Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Cancer Res ; 79(9): 2327-2338, 2019 05 01.
Article in English | MEDLINE | ID: mdl-31043430

ABSTRACT

When pancreatic cancer cannot be removed surgically, patients frequently experience morbidity and death from progression of their primary tumor. Radiation therapy (RT) cannot yet substitute for an operation because radiation causes fatal bleeding and ulceration of the nearby stomach and intestines before achieving tumor control. There are no FDA-approved medications that prevent or reduce radiation-induced gastrointestinal injury. Here, we overcome this fundamental problem of anatomy and biology with the use of the oral EGLN inhibitor FG-4592, which selectively protects the intestinal tract from radiation toxicity without protecting tumors. A total of 70 KPC mice with autochthonous pancreatic tumors received oral FG-4592 or vehicle control ± ablative RT to a cumulative 75 Gy administered in 15 daily fractions to a limited tumor field. Although ablative RT reduced complications from local tumor progression, fatal gastrointestinal bleeding was observed in 56% of mice that received high-dose RT with vehicle control. However, radiation-induced bleeding was completely ameliorated in mice that received high-dose RT with FG-4592 (0% bleeding, P < 0.0001 compared with vehicle). Furthermore, FG-4592 reduced epithelial apoptosis by half (P = 0.002) and increased intestinal microvessel density by 80% compared with vehicle controls. EGLN inhibition did not stimulate cancer growth, as treatment with FG-4592 alone, or overexpression of HIF2 within KPC tumors independently improved survival. Thus, we provide a proof of concept for the selective protection of the intestinal tract by the EGLN inhibition to enable ablative doses of cytotoxic therapy in unresectable pancreatic cancer by reducing untoward morbidity and death from radiation-induced gastrointestinal bleeding. SIGNIFICANCE: Selective protection of the intestinal tract by EGLN inhibition enables potentially definitive doses of radiation therapy. This might allow radiation to be a surgical surrogate for unresectable pancreatic cancer.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/79/9/2327/F1.large.jpg.


Subject(s)
Glycine/analogs & derivatives , Hypoxia-Inducible Factor-Proline Dioxygenases/antagonists & inhibitors , Isoquinolines/pharmacology , Pancreatic Neoplasms/mortality , Radiation Injuries/prevention & control , Radiation-Protective Agents/pharmacology , Radiotherapy/mortality , Animals , Apoptosis , Female , Glycine/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/radiotherapy , Proto-Oncogene Proteins p21(ras)/physiology , Radiation Injuries/etiology , Radiation Injuries/mortality , Radiotherapy/adverse effects , Transcription Factors/physiology , Tumor Suppressor Protein p53/physiology
2.
Nanomedicine (Lond) ; 9(8): 1209-22, 2014.
Article in English | MEDLINE | ID: mdl-24063415

ABSTRACT

AIM: We report a magneto-fluorescent theranostic nanocomplex targeted to neutrophil gelatinase-associated lipocalin (NGAL) for imaging and therapy of pancreatic cancer. MATERIALS & METHODS: Gold nanoshells resonant at 810 nm were encapsulated in silica epilayers doped with iron oxide and the near-infrared (NIR) dye indocyanine green, resulting in theranostic gold nanoshells (TGNS), which were subsequently conjugated with antibodies targeting NGAL in AsPC-1-derived xenografts in nude mice. RESULTS: Anti-NGAL-conjugated TGNS specifically targeted pancreatic cancer cells in vitro and in vivo providing contrast for both NIR fluorescence and T2-weighted MRI with higher tumor contrast than can be obtained using long-circulating, but nontargeted, PEGylated nanoparticles. The nanocomplexes also enabled highly specific cancer cell death via NIR photothermal therapy in vitro. CONCLUSION: TGNS with embedded NIR and magnetic resonance contrasts can be specifically targeted to pancreatic cancer cells with expression of early disease marker NGAL, and enable molecularly targeted imaging and photothermal therapy.


Subject(s)
Gold/therapeutic use , Nanoshells/therapeutic use , Pancreas/pathology , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/therapy , Acute-Phase Proteins/metabolism , Animals , Cell Line, Tumor , Contrast Media/chemistry , Contrast Media/therapeutic use , Drug Delivery Systems , Female , Fluorescent Dyes/chemistry , Fluorescent Dyes/therapeutic use , Gold/chemistry , Humans , Hyperthermia, Induced , Lipocalin-2 , Lipocalins/metabolism , Magnetic Resonance Imaging , Magnets/chemistry , Mice, Nude , Nanoshells/chemistry , Oncogene Proteins/metabolism , Optical Imaging , Pancreatic Neoplasms/pathology , Phototherapy
3.
Clin Cancer Res ; 18(18): 4942-53, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22832932

ABSTRACT

PURPOSE: Development of chemoresistance, poor prognosis, and metastasis often renders the current treatments for colorectal cancer (CRC) ineffective. Whether ursolic acid, a component of numerous medicinal plants, either alone or in combination with capecitabine, can inhibit the growth and metastasis of human CRC was investigated. EXPERIMENTAL DESIGN: The effect of ursolic acid on proliferation of CRC cell lines was examined by mitochondrial dye uptake assay, apoptosis by esterase staining, NF-κB activation by DNA-binding assay, and protein expression by Western blot. The effect of ursolic acid on the growth and chemosensitization was also examined in orthotopically implanted CRC in nude mice. RESULTS: We found that ursolic acid inhibited the proliferation of different colon cancer cell lines. This is correlated with inhibition of constitutive NF-κB activation and downregulation of cell survival (Bcl-xL, Bcl-2, cFLIP, and survivin), proliferative (cyclin D1), and metastatic (MMP-9, VEGF, and ICAM-1) proteins. When examined in an orthotopic nude mouse model, ursolic acid significantly inhibited tumor volume, ascites formation, and distant organ metastasis, and this effect was enhanced with capecitabine. Immunohistochemistry of tumor tissue indicated that ursolic acid downregulated biomarkers of proliferation (Ki-67) and microvessel density (CD31). This effect was accompanied by suppression of NF-κB, STAT3, and ß-catenin. In addition, ursolic acid suppressed EGF receptor (EGFR) and induced p53 and p21 expression. We also observed bioavailability of ursolic acid in the serum and tissue of animals. CONCLUSION: Overall, our results show that ursolic acid can inhibit the growth and metastasis of CRC and further enhance the therapeutic effects of capecitabine through the suppression of multiple biomarkers linked to inflammation, proliferation, invasion, angiogenesis, and metastasis.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/drug therapy , Signal Transduction/drug effects , Triterpenes/therapeutic use , Animals , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/pharmacology , Antimetabolites, Antineoplastic/toxicity , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Apoptosis/drug effects , Capecitabine , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Deoxycytidine/toxicity , Disease Models, Animal , Drug Resistance, Neoplasm/drug effects , Drug Synergism , ErbB Receptors/metabolism , Fluorouracil/administration & dosage , Fluorouracil/analogs & derivatives , Fluorouracil/pharmacology , Fluorouracil/toxicity , Humans , Ki-67 Antigen/metabolism , Male , Mice , Mice, Nude , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Neoplasm Metastasis/drug therapy , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , STAT3 Transcription Factor/metabolism , Triterpenes/administration & dosage , Tumor Suppressor Proteins/metabolism , Xenograft Model Antitumor Assays , beta Catenin/metabolism , Ursolic Acid
4.
Int J Cancer ; 131(3): E292-303, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-21935918

ABSTRACT

Agents that can potentiate the efficacy of standard chemotherapy against pancreatic cancer are of great interest. Because of their low cost and safety, patients commonly use a variety of dietary supplements, although evidence of their efficacy is often lacking. One such commonly used food supplement is Zyflamend, a polyherbal preparation with potent anti-inflammatory activities and preclinical efficacy against prostate and oral cancer. Whether Zyflamend has any efficacy against human pancreatic cancer alone or in combination with gemcitibine, a commonly used agent, was examined in cell cultures and in an orthotopic mouse model. In vitro, Zyflamend inhibited the proliferation of pancreatic cancer cell lines regardless of p53 status and also enhanced gemcitabine-induced apoptosis. This finding correlated with inhibition of NF-κB activation by Zyflamend and suppression of cyclin D1, c-myc, COX-2, Bcl-2, IAP, survivin, VEGF, ICAM-1 and CXCR4. In nude mice, oral administration of Zyflamend alone significantly inhibited the growth of orthotopically transplanted human pancreatic tumors, and when combined with gemcitabine, further enhanced the antitumor effects. Immunohistochemical and Western blot analyses of tumor tissue showed that the suppression of pancreatic cancer growth correlated with inhibition of proliferation index marker (Ki-67), COX-2, MMP-9, NF-κB and VEGF. Overall, these results suggest that the concentrated multiherb product Zyflamend alone can inhibit the growth of human pancreatic tumors and, in addition, can sensitize pancreatic cancers to gemcitabine through the suppression of multiple targets linked to tumorigenesis.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Deoxycytidine/analogs & derivatives , Pancreatic Neoplasms/drug therapy , Plant Extracts/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Deoxycytidine/pharmacology , Drug Synergism , Gene Expression Regulation, Neoplastic , Humans , Ki-67 Antigen/analysis , Male , Mice , Mice, Nude , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Tumor Suppressor Protein p53/analysis , Xenograft Model Antitumor Assays , Gemcitabine
5.
Int J Nanomedicine ; 6: 259-69, 2011.
Article in English | MEDLINE | ID: mdl-21423588

ABSTRACT

PURPOSE: Gold nanoshells (NSs) have already shown great promise as photothermal actuators for cancer therapy. Integrin αvß3 is a marker that is specifically and preferentially overexpressed on multiple tumor types and on angiogenic tumor neovasculature. Active targeting of NSs to integrin αvß3 offers the potential to increase accumulation preferentially in tumors and thereby enhance therapy efficacy. METHODS: Enzyme-linked immunosorbent assay (ELISA) and cell binding assay were used to study the in vitro binding affinities of the targeted nanoconjugate NS-RGDfK. In vivo biodistribution and tumor specificity were analyzed using 64Cu-radiolabeled untargeted and targeted NSs in live nude rats bearing head and neck squamous cell carcinoma (HNSCC) xenografts. The potential thermal therapy applications of NS-RGDfK were evaluated by subablative thermal therapy of tumor xenografts using untargeted and targeted NSs. RESULTS: ELISA and cell binding assay confirmed the binding affinity of NS-RGDfK to integrin αvß3. Positron emission tomography/computed tomography imaging suggested that tumor targeting is improved by conjugation of NSs to cyclo(RGDfK) and peaks at ~20 hours postinjection. In the subablative thermal therapy study, greater biological effectiveness of targeted NSs was implied by the greater degree of tumor necrosis. CONCLUSION: The results presented in this paper set the stage for the advancement of integrin αvß3-targeted NSs as therapeutic nanoconstructs for effective cancer therapy.


Subject(s)
Carcinoma, Squamous Cell/diagnostic imaging , Carcinoma, Squamous Cell/radiotherapy , Integrin alphaVbeta3 , Nanoconjugates/chemistry , Peptides, Cyclic/pharmacology , Animals , Carcinoma, Squamous Cell/blood supply , Cell Line, Tumor , Copper Radioisotopes , Enzyme-Linked Immunosorbent Assay , Gold/chemistry , Hot Temperature , Humans , Hyperthermia, Induced/methods , Integrin alphaVbeta3/antagonists & inhibitors , Integrin alphaVbeta3/metabolism , Laser Therapy , Mice , Mice, Nude , Models, Animal , Nanoshells/chemistry , Protein Binding , Rats , Rats, Nude , Tissue Distribution , Tomography, Emission-Computed/methods , Transplantation, Heterologous
6.
Nano Lett ; 8(5): 1492-500, 2008 May.
Article in English | MEDLINE | ID: mdl-18412402

ABSTRACT

We report noninvasive modulation of in vivo tumor radiation response using gold nanoshells. Mild-temperature hyperthermia generated by near-infrared illumination of gold nanoshell-laden tumors, noninvasively quantified by magnetic resonance temperature imaging, causes an early increase in tumor perfusion that reduces the hypoxic fraction of tumors. A subsequent radiation dose induces vascular disruption with extensive tumor necrosis. Gold nanoshells sequestered in the perivascular space mediate these two tumor vasculature-focused effects to improve radiation response of tumors. This novel integrated antihypoxic and localized vascular disrupting therapy can potentially be combined with other conventional antitumor therapies.


Subject(s)
Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Gold/therapeutic use , Hyperthermia, Induced/methods , Nanostructures/therapeutic use , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/therapy , Animals , Cell Line, Tumor , Humans , Light , Male , Mice , Mice, Nude , Nanomedicine/methods , Radiation Dosage , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL