were used to measure the expression of TM4SF1 in pancreatic cancer tissues and adjacent tissues. TM4SF1 was silenced using siRNA and shRNA to investigate the role of this protein in the proliferation and metastasis of pancreatic cancer cells. MTS and Transwell assays were used to examine the effect of TM4SF1 on pancreatic cancer cell lines. The expression and activity of MMP-2 and MMP-9 were determined by qRT-PCR, western blots and gelatin zymography. In vivo, orthotopic pancreatic tumor models were used to examine the formation of metastasis. RESULTS: qRT-PCR and immunohistochemical analyses showed that TM4SF1 was highly expressed in pancreatic cancer tissues compared with the adjacent tissues. In in vitro experiments the silencing of TM4SF1 reduced cell migration and invasion and down-regulated the expression and activity of MMP-2 and MMP-9. However, no significant difference in cell proliferation was detected after silencing TM4SF1. Additionally, knocking down TM4SF1 decreased the formation of lung and liver metastases in orthotopic pancreatic tumor models. CONCLUSION: Our results demonstrate that the expression of TM4SF1 is higher in pancreatic cancer tissues and pancreatic cancer cell lines than controls. Knockdown of TM4SF1 inhibited the migration and invasion of pancreatic cancer cells by regulating the expression and activity of MMP-2 and MMP-9, which suggests that TM4SF1 may play a significant role in metastasis in pancreatic cancer.


Subject(s)
Antigens, Surface/genetics , Cell Movement/genetics , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/genetics , Pancreatic Neoplasms/genetics , Animals , Antigens, Surface/metabolism , Cell Line, Tumor , Humans , Immunohistochemistry , Male , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Proteins/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/therapy , RNA Interference , RNAi Therapeutics , Reverse Transcriptase Polymerase Chain Reaction , Xenograft Model Antitumor Assays
5.
PLoS One ; 10(12): e0144969, 2015.
Article in English | MEDLINE | ID: mdl-26709920

ABSTRACT

BACKGROUND: TM4SF1 is overexpressed in pancreatic ductal adenocarcinoma (PDAC) and affects the development of this cancer. Also, multidrug resistance (MDR) is generally associated with tumor chemoresistance in pancreatic cancer. However, the correlation between TM4SF1 and MDR remains unknown. This research aims to investigate the effect of TM4SF1 on gemcitabine resistance in PDAC and explore the possible molecular mechanism between TM4SF1 and MDR. METHODS: The expression of TM4SF1 was evaluated in pancreatic cancer cell lines and human pancreatic duct epithelial (HPDE) cell lines by quantitative RT-PCR. TM4SF1 siRNA transfection was carried out using Hiperfect transfection reagent to knock down TM4SF1. The transcripts were analyzed by quantitative RT-PCR, RT-PCR and western blotting for further study. The cell proliferation and apoptosis were obtained to investigate the sensitivity to gemcitabine of pancreatic cancer cells after silencing TM4SF1 in vitro. We demonstrated that cell signaling of TM4SF1 mediated chemoresistance in cancer cells by assessing the expression of multidrug resistance (MDR) genes using quantitative RT-PCR. In vivo, we used orthotopic pancreatic tumor models to investigate the effect of proliferation after silencing TM4SF1 by a lentivirus-mediated shRNA in MIA PaCa-2 cell lines. RESULTS: The mRNA expression of TM4SF1 was higher in seven pancreatic cancer cell lines than in HPDE cell lines. In three gemcitabine-sensitive cell lines (L3.6pl, BxPC-3, SU86.86), the expression of TM4SF1 was lower than that in four gemcitabine-resistant cell lines (MIA PaCa-2, PANC-1, Hs766T, AsPC-1). We evaluated that TM4SF1 was a putative target for gemcitabine resistance in pancreatic cancer cells. Using AsPC-1, MIA PaCa-2 and PANC-1, we investigated that TM4SF1 silencing affected cell proliferation and increased the percentages of cell apoptosis mediated by treatment with gemcitabine compared with cells which were treated with negative control. This resistance was associated with the expression of multidrug resistance genes including ABCB1 and ABCC1. In vivo, silencing of TM4SF1 in MIA PaCa-2 cell lines increased the effectiveness of gemcitabine-based treatment in orthotopic pancreatic tumor models evaluated using noninvasive bioluminescent imaging. CONCLUSION: These findings suggest that TM4SF1 is a surface membrane antigen that is highly expressed in pancreatic cancer cells and increases the chemoresistance to gemcitabine. Thus, TM4SF1 may be a promising target to overcome the chemoresistance of pancreatic cancer.


Subject(s)
Antigens, Surface/genetics , Carcinoma, Pancreatic Ductal/genetics , Deoxycytidine/analogs & derivatives , Drug Resistance, Multiple/genetics , Drug Resistance, Neoplasm/genetics , Neoplasm Proteins/genetics , Pancreatic Neoplasms/drug therapy , ATP Binding Cassette Transporter, Subfamily B/biosynthesis , Animals , Apoptosis/drug effects , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Deoxycytidine/pharmacology , Humans , Male , Mice , Mice, Nude , Multidrug Resistance-Associated Proteins/biosynthesis , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , RNA Interference , RNA, Messenger/biosynthesis , RNA, Small Interfering/genetics , Signal Transduction/genetics , Gemcitabine
6.
Am J Physiol Gastrointest Liver Physiol ; 309(5): G283-91, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26159697

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is relatively rare but extremely lethal. Standard cytotoxic therapeutics provide little benefit. To date, newer targeted therapeutics have also not been highly successful. Often novel therapeutics that have appeared to perform well in preclinical models have failed in the clinic. Many factors contribute to these failures, but the one most often attributed is the shortcomings of the preclinical models. A plethora of animal models now exist for PDAC, including cell line xenografts, patient-derived xenografts, a wide variety of genetic mouse models, and syngeneic xenografts. These models have generated a tremendous amount of information useful for the understanding of PDAC. Yet none seems to well predict clinical outcomes of new treatments. This review will discuss how genetic instability and cellular heterogeneity make this disease so difficult to model accurately. We will also discuss the strengths and weaknesses of many of the popular models. Ultimately we will argue that there is no perfect model and that the best approach to understanding clinical performance is the use of multiple preclinical models with an understanding of their salient features.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Pancreatic Neoplasms/pathology , Xenograft Model Antitumor Assays/methods , Animals , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/physiopathology , Carcinoma, Pancreatic Ductal/therapy , Drug Evaluation, Preclinical/methods , Humans , Mice , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/physiopathology , Pancreatic Neoplasms/therapy , Species Specificity
7.
Mol Cancer Ther ; 14(4): 941-51, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25646014

ABSTRACT

Anterior gradient 2 (AGR2) promotes cancer growth, metastasis, and resistance to therapy via unknown mechanisms. We investigated the effects of extracellular AGR2 signaling through the orphan glycosylphosphatidylinositol-linked receptor C4.4A in pancreatic ductal adenocarcinoma (PDAC). Proliferation, migration, invasion, and apoptosis were measured using colorimetric, Boyden chamber, and FACS analyses. We developed blocking mAbs against AGR2 and C4.4A and tested their effects, along with siRNAs, on cancer cell functions and on orthotopic tumors in nude mice. Extracellular AGR2 stimulated proliferation, migration, invasion, and chemoresistance of PDAC cell lines. AGR2 interacted with C4.4A in cell lysates and mixtures of recombinant proteins. Knockdown of C4.4A reduced migration and resistance to gemcitabine. PDAC tissues, but not adjacent healthy pancreatic tissues, expressed high levels of AGR2 and C4.4A. AGR2 signaling through C4.4A required laminins 1 or 5 and integrin ß1. Administration of antibodies against AGR2 and C4.4A reduced growth and metastasis and caused regression of aggressive xenograft tumors, leading to increased survival of mice. These data support a model in which AGR2 binds and signals via C4.4A in an autocrine loop and promotes the growth of pancreas tumors in mice. Blocking mAbs against AGR2 and C4.4A may have therapeutic potential against PDAC.


Subject(s)
Antibodies, Blocking/pharmacology , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Cell Adhesion Molecules/metabolism , Pancreatic Neoplasms/metabolism , Proteins/metabolism , Signal Transduction/drug effects , Animals , Cell Adhesion Molecules/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Drug Resistance, Neoplasm , Extracellular Space/metabolism , Female , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Gene Expression , Humans , Integrin beta Chains/metabolism , Laminin/metabolism , Mice , Mucoproteins , Oncogene Proteins , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Protein Binding , Proteins/genetics , Xenograft Model Antitumor Assays , Kalinin , Pancreatic Neoplasms
8.
Mol Cancer Ther ; 13(11): 2583-94, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25193509

ABSTRACT

Pancreatic stellate cells (PSC) have been recognized as the principal cells responsible for the production of fibrosis in pancreatic ductal adenocarcinoma (PDAC). Recently, PSCs have been noted to share characteristics with cells of monocyte-macrophage lineage (MML cells). Thus, we tested whether PSCs could be targeted with the nitrogen-containing bisphosphonates (NBP; pamidronate or zoledronic acid), which are potent MML cell inhibitors. In addition, we tested NBPs treatment combination with nanoparticle albumin-bound paclitaxel (nab-paclitaxel) to enhance antitumor activity. In vitro, we observed that PSCs possess α-naphthyl butyrate esterase (ANBE) enzyme activity, a specific marker of MML cells. Moreover, NBPs inhibited PSCs proliferation, activation, release of macrophage chemoattractant protein-1 (MCP-1), and type I collagen expression. NBPs also induced PSCs apoptosis and cell-cycle arrest in the G1 phase. In vivo, NBPs inactivated PSCs; reduced fibrosis; inhibited tumor volume, tumor weight, peritoneal dissemination, angiogenesis, and cell proliferation; and increased apoptosis in an orthotopic murine model of PDAC. These in vivo antitumor effects were enhanced when NBPs were combined with nab-paclitaxel but not gemcitabine. Our study suggests that targeting PSCs and tumor cells with NBPs in combination with nab-paclitaxel may be a novel therapeutic approach to PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal/drug therapy , Diphosphonates/pharmacology , Nanoparticles/administration & dosage , Paclitaxel/administration & dosage , Pancreatic Neoplasms/drug therapy , Pancreatic Stellate Cells/drug effects , Adult , Albumins/administration & dosage , Animals , Apoptosis/drug effects , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Diphosphonates/administration & dosage , Drug Synergism , Female , Humans , Immunohistochemistry , Mice , Mice, Nude , Pamidronate , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/pathology , Xenograft Model Antitumor Assays
9.
Nucl Med Biol ; 41(10): 833-40, 2014.
Article in English | MEDLINE | ID: mdl-25189831

ABSTRACT

INTRODUCTION: Early detection of pancreatic cancer could save many thousands of lives. Non-invasive diagnostic imaging, including PET with [(18)F]FDG, has inadequate resolution for detection of small (2-3 mm) pancreatic tumours. We demonstrated the efficacy of PET imaging with an (18)F-labelled lactose derivative, [(18)F]FEDL, that targets HIP/PAP, a biomarker that is overexpressed in the peritumoural pancreas. We developed another analogue, 1-[(18)F]fluoroethyl lactose ([(18)F]FEL), which is simpler to synthesise, for the same application. We conducted a preliminary evaluation of the new probe and its efficacy in detecting orthotopic pancreatic carcinoma xenografts in mice. METHODS: Xenografts were developed in nude mice by injecting L3.6 pl/GL(+) pancreatic carcinoma cells into the pancreas of each mouse. Tumour growth was monitored by bioluminescence imaging (BLI); accuracy of BLI tumour size estimates was verified by MRI in two representative mice. When the tumour size reached approximately 2-3mm, the animals were injected with [(18)F]FEL (3.7 MBq) and underwent static PET/CT scans. Blood samples were collected at 2, 5, 10, 20 and 60 min after [(18)F]FEL injection to track blood clearance. Following imaging, animals were sacrificed and their organs and tumours/pancreatic tissue were collected and counted on a gamma counter. Pancreas, including tumour, was frozen, sliced and used for autoradiography and immunohistochemical analysis of HIP/PAP expression. RESULTS: Tumour growth was rapid, as observed by BLI and MRI. Blood clearance of [(18)F]FEL was bi-exponential, with half-lives of approximately 3.5 min and 40 min. Mean accumulation of [(18)F]FEL in the peritumoural pancreatic tissue was 1.29±0.295 %ID/g, and that in the normal pancreas of control animals was 0.090±0.101 %ID/g. [(18)F]FEL was cleared predominantly by the kidneys. Comparative analysis of autoradiographic images and immunostaining results demonstrated a correlation between [(18)F]FEL binding and HIP/PAP expression. CONCLUSION: [(18)F]FEL may be useful for non-invasive imaging of early-stage pancreatic tumours by PET. The results warrant further studies.


Subject(s)
Fluorine Radioisotopes , Lactose/analogs & derivatives , Pancreatic Neoplasms/diagnosis , Radiopharmaceuticals , Animals , Female , Fluorine Radioisotopes/pharmacokinetics , Humans , Immunoenzyme Techniques , Lactose/pharmacokinetics , Lactose/pharmacology , Luminescent Measurements , Magnetic Resonance Imaging , Mice , Mice, Nude , Pancreatic Neoplasms/metabolism , Pancreatitis-Associated Proteins , Positron-Emission Tomography , Proteins/metabolism , Radiopharmaceuticals/pharmacokinetics , Tissue Distribution , Tomography, X-Ray Computed , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
10.
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
11.
Cancer Res ; 74(18): 5301-10, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-24928781

ABSTRACT

The mechanisms that allow cancer cells to adapt to the typical tumor microenvironment of low oxygen and glucose and high lactate are not well understood. GPR81 is a lactate receptor recently identified in adipose and muscle cells that has not been investigated in cancer. In the current study, we examined GPR81 expression and function in cancer cells. We found that GPR81 was present in colon, breast, lung, hepatocellular, salivary gland, cervical, and pancreatic carcinoma cell lines. Examination of tumors resected from patients with pancreatic cancer indicated that 94% (148 of 158) expressed high levels of GPR81. Functionally, we observed that the reduction of GPR81 levels using shRNA-mediated silencing had little effect on pancreatic cancer cells cultured in high glucose, but led to the rapid death of cancer cells cultured in conditions of low glucose supplemented with lactate. We also observed that lactate addition to culture media induced the expression of genes involved in lactate metabolism, including monocarboxylase transporters in control, but not in GPR81-silenced cells. In vivo, GPR81 expression levels correlated with the rate of pancreatic cancer tumor growth and metastasis. Cells in which GPR81 was silenced showed a dramatic decrease in growth and metastasis. Implantation of cancer cells in vivo was also observed to lead to greatly elevated levels of GPR81. These data support that GPR81 is important for cancer cell regulation of lactate transport mechanisms. Furthermore, lactate transport is important for the survival of cancer cells in the tumor microenvironment. Cancer Res; 74(18); 5301-10. ©2014 AACR.


Subject(s)
Lactic Acid/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Receptors, G-Protein-Coupled/metabolism , Animals , Cell Death/physiology , Cell Line, Tumor , Cell Survival/physiology , HCT116 Cells , Hep G2 Cells , Heterografts , Humans , MCF-7 Cells , Mice, Nude , Receptors, G-Protein-Coupled/genetics , Transfection , Tumor Microenvironment
12.
Cancer Lett ; 350(1-2): 25-33, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24769074

ABSTRACT

Sulfated non-anticoagulant heparins (S-NACHs) might be preferred for potential clinical use in cancer patients without affecting hemostasis as compared to low molecular weight heparins (LMWHs). We investigated anti-tumor effects, anti-angiogenesis effects, and mechanisms of S-NACH in a mouse model of pancreatic cancer as compared to the LMWH tinzaparin. S-NACH or tinzaparin with or without gemcitabine were administered, and tumor luminescent signal intensity, tumor weight, and histopathology were assessed at the termination of the study. S-NACH and LMWH efficiently inhibited tumor growth and metastasis, without any observed bleeding events with S-NACH as compared to tinzaparin. S-NACH distinctly increased tumor necrosis and enhanced gemcitabine response in the mouse pancreatic cancer models. These data suggest the potential implication of S-NACH as a neoadjuvant in pancreatic cancer.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Fibrinolytic Agents/therapeutic use , Heparin, Low-Molecular-Weight/therapeutic use , Pancreatic Neoplasms/drug therapy , Animals , Cell Line, Tumor , Chick Embryo , Chorioallantoic Membrane/drug effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Enzyme Inhibitors/therapeutic use , Female , Humans , Mice , Mice, Nude , Neoplasm Metastasis , Neovascularization, Pathologic/drug therapy , Tinzaparin , Xenograft Model Antitumor Assays , Gemcitabine
13.
Sci Rep ; 4: 4410, 2014 Mar 19.
Article in English | MEDLINE | ID: mdl-24642931

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is the fourth leading cause of cancer death in the USA, accounting for ~40,000 deaths annually. The dismal prognosis for PDAC is largely due to its late diagnosis. Currently, the most sensitive diagnosis of PDAC requires invasive procedures, such as endoscopic ultrasonography, which has inherent risks and accuracy that is highly operator dependent. Here we took advantage of a general characteristic of solid tumors, the acidic microenvironment that is generated as a by-product of metabolism, to develop a novel approach of using pH (Low) Insertion Peptides (pHLIPs) for imaging of PDAC. We show that fluorescently labeled pHLIPs can localize and specifically detect PDAC in human xenografts as well as PDAC and PanIN lesions in genetically engineered mouse models. This novel approach may improve detection, differential diagnosis and staging of PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal/diagnosis , Diagnostic Imaging/methods , Pancreatic Neoplasms/diagnosis , Peptides , Peritoneal Neoplasms/diagnosis , Amino Acid Sequence , Animals , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/secondary , Cell Line, Tumor , Disease Models, Animal , Humans , Hydrogen-Ion Concentration , Mice , Mice, Nude , Molecular Sequence Data , Neoplasm Transplantation , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Peptides/chemical synthesis , Peptides/metabolism , Peritoneal Neoplasms/metabolism , Peritoneal Neoplasms/secondary , Prognosis , Tumor Microenvironment
14.
Mol Cancer Ther ; 12(5): 654-62, 2013 May.
Article in English | MEDLINE | ID: mdl-23303403

ABSTRACT

We have previously shown that the antiallergic drug cromolyn blocks S100P interaction with its receptor receptor for advanced glycation end product (RAGE) and improves gemcitabine effectiveness in pancreatic ductal adenocarcinoma (PDAC). However, the concentration required to achieve its effectiveness was high (100 µmol/L). In this study, we designed and synthesized analogs of cromolyn and analyzed their effectiveness compared with the parent molecule. An ELISA was used to confirm the binding of S100P with RAGE and to test the effectiveness of the different analogs. Analog 5-methyl cromolyn (C5OH) blocked S100P binding as well as the increases in NF-κB activity, cell growth, and apoptosis normally caused by S100P. In vivo C5OH systemic delivery reduced NF-κB activity to a greater extent than cromolyn and at 10 times lesser dose (50 mg vs. 5 mg). Treatment of mice-bearing syngeneic PDAC tumors showed that C5OH treatment reduced both tumor growth and metastasis. C5OH treatment of nude mice bearing orthotopic highly aggressive pancreatic Mpanc96 cells increased the overall animal survival. Therefore, the cromolyn analog, C5OH, was found to be more efficient and potent than cromolyn as a therapeutic for PDAC.


Subject(s)
Antineoplastic Agents/pharmacology , Calcium-Binding Proteins/antagonists & inhibitors , Carcinoma, Pancreatic Ductal/metabolism , Cromolyn Sodium/pharmacology , Neoplasm Proteins/antagonists & inhibitors , Pancreatic Neoplasms/metabolism , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Calcium-Binding Proteins/metabolism , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cromolyn Sodium/analogs & derivatives , Cromolyn Sodium/chemistry , Drug Design , Drug Evaluation, Preclinical , Humans , Male , Mice , Mice, Nude , NF-kappa B/metabolism , Neoplasm Proteins/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Protein Binding/drug effects , Receptor for Advanced Glycation End Products/metabolism , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
15.
Pancreas ; 42(3): 467-74, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23090667

ABSTRACT

OBJECTIVES: Pancreatic stellate cells are source of dense fibrotic stroma, a constant pathological feature of chronic pancreatitis and pancreatic adenocarcinoma. We observed correlation between levels of cyclooxygenase 2 (COX-2) and its product prostaglandin E2 (PGE2) and the extent of pancreatic fibrosis. The aims of this study were to delineate the effects of PGE2 on immortalized human pancreatic stellate cells (HPSCs) and to identify the receptor involved. METHODS: Immunohistochemistry, reverse transcription-polymerase chain reaction and quantitative reverse transcription-polymerase chain reaction were used to assess COX-2, extracellular matrix, and matrix metalloproteinase gene expression. Eicosanoid profile was determined by liquid chromatography-tandem mass spectrometry. Human pancreatic stellate cell proliferation was assessed by MTS assay, migration by Boyden chamber assay, and invasion using an invasion chamber. Transient silencing was obtained by small interfering RNA. RESULTS: Human pancreatic stellate cells express COX-2 and synthesize PGE2. Prostaglandin E2 stimulated HPSC proliferation, migration, and invasion and stimulated expression of both extracellular matrix and matrix metalloproteinase genes. Human pancreatic stellate cells expressed all 4 EP receptors. Only blocking the EP4 receptor resulted in abrogation of PGE2-mediated HPSC activation. Specificity of EP4 for the effects of PGE2 on stellate cells was confirmed using specific antagonists. CONCLUSIONS: Our data indicate that PGE2 regulates pancreatic stellate cell profibrotic activities via EP4 receptor, thus suggesting EP4 receptor as useful therapeutic target for pancreatic cancer to reduce desmoplasia.


Subject(s)
Cyclooxygenase 2/genetics , Dinoprostone/pharmacology , Pancreatic Stellate Cells/drug effects , Receptors, Prostaglandin E, EP4 Subtype/genetics , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Gene Expression/drug effects , Humans , Immunohistochemistry , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Naphthalenes/pharmacology , Pancreatic Stellate Cells/metabolism , Phenylbutyrates/pharmacology , RNA Interference , Receptors, Prostaglandin E, EP4 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
16.
Gastroenterology ; 143(6): 1510-1517.e1, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22960655

ABSTRACT

BACKGROUND & AIMS: New-onset diabetes in patients with pancreatic cancer is likely to be a paraneoplastic phenomenon caused by tumor-secreted products. We aimed to identify the diabetogenic secretory product(s) of pancreatic cancer. METHODS: Using microarray analysis, we identified adrenomedullin as a potential mediator of diabetes in patients with pancreatic cancer. Adrenomedullin was up-regulated in pancreatic cancer cell lines, in which supernatants reduced insulin signaling in beta cell lines. We performed quantitative reverse-transcriptase polymerase chain reaction and immunohistochemistry on human pancreatic cancer and healthy pancreatic tissues (controls) to determine expression of adrenomedullin messenger RNA and protein, respectively. We studied the effects of adrenomedullin on insulin secretion by beta cell lines and whole islets from mice and on glucose tolerance in pancreatic xenografts in mice. We measured plasma levels of adrenomedullin in patients with pancreatic cancer, patients with type 2 diabetes mellitus, and individuals with normal fasting glucose levels (controls). RESULTS: Levels of adrenomedullin messenger RNA and protein were increased in human pancreatic cancer samples compared with controls. Adrenomedullin and conditioned media from pancreatic cell lines inhibited glucose-stimulated insulin secretion from beta cell lines and islets isolated from mice; the effects of conditioned media from pancreatic cancer cells were reduced by small hairpin RNA-mediated knockdown of adrenomedullin. Conversely, overexpression of adrenomedullin in mice with pancreatic cancer led to glucose intolerance. Mean plasma levels of adrenomedullin (femtomoles per liter) were higher in patients with pancreatic cancer compared with patients with diabetes or controls. Levels of adrenomedullin were higher in patients with pancreatic cancer who developed diabetes compared those who did not. CONCLUSIONS: Adrenomedullin is up-regulated in patients with pancreatic cancer and causes insulin resistance in ß cells and mice.


Subject(s)
Adenocarcinoma/metabolism , Adrenomedullin/metabolism , Diabetes Mellitus, Type 2/metabolism , Insulin Resistance/physiology , Insulin-Secreting Cells/metabolism , Pancreatic Neoplasms/metabolism , Up-Regulation , Adenocarcinoma/pathology , Adrenomedullin/drug effects , Adrenomedullin/genetics , Aged , Animals , Cell Line, Tumor , Cells, Cultured , Diabetes Mellitus, Type 2/pathology , Female , Glucose/pharmacology , Humans , In Vitro Techniques , Insulin/metabolism , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/pathology , Male , Mice , Mice, Nude , Middle Aged , Models, Animal , Pancreas/metabolism , Pancreas/pathology , Pancreatic Neoplasms/pathology , RNA, Small Interfering/pharmacology , Rats , Transplantation, Heterologous
17.
Mol Cancer Res ; 10(9): 1147-57, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22859707

ABSTRACT

PURPOSE: The Hedgehog (Hh) pathway has emerged as an important pathway in multiple tumor types and is thought to be dependent on a paracrine signaling mechanism. The purpose of this study was to determine the role of pancreatic cancer-associated fibroblasts (human pancreatic stellate cells, HPSCs) in Hh signaling. In addition, we evaluated the efficacy of a novel Hh antagonist, AZD8542, on tumor progression with an emphasis on the role of the stroma compartment. EXPERIMENTAL DESIGN: Expression of Hh pathway members and activation of the Hh pathway were analyzed in both HPSCs and pancreatic cancer cells. We tested the effects of Smoothened (SMO) inhibition with AZD8542 on tumor growth in vivo using an orthotopic model of pancreatic cancer containing varying amounts of stroma. RESULTS: HPSCs expressed high levels of SMO receptor and low levels of Hh ligands, whereas cancer cells showed the converse expression pattern. HPSC proliferation was stimulated by Sonic Hedgehog with upregulation of downstream GLI1 mRNA. These effects were abrogated by AZD8542 treatment. In an orthotopic model of pancreatic cancer, AZD8542 inhibited tumor growth only when HPSCs were present, implicating a paracrine signaling mechanism dependent on stroma. Further evidence of paracrine signaling of the Hh pathway in prostate and colon cancer models is provided, demonstrating the broader applicability of our findings. CONCLUSION: Based on the use of our novel human-derived pancreatic cancer stellate cells, our results suggest that Hh-targeted therapies primarily affect the tumor-associated stroma, rather than the epithelial compartment.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Hedgehog Proteins/antagonists & inhibitors , Imidazoles/pharmacology , Pancreatic Neoplasms/drug therapy , Paracrine Communication/drug effects , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Antineoplastic Agents/chemistry , Benzamides/chemistry , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Disease Models, Animal , Female , Fibroblasts/metabolism , Gene Expression Regulation, Neoplastic , Humans , Imidazoles/chemistry , Liver Neoplasms/drug therapy , Male , Mice , Mice, Nude , Neoplasm Metastasis , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/physiology , Prostatic Neoplasms/drug therapy , Receptors, G-Protein-Coupled/antagonists & inhibitors , Smoothened Receptor , Up-Regulation
18.
PLoS One ; 7(7): e40420, 2012.
Article in English | MEDLINE | ID: mdl-22815748

ABSTRACT

At diagnosis, the majority of pancreatic cancer patients present with advanced disease when curative resection is no longer feasible and current therapeutic treatments are largely ineffective. An improved understanding of molecular targets for effective intervention of pancreatic cancer is thus urgent. The Met receptor tyrosine kinase is one candidate implicated in pancreatic cancer. Notably, Met is over expressed in up to 80% of invasive pancreatic cancers but not in normal ductal cells correlating with poor overall patient survival and increased recurrence rates following surgical resection. However the functional role of Met signaling in pancreatic cancer remains poorly understood. Here we used RNA interference to directly examine the pathobiological importance of increased Met signaling for pancreatic cancer. We show that Met knockdown in pancreatic tumor cells results in decreased cell survival, cell invasion, and migration on collagen I in vitro. Using an orthotopic model for pancreatic cancer, we provide in vivo evidence that Met knockdown reduced tumor burden correlating with decreased cell survival and tumor angiogenesis, with minimal effect on cell growth. Notably, we report that Met signaling regulates the secretion of the pro-angiogenic chemokine interleukin-8/CXCL8. Our data showing that the interleukin-8 receptors CXCR1 and CXCR2 are not expressed on pancreatic tumor cells, suggests a paracrine mechanism by which Met signaling regulates interleukin-8 secretion to remodel the tumor microenvironment, a novel finding that could have important clinical implications for improving the effectiveness of treatments for pancreatic cancer.


Subject(s)
Interleukin-8/metabolism , Neovascularization, Pathologic , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins c-met/metabolism , Signal Transduction , Animals , Blood Vessels/metabolism , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic , Gene Knockdown Techniques , Humans , Male , Mice , Mice, Nude , Pancreatic Neoplasms/metabolism , Paracrine Communication , Proto-Oncogene Proteins c-met/deficiency , Proto-Oncogene Proteins c-met/genetics , RNA Interference
19.
Clin Cancer Res ; 18(16): 4356-64, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22718861

ABSTRACT

PURPOSE: The receptor for advanced glycation end products (RAGE) contributes to multiple pathologies, including diabetes, arthritis, neurodegenerative diseases, and cancer. Despite the obvious need, no RAGE inhibitors are in common clinical use. Therefore, we developed a novel small RAGE antagonist peptide (RAP) that blocks activation by multiple ligands. EXPERIMENTAL DESIGN: RAGE and its ligands were visualized by immunohistochemical analysis of human pancreatic tissues, and siRNA was used to analyze their functions. Interactions between RAGE and S100P, S100A4, and HMGB-1 were measured by ELISA. Three S100P-derived small antagonistic peptides were designed, synthesized, and tested for inhibition of RAGE binding. The effects of the peptide blockers on NFκB-luciferase reporter activity was used to assess effects on RAGE-mediated signaling. The most effective peptide was tested on glioma and pancreatic ductal adenocarcinoma (PDAC) models. RESULTS: Immunohistochemical analysis confirmed the expression of RAGE and its ligands S100P, S100A4, and HMGB-1 in human PDAC. siRNA silencing of RAGE or its ligands reduced the growth and migration of PDAC cells in vitro. The most effective RAP inhibited the interaction of S100P, S100A4, and HMGB-1 with RAGE at micromolar concentrations. RAP also reduced the ability of the ligands to stimulate RAGE activation of NFκB in cancer cells in vitro and in vivo. Importantly, systemic in vivo administration of RAP reduced the growth and metastasis of pancreatic tumors and also inhibited glioma tumor growth. CONCLUSION: RAP shows promise as a tool for the investigation of RAGE function and as an in vivo treatment for RAGE-related disorders.


Subject(s)
Antineoplastic Agents/pharmacology , Calcium-Binding Proteins/chemistry , Neoplasm Proteins/chemistry , Neoplasms/metabolism , Peptide Fragments/pharmacology , Peptides/pharmacology , Receptors, Immunologic/antagonists & inhibitors , S100 Proteins/pharmacology , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , HMGB1 Protein/metabolism , Humans , Ligands , Male , Mice , Mice, Nude , NF-kappa B/metabolism , Neoplasm Metastasis , Neoplasms/genetics , Neoplasms/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Peptide Fragments/chemistry , Peptides/administration & dosage , Protein Binding/drug effects , Receptor for Advanced Glycation End Products , Receptors, Immunologic/metabolism , S100 Calcium-Binding Protein A4 , S100 Proteins/chemistry , S100 Proteins/metabolism
20.
Gut ; 61(9): 1315-22, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22068166

ABSTRACT

BACKGROUND AND AIMS: Pancreatic ductal adenocarcinoma (PDAC) is the fourth leading cause of cancer death in the USA. Surgical resection is the only effective treatment; however, only 20% of patients are candidates for surgery. The ability to detect early PDAC would increase the availability of surgery and improve patient survival. This study assessed the feasibility of using the enzymatic activity of cathepsin E (Cath E), a protease highly and specifically expressed in PDAC, as a novel biomarker for the detection of pancreas-bearing pancreatic intraepithelial neoplasia (PanIN) lesions and PDAC. METHODS: Pancreas from normal, chronic pancreatitis and PDAC patients was assessed for Cath E expression by quantitative real-time PCR and immunohistochemistry. Human PDAC xenografts and genetically engineered mouse models (GEMM) of PDAC were injected with a Cath E activity selective fluorescent probe and imaged using an optical imaging system. RESULTS: The specificity of Cath E expression in PDAC patients and GEMM of pancreatic cancer was confirmed by quantitative real-time PCR and immunohistochemistry. The novel probe for Cath E activity specifically detected PDAC in both human xenografts and GEMM in vivo. The Cath E sensitive probe was also able to detect pancreas with PanIN lesions in GEMM before tumour formation. CONCLUSIONS: The elevated Cath E expression in PanIN and pancreatic tumours allowed in-vivo detection of human PDAC xenografts and imaging of pancreas with PanIN and PDAC tumours in GEMM. Our results support the usefulness of Cath E activity as a potential molecular target for PDAC and early detection imaging.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma in Situ/diagnosis , Carcinoma, Pancreatic Ductal/diagnosis , Cathepsin E/metabolism , Diagnostic Imaging/methods , Pancreatic Neoplasms/diagnosis , Precancerous Conditions/diagnosis , Animals , Biomarkers, Tumor/genetics , Carcinoma in Situ/enzymology , Carcinoma, Pancreatic Ductal/enzymology , Cathepsin E/genetics , Cell Line, Tumor , DNA Primers/chemistry , Disease Models, Animal , Early Diagnosis , Feasibility Studies , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Mice , Oligonucleotide Probes/chemistry , Pancreatic Neoplasms/enzymology , Precancerous Conditions/enzymology , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...