Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Front Oncol ; 12: 1018767, 2022.
Article in English | MEDLINE | ID: mdl-36387154

ABSTRACT

The majority of colorectal cancers (CRCs) are microsatellite stable (MSS) and resistant to immunotherapy. The current study explores the possibility of using oncolytic reovirus to sensitize MSS CRC to immune checkpoint inhibition. While reovirus reduced metabolic activity among KRAS Mut cells, microarray/computational analysis revealed microsatellite status-oriented activation of immune-response pathways. Reovirus plus anti-PD-1 treatment increased cell death among MSS cells ex vivo. Reduced tumorigenicity and proliferative index, and increased apoptosis were evident among CT26 [MSS, KRAS Mut], but not in MC38 [microsatellite unstable/MSI, KRAS Wt] syngeneic mouse models under combinatorial treatment. PD-L1-PD-1 signaling axis were differentially altered among CT26/MC38 models. Combinatorial treatment activated the innate immune system, pattern recognition receptors, and antigen presentation markers. Furthermore, we observed the reduction of immunosuppressive macrophages and expansion of effector T cell subsets, as well as reduction in T cell exhaustion. The current investigation sheds light on the immunological mechanisms of the reovirus-anti-PD-1 combination to reduce the growth of MSS CRC.

2.
Clin Cancer Res ; 27(3): 865-876, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33168658

ABSTRACT

PURPOSE: To explore the effects of pelareorep on autophagy in multiple models of colorectal cancer, including patient-derived peripheral blood mononuclear cells (PBMCs). EXPERIMENTAL DESIGN: HCT116 [KRAS mutant (mut)] and Hke3 [KRAS wild-type (WT)] cells were treated with pelareorep (multiplicity of infection, 5) and harvested at 6 and 9 hours. LC3 A/B expression was determined by immunofluorescence and flow cytometry; five autophagic proteins were analyzed by Western blotting. The expression of 88 autophagy genes was determined by qRT-PCR. Syngeneic mouse models, CT26/Balb-C (KRAS mut) and MC38/C57B6 (KRAS WT), were developed and treated with pelareorep (10 × 106 plaque-forming unit/day) intraperitoneally. Protein and RNA were extracted from harvested tumor tissues. PBMCs from five experimental and three control patients were sampled at 0 (pre) and 48 hours, and on days 8 and 15. The gene expression normalized to "pre" was determined using 2-ΔΔC t method. RESULTS: Pelareorep induced significant upregulation of LC3 A/B in HCT116 as compared with Hke3 cells by immunofluorescence (3.24 × and 8.67 ×), flow cytometry (2.37 × and 2.58 ×), and autophagosome formation (2.02 × and 1.57 ×), at 6 and 9 hours, respectively; all P < 0.05. Western blot analysis showed an increase in LC3 A/B (2.38 × and 6.82 ×) and Beclin1 (1.17 × and 1.24 ×) at 6 and 9 hours, ATG5 (2.4 ×) and P-62 (1.52 ×) at 6 hours, and VPS-34 (1.39 ×) at 9 hours (all P < 0.05). Induction of 13 transcripts in cell lines (>4 ×; 6 and 9 hours; P < 0.05), 12 transcripts in CT26 (qRT-PCR), and 14 transcripts in human PBMCs (P < 0.05) was observed. LC3 A/B, RICTOR, and RASD1 expression was upregulated in all three model systems. CONCLUSIONS: Pelareorep hijacks host autophagic machinery in KRAS-mut conditions to augment its propagation and preferential oncolysis of the cancer cells.


Subject(s)
Autophagy/immunology , Colorectal Neoplasms/therapy , Oncolytic Virotherapy/methods , Oncolytic Viruses/immunology , Proto-Oncogene Proteins p21(ras)/genetics , Adult , Animals , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Autophagy/genetics , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Combined Modality Therapy , Disease Models, Animal , Female , Fluorouracil/administration & dosage , Gene Expression Regulation, Neoplastic/immunology , HCT116 Cells , Humans , Infusions, Intravenous , Injections, Intralesional , Leucovorin/administration & dosage , Male , Mice , Microtubule-Associated Proteins/genetics , Middle Aged , Rapamycin-Insensitive Companion of mTOR Protein/genetics , Up-Regulation , ras Proteins/genetics
3.
BMC Cancer ; 20(1): 569, 2020 Jun 18.
Article in English | MEDLINE | ID: mdl-32552875

ABSTRACT

BACKGROUND: KRAS mutations are prevalent in 40-45% of patients with colorectal cancer (CRC) and targeting this gene has remained elusive. Viruses are well known immune sensitizing agents. The therapeutic efficacy of oncolytic reovirus in combination with chemotherapy is examined in a phase 1 study of metastatic CRC. This study evaluates the nature of immune response by determining the cytokine expression pattern in peripheral circulation along with the distribution of antigen presenting cells (APCs) and activated T lymphocytes. Further the study evaluates the alterations in exosomal and cellular microRNA levels along with the effect of reovirus on leukocyte transcriptome. METHODS: Reovirus was administered as a 60-min intravenous infusion for 5 consecutive days every 28 days, at a tissue culture infective dose (TCID50) of 3 × 1010. Peripheral blood mononuclear cells (PBMC) were isolated from whole blood prior to reovirus administration and post-reovirus on days 2, 8, and 15. The expression profile of 25 cytokines in plasma was assessed (post PBMC isolation) on an EMD Millipore multiplex Luminex platform. Exosome and cellular levels of miR-29a-3p was determined in pre and post reovirus treated samples. Peripheral blood mononuclear cells were stained with fluorophore labelled antibodies against CD4, CD8, CD56, CD70, and CD123, fixed and evaluated by flow cytometry. The expression of granzyme B was determined on core biopsy of one patient. Finally, Clariom D Assay was used to determine the expression of 847 immune-related genes when compared to pre reovirus treatment by RNA sequencing analysis. A change was considered if the expression level either doubled or halved and the significance was determined at a p value of 0.001. RESULTS: Cytokine assay indicated upregulation at day 8 for IL-12p40 (2.95; p = 0.05); day 15 for GM-CSF (3.56; p = 0.009), IFN-y (1.86; p = 0.0004) and IL-12p70 (2.42; p = 0.02). An overall reduction in IL-8, VEGF and RANTES/CCL5 was observed over the 15-day period. Statistically significant reductions were observed at Day 15 for IL-8 (0.457-fold, 53.3% reduction; p = 0.03) and RANTES/CC5 (0.524-fold, 47.6% reduction; p = 0.003). An overall increase in IL-6 was observed, with statistical significance at day 8 (1.98- fold; 98% increase, p = 0.00007). APCs were stimulated within 48 h and activated (CD8+ CD70+) T cells within 168 h as determine by flow cytometry. Sustained reductions in exosomal and cellular levels of miR-29a-3p (a microRNA upregulated in CRC and associated with decreased expression of the tumor suppressor WWOX gene) was documented. Reovirus administration further resulted in increases in KRAS (33x), IFNAR1 (20x), STAT3(5x), and TAP1 (4x) genes after 2 days; FGCR2A (23x) and CD244 (3x) after 8 days; KLRD1 (14x), TAP1 (2x) and CD244(2x) after 15 days. Reductions (> 0.5x) were observed in VEGFA (2x) after 2 days; CXCR2 (2x), ITGAM (3x) after 15 days. CONCLUSIONS: Reovirus has profound immunomodulatory properties that span the genomic, protein and immune cell distribution levels. This is the first study with reovirus in cancer patients that demonstrates these multi- layered effects, demonstrating how reovirus can function as an immune stimulant (augmenting the efficacy of immuno-chemo-therapeutic drugs), and an oncolytic agent. Reovirus thus functions bimodally as an oncolytic agent causing lysis of tumor cells, and facilitator of immune-mediated recognition and destruction of tumor cells.


Subject(s)
Colorectal Neoplasms/therapy , Cytokines/metabolism , Gene Expression Regulation, Neoplastic/immunology , Oncolytic Virotherapy/methods , Oncolytic Viruses/immunology , Adult , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Bevacizumab/administration & dosage , Bevacizumab/adverse effects , Biopsy , Camptothecin/administration & dosage , Camptothecin/adverse effects , Camptothecin/analogs & derivatives , Colon/immunology , Colon/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Combined Modality Therapy/adverse effects , Combined Modality Therapy/methods , Female , Fluorouracil/administration & dosage , Fluorouracil/adverse effects , Gene Expression Profiling , Humans , Infusions, Intravenous , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Leucovorin/administration & dosage , Leucovorin/adverse effects , Lymphocyte Activation/immunology , Male , Middle Aged , Mutation , Oncolytic Virotherapy/adverse effects , Proto-Oncogene Proteins p21(ras)/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Treatment Outcome
4.
Int J Mol Sci ; 20(22)2019 Nov 13.
Article in English | MEDLINE | ID: mdl-31766149

ABSTRACT

Patients with KRAS mutated colorectal cancer (CRC) represent a cohort with unmet medical needs, with limited options of FDA-approved therapies. Representing 40-45% of all CRC patients, they are considered ineligible to receive anti-EGFR monoclonal antibodies that have added a significant therapeutic benefit for KRAS wild type CRC patients. Although several mouse models of CRC have been developed during the past decade, one genetically resembling the KRAS mutated CRC is yet to be established. In this study C57 BL/6 mice with truncated adenomatous polyposis coli (APC) floxed allele was crossed with heterozygous KRAS floxed outbred mice to generate an APCf/f KRAS+/f mouse colony. In another set of breeding, APC floxed mice were crossed with CDX2-Cre-ERT2 mice and selected for APCf/f CDX2-Cre-ERT2 after the second round of inbreeding. The final model of the disease was generated by the cross of the two parental colonies and viable APC f/f KRAS +/f CDX2-Cre-ERT2 (KPC: APC) were genotyped and characterized. The model animals were tamoxifen (TAM) induced to generate tumors. Micro-positron emission tomography (PET) scan was used to detect and measure tumor volume and standard uptake value (SUV). Hematoxylin and eosin (H&E) staining was performed to establish neoplasm and immunohistochemistry (IHC) was performed to determine histological similarities with human FFPE biopsies. The MSI/microsatellite stable (MSS) status was determined. Finally, the tumors were extensively characterized at the molecular level to establish similarities with human CRC tumors. The model KPC: APC animals are conditional mutants that developed colonic tumors upon induction with tamoxifen in a dose-dependent manner. The tumors were confirmed to be malignant within four weeks of induction by H&E staining and higher radioactive [18F] fluoro-2-deoxyglucose (FDG) uptake (SUV) in micro-PET scan. Furthermore, the tumors histologically and molecularly resembled human colorectal carcinoma. Post tumor generation, the KPC: APC animals died of cachexia and rectal bleeding. Implications: This model is an excellent preclinical platform to molecularly characterize the KRAS mutated colorectal tumors and discern appropriate therapeutic strategies to improve disease management and overall survival.


Subject(s)
Colorectal Neoplasms/genetics , Mutation , Proto-Oncogene Proteins p21(ras)/genetics , Adenomatous Polyposis Coli Protein/genetics , Animals , CDX2 Transcription Factor/genetics , Colorectal Neoplasms/pathology , Disease Models, Animal , Mice , Mice, Inbred C57BL
5.
Oncotarget ; 10(53): 5510-5522, 2019 Sep 17.
Article in English | MEDLINE | ID: mdl-31565185

ABSTRACT

BACKGROUND: ERCC1, a component of nucleotide excision repair pathway, is known to repair DNA breaks induced by platinum drugs. We sought to ascertain if ERCC1 expression dynamics and a single nucleotide polymorphism (SNP) rs11615 are biomarkers of sensitivity to oxaliplatin therapy in patients with colorectal cancer (CRC). METHODS: Western blot and qPCR for ERCC1 expression was performed from PBMCs isolated from patients receiving oxaliplatin-based therapy at specified timepoints. DNA was also isolated from 59 biorepository specimens for SNP analysis. Clinical benefit was determined using progression free survival (PFS) for metastatic CRC. RESULTS: ERCC1 was induced in PBMC in response to oxaliplatin in 13/25 patients with mCRC (52%). Median PFS with ERCC1 induction was 190d compared to 237d in non-induced patients (HR 2.35, CI 1.005-5.479; p=0.0182). ERCC1 rs11615 SNP analysis revealed that 43.3% harbored C/C, 41.2%-T/C and 15.5%-T/T genotype. Median PFS was significantly lower with C/C or T/C (211 and 196d) compared to T/T (590d; p=0.0310). CONCLUSIONS: ERCC1 was induced in a sub-population of patients undergoing oxaliplatin treatment, which was associated with poorer outcome, suggesting this could serve as a marker of oxaliplatin response. C/C or C/T genotype in ERCC1 rs11615 locus decreased benefit from oxaliplatin.

6.
NPJ Precis Oncol ; 3: 4, 2019.
Article in English | MEDLINE | ID: mdl-30729176

ABSTRACT

Cancer is an important global issue with increasing incidence and mortality, placing a substantial burden on the healthcare system. Colorectal cancer is the third most common cancer diagnosed among men and women in US. It is estimated that in 2018 there will be 319,160 new diagnosis and 160,820 deaths related to cancer of the digestive system including both genders in the United States alone. Considering limited success of chemotherapy, radiotherapy, and surgery in treatment of these cancer patients, new therapeutic avenues are under constant investigation. Therapy options have consistently moved away from typical cytotoxic chemotherapy where patients with a given type and stage of the disease were treated similarly, to an individualized approach where a tumor is defined by its specific tissue characteristics /epigenetic profile, protein expression and genetic mutations. This review takes a deeper look at the immune-biological aspects of cancers in the gastrointestinal tract (entire digestive tract extending from esophagus/stomach to rectum, including pancreatico-biliary apparatus) and discusses the different treatment modalities that are available or being developed to target the immune system for better disease outcome.

7.
Invest New Drugs ; 37(5): 948-960, 2019 10.
Article in English | MEDLINE | ID: mdl-30612311

ABSTRACT

Intended to explore synthetic lethality and develop better combinatorial regimens, we screened colorectal cancer (CRC) cells using poly ADP-ribose (PAR) polymerase (PARP) inhibitors and cytotoxic agents. We studied four PARP inhibitors and three DNA-damaging agents, and their combinations using sulforhodamine B assay. Rucaparib demonstrated the greatest synergy with irinotecan, followed by olaparib and PJ34. Rucaparib and irinotecan was further subjected to detailed examination to determine combination index (CI) and underlying mechanism of action. Effectiveness and sequence dependence of this combination were assessed in microsatellite stable (MSS) and unstable (MSI) CRC and HCT116 isogenic cell lines. The degree of cell cycle arrest and apoptosis was determined by FACS. In vivo studies were performed to confirm efficacy of this combination. PAR levels in MSI and PARP expression in MSI and MSS cell lines were diminished upon combinatorial treatment. HCT116 isogenic cells revealed the importance of p21, p53 and PTEN in exerting synergy. In MSI cells, administration of rucaparib prior to irinotecan enhanced cytotoxicity compared to other strategies explored. FACS revealed S-phase arrest and increased late-stage apoptosis in MSS, and G2-M arrest and total and early-stage apoptosis in MSI cells. In in vivo murine xenograft models, a significant reduction in tumor volume and expression of Ki67, pancytokeratin and RPS6KB1, and increase in expression of caspase 3 were observed with the combination. In conclusion, among the various combinations studied, rucaparib plus irinotecan was the most synergistic one. Alterations in cell cycle arrest and apoptosis were dependent on MSI status in CRC cells.


Subject(s)
Colorectal Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Indoles/pharmacology , Irinotecan/pharmacology , Animals , Apoptosis , Cell Cycle , Cell Proliferation , Colorectal Neoplasms/pathology , Drug Therapy, Combination , Humans , Mice , Mice, Nude , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Topoisomerase I Inhibitors/pharmacology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Genes Cancer ; 8(5-6): 550-558, 2017 May.
Article in English | MEDLINE | ID: mdl-28740573

ABSTRACT

Length of the telomere (TL), a structure at the tip of chromosome that protects and ensures stability, is determined by multi-protein complexes such as telosome/shelterin and telomerase. Earlier studies from our laboratory show that longer TL has potential to be positive predictive biomarker of clinical outcome to anti-epidermal growth factor receptor (EGFR) monoclonal antibody therapy in patients with KRAS WT metastatic colorectal cancer. Although there is extensive literature suggesting the role of shelterin and telomerase, not much literature exists that describes the role of EGFR and KRAS pathway in regulating TL. This detailed review focuses on an insight into various components, including proteins, enzymes and transcription factors, interlinking between EGFR pathways and telomerase that regulate TL.

9.
Oncotarget ; 8(21): 35138-35153, 2017 May 23.
Article in English | MEDLINE | ID: mdl-28422714

ABSTRACT

New therapeutic interventions are essential for improved management of patients with metastatic colorectal cancer (mCRC). This is especially critical for those patients whose tumors harbor a mutation in the KRAS oncogene (40-45% of all patients). This patient cohort is excluded from receiving anti-EGFR monoclonal antibodies that have added a significant therapeutic benefit for KRAS wild type CRC patients. Reovirus, a double stranded (ds) RNA virus is in clinical development for patients with chemotherapy refractory KRAS mutated tumors. Toll Like Receptor (TLR) 3, a member of the toll like receptor family of the host innate immune system is the pattern recognition motif for dsRNA pathogens. Using TLR3 expressing commercial HEK-BlueTM-hTLR3 cells we confirm that TLR3 is the host pattern recognition motif responsible for the detection of reovirus. Further, our investigation with KRAS mutated HCT116 cell line showed that effective expression of host TLR3 dampens the infection potential of reovirus by mounting a robust innate immune response. Down regulation of TLR3 expression with siRNA improves the anticancer activity of reovirus. In vivo experiments using human CRC cells derived xenografts in athymic mice further demonstrate the beneficial effects of TLR3 knock down by improving tumor response rates to reovirus. Strategies to mitigate the TLR3 response pathway can be utilized as a tool towards improved reovirus efficacy to specifically target the dissemination of KRAS mutated CRC.


Subject(s)
Colorectal Neoplasms/therapy , Orthoreovirus, Mammalian/physiology , Proto-Oncogene Proteins p21(ras)/genetics , Toll-Like Receptor 3/genetics , Animals , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Gene Knockdown Techniques , HCT116 Cells , Humans , Mice , Oncolytic Virotherapy , Orthoreovirus, Mammalian/immunology , Signal Transduction , Xenograft Model Antitumor Assays
10.
Oncotarget ; 5(9): 2807-19, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24798549

ABSTRACT

Reovirus is a double stranded RNA virus, with an intrinsic preference for replication in KRAS mutant cells. As 45% of human colorectal cancers (CRC) harbor KRAS mutations, we sought to investigate its efficacy in KRAS mutant CRC cells, and examine its impact in combination with the topoisimerase-1 inhibitor, irinotecan. Reovirus efficacy was examined in the KRAS mutant HCT116, and the isogenic KRAS WT Hke3 cell line, and in the non-malignant rat intestinal epithelial cell line. Apoptosis was determined by flow cytometry and TUNEL staining. Combination treatment with reovirus and irintoecan was investigated in 15 CRC cell lines, including the HCT116 p21 isogenic cell lines. Reovirus preferentially induced apoptosis in KRAS mutant HCT116 cells compared to its isogenic KRAS WT derivative, and in KRAS mutant IEC cells. Reovirus showed a greater degree of caspase 3 activation with PARP 1 cleavage, and preferential inhibition of p21 protein expression in KRAS mutant cells. Reovirus synergistically induced growth inhibition when combined with irinotecan. This synergy was lost upon p21 gene knock out. Reovirus preferentially induces apoptosis in KRAS mutant colon cancer cells. Reovirus and irinotecan combination therapy is synergistic, p21 mediated, and represents a novel potential treatment for patients with CRC.


Subject(s)
Antineoplastic Agents/therapeutic use , Camptothecin/analogs & derivatives , Colorectal Neoplasms/therapy , Mutation/genetics , Oncolytic Virotherapy , Proto-Oncogene Proteins/genetics , Reoviridae/physiology , ras Proteins/genetics , Animals , Apoptosis , Blotting, Western , Camptothecin/therapeutic use , Cell Cycle , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Combined Modality Therapy , Drug Synergism , Humans , Irinotecan , Microscopy, Electron, Scanning , Proto-Oncogene Proteins p21(ras) , Rats , Tumor Cells, Cultured
11.
Fundam Clin Pharmacol ; 26(2): 259-70, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21323996

ABSTRACT

Current study aims to evaluate the efficacy of baicalein (BE), a naturally occurring bioactive flavanoid (5,6,7-trihydroxy-flavone), at a dose of 12 mg/kg body wt in Swiss albino mice exposed to tobacco-specific carcinogen benzo(a)pyrene [B(a)P] (50 mg/kg body wt) for its ability to mitigate pulmonary adenoma formation and growth. Under coarse observation, B(a)P-administered mice, after 16 weeks, developed macroscopically detectable tumors, whereas oral treatment with BE to the lung cancer-induced mice significantly reduced tumor incidence in 16-week pre- and posttreated groups. A detailed histopathological examination of lung was conducted to determine the degree of cancer progression. Incidence of anaplasia, hyperplasia, dysplasia, severe dysplasia, and adenocarcinoma were evident in carcinogen-administrated group in 6, 10, 12, 14, and 16th weeks, respectively, whereas these anomalies were effectively reduced after pre- and posttreatment with BE. In the pretreatment group, BE significantly arrested tumor multiplicity by approximately 65% and tumor load by approximately 88%, while in the posttreatment, the compound significantly reduced the tumor multiplicity by approximately 48% and tumor load by approximately 61%. Further analysis of serum tumor markers like carcinoembryonic antigen, CK 19 fragments (CYFRA 21-1), and tissue marker enzymes like aryl hydrocarbon hydroxylase, adenosine deaminase, γ-glutamyl transpeptidase, 5'-nucleotidase, and lactate dehydrogenase in serum and lung homogenate was carried out to substantiate the anticarcinogenic effect of BE. The overall data from our experiments suggested that BE significantly inhibited pulmonary adenoma formation and growth, thus revealing its potent antitumorigenic effect.


Subject(s)
Adenocarcinoma/prevention & control , Anticarcinogenic Agents/pharmacology , Flavanones/pharmacology , Lung Neoplasms/prevention & control , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Administration, Oral , Animals , Anticarcinogenic Agents/administration & dosage , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/pharmacology , Benzo(a)pyrene/toxicity , Biomarkers, Tumor/blood , Disease Progression , Flavanones/administration & dosage , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Male , Mice , Time Factors
12.
Mol Cell Biochem ; 313(1-2): 53-61, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18373278

ABSTRACT

Silymarin is a naturally available bioflavonoid and is a strong antioxidant with a capacity to inhibit the formation of tumors in several cancer models. In the present study, we investigated whether dietary supplementation of silymarin has any role in lipid components, lipid-metabolizing enzymes, free fatty acid profile, and expression of cyclooxygenase-2 (COX-2) in N-nitrosodiethylamine (NDEA)-induced hepatocellular carcinoma in rats. NDEA-induced rats showed severe hyperlipidemia along with upregulated expression of COX-2 as revealed by western blotting and immunohistochemistry. Dietary silymarin supplementation attenuated this hyperlipidemia and downregulated the expression of COX-2. Thus we conclude that compounds like silymarin with potent hypolipidemic effect are strong candidates as chemopreventive agents for the treatment of liver cancer.


Subject(s)
Carcinoma, Hepatocellular/complications , Cyclooxygenase 2/metabolism , Down-Regulation/drug effects , Hyperlipidemias/complications , Hyperlipidemias/enzymology , Liver Neoplasms/complications , Silymarin/pharmacology , Animals , Arachidonic Acid/metabolism , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/enzymology , Cholesterol, HDL/blood , Diethylnitrosamine , Hydroxymethylglutaryl CoA Reductases/metabolism , Hyperlipidemias/drug therapy , Immunoblotting , Lipid Metabolism/drug effects , Liver/drug effects , Liver/enzymology , Liver/pathology , Liver Neoplasms/drug therapy , Liver Neoplasms/enzymology , Male , Phytotherapy , Rats , Rats, Wistar , Silymarin/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...