Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Clin Cancer Res ; 19(5): 1139-46, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23340293

ABSTRACT

PURPOSE: High-throughput chemosensitivity testing of low-passage cancer cell lines can be used to prioritize agents for personalized chemotherapy. However, generating cell lines from primary cancers is difficult because contaminating stromal cells overgrow the malignant cells. EXPERIMENTAL DESIGN: We produced a series of hypoxanthine phosphoribosyl transferase (hprt)-null immunodeficient mice. During growth of human cancers in these mice, hprt-null murine stromal cells replace their human counterparts. RESULTS: Pancreatic and ovarian cancers explanted from these mice were grown in selection media to produce pure human cancer cell lines. We screened one cell line with a 3,131-drug panel and identified 77 U.S. Food and Drug Administration (FDA)-approved drugs with activity, and two novel drugs to which the cell line was uniquely sensitive. Xenografts of this carcinoma were selectively responsive to both drugs. CONCLUSION: Chemotherapy can be personalized using patient-specific cell lines derived in biochemically selectable mice.


Subject(s)
Carcinoma, Pancreatic Ductal/drug therapy , Digitoxin/pharmacology , Nogalamycin/pharmacology , Ovarian Neoplasms/drug therapy , Pancreatic Neoplasms/drug therapy , Precision Medicine , Animals , Antibiotics, Antineoplastic/pharmacology , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cardiotonic Agents/pharmacology , Female , Humans , Hypoxanthine Phosphoribosyltransferase/genetics , Interleukin Receptor Common gamma Subunit , Male , Mice , Mice, Nude , Mice, SCID , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Stromal Cells/drug effects , Stromal Cells/metabolism , Stromal Cells/pathology , Tumor Cells, Cultured
2.
Pancreatology ; 12(4): 372-9, 2012.
Article in English | MEDLINE | ID: mdl-22898640

ABSTRACT

CONTEXT: Although pancreatic cancer is a common, highly lethal malignancy, the molecular events that enable precursor lesions to become invasive carcinoma remain unclear. We previously reported that the high-mobility group A1 (HMGA1) protein is overexpressed in >90% of primary pancreatic cancers, with absent or low levels in early precursor lesions. METHODS: Here, we investigate the role of HMGA1 in reprogramming pancreatic epithelium into invasive cancer cells. We assessed oncogenic properties induced by HMGA1 in non-transformed pancreatic epithelial cells expressing activated K-RAS. We also explored the HMGA1-cyclooxygenase (COX-2) pathway in human pancreatic cancer cells and the therapeutic effects of COX-2 inhibitors in xenograft tumorigenesis. RESULTS: HMGA1 cooperates with activated K-RAS to induce migration, invasion, and anchorage-independent cell growth in a cell line derived from normal human pancreatic epithelium. Moreover, HMGA1 and COX-2 expression are positively correlated in pancreatic cancer cell lines (r(2) = 0.93; p < 0.001). HMGA1 binds directly to the COX-2 promoter at an AT-rich region in vivo in three pancreatic cancer cell lines. In addition, HMGA1 induces COX-2 expression in pancreatic epithelial cells, while knock-down of HMGA1 results in repression of COX-2 in pancreatic cancer cells. Strikingly, we also discovered that Sulindac (a COX-1/COX-2 inhibitor) or Celecoxib (a more specific COX-2 inhibitor) block xenograft tumorigenesis from pancreatic cancer cells expressing high levels of HMGA1. CONCLUSIONS: Our studies identify for the first time an important role for the HMGA1-COX-2 pathway in pancreatic cancer and suggest that targeting this pathway could be effective to treat, or even prevent, pancreatic cancer.


Subject(s)
Adenocarcinoma/genetics , Cyclooxygenase 2/genetics , HMGA1a Protein/genetics , Pancreatic Neoplasms/genetics , Adenocarcinoma/drug therapy , Adenocarcinoma/prevention & control , Animals , Celecoxib , Cell Division/genetics , Cell Line, Tumor , Cell Movement/genetics , Cyclooxygenase 2/physiology , Cyclooxygenase Inhibitors/administration & dosage , Gene Expression , HMGA1a Protein/physiology , Humans , Mice , Mice, Nude , Neoplasm Invasiveness/genetics , Neoplasm Metastasis/genetics , Neoplasm Transplantation , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/prevention & control , Pyrazoles/administration & dosage , Sulfonamides/administration & dosage , Sulindac/administration & dosage , Transplantation, Heterologous , ras Proteins/physiology
3.
Oncotarget ; 3(6): 640-50, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22791660

ABSTRACT

Acquired chemotherapy resistance is a major contributor to treatment failure in oncology. For example, the efficacy of the common anticancer agent doxorubicin (DOX) is limited by the emergence of multidrug resistance (MDR) phenotype in cancer cells. While dose escalation of DOX can circumvent such resistance to a degree, this is precluded by the appearance of cardiotoxicity, a particularly debilitating condition in children. In vitro studies have established the ability of the natural phytochemical curcumin to overcome MDR; however, its widespread clinical application is restricted by poor solubility and low bioavailability. Building upon our recently developed polymer nanoparticle of curcumin (NanoCurc or NC) that significantly enhances the systemic bioavailability of curcumin, we synthesized a doxorubicin-curcumin composite nanoparticle formulation called NanoDoxCurc (NDC) for overcoming DOX resistance. Compared to DOX alone, NDC inhibited the MDR phenotype and caused striking growth inhibition both in vitro and in vivo in several models of DOX-resistant cancers (multiple myeloma, acute leukemia, prostate and ovarian cancers, respectively). Notably, NDC-treated mice also demonstrated complete absence of cardiac toxicity, as assessed by echocardiography, or any bone marrow suppression, even at cumulative dosages where free DOX and pegylated liposomal DOX (Doxil®) resulted in demonstrable attenuation of cardiac function and hematological toxicities. This improvement in safety profile was achieved through a reduction of DOX-induced intracellular oxidative stress, as indicated by total glutathione levels and glutathione peroxidase activity in cardiac tissue. A composite DOX-curcumin nanoparticle that overcomes both MDR-based DOX chemoresistance and DOX-induced cardiotoxicity holds promise for providing lasting and safe anticancer therapy.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Cardiomyopathies/prevention & control , Doxorubicin/analogs & derivatives , Doxorubicin/administration & dosage , Nanoparticles/administration & dosage , Animals , Antibiotics, Antineoplastic/adverse effects , Antibiotics, Antineoplastic/chemistry , Cardiomyopathies/chemically induced , Cardiomyopathies/drug therapy , Cell Line, Tumor , Curcumin/administration & dosage , Curcumin/analogs & derivatives , Curcumin/chemistry , Doxorubicin/adverse effects , Doxorubicin/chemistry , Drug Resistance, Neoplasm , Glutathione/metabolism , Humans , Male , Mice , Mice, Nude , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Nanoparticles/chemistry , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Random Allocation , Xenograft Model Antitumor Assays
4.
Proc Natl Acad Sci U S A ; 109(5): E252-9, 2012 Jan 31.
Article in English | MEDLINE | ID: mdl-22233809

ABSTRACT

Defining the molecular genetic alterations underlying pancreatic cancer may provide unique therapeutic insight for this deadly disease. Toward this goal, we report here an integrative DNA microarray and sequencing-based analysis of pancreatic cancer genomes. Notable among the alterations newly identified, genomic deletions, mutations, and rearrangements recurrently targeted genes encoding components of the SWItch/Sucrose NonFermentable (SWI/SNF) chromatin remodeling complex, including all three putative DNA binding subunits (ARID1A, ARID1B, and PBRM1) and both enzymatic subunits (SMARCA2 and SMARCA4). Whereas alterations of each individual SWI/SNF subunit occurred at modest-frequency, as mutational "hills" in the genomic landscape, together they affected at least one-third of all pancreatic cancers, defining SWI/SNF as a major mutational "mountain." Consistent with a tumor-suppressive role, re-expression of SMARCA4 in SMARCA4-deficient pancreatic cancer cell lines reduced cell growth and promoted senescence, whereas its overexpression in a SWI/SNF-intact line had no such effect. In addition, expression profiling analyses revealed that SWI/SNF likely antagonizes Polycomb repressive complex 2, implicating this as one possible mechanism of tumor suppression. Our findings reveal SWI/SNF to be a central tumor suppressive complex in pancreatic cancer.


Subject(s)
Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone/physiology , Genes, Tumor Suppressor , Pancreatic Neoplasms/metabolism , Transcription Factors/physiology , Chromosomal Proteins, Non-Histone/genetics , Gene Expression Profiling , Humans , Mutation , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Transcription Factors/genetics , Transcriptome
5.
Mol Cancer Ther ; 11(1): 165-73, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22027695

ABSTRACT

Aberrant activation of the hedgehog (Hh) signaling pathway is one of the most prevalent abnormalities in human cancer. Tumors with cell autonomous Hh activation (e.g., medulloblastomas) can acquire secondary mutations at the Smoothened (Smo) antagonist binding pocket, which render them refractory to conventional Hh inhibitors. A class of Hh pathway inhibitors (HPI) has been identified that block signaling downstream of Smo; one of these compounds, HPI-1, is a potent antagonist of the Hh transcription factor Gli1 and functions independent of upstream components in the pathway. Systemic administration of HPI-1 is challenging due to its minimal aqueous solubility and poor bioavailability. We engineered a polymeric nanoparticle from [poly(lactic-co-glycolic acid); (PLGA)] conjugated with polyethylene glycol (PEG), encapsulating HPI-1 (NanoHHI). NanoHHI particles have an average diameter of approximately 60 nm, forms uniform aqueous suspension, and improved systemic bioavailability compared with the parent compound. In contrast to the prototype targeted Smo antagonist, HhAntag (Genentech), NanoHHI markedly inhibits the growth of allografts derived from Ptch(-/+); Trp53(-/-) mouse medulloblastomas that harbor a Smo(D477G) binding site mutation (P < 0.001), which is accompanied by significant downregulation of mGli1 as well as bona fide Hh target genes (Akna, Cltb, and Olig2). Notably, NanoHHI combined with gemcitabine also significantly impedes the growth of orthotopic Pa03C pancreatic cancer xenografts that have a ligand-dependent, paracrine mechanism of Hh activation when compared with gemcitabine alone. No demonstrable hematologic or biochemical abnormalities were observed with NanoHHI administration. NanoHHI should be amenable to clinical translation in settings where tumors acquire mutational resistance to current Smo antagonists.


Subject(s)
Hedgehog Proteins/antagonists & inhibitors , Nanoparticles/chemistry , Pancreatic Neoplasms/metabolism , Receptors, G-Protein-Coupled/antagonists & inhibitors , Animals , Cell Line, Tumor , Cell Proliferation , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Hedgehog Proteins/metabolism , Humans , Male , Medulloblastoma/metabolism , Medulloblastoma/pathology , Mice , Mice, Nude , Patched Receptors , Patched-1 Receptor , Receptors, Cell Surface/genetics , Receptors, G-Protein-Coupled/genetics , Signal Transduction , Smoothened Receptor , Transcription Factors/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics , Xenograft Model Antitumor Assays , Zinc Finger Protein GLI1 , Gemcitabine
6.
Neoplasia ; 13(10): 923-30, 2011 Oct.
Article in English | MEDLINE | ID: mdl-22028618

ABSTRACT

Ligand-dependent activation of the Hedgehog (Hh) signaling pathway has been implicated in both tumor initiation and metastasis of pancreatic ductal adenocarcinoma (PDAC). Prior studies in genetically engineered mouse models (GEMMs) have assessed the role of Hh signaling by cell autonomous expression of a constitutively active Gli2 within epithelial cells. On the contrary, aberrant pathway reactivation in the human exocrine pancreas occurs principally as a consequence of Sonic Hh ligand (Shh) overexpression from epithelial cells. To recapitulate the cognate pathophysiology of Hh signaling observed in the human pancreas, we examined GEMM where Hh ligand is conditionally overexpressed within the mature exocrine pancreas using a tamoxifen-inducible Elastase-Cre promoter (Ela-CreERT2;LSL-mShh). We also facilitated potential cell autonomous epithelial responsiveness to secreted Hh ligand by generating compound transgenic mice with concomitant expression of the Hh receptor Smoothened (Ela-CreERT2;LSL-mShh;LSL-mSmo). Of interest, none of these mice developed intraductal precursor lesions or PDAC during the follow-up period of up to 12 months after tamoxifen induction. Instead, all animals demonstrated marked expansion of stromal cells, consistent with the previously described epithelial-to-stromal paracrine Hh signaling. Hh responsiveness was mirrored by the expression of primary cilia within the expanded mesenchymal compartment and the absence within mature acinar cells. In the absence of cooperating mutations, Hh ligand overexpression in the mature exocrine pancreas is insufficient to induce neoplasia, even when epithelial cells coexpress the Smo receptor. This autochthonous model serves as a platform for studying epithelial stromal interactions in pancreatic carcinogenesis.


Subject(s)
Hedgehog Proteins/metabolism , Pancreas/metabolism , Stromal Cells/metabolism , Acinar Cells/metabolism , Acinar Cells/pathology , Animals , Cell Proliferation , Estrogen Antagonists/pharmacology , Female , Genetic Engineering/methods , Hedgehog Proteins/genetics , Humans , Immunoblotting , Immunohistochemistry , Insulin/metabolism , Keratins/metabolism , Male , Metaplasia , Mice , Mice, Transgenic , Microscopy, Confocal , Pancreas/drug effects , Pancreas/pathology , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Smoothened Receptor , Stromal Cells/pathology , Tamoxifen/pharmacology , Tubulin/metabolism
7.
PLoS One ; 6(8): e23924, 2011.
Article in English | MEDLINE | ID: mdl-21887346

ABSTRACT

Pancreatic cancer is a deadly disease, and new therapeutic targets are urgently needed. We previously identified DNA amplification at 7q21-q22 in pancreatic cancer cell lines. Now, by high-resolution genomic profiling of human pancreatic cancer cell lines and human tumors (engrafted in immunodeficient mice to enrich the cancer epithelial fraction), we define a 325 Kb minimal amplicon spanning SMURF1, an E3 ubiquitin ligase and known negative regulator of transforming growth factor ß (TGFß) growth inhibitory signaling. SMURF1 amplification was confirmed in primary human pancreatic cancers by fluorescence in situ hybridization (FISH), where 4 of 95 cases (4.2%) exhibited amplification. By RNA interference (RNAi), knockdown of SMURF1 in a human pancreatic cancer line with focal amplification (AsPC-1) did not alter cell growth, but led to reduced cell invasion and anchorage-independent growth. Interestingly, this effect was not mediated through altered TGFß signaling, assayed by transcriptional reporter. Finally, overexpression of SMURF1 (but not a catalytic mutant) led to loss of contact inhibition in NIH-3T3 mouse embryo fibroblast cells. Together, these findings identify SMURF1 as an amplified oncogene driving multiple tumorigenic phenotypes in pancreatic cancer, and provide a new druggable target for molecularly directed therapy.


Subject(s)
Gene Amplification/physiology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Ubiquitin-Protein Ligases/genetics , Animals , Cell Communication , Cell Line , Cell Line, Tumor , Humans , In Situ Hybridization, Fluorescence , Mice , Neoplasm Invasiveness , Oncogenes
8.
Clin Cancer Res ; 17(17): 5793-800, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21742805

ABSTRACT

PURPOSE: The goal of this study was to evaluate prospectively the engraftment rate, factors influencing engraftment, and predictability of clinical outcome of low-passage xenografts from patients with resectable pancreatic ductal adenocarcinoma (PDA) and to establish a bank of PDA xenografts. EXPERIMENTAL DESIGN: Patients with resectable PDA scheduled for resection at the Johns Hopkins Hospital were eligible. Representative pieces of tumor were implanted in nude mice. The status of the SMAD4 gene and content of tumor-generating cells were determined by immunohistochemistry. Gene expression was carried out by using a U133 Plus 2.0 array. Patients were followed for progression and survival. RESULTS: A total of 94 patients with PDA were resected, 69 tumors implanted in nude mice, and 42 (61%) engrafted. Engrafted carcinomas were more often SMAD4 mutant, and had a metastatic gene expression signature and worse prognosis. Tumors from patients resistant to gemcitabine were enriched in stroma-related gene pathways. Tumors sensitive to gemcitabine were enriched in cell cycle and pyrimidine gene pathways. The time to progression for patients who received treatment with gemcitabine for metastatic disease (n = 7) was double in patients with xenografts sensitive to gemcitabine. CONCLUSION: A successful xenograft was generated in 61% of patients attempted, generating a pool of 42 PDA xenografts with significant biological information and annotated clinical data. Patients with PDA and SMAD4 inactivation have a better engraftment rate. Engraftment is a poor prognosis factor, and engrafted tumors have a metastatic gene expression signature. Tumors from gemcitabine-resistant patients were enriched in stromal pathways.


Subject(s)
Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Deoxycytidine/analogs & derivatives , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Smad4 Protein/genetics , Adult , Aged , Aged, 80 and over , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/surgery , Deoxycytidine/therapeutic use , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Profiling , Humans , Male , Mice , Mice, Nude , Middle Aged , Neoplasm Transplantation , Oligonucleotide Array Sequence Analysis , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/surgery , Prognosis , Transplantation, Heterologous , Treatment Outcome , Gemcitabine
9.
Cancer Biol Ther ; 12(7): 598-609, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21768779

ABSTRACT

Pancreatic cancer is one of the most lethal of human malignancies, and potent therapeutic options are lacking. Inhibition of cell cycle progression through pharmacological blockade of cyclin-dependent kinases (CDK) has been suggested as a potential treatment option for human cancers with deregulated cell cycle control. Dinaciclib (SCH727965) is a novel small molecule multi-CDK inhibitor with low nanomolar potency against CDK1, CDK2, CDK5 and CDK9 that has shown favorable toxicity and efficacy in preliminary mouse experiments, and has been well tolerated in Phase I clinical trials. In the current study, the therapeutic efficacy of SCH727965 on human pancreatic cancer cells was tested using in vitro and in vivo model systems. Treatment with SCH727965 significantly reduced in vitro cell growth, motility and colony formation in soft agar of MIAPaCa-2 and Pa20C cells. These phenotypic changes were accompanied by marked reduction of phosphorylation of Retinoblastoma (Rb) and reduced activation of RalA. Single agent therapy with SCH727965 (40 mg/kg i.p. twice weekly) for 4 weeks significantly reduced subcutaneous tumor growth in 10/10 (100%) of tested low-passage human pancreatic cancer xenografts. Treatment of low passage pancreatic cancer xenografts with a combination of SCH727965 and gemcitabine was significantly more effective than either agent alone. Gene Set Enrichment Analysis identified overrepresentation of the Notch and Transforming Growth Factor-ß (TGF-ß) signaling pathways in the xenografts least responsive to SCH727965 treatment. Treatment with the cyclin-dependent kinase inhibitor SCH727965 alone or in combination is a highly promising novel experimental therapeutic strategy against pancreatic cancer.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cyclin-Dependent Kinases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Pancreatic Neoplasms/drug therapy , Pyridinium Compounds/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cyclic N-Oxides , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Drug Resistance, Neoplasm/genetics , Humans , Indolizines , Male , Mice , Mice, Nude , Pancreatic Neoplasms/embryology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Phosphorylation/drug effects , Pyridinium Compounds/administration & dosage , Receptors, Notch/genetics , Receptors, Notch/metabolism , Retinoblastoma Protein/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Xenograft Model Antitumor Assays , ral GTP-Binding Proteins/metabolism , Gemcitabine
10.
Cancer Biol Ther ; 12(7): 617-28, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21750403

ABSTRACT

PURPOSE: To investigate the frequency of xenotropic murine leukemia virus (MLV) presence in human cell lines established from mouse xenografts and to search for the evidence of horizontal viral spread to other cell lines. RESULTS: Six of 23 (26%) mouse DNA free xenograft cultures were strongly positive for MLV and their sequences had greater than 99% homology to known MLV strains. Four of five available supernatant fluids from these viral positive cultures were strongly positive for RT activity. Three of these supernatant fluids were studied to confirm the infectivity of the released virions for other human culture cells. Of the 78 non-xenograft derived cell lines maintained in the xenograft culture-containing facilities, 13 (17%) were positive for MLV, including XMRV, a virus strain first identified in human tissues. By contrast, all 50 cultures maintained in a xenograft culture-free facility were negative for viral sequences. METHODOLOGY: We examined xenograft tumor cell lines from seven independent laboratories and 128 non-xenografted tumor cell lines. Cell line DNA was examined for mouse DNA contamination, and by 3 Taqman qPCR assays targeting the gag, env or pol regions of MLV. Sequencing was used for viral strain identification. Supernatant fluids were tested for reverse transcriptase (RT) activity. CONCLUSIONS: Human cultures derived after mouse xenografting frequently contain and release highly infectious xenotropic MLV viruses. Laboratories working with xenograft-derived human cultures should be aware of the risk of contamination with potentially biohazardous human-tropic mouse viruses and their horizontal spread to other cultures.


Subject(s)
Cell Line, Tumor/virology , Leukemia Virus, Murine/isolation & purification , Animals , DNA/analysis , Disease Transmission, Infectious , Genes, env , Genes, gag , Humans , Leukemia Virus, Murine/genetics , Leukemia Virus, Murine/pathogenicity , Mice/genetics , Retroviridae Infections/transmission , Transplantation, Heterologous , Virion
11.
Mol Cancer Ther ; 10(8): 1470-80, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21622730

ABSTRACT

Mis-expression of microRNAs (miRNA) is widespread in human cancers, including in pancreatic cancer. Aberrations of miRNA include overexpression of oncogenic miRs (Onco-miRs) or downregulation of so-called tumor suppressor TSG-miRs. Restitution of TSG-miRs in cancer cells through systemic delivery is a promising avenue for pancreatic cancer therapy. We have synthesized a lipid-based nanoparticle for systemic delivery of miRNA expression vectors to cancer cells (nanovector). The plasmid DNA-complexed nanovector is approximately 100 nm in diameter and shows no apparent histopathologic or biochemical evidence of toxicity upon intravenous injection. Two miRNA candidates known to be downregulated in the majority of pancreatic cancers were selected for nanovector delivery: miR-34a, which is a component of the p53 transcriptional network and regulates cancer stem cell survival, and the miR-143/145 cluster, which together repress the expression of KRAS2 and its downstream effector Ras-responsive element binding protein-1 (RREB1). Systemic intravenous delivery with either miR-34a or miR-143/145 nanovectors inhibited the growth of MiaPaCa-2 subcutaneous xenografts (P < 0.01 for miR-34a; P < 0.05 for miR-143/145); the effects were even more pronounced in the orthotopic (intrapancreatic) setting (P < 0.0005 for either nanovector) when compared with vehicle or mock nanovector delivering an empty plasmid. Tumor growth inhibition was accompanied by increased apoptosis and decreased proliferation. The miRNA restitution was confirmed in treated xenografts by significant upregulation of the corresponding miRNA and significant decreases in specific miRNA targets (SIRT1, CD44 and aldehyde dehydrogenase for miR34a, and KRAS2 and RREB1 for miR-143/145). The nanovector is a platform with potential broad applicability in systemic miRNA delivery to cancer cells.


Subject(s)
MicroRNAs/administration & dosage , Pancreatic Neoplasms/therapy , Animals , Cell Line, Tumor , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Gene Transfer Techniques , Humans , Male , Mice , Mice, Nude , MicroRNAs/genetics , MicroRNAs/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Xenograft Model Antitumor Assays
12.
Cancer Biol Ther ; 11(11): 959-68, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21455033

ABSTRACT

An inactivating germline mutation in BRCA2 is the most common known genetic basis for familial pancreatic cancer (FPC), accounting for 5-10% of inherited cases. A genetically engineered mouse model of pancreatic ductal adenocarcinoma (PDAC) arising on the backdrop of Brca2 deficiency is likely to elucidate valuable diagnostic and therapeutic insights for FPC. Both Brca2 alleles were conditionally deleted during development within the pancreatic epithelium by generating Pdx1-Cre; Brca2(f/f) (CB) mice; in addition, triple transgenic Pdx1-Cre; Brca2(f/f); LSL-Trp53(R172H) (CBP) mice were generated, in order to determine the impact of p53 deregulation on Brca2-deficient carcinogenesis. Both CB and CBP mice developed non-invasive ductal precursor lesions (murine pancreatic intraepithelial neoplasia or mPanIN), although these were observed at an earlier time point (5 versus 8 months) and with higher prevalence in CBP mice. A minority of CB mice (15%) developed invasive and metastatic PDAC at a latency of 15 months or greater; in contrast, CBP mice of comparable age uniformly developed PDAC with variable histological features. Mortality in the absence of neoplasia in CB and CBP mice was associated with profound loss of pancreatic parenchyma, consistent with progressive elimination of Brca2-deficient cells. Widespread DNA damage, as evidenced by overexpression of the phosphorylated histone H(2)AX(Ser139), was observed in the non-neoplastic exocrine pancreas, as well as in the mPanIN and PDAC lesions of Brca2-deficient mice, independent of p53 status. Loss of Brca2 function predisposes the exocrine pancreas to profound DNA damage, and the frequency of invasive neoplasia is accentuated by the concomitant deregulation of p53.


Subject(s)
Adenocarcinoma/genetics , BRCA2 Protein/genetics , Carcinoma in Situ/genetics , Carcinoma, Pancreatic Ductal/genetics , Neoplasms, Experimental/genetics , Pancreatic Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Animals , BRCA2 Protein/metabolism , Carcinoma in Situ/drug therapy , Carcinoma in Situ/pathology , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/pathology , DNA Damage , Gene Knockdown Techniques , Genetic Association Studies , Hedgehog Proteins/metabolism , Kaplan-Meier Estimate , Liver Neoplasms/secondary , Mice , Neoplasm Transplantation , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Pancreas/pathology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Splenic Neoplasms/secondary , Tumor Suppressor Protein p53/metabolism
13.
Cancer Biol Ther ; 10(10): 1009-18, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20818175

ABSTRACT

Esophageal adenocarcinoma (EAC) arises in the backdrop of reflux-induced metaplastic phenomenon known as Barrett esophagus. The prognosis of advanced EAC is dismal, and there is an urgent need for identifying molecular targets for therapy. Serial Analysis of Gene Expression (SAGE) was performed on metachronous mucosal biopsies from a patient who underwent progression to EAC during endoscopic surveillance. SAGE confirmed significant upregulation of Axl "tags" during the multistep progression of Barrett esophagus to EAC. In a cohort of 92 surgically resected EACs, Axl overexpression was associated with shortened median survival on both univariate (p < 0.004) and multivariate (p < 0.036) analysis. Genetic knockdown of Axl receptor tyrosine kinase (RTK) function was enabled in two EAC lines (OE33 and JH-EsoAd1) using lentiviral short hairpin RNA (shRNA). Genetic knockdown of Axl in EAC cell lines inhibited invasion, migration, and in vivo engraftment, which was accompanied by downregulation in the activity of the Ral GTPase proteins (RalA and RalB). Restoration of Ral activation rescued the transformed phenotype of EAC cell lines, suggesting a novel effector mechanism for Axl in cancer cells. Pharmacological inhibition of Axl was enabled using a small molecule antagonist, R428 (Rigel Pharmaceuticals). Pharmacological inhibition of Axl with R428 in EAC cell lines significantly reduced anchorage-independent growth, invasion and migration. Blockade of Axl function abrogated phosphorylation of ERBB2 (Her-2/neu) at the Tyr877 residue, indicative of receptor crosstalk. Axl RTK is an adverse prognostic factor in EAC. The availability of small molecule inhibitors of Axl function provides a tractable strategy for molecular therapy of established EAC.


Subject(s)
Adenocarcinoma/pathology , Barrett Esophagus/pathology , Esophageal Neoplasms/pathology , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction/drug effects , Adenocarcinoma/drug therapy , Adenocarcinoma/enzymology , Aged , Animals , Barrett Esophagus/drug therapy , Barrett Esophagus/enzymology , Benzocycloheptenes/pharmacology , Cell Movement/drug effects , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/enzymology , Female , Humans , Immunoenzyme Techniques , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Lapatinib , Lymphatic Metastasis , Male , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Quinazolines/pharmacology , RNA, Small Interfering/genetics , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/genetics , Survival Rate , Triazoles/pharmacology , ral GTP-Binding Proteins/antagonists & inhibitors , ral GTP-Binding Proteins/genetics , ral GTP-Binding Proteins/metabolism , Axl Receptor Tyrosine Kinase
14.
Mol Cancer Ther ; 9(8): 2255-64, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20647339

ABSTRACT

Curcumin or diferuloylmethane is a yellow polyphenol extracted from the rhizome of turmeric (Curcuma longa). A large volume (several hundreds) of published reports has established the anticancer and chemopreventative properties of curcumin in preclinical models of every known major cancer type. Nevertheless, the clinical translation of curcumin has been significantly hampered due to its poor systemic bioavailability, which mandates that patients consume up to 8 to 10 g of the free drug orally each day to achieve detectable levels in circulation. We have engineered a polymeric nanoparticle encapsulated curcumin formulation (NanoCurc) that shows remarkably higher systemic bioavailability in plasma and tissues compared with free curcumin upon parenteral administration. In xenograft models of human pancreatic cancer established in athymic mice, administration of parenteral NanoCurc significantly inhibits primary tumor growth in both subcutaneous and orthotopic settings. The combination of parenteral NanoCurc with gemcitabine results in enhanced tumor growth inhibition versus either single agent, suggesting an additive therapeutic influence in vivo. Furthermore, this combination completely abrogates systemic metastases in orthotopic pancreatic cancer xenograft models. Tumor growth inhibition is accompanied by significant reduction in activation of nuclear factor-kappaB, as well as significant reduction in expression of matrix metalloproteinase-9 and cyclin D1, in xenografts treated with NanoCurc and gemcitabine. NanoCurc is a promising new formulation that is able to overcome a major impediment for the clinical translation of curcumin to cancer patients by improving systemic bioavailability, and by extension, therapeutic efficacy.


Subject(s)
Curcumin/administration & dosage , Curcumin/therapeutic use , Nanoparticles/administration & dosage , Neoplasm Metastasis/prevention & control , Pancreatic Neoplasms/drug therapy , Polymers/administration & dosage , Xenograft Model Antitumor Assays , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biological Availability , Cell Line, Tumor , Cell Proliferation/drug effects , Curcumin/pharmacology , Cyclin D1/metabolism , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Disease Models, Animal , Down-Regulation/drug effects , Drug Synergism , Humans , Matrix Metalloproteinase 9/metabolism , Mice , NF-kappa B/metabolism , Neoplasm Metastasis/pathology , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/pathology , Subcutaneous Tissue/drug effects , Gemcitabine
15.
Lab Invest ; 90(5): 665-73, 2010 May.
Article in English | MEDLINE | ID: mdl-20231822

ABSTRACT

Intraductal papillary mucinous neoplasms (IPMNs) are one of the three known curable precursor lesions of invasive pancreatic ductal adenocarcinoma, an almost uniformly fatal disease. Cell lines from IPMNs and their invasive counterparts should be valuable to identify gene mutations critical to IPMN carcinogenesis, and permit high-throughput screening to identify drugs that cause regression of these lesions. To advance the study of the biological features of IPMNs, we attempted in vivo and in vitro growth of selected IPMNs based on the hypothesis that IPMNs could be grown in the most severely immunodeficient mice. We examined 14 cases by implanting them into nude, severe combined immunodeficient (SCID), and NOD/SCID/IL2Rgamma(null) (NOG) mice, in addition to direct culture, to generate tumor xenografts and cell lines. One sample was directly cultured only. Thirteen tumors were implanted into the three types of mice, including 10 tumors implanted into the triple immunodeficient NOG mice, in which the majority (8 of 10) grew. This included five IPMNs lacking an invasive component. One of the explanted IPMNs, with an associated invasive carcinoma, was successfully established as a cell line. Tumorigenicity was confirmed by growth in soft agar, growth in immunodeficient mice, and the homozygous deletion of p16/cdkn2a. Epithelial differentiation of the cell line was documented by cytokeratin expression. Patient origin was confirmed using DNA fingerprinting. Most non-invasive IPMNs grow in NOG mice. We successfully established one IPMN cell line, and plan to use it to clarify the molecular pathogenesis of IPMNs.


Subject(s)
Adenocarcinoma, Mucinous/pathology , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Papillary/pathology , Pancreatic Neoplasms/pathology , Adenocarcinoma, Mucinous/genetics , Adenocarcinoma, Mucinous/metabolism , Adult , Aged , Aged, 80 and over , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Papillary/genetics , Carcinoma, Papillary/metabolism , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , DNA Fingerprinting , Female , Humans , Immunohistochemistry , Male , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, Nude , Mice, SCID , Middle Aged , Neoplasm Transplantation , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Transplantation, Heterologous
16.
Pancreatology ; 10(1): 66-73, 2010.
Article in English | MEDLINE | ID: mdl-20332664

ABSTRACT

BACKGROUND/AIMS: Pancreatic intraepithelial neoplasia (PanIN) is the most common noninvasive precursor to invasive pancreatic adenocarcinoma. Misexpression of microRNAs (miRNAs) is commonly encountered in invasive neoplasia; however, miRNA abnormalities in PanIN lesions have not been documented. METHODS: Three candidate miRNAs (miR-21, miR-155, and miR-221) previously reported as overexpressed in pancreatic cancers were assessed in 31 microdissected PanINs (14 PanIN-1, 9 PanIN-2, 8 PanIN-3) using quantitative reverse transcription PCR (qRT-PCR). Subsequently, miR-155 was evaluated by locked nucleic acid in situ hybridization (LNA-ISH) in PanIN tissue microarrays. RESULTS: Relative to microdissected non-neoplastic ductal epithelium, significant overexpression of miR-155 was observed in both PanIN-2 (2.6-fold, p = 0.02) and in PanIN-3 (7.4-fold, p = 0.014), while borderline significant overexpression of miR-21 (2.5-fold, p = 0.049) was observed in PanIN-3 only. In contrast, no significant differences in miR-221 levels were observed between ductal epithelium and PanIN lesions by qRT-PCR. LNA-ISH confirmed the aberrant expression of miR-155 in PanIN-2 (9 of 20, 45%) and in PanIN-3 (8 of 13, 62%), respectively, when compared with normal ductal epithelium (0 of 10) (p < 0.01). CONCLUSIONS: Abnormalities of miRNA expression are observed in the multistep progression of pancreatic cancer, with miR-155 aberrations demonstrable at the stage of PanIN-2, and miR-21 abnormalities at the stage of PanIN-3 lesions. and IAP.


Subject(s)
Adenocarcinoma/genetics , MicroRNAs/biosynthesis , Pancreatic Neoplasms/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Carcinoma in Situ/genetics , Carcinoma in Situ/metabolism , Carcinoma in Situ/pathology , Humans , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology
17.
Med Oncol ; 27(4): 1096-101, 2010 Dec.
Article in English | MEDLINE | ID: mdl-19859844

ABSTRACT

Curcumin is a naturally occurring polyphenolic compound found in the turmeric, which is used as food additive in Indian cooking and as a therapeutic agent in traditional Indian medicine. Curcumin is currently under investigation as a chemotherapeutic and chemopreventive agent in adult cancer models at both pre-clinical and clinical levels. In this preliminary study, we show that curcumin is effective in causing cell cycle arrest, inducing apoptosis, and suppressing colony formation in the Ewing sarcoma cell line SK-NEP-1. Curcumin causes upregulation of cleaved caspase 3 and downregulation of phospho-Akt, producing apoptosis in Ewing sarcoma cells at an inhibitory concentration 50% (IC50) of approximately 4 µM. Our findings indicate a need for further evaluation of curcumin in chemotherapy and chemoprevention of Ewing sarcoma.


Subject(s)
Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Bone Neoplasms/pathology , Cell Cycle/drug effects , Curcumin/therapeutic use , Sarcoma, Ewing/pathology , Blotting, Western , Bone Neoplasms/drug therapy , Cell Line, Tumor , Flow Cytometry , Humans , Proto-Oncogene Proteins c-akt/metabolism , Sarcoma, Ewing/drug therapy
18.
J Mol Diagn ; 11(6): 583-9, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19815696

ABSTRACT

Patients with biliary tract carcinoma have a poor prognosis. Early detection efforts are urgently needed to ameliorate the dismal prognosis for these patients. Mutations of the KRAS2 gene are one of the most common genetic aberrations in this cancer. In this study, we used LigAmp, an ultrasensitive technology for detecting point mutations, to analyze KRAS2 mutations in patients with a variety of neoplastic and non-neoplastic pancreatobiliary diseases. DNA was isolated from 64 samples, including 44 bile samples and 20 serum samples. Oligonucleotides specific for KRAS2 G35A (GAT, G12D), G35T (GTT, G12V), and G34A (AGT, G12S) mutations were used. KRAS2 mutations were detected in 14 of 16 (87.5%) neoplastic bile samples and in 9 of 28 (32.1%) non-neoplastic bile samples. However, the mutation levels were significantly lower in the non-neoplastic bile (median = 0.4%) compared with those in the neoplastic bile (median = 5.1%). KRAS2 mutations were also detected in 9 of 11 (81.8%) serum samples from patients with biliary tract carcinoma, which was further confirmed by cloning BstN1-refractory PCR products and DNA sequencing. However, KRAS2 mutations were not present in the sera from eight patients with benign pancreatobiliary diseases. These data demonstrate that KRAS2 mutations are detectable in both bile and serum using LigAmp. This technology has the potential for early biliary tract carcinoma detection and possibly for residual disease monitoring post-therapy.


Subject(s)
Bile/metabolism , Biliary Tract Neoplasms/genetics , Carcinoma/genetics , DNA Mutational Analysis/methods , Proto-Oncogene Proteins/blood , Proto-Oncogene Proteins/genetics , ras Proteins/blood , ras Proteins/genetics , Biliary Tract Neoplasms/blood , Biliary Tract Neoplasms/pathology , Humans , Mutation , Proto-Oncogene Proteins p21(ras) , Reproducibility of Results
19.
Pancreatology ; 9(3): 293-301, 2009.
Article in English | MEDLINE | ID: mdl-19407485

ABSTRACT

Aberrant expression of microRNAs (miRNAs) has emerged as an important hallmark of cancer. However, the putative mechanisms regulating miRNAs per se are only partially known. It is well established that many tumor suppressor genes in human cancers are silenced by chromatin alterations, including promoter methylation and histone deacetylation. We postulated that miRNAs undergo similar epigenetic inactivation in pancreatic cancer. Two human pancreatic cancer cell lines - MiaPACA-2 and PANC-1 - were treated with the demethylating agent, 5-aza-2'-deoxycytidine (5-Aza-dC) or the histone deacetylase inhibitor, trichostatin A, as well as the combination of the two. Expression of miRNAs in control and treated cell lines was assessed using a custom microarray platform. Fourteen miRNAs were upregulated two-fold or greater in each of the cell lines following exposure to both chromatin-modifying agents, including 5 that were in common (miR-107, miR-103, miR-29a, miR-29b, and miR-320) to both MiaPACA-2 and PANC-1. The differential overexpression of miR-107 in the treated cancer cell lines was confirmed by Northern blot assays. Methylation-specific PCR assays for assessment of CpG island methylation status in the 5' promoter region of the miR-107 primary transcript demonstrated complete loss of methylation upon exposure to 5-Aza-dC. Enforced expression of miR-107 in MiaPACA-2 and PANC-1 cells downregulated in vitro growth, and this was associated with repression of the putative miR-107 target, cyclin-dependent kinase 6, thereby providing a functional basis for the epigenetic inactivation of this miRNA in pancreatic cancer.


Subject(s)
Cyclin-Dependent Kinase 6/genetics , Gene Silencing , MicroRNAs/genetics , Pancreatic Neoplasms/genetics , Conserved Sequence , DNA Primers , DNA, Neoplasm/genetics , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Oligonucleotide Array Sequence Analysis , Pancreatic Neoplasms/enzymology , Promoter Regions, Genetic , RNA, Neoplasm/genetics
20.
Cancer Biol Ther ; 8(7): 618-26, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19252414

ABSTRACT

BACKGROUND: Pancreatic cancer is a near uniformly lethal disease and a better understanding of the molecular basis of this malignancy may lead to improved therapeutics. The Axl receptor tyrosine kinase is implicated in cellular transformation and tumor progression, although its role in pancreatic cancer has not been previously documented. RESULTS: Axl labeling was present in 54 of 99 (55%), and was absent in 45 of 99 (45%) cases, respectively. Axl expression in pancreatic cancer was significantly associated with lymph node metastases (p < 0.01), and a shorter median survival (12 versus 18 months, p < 0.01), than in tumors with negative labeling. Stable knockdown of Axl resulted in significant reduction in cell viability (p < 0.001), anchorage independent growth (p = 0.0031), as well as attenuation of migratory (p < 0.001) and invasive properties (p < 0.005), compared to vector-transfected cells. Profound inhibition of p42/p44 MAP kinase and PI-3kinase/Akt effector pathways was observed in MIAPaCa-2 cells with loss of Axl function. The reduction in invasion and migration upon Axl knockdown was mirrored by a decrease in the amounts of activated (GTP-bound) GTPase proteins Rho and Rac, significant downregulation in transcript levels of the epithelial mesenchymal transition (EMT)-associated transcription factors slug, snail and twist, and significant decrease in matrix metalloproteinase MMP-9 mRNA levels. MATERIALS: The immunohistochemical expression of Axl protein was assessed in a panel of 99 archival pancreatic cancers. Endogenous Axl expression was stably downregulated by lentiviral short hairpin shRNA directed against AXL mRNA in MIAPaCa-2 cells, and the effects on cell viability, anchorage independent growth, invasion, migration and intracellular effector pathways was assessed, by comparing to lentiviral vector-transfected cells. CONCLUSION: Expression of Axl tyrosine kinase in pancreatic cancers confers an adverse prognostic influence, and represents a new therapeutic target in this malignancy.


Subject(s)
Carcinoma, Pancreatic Ductal/enzymology , Oncogene Proteins/biosynthesis , Pancreatic Neoplasms/enzymology , Receptor Protein-Tyrosine Kinases/biosynthesis , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/therapy , Cell Growth Processes/physiology , Cell Line, Tumor , Cell Movement/physiology , Female , Humans , Immunohistochemistry , Lymphatic Metastasis , Male , Neoplasm Invasiveness , Oncogene Proteins/antagonists & inhibitors , Oncogene Proteins/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/therapy , Prognosis , Proto-Oncogene Proteins , RNA, Small Interfering/genetics , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/genetics , Axl Receptor Tyrosine Kinase
SELECTION OF CITATIONS
SEARCH DETAIL
...