Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Nat Commun ; 12(1): 4671, 2021 08 03.
Article in English | MEDLINE | ID: mdl-34344863

ABSTRACT

Triple negative breast cancer (TNBC) remains challenging because of heterogeneous responses to chemotherapy. Incomplete response is associated with a greater risk of metastatic progression. Therefore, treatments that target chemotherapy-resistant TNBC and enhance chemosensitivity would improve outcomes for these high-risk patients. Breast cancer stem cell-like cells (BCSCs) have been proposed to represent a chemotherapy-resistant subpopulation responsible for tumor initiation, progression and metastases. Targeting this population could lead to improved TNBC disease control. Here, we describe a novel multi-kinase inhibitor, 108600, that targets the TNBC BCSC population. 108600 treatment suppresses growth, colony and mammosphere forming capacity of BCSCs and induces G2M arrest and apoptosis of TNBC cells. In vivo, 108600 treatment of mice bearing triple negative tumors results in the induction of apoptosis and overcomes chemotherapy resistance. Finally, treatment with 108600 and chemotherapy suppresses growth of pre-established TNBC metastases, providing additional support for the clinical translation of this agent to clinical trials.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Drug Resistance, Neoplasm/drug effects , Neoplastic Stem Cells/drug effects , Nitrobenzenes/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Thiazines/therapeutic use , Triple Negative Breast Neoplasms/drug therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Casein Kinase II/antagonists & inhibitors , Casein Kinase II/chemistry , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Mice , Neoplastic Stem Cells/pathology , Nitrobenzenes/chemistry , Nitrobenzenes/pharmacology , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/chemistry , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/chemistry , Thiazines/chemistry , Thiazines/pharmacology , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays , Dyrk Kinases
2.
Mol Cell ; 79(1): 180-190.e4, 2020 07 02.
Article in English | MEDLINE | ID: mdl-32619468

ABSTRACT

Rigosertib is a styryl benzyl sulfone that inhibits growth of tumor cells and acts as a RAS mimetic by binding to Ras binding domains of RAS effectors. A recent study attributed rigosertib's mechanism of action to microtubule binding. In that study, rigosertib was obtained from a commercial vendor. We compared the purity of clinical-grade and commercially sourced rigosertib and found that commercially sourced rigosertib contains approximately 5% ON01500, a potent inhibitor of tubulin polymerization. Clinical-grade rigosertib, which is free of this impurity, does not exhibit tubulin-binding activity. Cell lines expressing mutant ß-tubulin have also been reported to be resistant to rigosertib. However, our study showed that these cells failed to proliferate in the presence of rigosertib at concentrations that are lethal to wild-type cells. Rigosertib induced a senescence-like phenotype in the small percentage of surviving cells, which could be incorrectly scored as resistant using short-term cultures.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation , Glycine/analogs & derivatives , Lung Neoplasms/pathology , Sulfones/pharmacology , Tubulin/metabolism , Drug Contamination , Drug Resistance, Neoplasm , Glycine/pharmacology , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Mutation , Tubulin/chemistry , Tubulin/genetics , Tumor Cells, Cultured
3.
Oncotarget ; 10(20): 1932-1942, 2019 Mar 08.
Article in English | MEDLINE | ID: mdl-30956775

ABSTRACT

Aberrant signaling triggered by oncogenic or hyperactive RAS proteins contributes to the malignant phenotypes in a significant percentage of myeloid malignancies. Of these, juvenile myelomonocytic leukemia (JMML), an aggressive childhood cancer, is largely driven by mutations in RAS genes and those that encode regulators of these proteins. The Mx1-cre kras+/G12D mouse model mirrors several key features of this disease and has been used extensively to determine the utility and mechanism of small molecule therapeutics in the context of RAS-driven myeloproliferative disorders. Treatment of disease-bearing KRASG12D mice with rigosertib (RGS), a small molecule RAS mimetic that is in phase II and III clinical trials for MDS and AML, decreased the severity of leukocytosis and splenomegaly and extended their survival. RGS also increased the frequency of HSCs and rebalanced the ratios of myeloid progenitors. Further analysis of KRASG12D HSPCs in vitro revealed that RGS suppressed hyperproliferation in response to GM-CSF and inhibited the phosphorylation of key RAS effectors. Together, these data suggest that RGS might be of clinical benefit in RAS-driven myeloid disorders.

4.
Oncotarget ; 9(102): 37753-37765, 2018 Dec 28.
Article in English | MEDLINE | ID: mdl-30701029

ABSTRACT

Overexpression and constitutive activation of CYCLIN D1 and Casein Kinase 2 are common features of many hematologic malignancies, including mantle cell lymphoma (MCL) and leukemias such as T-cell acute lymphoblastic leukemia (T-ALL). Although both CK2 and CDK4 inhibitors have shown promising results against these tumor types, none of these agents have achieved objective responses in the clinic as monotherapies. Because both proteins play key roles in these and other hematological malignancies, we have analyzed the therapeutic potential of ON108110, a novel dual specificity ATP-competitive inhibitor of protein kinase CK2 as well as CDK4/6 in MCL and T-ALL. We show that in cell growth inhibition assays, MCL and T-ALL cell lines exhibited increased sensitivity to ON108110 when compared to other tumor types. Treatment with ON108110 reduced the level of phosphorylated RB-family proteins. In addition, ON108110 treatment resulted in concentration dependent inhibition of PTEN phosphorylation and a concomitant decrease in PI3K-AKT signaling mediated by CK2. Accordingly, cells treated with ON108110 rapidly accumulated in the G0/G1 stage of the cell cycle as a function of increasing concentration followed by rapid onset of apoptosis. Together, these results indicate that dual inhibition of CK2 and CDK4/6 may be an efficient treatment of MCL and T-ALLs displaying upregulation of CK2/PI3K and CDK4 signaling pathways.

5.
Cell ; 165(3): 643-55, 2016 Apr 21.
Article in English | MEDLINE | ID: mdl-27104980

ABSTRACT

Oncogenic activation of RAS genes via point mutations occurs in 20%-30% of human cancers. The development of effective RAS inhibitors has been challenging, necessitating new approaches to inhibit this oncogenic protein. Functional studies have shown that the switch region of RAS interacts with a large number of effector proteins containing a common RAS-binding domain (RBD). Because RBD-mediated interactions are essential for RAS signaling, blocking RBD association with small molecules constitutes an attractive therapeutic approach. Here, we present evidence that rigosertib, a styryl-benzyl sulfone, acts as a RAS-mimetic and interacts with the RBDs of RAF kinases, resulting in their inability to bind to RAS, disruption of RAF activation, and inhibition of the RAS-RAF-MEK pathway. We also find that ribosertib binds to the RBDs of Ral-GDS and PI3Ks. These results suggest that targeting of RBDs across multiple signaling pathways by rigosertib may represent an effective strategy for inactivation of RAS signaling.


Subject(s)
Glycine/analogs & derivatives , RNA-Binding Proteins/chemistry , Signal Transduction/drug effects , Sulfones/pharmacology , Amino Acid Sequence , Animals , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Cell Transformation, Neoplastic/drug effects , Crystallography, X-Ray , Dimerization , Glycine/administration & dosage , Glycine/chemistry , Glycine/pharmacology , Humans , MAP Kinase Signaling System , Mice , Mice, Nude , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Pancreatic Neoplasms/drug therapy , Phosphorylation , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins B-raf/chemistry , Proto-Oncogene Proteins B-raf/metabolism , RNA-Binding Proteins/metabolism , Sequence Alignment , Sulfones/administration & dosage , Sulfones/chemistry , ras Proteins/metabolism , Polo-Like Kinase 1
6.
Bioorg Med Chem ; 24(4): 521-44, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26762835

ABSTRACT

Several families of protein kinases have been shown to play a critical role in the regulation of cell cycle progression, particularly progression through mitosis. These kinase families include the Aurora kinases, the Mps1 gene product and the Polo Like family of protein kinases (PLKs). The PLK family consists of five members and of these, the role of PLK1 in human cancer is well documented. PLK2 (SNK), which is highly homologous to PLK1, has been shown to play a critical role in centriole duplication and is also believed to play a regulatory role in the survival pathway by physically stabilizing the TSC1/2 complex in tumor cells under hypoxic conditions. As a part of our research program, we have developed a library of novel ATP mimetic chemotypes that are cytotoxic against a panel of cancer cell lines. We show that one of these chemotypes, the 6-arylsulfonyl pyridopyrimidinones, induces apoptosis of human tumor cell lines in nanomolar concentrations. The most potent of these compounds, 7ao, was found to be a highly specific inhibitor of PLK2 when profiled against a panel of 288 wild type, 55 mutant and 12 lipid kinases. Here, we describe the synthesis, structure activity relationship, in vitro kinase specificity and biological activity of the lead compound, 7ao.


Subject(s)
Drug Discovery , Indoles/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyrimidinones/pharmacology , Dose-Response Relationship, Drug , Humans , Indoles/chemical synthesis , Indoles/chemistry , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/metabolism , Pyrimidinones/chemical synthesis , Pyrimidinones/chemistry , Structure-Activity Relationship
7.
Eur J Cancer ; 50(11): 1982-92, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24835034

ABSTRACT

Heat shock protein (Hsp) 90 is a key component of the super-chaperone complex that maintains functionally active conformation of various client proteins. Many of these client proteins regulate important nodal points in multiple signalling pathways that promote cancer cell growth and survival. Inhibitors of Hsp90, therefore, have the potential of functioning as anti-cancer agents with pleiotropic effects. Identification of novel Hsp90 inhibitors with more favourable pharmacological properties is a priority in cancer therapy. To achieve this goal, we screened a compound library using a biochemical assay based on refolding of denatured firefly luciferase. The assay revealed high sensitivity, reliability and reproducibility with a Z-factor of 0.81 ± 0.17. Six Hsp90 inhibitory compounds identified by this screening with IC50 values between 1.0 and 6 µM were further characterised for anti-proliferative activity by Cell Titer-Blue Cell Viability Assay using multiple tumour cell lines. Of particular interest was ONO4140 with lowest GI50 values in three different cancer cell lines viz; DU-145, BT-474 and K562 cell lines. This study also revealed that short-term exposure of tumour cells with ONO4140 is sufficient to inhibit the catalytic activity of Hsp90, evaluated through disruption of Hsp90-p23 association by immunoprecipitation. This short term exposure appears to initiate events like degradation of Hsp90 client proteins such as ErbB2/Her-2 and Akt with concomitant inhibition of survival signalling leading to the apoptotic death of tumour cells as seen by western blotting and Caspase Glow-3,7 assay. The study also reveals that apoptosis following Hsp90 inhibition with ONO4140 occurs via Caspase9-Caspase3 intrinsic apoptotic pathway, a process that is likely triggered by inactivation of Akt. In conclusion, we have identified a novel class of synthetic compounds which show potent Hsp90 inhibitory action in preclinical studies. The discovery of this novel class of synthetic Hsp90 inhibitors with simple chemical backbone allows us to conduct further structural modifications to improve their potency and pharmacokinetic properties for use in cancer therapy.


Subject(s)
Chalcones/pharmacology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Sulfones/pharmacology , Animals , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Female , HSP90 Heat-Shock Proteins/chemistry , HSP90 Heat-Shock Proteins/metabolism , Humans , K562 Cells , Luciferases, Firefly/chemistry , Luciferases, Firefly/metabolism , Rabbits , Signal Transduction
8.
J Med Chem ; 57(3): 578-99, 2014 Feb 13.
Article in English | MEDLINE | ID: mdl-24417566

ABSTRACT

The success of imatinib, a BCR-ABL inhibitor for the treatment of chronic myelogenous leukemia, has created a great impetus for the development of additional kinase inhibitors as therapeutic agents. However, the complexity of cancer has led to recent interest in polypharmacological approaches for developing multikinase inhibitors with low toxicity profiles. With this goal in mind, we analyzed more than 150 novel cyano pyridopyrimidine compounds and identified structure-activity relationship trends that can be exploited in the design of potent kinase inhibitors. One compound, 8-cyclopentyl-2-[4-(4-methyl-piperazin-1-yl)-phenylamino]-7-oxo-7,8-dihydro-pyrido[2,3-d]pyrimidine-6-carbonitrile (7x), was found to be the most active, inducing apoptosis of tumor cells at a concentration of approximately 30-100 nM. In vitro kinase profiling revealed that 7x is a multikinase inhibitor with potent inhibitory activity against the CDK4/CYCLIN D1 and ARK5 kinases. Here, we report the synthesis, structure-activity relationship, kinase inhibitory profile, in vitro cytotoxicity, and in vivo tumor regression studies by this lead compound.


Subject(s)
Antineoplastic Agents/chemical synthesis , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Pyridines/chemical synthesis , Pyrimidines/chemical synthesis , Repressor Proteins/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Drug Screening Assays, Antitumor , Female , Heterografts , Humans , Mice , Mice, Nude , Molecular Docking Simulation , Neoplasm Transplantation , Protein Kinases , Pyridines/chemistry , Pyridines/pharmacology , Pyrimidines/chemistry , Pyrimidines/pharmacology , Structure-Activity Relationship
9.
J Med Chem ; 56(13): 5562-86, 2013 Jul 11.
Article in English | MEDLINE | ID: mdl-23750455

ABSTRACT

A series of novel (E)-N-aryl-2-arylethenesulfonamides (6) were synthesized and evaluated for their anticancer activity. Some of the compounds in this series showed potent cytotoxicity against a wide spectrum of cancer cell-lines (IC50 values ranging from 5 to 10 nM) including all drug resistant cell-lines. Nude mice xenograft assays with compound (E)-N-(3-amino-4-methoxyphenyl)-2-(2',4',6'-trimethoxyphenyl)ethenesulfonamide (6t) showed dramatic reduction in tumor size, indicating their in vivo potential as anticancer agents. A preliminary drug development study with compound 6t is predicted to have increased blood-brain barrier permeability relative to many clinically used antimitotic agents. Mechanistic studies indicate that 6t and some other analogues disrupted microtubule formation, formation of mitotic spindles, and arrest of cells in mitotic phase. Compound 6t inhibited purified tubulin polymerization in vitro and in vivo and circumvented drug resistance mediated by P-glycoprotein. Compound 6t specifically competed with colchicine binding to tubulin and with similar avidity as podophylltoxin, indicating its binding site on tubulin.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Design , Microtubules/drug effects , Neoplasms/drug therapy , Sulfonamides/pharmacology , Xenograft Model Antitumor Assays , Administration, Oral , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacokinetics , Biological Availability , Blood-Brain Barrier/metabolism , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Drug Resistance, Neoplasm/drug effects , HCT116 Cells , Humans , K562 Cells , MCF-7 Cells , Mice , Mice, Nude , Microtubules/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Polymerization/drug effects , Sulfonamides/chemical synthesis , Sulfonamides/pharmacokinetics , Tubulin/metabolism , Tumor Burden/drug effects
10.
PLoS One ; 8(3): e58355, 2013.
Article in English | MEDLINE | ID: mdl-23505494

ABSTRACT

Development of radio-protective agents that are non-toxic is critical in light of ever increasing threats associated with proliferation of nuclear materials, terrorism and occupational risks associated with medical and space exploration. In this communication, we describe the discovery, characterization and mechanism of action of ON01210.Na, which effectively protects mouse and human bone marrow cells from radiation-induced damage both in vitro and in vivo. Our results show that treatment of normal fibroblasts with ON01210.Na before and after exposure to ionizing radiation provides dose dependent protection against radiation-induced damage. Treatment of mice with ON01210.Na prior to radiation exposure was found to result in a more rapid recovery of their hematopoietic system. The mechanistic studies described here show that ON01210.Na manifests its protective effects through the up-regulation of PI3-Kinase/AKT pathways in cells exposed to radiation. These results suggest that ON 01210.Na is a safe and effective radioprotectant and could be a novel agent for use in radiobiological disasters.


Subject(s)
DNA Damage/drug effects , DNA Damage/radiation effects , Proto-Oncogene Proteins c-akt/metabolism , Radiation-Protective Agents/pharmacology , Signal Transduction/drug effects , Signal Transduction/radiation effects , Sulfonamides/pharmacology , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/radiation effects , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cell Survival , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/radiation effects , Humans , Mice , Small Molecule Libraries
11.
Org Biomol Chem ; 11(12): 1964-77, 2013 Mar 28.
Article in English | MEDLINE | ID: mdl-23386308

ABSTRACT

A stereoselective and efficient method for free radical addition of benzyl thiol to aryl acetylene in the presence of Et3B-hexane has been developed for the synthesis of (Z) and (E)-styryl benzyl sulfides where base catalyzed hydrothiolations have failed. The scope of this reaction was successfully extended for the synthesis of (E)-ON 01910·Na, a phase III clinical stage anti-cancer agent and its inactive geometrical isomer (Z)-ON 01910·Na. It is interesting to note that all the E-isomers synthesized have shown better cytotoxicity profile on cancer cells compared to the Z-isomers.


Subject(s)
Alkynes/pharmacology , Antineoplastic Agents/pharmacology , Glycine/analogs & derivatives , Sulfhydryl Compounds/chemistry , Sulfones/pharmacology , Alkynes/chemical synthesis , Alkynes/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Clinical Trials, Phase III as Topic , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Glycine/chemical synthesis , Glycine/chemistry , Glycine/pharmacology , Humans , K562 Cells , Molecular Structure , Stereoisomerism , Structure-Activity Relationship , Sulfones/chemical synthesis , Sulfones/chemistry
12.
J Pharm Biomed Anal ; 75: 138-44, 2013 Mar 05.
Article in English | MEDLINE | ID: mdl-23261806

ABSTRACT

ON 013100, (E)-2,4,6-trimethoxystyryl-3-hydroxy-4-methoxybenzyl sulfone, is a potent kinase inhibitor whose phosphate form is in Phase I clinical trials in lymphoma and acute lymphoid leukemia. The objectives were to: (a) investigate the possible presence of the glucuronide metabolite of the drug in two representative colon cancer cell lines, a drug resistant (colo-205) and a drug sensitive (colo-320); (b) quantify the glucuronide metabolite and the unchanged drug in the cells after treatment with ON 013100. The glucuronide was synthesized and a selective LC/MS/MS method was developed and validated for the characterization and quantification of the metabolite. The glucuronide metabolite (570.6 Da) was found in the drug-resistant cells upon a 1h incubation with ON 013100 (20 µg/ml). After treatment with the drug, the concentration of the metabolite gradually decreased from 0.84 µg/ml at 0 h through 0.21 µg/ml at 6h to below detection limit of 8.0 ng/ml at 9 h. No glucuronide metabolite was detected in the drug-sensitive cells. The concentrations of intact ON 013100 in the drug-resistant cells gradually decreased from 0.41 µg/ml (0 h) to 0.06 µg/ml (9 h). The corresponding concentrations of the intact drug in the drug-sensitive cells were from 2.88 µg/ml to 0.94 µg/ml.


Subject(s)
Antineoplastic Agents/metabolism , Benzyl Compounds/metabolism , Colonic Neoplasms/metabolism , Glucuronides/analysis , Protein Kinase Inhibitors/metabolism , Styrenes/metabolism , Antineoplastic Agents/analysis , Antineoplastic Agents/pharmacology , Benzyl Compounds/analysis , Benzyl Compounds/pharmacology , Cell Line, Tumor , Chromatography, High Pressure Liquid , Colonic Neoplasms/drug therapy , Drug Resistance, Neoplasm , Glucuronides/chemistry , Glucuronides/metabolism , Humans , Kinetics , Limit of Detection , Metabolic Detoxication, Phase I , Molecular Weight , Protein Kinase Inhibitors/analysis , Protein Kinase Inhibitors/pharmacology , Spectrometry, Mass, Electrospray Ionization , Stereoisomerism , Styrenes/analysis , Styrenes/pharmacology , Sulfones , Tandem Mass Spectrometry
13.
J Med Chem ; 55(11): 5174-87, 2012 Jun 14.
Article in English | MEDLINE | ID: mdl-22587519

ABSTRACT

Tubulin, the major structural component of microtubules, is a target for the development of anticancer agents. A series of (Z)-1-aryl-3-arylamino-2-propen-1-one (10) were synthesized and evaluated for antiproliferative activity in cell-based assay. The most active compound (Z)-1-(2-bromo-3,4,5-trimethoxyphenyl)-3-(3-hydroxy-4-methoxyphenylamino)prop-2-en-1-one (10ae) was tested in 20 tumor cell lines including multidrug resistant phenotype and was found to induce apoptosis in all these cell lines with similar GI(50) values. Flow cytometry studies showed that 10ae arrested the cells in G2/M phase of cell cycle. In addition to G2/M block, these compounds caused microtubule stabilization like paclitaxel and induced apoptosis via activation of the caspase family. The observations made in this investigation demonstrate that (Z)-1-Aryl-3-arylamino-2-propen-1-one (10) represents a new class of microtubule-stabilizing agents.


Subject(s)
Alkenes/chemical synthesis , Aminophenols/chemical synthesis , Antineoplastic Agents/chemical synthesis , Tubulin Modulators/chemical synthesis , Tubulin/metabolism , Alkenes/chemistry , Alkenes/pharmacology , Aminophenols/chemistry , Aminophenols/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Division/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , G2 Phase/drug effects , Humans , Microtubules/drug effects , Microtubules/ultrastructure , Polymerization , Spindle Apparatus/drug effects , Spindle Apparatus/ultrastructure , Stereoisomerism , Structure-Activity Relationship , Tubulin Modulators/chemistry , Tubulin Modulators/pharmacology
14.
J Med Chem ; 54(18): 6254-76, 2011 Sep 22.
Article in English | MEDLINE | ID: mdl-21812421

ABSTRACT

Cyclin D proteins are elevated in many cancer cells, and targeted deletion of cyclin D1 gene in the mammary tissues protects mice from breast cancer. Accordingly, there is an increasing awareness of this novel nonenzymatic target for cancer therapeutics. We have developed novel, nonalkylating styrylbenzylsulfones that induce cell death in wide variety of cancer cells without affecting the proliferation and survival of normal cells. The development of derivatized styrylbenzylsulfones followed logically from a tumor cell cytotoxicity screen performed in our laboratory that did not have an a priori target profile. Modifications of some of the precursor molecules led to lead optimization with regard to tumor cell cytotoxicity. In this report we describe the synthesis and structure-activity relationships of novel, nonalkylating (E)-styrylbenzylsulfones and the development of the novel anticancer agent sodium (E)-2-{2-methoxy-5-[(2',4',6'-trimethoxystyrylsulfonyl)methyl]phenylamino}acetate (ON 01910.Na), which is in phase III trials for myelodysplastic syndromes (MDS) associated with aberrant expression of cyclin D proteins.


Subject(s)
Antineoplastic Agents/chemical synthesis , Glycine/analogs & derivatives , Protein Kinase Inhibitors/chemical synthesis , Sulfones/chemical synthesis , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis , Biological Availability , Cell Line, Tumor , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Female , Glycine/chemical synthesis , Glycine/chemistry , Glycine/pharmacology , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Stereoisomerism , Structure-Activity Relationship , Sulfones/chemistry , Sulfones/pharmacology , Transplantation, Heterologous
15.
Genes Cancer ; 1(4): 331-45, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20717479

ABSTRACT

Here we report the discovery of ON044580, an α-benzoyl styryl benzyl sulfide that possesses potent inhibitory activity against two unrelated kinases, JAK2 and BCR-ABL, and exhibits cytotoxicity to human tumor cells derived from chronic myelogenous leukemia (CML) and myelodysplasia (MDS) patients or cells harboring a mutant JAK2 kinase. This novel spectrum of activity is explained by the non-ATP-competitive inhibition of JAK2 and BCR-ABL kinases. ON044580 inhibits mutant JAK2 kinase and the proliferation of JAK2(V617F)-positive leukemic cells and blocks the IL-3-mediated phosphorylation of JAK2 and STAT5. Interestingly, this compound also directly inhibits the kinase activity of both wild-type and imatinib-resistant (T315I) forms of the BCR-ABL kinase. Finally, ON044580 effectively induces apoptosis of imatinib-resistant CML patient cells. The apparently unrelated JAK2 and BCR-ABL kinases share a common substrate, STAT5, and such substrate competitive inhibitors represent an alternative therapeutic strategy for development of new inhibitors. The novel mechanism of kinase inhibition exhibited by ON044580 renders it effective against mutant forms of kinases such as the BCR-ABL(T315I) and JAK2(V617F). Importantly, ON044580 selectively reduces the number of aneuploid cells in primary bone marrow samples from monosomy 7 MDS patients, suggesting another regulatory cascade amenable to this agent in these aberrant cells. Data presented suggest that this compound could have multiple therapeutic applications including monosomy 7 MDS, imatinib-resistant CML, and myeloproliferative neoplasms that develop resistance to ATP-competitive agents.

16.
Bioorg Med Chem ; 18(6): 2317-2326, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20188579

ABSTRACT

Novel (E)-alpha-benzylthio chalcones are reported with preliminary in vitro activity data indicating that several of them are potent inhibitors (comparable to imatinib, the reference compound) of BCR-ABL phosphorylation in leukemic K562 cells, known to express high levels of BCR-ABL. The ability of such compounds to significantly inhibit K562 cell proliferation suggests that this scaffold could be a promising lead for the development of anticancer agents that are able to block BCR-ABL phosphorylation in leukemic cells.


Subject(s)
Chalcones/chemical synthesis , Chalcones/pharmacology , Drug Design , Fusion Proteins, bcr-abl/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Cell Proliferation/drug effects , Chalcones/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Fusion Proteins, bcr-abl/metabolism , Humans , K562 Cells , Molecular Structure , Phosphorylation , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein-Tyrosine Kinases/metabolism , Stereoisomerism , Structure-Activity Relationship , Tumor Cells, Cultured
17.
Cancer Chemother Pharmacol ; 65(1): 177-86, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19466411

ABSTRACT

PURPOSE: ON 01910.Na is a novel targeted anti-cancer agent under clinical investigation in Phase I and II trials. The purpose of this research was to evaluate the pharmacokinetic profile of ON 01910.Na across several species, and to evaluate the effects of protein binding and duration of exposure on its in vitro cytotoxic activity. METHODS: Data were collated from several preclinical investigations, where the plasma disposition and tissue distribution of ON 01910.Na were assessed after administration (10-150 mg/kg, IP or IV) to several species (mouse, rat, and dog). Plasma protein binding was assessed using ultrafiltration. Cytotoxic activity of ON 01910.Na was determined in DU145 cells, and activity was correlated to unbound drug concentration and the duration of exposure. RESULTS: ON 01910.Na exhibits extensive plasma protein binding and the compound displays rapid elimination from the circulation in all three animal species (t(1/2) range 0.404-0.870 h). Tissue distribution studies in mice revealed highest drug accumulation in the liver, followed by the kidneys. ON 01910.Na is not extensively metabolized in vivo and urinary excretion is predominant at higher doses. ON 01910.Na cytotoxicity in DU145 cells was adversely affected by protein binding in the incubation medium. Drug cytotoxicity was greatly enhanced upon extending the duration of exposure at reduced drug concentrations. CONCLUSIONS: Due to the short half-life and rapid clearance of the drug, administration of ON 01910.Na by continuous IV infusion is a likely treatment option for cancer patients.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Glycine/analogs & derivatives , Kidney/metabolism , Liver/metabolism , Prostatic Neoplasms/drug therapy , Sulfones/pharmacokinetics , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Dogs , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Drug Screening Assays, Antitumor , Female , Glycine/administration & dosage , Glycine/pharmacokinetics , Glycine/pharmacology , Half-Life , Humans , Infusions, Intravenous , Male , Mice , Prostatic Neoplasms/metabolism , Protein Binding , Rats , Species Specificity , Sulfones/administration & dosage , Sulfones/pharmacology , Tissue Distribution
18.
J Med Chem ; 51(1): 86-100, 2008 Jan 10.
Article in English | MEDLINE | ID: mdl-18088089

ABSTRACT

Cell cycle progression is regulated by cyclins and cyclin-dependent kinases, which are formed at specific stages of the cell cycle and regulate the G1/S and G2/M phase transitions, employing a series of "checkpoints" governed by phosphorylation of their substrates. Tumor development is associated with the loss of these checkpoint controls, and this provides an approach for the development of therapeutic agents that can specifically target tumor cells. Here, we describe the synthesis and SAR of a novel group of cytotoxic molecules that selectively induce growth arrest of normal cells in the G1 phase while inducing a mitotic arrest of tumor cells resulting in selective killing of tumor cell populations with little or no effect on normal cell viability. The broad spectrum of antitumor activity in vitro and xenograft models, lack of in vivo toxicity, and drug resistance suggest potential for use of these agents in cancer therapy.


Subject(s)
Antineoplastic Agents/chemical synthesis , Styrenes/chemical synthesis , Sulfones/chemical synthesis , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Drug Design , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Female , Humans , Mice , Mice, Nude , Rats , Stereoisomerism , Structure-Activity Relationship , Styrenes/chemistry , Styrenes/pharmacology , Sulfones/chemistry , Sulfones/pharmacology , Toxicity Tests
19.
Nucl Med Biol ; 34(4): 371-81, 2007 May.
Article in English | MEDLINE | ID: mdl-17499726

ABSTRACT

UNLABELLED: Disintegrins, which contain an Arg-Gly-Asp sequence in their binding domains are antagonists of integrins such as alphavbeta3. The purpose of this study was to compare a range of disintegrins with different integrin selectivities for their binding behavior in vitro to vascular endothelial cells bearing alphavbeta3 and to cultured tumor cells which express alphavbeta3. METHODS: Five disintegrins (bitistatin, kistrin, flavoridin, VLO4 and echistatin) and a cyclic pentapeptide, c[RGDyK], were radiolabeled with (99m)Tc and tested for binding to cells in vitro. RESULTS: (99m)Tc-Kistrin, flavoridin and VLO4 had the highest binding, (99m)Tc-echistatin had moderate binding, and (99m)Tc-bitistatin and (99m)Tc-c[RGDyK] had low binding to cells. The observed binding was attributed to alphavbeta3 to various extents: echistatin, bitistatin>kistrin>flavoridin>VLO4. Cancer cells internalized bound disintegrins after binding, but endothelial cells did not. After binding to endothelial cells, (99m)Tc-kistrin was not displaced by competing peptide or plasma proteins. CONCLUSIONS: These data suggest that radiolabeled kistrin, flavoridin and VLO4 may have advantages over labeled bitistatin and small cyclic peptides for targeting alphavbeta3 in vivo. Since receptor-bound radioligand is not internalized by endothelial cells, disintegrins may provide an advantage for targeting alphavbeta3 on vasculature because they bind strongly to surface receptors and are not readily displaced.


Subject(s)
Disintegrins/chemical synthesis , Integrin alphaVbeta3/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Organotechnetium Compounds/chemical synthesis , Radiopharmaceuticals , Binding, Competitive/drug effects , Cell Line, Tumor , Disintegrins/pharmacokinetics , Endothelial Cells/metabolism , Humans , Isotope Labeling , Ligands , Organotechnetium Compounds/pharmacokinetics , Radioligand Assay , Radiopharmaceuticals/pharmacokinetics , Receptors, Cell Surface/metabolism
20.
Bioorg Med Chem ; 13(9): 3141-7, 2005 May 02.
Article in English | MEDLINE | ID: mdl-15809149

ABSTRACT

A series of novel coumarin carboxamides were synthesized, and their tumor cell cytotoxic activity was investigated. These compounds specifically inhibited the growth of cancer cells that have a high level of ErbB-2 expression. Immunoprecipitation analysis of the cell lysates prepared from carboxamide treated cancer cells showed the inhibition of ErbB-2 phosphorylation suggesting the interaction of these compounds with ErbB-2 receptor. The down regulation of the kinase activity was further confirmed by performing in vitro kinase assay with recombinant ErbB-2 incubated with carboxamides. The inhibition of ErbB-2 phosphorylation correlated with down-regulation of ERK1 MAP kinase activation that is involved in proliferative signaling pathway. Furthermore, the cell-killing activity of many of these inhibitors is restricted to tumor cells with no demonstrable cytotoxicity against normal human fibroblasts suggesting that these compounds are tumor-specific.


Subject(s)
Amides/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Coumarins/chemistry , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Receptor, ErbB-2/drug effects , Amides/chemical synthesis , Amides/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Coumarins/chemical synthesis , Female , Fibroblasts/drug effects , Humans , Mitogen-Activated Protein Kinase 3/metabolism , Molecular Structure , Receptor, ErbB-2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...