Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Biomedicines ; 11(4)2023 Apr 09.
Article in English | MEDLINE | ID: mdl-37189750

ABSTRACT

The survival rate of patients with osteosarcoma (OS) has not improved over the last 30 years. Mutations in the genes TP53, RB1 and c-Myc frequently occur in OS and enhance RNA Polymerase I (Pol I) activity, thus supporting uncontrolled cancer cell proliferation. We therefore hypothesised that Pol I inhibition may be an effective therapeutic strategy for this aggressive cancer. The Pol I inhibitor CX-5461 has demonstrated therapeutic efficacy in different cancers in pre-clinical and phase I clinical trials; thus, the effects were determined on ten human OS cell lines. Following characterisation using genome profiling and Western blotting, RNA Pol I activity, cell proliferation and cell cycle progression were evaluated in vitro, and the growth of TP53 wild-type and mutant tumours was measured in a murine allograft model and in two human xenograft OS models. CX-5461 treatment resulted in reduced ribosomal DNA (rDNA) transcription and Growth 2 (G2)-phase cell cycle arrest in all OS cell lines. Additionally, tumour growth in all allograft and xenograft OS models was effectively suppressed without apparent toxicity. Our study demonstrates the efficacy of Pol I inhibition against OS with varying genetic alterations. This study provides pre-clinical evidence to support this novel therapeutic approach in OS.

2.
Nat Cell Biol ; 24(6): 954-967, 2022 06.
Article in English | MEDLINE | ID: mdl-35637408

ABSTRACT

Epidemiological studies demonstrate an association between breast cancer (BC) and systemic dysregulation of glucose metabolism. However, how BC influences glucose homeostasis remains unknown. We show that BC-derived extracellular vesicles (EVs) suppress pancreatic insulin secretion to impair glucose homeostasis. EV-encapsulated miR-122 targets PKM in ß-cells to suppress glycolysis and ATP-dependent insulin exocytosis. Mice receiving high-miR-122 EVs or bearing BC tumours exhibit suppressed insulin secretion, enhanced endogenous glucose production, impaired glucose tolerance and fasting hyperglycaemia. These effects contribute to tumour growth and are abolished by inhibiting EV secretion or miR-122, restoring PKM in ß-cells or supplementing insulin. Compared with non-cancer controls, patients with BC have higher levels of circulating EV-encapsulated miR-122 and fasting glucose concentrations but lower fasting insulin; miR-122 levels are positively associated with glucose and negatively associated with insulin. Therefore, EV-mediated impairment of whole-body glycaemic control may contribute to tumour progression and incidence of type 2 diabetes in some patients with BC.


Subject(s)
Breast Neoplasms , Diabetes Mellitus, Type 2 , Extracellular Vesicles , MicroRNAs , Animals , Breast Neoplasms/pathology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Extracellular Vesicles/metabolism , Female , Glucose/metabolism , Homeostasis , Humans , Insulin/metabolism , Insulin Secretion , Mice , MicroRNAs/genetics , MicroRNAs/metabolism
3.
Invest New Drugs ; 40(3): 529-536, 2022 06.
Article in English | MEDLINE | ID: mdl-35201535

ABSTRACT

BACKGROUND: Uterine leiomyosarcoma is a rare aggressive smooth muscle cancer with poor survival rates. RNA Polymerase I (Pol I) activity is elevated in many cancers supporting tumour growth and prior studies in uterine leiomyosarcoma revealed enlarged nucleoli and upregulated Pol I activity-related genes. This study aimed to investigate the anti-tumour potential of CX-5461, a Pol I transcription inhibitor currently being evaluated in clinical trials for several cancers, against the human uterine leiomyosarcoma cell line, SK-UT-1. METHODS: SK-UT-1 was characterised using genome profiling and western blotting. The anti-tumour effects of CX-5461 were investigated using cell proliferation assays, expression analysis using qRT-PCR, and BrdU/PI based cell cycle analysis. RESULTS: Genetic analysis of SK-UT-1 revealed mutations in TP53, RB1, PTEN, APC and TSC1 & 2, all potentially associated with increased Pol I activity. Protein expression analysis showed dysregulated p53, RB1 and c-Myc. CX-5461 treatment resulted in an anti-proliferation response, G2 phase cell-cycle arrest and on-target activity demonstrated by reduced ribosomal DNA transcription. CONCLUSIONS: SK-UT-1 was confirmed as a representative model of uterine leiomyosarcoma and CX-5461 has significant potential as a novel adjuvant for this rare cancer.


Subject(s)
Benzothiazoles , Leiomyosarcoma , Naphthyridines , Uterine Neoplasms , Benzothiazoles/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Enzyme Inhibitors/pharmacology , Female , Humans , Leiomyosarcoma/drug therapy , Leiomyosarcoma/genetics , Leiomyosarcoma/metabolism , Naphthyridines/pharmacology , RNA Polymerase I/antagonists & inhibitors , RNA Polymerase I/metabolism , Signal Transduction/drug effects , Uterine Neoplasms/drug therapy , Uterine Neoplasms/genetics , Uterine Neoplasms/metabolism
4.
Nat Commun ; 10(1): 2110, 2019 05 08.
Article in English | MEDLINE | ID: mdl-31068593

ABSTRACT

Ribosome biogenesis is a canonical hallmark of cell growth and proliferation. Here we show that execution of Epithelial-to-Mesenchymal Transition (EMT), a migratory cellular program associated with development and tumor metastasis, is fueled by upregulation of ribosome biogenesis during G1/S arrest. This unexpected EMT feature is independent of species and initiating signal, and is accompanied by release of the repressive nucleolar chromatin remodeling complex (NoRC) from rDNA, together with recruitment of the EMT-driving transcription factor Snai1 (Snail1), RNA Polymerase I (Pol I) and the Upstream Binding Factor (UBF). EMT-associated ribosome biogenesis is also coincident with increased nucleolar recruitment of Rictor, an essential component of the EMT-promoting mammalian target of rapamycin complex 2 (mTORC2). Inhibition of rRNA synthesis in vivo differentiates primary tumors to a benign, Estrogen Receptor-alpha (ERα) positive, Rictor-negative phenotype and reduces metastasis. These findings implicate the EMT-associated ribosome biogenesis program with cellular plasticity, de-differentiation, cancer progression and metastatic disease.


Subject(s)
Epithelial-Mesenchymal Transition/physiology , G1 Phase Cell Cycle Checkpoints/physiology , Gene Expression Regulation, Developmental , Gene Expression Regulation, Neoplastic , Ribosomes/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Differentiation/physiology , Cell Line, Tumor/transplantation , Cell Movement/physiology , Cell Nucleolus/metabolism , Chick Embryo , Chromosomal Proteins, Non-Histone/metabolism , DNA, Ribosomal/metabolism , Disease Models, Animal , Female , Gene Expression Profiling , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA, Ribosomal/metabolism , Ribosomes/genetics
5.
Blood ; 129(21): 2882-2895, 2017 05 25.
Article in English | MEDLINE | ID: mdl-28283481

ABSTRACT

Despite the development of novel drugs, the prospects for many patients with acute myeloid leukemia (AML) remain dismal. This study reveals that the selective inhibitor of RNA polymerase I (Pol I) transcription, CX-5461, effectively treats aggressive AML, including mixed-lineage leukemia-driven AML, and outperforms standard chemotherapies. In addition to the previously characterized mechanism of action of CX-5461 (ie, the induction of p53-dependent apoptotic cell death), the inhibition of Pol I transcription also demonstrates potent efficacy in p53null AML in vivo. This significant survival advantage in both p53WT and p53null leukemic mice treated with CX-5461 is associated with activation of the checkpoint kinases 1/2, an aberrant G2/M cell-cycle progression and induction of myeloid differentiation of the leukemic blasts. The ability to target the leukemic-initiating cell population is thought to be essential for lasting therapeutic benefit. Most strikingly, the acute inhibition of Pol I transcription reduces both the leukemic granulocyte-macrophage progenitor and leukemia-initiating cell (LIC) populations, and suppresses their clonogenic capacity. This suggests that dysregulated Pol I transcription is essential for the maintenance of their leukemia-initiating potential. Together, these findings demonstrate the therapeutic utility of this new class of inhibitors to treat highly aggressive AML by targeting LICs.


Subject(s)
Benzothiazoles/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Naphthyridines/pharmacology , Neoplastic Stem Cells/enzymology , Pol1 Transcription Initiation Complex Proteins/antagonists & inhibitors , Transcription, Genetic/drug effects , Animals , Cell Division/drug effects , Cell Division/genetics , Cell Line, Tumor , Checkpoint Kinase 1/genetics , Checkpoint Kinase 1/metabolism , Checkpoint Kinase 2/genetics , Checkpoint Kinase 2/metabolism , G2 Phase/drug effects , G2 Phase/genetics , Humans , Leukemia, Myeloid, Acute/epidemiology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred NOD , Mice, Mutant Strains , Neoplastic Stem Cells/pathology , Pol1 Transcription Initiation Complex Proteins/genetics , Pol1 Transcription Initiation Complex Proteins/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
6.
Clin Cancer Res ; 22(22): 5539-5552, 2016 Nov 15.
Article in English | MEDLINE | ID: mdl-27486174

ABSTRACT

PURPOSE: The MYC oncogene is frequently overexpressed in prostate cancer. Upregulation of ribosome biogenesis and function is characteristic of MYC-driven tumors. In addition, PIM kinases activate MYC signaling and mRNA translation in prostate cancer and cooperate with MYC to accelerate tumorigenesis. Here, we investigate the efficacy of a single and dual approach targeting ribosome biogenesis and function to treat prostate cancer. EXPERIMENTAL DESIGN: The inhibition of ribosomal RNA (rRNA) synthesis with CX-5461, a potent, selective, and orally bioavailable inhibitor of RNA polymerase I (Pol I) transcription, has been successfully exploited therapeutically but only in models of hematologic malignancy. CX-5461 and CX-6258, a pan-PIM kinase inhibitor, were tested alone and in combination in prostate cancer cell lines, in Hi-MYC- and PTEN-deficient mouse models and in patient-derived xenografts (PDX) of metastatic tissue obtained from a patient with castration-resistant prostate cancer. RESULTS: CX-5461 inhibited anchorage-independent growth and induced cell-cycle arrest in prostate cancer cell lines at nanomolar concentrations. Oral administration of 50 mg/kg CX-5461 induced TP53 expression and activity and reduced proliferation (MKI67) and invasion (loss of ductal actin) in Hi-MYC tumors, but not in PTEN-null (low MYC) tumors. While 100 mg/kg CX-6258 showed limited effect alone, its combination with CX-5461 further suppressed proliferation and dramatically reduced large invasive lesions in both models. This rational combination strategy significantly inhibited proliferation and induced cell death in PDX of prostate cancer. CONCLUSIONS: Our results demonstrate preclinical efficacy of targeting the ribosome at multiple levels and provide a new approach for the treatment of prostate cancer. Clin Cancer Res; 22(22); 5539-52. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Prostatic Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , RNA Polymerase I/antagonists & inhibitors , Transcription, Genetic/drug effects , Animals , Azepines/pharmacology , Benzothiazoles/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Indoles/pharmacology , Male , Mice , Naphthyridines/pharmacology , PTEN Phosphohydrolase/metabolism , Prostate/drug effects , Prostate/metabolism , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays/methods
7.
Oncotarget ; 7(31): 49800-49818, 2016 Aug 02.
Article in English | MEDLINE | ID: mdl-27391441

ABSTRACT

RNA polymerase I (Pol I)-mediated transcription of the ribosomal RNA genes (rDNA) is confined to the nucleolus and is a rate-limiting step for cell growth and proliferation. Inhibition of Pol I by CX-5461 can selectively induce p53-mediated apoptosis of tumour cells in vivo. Currently, CX-5461 is in clinical trial for patients with advanced haematological malignancies (Peter Mac, Melbourne). Here we demonstrate that CX-5461 also induces p53-independent cell cycle checkpoints mediated by ATM/ATR signaling in the absence of DNA damage. Further, our data demonstrate that the combination of drugs targeting ATM/ATR signaling and CX-5461 leads to enhanced therapeutic benefit in treating p53-null tumours in vivo, which are normally refractory to each drug alone. Mechanistically, we show that CX-5461 induces an unusual chromatin structure in which transcriptionally competent relaxed rDNA repeats are devoid of transcribing Pol I leading to activation of ATM signaling within the nucleoli. Thus, we propose that acute inhibition of Pol transcription initiation by CX-5461 induces a novel nucleolar stress response that can be targeted to improve therapeutic efficacy.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , Benzothiazoles/pharmacology , Naphthyridines/pharmacology , Nucleic Acid Synthesis Inhibitors/pharmacology , RNA Polymerase I/antagonists & inhibitors , Signal Transduction , Animals , Apoptosis , Cell Enlargement , Cell Nucleolus/metabolism , Cell Proliferation , Chromatin/metabolism , Comet Assay , DNA Damage , DNA, Ribosomal/genetics , Fibroblasts/metabolism , Hematologic Neoplasms/metabolism , Humans , Mice , Mice, Inbred C57BL , RNA Polymerase I/metabolism , Tumor Suppressor Protein p53/metabolism
8.
Oncotarget ; 7(13): 15648-61, 2016 Mar 29.
Article in English | MEDLINE | ID: mdl-26871292

ABSTRACT

We previously showed how key pathways in cancer-related inflammation and Notch signaling are part of an autocrine malignant cell network in ovarian cancer. This network, which we named the "TNF network", has paracrine actions within the tumor microenvironment, influencing angiogenesis and the immune cell infiltrate.The aim of this study was to identify critical regulators in the signaling pathways of the TNF network in ovarian cancer cells that might be therapeutic targets. To achieve our aim, we used a systems biology approach, combining data from phospho-proteomic mass spectrometry and gene expression array analysis. Among the potential therapeutic kinase targets identified was the protein kinase Casein kinase II (CK2).Knockdown of CK2 expression in malignant cells by siRNA or treatment with the specific CK2 inhibitor CX-4945 significantly decreased Notch signaling and reduced constitutive cytokine release in ovarian cancer cell lines that expressed the TNF network as well as malignant cells isolated from high grade serous ovarian cancer ascites. The expression of the same cytokines was also inhibited after treatment with CX-4945 in a 3D organotypic model. CK2 inhibition was associated with concomitant inhibition of proliferative activity, reduced angiogenesis and experimental peritoneal ovarian tumor growth.In conclusion, we have identified kinases, particularly CK2, associated with the TNF network that may play a central role in sustaining the cytokine network and/or mediating its effects in ovarian cancer.


Subject(s)
Inflammation/enzymology , Ovarian Neoplasms/pathology , Signal Transduction/immunology , Animals , Casein Kinase II/metabolism , Cell Line, Tumor , Cytokines/immunology , Cytokines/metabolism , Female , Gene Expression Profiling/methods , Heterografts , Humans , Inflammation/immunology , Inflammation/pathology , Mice , Mice, Inbred BALB C , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/immunology , Proteomics/methods , Systems Biology/methods , Transcriptome , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
9.
Cancer Discov ; 6(1): 59-70, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26490423

ABSTRACT

UNLABELLED: Ribosome biogenesis and protein synthesis are dysregulated in many cancers, with those driven by the proto-oncogene c-MYC characterized by elevated Pol I-mediated ribosomal rDNA transcription and mTORC1/eIF4E-driven mRNA translation. Here, we demonstrate that coordinated targeting of rDNA transcription and PI3K-AKT-mTORC1-dependent ribosome biogenesis and protein synthesis provides a remarkable improvement in survival in MYC-driven B lymphoma. Combining an inhibitor of rDNA transcription (CX-5461) with the mTORC1 inhibitor everolimus more than doubled survival of Eµ-Myc lymphoma-bearing mice. The ability of each agent to trigger tumor cell death via independent pathways was central to their synergistic efficacy. CX-5461 induced nucleolar stress and p53 pathway activation, whereas everolimus induced expression of the proapoptotic protein BMF that was independent of p53 and reduced expression of RPL11 and RPL5. Thus, targeting the network controlling the synthesis and function of ribosomes at multiple points provides a potential new strategy to treat MYC-driven malignancies. SIGNIFICANCE: Treatment options for the high proportion of cancers driven by MYC are limited. We demonstrate that combining pharmacologic targeting of ribosome biogenesis and mTORC1-dependent translation provides a remarkable therapeutic benefit to Eµ-Myc lymphoma-bearing mice. These results establish a rationale for targeting ribosome biogenesis and function to treat MYC-driven cancer.


Subject(s)
Benzothiazoles/administration & dosage , DNA, Ribosomal/antagonists & inhibitors , Everolimus/administration & dosage , Lymphoma, B-Cell/therapy , Naphthyridines/administration & dosage , Proto-Oncogene Proteins c-myc/genetics , Animals , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzothiazoles/pharmacology , Drug Synergism , Everolimus/pharmacology , Humans , Lymphoma, B-Cell/genetics , Mice , Naphthyridines/pharmacology , Protein Biosynthesis/drug effects , Proto-Oncogene Mas , Signal Transduction/drug effects , Survival Analysis , Transcription, Genetic/drug effects , Treatment Outcome , Xenograft Model Antitumor Assays
10.
Drug Discov Today ; 19(3): 259-65, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23993916

ABSTRACT

The tumor suppressor protein p53 plays a crucial part in the cellular defense against malignancies. DNA-damaging chemotherapeutics rely on the activation of p53 for their anticancer activity at the expense of genotoxicity. Nongenotoxic approaches for p53 activation have been extensively investigated validating p53 as a therapeutic target. However, their development has been hampered by low efficacy and a narrow therapeutic window. An alternate nongenotoxic approach for cancer-specific activation of wild-type p53 has been recently identified. It relies on the activation of a cellular checkpoint mechanism termed 'nucleolar stress', which can be triggered by acute inhibition of rRNA biogenesis. CX5461, the first selective inhibitor of rRNA biogenesis, and thus a potent activator of nucleolar stress, is poised to enter clinical development.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Tumor Suppressor Protein p53/metabolism , Animals , Antineoplastic Agents/adverse effects , Benzothiazoles/pharmacology , Cell Nucleolus/metabolism , DNA Damage/drug effects , Drug Design , Humans , Molecular Targeted Therapy , Naphthyridines/pharmacology , Neoplasms/pathology , RNA, Ribosomal/metabolism
11.
Clin Cancer Res ; 19(23): 6484-94, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24036851

ABSTRACT

PURPOSE: Gliomas are the most frequently occurring primary malignancies in the brain, and glioblastoma is the most aggressive of these tumors. Protein kinase CK2 is composed of two catalytic subunits (α and/or α') and two ß regulatory subunits. CK2 suppresses apoptosis, promotes neoangiogenesis, and enhances activation of the JAK/STAT, NF-κB, PI3K/AKT, Hsp90, Wnt, and Hedgehog pathways. Aberrant activation of the NF-κB, PI3K/AKT, and JAK/STAT-3 pathways is implicated in glioblastoma progression. As CK2 is involved in their activation, the expression and function of CK2 in glioblastoma was evaluated. EXPERIMENTAL DESIGN AND RESULTS: Analysis of 537 glioblastomas from The Cancer Genome Atlas Project demonstrates the CSNK2A1 gene, encoding CK2α, has gene dosage gains in glioblastoma (33.7%), and is significantly associated with the classical glioblastoma subtype. Inhibition of CK2 activity by CX-4945, a selective CK2 inhibitor, or CK2 knockdown by siRNA suppresses activation of the JAK/STAT, NF-κB, and AKT pathways and downstream gene expression in human glioblastoma xenografts. On a functional level, CX-4945 treatment decreases the adhesion and migration of glioblastoma cells, in part through inhibition of integrin ß1 and α4 expression. In vivo, CX-4945 inhibits activation of STAT-3, NF-κB p65, and AKT, and promotes survival of mice with intracranial human glioblastoma xenografts. CONCLUSIONS: CK2 inhibitors may be considered for treatment of patients with glioblastoma.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/enzymology , Glioblastoma/enzymology , Naphthyridines/pharmacology , Signal Transduction , Animals , Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Casein Kinase II/genetics , Casein Kinase II/metabolism , Cell Adhesion , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Survival , Enzyme Activation , Female , Gene Amplification , Gene Dosage , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Mice , Mice, Nude , Naphthyridines/therapeutic use , Phenazines , Proto-Oncogene Proteins c-akt/metabolism , STAT3 Transcription Factor/metabolism , Xenograft Model Antitumor Assays
12.
Expert Opin Ther Targets ; 17(8): 873-8, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23862680

ABSTRACT

The nucleoli are the site of the production of ribosomes, the protein synthetic apparatus of the cell. The presence of enlarged nucleoli, reflecting increased ribosomal gene transcription, has long been used by pathologists as an indicator of aggressive tumors. However, over the last 10 years a growing body of evidence has revealed that the nucleolus contains a dynamic cohort of over 4500 proteins, the majority of which have no function in ribosome production. The activity of some of these proteins is modulated by their regulated sequestration and release from the nucleolus. In particular, the nucleolus plays a central role in sensing cellular stress to modulate the abundance of the critical tumor suppressor protein p53. The finding that p53 activity is dysregulated in up to 50% of all human cancers highlights the importance of the nucleolar stress response in limiting malignant transformation. The development of drugs to selectively inhibit transcription of the ribosomal RNA genes in the nucleolus has paved the way for a new therapeutic approach to hijack nucleolar stress to selectively and non-genotoxically activate p53 in tumor cells. Here, we describe the potential application of this exciting new class of drugs for the treatment of human cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , RNA Polymerase I/antagonists & inhibitors , Cell Nucleolus , Humans , RNA Polymerase I/genetics , Transcription, Genetic
14.
Cancer Cell ; 22(1): 51-65, 2012 Jul 10.
Article in English | MEDLINE | ID: mdl-22789538

ABSTRACT

Increased transcription of ribosomal RNA genes (rDNA) by RNA Polymerase I is a common feature of human cancer, but whether it is required for the malignant phenotype remains unclear. We show that rDNA transcription can be therapeutically targeted with the small molecule CX-5461 to selectively kill B-lymphoma cells in vivo while maintaining a viable wild-type B cell population. The therapeutic effect is a consequence of nucleolar disruption and activation of p53-dependent apoptotic signaling. Human leukemia and lymphoma cell lines also show high sensitivity to inhibition of rDNA transcription that is dependent on p53 mutational status. These results identify selective inhibition of rDNA transcription as a therapeutic strategy for the cancer specific activation of p53 and treatment of hematologic malignancies.


Subject(s)
Neoplasms/metabolism , RNA Polymerase I/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis , Benzothiazoles/pharmacology , DNA, Ribosomal/genetics , Female , Mice , Mice, Transgenic , Naphthyridines/pharmacology , Neoplasms/genetics , Neoplasms/pathology , RNA, Ribosomal/genetics , Transcription, Genetic
15.
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
16.
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
17.
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
18.
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
19.
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.

20.
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.

SELECTION OF CITATIONS
SEARCH DETAIL
...