Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Int J Nanomedicine ; 16: 2297-2309, 2021.
Article in English | MEDLINE | ID: mdl-33776434

ABSTRACT

PURPOSE: Accurate tumor identification and staging can be difficult. Aptamer-targeted indocyanine green (ICG)-nanoparticles can enhance near-infrared fluorescent imaging of pancreatic and prostate tumors and could improve early cancer detection. This project explored whether calcium-phosphosilicate nanoparticles, also known as NanoJackets (NJs), that were bioconjugated with a tumor-specific targeting DNA aptamer could improve the non-invasive detection of pancreatic and prostate tumors. METHODS: Using in vivo near-infrared optical imaging and ex vivo fluorescence analysis, DNA aptamer-targeted ICG-loaded NJs were compared to untargeted NJs for detection of tumors. RESULTS: Nanoparticles were bioconjugated with the DNA aptamer AP1153, which binds to the CCK-B receptor (CCKBR). Aptamer bioconjugated NJs were not significantly increased in size compared with unconjugated nanoparticles. AP1153-ICG-NJ accumulation in orthotopic pancreatic tumors peaked at 18 h post-injection and the ICG signal was cleared by 36 h with no evidence on uptake by non-tumor tissues. Ex vivo tumor imaging confirmed the aptamer-targeted NJs accumulated to higher levels than untargeted NJs, were not taken up by normal pancreas, exited from the tumor vasculature, and were well-dispersed throughout pancreatic and prostate tumors despite extensive fibrosis. Specificity for AP1153-NJ binding to the CCK-B receptor on pancreatic tumor cells was confirmed by pre-treating tumor-bearing mice with the CCK receptor antagonist proglumide. Proglumide pre-treatment reduced the in vivo tumoral accumulation of AP1153-NJs to levels comparable to that of untargeted NJs. CONCLUSION: Through specific interactions with CCK-B receptors, tumor-targeted nanoparticles containing either ICG or rhodamine WT were well distributed throughout the matrix of both pancreatic and prostate tumors. Tumor-targeted NJs carrying various imaging agents can enhance tumor detection.


Subject(s)
Aptamers, Nucleotide/chemistry , Diagnostic Imaging , Nanoparticles/chemistry , Pancreatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/diagnostic imaging , Silicates/chemistry , Animals , Calcium , Cell Line, Tumor , Coloring Agents , Fluorescence , Humans , Indocyanine Green/chemistry , Infrared Rays , Male , Mice , Neovascularization, Pathologic/diagnostic imaging , Pancreatic Neoplasms/blood supply , Prostatic Neoplasms/blood supply , Receptors, Cholecystokinin/metabolism , Rhodamines/chemistry , Tumor Microenvironment
2.
Nanomedicine ; 13(7): 2313-2324, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28673852

ABSTRACT

Drug resistant cancers like pancreatic ductal adenocarcinoma (PDAC) are difficult to treat, and nanoparticle drug delivery systems can overcome some of the limitations of conventional systemic chemotherapy. In this study, we demonstrate that FdUMP and dFdCMP, the bioactive, phosphorylated metabolites of the chemotherapy drugs 5-FU and gemcitabine, can be encapsulated into calcium phosphosilicate nanoparticles (CPSNPs). The non-phosphorylated drug analogs were not well encapsulated by CPSNPs, suggesting the phosphate modification is essential for effective encapsulation. In vitro proliferation assays, cell cycle analyses and/or thymidylate synthase inhibition assays verified that CPSNP-encapsulated phospho-drugs retained biological activity. Analysis of orthotopic tumors from mice treated systemically with tumor-targeted FdUMP-CPSNPs confirmed the in vivo up take of these particles by PDAC tumor cells and release of active drug cargos intracellularly. These findings demonstrate a novel methodology to efficiently encapsulate chemotherapeutic agents into the CPSNPs and to effectively deliver them to pancreatic tumor cells.


Subject(s)
Antineoplastic Agents/administration & dosage , Calcium Compounds/chemistry , Carcinoma, Pancreatic Ductal/drug therapy , Deoxycytidine/analogs & derivatives , Fluorouracil/administration & dosage , Nanoparticles/chemistry , Pancreatic Neoplasms/drug therapy , Silicates/chemistry , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Deoxycytidine/administration & dosage , Deoxycytidine/chemistry , Deoxycytidine/therapeutic use , Drug Carriers/chemistry , Drug Delivery Systems , Fluorouracil/analogs & derivatives , Fluorouracil/therapeutic use , Humans , Male , Mice , Mice, Nude , Nanoparticles/ultrastructure , Phosphorylation , Xenograft Model Antitumor Assays , Gemcitabine
3.
Nucleic Acid Ther ; 27(1): 23-35, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27754762

ABSTRACT

Pancreatic ductal adenocarcinomas (PDACs) constitutively express the G-protein-coupled cholecystokinin B receptor (CCKBR). In this study, we identified DNA aptamers (APs) that bind to the CCKBR and describe their characterization and targeting efficacy. Using dual SELEX selection against "exposed" CCKBR peptides and CCKBR-expressing PDAC cells, a pool of DNA APs was identified. Further downselection was based on predicted structures and properties, and we selected eight APs for initial characterizations. The APs bound specifically to the CCKBR, and we showed not only that they did not stimulate proliferation of PDAC cell lines but rather inhibited their proliferation. We chose one AP, termed AP1153, for further binding and localization studies. We found that AP1153 did not activate CCKBR signaling pathways, and three-dimensional Confocal microscopy showed that AP1153 was internalized by PDAC cells in a receptor-mediated manner. AP1153 showed a binding affinity of 15 pM. Bioconjugation of AP1153 to the surface of fluorescent NPs greatly facilitated delivery of NPs to PDAC tumors in vivo. The selectivity of this AP-targeted NP delivery system holds promise for enhanced early detection of PDAC lesions as well as improved chemotherapeutic treatments for PDAC patients.


Subject(s)
Aptamers, Nucleotide/therapeutic use , Carcinoma, Pancreatic Ductal/therapy , Nanoconjugates/administration & dosage , Pancreatic Neoplasms/therapy , Receptor, Cholecystokinin B/therapeutic use , Animals , Aptamers, Nucleotide/genetics , Aptamers, Nucleotide/metabolism , COS Cells , Carcinoma, Pancreatic Ductal/metabolism , Cell Line, Tumor , Chlorocebus aethiops , Drug Delivery Systems , Humans , Imaging, Three-Dimensional , Male , Mice , Mice, Nude , Microscopy, Confocal , Nanoconjugates/chemistry , Optical Imaging , Pancreatic Neoplasms/metabolism , Receptor, Cholecystokinin B/genetics , Receptor, Cholecystokinin B/metabolism , Theranostic Nanomedicine , Xenograft Model Antitumor Assays
4.
Clin Transl Gastroenterol ; 7: e134, 2016 Jan 07.
Article in English | MEDLINE | ID: mdl-26741064

ABSTRACT

OBJECTIVES: Today, genetic biomarkers have been demonstrated to play an important role in identifying at-risk subjects for familial or inherited cancers. We have identified a single-nucleotide polymorphism (SNP) that results in missplicing of the cholecystokinin (CCK) receptor gene and expressing a larger mutated receptor in pancreatic cancer. The purpose of this study was to evaluate the significance and specificity of this SNP as a potential biomarker in patients with pancreatic cancer compared with other gastrointestinal (GI) cancers that also have CCK receptors. METHODS: DNA was isolated and genotyped for the CCK receptor SNP from frozen tumor tissue from banked specimens of patients with pancreas, gastric, or colon cancer and from human cancer cell lines. Genotype and allelic frequencies were compared between the cancer cohort and two normal control databases using Fisher's exact test and odds ratio (OR). The Kaplan-Meier method was used to estimate the survival for patients with the CCK-B receptor SNP compared with those with the wild-type genotype. Immunohistochemical staining of cancer cells was done to detect the mutated receptor. RESULTS: Colon and gastric cancer patients had similar genotype frequencies for the CCK receptor SNP as that reported in the normal population. In contrast, the prevalence of the SNP in subjects with pancreatic cancer was twice that of controls and other GI cancers. Survival was adversely affected by the presence of the SNP only in those with pancreatic cancer. Immunoreactivity for the mutated receptor was positive in pancreatic cancer tissues with the SNP but absent in other GI cancers. CONCLUSIONS: A SNP of the CCK receptor is significantly increased in patients with pancreatic cancer but not in those with other GI malignancies. Therefore, this SNP may be a potential biomarker for pancreatic cancer.

5.
Pancreas ; 43(7): 1050-9, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25058882

ABSTRACT

OBJECTIVES: Exogenous administration of cholecystokinin (CCK) induces hypertrophy and hyperplasia of the pancreas with an increase in DNA content. We hypothesized that endogenous CCK is involved in the malignant progression of pancreatic intraepithelial neoplasia (PanIN) lesions and the fibrosis associated with pancreatic cancer. METHODS: The presence of CCK receptors in early PanIN lesions was examined by immunohistochemistry in mouse and human pancreas. Pdx1-Cre/LSL-Kras transgenic mice were randomized to receive either untreated drinking water or water supplemented with a CCK receptor antagonist (proglumide, 0.1 mg/mL). Pancreas from the mice were removed and examined histologically for number and grade of PanINs after 1, 2, or 4 months of antagonist therapy. RESULTS: Both CCK-A and CCK-B receptors were identified in early stage PanINs from mouse and human pancreas. The grade of PanIN lesions was reversed, and progression to advanced lesions arrested in mice treated with proglumide compared with the controls (P = 0.004). Furthermore, pancreatic fibrosis was significantly reduced in antagonist-treated animals compared with vehicle (P < 0.001). CONCLUSIONS: These findings demonstrate that endogenous CCK is in part responsible for the development and progression of pancreatic cancer. The use of CCK receptor antagonists may have a role in cancer prophylaxis in high-risk subjects and may reduce fibrosis in the microenvironment.


Subject(s)
Carcinoma in Situ/prevention & control , Pancreas/drug effects , Pancreatic Neoplasms/prevention & control , Precancerous Conditions/drug therapy , Proglumide/therapeutic use , Receptor, Cholecystokinin A/antagonists & inhibitors , Receptor, Cholecystokinin B/antagonists & inhibitors , Animals , Carcinoma in Situ/chemistry , Carcinoma in Situ/drug therapy , Carcinoma in Situ/pathology , Cholecystokinin/physiology , Disease Progression , Drug Screening Assays, Antitumor , Fibrosis , Humans , Mice , Mice, Transgenic , Pancreas/chemistry , Pancreas/pathology , Pancreatic Neoplasms/chemistry , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Pancreatitis/prevention & control , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , Proglumide/pharmacology , Random Allocation , Receptor, Cholecystokinin A/analysis , Receptor, Cholecystokinin B/analysis
6.
Dig Dis Sci ; 59(6): 1180-91, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24817409

ABSTRACT

BACKGROUND: Obesity and dietary fat are associated with increased risk of several malignancies including pancreatic cancer. The incidence of pancreatic cancer is increased in countries that consume diets high in fat. AIM: The purpose of this study was to assess the relationship and mechanism of action between dietary fat and endogenous cholecystokinin (CCK) on pancreatic tumor growth and metastasis in an immunocompetent animal model. METHODS: C57BL/6 mice were placed on regular, low-fat, or high-fat diets for 8 weeks before establishment of Panc-02 orthotopic pancreatic tumors. Mice were then treated with a CCK-A receptor antagonist, devazepide, or vehicle for an additional 2.5 weeks. Pancreas tumors were weighed and metastases counted. Blood CCK levels were measured by radioimmunoassay (RIA). Tissues were examined histologically and studied for genes associated with metastasis by RT-PCR array. Effects of the CCK antagonist on Panc-02 cells invasiveness was assessed in a Matrigel invasion assay. RESULTS: Mice that received the high-fat diet had larger tumors and tenfold higher serum CCK levels by RIA compared to normal diet controls (p < 0.01). Pancreatic tumors in high-fat diet mice treated with the antagonist had fewer intravascular tumor emboli and metastases compared to controls. The reduction in tumor emboli correlated with decreased vascular endothelial growth factor-A (VEGF-A) expression in tumors (p < 6 × 10(-9)). In vitro invasiveness of Panc-02 cells also was reduced by CCK-A receptor antagonist treatment (p = 1.33 × 10(-6)). CONCLUSION: CCK is a mediator of dietary fat-associated pancreatic cancer. CCK is also involved in the invasiveness of pancreatic tumors through a mechanism involving VEGF-A.


Subject(s)
Cholecystokinin/metabolism , Dietary Fats/adverse effects , Pancreatic Neoplasms/metabolism , Animals , Blood Glucose , Cell Line, Tumor , Devazepide/pharmacology , Dietary Fats/administration & dosage , Dose-Response Relationship, Drug , Embolism/prevention & control , Hormone Antagonists/pharmacology , Male , Mice , Mice, Inbred C57BL , Neoplasm Invasiveness , Neoplasm Metastasis/prevention & control , Pancreatic Neoplasms/pathology , Radioimmunoassay
7.
Am J Physiol Gastrointest Liver Physiol ; 302(11): G1244-52, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22442157

ABSTRACT

Gastrin stimulates the growth of pancreatic cancer cells through the activation of the cholecystokinin-B receptor (CCK-BR), which has been found to be overexpressed in pancreatic cancer. In this study, we proposed that the CCK-BR drives growth of pancreatic cancer; hence, interruption of CCK-BR activity could potentially be an ideal target for cancer therapeutics. The effect of CCK-BR downregulation in the human pancreatic adenocarcinoma cells was examined by utilizing specific CCK-BR-targeted RNA interference reagents. The CCK-BR receptor expression was both transiently and stably downregulated by transfection with selective CCK-BR small-interfering RNA or short-hairpin RNA, respectively, and the effects on cell growth and apoptosis were assessed. CCK-BR downregulation resulted in reduced cancer cell proliferation, decreased DNA synthesis, and cell cycle arrest as demonstrated by an inhibition of G(1) to S phase progression. Furthermore, CCK-BR downregulation increased caspase-3 activity, TUNEL-positive cells, and decreased X-linked inhibitor of apoptosis protein expression, suggesting apoptotic activity. Pancreatic cancer cell mobility was decreased when the CCK-BR was downregulated, as assessed by a migration assay. These results show the importance of the CCK-BR in regulation of growth and apoptosis in pancreatic cancer. Strategies to decrease the CCK-BR expression and activity may be beneficial for the development of new methods to improve the treatment for patients with pancreatic cancer.


Subject(s)
Apoptosis/genetics , Cell Proliferation , Down-Regulation , Pancreas/metabolism , Receptor, Cholecystokinin B/genetics , Caspase 3/genetics , Caspase 3/metabolism , Cell Cycle Checkpoints , Cell Line, Tumor , Cell Movement/genetics , Gene Expression Regulation, Neoplastic , Humans , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , RNA Interference , Receptor, Cholecystokinin B/metabolism , X-Linked Inhibitor of Apoptosis Protein/genetics , X-Linked Inhibitor of Apoptosis Protein/metabolism
8.
Cancer Biol Ther ; 13(3): 164-74, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22277584

ABSTRACT

There currently are no tests available for early diagnosis or for the identification of patients at risk for development of pancreatic cancer. We report the discovery of single nucleotide polymorphism (SNP) in the cholecystokinin B receptor (CCKBR) gene predicts survival and risk of pancreatic cancer. Growth of human pancreatic cancer is stimulated by gastrin through the CCKBR and an alternatively spliced isoform of the CCKBR gene called CCKCR. One hundred and ten surgically resected benign and malignant pancreatic tissues as well as normal pancreas were prospectively evaluated for CCKBR genotype and protein expression. Analysis demonstrated the expression of the spliced isoform, CCKCR, was associated with a (SNP) (C > A) at position 32 of the intron 4 (IVS 4) of the CCKBR gene. Since the SNP is within an intron, it has not previously been identified in the GWAS studies. Only patients with the A/A or A/C genotypes, exhibited immunoreactivity to a selective CCKCR antibody. Survival among pancreatic cancer patients with the A-SNP was significantly shorter (p = 0.0001, hazard ratio = 3.63) compared with individuals with C/C genotype. Other variables such as surgical margins, lymph node status, histologic grade or adjuvant chemotherapy were not associated with survival. Furthermore, having one or two of the A-alleles was found to increase the risk of pancreatic adenocarcinoma by 174% (p = 0.0192) compared with the C/C wild type. Cancer cells transfected to overexpress the CCKCR demonstrated increased proliferation over controls. Genetic screening for this SNP may aid in early detection of pancreatic cancer in high risk subjects.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Pancreatic Neoplasms/genetics , Receptor, Cholecystokinin B/genetics , Aged , Antibodies, Monoclonal, Murine-Derived/chemistry , Antibody Specificity , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/pathology , Case-Control Studies , Cell Line, Tumor , Cell Proliferation , Female , Genome-Wide Association Study , Genotype , Humans , Kaplan-Meier Estimate , Male , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Polymorphism, Single Nucleotide , Protein Isoforms/genetics , Protein Isoforms/immunology , Protein Isoforms/metabolism , Receptor, Cholecystokinin B/immunology , Receptor, Cholecystokinin B/metabolism , Risk Factors
9.
Dig Dis Sci ; 56(7): 2088-97, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21380937

ABSTRACT

BACKGROUND: Endogenous opioid peptides have been shown to play a role in the development and/or perpetuation of inflammation. We hypothesize that the endogenous opioid system is involved in inflammatory bowel disease, and antagonism of the opioid-opioid receptor will lead to reversal of inflammation. AIMS: A randomized double-blind placebo-controlled study was designed to test the efficacy and safety of an opioid antagonist for 12 weeks in adults with active Crohn's disease. METHODS: Forty subjects with active Crohn's disease were enrolled in the study. Randomized patients received daily oral administration of 4.5-mg naltrexone or placebo. Providers and patients were masked to treatment assignment. The primary outcome was the proportion of subjects in each arm with a 70-point decline in Crohn's Disease Activity Index score (CDAI). The secondary outcome included mucosal healing based upon colonoscopy appearance and histology. RESULTS: Eighty-eight percent of those treated with naltrexone had at least a 70-point decline in CDAI scores compared to 40% of placebo-treated patients (p = 0.009). After 12 weeks, 78% of subjects treated with naltrexone exhibited an endoscopic response as indicated by a 5-point decline in the Crohn's disease endoscopy index severity score (CDEIS) from baseline compared to 28% response in placebo-treated controls (p = 0.008), and 33% achieved remission with a CDEIS score <6, whereas only 8% of those on placebo showed the same change. Fatigue was the only side effect reported that was significantly greater in subjects receiving placebo. CONCLUSIONS: Naltrexone improves clinical and inflammatory activity of subjects with moderate to severe Crohn's disease compared to placebo-treated controls. Strategies to alter the endogenous opioid system provide promise for the treatment of Crohn's disease.


Subject(s)
Crohn Disease/drug therapy , Intestinal Mucosa/drug effects , Naltrexone/therapeutic use , Narcotic Antagonists/therapeutic use , Adult , Aged , Colonoscopy , Fatigue/chemically induced , Female , Humans , Male , Middle Aged , Naltrexone/adverse effects , Narcotic Antagonists/adverse effects , Remission Induction , Severity of Illness Index , Treatment Outcome , Young Adult
10.
Pancreas ; 38(5): e151-61, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19465883

ABSTRACT

OBJECTIVES: This study evaluated the effects of gastrin messenger RNA (mRNA) down-regulation on growth of human pancreatic cancer. METHODS: Gastrin expression was examined in human pancreatic cancer cell lines by reverse transcriptase-polymerase chain reaction, and peptide expression was assessed by immunocytochemistry. Gastrin was down-regulated using either stable transfection of an antisense gastrin cDNA or 1 of 3 shRNA (short hairpin RNA) constructs. Tumor formation was evaluated after either subcutaneous or orthotopic injections into nude mice. The effect of nanoliposomes loaded with gastrin siRNA (small interfering RNA) was tested in mice bearing pancreatic tumors. RESULTS: Stable transfection of gastrin antisense or shRNAs into BxPC-3 cells resulted in clones with more than 90% reduction in gastrin mRNA. Tumor growth rate and incidence of metastases in both wild-type and transfected pancreatic cancer cells were directly proportional to the degrees of gastrin mRNA expression. Immunofluorescence analysis confirmed that gastrin peptide levels were decreased in antisense and shRNA tumors. Gastrin knockdown clones had lower Ki-67 and increased cleaved caspase-3 staining, consistent with known effects of gastrin on proliferation and apoptosis. Tumors in mice treated with gastrin siRNA were smaller than controls. CONCLUSIONS: These results suggest that RNAi targeting of gastrin could serve as an effective treatment for pancreatic cancer.


Subject(s)
Cell Proliferation , Gastrins/genetics , Pancreatic Neoplasms/pathology , RNA Interference , Animals , Cell Line, Tumor , Chromatography, High Pressure Liquid , Fluorescent Antibody Technique , Gastrins/metabolism , Humans , Ki-67 Antigen/analysis , Liposomes , Mass Spectrometry , Mice , Mice, Nude , Nanotechnology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/therapy , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Tumor Burden , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...