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










Publication year range
1.
Bioorg Med Chem Lett ; 28(5): 934-941, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29433928

ABSTRACT

Microtubule-targeting agents are important tools in cancer treatment. Generating novel microtubule targeting agents with novel pharmacology could dramatically expand the utility of this class of drugs. Here we characterize the pharmacology of recently described small molecule microtubule polymerization inhibitors. Pharmacokinetic experiments show oral bioavailability through gastric absorption. In vitro assays designed to predict absorption, distribution, metabolism, and excretion (ADME) and safety reveal a scaffold that is metabolically stable, evades P-glycoprotein, does not inhibit CYP enzymes, occurs as a significant free fraction in serum, and has exceptionally high cellular permeability. Together with in vivo efficacy models, pharmacology supports further development as a treatment for solid tumors.


Subject(s)
Amides/pharmacology , Microtubules/drug effects , Pyridines/pharmacology , Pyrimidines/pharmacology , Amides/chemical synthesis , Amides/chemistry , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Dogs , Dose-Response Relationship, Drug , Humans , Mice , Microsomes, Liver/chemistry , Microsomes, Liver/metabolism , Microtubules/metabolism , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Polymerization/drug effects , Pyridines/chemical synthesis , Pyridines/chemistry , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Rats , Structure-Activity Relationship
2.
Bioorg Med Chem Lett ; 27(17): 3992-4000, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28780159

ABSTRACT

Stimulation of cultured epithelial cells with scatter factor/hepatocyte growth factor (HGF) results in individual cells detaching and assuming a migratory and invasive phenotype. Epithelial scattering recapitulates cancer progression and studies have implicated HGF signaling as a driver of cancer metastasis. Inhibitors of HGF signaling have been proposed to act as anti-cancer agents. We previously screened a small molecule library for compounds that block HGF-induced epithelial scattering. Most hits identified in this screen exhibit anti-mitotic properties. Here we assess the biological mechanism of a compound that blocks HGF-induced scattering with limited anti-mitotic activity. Analogs of this compound have one of two distinct activities: inhibiting either cell migration or cell proliferation with cell cycle arrest in G2/M. Each activity bears unique structure-activity relationships. The mechanism of action of anti-mitotic compounds is by inhibition of microtubule polymerization; these compounds entropically and enthalpically bind tubulin in the colchicine binding site, generating a conformational change in the tubulin dimer.


Subject(s)
Amides/pharmacology , Antineoplastic Agents/pharmacology , Epithelial Cells/drug effects , Hepatocyte Growth Factor/antagonists & inhibitors , Pyridines/pharmacology , Pyrimidines/pharmacology , Amides/chemical synthesis , Amides/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Epithelial Cells/metabolism , Hepatocyte Growth Factor/metabolism , Humans , MCF-7 Cells , Molecular Structure , Pyridines/chemical synthesis , Pyridines/chemistry , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Structure-Activity Relationship
3.
Oncotarget ; 8(63): 107206-107222, 2017 Dec 05.
Article in English | MEDLINE | ID: mdl-29291023

ABSTRACT

More effective treatment options for elderly acute myeloid leukemia (AML) patients are needed as only 25-50% of patients respond to standard-of-care therapies, response duration is typically short, and disease progression is inevitable even with some novel therapies and ongoing clinical trials. Anti-apoptotic BCL-2 family inhibitors, such as venetoclax, are promising therapies for AML. Nonetheless, resistance is emerging. We demonstrate that venetoclax combined with cyclin-dependent kinase (CDK) inhibitor alvocidib is potently synergistic in venetoclax-sensitive and -resistant AML models in vitro, ex vivo and in vivo. Alvocidib decreased MCL-1, and/or increased pro-apoptotic proteins such as BIM or NOXA, often synergistically with venetoclax. Over-expression of BCL-XL diminished synergy, while knock-down of BIM almost entirely abrogated synergy, demonstrating that the synergistic interaction between alvocidib and venetoclax is primarily dependent on intrinsic apoptosis. CDK9 inhibition predominantly mediated venetoclax sensitization, while CDK4/6 inhibition with palbociclib did not potentiate venetoclax activity. Combined, venetoclax and alvocidib modulate the balance of BCL-2 family proteins through complementary, yet variable mechanisms favoring apoptosis, highlighting this combination as a promising therapy for AML or high-risk MDS with the capacity to overcome intrinsic apoptosis mechanisms of resistance. These results support clinical testing of combined venetoclax and alvocidib for the treatment of AML and advanced MDS.

4.
Nat Chem ; 5(3): 153-5, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23422553
5.
Bioorg Med Chem Lett ; 22(9): 3327-31, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22460033

ABSTRACT

A novel family of potent dual inhibitors of CK2 and the Pim kinases was discovered by modifying the scaffolds of tricyclic Pim inhibitors. Several analogs were active at single digit nanomolar IC(50) values against CK2 and the Pim isoforms Pim-1 and Pim-2. The molecules displayed antiproliferative activity in various cell phenotypes in the low micromolar and submicromolar range, providing an excellent starting point for further drug discovery optimization.


Subject(s)
Antineoplastic Agents/pharmacology , Casein Kinase II/antagonists & inhibitors , Cell Proliferation/drug effects , Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Cell Line, Tumor , Drug Discovery , Humans , Inhibitory Concentration 50 , Neoplasms/drug therapy
6.
Cancer Lett ; 322(1): 113-8, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22387988

ABSTRACT

Ser/Thr protein kinase CK2 regulates multiple processes that play important roles in the sensitivity of cancer to epidermal growth factor receptor targeting therapeutics, including PI3K-Akt-mTOR signaling, Hsp90 activity, and inhibition of apoptosis. We hypothesized that top-down inhibition of EGFR, combined with lateral suppression of multiple oncogenic pathways by targeting CK2, would create a pharmacologic synthetic lethal event and result in an improved cancer therapy compared to EGFR inhibition alone. This hypothesis was tested by combining CX-4945, a first-in-class clinical stage inhibitor of CK2, with the EGFR tyrosine kinase inhibitor, erlotinib, in vitro and in vivo in models of non-small cell lung carcinoma, NCI-H2170, and squamous cell carcinoma, A431. Our results demonstrate that combination of CX-4945 with erlotinib results in enhanced attenuation of the PI3K-Akt-mTOR pathway. We also observed an increase in apoptosis, synergistic killing of cancer cells in vitro, as well as improved antitumor efficacy in vivo. Taken together, these data position CK2 as a valid pharmacologic target for drug combinations and support further evaluation of CX-4945 in combination with EGFR targeting agents.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Casein Kinase II/antagonists & inhibitors , ErbB Receptors/antagonists & inhibitors , Naphthyridines/pharmacology , Neoplasms/drug therapy , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology , Quinazolines/pharmacology , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/physiology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Erlotinib Hydrochloride , Female , Humans , Mice , Neoplasms/pathology , Phenazines
7.
Mol Cancer Ther ; 11(4): 994-1005, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22267551

ABSTRACT

Drug combination therapies are commonly used for the treatment of cancers to increase therapeutic efficacy, reduce toxicity, and decrease the incidence of drug resistance. Although drug combination therapies were originally devised primarily by empirical methods, the increased understanding of drug mechanisms and the pathways they modulate provides a unique opportunity to design combinations that are based on mechanistic rationale. We have identified protein kinase CK2 as a promising therapeutic target for combination therapy, because CK2 regulates not just one but many oncogenic pathways and processes that play important roles in drug resistance, including DNA repair, epidermal growth factor receptor signaling, PI3K/AKT/mTOR signaling, Hsp90 machinery activity, hypoxia, and interleukin-6 expression. In this article, we show that CX-4945, a clinical stage selective small molecule inhibitor of CK2, blocks the DNA repair response induced by gemcitabine and cisplatin and synergizes with these agents in models of ovarian cancer. Mechanistic studies show that the enhanced activity is a result of inactivation of XRCC1 and MDC1, two mediator/adaptor proteins that are essential for DNA repair and that require phosphorylation by CK2 for their function. These data position CK2 as a valid pharmacologic target for intelligent drug combinations and support the evaluation of CX-4945 in combination with gemcitabine and platinum-based chemotherapeutics in the clinical setting.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Casein Kinase II/antagonists & inhibitors , DNA Repair/drug effects , Naphthyridines/pharmacology , Neoplasms/drug therapy , Ovarian Neoplasms/drug therapy , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Cell Line, Tumor , Checkpoint Kinase 2 , Drug Synergism , Female , Humans , Mice , Naphthyridines/administration & dosage , Neoplasms/genetics , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/genetics , Phenazines , Phosphorylation , Random Allocation , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
8.
Bioorg Med Chem Lett ; 22(1): 45-8, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22169261

ABSTRACT

Protein kinase CK2 is a potential drug target for many diseases including cancer and inflammation disorders. The crystal structure of clinical candidate CX-4945 1 with CK2 revealed an indirect interaction with the protein through hydrogen bonding between the NH of the 3-chlorophenyl amine and a water molecule. Herein, we investigate the relevance of this hydrogen bond by preparing several novel tricyclic derivatives lacking a NH moiety at the same position. This SAR study allowed the discovery of highly potent CK2 inhibitors.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Casein Kinase II/antagonists & inhibitors , Quinolines/chemistry , Casein Kinase II/chemistry , Cell Line, Tumor , Chemistry, Pharmaceutical/methods , Crystallography, X-Ray/methods , Drug Design , Drug Screening Assays, Antitumor , Humans , Inhibitory Concentration 50 , Models, Chemical , Models, Molecular , Protein Conformation , Quinolines/chemical synthesis , Structure-Activity Relationship
9.
ACS Med Chem Lett ; 3(2): 135-9, 2012 Feb 09.
Article in English | MEDLINE | ID: mdl-24900437

ABSTRACT

Structure-activity relationship analysis in a series of 3-(5-((2-oxoindolin-3-ylidene)methyl)furan-2-yl)amides identified compound 13, a pan-Pim kinases inhibitor with excellent biochemical potency and kinase selectivity. Compound 13 exhibited in vitro synergy with chemotherapeutics and robust in vivo efficacy in two Pim kinases driven tumor models.

10.
ACS Med Chem Lett ; 3(7): 602-6, 2012 Jul 12.
Article in English | MEDLINE | ID: mdl-24900516

ABSTRACT

Accelerated proliferation of solid tumor and hematologic cancer cells is linked to accelerated transcription of rDNA by the RNA polymerase I (Pol I) enzyme to produce elevated levels of rRNA (rRNA). Indeed, upregulation of Pol I, frequently caused by mutational alterations among tumor suppressors and oncogenes, is required for maintenance of the cancer phenotype and forms the basis for seeking selective inhibitors of Pol I as anticancer therapeutics. 2-(4-Methyl-[1,4]diazepan-1-yl)-5-oxo-5H-7-thia-1,11b-diaza-benzo[c]fluorene-6-carboxylic acid (5-methyl-pyrazin-2-ylmethyl)-amide (CX-5461, 7c) has been identified as the first potent, selective, and orally bioavailable inhibitor of RNA Pol I transcription with in vivo activity in tumor growth efficacy models. The preclinical data support the development of CX-5461 as an anticancer drug with potential for activity in several types of cancer.

11.
Biochem Biophys Res Commun ; 415(1): 163-7, 2011 Nov 11.
Article in English | MEDLINE | ID: mdl-22027148

ABSTRACT

Inflammatory breast cancer is driven by pro-angiogenic and pro-inflammatory cytokines. One of them Interleukin-6 (IL-6) is implicated in cancer cell proliferation and survival, and promotes angiogenesis, inflammation and metastasis. While IL-6 has been shown to be upregulated by several oncogenes, the mechanism behind this phenomenon is not well characterized. Here we demonstrate that the pleotropic Serine/Threonine kinase CK2 is implicated in the regulation of IL-6 expression in a model of inflammatory breast cancer. We used siRNAs targeted toward CK2 and a selective small molecule inhibitor of CK2, CX-4945, to inhibit the expression and thus suppress the secretion of IL-6 in in vitro as well as in vivo models. Moreover, we report that in a clinical trial, CX-4945 was able to dramatically reduce IL-6 levels in plasma of an inflammatory breast cancer patient. Our data shed a new light on the regulation of IL-6 expression and position CX-4945 and potentially other inhibitors of CK2, for the treatment of IL-6-driven cancers and possibly other diseases where IL-6 is instrumental, including rheumatoid arthritis.


Subject(s)
Casein Kinase II/metabolism , Inflammatory Breast Neoplasms/metabolism , Interleukin-6/biosynthesis , Animals , Casein Kinase II/antagonists & inhibitors , Casein Kinase II/genetics , Cell Line, Tumor , Clinical Trials, Phase II as Topic , Female , Humans , Inflammatory Breast Neoplasms/blood , Inflammatory Breast Neoplasms/drug therapy , Interleukin-6/antagonists & inhibitors , Interleukin-6/blood , Mice , Mice, Inbred BALB C , Naphthyridines/therapeutic use , Phenazines , Protein Kinase Inhibitors/therapeutic use , RNA, Small Interfering/genetics , STAT3 Transcription Factor/metabolism
12.
Bioorg Med Chem Lett ; 21(22): 6687-92, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21982499

ABSTRACT

A novel class of pan-Pim kinase inhibitors was designed by modifying the CK2 inhibitor CX-4945. Introduction of a triazole or secondary amide functionality on the C-7 position and 2'-halogenoanilines on C-5 resulted in potent inhibitors of the Pim-1 and Pim-2 isoforms, with many analogs active at single digit nanomolar concentrations. The molecules inhibited the phosphorylation at Serine 112 of the apoptosis effector BAD, and had potent antiproliferative effects on the AML cell line MV-4-11 (IC(50) <30 nM). This work delivers an excellent lead-optimization platform for Pim targeting anticancer therapies.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Naphthyridines/chemistry , Naphthyridines/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Leukemia/drug therapy , Neoplasms/drug therapy , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-pim-1/metabolism , Triazoles/chemistry , Triazoles/pharmacology
13.
Biochemistry ; 50(39): 8478-88, 2011 Oct 04.
Article in English | MEDLINE | ID: mdl-21870818

ABSTRACT

5-(3-Chlorophenylamino)benzo[c][2,6]naphthyridine-8-carboxylic acid (CX-4945), the first clinical stage inhibitor of protein kinase CK2 for the treatment of cancer, is representative of a new class of CK2 inhibitors with K(i) values in the low nanomolar range and unprecedented selectivity versus other kinases. Here we present the crystal structure of the complexes of CX-4945 and two analogues (CX-5011 and CX-5279) with the catalytic subunit of human CK2. Consistent with their ATP-competitive mode of inhibition, all three compounds bind in the active site of CK2 (type I inhibitors). The tricyclic scaffold of the inhibitors superposes on the adenine of ATP, establishing multiple hydrophobic interactions with the binding cavity. The more extended scaffold, as compared to that of ATP, allows the carboxylic function, shared by all three ligands, to penetrate into the deepest part of the active site where it makes interactions with conserved water W1 and Lys-68, thus accounting for the crucial role of this negatively charged group in conferring high potency to this class of inhibitors. The presence of a pyrimidine in CX-5011 and in CX-5279 instead of a pyridine (as in CX-4945) ring is likely to account for the higher specificity of these compounds whose Gini coefficients, calculated by profiling them against panels of 102 and/or 235 kinases, are significantly higher than that of CX-4945 (0.735 and 0.755, respectively, vs 0.615), marking the highest selectivity ever reported for CK2 inhibitors.


Subject(s)
Casein Kinase II/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Catalytic Domain , Cell Survival/drug effects , Crystallography, X-Ray , Humans , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Naphthyridines/chemistry , Naphthyridines/pharmacology , Neoplasms/drug therapy , Phenazines , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/chemistry , Pyrimidines/pharmacology , Quinolines/chemistry , Quinolines/pharmacology
14.
Mol Cell Biochem ; 356(1-2): 37-43, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21755459

ABSTRACT

In this article we describe the preclinical characterization of 5-(3-chlorophenylamino) benzo[c][2,6]naphthyridine-8-carboxylic acid (CX-4945), the first orally available small molecule inhibitor of protein CK2 in clinical trials for cancer. CX-4945 was optimized as an ATP-competitive inhibitor of the CK2 holoenzyme (Ki = 0.38 nM). Iterative synthesis and screening of analogs, guided by molecular modeling, led to the discovery of orally available CX-4945. CK2 promotes signaling in the Akt pathway and CX-4945 suppresses the phosphorylation of Akt as well as other key downstream mediators of the pathway such as p21. CX-4945 induced apoptosis and caused cell cycle arrest in cancer cells in vitro. CX-4945 exhibited a dose-dependent antitumor activity in a xenograft model of PC3 prostate cancer model and was well tolerated. In vivo time-dependent reduction in the phosphorylation of the biomarker p21 at T145 was observed by immunohistochemistry. Inhibition of the newly validated CK2 target by CX-4945 represents a fresh therapeutic strategy for cancer.


Subject(s)
Casein Kinase II/antagonists & inhibitors , Naphthyridines/therapeutic use , Prostatic Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Small Molecule Libraries/therapeutic use , Xenograft Model Antitumor Assays , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Casein Kinase II/metabolism , Caspase 3/metabolism , Caspase 7/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Humans , Immunohistochemistry , Male , Mice , Naphthyridines/chemistry , Naphthyridines/pharmacology , Phenazines , Phosphorylation/drug effects , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Small Molecule Libraries/administration & dosage , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Structure-Activity Relationship
15.
Bioorg Med Chem Lett ; 21(6): 1687-91, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21316963

ABSTRACT

We describe the discovery of novel potent substituted pyrimido[4,5-c]quinoline ATP-competitive inhibitors of protein kinase CK2. A binding model of the inhibitors with the protein was elaborated on the basis of SAR and revealed various modes of interaction with the hinge region. Representative analog 14k (CK2 IC(50)=9 nM) showed anti-viral activity at nanomolar concentrations against HIV-1. Orally available compound 7e (CK2 IC(50)=3 nM) reduced pain in the phase II of a murine formalin model. These preliminary data confirm that properly optimized CK2 inhibitors may be used for anti-viral and pain therapy.


Subject(s)
Analgesics/pharmacology , Antiviral Agents/pharmacology , Casein Kinase II/antagonists & inhibitors , Quinolines/pharmacology , Analgesics/chemistry , Antiviral Agents/chemistry , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Hydrogen Bonding , Quinolines/chemistry , Structure-Activity Relationship
16.
J Med Chem ; 54(2): 635-54, 2011 Jan 27.
Article in English | MEDLINE | ID: mdl-21174434

ABSTRACT

Herein we chronicle the discovery of CX-4945 (25n), a first-in-class, orally bioavailable ATP-competitive inhibitor of protein kinase CK2 in clinical trials for cancer. CK2 has long been considered a prime cancer drug target because of the roles of deregulated and overexpressed CK2 in cancer-promoting prosurvival and antiapoptotic pathways. These biological properties as well as the suitability of CK2's small ATP binding site for the design of selective inhibitors, led us to fashion novel therapeutic agents for cancer. The optimization leading to 25n (K(i) = 0.38 nM) was guided by molecular modeling, suggesting a strong binding of 25n resulting from a combination of hydrophobic interactions, an ionic bridge with Lys68, and hydrogen bonding with the hinge region. 25n was found to be highly selective, orally bioavailable across species (20-51%) and efficacious in xenograft models. The discovery of 25n will allow the therapeutic targeting of CK2 in humans for the first time.


Subject(s)
Antineoplastic Agents/chemical synthesis , Casein Kinase II/antagonists & inhibitors , Naphthyridines/chemical synthesis , Adenosine Triphosphate/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Binding, Competitive , Biological Availability , Cell Line, Tumor , Dogs , Drug Screening Assays, Antitumor , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Mice , Mice, Inbred ICR , Mice, Nude , Models, Molecular , Naphthyridines/chemistry , Naphthyridines/pharmacology , Neoplasm Transplantation , Phenazines , Rats , Structure-Activity Relationship , Transplantation, Heterologous
17.
Cancer Res ; 71(4): 1418-30, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21159662

ABSTRACT

Deregulated ribosomal RNA synthesis is associated with uncontrolled cancer cell proliferation. RNA polymerase (Pol) I, the multiprotein complex that synthesizes rRNA, is activated widely in cancer. Thus, selective inhibitors of Pol I may offer a general therapeutic strategy to block cancer cell proliferation. Coupling medicinal chemistry efforts to tandem cell- and molecular-based screening led to the design of CX-5461, a potent small-molecule inhibitor of rRNA synthesis in cancer cells. CX-5461 selectively inhibits Pol I-driven transcription relative to Pol II-driven transcription, DNA replication, and protein translation. Molecular studies demonstrate that CX-5461 inhibits the initiation stage of rRNA synthesis and induces both senescence and autophagy, but not apoptosis, through a p53-independent process in solid tumor cell lines. CX-5461 is orally bioavailable and demonstrates in vivo antitumor activity against human solid tumors in murine xenograft models. Our findings position CX-5461 for investigational clinical trials as a potent, selective, and orally administered agent for cancer treatment.


Subject(s)
Benzothiazoles/pharmacology , Cell Proliferation/drug effects , Naphthyridines/pharmacology , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , RNA Polymerase I/antagonists & inhibitors , RNA, Ribosomal/biosynthesis , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Benzothiazoles/administration & dosage , Benzothiazoles/therapeutic use , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Female , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , HeLa Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Targeted Therapy/methods , Naphthyridines/administration & dosage , Naphthyridines/therapeutic use , Neoplasms/metabolism , RNA Polymerase I/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
18.
Cancer Res ; 70(24): 10288-98, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21159648

ABSTRACT

Malignant transformation and maintenance of the malignant phenotype depends on oncogenic and non-oncogenic proteins that are essential to mediate oncogene signaling and to support the altered physiologic demands induced by transformation. Protein kinase CK2 supports key prosurvival signaling pathways and represents a prototypical non-oncogene. In this study, we describe CX-4945, a potent and selective orally bioavailable small molecule inhibitor of CK2. The antiproliferative activity of CX-4945 against cancer cells correlated with expression levels of the CK2α catalytic subunit. Attenuation of PI3K/Akt signaling by CX-4945 was evidenced by dephosphorylation of Akt on the CK2-specific S129 site and the canonical S473 and T308 regulatory sites. CX-4945 caused cell-cycle arrest and selectively induced apoptosis in cancer cells relative to normal cells. In models of angiogenesis, CX-4945 inhibited human umbilical vein endothelial cell migration, tube formation, and blocked CK2-dependent hypoxia-induced factor 1 alpha (HIF-1α) transcription in cancer cells. When administered orally in murine xenograft models, CX-4945 was well tolerated and demonstrated robust antitumor activity with concomitant reductions of the mechanism-based biomarker phospho-p21 (T145). The observed antiproliferative and anti-angiogenic responses to CX-4945 in tumor cells and endothelial cells collectively illustrate that this compound exerts its antitumor effects through inhibition of CK2-dependent signaling in multiple pathways. Finally, CX-4945 is the first orally bioavailable small molecule inhibitor of CK2 to advance into human clinical trials, thereby paving the way for an entirely new class of targeted treatment for cancer.


Subject(s)
Casein Kinase II/antagonists & inhibitors , Inflammatory Breast Neoplasms/drug therapy , Naphthyridines/pharmacology , Pancreatic Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Administration, Oral , Animals , Biological Availability , Cell Line, Tumor , Endothelial Cells/cytology , Endothelial Cells/drug effects , Female , HeLa Cells , Humans , Inflammatory Breast Neoplasms/blood supply , Inflammatory Breast Neoplasms/enzymology , Mice , Naphthyridines/pharmacokinetics , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/enzymology , Phenazines , Protein Kinase Inhibitors/pharmacokinetics , Random Allocation , Xenograft Model Antitumor Assays
19.
Cancer Res ; 69(19): 7653-61, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19738048

ABSTRACT

Hallmark deregulated signaling in cancer cells drives excessive ribosome biogenesis within the nucleolus, which elicits unbridled cell growth and proliferation. The rate-limiting step of ribosome biogenesis is synthesis of rRNA (building blocks of ribosomes) by RNA Polymerase I (Pol I). Numerous kinase pathways and products of proto-oncogenes can up-regulate Pol I, whereas tumor suppressor proteins can inhibit rRNA synthesis. In tumorigenesis, activating mutations in certain cancer-associated kinases and loss-of-function mutations in tumor suppressors lead to deregulated signaling that stimulates Pol I transcription with resultant increases in ribosome biogenesis, protein synthesis, cell growth, and proliferation. Certain anticancer therapeutics, such as cisplatin and 5-fluorouracil, reportedly exert, at least partially, their activity through disruption of ribosome biogenesis, yet many prime targets for anticancer drugs within the ribosome synthetic machinery of the nucleolus remain largely unexploited. Herein, we describe CX-3543, a small molecule nucleolus-targeting agent that selectively disrupts nucleolin/rDNA G-quadruplex complexes in the nucleolus, thereby inhibiting Pol I transcription and inducing apoptosis in cancer cells. CX-3543 is the first G-quadruplex interactive agent to enter human clinical trials, and it is currently under evaluation against carcinoid/neuroendocrine tumors in a phase II clinical trial.


Subject(s)
Benzoxazines/pharmacology , Neoplasms/drug therapy , Quinolones/pharmacology , RNA, Ribosomal/antagonists & inhibitors , RNA, Ribosomal/biosynthesis , Animals , Apoptosis/drug effects , Benzoxazines/pharmacokinetics , Cell Line, Tumor , Cell Nucleolus/drug effects , Cell Nucleolus/metabolism , DNA Polymerase I/antagonists & inhibitors , DNA Polymerase I/genetics , DNA Polymerase I/metabolism , DNA Topoisomerases, Type I/metabolism , DNA Topoisomerases, Type II/metabolism , Female , G-Quadruplexes/drug effects , HL-60 Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Mice, SCID , Neoplasms/genetics , Neoplasms/metabolism , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/genetics , Phosphoproteins/metabolism , Quinolones/pharmacokinetics , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Telomere/drug effects , Transcription, Genetic/drug effects , Xenograft Model Antitumor Assays , Nucleolin
20.
Semin Oncol ; 33(4): 498-512, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16890804

ABSTRACT

In this review, we describe the evidence for a parallel-stranded G-quadruplex in the purine-rich strand of the nuclease hypersensitivity element III(1) (NHE III(1)) of the promoter of c-MYC upstream of the P1 and P2 promoters. This biologically relevant G-quadruplex is a mixture of four loop isomers. The folding pattern of a nuclear magnetic resonance (NMR)-derived structure for the predominant loop isomer of this G-quadruplex has been obtained. This G-quadruplex has been demonstrated to be a silencer element, and the cationic porphyrin TMPyP4 has been shown to stabilize this G-quadruplex. Furthermore, TMPyP4 has been shown to repress c-MYC expression, and this effect is mediated through the silencer element. Last, the in vivo activity of TMPyP4 in xenograph models is presented.


Subject(s)
Genes, myc/drug effects , Porphyrins/pharmacology , Proto-Oncogene Proteins c-myc/metabolism , Animals , Base Sequence , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Gene Silencing , Humans , Isomerism , Porphyrins/chemistry , Promoter Regions, Genetic , Proto-Oncogene Proteins c-myc/genetics , Silencer Elements, Transcriptional , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...