Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 170
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Pancreatology ; 20(3): 394-408, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32173257

ABSTRACT

BACKGROUND: Death from pancreatic ductal adenocarcinoma (PDAC) is rising across the world and PDAC is predicted to be the second most common cause of cancer death in the USA by 2030. Development of effective biotherapies for PDAC are hampered by late presentation, a low number of differentially expressed molecular targets and a tumor-promoting microenvironment that forms both a physical, collagen-rich barrier and is also immunosuppressive. In 2017 Pancreatic Cancer UK awarded its first Grand Challenge Programme award to tackle this problem. The team plan to combine the use of novel CAR T cells with strategies to overcome the barriers presented by the tumor microenvironment. In advance of publication of those data this review seeks to highlight the key problems in effective CAR T cell therapy of PDAC and to describe pre-clinical and clinical progress in CAR T bio-therapeutics.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Gene Expression Profiling , Pancreatic Neoplasms/genetics , Humans , Immunotherapy, Adoptive , Male , Middle Aged , United Kingdom
2.
Gene Ther ; 22(6): 476-84, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25876464

ABSTRACT

Vaccinia virus (VV) has many attractive characteristics as a potential cancer therapeutic. There are several strains of VV. The nonvaccine strain Western Reserve VV with deletion of both the thymidine kinase and the viral growth factor genes (known as WRDD) has been reported as the most potent tumor-targeted oncolytic VV. Other strains, such as the European vaccine Lister strain, are largely untested. This study evaluated the antitumor potency and biodistribution of different VV strains using in vitro and in vivo models of cancer. Lister strain virus with thymidine kinase gene deletion (VVΔTK) demonstrated superior antitumor potency and cancer-selective replication in vitro and in vivo, compared with WRDD, especially in human cancer cell lines and immune-competent hosts. Further investigation of functional mechanisms revealed that Lister VVΔTK presented favorable viral biodistribution within the tumors, with lower levels of proinflammatory cytokines compared with WRDD, suggesting that Lister strain may induce a diminished host inflammatory response. This study indicates that the Lister strain VVΔTK may be a particularly promising VV strain for the development of the next generation of tumor-targeted oncolytic therapeutics.


Subject(s)
Gene Deletion , Oncolytic Viruses/physiology , Thymidine Kinase/genetics , Vaccinia virus/genetics , Animals , Cell Line , Cell Line, Tumor , Chlorocebus aethiops , Disease Models, Animal , Genetic Therapy , Humans , Kidney/cytology , Mice , Mutation , Neoplasms/therapy , Oncolytic Viruses/genetics , Smallpox Vaccine/genetics , Smallpox Vaccine/therapeutic use , Virus Replication
3.
Gene Ther ; 18(12): 1157-65, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21975464

ABSTRACT

Pancreatic adenocarcinomas are aggressive and frequently develop resistance to all current therapies. Replication-selective adenoviruses can overcome resistance to chemotherapeutics through their sensitizing effects on drug-induced cell killing. We previously found that adenovirus deleted in the anti-apoptotic E1B19K gene enhanced gemcitabine-induced apoptotis. Here we demonstrate that our engineered double-deleted AdΔΔ mutant (deleted in the pRb-binding E1ACR2 region and E1B19K) selectively replicates and enhances cell killing in combination with DNA-damaging cytotoxic drugs in pancreatic cancer cells. Combinations of AdΔΔ with gemcitabine, irinotecan or cisplatin resulted in two- to fourfold decreases in EC(50) (half maximal effective concentration) values and was more efficent than similar combinations with wild-type virus, the dl1520 (ONYX-015) and dl922-947 mutants. AdΔΔ replication was impaired in normal bronchial human epithelial cells and did not sensitize the cells to drugs. Gemcitabine-insensitive AsPC-1, BxPC-3 and PANC-1 cells were efficiently killed by irinotecan in combination with AdΔΔ. Suboptimal doses of AdΔΔ and gemcitabine significantly prolonged time to tumor progression in two human pancreatic tumor xenograft in vivo models, PT45 and SUIT-2. We conclude that AdΔΔ has low toxicity to normal cells while potently sensitizing pancreatic cancer cells to DNA-damaging drugs, and holds promise as an improved therapeutic strategy for pancreatic cancer.


Subject(s)
Adenocarcinoma/therapy , Adenoviridae/genetics , Antineoplastic Agents/administration & dosage , Oncolytic Viruses/genetics , Pancreatic Neoplasms/therapy , Animals , Apoptosis , Cell Line, Tumor , Combined Modality Therapy , Drug Resistance, Neoplasm , Genetic Vectors , Mice , Mice, Inbred C57BL , Mice, Nude , Viral Vaccines , Virus Replication , Virus Shedding , Xenograft Model Antitumor Assays
5.
Gene Ther ; 17(2): 281-7, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19890355

ABSTRACT

Hypoxia contributes to the aggressive and treatment-resistant phenotype of pancreatic ductal adenocarcinoma. Oncolytic vaccinia virus has potential as an anti-tumour agent, but the ability to lyse hypoxic tumour cells is vital for clinical efficacy. We hypothesized that unique aspects of the poxvirus life cycle would protect it from attenuation in hypoxic conditions. We characterized and compared the viral protein production, viral replication, cytotoxicity and transgene expression of Lister strain vaccinia virus in a panel of pancreatic cancer cell lines after exposure to normoxic or hypoxic conditions. Viral protein production was not affected by hypoxia, and high viral titres were produced in both normoxic and hypoxic conditions. Interestingly, there was a 3.5-fold (P<0.001) and 20-fold (P<0.0001) increase in viral cytotoxicity for CFPac1 and MiaPaca2 cell lines, respectively, in hypoxic conditions. Cytotoxicity was equivalent in the remaining cell lines. Levels of transgene expression (luciferase reporter gene) from the vaccinia viral vector were comparable, regardless of the ambient oxygen concentration. The present study suggests that the vaccinia virus is a promising vector for targeting pancreatic cancer and potentially other hypoxic tumour types.


Subject(s)
Cell Hypoxia , Gene Transfer Techniques , Genetic Vectors , Oncolytic Virotherapy/methods , Vaccinia virus/genetics , Cell Line, Tumor , Humans , Oncolytic Viruses/genetics , Transgenes , Viral Proteins/genetics , Virus Replication
6.
Gene Ther ; 16(10): 1223-33, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19587709

ABSTRACT

Survival after pancreatic cancer remains poor despite incremental advances in surgical and adjuvant therapy, and new strategies for treatment are needed. Oncolytic virotherapy is an attractive approach for cancer treatment. In this study, we have evaluated the effectiveness of the Lister vaccine strain of vaccinia virus armed with the endostatin-angiostatin fusion gene (VVhEA) as a novel therapeutic approach for pancreatic cancer. The Lister vaccine strain of vaccinia virus was effective against all human pancreatic carcinoma cells tested in vitro, especially those insensitive to oncolytic adenovirus. The virus displayed inherently high selectivity for cancer cells, sparing normal cells both in vitro and in vivo, with effective infection of tumors after both intravenous (i.v.) and intratumoral (i.t.) administrations. The expression of the endostatin-angiostatin fusion protein was confirmed in a pancreatic cancer model both in vitro and in vivo, with evidence of inhibition of angiogenesis. This novel vaccinia virus showed significant antitumor potency in vivo against the Suit-2 model by i.t. administration. This study suggests that the novel Lister strain of vaccinia virus armed with the endostatin-angiostatin fusion gene is a potential therapeutic agent for pancreatic cancer.


Subject(s)
Genetic Therapy/methods , Oncolytic Virotherapy/methods , Pancreatic Neoplasms/therapy , Vaccinia virus/genetics , Angiostatins/genetics , Animals , Artificial Gene Fusion , Combined Modality Therapy , Endostatins/genetics , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Pancreatic Neoplasms/pathology , Recombinant Fusion Proteins , Tumor Cells, Cultured , Vaccinia virus/physiology , Virus Replication , Xenograft Model Antitumor Assays
7.
Cell Death Differ ; 15(11): 1752-9, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18617895

ABSTRACT

Yes-associated protein (YAP) has been shown to positively regulate p53 family members and to be negatively regulated by the AKT proto-oncogene product in promoting apoptosis. On the basis of this function and its location at 11q22.2, a site of frequent loss of heterozygosity (LOH) in breast cancer, we investigated whether YAP is a tumor suppressor in breast. Examination of tumors by immunohistochemistry demonstrated significant loss of YAP protein. LOH analysis revealed that protein loss correlates with specific deletion of the YAP gene locus. Functionally, short hairpin RNA knockdown of YAP in breast cell lines suppressed anoikis, increased migration and invasiveness, inhibited the response to taxol and enhanced tumor growth in nude mice. This is the first report indicating YAP as a tumor suppressor, revealing its decreased expression in breast cancer as well as demonstrating the functional implications of YAP loss in several aspects of cancer signaling.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Breast/metabolism , Phosphoproteins/metabolism , Tumor Suppressor Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Anoikis/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cytoprotection/drug effects , Drug Resistance/drug effects , Epithelium/drug effects , Epithelium/metabolism , Gene Deletion , Gene Silencing/drug effects , Humans , Mice , Mice, Nude , Neoplasm Invasiveness , Paclitaxel/pharmacology , Phosphoproteins/genetics , Proto-Oncogene Mas , Transcription Factors , Xenograft Model Antitumor Assays , YAP-Signaling Proteins
8.
Pancreatology ; 9(1-2): 25-33, 2009.
Article in English | MEDLINE | ID: mdl-19077452

ABSTRACT

PURPOSE: Although ductal adenocarcinoma is the most common and well known pancreatic tumor type, other distinct epithelial neoplasms affecting the pancreas that show different symptoms, biological behaviors and outcomes are becoming more frequently recognized and documented. Pancreatic epithelial tumors may be separated into ductal and nonductal neoplasms. The former group includes pancreatic ductal adenocarcinoma, intraductal papillary-mucinous tumor, mucinous cystic tumor and serous cystic tumor. The latter group includes pancreatic endocrine tumor, pancreatic acinar cell carcinoma, pancreatoblastoma and solid-pseudopapillary tumor. The aim of this review is to summarize recently acquired knowledge regarding the molecular characterization of these uncommon pancreatic epithelial neoplasms. RECENT FINDINGS: Molecular studies of uncommon pancreatic epithelial tumors suggest that the different morphological entities are associated with distinct molecular profiles, highlighting the involvement of different molecular pathways leading to the development of each subtype of pancreatic neoplasm. CONCLUSION: The correct classification of rare pancreatic epithelial tumors and the identification of their characteristic molecular aspects is the fundamental starting point in identifying novel diagnostic molecular tools and new targets for innovative therapeutic strategies.


Subject(s)
Pancreatic Neoplasms/genetics , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adenocarcinoma, Mucinous/genetics , Adenocarcinoma, Mucinous/pathology , Carcinoma, Acinar Cell/genetics , Carcinoma, Acinar Cell/pathology , Cystadenoma, Mucinous/genetics , Cystadenoma, Mucinous/pathology , Humans , Pancreatic Neoplasms/pathology
9.
Oncogene ; 26(11): 1533-45, 2007 Mar 08.
Article in English | MEDLINE | ID: mdl-16983343

ABSTRACT

Sperm-associated antigen 1 (SPAG1) was recently identified in a rare form of infertility where anti-SPAG1 antibodies derived from the serum of an infertile woman were reported to cause sperm agglutination. Except for its expression and potential role in spermatogenesis, the function of SPAG1 is completely unknown. The unexpected finding of high levels of SPAG1 expression in pancreatic adenocarcinoma compared to normal pancreatic tissue in our previous cDNA array experiments prompted us to look in more detail at the expression and role of this gene in a panel of normal and malignant human tissues as well as in a larger series of pancreatic cancer specimens. We have generated an SPAG1-specific monoclonal antibody and showed high levels of SPAG1 protein in testis and in a large proportion of pancreatic ductal adenocarcinomas (PDAC). In the latter, SPAG1 expression was predominantly cytoplasmic and confined to malignant cells. Furthermore, the extent and intensity of SPAG1 expression was shown to be associated with stage and tumour nodal status, while analysis of precursor lesions, pancreatic intraepithelial neoplasias (PanINs), demonstrated its increased immunoreactivity with increasing PanIN grade, suggesting that SPAG1 is a novel marker of PDAC progression. Immunocytochemical analysis demonstrated colocalization of SPAG1 with microtubules, and their association was confirmed by co-immunoprecipitation; subsequent motility assays further substantiated a potential role of SPAG1 in cancer cell motility. Combined with the finding of its early expression in PDAC development, our data suggest that SPAG1 could contribute to the early spread and poor prognosis of pancreatic adenocarcinoma.


Subject(s)
Adenocarcinoma/metabolism , Antigens, Surface/metabolism , Cell Movement/physiology , GTP-Binding Proteins/metabolism , Pancreatic Neoplasms/metabolism , Adenocarcinoma/pathology , Amino Acid Sequence , Antigens, Surface/genetics , Cell Line, Tumor , DNA Primers , Down-Regulation , Enzyme-Linked Immunosorbent Assay , GTP-Binding Proteins/genetics , Humans , Pancreatic Neoplasms/pathology , RNA, Messenger/genetics , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction
10.
Oncogene ; 26(14): 2082-94, 2007 Mar 29.
Article in English | MEDLINE | ID: mdl-17043657

ABSTRACT

Pancreatic ductal adenocarcinoma is a devastating disease, characterized by a rapid progression and poor treatment response. Using gene expression profiling of pancreatic cancer tissues, we previously identified periostin as a potential diagnostic and therapeutic target. In this study, we report the overexpression of periostin in a larger set of pancreatic cancer tissues and show that although the periostin transcript is exclusively expressed in tumour cells, the protein product is only detected in the extracellular matrix adjacent to cancer cells. Using an enzyme-linked immunosorbent assay (ELISA) assay, we show significantly increased levels of periostin in the sera of pancreatic cancer patients compared to non-cancer controls. We demonstrate that periostin promotes the invasiveness of tumour cells by increasing the motility of cells without inducing expression of proteases, and enhances the survival of tumour cells exposed to hypoxic conditions. At the molecular level, we provide evidence that the alpha(6)beta(4) integrin complex acts as the cell receptor of periostin in pancreatic cancer cells and that interaction promotes phosphorylation of focal adhesion kinase (FAK) and protein kinase B (AKT) though activation of the PI3 kinase pathway, but not the RAS/MEK/ERK pathway. These findings suggest an important role of periostin in pancreatic cancer and provide a rationale to study periostin for diagnostic and therapeutic applications.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Cell Adhesion Molecules/physiology , Integrin beta4/metabolism , Pancreatic Neoplasms/pathology , Phosphatidylinositol 3-Kinases/metabolism , Apoptosis , Carcinoma, Pancreatic Ductal/chemistry , Carcinoma, Pancreatic Ductal/metabolism , Cell Adhesion Molecules/analysis , Cell Adhesion Molecules/genetics , Cell Line, Tumor , Cell Movement/genetics , Focal Adhesion Kinase 1/metabolism , Humans , Hypoxia/metabolism , Integrin alpha6beta4/metabolism , Neoplasm Invasiveness , Pancreatic Neoplasms/chemistry , Pancreatic Neoplasms/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/analysis , RNA, Messenger/metabolism , Transcription, Genetic
11.
Br Med Bull ; 87: 97-130, 2008.
Article in English | MEDLINE | ID: mdl-18753179

ABSTRACT

INTRODUCTION: Pancreatic cancer is a devastating malignancy and a leading cause of cancer mortality. Furthermore, early diagnosis represents a serious hurdle for clinicians, as symptoms are non-specific and usually manifest in advanced, treatment-resistant stages of the disease. SOURCES OF DATA: Here, we review the rationale and progress of targeted therapies currently under investigation. AREAS OF AGREEMENT: At present, chemoradiation regimes are administered palliatively, and produce only marginal survival benefits, underscoring a desperate need for more effective treatment modalities. AREAS OF CONTROVERSY: Questions have been raised as to whether erlotinib, the only targeted therapy to attain a statistically significant increase in median survival, is cost-effective. GROWING POINTS: The last decade of research has provided us with a wealth of information regarding the molecular nature of pancreatic cancer, leading to the identification of signalling pathways and their respective components which are critical for the maintenance of the malignant phenotype. AREAS TIMELY FOR DEVELOPING RESEARCH: These proteins thus represent ideal targets for novel molecular therapies which embody an urgently needed novel treatment strategy.


Subject(s)
Pancreatic Neoplasms/therapy , Antineoplastic Agents/administration & dosage , Clinical Trials as Topic , Humans , Pancreatic Neoplasms/genetics , Protein Kinase Inhibitors/administration & dosage , Signal Transduction/genetics
12.
Trends Genet ; 12(10): 400-5, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8909136

ABSTRACT

Genetic intervention for the therapy of human disease has long been a dream for scientists and clinicians alike, and the first steps towards reality have already been taken in clinical trials involving over 1000 patients around the world. The technology used in these initial experiments has limited potential for therapeutic effect and it is now appreciated that improvements in targeting gene delivery and gene expression are both required before real clinical benefit is achieved. The enormous advances made in the fields of molecular genetics and molecular biology of the last few years have set the scene for their translation into novel approaches to gene transfer and control, for gene therapy applications.


Subject(s)
Adenoviridae , Genetic Therapy/methods , Retroviridae , Animals , Genetic Vectors , Humans , Liposomes , Promoter Regions, Genetic , Transcription, Genetic , Viral Envelope Proteins
13.
Mol Cell Biol ; 10(10): 5365-77, 1990 Oct.
Article in English | MEDLINE | ID: mdl-1697930

ABSTRACT

To overcome the difficulty of assessing oncogene action in human epithelial cell types, such as thyroid, which have limited proliferative potential in culture, we have explored the use of temperature-sensitive (ts) mutants of simian virus 40 (SV40) early region to create conditionally immortalized epithelial cell lines. Normal primary cultures of human thyroid follicular cells were transfected with a plasmid containing the SV40 early region from mutant tsA58. Expanding epithelial colonies were observed after 2 to 3 months, all of which grew to greater than 200 population doublings without crisis. All showed tight temperature dependence for growth. After switch-up to the restrictive temperature (40.5 degrees C), no further increase in cell number was seen after 1 to 2 days. However, DNA synthesis declined much more slowly; the dissociation from cell division led to marked polyploidy. Viability was maintained for up to 2 weeks. Introduction of an inducible mutant ras gene into ts thyroid cells led, as expected, to morphological transformation at the permissive temperature when ras was induced. Interestingly, this was associated with a marked reduction in net growth rate. At the restrictive temperature, induction of mutant ras caused rapid cell death. These results demonstrate the utility of a ts SV40 mutant to permit the study of oncogene action in an otherwise nonproliferative target cell and reveal important differences in the interaction between ras and SV40 T in these epithelial cells compared with previously studied cell types.


Subject(s)
Antigens, Viral, Tumor/genetics , Cell Transformation, Neoplastic/genetics , Thyroid Gland/cytology , Animals , Blotting, Northern , Cell Adhesion , Cell Differentiation , Cell Division , Clone Cells , Epithelial Cells , Gene Expression , Humans , In Vitro Techniques , Iodine/metabolism , Karyotyping , Keratins/metabolism , Mice , Mice, Nude , Mutation , Neoplasms, Experimental/pathology , Oncogene Protein p21(ras)/genetics , Phenotype , RNA, Messenger/genetics , Temperature , Thyroglobulin/metabolism , Transfection
14.
Oncogene ; 36(22): 3094-3103, 2017 06 01.
Article in English | MEDLINE | ID: mdl-27941872

ABSTRACT

The mechanisms of initiation of pancreatic ductal adenocarcinoma (PDAC) are still largely unknown. In the present study, we analysed the role of anterior gradient-2 (AGR2) in the earliest stages of pancreatic neoplasia. Immunohistochemical analysis of chronic pancreatitis (CP) and peritumoral areas in PDAC tissues showed that AGR2 was present in tubular complexes (TC) and early pancreatic intraepithelial neoplasia (PanINs). Moreover, AGR2 was also found in discrete subpopulations of non-transformed cells neighbouring these pre-neoplastic lesions. In primary cells derived from human patient-derived xenograft (PDX) model, flow-cytometry revealed that AGR2 was overexpressed in pancreatic cancer stem cells (CSC) compared with non-stem cancer cells. In LSL-KrasG12D;Pdx1-Cre (KC) mouse model Agr2 induction preceded the formation of pre-neoplastic lesions and their development was largely inhibited by Agr2 deletion in engineered LSL-KrasG12D;Pdx1-Cre; Agr2-/- mice. In vitro, AGR2 expression was stimulated by tunicamycin-induced endoplasmic reticulum (ER) stress in both KRAS wild-type normal pancreas cells, as well as in KRAS mutated pancreatic cancer cells and was essential for ER homoeostasis. The unfolded protein response proteins GRP78, ATF6 and XBP1s were found expressed in CP and PDAC peritumoral tissues, but in contrast to AGR2, their expression was switched off during TC and PanIN formation. Real-time PCR and ELISA analyses showed that ER stress induced a pro-inflammatory phenotype in pancreatic normal, cancer and stellate cells. Moreover, AGR2 expression was inducible by paracrine transfer of ER stress and pro-inflammation between different pancreatic cell types. Our findings demonstrate that AGR2 induced in ER-stressed and inflammatory pre-neoplastic pancreas is a potential marker of cancer progenitor cells with an important functional role in PDAC initiation.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Endoplasmic Reticulum Stress/physiology , Mucoproteins/metabolism , Pancreatic Neoplasms/pathology , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Disease Models, Animal , Endoplasmic Reticulum Chaperone BiP , Humans , Mice , Mucoproteins/biosynthesis , Mucoproteins/deficiency , Mucoproteins/genetics , Oncogene Proteins , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism
15.
Endocr Relat Cancer ; 13(2): 541-58, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16728581

ABSTRACT

The intrinsic nature of tumour behaviour (stable vs progressive) and the presence of liver metastases are key factors in determining the outcome of patients with a pancreatic endocrine tumour (PET). Previous expression profile analyses of PETs were limited to non-homogeneous groups or to primary lesions only. The aim of this study was to investigate the gene expression profiles of a more uniform series of sporadic, non-functioning (NF) PETs with progressive disease and, for the first time, their liver metastases, on the Affymetrix human genome U133A and B GeneChip set. Thirteen NF PET samples (eight primaries and five liver metastases) from ten patients with progressive, metastatic disease, three cell lines (BON, QGP and CM) and four purified islet samples were analysed. The same samples were employed for confirmation of candidate gene expression by means of quantitative RT-PCR, while a further 37 PET and 15 carcinoid samples were analysed by immunohistochemistry. Analysis of genes differentially expressed between islets and primaries and metastases revealed 667 up- and 223 down-regulated genes, most of which have not previously been observed in PETs, and whose gene ontology molecular function has been detailed. Overexpression of bridging integrator 1 (BIN1) and protein Z dependent protease inhibitor (SERPINA10) which may represent useful biomarkers, and of lymphocyte specific protein tyrosine kinase (LCK) and bone marrow stromal cell antigen (BST2) which could be used as therapeutic targets, has been validated. When primary tumours were compared with metastatic lesions, no significantly differentially expressed genes were found, in accord with cluster analysis which revealed a striking similarity between primary and metastatic lesions, with the cell lines clustering separately. We have provided a comprehensive list of differentially expressed genes in a uniform set of aggressive NF PETs. A number of dysregulated genes deserve further in-depth study as potentially promising candidates for new diagnostic and treatment strategies. The analysis of liver metastases revealed a previously unknown high level of similarity with the primary lesions.


Subject(s)
Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Gene Expression Profiling , Genes, Neoplasm/genetics , Liver Neoplasms/genetics , Pancreatic Neoplasms/genetics , Adult , Aged , Cluster Analysis , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/secondary , Liver Neoplasms/therapy , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/therapy , RNA, Messenger/analysis
16.
Cancer Res ; 55(5): 1129-38, 1995 Mar 01.
Article in English | MEDLINE | ID: mdl-7866999

ABSTRACT

The c-MET oncogene encodes the receptor for hepatocyte growth factor (HGF) scatter factor, a multifunctional cytokine able to mediate morphogenesis as well as invasive growth of epithelial cells. The c-MET-encoded receptor is detectable only at low levels in the normal human exocrine pancreas, but it is up-regulated in the majority of pancreatic ductal adenocarcinomas. The c-MET-encoded HGF receptor is also overexpressed in a proportion of the panel of 31 human pancreatic cancer cell lines examined, which have a range of different growth properties and degrees of differentiation. In most cases the HGF receptor found in the malignant cells has features of the normal receptor. When added to pancreatic cancer cell lines, HGF triggers receptor phosphorylation and stimulates cells to move and proliferate. In overexpressing cell lines, the Met/HGF receptor is phosphorylated in the absence of endogenously produced or exogenously added ligand. These data suggest that the Met/HGF receptor may be involved in the growth and behavior of pancreatic cancer and may contribute to the ductal phenotype of these tumors.


Subject(s)
Pancreatic Neoplasms/genetics , Receptor Protein-Tyrosine Kinases/genetics , Base Sequence , Cell Division/drug effects , Cell Movement/drug effects , Gene Expression , Glycosylation , Hepatocyte Growth Factor/pharmacology , Humans , In Situ Hybridization , Molecular Sequence Data , Pancreas/physiology , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/ultrastructure , Proto-Oncogene Proteins c-met , Receptor Protein-Tyrosine Kinases/metabolism , Stimulation, Chemical , Tumor Cells, Cultured/drug effects
17.
Cancer Res ; 59(12): 2802-5, 1999 Jun 15.
Article in English | MEDLINE | ID: mdl-10383135

ABSTRACT

Dendritic cells (DCs) are potent antigen-presenting cells that are capable of priming systemic antitumor immune responses in animal tumor models. However, many of the model tumor systems tested need definition of the specific tumor antigens involved. To use DCs in situations that are more relevant to the majority of human cancers, where the antigens are unknown, we have tested the adoptive transfer of immature DCs in mouse colorectal and melanoma models of varying immunogenicity but with undefined antigens. When DCs admixed with a syngeneic primary tumor inoculum were seeded s.c., the growth of the primary tumor was unchanged; however, if the primary tumor was then surgically excised and the animal was rechallenged with the same tumor, significant protection (75%) was generated when DCs were present in the original primary inoculum of a moderately immunogenic colorectal model (CMT93tk). This effect was not observed when a nonimmunogenic melanoma (B16) was tested in an identical protocol. Next, DCs were injected directly into 6-9-mm established tumors; again, protection (55%) was achieved against a secondary tumor challenge following excision of the primary, but only in the CMT93tk model of moderate immunogenicity. To increase the clinical relevance of this approach still further, we tested irradiated allogeneic K1735 melanoma cells mixed with syngeneic DCs as a vaccine against subsequent challenge with the poorly immunogenic syngeneic melanoma B16. The allogeneic vaccine alone was ineffective, but when admixed with DCs, a significant number of animals rejected a subsequent B16 challenge, suggesting that DCs are able to prime an immune response against melanoma antigens shared between K1735 and B16. The generation of systemic antitumor immunity by adoptive transfer of DCs has significant clinical potential because it is technically straightforward and does not require the definition of specific tumor antigens.


Subject(s)
Dendritic Cells/transplantation , Immunotherapy, Adoptive , Melanoma, Experimental/therapy , Animals , Cancer Vaccines/immunology , Cells, Cultured , Disease Models, Animal , Immunity , Immunotherapy , Isoantigens , Melanoma, Experimental/immunology , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Transplantation, Autologous
18.
Cancer Res ; 58(19): 4461-7, 1998 Oct 01.
Article in English | MEDLINE | ID: mdl-9766679

ABSTRACT

Overexpression of tissue factor (TF) is characteristically observed in advanced pancreatic cancer and has been associated with invasion and metastasis. Functional responses of TF activation are here investigated using as a model system the human pancreatic cancer cell lines SW979 (which overexpresses TF) and MIAPaCa2 (which does not express detectable levels). After stimulation of these cell lines with factor VIIa (FVIIa), the only known TF ligand, expression of urokinase receptor (uPAR) gene was up-regulated in SW979 cells in a dose-dependent manner but not in MIAPaCa2 cells. Interestingly, urokinase (uPA) and its specific inhibitor PAI-1 were not up-regulated. Exposure to functionally inactivated FVIIa did not show any effect on uPAR expression on SW979 cells despite binding to TF with higher efficiency. The neutralizing anti-TF antibody 5G9 blocked the FVIIa-induced up-regulation of uPAR completely, whereas hirudin failed to block this up-regulation. Treatment of SW979 cells with Factor Xa did not up-regulate the expression of uPAR gene, whereas treatment with FVII induced the same level of enhanced uPAR gene expression as that with FVIIa. In the matrigel invasion assay, enhanced invasion of SW979 cell line induced by FVIIa was completely inhibited by anti-TF antibody and alpha2-antiplasmin. Moreover, the endogenous levels of uPAR gene expression were significantly correlated with the level of TF gene expression in 19 human cancer cell lines (P < 0.05). These data suggest that up-regulation of uPAR expression by tumor cells leading to tumor invasion is induced through the TF-FVIIa pathway rather than TF-initiated thrombin generation. This is the first report that TF may be one of the key receptors that can up-regulate expression of the plasminogen activator receptor in human cancer cells to enhance tumor invasion and metastasis.


Subject(s)
Factor VIIa/physiology , Gene Expression Regulation, Neoplastic , Pancreatic Neoplasms/genetics , Receptors, Cell Surface/genetics , Thromboplastin/metabolism , Breast Neoplasms , Cell Movement , Factor VIIa/pharmacology , Factor Xa/pharmacology , Factor Xa/physiology , Female , Flow Cytometry , Humans , Pancreatic Neoplasms/metabolism , Receptors, Cell Surface/biosynthesis , Receptors, Urokinase Plasminogen Activator , Thromboplastin/genetics , Transcription, Genetic , Tumor Cells, Cultured , Up-Regulation , Urokinase-Type Plasminogen Activator/metabolism
19.
Cancer Res ; 48(16): 4459-63, 1988 Aug 15.
Article in English | MEDLINE | ID: mdl-3293774

ABSTRACT

Human thyroid epithelial (follicular) cells give rise to two malignant tumors--"follicular" carcinomas, which metastasize almost exclusively via the bloodstream, and "papillary" carcinomas, which metastasize predominantly via lymphatics (Williams, E. D. In: W. Duncan (ed.), Recent Results in Cancer Research: Thyroid Cancer, pp. 47-55. Berlin: Springer-Verlag, 1980). We have investigated whether this contrast in biological behavior might be associated with different patterns of oncogene activation. DNA transfection analysis of five follicular and ten papillary cancers indeed showed a statistically significant difference in the pattern of genes responsible, activated ras oncogenes being found in 80% of follicular tumors but only 20% of papillary tumors. In addition, in follicular cancers we have found activation of all three ras oncogenes (H-ras, K-ras, and N-ras), the first time that this has been demonstrated in a primary human tumor type (as opposed to cell lines). We suggest therefore that ras activation may be an important determinant of metastatic capability in these epithelial cancers.


Subject(s)
Genes, ras , Thyroid Neoplasms/genetics , Animals , Humans , Mice , Mutation , Neoplasm Metastasis , Transfection
20.
Oncogene ; 6(3): 471-3, 1991 Mar.
Article in English | MEDLINE | ID: mdl-2011403

ABSTRACT

Activated ras oncogenes in experimentally induced rodent tumours have been demonstrated to show specific patterns of oncogene activation which depend on the inducing agent, with H-ras activation in nitrosomethylurea (NMU) induced tumours, and K-ras activation in tumours induced by ionising radiation. We report a study of 12 radiation-associated human thyroid tumours, using polymerase chain reaction (PCR) amplification of paraffin-embedded material and allele-specific hybridisation with mutant-specific probes for the 3 ras oncogenes. Compared to 68 'spontaneous' human thyroid tumours, the radiation-associated cases show the same overall prevalence of ras mutation. However there is a significantly higher rate of K-ras mutation in radiation-associated follicular carcinomas than in 'spontaneous' follicular carcinomas (60% compared to 6%, P less than 0.05), suggesting that radiation may preferentially activate K-ras in human as well as rodent tumours.


Subject(s)
Adenocarcinoma/genetics , Carcinoma, Papillary/genetics , Carcinoma/genetics , Genes, ras/genetics , Neoplasms, Radiation-Induced/genetics , Thyroid Neoplasms/genetics , Adenocarcinoma/etiology , Adolescent , Adult , Alleles , Carcinoma/etiology , Carcinoma, Papillary/etiology , Child , Female , Humans , Male , Middle Aged , Mutation , Nucleic Acid Hybridization , Polymerase Chain Reaction , Thyroid Neoplasms/etiology
SELECTION OF CITATIONS
SEARCH DETAIL