Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Cell Chem Biol ; 28(12): 1693-1702.e6, 2021 12 16.
Article in English | MEDLINE | ID: mdl-34192523

ABSTRACT

Ganciclovir (GCV) is the first-line therapy against human cytomegalovirus (HCMV), a widespread infection that is particularly dangerous for immunodeficient individuals. Closely resembling deoxyguanosine triphosphate, the tri-phosphorylated metabolite of GCV (GCV-TP) is preferentially incorporated by the viral DNA polymerase, thereby terminating chain extension and, eventually, viral replication. However, the treatment outcome of GCV varies greatly among individuals, therefore warranting better understanding of its metabolism. Here we show that NUDT15, a Nudix hydrolase known to metabolize thiopurine triphosphates, can similarly hydrolyze GCV-TP through biochemical studies and co-crystallization of the NUDT15/GCV-TP complex. More critically, GCV efficacy was potentiated in HCMV-infected cells following NUDT15 depletion by RNAi or inhibition by an in-house-developed, nanomolar NUDT15 inhibitor, TH8321, suggesting that pharmacological targeting of NUDT15 is a possible avenue to improve existing anti-HCMV regimens. Collectively, the data further implicate NUDT15 as a broad-spectrum metabolic regulator of nucleoside analog therapeutics, such as thiopurines and GCV.


Subject(s)
Antiviral Agents/pharmacology , Cytomegalovirus/drug effects , Ganciclovir/pharmacology , Pyrophosphatases/metabolism , Antiviral Agents/chemistry , Cell Line, Tumor , Female , Ganciclovir/chemistry , Humans , Hydrolysis , Microbial Sensitivity Tests , Recombinant Proteins/metabolism
2.
Nat Chem Biol ; 16(10): 1120-1128, 2020 10.
Article in English | MEDLINE | ID: mdl-32690945

ABSTRACT

The NUDIX hydrolase NUDT15 was originally implicated in sanitizing oxidized nucleotides, but was later shown to hydrolyze the active thiopurine metabolites, 6-thio-(d)GTP, thereby dictating the clinical response of this standard-of-care treatment for leukemia and inflammatory diseases. Nonetheless, its physiological roles remain elusive. Here, we sought to develop small-molecule NUDT15 inhibitors to elucidate its biological functions and potentially to improve NUDT15-dependent chemotherapeutics. Lead compound TH1760 demonstrated low-nanomolar biochemical potency through direct and specific binding into the NUDT15 catalytic pocket and engaged cellular NUDT15 in the low-micromolar range. We also employed thiopurine potentiation as a proxy functional readout and demonstrated that TH1760 sensitized cells to 6-thioguanine through enhanced accumulation of 6-thio-(d)GTP in nucleic acids. A biochemically validated, inactive structural analog, TH7285, confirmed that increased thiopurine toxicity takes place via direct NUDT15 inhibition. In conclusion, TH1760 represents the first chemical probe for interrogating NUDT15 biology and potential therapeutic avenues.


Subject(s)
Pyrophosphatases/antagonists & inhibitors , Pyrophosphatases/metabolism , Binding Sites , Cell Line , Drug Design , Drug Development , Escherichia coli , Humans , Inorganic Pyrophosphatase/antagonists & inhibitors , Inorganic Pyrophosphatase/genetics , Inorganic Pyrophosphatase/metabolism , Models, Molecular , Protein Binding , Protein Conformation , Pyrophosphatases/chemistry , Pyrophosphatases/genetics , Structure-Activity Relationship
4.
Mol Cancer Ther ; 17(8): 1637-1647, 2018 08.
Article in English | MEDLINE | ID: mdl-29769307

ABSTRACT

Inhibition of ataxia-telangiectasia mutated (ATM) during radiotherapy of glioblastoma multiforme (GBM) may improve tumor control by short-circuiting the response to radiation-induced DNA damage. A major impediment for clinical implementation is that current inhibitors have limited central nervous system (CNS) bioavailability; thus, the goal was to identify ATM inhibitors (ATMi) with improved CNS penetration. Drug screens and refinement of lead compounds identified AZ31 and AZ32. The compounds were then tested in vivo for efficacy and impact on tumor and healthy brain. Both AZ31 and AZ32 blocked the DNA damage response and radiosensitized GBM cells in vitro AZ32, with enhanced blood-brain barrier (BBB) penetration, was highly efficient in vivo as radiosensitizer in syngeneic and human, orthotopic mouse glioma model compared with AZ31. Furthermore, human glioma cell lines expressing mutant p53 or having checkpoint-defective mutations were particularly sensitive to ATMi radiosensitization. The mechanism for this p53 effect involves a propensity to undergo mitotic catastrophe relative to cells with wild-type p53. In vivo, apoptosis was >6-fold higher in tumor relative to healthy brain after exposure to AZ32 and low-dose radiation. AZ32 is the first ATMi with oral bioavailability shown to radiosensitize glioma and improve survival in orthotopic mouse models. These findings support the development of a clinical-grade, BBB-penetrating ATMi for the treatment of GBM. Importantly, because many GBMs have defective p53 signaling, the use of an ATMi concurrent with standard radiotherapy is expected to be cancer-specific, increase the therapeutic ratio, and maintain full therapeutic effect at lower radiation doses. Mol Cancer Ther; 17(8); 1637-47. ©2018 AACR.


Subject(s)
Blood-Brain Barrier/metabolism , Glioma/drug therapy , Protein Kinase Inhibitors/therapeutic use , Radiation-Sensitizing Agents/therapeutic use , Administration, Oral , Animals , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Cell Line, Tumor , Humans , Mice , Mice, Nude , Protein Kinase Inhibitors/pharmacology , Radiation-Sensitizing Agents/pharmacology
5.
Nat Commun ; 9(1): 250, 2018 01 17.
Article in English | MEDLINE | ID: mdl-29343827

ABSTRACT

With a diverse network of substrates, NUDIX hydrolases have emerged as a key family of nucleotide-metabolizing enzymes. NUDT5 (also called NUDIX5) has been implicated in ADP-ribose and 8-oxo-guanine metabolism and was recently identified as a rheostat of hormone-dependent gene regulation and proliferation in breast cancer cells. Here, we further elucidate the physiological relevance of known NUDT5 substrates and underscore the biological requirement for NUDT5 in gene regulation and proliferation of breast cancer cells. We confirm the involvement of NUDT5 in ADP-ribose metabolism and dissociate a relationship to oxidized nucleotide sanitation. Furthermore, we identify potent NUDT5 inhibitors, which are optimized to promote maximal NUDT5 cellular target engagement by CETSA. Lead compound, TH5427, blocks progestin-dependent, PAR-derived nuclear ATP synthesis and subsequent chromatin remodeling, gene regulation and proliferation in breast cancer cells. We herein present TH5427 as a promising, targeted inhibitor that can be used to further study NUDT5 activity and ADP-ribose metabolism.


Subject(s)
Enzyme Inhibitors/pharmacology , Progestins/metabolism , Pyrophosphatases/antagonists & inhibitors , Signal Transduction/drug effects , Adenosine Diphosphate Ribose/metabolism , Adenosine Triphosphate/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Cell Proliferation/genetics , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Female , HL-60 Cells , Humans , Molecular Structure , Pyrophosphatases/genetics , Pyrophosphatases/metabolism , RNA Interference , Substrate Specificity
7.
J Med Chem ; 60(10): 4279-4292, 2017 05 25.
Article in English | MEDLINE | ID: mdl-28508636

ABSTRACT

The dCTP pyrophosphatase 1 (dCTPase) is a nucleotide pool "housekeeping" enzyme responsible for the catabolism of canonical and noncanonical nucleoside triphosphates (dNTPs) and has been associated with cancer progression and cancer cell stemness. We have identified a series of piperazin-1-ylpyridazines as a new class of potent dCTPase inhibitors. Lead compounds increase dCTPase thermal and protease stability, display outstanding selectivity over related enzymes and synergize with a cytidine analogue against leukemic cells. This new class of dCTPase inhibitors lays the first stone toward the development of drug-like probes for the dCTPase enzyme.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Piperazines/chemistry , Piperazines/pharmacology , Pyridazines/chemistry , Pyridazines/pharmacology , Pyrophosphatases/antagonists & inhibitors , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Humans , Leukemia/drug therapy , Leukemia/enzymology , Molecular Docking Simulation , Pyrophosphatases/metabolism
8.
Nat Med ; 23(2): 256-263, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28067901

ABSTRACT

The cytostatic deoxycytidine analog cytarabine (ara-C) is the most active agent available against acute myelogenous leukemia (AML). Together with anthracyclines, ara-C forms the backbone of AML treatment for children and adults. In AML, both the cytotoxicity of ara-C in vitro and the clinical response to ara-C therapy are correlated with the ability of AML blasts to accumulate the active metabolite ara-C triphosphate (ara-CTP), which causes DNA damage through perturbation of DNA synthesis. Differences in expression levels of known transporters or metabolic enzymes relevant to ara-C only partially account for patient-specific differential ara-CTP accumulation in AML blasts and response to ara-C treatment. Here we demonstrate that the deoxynucleoside triphosphate (dNTP) triphosphohydrolase SAM domain and HD domain 1 (SAMHD1) promotes the detoxification of intracellular ara-CTP pools. Recombinant SAMHD1 exhibited ara-CTPase activity in vitro, and cells in which SAMHD1 expression was transiently reduced by treatment with the simian immunodeficiency virus (SIV) protein Vpx were dramatically more sensitive to ara-C-induced cytotoxicity. CRISPR-Cas9-mediated disruption of the gene encoding SAMHD1 sensitized cells to ara-C, and this sensitivity could be abrogated by ectopic expression of wild-type (WT), but not dNTPase-deficient, SAMHD1. Mouse models of AML lacking SAMHD1 were hypersensitive to ara-C, and treatment ex vivo with Vpx sensitized primary patient-derived AML blasts to ara-C. Finally, we identified SAMHD1 as a risk factor in cohorts of both pediatric and adult patients with de novo AML who received ara-C treatment. Thus, SAMHD1 expression levels dictate patient sensitivity to ara-C, providing proof-of-concept that the targeting of SAMHD1 by Vpx could be an attractive therapeutic strategy for potentiating ara-C efficacy in hematological malignancies.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Cytarabine/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Monomeric GTP-Binding Proteins/drug effects , Viral Regulatory and Accessory Proteins/pharmacology , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Antimetabolites, Antineoplastic/therapeutic use , Arabinofuranosylcytosine Triphosphate/metabolism , Child , Child, Preschool , Cytarabine/therapeutic use , Disease Models, Animal , Female , Humans , In Vitro Techniques , Infant , Leukemia, Myeloid, Acute/metabolism , Male , Mice , Molecular Targeted Therapy , Monomeric GTP-Binding Proteins/metabolism , Prognosis , SAM Domain and HD Domain-Containing Protein 1
9.
Cancer Res ; 76(18): 5501-11, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27530327

ABSTRACT

Thiopurines are a standard treatment for childhood leukemia, but like all chemotherapeutics, their use is limited by inherent or acquired resistance in patients. Recently, the nucleoside diphosphate hydrolase NUDT15 has received attention on the basis of its ability to hydrolyze the thiopurine effector metabolites 6-thio-deoxyGTP (6-thio-dGTP) and 6-thio-GTP, thereby limiting the efficacy of thiopurines. In particular, increasing evidence suggests an association between the NUDT15 missense variant, R139C, and thiopurine sensitivity. In this study, we elucidated the role of NUDT15 and NUDT15 R139C in thiopurine metabolism. In vitro and cellular results argued that 6-thio-dGTP and 6-thio-GTP are favored substrates for NUDT15, a finding supported by a crystallographic determination of NUDT15 in complex with 6-thio-GMP. We found that NUDT15 R139C mutation did not affect enzymatic activity but instead negatively influenced protein stability, likely due to a loss of supportive intramolecular bonds that caused rapid proteasomal degradation in cells. Mechanistic investigations in cells indicated that NUDT15 ablation potentiated induction of the DNA damage checkpoint and cancer cell death by 6-thioguanine. Taken together, our results defined how NUDT15 limits thiopurine efficacy and how genetic ablation via the R139C missense mutation confers sensitivity to thiopurine treatment in patients. Cancer Res; 76(18); 5501-11. ©2016 AACR.


Subject(s)
Antimetabolites, Antineoplastic/metabolism , Antimetabolites, Antineoplastic/pharmacology , Pyrophosphatases/metabolism , Thioguanine/metabolism , Thioguanine/pharmacology , Blotting, Western , Cell Line, Tumor , Chromatography, High Pressure Liquid , Crystallography, X-Ray , Humans , Mutation, Missense , Protein Stability , Pyrophosphatases/chemistry , Pyrophosphatases/genetics
10.
DNA Repair (Amst) ; 44: 193-204, 2016 08.
Article in English | MEDLINE | ID: mdl-27311542

ABSTRACT

Artificially modified nucleotides, in the form of nucleoside analogues, are widely used in the treatment of cancers and various other diseases, and have become important tools in the laboratory to characterise DNA repair pathways. In contrast, the role of endogenously occurring nucleotide modifications in genome stability is little understood. This is despite the demonstration over three decades ago that the cellular DNA precursor pool is orders of magnitude more susceptible to modification than the DNA molecule itself. More recently, underscoring the importance of this topic, oxidation of the cellular nucleotide pool achieved through targeting the sanitation enzyme MTH1, appears to be a promising anti-cancer strategy. This article reviews our current understanding of modified DNA precursors in genome stability, with a particular focus upon oxidised nucleotides, and outlines some important outstanding questions.


Subject(s)
DNA Repair Enzymes/metabolism , DNA Repair , DNA/metabolism , Nucleic Acid Precursors/metabolism , Nucleotides/biosynthesis , Phosphoric Monoester Hydrolases/metabolism , Pyrophosphatases/metabolism , DNA/chemistry , DNA/genetics , DNA Damage , DNA Repair Enzymes/genetics , Genome, Human , Genomic Instability , Humans , Nucleic Acid Precursors/chemistry , Nucleic Acid Precursors/genetics , Nucleotides/chemistry , Oxidation-Reduction , Oxidative Stress , Phosphoric Monoester Hydrolases/genetics , Pyrophosphatases/genetics , Nudix Hydrolases
11.
J Med Chem ; 59(3): 1140-1148, 2016 Feb 11.
Article in English | MEDLINE | ID: mdl-26771665

ABSTRACT

The dCTPase pyrophosphatase 1 (dCTPase) regulates the intracellular nucleotide pool through hydrolytic degradation of canonical and noncanonical nucleotide triphosphates (dNTPs). dCTPase is highly expressed in multiple carcinomas and is associated with cancer cell stemness. Here we report on the development of the first potent and selective dCTPase inhibitors that enhance the cytotoxic effect of cytidine analogues in leukemia cells. Boronate 30 displays a promising in vitro ADME profile, including plasma and mouse microsomal half-lives, aqueous solubility, cell permeability and CYP inhibition, deeming it a suitable compound for in vivo studies.


Subject(s)
Drug Discovery , Enzyme Inhibitors/pharmacology , Pyrophosphatases/antagonists & inhibitors , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , HL-60 Cells , Humans , Ligands , Molecular Structure , Pyrophosphatases/metabolism , Structure-Activity Relationship
12.
Nat Commun ; 6: 7871, 2015 Aug 04.
Article in English | MEDLINE | ID: mdl-26238318

ABSTRACT

Deregulated redox metabolism in cancer leads to oxidative damage to cellular components including deoxyribonucleoside triphosphates (dNTPs). Targeting dNTP pool sanitizing enzymes, such as MTH1, is a highly promising anticancer strategy. The MTH2 protein, known as NUDT15, is described as the second human homologue of bacterial MutT with 8-oxo-dGTPase activity. We present the first NUDT15 crystal structure and demonstrate that NUDT15 prefers other nucleotide substrates over 8-oxo-dGTP. Key structural features are identified that explain different substrate preferences for NUDT15 and MTH1. We find that depletion of NUDT15 has no effect on incorporation of 8-oxo-dGTP into DNA and does not impact cancer cell survival in cell lines tested. NUDT17 and NUDT18 were also profiled and found to have far less activity than MTH1 against oxidized nucleotides. We show that NUDT15 is not a biologically relevant 8-oxo-dGTPase, and that MTH1 is the most prominent sanitizer of the cellular dNTP pool known to date.


Subject(s)
DNA Repair Enzymes/metabolism , Deoxyguanine Nucleotides/metabolism , Deoxyribonucleotides/metabolism , Oxidation-Reduction , Oxidative Stress , Phosphoric Monoester Hydrolases/metabolism , Pyrophosphatases/metabolism , Blotting, Western , Cell Line, Tumor , Cell Survival , Crystallization , HCT116 Cells , HeLa Cells , Humans , MCF-7 Cells , Pyrophosphatases/chemistry , Substrate Specificity
13.
Clin Cancer Res ; 19(12): 3189-200, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23620409

ABSTRACT

PURPOSE: Glioblastoma multiforme (GBM) is the most lethal form of brain cancer with a median survival of only 12 to 15 months. Current standard treatment consists of surgery followed by chemoradiation. The poor survival of patients with GBM is due to aggressive tumor invasiveness, an inability to remove all tumor tissue, and an innate tumor chemo- and radioresistance. Ataxia-telangiectasia mutated (ATM) is an excellent target for radiosensitizing GBM because of its critical role in regulating the DNA damage response and p53, among other cellular processes. As a first step toward this goal, we recently showed that the novel ATM kinase inhibitor KU-60019 reduced migration, invasion, and growth, and potently radiosensitized human glioma cells in vitro. EXPERIMENTAL DESIGN: Using orthotopic xenograft models of GBM, we now show that KU-60019 is also an effective radiosensitizer in vivo. Human glioma cells expressing reporter genes for monitoring tumor growth and dispersal were grown intracranially, and KU-60019 was administered intratumorally by convection-enhanced delivery or osmotic pump. RESULTS: Our results show that the combined effect of KU-60019 and radiation significantly increased survival of mice 2- to 3-fold over controls. Importantly, we show that glioma with mutant p53 is much more sensitive to KU-60019 radiosensitization than genetically matched wild-type glioma. CONCLUSIONS: Taken together, our results suggest that an ATM kinase inhibitor may be an effective radiosensitizer and adjuvant therapy for patients with mutant p53 brain cancers.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/genetics , Brain Neoplasms/therapy , Glioma/therapy , Morpholines/administration & dosage , Thioxanthenes/administration & dosage , Animals , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Brain Neoplasms/pathology , Cell Line, Tumor , Flow Cytometry , Gene Expression Regulation, Neoplastic/drug effects , Glioma/pathology , Humans , Mice , Mutation , Radiation Tolerance/drug effects , Radiation, Ionizing , Tumor Suppressor Protein p53/genetics
14.
Biochem Pharmacol ; 85(7): 888-97, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23287412

ABSTRACT

Glioblastoma multiforme (GBM) are brain tumors that are exceptionally resistant to both radio- and chemotherapy regimens and novel approaches to treatment are needed. T-type calcium channels are one type of low voltage-gated channel (LVCC) involved in embryonic cell proliferation and differentiation; however they are often over-expressed in tumors, including GBM. In this study, we found that inhibition of T-type Ca(2+) channels in GBM cells significantly reduced their survival and resistance to therapy. Moreover, either T-type selective antagonists, such as mibefradil, or siRNA-mediated knockdown of the T-type channel alpha subunits not only reduced cell viability and clonogenic potential, but also induced apoptosis. In response to channel blockade or ablation, we observed reduced phosphorylation of Akt and Rictor, suggesting inhibition of the mTORC2/Akt pathway. This was followed by reduction in phosphorylation of anti-apoptotic Bad and caspases activation. The apoptotic response was specific for T-type Ca(2+) channels, as inhibition of L-type Ca(2+) channels did not induce similar effects. Our results implicate T-type Ca(2+) channels as distinct entities for survival signaling in GBM cells and suggest that they are a novel molecular target for tumor therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Brain Neoplasms/drug therapy , Calcium Channel Blockers/pharmacology , Calcium Channels, T-Type/metabolism , Glioblastoma/drug therapy , Proto-Oncogene Proteins c-akt/metabolism , Radiation-Sensitizing Agents/pharmacology , Calcium Channels, T-Type/genetics , Cell Line, Tumor/drug effects , Cell Line, Tumor/radiation effects , Cell Survival/drug effects , Humans , Mechanistic Target of Rapamycin Complex 2 , Mibefradil/pharmacology , Multiprotein Complexes/metabolism , Phosphorylation , RNA, Small Interfering/genetics , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , bcl-Associated Death Protein/metabolism
15.
J Biol Chem ; 287(12): 9230-9, 2012 Mar 16.
Article in English | MEDLINE | ID: mdl-22298787

ABSTRACT

DNA-dependent protein kinase (DNA-PK) becomes activated in response to DNA double strand breaks, initiating repair by the non-homologous end joining pathway. DNA·PK complexes with the regulatory subunit SAPSR1 (R1) of protein phosphatase-6 (PP6). Knockdown of either R1 or PP6c prevents DNA-PK activation in response to ionizing radiation-induced DNA damage and radiosensitizes glioblastoma cells. Here, we demonstrate that R1 is necessary for and bridges the interaction between DNA-PK and PP6c. Using R1 deletion mutants, DNA-PK binding was mapped to two distinct regions of R1 spanning residues 1-326 and 522-700. Either region expressed alone was sufficient to bind DNA-PK, but only deletion of residues 1-326, not 522-700, eliminated interaction of R1 with DNA-PK. We assign 1-326 as the dominant domain and 522-700 as the supporting region. These results demonstrate that R1 acts as a bidentate anchor to DNA-PK and recruits PP6c. Targeting the dominant interface with small molecule or peptidomimetic inhibitors could specifically prevent activation of DNA-PK and thereby sensitize cells to ionizing radiation and other genotoxic agents.


Subject(s)
DNA-Activated Protein Kinase/metabolism , Phosphoprotein Phosphatases/metabolism , Amino Acid Motifs , Cell Line , DNA-Activated Protein Kinase/chemistry , DNA-Activated Protein Kinase/genetics , Humans , Phosphoprotein Phosphatases/chemistry , Phosphoprotein Phosphatases/genetics , Protein Binding
16.
Cancer Res ; 71(21): 6817-26, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21903767

ABSTRACT

Radiotherapy combined with androgen depletion is generally successful for treating locally advanced prostate cancer. However, radioresistance that contributes to recurrence remains a major therapeutic problem in many patients. In this study, we define the high-affinity neurotensin receptor 1 (NTR1) as a tractable new molecular target to radiosensitize prostate cancers. The selective NTR1 antagonist SR48692 sensitized prostate cancer cells in a dose- and time-dependent manner, increasing apoptotic cell death and decreasing clonogenic survival. The observed cancer selectivity for combinations of SR48692 and radiation reflected differential expression of NTR1, which is highly expressed in prostate cancer cells but not in normal prostate epithelial cells. Radiosensitization was not affected by androgen dependence or androgen receptor expression status. NTR1 inhibition in cancer cell-attenuated epidermal growth factor receptor activation and downstream signaling, whether induced by neurotensin or ionizing radiation, establish a molecular mechanism for sensitization. Most notably, SR48692 efficiently radiosensitized PC-3M orthotopic human tumor xenografts in mice, and significantly reduced tumor burden. Taken together, our findings offer preclinical proof of concept for targeting the NTR1 receptor as a strategy to improve efficacy and outcomes of prostate cancer treatments using radiotherapy.


Subject(s)
Adenocarcinoma/radiotherapy , Neoplasm Proteins/antagonists & inhibitors , Prostatic Neoplasms/radiotherapy , Pyrazoles/therapeutic use , Quinolines/therapeutic use , Radiation-Sensitizing Agents/therapeutic use , Receptors, Neurotensin/antagonists & inhibitors , Adenocarcinoma/pathology , Androgens , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Line, Tumor/drug effects , Cell Line, Tumor/radiation effects , Epidermal Growth Factor/pharmacology , ErbB Receptors/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice , Mice, Nude , Neoplasm Proteins/physiology , Neoplasms, Hormone-Dependent/pathology , Neoplasms, Hormone-Dependent/radiotherapy , Phosphorylation/drug effects , Phosphorylation/radiation effects , Prostatic Neoplasms/pathology , Protein Processing, Post-Translational/drug effects , Protein Processing, Post-Translational/radiation effects , Pyrazoles/pharmacology , Quinolines/pharmacology , Radiation Tolerance/drug effects , Radiation Tolerance/physiology , Radiation-Sensitizing Agents/pharmacology , Receptors, Androgen/analysis , Receptors, Neurotensin/physiology , Tumor Stem Cell Assay , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...