phenanthrenes has been researched along with Carcinoma--Pancreatic-Ductal* in 8 studies
8 other study(ies) available for phenanthrenes and Carcinoma--Pancreatic-Ductal
Article | Year |
---|---|
Sequential delivery of nanoformulated α-mangostin and triptolide overcomes permeation obstacles and improves therapeutic effects in pancreatic cancer.
Pancreatic ductal adenocarcinoma (PDAC) is a devastating disease exhibiting the poorest prognosis among solid tumors. The efficacy of conventional therapies has been hindered largely due to the insufficient chemotherapeutic delivery to the dense desmoplastic tumor stroma, and the extremely high or toxic dose needed for chemotherapy. Traditional Chinese Medicine (TCM) contains effective components that can effectively regulate tumor microenvironment and kill tumor cells, providing promising alternatives to PDAC chemotherapy. In this study, two active drug monomers of TCM were screened out and a sequentially targeting delivery regimen was developed to realize the optimized combinational therapy. Transforming growth factor-β (TGF-β) plays an indispensable role in promoting cancer-associated fibroblasts (CAFs) activation and proliferation, and CAFs have caused major physical barriers for chemotherapeutic drug delivery. Herein, CAFs-targeting biodegradable polymer nanoparticle (CRE-NP(α-M)) coated with CREKA peptide and loaded with TCM α-mangostin (α-M) was developed to modulate tumor microenvironment by interfering of TGF-β/Smad signaling pathway. Low pH-triggered micelle modified with CRPPR peptide and loaded with another TCM triptolide was constructed to increase the therapeutic effect of triptolide at the tumor sites and reduced its damage to main organs. As expected, CRE-NP(α-M) effectively inactived CAFs, reduced extracellular matrix production, promoted tumor vascular normalization and enhanced blood perfusion at the tumor site. The sequentially targeting drug delivery regimen, CRP-MC(Trip) following CRE-NP(α-M) pretreatment, exhibited strong tumor growth inhibition effect in the orthotopic tumor model. Hence, sequentially targeting delivery of nanoformulated TCM offers an efficient approach to overcome the permeation obstacles and improve the effect of chemotherapy on PDAC, and provides a novel option to treat desmoplastic tumors. Topics: Carcinoma, Pancreatic Ductal; Diterpenes; Epoxy Compounds; Humans; Pancreatic Neoplasms; Phenanthrenes; Tumor Microenvironment; Xanthones | 2020 |
Inactivation of Cancer-Associated-Fibroblasts Disrupts Oncogenic Signaling in Pancreatic Cancer Cells and Promotes Its Regression.
Resident fibroblasts that contact tumor epithelial cells (TEC) can become irreversibly activated as cancer-associated-fibroblasts (CAF) that stimulate oncogenic signaling in TEC. In this study, we evaluated the cross-talk between CAF and TEC isolated from tumors generated in a mouse model of KRAS/mut p53-induced pancreatic cancer (KPC mice). Transcriptomic profiling conducted after treatment with the anticancer compound Minnelide revealed deregulation of the TGFβ signaling pathway in CAF, resulting in an apparent reversal of their activated state to a quiescent, nonproliferative state. TEC exposed to media conditioned by drug-treated CAFs exhibited a decrease in oncogenic signaling, as manifested by downregulation of the transcription factor Sp1. This inhibition was rescued by treating TEC with TGFβ. Given promising early clinical studies with Minnelide, our findings suggest that approaches to inactivate CAF and prevent tumor-stroma cross-talk may offer a viable strategy to treat pancreatic cancer. Topics: Animals; Apoptosis; Cancer-Associated Fibroblasts; Carcinogenesis; Carcinoma, Pancreatic Ductal; Cell Proliferation; Disease Models, Animal; Diterpenes; Epithelial Cells; Epoxy Compounds; Gene Expression Profiling; Gene Expression Regulation, Neoplastic; Humans; Mice; Mice, Inbred C57BL; Mutation; Organophosphates; Pancreatic Neoplasms; Phenanthrenes; Proto-Oncogene Proteins p21(ras); Signal Transduction; Transforming Growth Factor beta; Tumor Cells, Cultured; Tumor Suppressor Protein p53 | 2018 |
Screening of Conditionally Reprogrammed Patient-Derived Carcinoma Cells Identifies ERCC3-MYC Interactions as a Target in Pancreatic Cancer.
Even when diagnosed prior to metastasis, pancreatic ductal adenocarcinoma (PDAC) is a devastating malignancy with almost 90% lethality, emphasizing the need for new therapies optimally targeting the tumors of individual patients.. We first developed a panel of new physiologic models for study of PDAC, expanding surgical PDAC tumor samples in culture using short-term culture and conditional reprogramming with the Rho kinase inhibitor Y-27632, and creating matched patient-derived xenografts (PDX). These were evaluated for sensitivity to a large panel of clinical agents, and promising leads further evaluated mechanistically.. Only a small minority of tested agents was cytotoxic in minimally passaged PDAC cultures in vitro Drugs interfering with protein turnover and transcription were among most cytotoxic. Among transcriptional repressors, triptolide, a covalent inhibitor of ERCC3, was most consistently effective in vitro and in vivo causing prolonged complete regression in multiple PDX models resistant to standard PDAC therapies. Importantly, triptolide showed superior activity in MYC-amplified PDX models and elicited rapid and profound depletion of the oncoprotein MYC, a transcriptional regulator. Expression of ERCC3 and MYC was interdependent in PDACs, and acquired resistance to triptolide depended on elevated ERCC3 and MYC expression. The Cancer Genome Atlas analysis indicates ERCC3 expression predicts poor prognosis, particularly in CDKN2A-null, highly proliferative tumors.. This provides initial preclinical evidence for an essential role of MYC-ERCC3 interactions in PDAC, and suggests a new mechanistic approach for disruption of critical survival signaling in MYC-dependent cancers. Clin Cancer Res; 22(24); 6153-63. ©2016 AACR. Topics: Amides; Animals; Carcinoma, Pancreatic Ductal; Cell Line; Cell Line, Tumor; Cell Proliferation; Diterpenes; DNA Helicases; DNA-Binding Proteins; Epoxy Compounds; Gene Expression Regulation, Neoplastic; Heterografts; Humans; Mice; Mice, SCID; NIH 3T3 Cells; Pancreatic Neoplasms; Phenanthrenes; Proto-Oncogene Proteins c-myc; Pyridines; rho-Associated Kinases; Signal Transduction; Transcription, Genetic | 2016 |
Minnelide effectively eliminates CD133(+) side population in pancreatic cancer.
Pancreatic Ductal Adenocarcinoma (PDAC) is a devastating disease hallmarked by limited patient survival. Resistance to chemotherapy, a major cause of treatment failure in PDAC patients, is often attributed to Cancer Stem Cells (CSCs). Pancreatic CSCs are a small subset of quiescent cells within a tumor represented by surface markers like CD133. These cells are responsible not only for tumor recurrence, but also poor prognosis based on their "stem-like" characteristics. At present, conventional therapy is directed towards rapidly dividing PDAC cells and thus fails to target the CSC population.. MIA PaCa-2, S2-013 and AsPC-1 were treated with 12.5 nM triptolide (12 T cells) for 7 days. The surviving cells were recovered briefly in drug-free growth media and then transferred to Cancer Stem cell Media (CSM). As a control, untreated cells were also transferred to CSM media (CSM). The 12 T and CSM cells were tested for stemness properties using RNA and protein markers. Low numbers of CSM and 12 T cells were implanted subcutaneously in athymic nude mice to study their tumorigenic potential. 12 T and CSM cells were sorted for CD133 expression and assayed for their colony forming ability and sphere forming ability. Invasiveness of 12 T cells, CSM and MIA PaCa-2 were compared using Boyden chamber assays.. Treated 12 T cells displayed increased expression of the surface marker CD133 and the drug transporter ABCG2 compared to untreated cells (CSM cells). Both 12 T and CSM cells formed subcutaneous tumors in mice confirming their tumor-initiating properties. When tested for invasion, 12 T cells had increased invasiveness compared to CSM cells. CD133(+) cells in both CSM and 12 T showed greater colony and sphere forming ability compared to CD133(-) cells from each group. Consistent with these data, when injected subcutaneously in mice, CD133(-) cells from CSM or 12 T did not form any tumors whereas CD133(+) cells from both groups showed tumor formation at a very low cell number. Despite pre-exposure to triptolide in 12 T CD133(+) cells, treatment of tumors formed by these cells with Minnelide, a triptolide pro-drug, showed significant tumor regression.. Our results indicated that triptolide enhanced and enriched the "stemness" in the PDAC cell lines at a low dose of 12.5 nM, but also resulted in the regression of tumors derived from these cells. Topics: AC133 Antigen; Antigens, CD; Antineoplastic Agents; Carcinoma, Pancreatic Ductal; Cell Line, Tumor; Cell Survival; Diterpenes; Drug Resistance, Neoplasm; Drug Screening Assays, Antitumor; Epoxy Compounds; Glycoproteins; Humans; Neoplastic Stem Cells; Organophosphates; Pancreatic Neoplasms; Peptides; Phenanthrenes; Side-Population Cells | 2015 |
Triptolide reverses hypoxia-induced epithelial-mesenchymal transition and stem-like features in pancreatic cancer by NF-κB downregulation.
Pancreatic ductal adenocarcinoma (PDA) is one of the most lethal malignancies characterized by an intense tumor stroma with hypoperfused regions, a significant inflammatory response and pronounced therapy resistance. New therapeutic agents are urgently needed. The plant-derived agent triptolide also known as "thunder god vine" has a long history in traditional Chinese medicine for treatment of rheumatoid arthritis and cancer and is now in a clinical phase II trial for establishing the efficacy against a placebo. The authors mimicked the situation in patient tumors by induction of hypoxia in experimental models of pancreatic cancer stem cells (CSCs) and evaluated the therapeutic effect of triptolide. Hypoxia led to induction of colony and spheroid formation, aldehyde dehydrogenase 1 (ALDH1) and NF-κB activity, migratory potential and a switch in morphology to a fibroblastoid phenotype, as well as stem cell- and epithelial-mesenchymal transition-associated protein expression. Triptolide efficiently inhibited hypoxia-induced transcriptional signaling and downregulated epithelial-mesenchymal transition (EMT) and CSC features in established highly malignant cell lines, whereas sensitive cancer cells or nonmalignant cells were less affected. In vivo triptolide inhibited tumor take and tumor growth. In primary CSCs isolated from patient tumors, triptolide downregulated markers of CSCs, proliferation and mesenchymal cells along with upregulation of markers for apoptosis and epithelial cells. This study is the first to show that triptolide reverses EMT and CSC characteristics and therefore may be superior to current chemotherapeutics for treatment of PDA. Topics: Aldehyde Dehydrogenase 1 Family; Animals; Antineoplastic Agents, Alkylating; Biomarkers, Tumor; Blotting, Western; Carcinoma, Pancreatic Ductal; Cell Hypoxia; Cell Line, Tumor; Cell Movement; Chick Embryo; Diterpenes; Down-Regulation; Epithelial-Mesenchymal Transition; Epoxy Compounds; Humans; Isoenzymes; Mice; Mice, Inbred Strains; Mice, Nude; Neoplastic Stem Cells; NF-kappa B; Pancreatic Neoplasms; Phenanthrenes; Proto-Oncogene Proteins c-rel; Retinal Dehydrogenase; RNA Interference; Spheroids, Cellular; Xenograft Model Antitumor Assays | 2014 |
Establishment of hypoxia induction in an in vivo animal replacement model for experimental evaluation of pancreatic cancer.
Transplantation of tumor xenografts to fertilized chicken eggs is a promising animal replacement method, which has successfully been used for xenotransplantation of pancreatic ductal adenocarcinoma (PDA) cells. PDA is characterized by a pronounced tumor hypoxia, which mediates aggressive growth, therapy resistance and cancer stem cell (CSC) features. For in vivo experimental evaluation of hypoxia-targeting therapeutic strategies, the xenografting of tumors to chicken eggs combined with the induction of hypoxia is necessary. However, the chicken embryos do not survive the conventional method of hypoxia induction by a gas mixture of 1% O2, 5% CO2, 94% N2, not even when hypoxia is applied for only 30 min. Therefore, we employed chemical induction of hypoxia by the hypoxia mimetic agent cobalt chloride (CoCl2). Whereas CoCl2 did not further increase tumor growth, it mediated the induction of carbonic anhydrase IX (CAIX) in the tumor xenografts and led to enhanced expression of the human CSC markers CD133, Sox2 and CD44. Side-effects in chicken embryos were not observed as evaluated by H&E staining of embryo-derived liver sections and the determination of the embryo weight. These results suggest the successful induction of hypoxia in chicken eggs and xenografted tumors by CoCl2. For therapeutic intervention and as a control, we treated the eggs with the plant-derived anti-inflammatory agent triptolide, which recently showed promising effects toward hypoxia-induced tumor progression in experimental PDA. Triptolide abolished tumor growth and the CoCl2-induced hypoxic effects, without inducing obvious side-effects. Collectively, our data present a new in vivo animal replacement method for the successful induction of tumor hypoxia in PDA. Topics: Animals; Antineoplastic Agents, Alkylating; Biomarkers, Tumor; Carbonic Anhydrases; Carcinoma, Pancreatic Ductal; Cell Hypoxia; Chick Embryo; Cobalt; Diterpenes; Epoxy Compounds; Gene Expression Regulation, Neoplastic; Humans; Neoplasms, Experimental; Neoplastic Stem Cells; Pancreatic Neoplasms; Phenanthrenes | 2014 |
Triptolide induces the expression of miR-142-3p: a negative regulator of heat shock protein 70 and pancreatic cancer cell proliferation.
Pancreatic ductal adenocarcinoma (PDAC), one of the deadliest malignancies, is resistant to current chemotherapies. We previously showed that triptolide inhibits PDAC cell growth in vitro and blocks metastatic spread in vivo. Triptolide downregulates HSP70, a molecular chaperone upregulated in several tumor types. This study investigates the mechanism by which triptolide inhibits HSP70. Because microRNAs (miRNA) are becoming increasingly recognized as negative regulators of gene expression, we tested whether triptolide regulates HSP70 via miRNAs. Here, we show that triptolide as well as quercetin, but not gemcitabine, upregulated miR-142-3p in PDAC cells (MIA PaCa-2, Capan-1, and S2-013). Ectopic expression of miR-142-3p inhibited cell proliferation, measured by electric cell-substrate impedance sensing, and decreased HSP70 expression, measured by real-time PCR and immunoblotting, compared with controls. We showed that miR-142-3p directly binds to the 3'UTR of HSP70, and that this interaction is important as HSP70 overexpression rescued miR-142-3p-induced cell death. We found that miR-142-3p regulates HSP70 independently of heat shock factor 1. Furthermore, Minnelide, a water-soluble prodrug of triptolide, induced the expression of miR-142-3p in vivo. This is the first description of an miRNA-mediated mechanism of HSP70 regulation in cancer, making miR-142-3p an attractive target for PDAC therapeutic intervention. Topics: Animals; Antineoplastic Agents, Alkylating; Apoptosis; Carcinoma, Pancreatic Ductal; Cell Line, Tumor; Cell Proliferation; Diterpenes; Epoxy Compounds; Female; Gene Expression; HEK293 Cells; HSP70 Heat-Shock Proteins; Humans; Mice; Mice, SCID; MicroRNAs; Pancreatic Neoplasms; Phenanthrenes; Random Allocation; Transfection; Xenograft Model Antitumor Assays | 2013 |
Pancreas cancer meets the thunder god.
A new formulation of a natural product shows remarkable activity against pancreatic ductal adenocarcinoma across a number of preclinical model systems. These findings set the stage for a clinical trial. Topics: Animals; Antineoplastic Agents, Alkylating; Carcinoma, Pancreatic Ductal; Clinical Trials as Topic; Diterpenes; Drug Evaluation, Preclinical; Epoxy Compounds; Humans; Medical Oncology; Medicine, Chinese Traditional; Organophosphates; Pancreatic Neoplasms; Phenanthrenes; Research Design; Translational Research, Biomedical | 2012 |