benzofurans and Pancreatic-Neoplasms

benzofurans has been researched along with Pancreatic-Neoplasms* in 18 studies

Trials

1 trial(s) available for benzofurans and Pancreatic-Neoplasms

ArticleYear
CanStem111P trial: a Phase III study of napabucasin plus nab-paclitaxel with gemcitabine.
    Future oncology (London, England), 2019, Volume: 15, Issue:12

    Napabucasin (also known as BBI-608 or BBI608) is an investigational, oral agent hypothesized to inhibit multiple oncogenic pathways. In this article, we describe the design and rationale for the CanStem111P clinical trial, a multicenter, randomized, open-label, Phase III study designed to determine the efficacy and safety of combining napabucasin with nab-paclitaxel and gemcitabine for first-line treatment of patients with metastatic pancreatic adenocarcinoma (NCT02993731). Patients were randomized in a 1:1 fashion to receive weekly gemcitabine and nab-paclitaxel with or without napabucasin. The results of this study will help define the role of this novel agent in the management of advanced pancreatic cancer.

    Topics: Administration, Oral; Adult; Albumins; Antineoplastic Combined Chemotherapy Protocols; Benzofurans; Carcinoma, Pancreatic Ductal; Clinical Trials, Phase III as Topic; Deoxycytidine; Drug Administration Schedule; Gemcitabine; Humans; Multicenter Studies as Topic; Naphthoquinones; Paclitaxel; Pancreatic Neoplasms; Progression-Free Survival; Randomized Controlled Trials as Topic; Young Adult

2019

Other Studies

17 other study(ies) available for benzofurans and Pancreatic-Neoplasms

ArticleYear
Marine bioactive compound dieckol induces apoptosis and inhibits the growth of human pancreatic cancer cells PANC-1.
    Journal of biochemical and molecular toxicology, 2021, Volume: 35, Issue:2

    Pancreatic cancer, which threatens the global population, is a very aggressive disease with an increased mortality rate. Regarding the types of cancer, pancreatic cancer is prone to display significant resistance to conventional therapy, therefore there 5-year survival rate is only 2% to 9%. Bioactive metabolites of marine algae such as polysaccharides, chitin, carternoids, and sterols possess immense pharmacological properties and tend to be promising alternatives for cancer treatment. Dieckol is one such polyphenolic bioactive compound extracted from brown algae Ecklonia cava, which is proven to possess antioxidant, anti-inflammatory, antibacterial, antidiabetic properties. Therefore in the present study, we analyzed the anticancer property of dieckol on PANC-1 pancreatic carcinoma cells. The cytotoxicity property of dieckol against PANC-1 cells was assessed with 3-(4,5-dimethylthiazol-2-yl)-2, 5-diphenyl tetrazolium bromide assay, and cell morphological analysis. The generation of reactive oxygen species by dieckol on PANC-1 was analyzed with DCFH-DA staining and confirmed by quantifying antioxidants levels in untreated and dieckol-treated PANC-1 cells. The induction of apoptosis was further evaluated with different staining techniques such as Rhodamine 123 staining, acridine orange/ethidium bromide staining, DAPI staining, propidium iodide staining and was confirmed by estimating the protein expression of apoptotic genes, Bax and Bcl2. Cell adhesion assay and estimation of inflammatory cytokines were performed to detect the inhibitory effect of dieckol against cancer cell progression. It is further confirmed by analyzing cancer cell progression proteins, that is, proliferating cell nuclear antigen and cyclin D1 expressions in untreated and dieckol-treated PANC-1 cells. Our overall results authentically prove dieckol persuasively induces apoptosis and inhibits the progression of human pancreatic cancer cells in vitro, suggesting dieckol as a potent marine-based phytochemical to treat pancreatic cancer.

    Topics: Apoptosis; Benzofurans; Cell Line, Tumor; Cell Proliferation; Humans; Membrane Potential, Mitochondrial; Pancreatic Neoplasms; Reactive Oxygen Species; Seawater

2021
Combined inhibition of Ref-1 and STAT3 leads to synergistic tumour inhibition in multiple cancers using 3D and in vivo tumour co-culture models.
    Journal of cellular and molecular medicine, 2021, Volume: 25, Issue:2

    With a plethora of molecularly targeted agents under investigation in cancer, a clear need exists to understand which pathways can be targeted simultaneously with multiple agents to elicit a maximal killing effect on the tumour. Combination therapy provides the most promise in difficult to treat cancers such as pancreatic. Ref-1 is a multifunctional protein with a role in redox signalling that activates transcription factors such as NF-κB, AP-1, HIF-1α and STAT3. Formerly, we have demonstrated that dual targeting of Ref-1 (redox factor-1) and STAT3 is synergistic and decreases cell viability in pancreatic cancer cells. Data presented here extensively expands upon this work and provides further insights into the relationship of STAT3 and Ref-1 in multiple cancer types. Using targeted small molecule inhibitors, Ref-1 redox signalling was blocked along with STAT3 activation, and tumour growth evaluated in the presence and absence of the relevant tumour microenvironment. Our study utilized qPCR, cytotoxicity and in vivo analysis of tumour and cancer-associated fibroblasts (CAF) response to determine the synergy of Ref-1 and STAT3 inhibitors. Overall, pancreatic tumours grown in the presence of CAFs were sensitized to the combination of STAT3 and Ref-1 inhibition in vivo. In vitro bladder and pancreatic cancer demonstrated the most synergistic responses. By disabling both of these important pathways, this combination therapy has the capacity to hinder crosstalk between the tumour and its microenvironment, leading to improved tumour response.

    Topics: Animals; Benzofurans; Blotting, Western; Cell Line, Tumor; DNA-(Apurinic or Apyrimidinic Site) Lyase; HCT116 Cells; Humans; Immunohistochemistry; Mice; Naphthoquinones; Nitriles; Pancreatic Neoplasms; Pyrazoles; Pyrimidines; Reactive Oxygen Species; STAT3 Transcription Factor; Tumor Microenvironment

2021
Discovery of a Napabucasin PROTAC as an Effective Degrader of the E3 Ligase ZFP91.
    Journal of medicinal chemistry, 2021, 02-11, Volume: 64, Issue:3

    Napabucasin, undergoing multiple clinical trials, was reported to inhibit the signal transducer and transcription factor 3 (STAT3). To better elucidate its mechanism of action, we designed a napabucasin-based proteolysis targeting chimera (PROTAC),

    Topics: Benzofurans; Cell Line, Tumor; Drug Design; Drug Screening Assays, Antitumor; Gene Knockdown Techniques; Humans; Hypoxia-Inducible Factor 1, alpha Subunit; Mutant Chimeric Proteins; NAD(P)H Dehydrogenase (Quinone); Naphthoquinones; Pancreatic Neoplasms; Proteolysis; Signal Transduction; STAT3 Transcription Factor; Structure-Activity Relationship; Ubiquitin-Protein Ligases

2021
Licocoumarone induces BxPC-3 pancreatic adenocarcinoma cell death by inhibiting DYRK1A.
    Chemico-biological interactions, 2020, Jan-25, Volume: 316

    Protein kinases play an indispensable role in signaling pathways that regulate tumor cell functions, which represent potent therapeutic targets in cancers. Dual-specificity tyrosine phosphorylation regulated kinase 1A (DYRK1A) as a serine/threonine kinase has recently been reported to be upregulated in pancreatic ductal adenocarcinoma (PDAC) and show protumorigenic effect. By activity-guided phytochemical investigation of the extracts from Glycyrrhiza uralensis Fisch, we expect to find the effective constituents that can suppress pancreatic cancer cell proliferation and/or induce cells apoptotic by inhibiting DYRK1A. Eight isopentenyl-substituted compounds (1-8), including four coumarins (1-4), one benzofuran (5), and three flavonoids (6-8), were isolated and identified from G. uralensis Fisch. Among them, licocoumarone (LC, 5) showed effective inhibitory activity against DYRK1A with an IC

    Topics: Apoptosis; Benzofurans; Binding Sites; Catalytic Domain; Cell Line, Tumor; Down-Regulation; Dyrk Kinases; Glycyrrhiza; Humans; Hydrogen Bonding; Molecular Docking Simulation; Pancreatic Neoplasms; Plant Roots; Protein Serine-Threonine Kinases; Protein-Tyrosine Kinases; Proto-Oncogene Proteins c-met; Resorcinols

2020
Bioactivation of Napabucasin Triggers Reactive Oxygen Species-Mediated Cancer Cell Death.
    Clinical cancer research : an official journal of the American Association for Cancer Research, 2019, 12-01, Volume: 25, Issue:23

    Napabucasin (2-acetylfuro-1,4-naphthoquinone or BBI-608) is a small molecule currently being clinically evaluated in various cancer types. It has mostly been recognized for its ability to inhibit STAT3 signaling. However, based on its chemical structure, we hypothesized that napabucasin is a substrate for intracellular oxidoreductases and therefore may exert its anticancer effect through redox cycling, resulting in reactive oxygen species (ROS) production and cell death.. Binding of napabucasin to NAD(P)H:quinone oxidoreductase-1 (NQO1), and other oxidoreductases, was measured. Pancreatic cancer cell lines were treated with napabucasin, and cell survival, ROS generation, DNA damage, transcriptomic changes, and alterations in STAT3 activation were assayed. Napabucasin was found to bind with high affinity to NQO1 and to a lesser degree to cytochrome P450 oxidoreductase (POR). Treatment resulted in marked induction of ROS and DNA damage with an NQO1- and ROS-dependent decrease in STAT3 phosphorylation. Differential cytotoxic effects were observed, where NQO1-expressing cells generating cytotoxic levels of ROS at low napabucasin concentrations were more sensitive. Cells with low or no baseline NQO1 expression also produced ROS in response to napabucasin, albeit to a lesser extent, through the one-electron reductase POR.. Napabucasin is bioactivated by NQO1, and to a lesser degree by POR, resulting in futile redox cycling and ROS generation. The increased ROS levels result in DNA damage and multiple intracellular changes, one of which is a reduction in STAT3 phosphorylation.

    Topics: Apoptosis; Benzofurans; Cell Proliferation; DNA Damage; Humans; NAD(P)H Dehydrogenase (Quinone); Naphthoquinones; Oxidation-Reduction; Pancreatic Neoplasms; Reactive Oxygen Species; STAT3 Transcription Factor; Tumor Cells, Cultured

2019
Sequential Targeting TGF-β Signaling and KRAS Mutation Increases Therapeutic Efficacy in Pancreatic Cancer.
    Small (Weinheim an der Bergstrasse, Germany), 2019, Volume: 15, Issue:24

    Pancreatic cancer is a highly aggressive malignancy that strongly resists extant treatments. The failure of existing therapies is majorly attributed to the tough tumor microenvironment (TME) limiting drug access and the undruggable targets of tumor cells. The formation of suppressive TME is regulated by transforming growth factor beta (TGF-β) signaling, while the poor response and short survival of almost 90% of pancreatic cancer patients results from the oncogenic KRAS mutation. Hence, simultaneously targeting both the TGF-β and KRAS pathways might dismantle the obstacles of pancreatic cancer therapy. Here, a novel sequential-targeting strategy is developed, in which antifibrotic fraxinellone-loaded CGKRK-modified nanoparticles (Frax-NP-CGKRK) are constructed to regulate TGF-β signaling and siRNA-loaded lipid-coated calcium phosphate (LCP) biomimetic nanoparticles (siKras-LCP-ApoE3) are applied to interfere with the oncogenic KRAS. Frax-NP-CGKRK successfully targets the tumor sites through the recognition of overexpressed heparan sulfate proteoglycan, reverses the activated cancer-associated fibroblasts (CAFs), attenuates the dense stroma barrier, and enhances tumor blood perfusion. Afterward, siKras-LCP-ApoE3 is efficiently internalized by the tumor cells through macropinocytosis and specifically silencing KRAS mutation. Compared with gemcitabine, this sequential-targeting strategy significantly elongates the lifespans of pancreatic tumor-bearing animals, hence providing a promising approach for pancreatic cancer therapy.

    Topics: Animals; Antineoplastic Combined Chemotherapy Protocols; Apolipoprotein E3; Benzofurans; Drug Administration Schedule; Drug Carriers; Gene Expression Regulation, Neoplastic; Humans; Mice; Mice, Nude; Molecular Targeted Therapy; Mutation; Nanoparticles; NIH 3T3 Cells; Pancreatic Neoplasms; Peptide Fragments; Proto-Oncogene Proteins p21(ras); RNA, Small Interfering; Signal Transduction; Transforming Growth Factor beta; Tumor Cells, Cultured; Xenograft Model Antitumor Assays

2019
Nucleus-Targeted, Echogenic Polymersomes for Delivering a Cancer Stemness Inhibitor to Pancreatic Cancer Cells.
    Biomacromolecules, 2018, 10-08, Volume: 19, Issue:10

    Chemotherapeutic agents for treating cancers show considerable side effects, toxicity, and drug resistance. To mitigate the problems, we designed nucleus-targeted, echogenic, stimuli-responsive polymeric vesicles (polymersomes) to transport and subsequently release the encapsulated anticancer drugs within the nuclei of pancreatic cancer cells. We synthesized an alkyne-dexamethasone derivative and conjugated it to N

    Topics: Antineoplastic Agents; Benzofurans; Cell Nucleus; Cell Survival; Drug Carriers; Drug Delivery Systems; Humans; Naphthoquinones; Neoplastic Stem Cells; Pancreatic Neoplasms; Polymers; Tumor Cells, Cultured

2018
Design, synthesis, and evaluation of benzofuran derivatives as novel anti-pancreatic carcinoma agents via interfering the hypoxia environment by targeting HIF-1α pathway.
    European journal of medicinal chemistry, 2017, Sep-08, Volume: 137

    Pancreatic ductal adenocarcinoma (PDAC) is one of the most common type of pancreatic cancer, and has still been the medicinal mystery. New drugs and treatment strategies are urgently needed. In this study, 32 benzofuran derivatives are designed, synthesized and evaluated as potential agents against the pancreatic cancer. Among them, compound 9o with the best physicochemical and pharmacokinetic properties exhibited excellent cytotoxicity against many tumor cell lines. In vivo study showed that compound 9o dramatically suppressed the tumor growth of nude mice. Furthermore, compound 9o could affect the hypoxia environment through Hif-1α/VEGF pathway, resulting in the anti-angiogenic activity. These studies indicated that compound 9o was a promising candidate for the treatment of PDAC, deserving further studies.

    Topics: Animals; Antineoplastic Agents; Benzofurans; Cell Proliferation; Dose-Response Relationship, Drug; Drug Design; Drug Screening Assays, Antitumor; Humans; Hypoxia; Hypoxia-Inducible Factor 1, alpha Subunit; Mice; Molecular Structure; Neoplasms, Experimental; Pancreatic Neoplasms; Structure-Activity Relationship; Tumor Cells, Cultured

2017
Cellular mechanisms of the anticancer effects of the lichen compound usnic acid.
    Planta medica, 2010, Volume: 76, Issue:10

    The lichen compound usnic acid is used for its antimicrobial activities in cosmetic products and is also a component of slimming agents. Its effect against cancer cells was first noted over 30 years ago. In this study possible mechanisms of this effect were investigated using two human cell lines, the breast cancer cell line T-47D and the pancreatic cancer cell line Capan-2. Pure (+)-usnic acid from CLADONIA ARBUSCULA and (-)-usnic acid from ALECTORIA OCHROLEUCA were shown to be equally effective inhibitors of DNA synthesis, with IC (50) 4.2 microg/mL and 4.0 microg/mL for (+) and (-)-usnic acid against T-47D, and 5.3 microg/mL and 5.0 microg/mL against Capan-2, respectively. Flow cytometric analysis confirmed the inhibited entry into the S-phase and showed reduction in cell size. Classical apoptosis, as assessed by TUNEL staining, was not observed. Necrosis, measured by LDH release, was seen only in Capan-2 after exposure for 48 hours. Staining with the mitochondrial dye JC-1 demonstrated dose-dependent loss of mitochondrial membrane potential following treatment with usnic acid in both cell lines. In conclusion, usnic acid had a marked inhibitory effect on growth and proliferation of two different human cancer cell lines and led to loss of mitochondrial membrane potential. Cell survival was little affected; late necrosis was seen in one of the cell lines. No difference was noted between the two enantiomers.

    Topics: Antineoplastic Agents, Phytogenic; Benzimidazoles; Benzofurans; Breast Neoplasms; Carbocyanines; Cell Cycle; Cell Line, Tumor; Cell Proliferation; DNA; Dose-Response Relationship, Drug; Female; Flow Cytometry; Humans; Inhibitory Concentration 50; Lichens; Membrane Potential, Mitochondrial; Necrosis; Pancreatic Neoplasms; Phytotherapy; Plant Extracts

2010
From a natural product lead to the identification of potent and selective benzofuran-3-yl-(indol-3-yl)maleimides as glycogen synthase kinase 3beta inhibitors that suppress proliferation and survival of pancreatic cancer cells.
    Journal of medicinal chemistry, 2009, Apr-09, Volume: 52, Issue:7

    Recent studies have demonstrated that glycogen synthase kinase 3beta (GSK-3beta) is overexpressed in human colon and pancreatic carcinomas, contributing to cancer cell proliferation and survival. Here, we report the design, synthesis, and biological evaluation of benzofuran-3-yl-(indol-3-yl)maleimides, potent GSK-3beta inhibitors. Some of these compounds show picomolar inhibitory activity toward GSK-3beta and an enhanced selectivity against cyclin-dependent kinase 2 (CDK-2). Selected GSK-3beta inhibitors were tested in the pancreatic cancer cell lines MiaPaCa-2, BXPC-3, and HupT3. We determined that some of these compounds, namely compounds 5, 6, 11, 20, and 26, demonstrate antiproliferative activity against some or all of the pancreatic cancer cells at low micromolar to nanomolar concentrations. We found that the treatment of pancreatic cancer cells with GSK-3beta inhibitors 5 and 26 resulted in suppression of GSK-3beta activity and a distinct decrease of the X-linked inhibitor of apoptosis (XIAP) expression, leading to significant apoptosis. The present data suggest a possible role for GSK-3beta inhibitors in cancer therapy, in addition to their more prominent applications in CNS disorders.

    Topics: Antineoplastic Agents; Apoptosis; Benzofurans; Cell Line, Tumor; Cell Proliferation; Cell Survival; Cyclin-Dependent Kinase 2; Drug Screening Assays, Antitumor; Glycogen Synthase Kinase 3; Glycogen Synthase Kinase 3 beta; Humans; Indoles; Maleimides; Models, Molecular; Pancreatic Neoplasms; Structure-Activity Relationship; X-Linked Inhibitor of Apoptosis Protein

2009
Preparation of leukotriene B(4) inhibitory active 2- and 3-(2-aminothiazol-4-yl)benzo[b]furan derivatives and their growth inhibitory activity on human pancreatic cancer cells.
    Organic & biomolecular chemistry, 2008, Aug-07, Volume: 6, Issue:15

    A series of 2-(2-aminothiazol-4-yl)benzo[b]furan and 3-(2-aminothiazol-4-yl)benzo[b]furan derivatives were prepared, and their leukotriene B(4) inhibitory activity and growth inhibitory activity in cancer cell lines were evaluated. Several compounds showed strong inhibition of calcium mobilization in CHO cells overexpressing human BLT(1) and BLT(2) receptors and growth inhibition to human pancreatic cancer cells MIA PaCa-2. 3-(4-Chlorophenyl)-2-[5-formyl-2-[(dimethylamino)methyleneamino]thiazol-4-yl]-5-methoxybenzo[b]furan 8b showed the most potent and selective inhibition for the human BLT(2) receptor, and its IC(50) value was smaller than that of the selected positive control compound, ZK-158252. 3-(4-Chlorophenyl)-2-[2-[(dimethylamino)methyleneamino]-5-(2-hydroxyethyliminomethyl)thiazol-4-yl]-5-methoxybenzo[b]furan 9a displayed growth inhibitory activity towards MIA PaCa-2.

    Topics: Animals; Antineoplastic Agents; Benzofurans; Cell Line, Tumor; CHO Cells; Cricetinae; Cricetulus; Drug Screening Assays, Antitumor; Humans; Inhibitory Concentration 50; Molecular Structure; Pancreatic Neoplasms; Receptors, Leukotriene B4; Thiazoles

2008
BPDZ 154 activates adenosine 5'-triphosphate-sensitive potassium channels: in vitro studies using rodent insulin-secreting cells and islets isolated from patients with hyperinsulinism.
    The Journal of clinical endocrinology and metabolism, 2002, Volume: 87, Issue:11

    A novel ATP-sensitive potassium channel (K(ATP)) channel agonist, BPDZ 154 (6,7-dichloro-3-isopropylamino-4H-1,2,4-benzothiadiazine 1,1-dioxide), was synthesized, and its effects on insulin-secreting cells were evaluated using electrophysiology, (86)Rb(+) and (45)Ca(2+) efflux, and RIA determinations of insulin secretion. BPDZ 154, an analog of diazoxide, inhibited both glucose-induced insulin secretion from isolated perifused islets and the secretion of insulin induced by glucose and tolbutamide. These effects were mediated by the activation of ATP-sensitive potassium channels because BPDZ 154 induced a concentration-dependent increase in channel activity that was inhibited by the sulfonylurea tolbutamide and the imidazoline efaroxan. In beta-cells isolated from patients with either nontypical hyperinsulinism (preserved K(ATP) channel function) or from the control areas of the pancreas of patients with focal hyperinsulinism, BPDZ 154 activated K(ATP) channels and was found to be more effective and less readily reversible than diazoxide. By contrast, it was not possible to activate K(ATP) channels by either diazoxide or BPDZ 154 in beta-cells from patients with hyperinsulinism as a consequence of defects in K(ATP) channel function. In beta-cells isolated from a patient with pancreatic insulinoma, K(ATP) channels were readily recorded and modulated by BPDZ 154. These data suggest that BPDZ 154 or BPDZ 154-like compounds may have therapeutic potential in the treatment of certain forms of hyperinsulinism.

    Topics: Adenosine Triphosphate; Adolescent; Adrenergic alpha-Antagonists; Animals; Benzofurans; Benzothiadiazines; Calcium Radioisotopes; Cell Line; Child, Preschool; Cyclic S-Oxides; Female; Glucose; Humans; Hyperinsulinism; Hypoglycemic Agents; Imidazoles; Infant; Insulin; Insulin Secretion; Insulinoma; Islets of Langerhans; Male; Pancreatic Neoplasms; Potassium Channels; Rats; Rats, Wistar; Rubidium Radioisotopes; Tolbutamide

2002
ATP-sensitive potassium channels and efaroxan-induced insulin release in the electrofusion-derived BRIN-BD11 beta-cell line.
    Diabetes, 1999, Volume: 48, Issue:12

    The properties of ATP-sensitive K+ (K(ATP)) channels were explored in the electrofusion-derived, glucose-responsive, insulin-secreting cell line BRIN-BD11 using patch-clamp techniques. In intact cells, K(ATP) channels were inhibited by glucose, the sulfonylurea tolbutamide, and the imidazoline compounds efaroxan and phentolamine. Each of these agents initiated insulin secretion and potentiated the actions of glucose. K(ATP) channels were blocked by ATP in a concentration-dependent manner and activated by ADP in the presence of ATP. In both intact cells and excised inside-out patches, the K(ATP) channel agonists diazoxide and pinacidil activated channels, and both compounds inhibited insulin secretion evoked by glucose, tolbutamide, and imidazolines. The mechanisms of action of imidazolines were examined in more detail. Pre-exposure of BRIN-BD11 cells to either efaroxan or phentolamine selectively inhibited imidazoline-induced insulin secretion but not the secretory responses of cells to glucose, tolbutamide, or a depolarizing concentration of KCl. These conditions did not result in the loss of depolarization-dependent rises in intracellular Ca2+ ([Ca2+]i), K(ATP) channel operation, or the actions of either ATP or efaroxan on K(ATP) channels. Desensitization of the imidazoline receptor following exposure to high concentrations of efaroxan, however, was found to result in an increase in SUR1 protein expression and, as a consequence, an upregulation of K(ATP) channel density. Our data provide 1) the first characterization of K(ATP) channels in BRIN-BD11 cells, a novel insulin-secreting cell line produced by electrofusion techniques, and 2) a further analysis of the role of imidazolines in the control of insulin release.

    Topics: Adenosine Diphosphate; Adenosine Triphosphate; Adrenergic alpha-Antagonists; Animals; Benzofurans; Cell Fusion; Cell Line; Diazoxide; Glucose; Imidazoles; Insulin; Insulin Secretion; Insulinoma; Islets of Langerhans; Membrane Potentials; Pancreatic Neoplasms; Phentolamine; Pinacidil; Potassium Channels; Tolbutamide; Tumor Cells, Cultured

1999
Imidazolines stimulate release of insulin from RIN-5AH cells independently from imidazoline I1 and I2 receptors.
    European journal of pharmacology, 1994, Sep-01, Volume: 262, Issue:1-2

    The effect on insulin release of efaroxan, an alpha 2-adrenoceptor antagonist and a highly potent drug at imidazoline I1 receptors, and the effects of seven other imidazoline compounds selective for the imidazoline I1 or I2 receptors, were studied in the rat insulinoma cell line RIN-5AH. The cells released insulin in response to glucose (0.3-10 mM), and efaroxan (100 microM) potentiated glucose-induced insulin release. (-)-Adrenaline completely displaced the binding of [125I]p-iodoclonidine to membranes of RIN-5AH cells, indicating that these cells do not express imidazoline I1 receptors. Cirazoline and idazoxan (100 microM), both highly potent drugs at imidazoline I2 receptors, and the guanidines guanoxan and amiloride (200 microM), also promoted insulin release from RIN-5AH cells. Irreversible blockade of imidazoline I2 receptors with 10 microM clorgyline did not prevent the stimulatory effects of cirazoline or idazoxan; however, these compounds completely reversed the inhibition by diazoxide (250 microM), an opener of ATP-dependent K+ channels (K+ATP channels), of glucose-induced insulin release. These data indicate that the imidazoline/guanidine compounds promote insulin release from RIN-5AH cells, by interacting with a novel binding site related to K+ATP channels that does not represent any of the known imidazoline I1 or I2 receptors.

    Topics: Adrenergic alpha-Agonists; Adrenergic alpha-Antagonists; Adrenergic beta-Agonists; Affinity Labels; Animals; Benzofurans; Binding, Competitive; Clonidine; Clorgyline; Diazoxide; Dioxanes; Guanidines; Idazoxan; Imidazoles; Imidazoline Receptors; Insulin; Insulinoma; Ligands; Pancreatic Neoplasms; Potassium Channels; Rats; Receptors, Drug; Regression Analysis; Software; Tumor Cells, Cultured

1994
Cytotoxicity and antitumor activity of carzelesin, a prodrug cyclopropylpyrroloindole analogue.
    Cancer research, 1992, Sep-15, Volume: 52, Issue:18

    The cyclopropylpyrroloindole analogues are DNA minor-groove binders containing a cyclopropyl group, which mediates N3-adenine covalent adduct formation in a sequence-selective fashion. Carzelesin (U-80244) is a cyclopropylpyrroloindole prodrug containing a relatively nonreactive chloromethyl precursor to the cyclopropyl function. Activation of carzelesin requires two steps, (a) hydrolysis of a phenylurethane substituent to form U-76073, followed by (b) ring closure to form the cyclopropyl-containing DNA-reactive U-76074. The formation of the DNA-reactive U-76074, via U-76073, from carzelesin was shown to proceed very slowly in phosphate-buffered saline (t1/2 greater than 24 h) but to occur rapidly in plasma from mouse, rat, dog, and human (initial t1/2 values ranging from 18 min for mouse to 52 min for rat) and in cell culture medium (t1/2 approximately 40 min). Although carzelesin was less potent in terms of in vitro cytotoxicity and in vivo optimal dosage and showed low affinity for binding to DNA, it was therapeutically more efficacious against mouse L1210 leukemia than was U-76074 or adozelesin (U-73975), another cyclopropylpyrroloindole analogue which is currently in phase I clinical trials. Carzelesin also proved to be more efficacious than U-76074 or adozelesin against mouse pancreatic ductal 02 adenocarcinoma, a system reported to be resistant to every agent tested. Carzelesin was highly effective against this tumor and produced 97% tumor growth inhibition. In addition, i.v. administered carzelesin showed significant activity (National Cancer Institute criteria) against i.v. or s.c. implanted Lewis lung carcinoma, i.p. or s.c. implanted B16 melanoma, s.c. implanted colon 38 carcinoma, and five s.c. implanted human tumor xenografts, including clear cell Caki-1 carcinoma, colon CX-1 adenocarcinoma, lung LX-1 tumor, ovarian 2780 carcinoma, and prostatic DU-145 carcinoma. Carzelesin treatment produced 100% complete remissions (no palpable tumor mass at the termination of the experiment) in mice bearing early-stage human ovarian 2780. Pharmacologically, carzelesin proved to be relatively schedule and route independent and was highly active against i.p. implanted L1210 leukemia, regardless of whether the analogue was given i.v., i.p., s.c., or p.o. These results, collectively, suggest that carzelesin is absorbed and distributed well. Both carzelesin and adozelesin caused marked tumor shrinkage in mice bearing human lung LX-1 or advanced-stage human ovar

    Topics: Adenocarcinoma; Animals; Antineoplastic Agents; Benzofurans; Cell Survival; Colonic Neoplasms; Culture Media; Duocarmycins; Indoles; Leukemia L1210; Metabolic Clearance Rate; Mice; Mice, Nude; Neoplasm Transplantation; Pancreatic Neoplasms; Plasma; Prodrugs; Transplantation, Heterologous

1992
Adozelesin, a selected lead among cyclopropylpyrroloindole analogs of the DNA-binding antibiotic, CC-1065.
    Investigational new drugs, 1991, Volume: 9, Issue:2

    Adozelesin (U-73975) is a potent synthetic cyclopropylpyrroloindole (CPI) analog of the cytotoxic DNA-binding antibiotic, CC-1065. In contrast to the natural product, adozelesin and related CPI analogs do not cause delayed death in non-tumored mice. Adozelesin, selected from a series of analogs for its superior in vivo antitumor activity and ease of formulation, is highly active when administered i.v. against i.p. - or s.c.- implanted murine tumors, including L1210 leukemia, B16 melanoma, M5076 sarcoma, and colon 38 carcinoma, and produces long-term survivors in mice bearing i.v.-inoculated L1210 and Lewis lung carcinoma. Modest activity is shown against the highly drug-resistant pancreas 02 carcinoma. Adozelesin is also highly effective against human tumor xenografts s.c.-implanted in athymic (nude) mice, including colon CX-1 adenocarcinoma, lung LX-1 tumor, clear cell Caki-1 carcinoma, and ovarian 2780 carcinoma. Its broad spectrum of in vivo activity compares favorably with three widely used antitumor drugs, i.e. cisplatin, cyclophosphamide, and doxorubicin. Adozelesin appears to be more effective than these drugs in the treatment of very resistant tumors such as s.c.-implanted mouse B16 melanoma, pancreatic 02 carcinoma, and human colon CX-1 and human lung LX-1 tumor xenografts. Based on its high potency and high efficacy against a broad spectrum of experimental tumors, adozelesin was chosen for clinical investigation and development.

    Topics: Animals; Antineoplastic Agents; Benzofurans; Colonic Neoplasms; Cyclohexanecarboxylic Acids; Cyclohexenes; DNA, Neoplasm; Duocarmycins; Female; Humans; Indoles; Leukemia L1210; Leukemia, Experimental; Lung Neoplasms; Male; Melanoma, Experimental; Mice; Mice, Nude; Molecular Structure; Neoplasm Transplantation; Pancreatic Neoplasms; Sarcoma, Experimental

1991
Phorbol ester-stimulated insulin secretion by RINm5F insulinoma cells is linked with membrane depolarization and an increase in cytosolic free Ca2+ concentration.
    The Journal of biological chemistry, 1989, Feb-15, Volume: 264, Issue:5

    In studying the regulation of insulin secretion by phorbol esters, we examined their effects on the cytosolic free Ca2+ concentration ([Ca2+]i), using the Ca2+ indicator fura-2 in the rat insulin-secreting beta-cell line RINm5F. [Ca2+]i was measured in parallel with the rate of insulin release. 50 nM 12-O-tetradecanoylphorbol-13-acetate (TPA), which may act via protein kinase C, stimulated insulin release and caused an increase in [Ca2+]i. Ca2+-free conditions eliminated the increase in [Ca2+]i and resulted in a reduced stimulation of insulin release by TPA. The Ca2+ channel blocker nitrendipine (300 nM) inhibited both the increase in [Ca2+]i and the increased rate of insulin secretion. Another phorbol ester, 4 beta-phorbol 12,13-didecanoate, which activates protein kinase C, also induced an increase in [Ca2+]i and in the rate of insulin release, while 4 alpha-phorbol 12,13-didecanoate, which fails to stimulate protein kinase C, was without effect. Further studies with bis-oxonol as an indicator of membrane potential showed that TPA depolarized the beta-cell plasma membrane. From these results, it is concluded that TPA depolarizes the plasma membrane, induces the opening of Ca2+ channels in the RINm5F beta-cell plasma membrane, increases [Ca2+]i, and results in insulin secretion. The action of TPA was next compared with that of a depolarizing concentration of KC1 (25 mM), which stimulates insulin secretion simply by opening Ca2+ channels. TPA consistently elicited less depolarization, a smaller rise of [Ca2+]i, but a greater release of insulin than KC1. Therefore an additional action of TPA is suggested, which potentiates the action of the elevated [Ca2+]i on insulin secretion.

    Topics: Adenoma, Islet Cell; Animals; Benzofurans; Calcium; Carbachol; Cell Line; Cell Membrane; Cytosol; Ethers; Fluorescent Dyes; Fura-2; Insulinoma; Ionomycin; Kinetics; Membrane Potentials; Pancreatic Neoplasms; Potassium Chloride; Rats; Tetradecanoylphorbol Acetate

1989