Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Cancer Lett ; 394: 76-87, 2017 05 28.
Article in English | MEDLINE | ID: mdl-28254412

ABSTRACT

Bromodomain inhibitors (JQ1 and I-BET 762) are a new generation of selective, small molecule inhibitors that target BET (bromodomain and extra terminal) proteins. By impairing their ability to bind to acetylated lysines on histones, bromodomain inhibitors interfere with transcriptional initiation and elongation. BET proteins regulate several genes responsible for cell cycle, apoptosis and inflammation. In this study, JQ1 and I-BET 762 decreased c-Myc and p-Erk 1/2 protein levels and inhibited proliferation in pancreatic cancer cells. The tumor microenvironment is known to play an important role in pancreatic cancer, and these drugs suppressed the production of nitric oxide and a variety of inflammatory cytokines, including IL-6, CCL2, and GM-CSF, in both immune and pancreatic cancer cells in vitro. Notably, the bromodomain inhibitors also reduced protein levels of p-Erk 1/2 and p-STAT3 in mouse models of pancreatic cancer. All of these proteins are essential for tumor promotion, progression and metastasis. In conclusion, the bromodomain inhibitors JQ1 and I-BET 762 targeted and suppressed multiple pathways in pancreatic cancer. I-BET 762 and a number of other bromodomain inhibitors are currently being tested in several clinical trials, making them potentially promising drugs for the treatment of pancreatic cancer, an often-fatal disease.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antineoplastic Agents/pharmacology , Azepines/pharmacology , Benzodiazepines/pharmacology , Pancreatic Neoplasms/drug therapy , Triazoles/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Ceruletide , Cytokines/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Inflammation Mediators/metabolism , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nitric Oxide/metabolism , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pancreatitis/chemically induced , Pancreatitis/drug therapy , Pancreatitis/immunology , Pancreatitis/metabolism , Phosphorylation , Proto-Oncogene Proteins c-myc/metabolism , RAW 264.7 Cells , Signal Transduction/drug effects
2.
Cancer Prev Res (Phila) ; 9(1): 105-14, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26554632

ABSTRACT

LG101506 was originally synthesized to overcome some of the undesirable side effects of rexinoids. We compared the anticarcinogenic action of LG101506 and LG100268 and for the first time showed that both drugs are useful for prevention of lung cancer in A/J mice. These molecules markedly reduced tumor number, tumor size, and total tumor burden, when chronically administered to A/J mice that had been initiated with the mutagenic carcinogen, vinyl carbamate. Moreover, LG100268 synergized with the histone deacetylase inhibitor, vorinostat, for prevention of experimental lung cancer and enhanced the effect of carboplatin/paclitaxel for treatment of experimental lung cancer. Both rexinoids diminished the percentage of high-grade, highly malignant adenocarcinomas found at autopsy. In cell culture studies, the rexinoids exhibited potent anti-inflammatory properties at nanoMolar concentrations. These drugs suppressed the ability of lipopolysaccharide to stimulate the synthesis and secretion of nitric oxide and inflammatory cytokines and chemokines, such as IL6, IL1ß, CXCL2, and CSF3, in macrophage-like RAW264.7 cells. The present results suggest that LG100268, LG101506, or a related rexinoid may have useful clinical applications in the field of oncology.


Subject(s)
Anticarcinogenic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Carcinogenesis , Fatty Acids, Unsaturated/pharmacology , Lung Neoplasms/drug therapy , Nicotinic Acids/pharmacology , Phenyl Ethers/pharmacology , Tetrahydronaphthalenes/pharmacology , Animals , Carboplatin/administration & dosage , Cell Line , Chemokines/metabolism , Cytokines/metabolism , Female , Flow Cytometry , Humans , Hydroxamic Acids/administration & dosage , Inflammation , Lipopolysaccharides/chemistry , Lipoproteins/blood , Mice , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Oligonucleotide Array Sequence Analysis , Paclitaxel/administration & dosage , Random Allocation , Real-Time Polymerase Chain Reaction , U937 Cells , Vorinostat
3.
Pharmacol Res ; 100: 135-47, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26238177

ABSTRACT

Two new analogues of CDDO-Imidazolide (CDDO-Im), namely 1-[2-Cyano-3,12-dioxooleana-1,9(11)-dien-28-oyl]-4(-pyridin-2-yl)-1H-imidazole ("CDDO-2P-Im") and 1-[2-Cyano-3,12-dioxooleana-1,9(11)-dien-28-oyl]-4(-pyridin-3-yl)-1H-imidazole ("CDDO-3P-Im") have been synthesized and tested for their potential use as chemopreventive drugs. At nanomolar concentrations, they were equipotent to CDDO-Im for inducing differentiation and apoptosis in U937 leukemia cells. As inflammation and oxidative stress contribute to carcinogenesis, we also assessed their cytoprotective potential. The new compounds suppressed inducible nitric oxide synthase (iNOS) expression in RAW264.7 macrophage-like cells and significantly elevated heme oxygenase-1 (HO-1) and quinone reductase (NQO1) mRNA and protein levels in various mouse tissues in vivo. Most importantly, pharmacokinetic studies performed in vitro in human plasma and in vivo showed that each new analogue was more stable than CDDO-Im. Much higher concentrations of the new derivatives were found in mouse liver, lung, pancreas and kidney after gavage in contrast to CDDO-Im. Because of their better bioavailability and their excellent anti-inflammatory profile in vitro, CDDO-2P-Im and CDDO-3P-Im were tested for prevention in a highly relevant mouse lung cancer model, in which A/J mice develop lung carcinomas after injection of vinyl carbamate, a potent carcinogen. CDDO-2P-Im and CDDO-3P-Im were as effective as CDDO-Im for reducing the size and the severity of the lung tumors.


Subject(s)
Anticarcinogenic Agents/pharmacology , Antineoplastic Agents/pharmacology , Imidazoles/pharmacology , Neoplasms/prevention & control , Oleanolic Acid/analogs & derivatives , Animals , Cell Line, Tumor , Cells, Cultured , Female , Heme Oxygenase-1/metabolism , Humans , Mice , NAD(P)H Dehydrogenase (Quinone)/metabolism , Neoplasms/metabolism , Nitric Oxide Synthase Type II/metabolism , Oleanolic Acid/pharmacology , Oxidative Stress/drug effects , RNA, Messenger/metabolism , U937 Cells
4.
Carcinogenesis ; 36(7): 769-81, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25939751

ABSTRACT

Lung cancer accounts for the highest number of cancer-related deaths in the USA, highlighting the need for better prevention and therapy. Activation of the Nrf2 pathway detoxifies harmful insults and reduces oxidative stress, thus preventing carcinogenesis in various preclinical models. However, constitutive activation of the Nrf2 pathway has been detected in numerous cancers, which confers a survival advantage to tumor cells and a poor prognosis. In our study, we compared the effects of two clinically relevant classes of Nrf2 activators, dimethyl fumarate (DMF) and the synthetic oleanane triterpenoids, CDDO-imidazolide (CDDO-Im) and CDDO-methyl ester (CDDO-Me) in RAW 264.7 mouse macrophage-like cells, in VC1 lung cancer cells and in the A/J model of lung cancer. Although the triterpenoids and DMF both activated the Nrf2 pathway, CDDO-Im and CDDO-Me were markedly more potent than DMF. All of these drugs reduced the production of reactive oxygen species and inhibited nitric oxide production in RAW264.7 cells, but the triterpenoids were 100 times more potent than DMF in these assays. Microarray analysis revealed that only 52 of 99 Nrf2-target genes were induced by all three compounds, and each drug regulated a unique subset of Nrf2 genes. These drugs also altered the expression of other genes important in lung cancer independent of Nrf2. Although all three compounds enhanced the phosphorylation of CREB, only DMF increased the phosphorylation of Akt. CDDO-Me, at either 12.5 or 50mg/kg of diet, was the most effective drug in our lung cancer mouse model. Specifically, CDDO-Me significantly reduced the average tumor number, size and burden compared with the control group (P < 0.05). Additionally, 52% of the tumors in the control group were high-grade tumors compared with only 14% in the CDDO-Me group. Though less potent, CDDO-Im had similar activity as CDDO-Me. In contrast, 61-63% of the tumors in the DMF groups (400-1200mg/kg diet) were high-grade tumors compared with 52% for the controls (P < 0.05). Additionally, DMF significantly increased the average number of tumors compared with the controls (P < 0.05). Thus, in contrast to the triterpenoids, which effectively reduced pathogenesis in A/J mice, DMF enhanced the severity of lung carcinogenesis in these mice. Collectively, these results suggest that although CDDO-Im, CDDO-Me and DMF all activate the Nrf2 pathway, they target distinct genes and signaling pathways, resulting in opposite effects for the prevention of experimental lung cancer.


Subject(s)
Fumarates/pharmacology , Imidazoles/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Oleanolic Acid/analogs & derivatives , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Dimethyl Fumarate , Female , Gene Expression Regulation, Neoplastic/drug effects , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice, Inbred Strains , Mice, Knockout , Molecular Targeted Therapy , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Neoplasms, Experimental , Oleanolic Acid/pharmacology , Oligonucleotide Array Sequence Analysis , Oxidative Stress/drug effects
5.
Cell Cycle ; 14(4): 648-55, 2015.
Article in English | MEDLINE | ID: mdl-25590338

ABSTRACT

Approximately 25% of breast cancers overexpress and depend on the receptor tyrosine kinase ERBB2, one of 4 ERBB family members. Targeted therapies directed against ERBB2 have been developed and used clinically, but many patients continue to develop resistance to such therapies. Although much effort has been focused on elucidating the mechanisms of acquired resistance to ERBB2-targeted therapies, the involvement of ERBB4 remains elusive and controversial. We demonstrate that genetic ablation of ERBB4, but not ERBB1-3, led to apoptosis in lapatinib-resistant cells, suggesting that the efficacy of pan-ERBB inhibitors was, at least in part, mediated by the inhibition of ERBB4. Moreover, ERBB4 was upregulated at the protein level in ERBB2+ breast cancer cell lines selected for acquired lapatinib resistance in vitro and in MMTV-Neu mice following prolonged lapatinib treatment. Knockdown of ERBB4 caused a decrease in AKT phosphorylation in resistant cells but not in sensitive cells, suggesting that ERBB4 activated the PI3K/AKT pathway in lapatinib-resistant cells. Importantly, ERBB4 knockdown triggered apoptosis not only in lapatinib-resistant cells but also in trastuzumab-resistant cells. Our results suggest that although ERBB4 is dispensable for naïve ERBB2+ breast cancer cells, it may play a key role in the survival of ERBB2+ cancer cells after they develop resistance to ERBB2 inhibitors, lapatinib and trastuzumab.


Subject(s)
Breast Neoplasms/metabolism , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/physiology , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-4/metabolism , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Line, Tumor , Female , Gene Knockdown Techniques , Lapatinib , Mice , Quinazolines , Receptor, ErbB-4/genetics , Trastuzumab
6.
Cancer Prev Res (Phila) ; 7(7): 698-707, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24817481

ABSTRACT

Poly-ADP ribose polymerase (PARP) inhibitors are effective for the treatment of BRCA-deficient tumors. Women with these mutations have an increased risk of developing breast cancer and would benefit from effective chemoprevention. This study examines whether the PARP inhibitors, veliparib and olaparib, delay mammary gland tumor development in a BRCA1-deficient (BRCA1(Co/Co);MMTV-Cre;p53(+/-)) mouse model. In dose de-escalation studies, mice were fed with control, veliparib (100 mg/kg diet), or olaparib (200, 100, 50, or 25 mg/kg diet) continuously for up to 43 weeks. For intermittent dosing studies, mice cycled through olaparib (200 mg/kg diet) for 2 weeks followed by a 4-week rest period on control diet. To examine biomarkers, mice were fed with olaparib using the intermittent dosing regimen and mammary glands were evaluated by immunohistochemistry. In mice treated with veliparib or olaparib (200 mg/kg diet), the average age of the first detectable tumor was delayed by 2.4 and 6.5 weeks, respectively, compared with controls. Olaparib also increased the average lifespan of mice by 7 weeks. In dose de-escalation studies, lower concentrations of olaparib delayed tumor development but were less effective than the highest dose. When fed intermittently, olaparib delayed the onset of the first palpable tumor by 5.7 weeks and significantly reduced proliferation and induced apoptosis in hyperplastic mammary glands. In summary, veliparib and olaparib are effective for delaying tumor development and extending the lifespan of BRCA1-deficient mice, and intermittent dosing with olaparib was as effective as continuous dosing. These results suggest that the use of PARP inhibitors is a promising chemopreventive option.


Subject(s)
Antineoplastic Agents/therapeutic use , BRCA1 Protein/physiology , Benzimidazoles/therapeutic use , Mammary Neoplasms, Animal/prevention & control , Phthalazines/therapeutic use , Piperazines/therapeutic use , Poly(ADP-ribose) Polymerase Inhibitors , Animals , Apoptosis/drug effects , Biomarkers/analysis , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Female , Humans , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/pathology , Mice , Mice, Knockout , Tumor Suppressor Protein p53/physiology
7.
Antioxid Redox Signal ; 18(2): 139-57, 2013 Jan 10.
Article in English | MEDLINE | ID: mdl-22746536

ABSTRACT

UNLABELLED: Although the etiology of Parkinson's disease (PD) remains unclear, ample empirical evidence suggests that oxidative stress is a major player in the development of PD and in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) neurotoxicity. Nuclear factor E2-related factor 2 (Nrf2) is a redox-sensitive transcription factor that upregulates a battery of antioxidant response element (ARE)-driven antioxidative and cytoprotective genes that defend against oxidative stress. AIMS: We evaluated whether the strategy of activation of Nrf2 and its downstream network of cytoprotective genes with small molecule synthetic triterpenoids (TP) attenuate MPTP-induced PD in mice. RESULTS: We show that synthetic TP are thus far the most potent and direct activators of the Nrf2 pathway using a novel Neh2-luciferase reporter. They upregulate several cytoprotective genes, including those involved in glutathione biosynthesis in vitro. Oral administration of TP that were structurally modified to penetrate the brain-induced messenger RNA and protein levels for a battery of Nrf2-dependent cytoprotective genes reduced MPTP-induced oxidative stress and inflammation, and ameliorated dopaminergic neurotoxicity in mice. The neuroprotective effect of these TP against MPTP neurotoxicity was dependent on Nrf2, since treatment with TP in Nrf2 knockout mice failed to block against MPTP neurotoxicity and induce Nrf2-dependent cytoprotective genes. INNOVATION: Extremely potent synthetic TP that are direct activators of the Nrf2 pathway block dopaminergic neurodegeneration in the MPTP mouse model of PD. CONCLUSION: Our results indicate that activation of Nrf2/antioxidant response element (ARE) signaling by synthetic TP is directly associated with their neuroprotective effects against MPTP neurotoxicity and suggest that targeting the Nrf2/ARE pathway is a promising approach for therapeutic intervention in PD.


Subject(s)
Disease Models, Animal , Dopamine/metabolism , MPTP Poisoning/complications , NF-E2-Related Factor 2/genetics , Parkinson Disease/metabolism , Transcription, Genetic/drug effects , Triterpenes/pharmacology , Administration, Oral , Animals , Mice , Mice, Knockout , Parkinson Disease/etiology , Parkinson Disease/genetics , Transcription, Genetic/physiology , Triterpenes/administration & dosage
8.
Carcinogenesis ; 34(1): 199-210, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23042302

ABSTRACT

Novel drugs and drug combinations are needed for the chemoprevention and treatment of cancer. We show that the histone deacetylase inhibitor vorinostat [suberoylanilide hydroxamic acid (SAHA)] and the methyl ester or ethyl amide derivatives of the synthetic triterpenoid 2-cyano-3,12-dioxooleana-1,9(11)-dien-28-oic acid (CDDO-Me and CDDO-Ea, respectively) cooperated to inhibit the de novo synthesis of nitric oxide in RAW 264.7 macrophage-like cells and in primary mouse peritoneal macrophages. Additionally, SAHA enhanced the ability of synthetic triterpenoids to delay formation of estrogen receptor-negative mammary tumors in MMTV-polyoma middle T (PyMT) mice. CDDO-Me (50 mg/kg diet) and SAHA (250 mg/kg diet) each significantly delayed the initial development of tumors by 4 (P < 0.001) and 2 (P < 0.05) weeks, respectively, compared with the control group in the time required to reach 50% tumor incidence. CDDO-Ea (400 mg/kg diet), as a single agent, did not delay tumor development. The combination of either triterpenoid with SAHA was significantly more potent than the individual drugs for delaying tumor development, with a 7 week (P < 0.001) delay before 50% tumor incidence was reached. SAHA, alone and in combination with CDDO-Me, also significantly (P < 0.05) inhibited the infiltration of tumor-associated macrophages into the mammary glands of PyMT mice and levels of the chemokine macrophage colony-stimulating factor in primary PyMT tumor cells. In addition, SAHA and the synthetic triterpenoids cooperated to suppress secreted levels of the pro-angiogenic factor matrix metalloproteinase-9. Similar results were observed in mouse models of pancreatic and lung cancer. At concentrations that were anti-inflammatory, SAHA had no effect on histone acetylation. These studies suggest that both SAHA and triterpenoids effectively delay tumorigenesis, thereby demonstrating a promising, novel drug combination for chemoprevention.


Subject(s)
Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Mammary Neoplasms, Experimental/prevention & control , Triterpenes/pharmacology , Animals , Cell Proliferation , Disease Models, Animal , Drug Therapy, Combination , Enzyme-Linked Immunosorbent Assay , Histone Deacetylase Inhibitors/administration & dosage , Hydroxamic Acids/administration & dosage , Mice , Triterpenes/administration & dosage , Vorinostat
9.
Cancer Prev Res (Phila) ; 5(5): 726-34, 2012 May.
Article in English | MEDLINE | ID: mdl-22401982

ABSTRACT

Novel drugs are needed for the prevention and treatment of breast cancer. Synthetic triterpenoids are a promising new class of compounds with activity in a variety of preclinical cancer models. We tested activity of the methyl ester derivative of the synthetic triterpenoid, 2-cyano-3,12-dioxooleana-1,9(11)-dien-28-oic acid (CDDO-Me), in a relevant model of estrogen receptor-negative breast cancer, the polyoma-middle T (PyMT), in which the oncoprotein drives carcinogenesis. The developing tumors recapitulate key features of the human disease. Mice were fed CDDO-Me (50 mg/kg diet), starting at 4 weeks of age. CDDO-Me significantly increased the age of mice at onset of first tumor (P < 0.001) by an average of 4.3 weeks and overall survival (P < 0.001) by 5.2 weeks. The drug also inhibited the infiltration of tumor-associated macrophages into mammary glands of PyMT mice at 12 weeks of age and reduced levels of the chemokines CXCL12 and CCL2 in primary PyMT mammary tumor cells. Treatment with this multifunctional drug also inhibited secretion of matrix metalloproteinase-9 in primary tumor cells from PyMT mice and decreased proliferation of these cells by inhibiting cyclin D1 and decreasing phosphorylation of epidermal growth factor receptor and STAT3.


Subject(s)
Antigens, Polyomavirus Transforming/genetics , Carcinoma/prevention & control , Cell Transformation, Neoplastic/drug effects , Mammary Neoplasms, Experimental/prevention & control , Oleanolic Acid/analogs & derivatives , Animals , Anticarcinogenic Agents/pharmacology , Anticarcinogenic Agents/therapeutic use , Carcinoma/genetics , Carcinoma/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Cells, Cultured , Drug Evaluation, Preclinical , Female , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Transgenic , Oleanolic Acid/pharmacology , Oleanolic Acid/therapeutic use , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Time Factors , Triterpenes/chemical synthesis , Triterpenes/pharmacology , Triterpenes/therapeutic use
10.
Cancer Prev Res (Phila) ; 5(1): 89-97, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21933912

ABSTRACT

The breast cancer-associated gene 1 (BRCA1) is the most frequently mutated tumor suppressor gene in familial breast cancers. Mutations in BRCA1 also predispose to other types of cancers, pointing to a fundamental role of this pathway in tumor suppression and emphasizing the need for effective chemoprevention in these high-risk patients. Because the methyl ester of the synthetic triterpenoid 2-cyano-3,12-dioxooleana-1,9(11)-dien-28-oic acid (CDDO-Me) is a potent chemopreventive agent, we tested its efficacy in a highly relevant mouse model of BRCA1-mutated breast cancer. Beginning at 12 weeks of age, Brca1(Co/Co); MMTV-Cre;p53(+/-) mice were fed powdered control diet or diet containing CDDO-Me (50 mg/kg diet). CDDO-Me significantly (P < 0.05) delayed tumor development in the Brca1-mutated mice by an average of 5.2 weeks. We also observed that levels of ErbB2, p-ErbB2, and cyclin D1 increased in a time-dependent manner in the mammary glands in Brca1-deficient mice, and CDDO-Me inhibited the constitutive phosphorylation of ErbB2 in tumor tissues from these mice. In BRCA1-deficient cell lines, the triterpenoids directly interacted with ErbB2, decreased constitutive phosphorylation of ErbB2, inhibited proliferation, and induced G(0)-G(1) arrest. These results suggest that CDDO-Me has the potential to prevent BRCA1-mutated breast cancer.


Subject(s)
Anticarcinogenic Agents/pharmacology , BRCA1 Protein/genetics , Genes, BRCA1 , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/prevention & control , Oleanolic Acid/analogs & derivatives , Animals , Cell Cycle , Female , Genes, p53 , Mice , Mutation , Oleanolic Acid/pharmacology , Phosphorylation , Receptor, ErbB-2/metabolism
11.
Cancer Prev Res (Phila) ; 3(11): 1427-34, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20959520

ABSTRACT

Pancreatic cancer is the fourth leading cause of cancer-related deaths in the United States and is nearly always fatal. Whereas early detection offers the most promising approach for reducing the mortality of this disease, there is still a need to develop effective drugs for the prevention and treatment of pancreatic cancer. We tested two promising classes of noncytotoxic drugs, synthetic oleanane triterpenoids and rexinoids, for the prevention of carcinogenesis in the highly relevant LSL-Kras(G12D/+);LSL-Trp53(R127H/+);Pdx-1-Cre (KPC) mouse model of pancreatic cancer. KPC transgenic mice closely recapitulate the genetic mutations, clinical symptoms, and histopathology found in human pancreatic cancer. Beginning at 4 weeks of age, mice were fed powdered control diet or a diet containing the triterpenoids CDDO-methyl ester (CDDO-Me) or CDDO-ethyl amide, the rexinoid LG100268 (LG268), or the combination, until the mice displayed overt symptoms of pancreatic cancer. CDDO-Me, LG268, the combination of CDDO-Me and LG268, and the combination of CDDO-ethyl amide and LG268, all significantly (P < 0.05) increased survival in the KPC mice by 3 to 4 weeks. Recent studies have shown that gemcitabine, the current standard of care for human pancreatic cancer, does not extend survival in KPC mice. In cell lines developed from the KPC mice, the triterpenoids directly interact with both signal transducer and activator of transcription 3 and IκB kinase (IKK) to decrease constitutive interleukin-6 secretion, inhibit constitutive signal transducer and activator of transcription 3 phosphorylation, and block the degradation of IκBα when challenged with tumor necrosis factor α. These results suggest that oleanane triterpenoids and rexinoids have the potential to prevent pancreatic cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Nicotinic Acids/pharmacology , Oleanolic Acid/analogs & derivatives , Pancreatic Neoplasms/prevention & control , Tetrahydronaphthalenes/pharmacology , Animals , Blotting, Western , Disease Models, Animal , Mice , Mice, Transgenic , Oleanolic Acid/pharmacology , Signal Transduction/drug effects
12.
Cancer Prev Res (Phila) ; 2(12): 1050-8, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19952361

ABSTRACT

We tested members of two noncytotoxic classes of drugs, synthetic oleanane triterpenoids and rexinoids, both as individual agents and in combination, for the prevention and treatment of carcinogenesis in a highly relevant animal model of lung cancer. Lung adenocarcinomas were induced in A/J mice by injection of the carcinogen vinyl carbamate. Mice were fed drugs in diet, beginning 1 week after the carcinogen challenge for prevention or 8 weeks later for treatment. The number, size, and severity of tumors in the lungs were then evaluated. In the prevention studies, the triterpenoids CDDO-ethyl amide and CDDO-methyl ester reduced the average tumor burden (ATB) in the lungs 86% to 92%, respectively, compared with the controls, and the rexinoid LG100268 (268) reduced ATB by 50%. The combination of CDDO-ethyl amide and 268 reduced ATB by 93%. We show for the first time that these drugs also were highly effective for treatment of experimental lung cancer, and all triterpenoid and rexinoid combinations reduced ATB 85% to 87% compared with the control group. The triterpenoids also potently inhibited proliferation of VC1 mouse lung carcinoma cells and directly interacted with key regulatory proteins in these cells. In contrast, the rexinoids had little antiproliferative activity in VC1 cells but were potent inhibitors of the toll-like receptor pathway in macrophage-like cells. Triterpenoids and rexinoids are multifunctional, well-tolerated drugs that target different signaling pathways and are thus highly effective for prevention and treatment of experimental lung cancer.


Subject(s)
Adenocarcinoma/prevention & control , Fatty Acids, Unsaturated/therapeutic use , Lung Neoplasms/prevention & control , Nicotinic Acids/therapeutic use , Oleanolic Acid/analogs & derivatives , Tetrahydronaphthalenes/therapeutic use , Animals , Cell Proliferation/drug effects , Female , Ligands , Macrophages/cytology , Macrophages/drug effects , Mice , Mice, Inbred A , Oleanolic Acid/therapeutic use , Rats , Urethane/analogs & derivatives , Urethane/pharmacology
13.
Carcinogenesis ; 30(6): 1024-31, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19386581

ABSTRACT

Loss of NF-E2-related factor 2 (Nrf2) signaling increases susceptibility to acute toxicity, inflammation and carcinogenesis in mice due to the inability to mount adaptive responses. In contrast, disruption of Keap1 (a cytoplasmic modifier of Nrf2 turnover) protects against these stresses in mice, although inactivating mutations in Keap1 have been identified recently in some human cancers. Global characterization of Nrf2 activation is important to exploit this pathway for chemoprevention in healthy, yet at-risk individuals and also to elucidate the consequences of hijacking the pathway in Keap1-mutant human cancers. Liver-targeted conditional Keap1-null, Albumin-Cre:Keap1((flox/-)) (CKO) mice provide a model of genetic activation of Nrf2 signaling. By coupling global gene expression analysis of CKO mice with analysis of pharmacologic activation using the synthetic oleanane triterpenoid 1-[2-cyano-3,12-dioxooleana-1,9(11)-dien-28-oyl]imidazole (CDDO-Im), we are able to gain insight into pathways affected by Nrf2 activation. CDDO-Im is an extremely potent activator of Nrf2 signaling. CKO mice were used to identify genes modulated by genetic activation of Nrf2 signaling. The CKO response was compared with hepatic global gene expression changes in wild-type mice treated with CDDO-Im at a maximal Nrf2 activating dose. The results show that genetic and pharmacologic activation of Nrf2 signaling modulates pathways beyond detoxication and cytoprotection, with the largest cluster of genes associated with lipid metabolism. Genetic activation of Nrf2 results in much larger numbers of detoxication and lipid metabolism gene changes. Additionally, analysis of pharmacologic activation suggests that Nrf2 is the primary mediator of CDDO-Im activity, though other cell-signaling targets are also modulated following an oral dose of 30 micromol/kg.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cytoskeletal Proteins/metabolism , Imidazoles/pharmacology , Liver/metabolism , NF-E2-Related Factor 2/metabolism , Oleanolic Acid/analogs & derivatives , Adaptor Proteins, Signal Transducing/genetics , Animals , Cytoskeletal Proteins/genetics , Kelch-Like ECH-Associated Protein 1 , Lipid Metabolism/drug effects , Male , Mice , Mice, Knockout , NF-E2-Related Factor 2/genetics , Oleanolic Acid/pharmacology , Oligonucleotide Array Sequence Analysis , Signal Transduction
14.
Cancer Res ; 68(16): 6727-33, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18701497

ABSTRACT

A novel acetylenic tricyclic bis-(cyano enone), TBE-31, is a lead compound in a series of tricyclic compounds with enone functionalities in rings A and C. Nanomolar concentrations of this potent multifunctional molecule suppress the induction of the inflammatory protein, inducible nitric oxide synthase, activate phase 2 cytoprotective enzymes in vitro and in vivo, block cell proliferation, and induce differentiation and apoptosis of leukemia cells. Oral administration of TBE-31 also significantly reduces formation of aflatoxin-DNA adducts and decreases size and number of aflatoxin-induced preneoplastic hepatic lesions in rats by >90%. Because of the two cyano enones in rings A and C, TBE-31 may directly interact with DTT and protein targets such as Keap1 that contain reactive cysteine residues. The above findings suggest that TBE-31 should also be tested for chemoprevention and chemotherapy in relevant models of cancer and against other chronic, degenerative diseases in which inflammation and oxidative stress contribute to disease pathogenesis.


Subject(s)
Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Transformation, Neoplastic/drug effects , Liver Neoplasms/prevention & control , Nitric Oxide Synthase Type II/antagonists & inhibitors , Phenanthrenes/pharmacology , Administration, Oral , Aflatoxin B1/metabolism , Animals , Cell Proliferation/drug effects , Cells, Cultured , DNA Adducts/metabolism , Heme Oxygenase-1/metabolism , Humans , Imidazoles/pharmacology , Leukemia/drug therapy , Liver Neoplasms/chemically induced , Liver Neoplasms/pathology , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Molecular Structure , NAD(P)H Dehydrogenase (Quinone)/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Oleanolic Acid/analogs & derivatives , Oleanolic Acid/pharmacology , Phenanthrenes/chemistry , Rats , Rats, Inbred F344 , Reactive Oxygen Species/metabolism
15.
Clin Cancer Res ; 14(14): 4556-63, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18628471

ABSTRACT

PURPOSE: To test whether the triterpenoid 2-cyano-3,12-dioxooleana-1,9(11)-dien-28-oic acid methyl ester (CDDO-Me) and the rexinoid LG100268 (268) prevent the formation of estrogen receptor (ER)-negative mammary tumors or either arrest the growth or cause regression of established tumors in MMTV-neu mice. EXPERIMENTAL DESIGN: For prevention, mice were fed control diet, CDDO-Me (60 mg/kg diet), 268 (20 mg/kg diet), or the combination for 45 weeks. For treatment, mice with established tumors at least 4 mm in diameter were fed control diet, CDDO-Me (100 mg/kg diet), 268 (60 mg/kg diet), or the combination for 4 weeks. RESULTS: CDDO-Me and 268 significantly delayed the development of ER-negative tumors, with a 14- and 24-week delay, respectively, compared with the control group for the time required to reach 50% tumor incidence. The combination of CDDO-Me and 268 was significantly more potent than the individual drugs, as only one tumor was found in the combination group, after 45 weeks on diet, at which time all control animals had tumors. Treating established tumors with CDDO-Me arrested the growth of 86% of the tumors, and 268 induced tumor regression in 85% of tumors. CDDO-Me and 268 target different signaling pathways and cell types. CDDO-Me inhibited constitutive STAT3 phosphorylation and the degradation of IKBalpha in ER-negative breast cancer cells, whereas 268 blocked IKBalpha degradation and the release of interleukin-6 in RAW264.7 macrophage-like cells, inhibited the ability of endothelial cells to organize into networks, and blocked angiogenesis in vivo. CONCLUSIONS: CDDO-Me and 268 are useful as individual drugs to prevent ER-negative mammary tumorigenesis and to treat established tumors. They synergize when used in combination for prevention.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Mammary Neoplasms, Experimental/drug therapy , Nicotinic Acids/pharmacology , Oleanolic Acid/analogs & derivatives , Tetrahydronaphthalenes/pharmacology , Administration, Oral , Animals , Antineoplastic Agents/pharmacology , Cell Movement/drug effects , Drug Synergism , Endothelial Cells/drug effects , Female , Humans , Mammary Tumor Virus, Mouse/genetics , Mice , Mice, Transgenic , Neovascularization, Pathologic/drug therapy , Oleanolic Acid/pharmacology , Receptor, ErbB-2/genetics , Receptors, Estrogen/biosynthesis
16.
Mol Cancer Ther ; 7(5): 1251-7, 2008 May.
Article in English | MEDLINE | ID: mdl-18483313

ABSTRACT

Female A/J mice injected with the carcinogen vinyl carbamate develop atypical adenomatous hyperplasias in lungs 4 weeks after injection with the carcinogen. The number and severity of tumors then increase over time, making these mice a useful model for evaluating potential chemopreventive agents. The rexinoid LG100268 (LG268), a selective ligand for the retinoid X receptor, and the methyl amide of the synthetic triterpenoid 2-cyano-3,12-dioxooleana-1,9(11)-dien-28-oic acid (CDDO) both significantly reduced the number, size, and severity of the histopathology of lung tumors in female A/J mice when fed in diet for 14 to 20 weeks. The total tumor burden was 85% to 87% lower in mice fed LG268 and CDDO-MA than in controls, and the percentage of high-grade tumors decreased from 59% in the controls to 25% or 30% with CDDO-MA and LG268. Erlotinib, which is used to treat lung cancer patients and is an inhibitor of the epidermal growth factor receptor, was less effective in this model. Immunohistochemical staining of geminin, a marker of cell cycle progression, was higher in lung sections from control mice than in mice treated with LG268. Because rexinoids and triterpenoids signal through different biological pathways, they should be tested in combination for the prevention of lung cancer.


Subject(s)
Anticarcinogenic Agents/therapeutic use , Lung Neoplasms/prevention & control , Lung/drug effects , Nicotinic Acids/therapeutic use , Oleanolic Acid/analogs & derivatives , Quinazolines/therapeutic use , Tetrahydronaphthalenes/therapeutic use , Animals , Cell Line, Tumor , Erlotinib Hydrochloride , Female , Lung Neoplasms/pathology , Mice , Oleanolic Acid/therapeutic use , Protein Kinase Inhibitors/therapeutic use
17.
Clin Cancer Res ; 13(20): 6237-43, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17947492

ABSTRACT

PURPOSE: We evaluated the anti-inflammatory and growth-inhibitory properties of the novel rexinoid NRX194204 (4204) in vitro and then tested its ability to prevent and/or treat experimental lung and estrogen receptor (ER)-negative breast cancer in vivo. EXPERIMENTAL DESIGN: In cell culture studies, we measured the ability of 4204 to block the effects of lipopolysaccharide and induce apoptosis. For the lung cancer prevention studies, A/J mice were injected with the carcinogen vinyl carbamate and then fed 4204 (30-60 mg/kg diet) for 15 weeks, beginning 1 week after the administration of the carcinogen. For breast cancer prevention studies, mouse mammary tumor virus-neu mice were fed control diet or 4204 (20 mg/kg diet) for 50 weeks; for treatment, tumors at least 32 mm3 in size were allowed to form, and then mice were fed control diet or 4204 (60 mg/kg diet) for 4 weeks. RESULTS: Low nanomolar concentrations of 4204 blocked the ability of lipopolysaccharide and tumor necrosis factor-alpha to induce the release of nitric oxide and interleukin 6 and the degradation of IKBalpha in RAW264.7 macrophage-like cells. In the A/J mouse model of lung cancer, 4204 significantly (P < 0.05) reduced the number and size of tumors on the surface of the lungs and reduced the total tumor volume per slide by 64% to 81% compared with the control group. In mouse mammary tumor virus-neu mice, 4204 not only delayed the development of ER-negative mammary tumors in the prevention studies but also caused marked tumor regression (92%) or growth arrest (8%) in all of the mammary tumors when used therapeutically. CONCLUSIONS: The combined anti-inflammatory and anticarcinogenic actions of 4204 suggest that it is a promising new rexinoid that should be considered for future clinical trials.


Subject(s)
Breast Neoplasms/prevention & control , Fatty Acids, Unsaturated/pharmacology , Lung Neoplasms/prevention & control , Receptors, Retinoic Acid/metabolism , Tetrahydronaphthalenes/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Anticarcinogenic Agents/pharmacology , Apoptosis , Breast Neoplasms/metabolism , Carcinogens/chemistry , Fatty Acids, Unsaturated/chemistry , Humans , Interleukin-6/metabolism , Lipopolysaccharides/metabolism , Lung Neoplasms/metabolism , Macrophages/metabolism , Mammary Tumor Virus, Mouse/metabolism , Mice , Models, Biological , Models, Chemical , Receptors, Estrogen/metabolism , Tetrahydronaphthalenes/chemistry
18.
Mol Cancer Ther ; 6(7): 2113-9, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17620440

ABSTRACT

Betulinic acid (BA), a pentacyclic triterpene isolated from birch bark and other plants, selectively inhibits the growth of human cancer cell lines. However, the poor potency of BA hinders its clinical development, despite a lack of toxicity in animal studies even at high concentrations. Here, we describe six BA derivatives that are markedly more potent than BA for inhibiting inducible nitric oxide synthase, activating phase 2 cytoprotective enzymes, and inducing apoptosis in cancer cells and in Bax/Bak(-/-) fibroblasts, which lack two key proteins involved in the intrinsic, mitochondrial-dependent apoptotic pathway. Notably, adding a cyano-enone functionality in the A ring of BA enhanced its cytoprotective properties, but replacing the cyano group with a methoxycarbonyl strikingly increased potency in the apoptosis assays. Higher plasma and tissue levels were obtained with the new BA analogues, especially CBA-Im [1-(2-cyano-3-oxolupa-1,20(29)-dien-28-oyl)imidazole], compared with BA itself and at concentrations that were active in vitro. These results suggest that BA is a useful platform for drug development, and the enhanced potency and varied biological activities of CBA-Im make it a promising candidate for further chemoprevention or chemotherapeutic studies.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Cytoprotection/drug effects , Triterpenes/pharmacology , Animals , Antineoplastic Agents, Phytogenic/blood , Antineoplastic Agents, Phytogenic/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Fibroblasts/cytology , Fibroblasts/drug effects , Humans , Inflammation , Macrophages/drug effects , Macrophages/enzymology , Male , Mice , NAD(P)H Dehydrogenase (Quinone)/metabolism , Nitric Oxide/biosynthesis , Pentacyclic Triterpenes , Triterpenes/blood , Triterpenes/chemistry , bcl-2-Associated X Protein/metabolism , Betulinic Acid
19.
Cancer Res ; 67(6): 2414-9, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17363558

ABSTRACT

We report the first use of new synthetic triterpenoids to prevent lung cancer in experimental animals. Female A/J mice were treated with the mutagenic carcinogen vinyl carbamate, which induces adenocarcinoma of the lung in all animals within 16 weeks. If mice were fed either the methyl ester or the ethyl amide derivative of the synthetic triterpenoid 2-cyano-3,12-dioxooleana-1,9(11)-dien-28-oic acid (CDDO-ME and CDDO-EA, respectively), beginning 1 week after dosing with carcinogen, the number, size, and severity of lung carcinomas were markedly reduced. The mechanisms of action of CDDO-ME and CDDO-EA that are germane to these in vivo findings are the following results shown here in cell culture: (a) suppression of the ability of IFN-gamma to induce de novo formation of nitric oxide synthase in a macrophage-like cell line RAW264.7, (b) induction of heme oxygenase-1 in these RAW cells, and (c) suppression of phosphorylation of the transcription factor signal transducers and activators of transcription 3 as well as induction of apoptosis in human lung cancer cell lines.


Subject(s)
Adenocarcinoma/prevention & control , Anticarcinogenic Agents/pharmacology , Lung Neoplasms/prevention & control , Oleanolic Acid/analogs & derivatives , Adenocarcinoma/chemically induced , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Anticarcinogenic Agents/blood , Anticarcinogenic Agents/pharmacokinetics , Apoptosis/drug effects , Cell Line, Tumor , Humans , Lung/metabolism , Lung Neoplasms/chemically induced , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Inbred A , Oleanolic Acid/blood , Oleanolic Acid/pharmacokinetics , Oleanolic Acid/pharmacology , Phosphorylation , STAT3 Transcription Factor/metabolism , Urethane/analogs & derivatives
20.
Clin Cancer Res ; 12(19): 5902-9, 2006 Oct 01.
Article in English | MEDLINE | ID: mdl-17020999

ABSTRACT

PURPOSE: We tested whether a selective estrogen receptor modulator (SERM) and a rexinoid are active for prevention and treatment in the mouse mammary tumor virus-neu mouse model of estrogen receptor-negative breast cancer. EXPERIMENTAL DESIGN: For prevention, mice were fed a powdered control diet, the SERM arzoxifene (Arz, 20 mg/kg diet), the rexinoid LG100268 (268, 30 mg/kg diet), or the combination for 60 weeks. In a second prevention study, mice were fed Arz (6 mg/kg diet), 268 (30 mg/kg diet), the combination of Arz and 268, the SERM acolbifene (Acol, 3 mg/kg diet), or the combination of Acol and 268 for 52 weeks. For the treatment studies, mice with tumors were fed combinations of a SERM and 268 for 4 weeks. RESULTS: The rexinoid 268 and the SERMs Arz and Acol, as individual drugs, delayed the development of estrogen receptor-negative tumors. Moreover, the combination of a SERM and 268 was strikingly synergistic, as no tumors developed in any mouse fed the combination of 268 and a SERM. Moreover, this drug combination also induced significant tumor regression when used therapeutically. These drugs did not inhibit transgene expression in vitro or in vivo, and the combination of Arz and 268 inhibited proliferation and induced apoptosis in the tumors. CONCLUSION: The combination of a rexinoid and SERM should be considered for future clinical trials.


Subject(s)
Disease Models, Animal , Mammary Neoplasms, Experimental/prevention & control , Nicotinic Acids/therapeutic use , Piperidines/therapeutic use , Receptors, Estrogen/metabolism , Selective Estrogen Receptor Modulators/pharmacology , Tetrahydronaphthalenes/therapeutic use , Thiophenes/therapeutic use , Animals , Cell Division/drug effects , Drug Synergism , Drug Therapy, Combination , Female , Humans , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Transgenic , Survival Rate
SELECTION OF CITATIONS
SEARCH DETAIL
...