Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Development ; 149(13)2022 07 01.
Article in English | MEDLINE | ID: mdl-35723257

ABSTRACT

Precise vascular patterning is crucial for normal growth and development. The ERG transcription factor drives Delta-like ligand 4 (DLL4)/Notch signalling and is thought to act as a pivotal regulator of endothelial cell (EC) dynamics and developmental angiogenesis. However, molecular regulation of ERG activity remains obscure. Using a series of EC-specific focal adhesion kinase (FAK)-knockout (KO) and point-mutant FAK-knock-in mice, we show that loss of ECFAK, its kinase activity or phosphorylation at FAK-Y397, but not FAK-Y861, reduces ERG and DLL4 expression levels together with concomitant aberrations in vascular patterning. Rapid immunoprecipitation mass spectrometry of endogenous proteins identified that endothelial nuclear-FAK interacts with the deubiquitinase USP9x and the ubiquitin ligase TRIM25. Further in silico analysis confirms that ERG interacts with USP9x and TRIM25. Moreover, ERG levels are reduced in FAKKO ECs via a ubiquitin-mediated post-translational modification programme involving USP9x and TRIM25. Re-expression of ERG in vivo and in vitro rescues the aberrant vessel-sprouting defects observed in the absence of ECFAK. Our findings identify ECFAK as a regulator of retinal vascular patterning by controlling ERG protein degradation via TRIM25/USP9x.


Subject(s)
Endothelial Cells , Transcription Factors , Animals , Endothelial Cells/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Mice , Neovascularization, Physiologic/genetics , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , Ubiquitins/metabolism
2.
Mol Ther ; 28(5): 1263-1275, 2020 05 06.
Article in English | MEDLINE | ID: mdl-32145202

ABSTRACT

Tumor-targeting oncolytic viruses such as vaccinia virus (VV) are attractive cancer therapeutic agents that act through multiple mechanisms to provoke both tumor lysis and anti-tumor immune responses. However, delivery of these agents remains restricted to intra-tumoral administration, which prevents effective targeting of inaccessible and disseminated tumor cells. In the present study we have identified transient pharmacological inhibition of the leukocyte-enriched phosphoinositide 3-kinase δ (PI3Kδ) as a novel mechanism to potentiate intravenous delivery of oncolytic VV to tumors. Pre-treatment of immunocompetent mice with the PI3Kδ-selective inhibitor IC87114 or the clinically approved idelalisib (CAL-101), prior to intravenous delivery of a tumor-tropic VV, dramatically improved viral delivery to tumors. This occurred via an inhibition of viral attachment to, but not internalization by, systemic macrophages through perturbation of signaling pathways involving RhoA/ROCK, AKT, and Rac. Pre-treatment using PI3Kδ-selective inhibitors prior to intravenous delivery of VV resulted in enhanced anti-tumor efficacy and significantly prolonged survival compared to delivery without PI3Kδ inhibition. These results indicate that effective intravenous delivery of oncolytic VV may be clinically achievable and could be useful in improving anti-tumor efficacy of oncolytic virotherapy.


Subject(s)
Adenine/analogs & derivatives , Administration, Intravenous/methods , Antineoplastic Agents/therapeutic use , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Immunotherapy/methods , Oncolytic Virotherapy/methods , Oncolytic Viruses/immunology , Purines/therapeutic use , Quinazolines/therapeutic use , Quinazolinones/therapeutic use , Vaccinia virus/immunology , Adenine/pharmacology , Adenine/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival , Combined Modality Therapy/methods , Female , Mice , Mice, Inbred BALB C , Purines/pharmacology , Quinazolines/pharmacology , Quinazolinones/pharmacology , Transplantation, Homologous , Treatment Outcome , Tumor Burden
4.
J Virol ; 88(4): 2291-300, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24335311

ABSTRACT

Oncolytic viruses based on adenovirus type 5 (Ad5) have been developed as a new class of therapeutic agents for cancers that are resistant to conventional therapies. Clinical experience shows that these agents are safe, but virotherapy alone has not achieved long-term cure in cancer patients. The vast majority of oncolytic adenoviruses used in clinical trials to date have deletion of the E3B genes. It has been demonstrated that the antitumor potency of the E3B-deleted mutant (dl309) is inferior to adenovirus with E3B genes intact. Tumors treated with dl309 show markedly greater macrophage infiltration than E3B-intact adenovirus. However, the functional mechanisms for this were not previously known. Here, we demonstrate that deletion of E3B genes increases production of chemokines by monocytes after adenovirus infection and increases monocyte migration. The E3B 14,700-Da protein (E3B-14.7K) inhibits STAT1 function by preventing its phosphorylation and nuclear translocation. The STAT1 inhibitor, fludarabine, rescues the effect of E3B-14.7K deletion by downregulating target chemokine expression in human and murine monocytes and results in an enhanced antitumor efficacy with dl309 in vivo. These findings have important implications for clinical use of E3B-deleted oncolytic adenovirus and other E3B-deleted adenovirus vector-based therapy.


Subject(s)
Adenoviridae/physiology , Adenovirus E3 Proteins/metabolism , Monocytes/metabolism , Oncolytic Viruses/physiology , STAT1 Transcription Factor/metabolism , Active Transport, Cell Nucleus/drug effects , Adenoviridae/metabolism , Adenovirus E3 Proteins/genetics , Analysis of Variance , Animals , Blotting, Western , Cell Line , DNA, Complementary/biosynthesis , Enzyme-Linked Immunosorbent Assay , Gene Deletion , Humans , Immunoprecipitation , Mice , Microscopy, Confocal , Oncolytic Viruses/metabolism , Phosphorylation/drug effects , Real-Time Polymerase Chain Reaction , STAT1 Transcription Factor/antagonists & inhibitors , Vidarabine/analogs & derivatives , Vidarabine/pharmacology
5.
J Virol ; 87(5): 2781-90, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23269798

ABSTRACT

Vaccinia virus (VV) is an enveloped DNA virus from the poxvirus family and has played a crucial role in the eradication of smallpox. It continues to be used in immunotherapy for the prevention of infectious diseases and treatment of cancer. However, the mechanisms of poxvirus entry, the host factors that affect viral virulence, and the reasons for its natural tropism for tumor cells are incompletely understood. By studying the effect of hypoxia on VV infection, we found that vascular endothelial growth factor A (VEGF-A) augments oncolytic VV cytotoxicity. VEGF derived from tumor cells acts to increase VV internalization, resulting in increased replication and cytotoxicity in an AKT-dependent manner in both tumor cells and normal respiratory epithelial cells. Overexpression of VEGF also enhances VV infection within tumor tissue in vivo after systemic delivery. These results highlight the importance of VEGF expression in VV infection and have potential implications for the design of new strategies to prevent poxvirus infection and the development of future generations of oncolytic VV in combination with conventional or biological therapies.


Subject(s)
Proto-Oncogene Proteins c-akt/metabolism , Vaccinia virus/pathogenicity , Vascular Endothelial Growth Factor A/metabolism , Virus Internalization , Animals , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/virology , Cell Line, Tumor , Epithelial Cells/virology , Genes, Reporter , Humans , Hypoxia , Mice , Mice, Inbred BALB C , Mice, Nude , RNA Interference , RNA, Small Interfering , Respiratory Mucosa/virology , Vaccinia/metabolism , Vaccinia/virology , Vaccinia virus/genetics , Vascular Endothelial Growth Factor A/genetics , Viral Tropism , Virus Replication/genetics
7.
Mol Ther ; 20(2): 306-16, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22086234

ABSTRACT

Oncolytic adenoviruses based on serotype 5 (Ad5) have several shortcomings, including the downregulation of its receptor in cancer cells, high prevalence of neutralizing antibodies and hepatotoxicity. Another adenoviral serotype, Ad11, could overcome these obstacles. Here, we show that human cancer cell lines express higher levels of the Ad11 receptor CD46, resulting in much better infectivity than Ad5. Surprisingly, only 36% (9/25) of the cell lines were more sensitive to Ad11- than to Ad5-mediated cytotoxicity. Investigations revealed that it was the transcription of Ad11 E1A, not CD46 expression or virus infectivity, which determined the cell's sensitivity to Ad11 killing. Ad11 E1A mRNA levels have an effect on viral DNA replication, structural protein synthesis and infectious particle production. To test the hypothesis that increased E1A transcription would lead to improved Ad11 replication in Ad5-sensitive (but Ad11-less sensitive) cells, two Ad11 mutants (Ad11-Ad5-P and Ad11-Ad5-EP) were constructed where either the E1A promoter or enhancer-promoter, respectively, was replaced by that of Ad5. Ad11-Ad5-EP demonstrated increased E1A mRNA levels and replication, together with enhanced oncolytic potency in vitro and in vivo. This effect was found in both the Ad5-sensitive and Ad11-sensitive cancer cells, broadening the range of tumors that could be effectively killed by Ad11-Ad5-EP.


Subject(s)
Adenoviridae/genetics , Adenovirus E1A Proteins/genetics , Enhancer Elements, Genetic , Genetic Vectors/genetics , Oncolytic Viruses/genetics , Promoter Regions, Genetic , Animals , Capsid Proteins/genetics , Capsid Proteins/metabolism , Cell Line, Tumor , Cytopathogenic Effect, Viral/genetics , Desmoglein 2/genetics , Genetic Vectors/administration & dosage , Humans , Membrane Cofactor Protein/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms/genetics , Neoplasms/mortality , Neoplasms/therapy , Oncolytic Virotherapy , Survival Analysis , Transcription, Genetic , Virus Replication/genetics , Xenograft Model Antitumor Assays
8.
J Immunother Cancer ; 9(1)2021 01.
Article in English | MEDLINE | ID: mdl-33500259

ABSTRACT

BACKGROUND: Pancreatic cancer remains one of the most lethal cancers and is refractory to immunotherapeutic interventions. Oncolytic viruses are a promising new treatment option, but current platforms demonstrate limited efficacy, especially for inaccessible and metastatic cancers that require systemically deliverable therapies. We recently described an oncolytic vaccinia virus (VV), VVLΔTKΔN1L, which has potent antitumor activity, and a regime to enhance intravenous delivery of VV by pharmacological inhibition of pharmacological inhibition of PI3 Kinase δ (PI3Kδ) to prevent virus uptake by macrophages. While these platforms improve the clinical prospects of VV, antitumor efficacy must be improved. METHODS: VVLΔTKΔN1L was modified to improve viral spread within and between tumors via viral B5R protein modification, which enhanced production of the extracellular enveloped virus form of VV. Antitumor immunity evoked by viral treatment was improved by arming the virus with interleukin-21, creating VVL-21. Efficacy, functional activity and synergy with α-programmed cell death protein 1 (α-PD1) were assessed after systemic delivery to murine and Syrian hamster models of pancreatic cancer. RESULTS: VVL-21 could reach tumors after systemic delivery and demonstrated antitumor efficacy in subcutaneous, orthotopic and disseminated models of pancreatic cancer. The incorporation of modified B5R improved intratumoural accumulation of VV. VVL-21 treatment increased the numbers of effector CD8+ T cells within the tumor, increased circulating natural killer cells and was able to polarize macrophages to an M1 phenotype in vivo and in vitro. Importantly, treatment with VVL-21 sensitized tumors to the immune checkpoint inhibitor α-PD1. CONCLUSIONS: Intravenously administered VVL-21 successfully remodeled the suppressive tumor-microenvironment to promote antitumor immune responses and improve long-term survival in animal models of pancreatic cancer. Importantly, treatment with VVL-21 sensitized tumors to the immune checkpoint inhibitor α-PD1. Combination of PI3Kδ inhibition, VVL-21 and α-PD1 creates an effective platform for treatment of pancreatic cancer.


Subject(s)
Immune Checkpoint Inhibitors/administration & dosage , Interleukin-12/genetics , Membrane Glycoproteins/metabolism , Pancreatic Neoplasms/therapy , Protein Kinase Inhibitors/administration & dosage , Vaccinia virus/physiology , Viral Envelope Proteins/metabolism , Administration, Intravenous , Animals , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Combined Modality Therapy , Humans , Immune Checkpoint Inhibitors/pharmacology , Interleukin-12/metabolism , Male , Membrane Glycoproteins/genetics , Mesocricetus , Mice , Oncolytic Virotherapy , Oncolytic Viruses/physiology , Pancreatic Neoplasms/immunology , Protein Kinase Inhibitors/pharmacology , Tumor Microenvironment , Viral Envelope Proteins/genetics , Xenograft Model Antitumor Assays
9.
Clin Cancer Res ; 18(24): 6679-89, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23091113

ABSTRACT

PURPOSE: The efficacy of oncolytic viruses depends on multiple actions including direct tumor lysis, modulation of tumor perfusion, and stimulation of tumor-directed immune responses. In this study, we investigated whether a sequential combination of immunologically distinct viruses might enhance antitumor efficacy through the induction of tumor-specific immunity and circumvention or mitigation of antiviral immune responses. EXPERIMENTAL DESIGN: The Syrian hamster as an immune-competent model that supports replication of both adenovirus and vaccinia virus was evaluated in vitro and in vivo. The antitumor efficacy of either virus alone or sequential combination of the two viruses was examined in pancreatic and kidney cancer models. The functional mechanism of the regimen developed here was investigated by histopathology, immunohistochemistry staining, CTL assay, and T-cell depletion. RESULTS: The Syrian hamster is a suitable model for assessment of oncolytic adenovirus and vaccinia virus. Three low doses of adenovirus followed by three low doses of vaccinia virus resulted in a superior antitumor efficacy to the reverse combination, or six doses of either virus alone, against pancreatic and kidney tumors in Syrian hamsters. A total of 62.5% of animals bearing either tumor type treated with the sequential combination became tumor-free, accompanied by the induction of effective tumor-specific immunity. This enhanced efficacy was ablated by CD3+ T-cell depletion but was not associated with humoral immunity against the viruses. CONCLUSION: These findings show that sequential treatment of tumors with oncolytic adenovirus and vaccinia virus is a promising approach for cancer therapy and that T-cell responses play a critical role.


Subject(s)
Kidney Neoplasms/therapy , Oncolytic Virotherapy , Pancreatic Neoplasms/therapy , Adenoviridae/genetics , Adenoviridae/immunology , Adenoviridae/physiology , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Apoptosis , Cells, Cultured , Cricetinae , Female , Humans , Immunocompetence , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Mesocricetus , Oncolytic Viruses/genetics , Oncolytic Viruses/immunology , Oncolytic Viruses/physiology , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/virology , Tumor Burden , Vaccinia virus/genetics , Vaccinia virus/immunology , Vaccinia virus/physiology , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication , Xenograft Model Antitumor Assays
10.
Hum Gene Ther ; 22(9): 1101-8, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21361787

ABSTRACT

Oncolytic viral therapy represents a promising strategy for the treatment of head and neck squamous cell carcinoma (HNSCC), with dl1520 (ONYX-015) the most widely used oncolytic adenovirus in clinical trials. This study aimed to determine the effectiveness of the Lister vaccine strain of vaccinia virus as well as a vaccinia virus armed with the endostatin-angiostatin fusion gene (VVhEA) as a novel therapy for HNSCC and to compare them with dl1520. The potency and replication of the Lister strain and VVhEA and the expression and function of the fusion protein were determined in human HNSCC cells in vitro and in vivo. Finally, the efficacy of VVhEA was compared with dl1520 in vivo in a human HNSCC model. The Lister vaccine strain of vaccinia virus was more effective than the adenovirus against all HNSCC cell lines tested in vitro. Although the potency of VVhEA was attenuated in vitro, the expression and function of the endostatin-angiostatin fusion protein was confirmed in HNSCC models both in vitro and in vivo. This novel vaccinia virus (VVhEA) demonstrated superior antitumor potency in vivo compared with both dl1520 and the control vaccinia virus. This study suggests that the Lister strain vaccinia virus armed with an endostatin-angiostatin fusion gene may be a potential therapeutic agent for HNSCC.


Subject(s)
Angiostatins/genetics , Carcinoma, Squamous Cell/therapy , Endostatins/genetics , Genetic Vectors/genetics , Head and Neck Neoplasms/therapy , Oncolytic Viruses/genetics , Vaccinia virus/genetics , Adenoviridae/genetics , Angiostatins/metabolism , Animals , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Cytopathogenic Effect, Viral , Endostatins/metabolism , Female , Gene Expression Regulation, Viral , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Humans , Kaplan-Meier Estimate , Mice , Mice, Inbred BALB C , Mice, Nude , Oncolytic Virotherapy , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Squamous Cell Carcinoma of Head and Neck , Viral Vaccines , Virus Replication , Xenograft Model Antitumor Assays
11.
J Clin Invest ; 119(6): 1604-15, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19411761

ABSTRACT

The changes in cancer cell surface molecules and intracellular signaling pathways during tumorigenesis make delivery of adenovirus-based cancer therapies inefficient. Here we have identified carcinoembryonic antigen- related cell adhesion molecule 6 (CEACAM6) as a cellular protein that restricts the ability of adenoviral vectors to infect cancer cells. We have demonstrated that CEACAM6 can antagonize the Src signaling pathway, downregulate cancer cell cytoskeleton proteins, and block adenovirus trafficking to the nucleus of human pancreatic cancer cells. Similar to CEACAM6 overexpression, treatment with a Src-selective inhibitor significantly reduced adenovirus replication in these cancer cells and normal human epithelial cells. In a mouse xenograft tumor model, siRNA-mediated knockdown of CEACAM6 also significantly enhanced the antitumor effect of an oncolytic adenovirus. We propose that CEACAM6-associated signaling pathways could be potential targets for the development of biomarkers to predict the response of patients to adenovirus-based therapies, as well as for the development of more potent adenovirus-based therapeutics.


Subject(s)
Adenoviridae/physiology , Antigens, CD/metabolism , Cell Adhesion Molecules/metabolism , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Adenoviridae/ultrastructure , Antigens, CD/genetics , Cell Adhesion Molecules/genetics , Cell Line , Colorectal Neoplasms/genetics , Cytoskeleton/metabolism , Down-Regulation , GPI-Linked Proteins , Gene Expression Regulation, Neoplastic , Humans , Microscopy, Electron, Transmission , Pancreatic Neoplasms/genetics , RNA, Small Interfering/genetics , Substrate Specificity , src-Family Kinases/metabolism
12.
Int J Cancer ; 120(11): 2344-52, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17311258

ABSTRACT

Pancreatic cancer is one of the most aggressive human tumors with a 5-year survival rate of only 3% and a striking resistance to chemotherapy and radiotherapy. The search for new therapeutic approaches includes strategies exploiting the deregulation of apoptotic pathways commonly found in cancer cells. The IAP proteins are inhibitors of apoptosis that have altered activity in numerous cancer types and are implicated in resistance to chemotherapy, and therefore are potentially interesting as therapeutic targets. We investigated alterations in the expression of IAPs and their inhibitors in pancreatic adenocarcinoma by using real-time PCR, in situ hybridization and immunohistochemistry. We found differential expression of various IAPs in this malignancy, and particularly we observed overexpression of cIAP-2, survivin, livin and XIAP. We also looked for correlations between the expression of IAPs and resistance to paclitaxel, doxorubicin, CDDP and 5-fluorouracil, and found that resistance to these drugs correlates most significantly with expression of cIAP-2. Using RNAi to downregulate these proteins we further confirmed that the levels of cIAP-2 and XIAP influence the response to the anti-cancer drugs, although only marginally for 5-FU. We conclude that anti-tumor strategies based on the inhibition of particular IAPs can be useful in targeting pancreatic adenocarcinoma.


Subject(s)
Antineoplastic Agents/therapeutic use , Inhibitor of Apoptosis Proteins/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/physiopathology , Area Under Curve , Base Sequence , DNA Primers , Drug Resistance, Neoplasm , Humans , Immunohistochemistry , In Situ Hybridization , Inhibitor of Apoptosis Proteins/physiology , Polymerase Chain Reaction , RNA Interference
13.
Am J Pathol ; 166(1): 81-92, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15632002

ABSTRACT

S100P is a member of the S100 family of calcium-binding proteins and there have been several recent reports of its overexpression in pancreatic ductal adenocarcinoma (PDAC). We have used Far Western screening and in vitro interaction assays to identify and confirm a novel target protein for S100P. We have named this protein S100PBPR, and shown that its interaction with S100P is dependent on Ca(2+) or Mg(2+). S100PBPR was found to localize to cell nuclei where S100P is also present, and the two proteins co-immunoprecipitate. By in situ hybridization, S100PBPR transcript was found in islet cells but not duct cells of the healthy pancreas. Both S100P and S100PBPR were detected by quantitative real-time polymerase chain reaction in pancreatic intraepithelial neoplasia (PanIN) and PDAC samples, and in situ hybridization revealed the presence of S100PBPR transcript in malignant PDAC cells. These data suggest that an interaction between S100P and S100PBPR may be involved in early pancreatic cancer. S100P was further investigated in PanIN lesions and immunohistochemical analysis showed its expression to correlate significantly with increasing grade of PanINs, being found as early as PanIN-1 with more prevalent expression in PanIN-2 and -3. These data suggest that S100P can be added to the genetic progression model for PDAC.


Subject(s)
Calcium-Binding Proteins/genetics , Carrier Proteins/genetics , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , Pancreatic Neoplasms/genetics , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Base Sequence , Calcium/metabolism , Carrier Proteins/metabolism , Cloning, Molecular , DNA Primers , Disease Progression , Humans , In Situ Hybridization , Magnesium/metabolism , Pancreatic Neoplasms/pathology , Polymerase Chain Reaction , RNA, Messenger/genetics , Transfection
14.
Gastroenterology ; 129(5): 1454-63, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16285947

ABSTRACT

BACKGROUND & AIMS: Markers to differentiate among pancreatic adenocarcinoma, chronic pancreatitis, and normal pancreas would be of significant clinical utility. This study was therefore designed to analyze the proteome of such specimens and identify new candidate proteins for differential diagnosis. METHODS: A PowerBlot analysis with more than 900 well-characterized antibodies was performed with tissue specimens from patients with chronic pancreatitis, pancreatic adenocarcinoma, and normal pancreas. Differential expression of selected proteins was confirmed on a larger scale by quantitative reverse transcription-polymerase chain reaction and immunohistochemistry using tissue arrays. RESULTS: A total of 30 and 102 proteins showed significant deregulation between normal pancreas when compared with chronic pancreatitis and pancreatic adenocarcinoma, respectively, and although a substantial proportion were found similarly dysregulated in both chronic pancreatitis and pancreatic adenocarcinoma, several proteins were identified as potential disease-specific markers. CONCLUSIONS: A large number of proteins are differentially expressed in chronic pancreatitis and pancreatic adenocarcinoma compared with normal pancreas. Among these, expression analysis of UHRF1, ATP7A, and aldehyde oxidase 1 in combination could potentially provide a useful additional diagnostic tool for fine-needle aspirated or cytological specimens obtained during endoscopic investigations.


Subject(s)
Adenocarcinoma/physiopathology , Pancreatic Neoplasms/physiopathology , Pancreatitis, Chronic/physiopathology , Protein Array Analysis , Proteomics , Adenocarcinoma/genetics , Biomarkers, Tumor , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Pancreatic Neoplasms/genetics , Pancreatitis, Chronic/genetics , Reverse Transcriptase Polymerase Chain Reaction
15.
J Pathol ; 201(1): 63-74, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12950018

ABSTRACT

In order to expand our understanding of the molecular changes underlying the complex pathology of pancreatic malignancy, global gene expression profiling of pancreatic adenocarcinoma compared with normal pancreatic tissue was performed. Human cDNA arrays comprising 9932 elements were interrogated with fluorescence-labelled normal and adenocarcinoma samples (nine tumours, three normal pancreata, and three cell lines). The data were analysed for differential gene expression, which was confirmed by serial analysis of gene expression (SAGE), digital differential display (DDD) analysis, and immunohistochemistry for selected cases. The array data were filtered to produce lists of a total of 75 genes significantly up-regulated or down-regulated in pancreatic adenocarcinoma. Two of those showing the highest differential were members of the S100 family of Ca-binding proteins, namely S100P and S100A6, and therefore the S100 genes were studied in more detail. By immunohistochemical analysis of custom-built, pancreas-specific tissue arrays and commercially available, normal/cancer tissue arrays that included a wide variety of different tumour types, differential expression of S100P protein was found to be almost exclusive to pancreatic cancer. S100P could therefore represent a useful biomarker for pancreatic adenocarcinomas.


Subject(s)
Adenocarcinoma/genetics , Biomarkers, Tumor/metabolism , Cell Cycle Proteins , Gene Expression Regulation, Neoplastic , Neoplasm Proteins , Pancreatic Neoplasms/genetics , S100 Proteins/genetics , Adenocarcinoma/metabolism , Calcium-Binding Proteins/genetics , DNA, Neoplasm/genetics , Gene Expression Profiling , Humans , Oligonucleotide Array Sequence Analysis , Pancreatic Neoplasms/metabolism , S100 Calcium Binding Protein A6 , S100 Proteins/metabolism , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL