Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Immunity ; 42(3): 484-98, 2015 Mar 17.
Article in English | MEDLINE | ID: mdl-25746953

ABSTRACT

Sepsis is characterized by a dysregulated inflammatory response to infection. Despite studies in mice, the cellular and molecular basis of human sepsis remains unclear and effective therapies are lacking. Blood monocytes serve as the first line of host defense and are equipped to recognize and respond to infection by triggering an immune-inflammatory response. However, the response of these cells in human sepsis and their contribution to sepsis pathogenesis is poorly understood. To investigate this, we performed a transcriptomic, functional, and mechanistic analysis of blood monocytes from patients during sepsis and after recovery. Our results revealed the functional plasticity of monocytes during human sepsis, wherein they transited from a pro-inflammatory to an immunosuppressive phenotype, while enhancing protective functions like phagocytosis, anti-microbial activity, and tissue remodeling. Mechanistically, hypoxia inducible factor-1α (HIF1α) mediated this functional re-programming of monocytes, revealing a potential mechanism for their therapeutic targeting to regulate human sepsis.


Subject(s)
Cellular Reprogramming/immunology , Hypoxia-Inducible Factor 1, alpha Subunit/immunology , Immunocompromised Host , Interleukin-1 Receptor-Associated Kinases/immunology , Sepsis/immunology , Adaptive Immunity , Convalescence , Cytokines/genetics , Cytokines/immunology , Gene Expression Profiling , Gene Expression Regulation , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Immunity, Innate , Interleukin-1 Receptor-Associated Kinases/genetics , Monocytes/immunology , Monocytes/pathology , Phagocytosis , Sepsis/genetics , Sepsis/pathology , Signal Transduction , Transcriptome/immunology
2.
Invest New Drugs ; 32(2): 340-6, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24242862

ABSTRACT

BACKGROUND: Anti-angiogenic therapies such as bevacizumab upregulate hypoxia-inducible factor-1α (HIF-1α), a possible mechanism of drug resistance. Camptothecin analogues, including SN-38, have been shown to reduce the expression and transcriptional activity of HIF-1α in preclinical models. We hypothesized that co-administration of pegylated SN-38 (EZN-2208) may offset the induction of HIF-1α following bevacizumab treatment, resulting in synergistic antitumor effects. PATIENTS AND METHODS: Patients with refractory solid tumors were enrolled. Objectives were to evaluate the modulation of HIF-1α protein and target genes in tumor biopsies following administration of the combination of EZN-2208 administered weekly × 3 (days 1, 8, 15) and bevacizumab administered every 2 weeks, in 28-day cycles, and to establish the safety and tolerability of the combination. Tumor biopsies and dynamic contrast enhanced MRI (DCE-MRI) were obtained following bevacizumab alone (before EZN-2208) and after administration of both study drugs. RESULTS: Twelve patients were enrolled; ten were evaluable for response. Prolonged stable disease was observed in 2 patients, one with HCC (16 cycles) and another with desmoplastic round cell tumor (7 cycles). Reduction in HIF-1α protein levels in tumor biopsies compared to baseline was observed in 5 of 7 patients. Quantitative analysis of DCE-MRI from 2 patients revealed changes in K(trans) and k(ep). The study closed prematurely as further clinical development of EZN-2208 was suspended by the pharmaceutical sponsor. CONCLUSION: Preliminary proof-of-concept for modulation of HIF-1α protein in tumor biopsies following administration of EZN-2208 was observed. Two of 10 patients had prolonged disease stabilization following treatment with the EZN-2208 and bevacizumab combination.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neoplasms/drug therapy , Adult , Aged , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/adverse effects , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Bevacizumab , Camptothecin/administration & dosage , Camptothecin/adverse effects , Camptothecin/analogs & derivatives , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Male , Middle Aged , Neoplasms/metabolism , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/adverse effects , RNA, Messenger/metabolism , Young Adult
3.
Oncotarget ; 12(21): 2114-2130, 2021 Oct 12.
Article in English | MEDLINE | ID: mdl-34676045

ABSTRACT

The therapeutic efficacy of temozolomide (TMZ) is hindered by inherent and acquired resistance. Biomarkers such as MGMT expression and MMR proficiency are used as predictors of response. However, not all MGMTlow/-ve/MMRproficient patients benefit from TMZ treatment, indicating a need for additional patient selection criteria. We explored the role of ATR in mediating TMZ resistance and whether ATR inhibitors (ATRi) could reverse this resistance in multiple cancer lines. We observed that only 31% of MGMTlow/-ve/MMRproficient patient-derived and established cancer lines are sensitive to TMZ at clinically relevant concentrations. TMZ treatment resulted in DNA damage signaling in both sensitive and resistant lines, but prolonged G2/M arrest and cell death were exclusive to sensitive models. Inhibition of ATR but not ATM, sensitized the majority of resistant models to TMZ and resulted in measurable DNA damage and persistent growth inhibition. Also, compromised homologous recombination (HR) via RAD51 or BRCA1 loss only conferred sensitivity to TMZ when combined with an ATRi. Furthermore, low REV3L mRNA expression correlated with sensitivity to the TMZ and ATRi combination in vitro and in vivo. This suggests that HR defects and low REV3L levels could be useful selection criteria for enhanced clinical efficacy of an ATRi plus TMZ combination.

4.
J Exp Med ; 198(9): 1391-402, 2003 Nov 03.
Article in English | MEDLINE | ID: mdl-14597738

ABSTRACT

Cell adaptation to hypoxia (Hyp) requires activation of transcriptional programs that coordinate expression of genes involved in oxygen delivery (via angiogenesis) and metabolic adaptation (via glycolysis). Here, we describe that oxygen availability is a determinant parameter in the setting of chemotactic responsiveness to stromal-derived factor 1 (CXCL12). Low oxygen concentration induces high expression of the CXCL12 receptor, CXC receptor 4 (CXCR4), in different cell types (monocytes, monocyte-derived macrophages, tumor-associated macrophages, endothelial cells, and cancer cells), which is paralleled by increased chemotactic responsiveness to its specific ligand. CXCR4 induction by Hyp is dependent on both activation of the Hyp-inducible factor 1 alpha and transcript stabilization. In a relay multistep navigation process, the Hyp-Hyp-inducible factor 1 alpha-CXCR4 pathway may regulate trafficking in and out of hypoxic tissue microenvironments.


Subject(s)
Cell Hypoxia , Phagocytes/metabolism , Receptors, CXCR4/physiology , Animals , Base Sequence , Cells, Cultured , DNA Primers , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Humans , Mice , RNA, Messenger/genetics , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism
5.
Drug Resist Updat ; 12(3): 74-80, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19394890

ABSTRACT

Angiogenesis, a key process for the growth of human cancers, has recently been exploited for the development of a novel class of cancer therapeutics that was thought to have wide applications and not to induce resistance in the clinical setting. Indeed, anti-angiogenic therapy has become an important option for the management of several human malignancies. However, a significant number of patients either do not respond to anti-angiogenic agents or fairly rapidly develop resistance. In addition, the benefit of anti-angiogenic therapy is relatively short-lived and the majority of patients eventually relapses and progresses. Several mechanisms of resistance to anti-angiogenic therapy have been recently proposed. The current review focuses on the role of intra-tumor hypoxia as a mechanism of resistance to anti-angiogenic agents and speculates on therapeutic approaches that might circumvent resistance and thereby improve clinical outcome.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Cell Hypoxia/drug effects , Cell Hypoxia/physiology , Drug Resistance, Neoplasm/physiology , Neoplasms/blood supply , Neoplasms/physiopathology , Neovascularization, Pathologic/physiopathology , Angiogenesis Inhibitors/therapeutic use , Combined Modality Therapy , Humans , Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy
6.
Clin Epigenetics ; 12(1): 93, 2020 06 25.
Article in English | MEDLINE | ID: mdl-32586373

ABSTRACT

BACKGROUND: Small cell lung cancer (SCLC) is an aggressive neuroendocrine lung cancer. SCLC progression and treatment resistance involve epigenetic processes. However, links between SCLC DNA methylation and drug response remain unclear. We performed an epigenome-wide study of 66 human SCLC cell lines using the Illumina Infinium MethylationEPIC BeadChip array. Correlations of SCLC DNA methylation and gene expression with in vitro response to 526 antitumor agents were examined. RESULTS: We found multiple significant correlations between DNA methylation and chemosensitivity. A potentially important association was observed for TREX1, which encodes the 3' exonuclease I that serves as a STING antagonist in the regulation of a cytosolic DNA-sensing pathway. Increased methylation and low expression of TREX1 were associated with the sensitivity to Aurora kinase inhibitors AZD-1152, SCH-1473759, SNS-314, and TAK-901; the CDK inhibitor R-547; the Vertex ATR inhibitor Cpd 45; and the mitotic spindle disruptor vinorelbine. Compared with cell lines of other cancer types, TREX1 had low mRNA expression and increased upstream region methylation in SCLC, suggesting a possible relationship with SCLC sensitivity to Aurora kinase inhibitors. We also identified multiple additional correlations indicative of potential mechanisms of chemosensitivity. Methylation of the 3'UTR of CEP350 and MLPH, involved in centrosome machinery and microtubule tracking, respectively, was associated with response to Aurora kinase inhibitors and other agents. EPAS1 methylation was associated with response to Aurora kinase inhibitors, a PLK-1 inhibitor and a Bcl-2 inhibitor. KDM1A methylation was associated with PLK-1 inhibitors and a KSP inhibitor. Increased promoter methylation of SLFN11 was correlated with resistance to DNA damaging agents, as a result of low or no SLFN11 expression. The 5' UTR of the epigenetic modifier EZH2 was associated with response to Aurora kinase inhibitors and a FGFR inhibitor. Methylation and expression of YAP1 were correlated with response to an mTOR inhibitor. Among non-neuroendocrine markers, EPHA2 was associated with response to Aurora kinase inhibitors and a PLK-1 inhibitor and CD151 with Bcl-2 inhibitors. CONCLUSIONS: Multiple associations indicate potential epigenetic mechanisms affecting SCLC response to chemotherapy and suggest targets for combination therapies. While many correlations were not specific to SCLC lineages, several lineage markers were associated with specific agents.


Subject(s)
Cell Line, Tumor/drug effects , DNA Methylation/genetics , Epigenome/genetics , Small Cell Lung Carcinoma/genetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Aurora Kinases/antagonists & inhibitors , Cell Cycle Proteins/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor Proteins/pharmacology , DNA Methylation/drug effects , Drug Therapy, Combination/statistics & numerical data , Exodeoxyribonucleases/genetics , Exodeoxyribonucleases/metabolism , Gene Expression/drug effects , Gene Expression/genetics , Gene Expression Regulation, Neoplastic/drug effects , High-Throughput Nucleotide Sequencing/methods , Histone Demethylases/drug effects , Histone Demethylases/genetics , Humans , Lung Neoplasms/pathology , Membrane Proteins/antagonists & inhibitors , Nuclear Proteins/drug effects , Nuclear Proteins/genetics , Phosphoproteins/genetics , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Small Cell Lung Carcinoma/diagnosis , Polo-Like Kinase 1
7.
J Cell Mol Med ; 13(9A): 2780-6, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19674190

ABSTRACT

Intratumour hypoxia has long been considered a driving force of tumour progression and a negative prognostic factor in human cancers. The discovery of hypoxia inducible factors (HIFs), which mediate transcriptional responses to changes in oxygen levels, has renewed enthusiasm for the discovery and development of targeted therapies exploiting the hypoxic tumour microenvironment. In spite of an ever increasing number of putative small molecule inhibitors of HIF, only few progress through pre-clinical and early clinical development. In this review, we will focus primarily on: (1) HIF inhibitors that have been more recently described and (2) small molecules targeting HIF that are being tested in early clinical trials or that are already approved for use in patients. A rigorous 'validation' of HIF targeted therapies in relevant pre-clinical models and eventually in pharmacodynamic-based early clinical trials is essential for 'credentialing' HIF-1 as a legitimate target that can be pharmacologically modulated in cancer patients.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Drug Discovery , Hypoxia-Inducible Factor 1/antagonists & inhibitors , Neoplasms/drug therapy , Animals , Humans
8.
Clin Cancer Res ; 13(3): 1010-8, 2007 Feb 01.
Article in English | MEDLINE | ID: mdl-17289897

ABSTRACT

PURPOSE: The discovery and development of small-molecule inhibitors of hypoxia-inducible factor-1 (HIF-1) is an attractive, yet challenging, strategy for the development of new cancer therapeutic agents. Here, we report on a novel tricyclic carboxamide inhibitor of HIF-1alpha, NSC 644221. EXPERIMENTAL DESIGN: We investigated the mechanism by which the novel compound NSC 644221 inhibited HIF-1alpha. RESULTS: NSC 644221 inhibited HIF-1-dependent, but not constitutive, luciferase expression in U251-HRE and U251-pGL3 cells, respectively, as well as hypoxic induction of vascular endothelial growth factor mRNA expression in U251 cells. HIF-1alpha, but not HIF-1beta, protein expression was inhibited by NSC 644221 in a time- and dose-dependent fashion. Interestingly, NSC 644221 was unable to inhibit HIF-1alpha protein accumulation in the presence of the proteasome inhibitors MG132 or PS341, yet it did not directly affect the degradation of HIF-1alpha as shown by experiments done in the presence of cyclohexamide or pulse-chase labeling using [35S]methionine. In contrast, NSC 644221 decreased the rate of HIF-1alpha translation relative to untreated controls. Silencing of topoisomerase (topo) IIalpha, but not topo I, by specific small interfering RNA completely blocked the ability of NSC 644221 to inhibit HIF-1alpha. The data presented show that topo II is required for the inhibition of HIF-1alpha by NSC 644221. Furthermore, although NSC 644221 induced p21 expression, gammaH2A.X, and G2-M arrest in the majority of cell lines tested, it only inhibited HIF-1alpha in a distinct subset of cells, raising the possibility of pathway-specific "resistance" to HIF-1 inhibition in cancer cells. CONCLUSIONS: NSC 644221 is a novel HIF-1 inhibitor with potential for use as both an analytic tool and a therapeutic agent. Our data provide a strong rationale for pursuing the preclinical development of NSC 644221 as a HIF-1 inhibitor.


Subject(s)
Amides/pharmacology , Antineoplastic Agents/pharmacology , DNA Topoisomerases, Type II/metabolism , Gene Expression Regulation, Neoplastic , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Neoplasms/drug therapy , Polycyclic Compounds/pharmacology , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Humans , Luciferases/metabolism , Models, Chemical , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Temperature , Time Factors , Transfection
9.
Cancer Res ; 78(24): 6807-6817, 2018 12 15.
Article in English | MEDLINE | ID: mdl-30355619

ABSTRACT

: The intracellular effects and overall efficacies of anticancer therapies can vary significantly by tumor type. To identify patterns of drug-induced gene modulation that occur in different cancer cell types, we measured gene-expression changes across the NCI-60 cell line panel after exposure to 15 anticancer agents. The results were integrated into a combined database and set of interactive analysis tools, designated the NCI Transcriptional Pharmacodynamics Workbench (NCI TPW), that allows exploration of gene-expression modulation by molecular pathway, drug target, and association with drug sensitivity. We identified common transcriptional responses across agents and cell types and uncovered gene-expression changes associated with drug sensitivity. We also demonstrated the value of this tool for investigating clinically relevant molecular hypotheses and identifying candidate biomarkers of drug activity. The NCI TPW, publicly available at https://tpwb.nci.nih.gov, provides a comprehensive resource to facilitate understanding of tumor cell characteristics that define sensitivity to commonly used anticancer drugs. SIGNIFICANCE: The NCI Transcriptional Pharmacodynamics Workbench represents the most extensive compilation to date of directly measured longitudinal transcriptional responses to anticancer agents across a thoroughly characterized ensemble of cancer cell lines.


Subject(s)
Drug Screening Assays, Antitumor/methods , Gene Expression Profiling , National Cancer Institute (U.S.) , Translational Research, Biomedical/methods , Antineoplastic Agents/pharmacology , Biomarkers, Tumor , Cell Line, Tumor , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Dose-Response Relationship, Drug , Early Growth Response Protein 1/metabolism , Erlotinib Hydrochloride/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Internet , Oligonucleotide Array Sequence Analysis , Signal Transduction , United States , Vorinostat/pharmacology , Gemcitabine
10.
Cancer Res ; 65(11): 4775-81, 2005 Jun 01.
Article in English | MEDLINE | ID: mdl-15930297

ABSTRACT

The extent of angiogenesis and/or vascular endothelial growth factor (VEGF) expression in neuroblastoma tumors correlates with metastases, N-myc amplification, and poor clinical outcome. Understanding the mechanisms regulating VEGF expression in neuroblastoma cells provides additional therapeutic options to control neuroblastoma tumor growth. VEGF mRNA is controlled by growth factors and hypoxia via the transcription factor hypoxia-inducible factor (HIF-1alpha). HIF-1alpha protein levels are regulated by the von Hippel Lindau tumor suppressor gene, VHL, which targets HIF-1alpha degradation. To determine whether the levels of VEGF in neuroblastomas are due to mutations in VHL, we evaluated genomic DNA from 15 neuroblastoma cell lines using PCR. We found no mutations in exons 1, 2, or 3 of the VHL gene. VEGF mRNA levels in neuroblastoma cells cultured in serum-free medium increased after 8 to 16 hours in serum, insulin-like growth factor-I (IGF-I), epidermal growth factor, or platelet-derived growth factor. Serum/IGF-I induced increases in HIF-1alpha protein that temporally paralleled increases in VEGF mRNA, whereas HIF-1beta levels were unaffected. VEGF and HIF-1alpha levels were blocked by inhibitors of phosphatidylinositol 3-kinase and mammalian target of rapamycin. Furthermore, we confirmed that HIF-1alpha mediates approximately 40% of the growth factor activity stimulating VEGF protein expression. Topotecan blocked the IGF-I-stimulated increase in HIF-1alpha but not HIF-1beta, and this resulted in a decrease in VEGF in four neuroblastoma cell lines tested. These data indicate that growth factors in an autocrine or paracrine manner play a major role in regulating VEGF levels in neuroblastoma cells and that targeted therapies to phosphatidylinositol 3-kinase, mammalian target of rapamycin, and/or HIF-1alpha have the potential to inhibit VEGF expression and limit neuroblastoma tumor growth.


Subject(s)
Insulin-Like Growth Factor I/pharmacology , Neuroblastoma/drug therapy , Topotecan/pharmacology , Transcription Factors/antagonists & inhibitors , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Aryl Hydrocarbon Receptor Nuclear Translocator , Cell Line, Tumor , Culture Media, Conditioned , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/biosynthesis , Humans , Hypoxia-Inducible Factor 1, alpha Subunit , Insulin-Like Growth Factor I/antagonists & inhibitors , MAP Kinase Signaling System , Neuroblastoma/enzymology , Neuroblastoma/genetics , Neuroblastoma/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , RNA, Small Interfering/genetics , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Receptors, Aryl Hydrocarbon/biosynthesis , Transcription Factors/biosynthesis , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/genetics , Von Hippel-Lindau Tumor Suppressor Protein
11.
Oncogene ; 24(30): 4829-38, 2005 Jul 14.
Article in English | MEDLINE | ID: mdl-15897903

ABSTRACT

In the presence of oxygen and iron, hypoxia-inducible factor (HIF-1alpha) is rapidly degraded via the prolyl hydroxylases (PHD)/VHL pathways. Given striking similarities between p53 and HIF-1alpha regulation, we previously suggested that HIF-1 transcriptionally initiates its own degradation and therefore inhibitors of transcription must induce HIF-1alpha. Under normoxia, while inducing p53, inhibitors of transcription did not induce HIF-1alpha. Under hypoxia or low iron (DFX), inhibitors of transcription dramatically super-induced HIF-1alpha. Removal of inhibitors resulted in outburst of the HIF-1-dependent transcription followed by depletion of HIF-1alpha. Although hypoxia/DFX induced PHD3, we excluded the PHD/VHL pathway in the regulation of HIF-1alpha under hypoxia/DFX. The transcription-dependent degradation of HIF-1alpha under hypoxia occurs via the proteasome and is accelerated by protein acetylation. Thus, HIF-1alpha is regulated by two distinct mechanisms. Under normoxia, HIF-1alpha is degraded via the classic PHD/VHL pathway, is expressed at low levels and therefore does not activate the feedback loop. But under hypoxia, HIF-1alpha accumulates and transcriptionally activates its own degradation that is independent from the PHD/VHL pathway.


Subject(s)
Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic/genetics , Cell Hypoxia/drug effects , Cell Hypoxia/genetics , Cell Hypoxia/physiology , Cell Line, Tumor , Dactinomycin/pharmacology , Depsipeptides/pharmacology , Dioxygenases , Flavonoids/pharmacology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit , Hypoxia-Inducible Factor-Proline Dioxygenases , Iron/pharmacology , Piperidines/pharmacology , Procollagen-Proline Dioxygenase/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factors/biosynthesis , Transcription Factors/deficiency , Transcription, Genetic/drug effects , Tumor Suppressor Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Von Hippel-Lindau Tumor Suppressor Protein
12.
Cancer Res ; 64(4): 1475-82, 2004 Feb 15.
Article in English | MEDLINE | ID: mdl-14983893

ABSTRACT

We have shown previously that the camptothecin analogue topotecan (TPT), a topoisomerase I (Top 1) poison, inhibits hypoxia-inducible factor 1 (HIF-1) transcriptional activity and HIF-1alpha protein accumulation in hypoxia-treated U251 human glioma cells. In this article, we demonstrate that TPT does not affect HIF-1alpha protein accumulation but inhibits its translation. In addition, we demonstrate that Top 1 is required for the inhibition of HIF-1alpha protein accumulation by TPT as shown by experiments performed using camptothecin-resistant cell lines with known Top 1 alterations. Experiments performed with aphidicolin indicated that TPT inhibited HIF-1alpha protein accumulation in the absence of DNA replication. DNA-damaging agents, such as ionizing radiation and doxorubicin, did not affect HIF-1alpha protein accumulation. Ongoing transcription was essential for the inhibition of HIF-1alpha protein accumulation by TPT. Our results demonstrate the existence of a novel pathway connecting Top 1-dependent signaling events and the regulation of HIF-1alpha protein expression and function. In addition, our findings dissociate the cytotoxic activity of TPT from the inhibition of the HIF-1 pathway and raise the possibility of novel clinical applications of TPT aimed at targeting HIF-1-dependent responses.


Subject(s)
DNA Topoisomerases, Type I/physiology , Protein Serine-Threonine Kinases , Transcription Factors/antagonists & inhibitors , Cell Line, Tumor , Cysteine Endopeptidases/physiology , DNA Damage , DNA Replication , Dose-Response Relationship, Drug , Humans , Hypoxia-Inducible Factor 1, alpha Subunit , Multienzyme Complexes/physiology , Phosphatidylinositol 3-Kinases/physiology , Proteasome Endopeptidase Complex , Protein Biosynthesis , Protein Kinases/physiology , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-akt , RNA, Messenger/analysis , TOR Serine-Threonine Kinases , Topoisomerase I Inhibitors , Topotecan/pharmacology , Topotecan/therapeutic use , Transcription Factors/genetics , Transcription, Genetic
13.
Cancer Res ; 62(15): 4316-24, 2002 Aug 01.
Article in English | MEDLINE | ID: mdl-12154035

ABSTRACT

Hypoxia-inducible factor 1 (HIF-1) is a master regulator of the transcriptional response to oxygen deprivation. HIF-1 has been implicated in the regulation of genes involved in angiogenesis [e.g., vascular endothelial growth factor (VEGF) and inducible nitric oxide synthase] and anaerobic metabolism (e.g., glycolytic enzymes). HIF-1 is essential for angiogenesis and is associated with tumor progression. In addition, overexpression of HIF-1 alpha has been demonstrated in many common human cancers. Therefore, HIF-1 is an attractive molecular target for development of novel cancer therapeutics. We have developed a cell-based high-throughput screen for the identification of small molecule inhibitors of the HIF-1 pathway. We have genetically engineered U251 human glioma cells to stably express a recombinant vector in which the luciferase reporter gene is under control of three copies of a canonical hypoxia-responsive element (U251-HRE). U251-HRE cells consistently expressed luciferase in a hypoxia- and HIF-1-dependent fashion. We now report the results of a pilot screen of the National Cancer Institute "Diversity Set," a collection of approximately 2000 compounds selected to represent the greater chemical diversity of the National Cancer Institute chemical repository. We found four compounds that specifically inhibited HIF-1-dependent induction of luciferase but not luciferase expression driven by a constitutive promoter. In addition, these compounds inhibited hypoxic induction of VEGF mRNA and protein expression in U251 cells. Interestingly, three compounds are closely related camptothecin analogues and topoisomerase (Topo)-I inhibitors. We show that concomitant with HIF-1 and VEGF inhibition, the activity of the Topo-I inhibitors tested is associated with induction of cyclooxygenase 2 mRNA expression. The luciferase-based high-throughput screen is a feasible tool for the identification of small molecule inhibitors of HIF-1 transcriptional activation. In addition, our results suggest that altered Topo-I function may be associated with repression of HIF-1-dependent induction of gene expression.


Subject(s)
Antineoplastic Agents/pharmacology , DNA-Binding Proteins/antagonists & inhibitors , Nuclear Proteins/antagonists & inhibitors , Transcription Factors , Transcriptional Activation/drug effects , Camptothecin/analogs & derivatives , DNA, Neoplasm/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , Drug Screening Assays, Antitumor , Endothelial Growth Factors/antagonists & inhibitors , Endothelial Growth Factors/biosynthesis , Endothelial Growth Factors/genetics , Enzyme Inhibitors/pharmacology , Gene Expression/drug effects , Glioma/drug therapy , Glioma/genetics , Glioma/metabolism , Humans , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Luciferases/antagonists & inhibitors , Luciferases/biosynthesis , Luciferases/genetics , Lymphokines/antagonists & inhibitors , Lymphokines/biosynthesis , Lymphokines/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/physiology , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Response Elements/drug effects , Response Elements/genetics , Topotecan/pharmacology , Transcriptional Activation/physiology , Transfection , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
14.
Cancer Res ; 64(19): 6845-8, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15466170

ABSTRACT

We have previously shown that topotecan, a topoisomerase I poison, inhibits hypoxia-inducible factor (HIF)-1alpha protein accumulation by a DNA damage-independent mechanism. Here, we report that daily administration of topotecan inhibits HIF-1alpha protein expression in U251-HRE glioblastoma xenografts. Concomitant with HIF-1alpha inhibition, topotecan caused a significant tumor growth inhibition associated with a marked decrease of angiogenesis and expression of HIF-1 target genes in tumor tissue. These results provide a compelling rationale for testing topotecan in clinical trials to target HIF-1 in cancer patients.


Subject(s)
Antineoplastic Agents/administration & dosage , Glioblastoma/drug therapy , Glioblastoma/metabolism , Neovascularization, Pathologic/drug therapy , Topotecan/administration & dosage , Transcription Factors/antagonists & inhibitors , Animals , Cell Division/drug effects , Cell Line, Tumor , Drug Administration Schedule , Enzyme Inhibitors/pharmacology , Female , Glioblastoma/blood supply , Glioblastoma/pathology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit , Luciferases/antagonists & inhibitors , Luciferases/biosynthesis , Luciferases/genetics , Mice , Mice, Nude , Neovascularization, Pathologic/metabolism , Topoisomerase I Inhibitors , Transcription Factors/biosynthesis , Transcription Factors/genetics , Xenograft Model Antitumor Assays
15.
Cell Cycle ; 3(2): 172-5, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14712084

ABSTRACT

HIF-1 is a key factor in cancer progression. Efforts are underway to identify and develop small molecules that inhibit HIF-1 transcriptional activity. What are the best targets and the best ways to develop HIF-1 inhibitors are open questions. However, several "nonselective" HIF-1 inhibitors have been identified, which are either in the clinic or under development. In this article, we discuss how topoisomerase I poisons, which inhibit HIF-1a protein accumulation and transcriptional activity, can be "rationally" used to target HIF-1 for cancer therapy.


Subject(s)
DNA Topoisomerases, Type I/metabolism , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Topotecan/pharmacology , Transcription Factors , Antineoplastic Agents/pharmacology , Cell Hypoxia/physiology , Cyclooxygenase 2 , DNA-Binding Proteins/antagonists & inhibitors , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/physiology , Helix-Loop-Helix Motifs/physiology , Humans , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Isoenzymes/metabolism , Membrane Proteins , Nuclear Proteins/antagonists & inhibitors , Prostaglandin-Endoperoxide Synthases/metabolism , Signal Transduction/physiology , Topoisomerase I Inhibitors , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A/metabolism
16.
Adv Exp Med Biol ; 527: 55-65, 2003.
Article in English | MEDLINE | ID: mdl-15206716

ABSTRACT

Recent studies have suggested a role for aminoacid catabolites as important regulators of macrophage (Mphi) activities. We reported previously that picolinic acid (PA), a tryptophan catabolite produced under inflammatory conditions and a costimulus with IFNgamma of Mphi effector functions, is a selective inducer of the Mphi inflammatory protein-1alpha (MIP-1alpha) and -1beta (MIPs), two CC-chemokines involved in the elicitation of the inflammatory reactions and in the development of the Th1 responses. In this study, we have investigated the effects of IFNgamma on PA-induced MIPs expression and secretion by mouse Mphi as well as the regulation of MIP-1alpha/beta receptor, CCR5, by both stimuli alone or in combination. We demonstrated that IFNgamma inhibited MIPs mRNA stimulation by PA in a dose-and time-dependent fashion, despite its ability to induce other CC- or CXC chemokines. MIPs mRNA down-regulation was associated with decreased intracellular chemokine expression and secretion and was dependent on both mRNA destabilization and gene transcription inhibition. Moreover, IFNgamma inhibitory effects were stimulus-specific because MIPs induction by PA was either unaffected or increased by the anti-inflammatory cytokines, IL-10 and IL-4, or the pro-inflammatory stimulus, LPS, respectively. In contrast, we found that IFNgamma increased CCR5 basal expression, whereas PA down-regulated both constitutive and IFNgamma-induced CCR5 mRNA and protein levels. These results demonstrate that IFNgamma and PA have reciprocal effects on the production of MIPs chemokines and the expression of their receptor. The concerted action of IFNgamma and PA on MIP-1alpha/beta chemokine/receptor system is likely to be of pathophysiological significance and to represent an important regulatory mechanism for leukocyte recruitment and distribution into damaged tissues during inflammatory responses.


Subject(s)
Macrophage Activation/drug effects , Picolinic Acids/metabolism , Picolinic Acids/pharmacology , Tryptophan/metabolism , Animals , Chemokine CCL3 , Chemokine CCL4 , Chemokines/biosynthesis , Humans , Interferon-gamma/pharmacology , Macrophage Inflammatory Proteins/biosynthesis , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice , Recombinant Proteins
17.
Cancer Chemother Pharmacol ; 73(2): 343-8, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24292632

ABSTRACT

PURPOSE: Hypoxia-inducible factor-1 (HIF-1) facilitates the adaptation of normal and tumor tissues to oxygen deprivation. HIF-1 is frequently overexpressed in cancer cells, where it is involved in the upregulation of many genes necessary for survival. EZN-2968 is an antisense oligodeoxynucleotide that specifically targets HIF-1α, one of the subunits of HIF-1. We conducted a trial of EZN-2968 in patients with refractory solid tumors to evaluate antitumor response and to measure modulation of HIF-1α mRNA and protein levels as well as HIF-1 target genes. METHODS: Adult patients with refractory advanced solid tumors were administered EZN-2968 as a 2-h IV infusion at a dose of 18 mg/kg once a week for three consecutive weeks followed by 3-week off; in a 6-week cycle. Tumor biopsies and dynamic contrast enhanced MRI (DCE-MRI) were performed at baseline and after the third dose. RESULTS: Ten patients were enrolled, of whom all were evaluable for response; one patient with a duodenal neuroendocrine tumor had prolonged stabilization of disease (24 weeks). Reduction in HIF-1α mRNA levels compared to baseline was demonstrated in 4 of 6 patients with paired tumor biopsies. Reductions in levels of HIF-1α protein and mRNA levels of some target genes were observed in two patients. Quantitative analysis of DCE-MRI from two patients revealed changes in K (trans) and k ep. The trial was closed prematurely when the sponsor suspended development of this agent. CONCLUSION: This trial provides preliminary proof of concept for modulation of HIF-1α mRNA and protein expression and target genes in tumor biopsies following the administration of EZN-2968.


Subject(s)
Hypoxia-Inducible Factor 1/antagonists & inhibitors , Neoplasms/drug therapy , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides/administration & dosage , Aged , Biopsy , Cell Line, Tumor , Female , Humans , Hypoxia-Inducible Factor 1/genetics , Hypoxia-Inducible Factor 1/metabolism , Infusions, Intravenous , Male , Middle Aged , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Oligonucleotides/adverse effects , Oligonucleotides, Antisense/adverse effects , Pilot Projects , RNA, Messenger/genetics , RNA, Messenger/metabolism
18.
J Clin Invest ; 123(4): 1615-29, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23549086

ABSTRACT

IL-11 and its receptor, IL-11Ra, are expressed in human cancers; however, the functional role of IL-11 in tumor progression is not known. We found that IL11 is a hypoxia-inducible, VHL-regulated gene in human cancer cells and that expression of IL11 mRNA was dependent, at least in part, on HIF-1. A cooperative interaction between HIF-1 and AP-1 mediated transcriptional activation of the IL11 promoter. Additionally, we found that human cancer cells expressed a functional IL-11Ra subunit, which triggered signal transduction either by exogenous recombinant human IL-11 or by autocrine production of IL-11 in cells cultured under hypoxic conditions. Silencing of IL11 dramatically abrogated the ability of hypoxia to increase anchorage-independent growth and significantly reduced tumor growth in xenograft models. Notably, these results were phenocopied by partial knockdown of STAT1 in a human prostate cancer cell line (PC3), suggesting that this pathway may play an important role in mediating the effects of IL-11 under hypoxic conditions. In conclusion, these results identify IL11 as an oxygen- and VHL-regulated gene and provide evidence of a pathway "hijacked" by hypoxic cancer cells that may contribute to tumor progression.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Gene Expression Regulation, Neoplastic , Interleukin-11 Receptor alpha Subunit/metabolism , Interleukin-11/metabolism , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Autocrine Communication , Basic Helix-Loop-Helix Transcription Factors/physiology , Binding Sites , Carbonic Anhydrase IX , Carbonic Anhydrases/genetics , Carbonic Anhydrases/metabolism , Cell Hypoxia , Cell Proliferation , Female , Gene Knockdown Techniques , HCT116 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Interleukin-11/genetics , Interleukin-11/physiology , Interleukin-11 Receptor alpha Subunit/genetics , Interleukin-11 Receptor alpha Subunit/physiology , MAP Kinase Signaling System , Mice , Mice, Nude , Neoplasm Transplantation , Phosphorylation , Protein Processing, Post-Translational , RNA Interference , Response Elements , STAT1 Transcription Factor/metabolism , Transcription Factor AP-1/physiology , Transcriptional Activation , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/physiology
19.
Adv Cancer Res ; 114: 237-67, 2012.
Article in English | MEDLINE | ID: mdl-22588059

ABSTRACT

New vessel formation (angiogenesis) is an essential physiological process for embryologic development, normal growth, and tissue repair. Angiogenesis is tightly regulated at the molecular level; however, this process is dysregulated in several pathological conditions such as cancer. The imbalance between pro- and antiangiogenic signaling molecules within tumors creates an abnormal vascular network that is characterized by dilated, tortuous, and leaky vessels. The pathophysiological consequences of these vascular abnormalities include temporal and spatial heterogeneity in tumor blood flow, oxygenation, and increased tumor interstitial fluid pressure. The resultant microenvironment deeply impacts on tumor progression, and also leads to a reduction in therapy efficacy. The discovery of vascular endothelial growth factor (VEGF) as a major driver of tumor angiogenesis has led to efforts to develop novel therapeutics aimed at inhibiting its activity. Anti-VEGF therapy has become an important option for the management of several human malignancies; however, a significant number of patients do not respond to anti-VEGF therapy when used either as single agent or in combination with chemotherapy. In addition, the benefit of antiangiogenic therapy is relatively short lived and the majority of patients relapse and progress. An increasing amount of reports suggest several potential mechanisms of resistance to antiangiogenic therapy including, but not limited to, tumor hypoxia. This chapter discusses the role of the VEGF axis in tumor biology and highlights the clinical application of anti-VEGF therapies elaborating on pitfalls and strategies to improve clinical outcome.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Receptors, Vascular Endothelial Growth Factor/metabolism , Vascular Endothelial Growth Factor A/metabolism , Angiogenesis Inhibitors/pharmacology , Animals , Cell Transformation, Neoplastic/drug effects , Humans , Neoplasms/blood supply , Neoplasms/pathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism
20.
Nat Rev Clin Oncol ; 9(7): 378-90, 2012 Apr 24.
Article in English | MEDLINE | ID: mdl-22525710

ABSTRACT

Cancer cells rely on angiogenesis to fulfil their need for oxygen and nutrients; hence, agents targeting angiogenic pathways and mediators have been investigated as potential cancer drugs. Although this strategy has demonstrated delayed tumour progression--leading to progression-free survival and overall survival benefits compared with standard therapy--in some patients, the results are more modest than predicted. A significant number of patients either do not respond to antiangiogenic agents or fairly rapidly develop resistance to them, which raises questions about how resistance develops and how it can be overcome. Furthermore, whether cancers, once they develop resistance, become more invasive or lead to metastatic disease remains unclear. Several mechanisms of resistance have been recently proposed and emerging evidence indicates that, under certain experimental conditions, antiangiogenic agents increase intratumour hypoxia by promoting vessel pruning and inhibiting neoangiogenesis. Indeed, several studies have highlighted the possibility that inhibitors of VEGF (and its receptors) can promote an invasive metastatic switch, in part by creating an increasingly hypoxic tumour microenvironment. As a potential remedy, a number of therapeutic approaches have been investigated that target the hypoxic tumour compartment to improve the clinical outcome of antiangiogenic therapy.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Drug Resistance, Neoplasm/drug effects , Hypoxia/drug therapy , Neoplasms/blood supply , Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Animals , Humans
SELECTION OF CITATIONS
SEARCH DETAIL