Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Br J Pharmacol ; 178(3): 600-613, 2021 02.
Article in English | MEDLINE | ID: mdl-33125717

ABSTRACT

BACKGROUND AND PURPOSE: Savolitinib (AZD6094, HMPL-504, volitinib) is an oral, potent, and highly MET receptor TK inhibitor. This series of studies aimed to develop a pharmacokinetic-pharmacodynamic (PK/PD) model to link inhibition of MET phosphorylation (pMET) by savolitinib with anti-tumour activity. EXPERIMENTAL APPROACH: Cell line-derived xenograft (CDX) experiments using human lung cancer (EBC-1) and gastric cancer (MKN-45) cells were conducted in athymic nude mice using a variety of doses and schedules of savolitinib. Tumour pMET changes and growth inhibition were calculated after 28 days. Population PK/PD techniques were used to construct a PK/PD model for savolitinib. KEY RESULTS: Savolitinib showed dose- and dose frequency-dependent anti-tumour activity in the CDX models, with more frequent, lower dosing schedules (e.g., twice daily) being more effective than intermittent, higher dosing schedules (e.g., 4 days on/3 days off or 2 days on/5 days off). There was a clear exposure-response relationship, with maximal suppression of pMET of >90%. Data from additional CDX and patient-derived xenograft (PDX) models overlapped, allowing calculation of a single EC50 of 0.38 ng·ml-1 . Tumour growth modelling demonstrated that prolonged, high levels of pMET inhibition (>90%) were required for tumour stasis and regression in the models. CONCLUSION AND IMPLICATIONS: High and persistent levels of MET inhibition by savolitinib were needed for optimal monotherapy anti-tumour activity in preclinical models. The modelling framework developed here can be used to translate tumour growth inhibition from the mouse to human and thus guide choice of clinical dose and schedule.


Subject(s)
Antineoplastic Agents , Proto-Oncogene Proteins c-met , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Mice , Mice, Nude , Protein Kinase Inhibitors/pharmacology , Pyrazines , Triazines , Xenograft Model Antitumor Assays
2.
Mol Cancer ; 16(1): 177, 2017 12 06.
Article in English | MEDLINE | ID: mdl-29212548

ABSTRACT

Efforts to develop effective cancer therapeutics have been hindered by a lack of clinically predictive preclinical models which recapitulate this complex disease. Patient derived xenograft (PDX) models have emerged as valuable tools for translational research but have several practical limitations including lack of sustained growth in vitro. In this study, we utilized Conditional Reprogramming (CR) cell technology- a novel cell culture system facilitating the generation of stable cultures from patient biopsies- to establish PDX-derived cell lines which maintain the characteristics of the parental PDX tumor. Human lung and ovarian PDX tumors were successfully propagated using CR technology to create stable explant cell lines (CR-PDX). These CR-PDX cell lines maintained parental driver mutations and allele frequency without clonal drift. Purified CR-PDX cell lines were amenable to high throughput chemosensitivity screening and in vitro genetic knockdown studies. Additionally, re-implanted CR-PDX cells proliferated to form tumors that retained the growth kinetics, histology, and drug responses of the parental PDX tumor. CR technology can be used to generate and expand stable cell lines from PDX tumors without compromising fundamental biological properties of the model. It offers the ability to expand PDX cells in vitro for subsequent 2D screening assays as well as for use in vivo to reduce variability, animal usage and study costs. The methods and data detailed here provide a platform to generate physiologically relevant and predictive preclinical models to enhance drug discovery efforts.


Subject(s)
Cell Line, Tumor/cytology , Cellular Reprogramming Techniques/methods , Lung Neoplasms/pathology , Ovarian Neoplasms/pathology , Animals , Cell Line, Tumor/pathology , Drug Screening Assays, Antitumor , Female , Humans , Lung Neoplasms/genetics , Male , Mice , Mutation , Ovarian Neoplasms/genetics , Xenograft Model Antitumor Assays
3.
Oncotarget ; 7(36): 57651-57670, 2016 Sep 06.
Article in English | MEDLINE | ID: mdl-27472392

ABSTRACT

Lung cancer is the most common cause of cancer death globally with a significant, unmet need for more efficacious treatments. The receptor tyrosine kinase MET has been implicated as an oncogene in numerous cancer subtypes, including non-small cell lung cancer (NSCLC). Here we explore the therapeutic potential of savolitinib (volitinib, AZD6094, HMPL-504), a potent and selective MET inhibitor, in NSCLC. In vitro, savolitinib inhibits MET phosphorylation with nanomolar potency, which correlates with blockade of PI3K/AKT and MAPK signaling as well as MYC down-regulation. In vivo, savolitinib causes inhibition of these pathways and significantly decreases growth of MET-dependent xenografts. To understand resistance mechanisms, we generated savolitinib resistance in MET-amplified NSCLC cell lines and analyzed individual clones. We found that upregulation of MYC and constitutive mTOR pathway activation is a conserved feature of resistant clones that can be overcome by knockdown of MYC or dual mTORC1/2 inhibition. Lastly, we demonstrate that mechanisms of resistance are heterogeneous, arising via a switch to EGFR dependence or by a requirement for PIM signaling. This work demonstrates the efficacy of savolitinib in NSCLC and characterizes acquired resistance, identifying both known and novel mechanisms that may inform combination strategies in the clinic.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Drug Resistance, Neoplasm , Lung Neoplasms/drug therapy , Proto-Oncogene Proteins c-myc/metabolism , Pyrazines/chemistry , TOR Serine-Threonine Kinases/metabolism , Triazines/chemistry , Animals , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Survival , Down-Regulation , ErbB Receptors/metabolism , Female , Humans , Lung Neoplasms/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-met/metabolism
4.
Oncotarget ; 7(34): 54120-54136, 2016 Aug 23.
Article in English | MEDLINE | ID: mdl-27472462

ABSTRACT

Although endocrine therapy is successfully used to treat patients with estrogen receptor (ER) positive breast cancer, a substantial proportion of this population will relapse. Several mechanisms of acquired resistance have been described including activation of the mTOR pathway, increased activity of CDK4 and activating mutations in ER. Using a patient derived xenograft model harboring a common activating ER ligand binding domain mutation (D538G), we evaluated several combinatorial strategies using the selective estrogen receptor degrader (SERD) fulvestrant in combination with chromatin modifying agents, and CDK4/6 and mTOR inhibitors. In this model, fulvestrant binds WT and MT ER, reduces ER protein levels, and downregulated ER target gene expression. Addition of JQ1 or vorinostat to fulvestrant resulted in tumor regression (41% and 22% regression, respectively) though no efficacy was seen when either agent was given alone. Interestingly, although the CDK4/6 inhibitor palbociclib and mTOR inhibitor everolimus were efficacious as monotherapies, long-term delayed tumor growth was only observed when co-administered with fulvestrant. This observation was consistent with a greater inhibition of compensatory signaling when palbociclib and everolimus were co-dosed with fulvestrant. The addition of fulvestrant to JQ1, vorinostat, everolimus and palbociclib also significantly reduced lung metastatic burden as compared to monotherapy. The combination potential of fulvestrant with palbociclib or everolimus were confirmed in an MCF7 CRISPR model harboring the Y537S ER activating mutation. Taken together, these data suggest that fulvestrant may have an important role in the treatment of ER positive breast cancer with acquired ER mutations.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Mutation , Receptors, Estrogen/genetics , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Drug Resistance, Neoplasm , Estradiol/administration & dosage , Estradiol/analogs & derivatives , Everolimus/administration & dosage , Female , Fulvestrant , Humans , MCF-7 Cells , Mice , Piperazines/administration & dosage , Pyridines/administration & dosage , Receptors, Estrogen/analysis , Xenograft Model Antitumor Assays
5.
Cancer Res ; 76(11): 3307-18, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27020862

ABSTRACT

Fulvestrant is an estrogen receptor (ER) antagonist administered to breast cancer patients by monthly intramuscular injection. Given its present limitations of dosing and route of administration, a more flexible orally available compound has been sought to pursue the potential benefits of this drug in patients with advanced metastatic disease. Here we report the identification and characterization of AZD9496, a nonsteroidal small-molecule inhibitor of ERα, which is a potent and selective antagonist and downregulator of ERα in vitro and in vivo in ER-positive models of breast cancer. Significant tumor growth inhibition was observed as low as 0.5 mg/kg dose in the estrogen-dependent MCF-7 xenograft model, where this effect was accompanied by a dose-dependent decrease in PR protein levels, demonstrating potent antagonist activity. Combining AZD9496 with PI3K pathway and CDK4/6 inhibitors led to further growth-inhibitory effects compared with monotherapy alone. Tumor regressions were also seen in a long-term estrogen-deprived breast model, where significant downregulation of ERα protein was observed. AZD9496 bound and downregulated clinically relevant ESR1 mutants in vitro and inhibited tumor growth in an ESR1-mutant patient-derived xenograft model that included a D538G mutation. Collectively, the pharmacologic evidence showed that AZD9496 is an oral, nonsteroidal, selective estrogen receptor antagonist and downregulator in ER(+) breast cells that could provide meaningful benefit to ER(+) breast cancer patients. AZD9496 is currently being evaluated in a phase I clinical trial. Cancer Res; 76(11); 3307-18. ©2016 AACR.


Subject(s)
Breast Neoplasms/drug therapy , Cinnamates/pharmacology , Estrogen Receptor Modulators/pharmacology , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/genetics , Indoles/pharmacology , Mutation/genetics , Administration, Oral , Animals , Apoptosis/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Cinnamates/administration & dosage , Drug Evaluation, Preclinical , Estrogen Receptor Modulators/administration & dosage , Estrogen Receptor alpha/chemistry , Female , Humans , Indoles/administration & dosage , Mice , Mice, Inbred NOD , Mice, SCID , Protein Conformation , Rats , Tumor Cells, Cultured , Uterus/metabolism , Uterus/pathology , Xenograft Model Antitumor Assays
6.
Oncotarget ; 7(15): 20773-87, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-26980748

ABSTRACT

The tumor microenvironment is emerging as a key regulator of cancer growth and progression, however the exact mechanisms of interaction with the tumor are poorly understood. Whilst the majority of genomic profiling efforts thus far have focused on the tumor, here we investigate RNA-Seq as a hypothesis-free tool to generate independent tumor and stromal biomarkers, and explore tumor-stroma interactions by exploiting the human-murine compartment specificity of patient-derived xenografts (PDX).Across a pan-cancer cohort of 79 PDX models, we determine that mouse stroma can be separated into distinct clusters, each corresponding to a specific stromal cell type. This implies heterogeneous recruitment of mouse stroma to the xenograft independent of tumor type. We then generate cross-species expression networks to recapitulate a known association between tumor epithelial cells and fibroblast activation, and propose a potentially novel relationship between two hypoxia-associated genes, human MIF and mouse Ddx6. Assessment of disease subtype also reveals MMP12 as a putative stromal marker of triple-negative breast cancer. Finally, we establish that our ability to dissect recruited stroma from trans-differentiated tumor cells is crucial to identifying stem-like poor-prognosis signatures in the tumor compartment.In conclusion, RNA-Seq is a powerful, cost-effective solution to global analysis of human tumor and mouse stroma simultaneously, providing new insights into mouse stromal heterogeneity and compartment-specific disease markers that are otherwise overlooked by alternative technologies. The study represents the first comprehensive analysis of its kind across multiple PDX models, and supports adoption of the approach in pre-clinical drug efficacy studies, and compartment-specific biomarker discovery.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/pathology , Epithelial Cells/pathology , Gene Expression Profiling/methods , Stromal Cells/pathology , Transcriptome , Tumor Microenvironment/genetics , Animals , Breast Neoplasms/genetics , Epithelial Cells/metabolism , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Stromal Cells/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
7.
Mol Cancer Ther ; 14(11): 2508-18, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26358751

ABSTRACT

mTOR is an atypical serine threonine kinase involved in regulating major cellular functions, such as nutrients sensing, growth, and proliferation. mTOR is part of the multiprotein complexes mTORC1 and mTORC2, which have been shown to play critical yet functionally distinct roles in the regulation of cellular processes. Current clinical mTOR inhibitors only inhibit the mTORC1 complex and are derivatives of the macrolide rapamycin (rapalogs). Encouraging effects have been observed with rapalogs in estrogen receptor-positive (ER(+)) breast cancer patients in combination with endocrine therapy, such as aromatase inhibitors. AZD2014 is a small-molecule ATP competitive inhibitor of mTOR that inhibits both mTORC1 and mTORC2 complexes and has a greater inhibitory function against mTORC1 than the clinically approved rapalogs. Here, we demonstrate that AZD2014 has broad antiproliferative effects across multiple cell lines, including ER(+) breast models with acquired resistance to hormonal therapy and cell lines with acquired resistance to rapalogs. In vivo, AZD2014 induces dose-dependent tumor growth inhibition in several xenograft and primary explant models. The antitumor activity of AZD2014 is associated with modulation of both mTORC1 and mTORC2 substrates, consistent with its mechanism of action. In combination with fulvestrant, AZD2014 induces tumor regressions when dosed continuously or using intermittent dosing schedules. The ability to dose AZD2014 intermittently, together with its ability to block signaling from both mTORC1 and mTORC2 complexes, makes this compound an ideal candidate for combining with endocrine therapies in the clinic. AZD2014 is currently in phase II clinical trials.


Subject(s)
Breast Neoplasms/drug therapy , Morpholines/pharmacology , Multiprotein Complexes/antagonists & inhibitors , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzamides , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Administration Schedule , Estradiol/administration & dosage , Estradiol/analogs & derivatives , Estradiol/pharmacology , Female , Fulvestrant , HEK293 Cells , Humans , Immunoblotting , MCF-7 Cells , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Morpholines/administration & dosage , Morpholines/chemistry , Multiprotein Complexes/metabolism , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Pyrimidines , Receptors, Estrogen/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Tumor Burden/drug effects , Xenograft Model Antitumor Assays/methods
8.
Mol Cancer Ther ; 14(11): 2441-51, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26351323

ABSTRACT

AKT1(E17K) mutations occur at low frequency in a variety of solid tumors, including those of the breast and urinary bladder. Although this mutation has been shown to transform rodent cells in culture, it was found to be less oncogenic than PIK3CA mutations in breast epithelial cells. Moreover, the therapeutic potential of AKT inhibitors in human tumors with an endogenous AKT1(E17K) mutation is not known. Expression of exogenous copies of AKT1(E17K) in MCF10A breast epithelial cells increased phosphorylation of AKT and its substrates, induced colony formation in soft agar, and formation of lesions in the mammary fat pad of immunodeficient mice. These effects were inhibited by the allosteric and catalytic AKT inhibitors MK-2206 and AZD5363, respectively. Both AKT inhibitors caused highly significant growth inhibition of breast cancer explant models with AKT1(E17K) mutation. Furthermore, in a phase I clinical study, the catalytic Akt inhibitor AZD5363 induced partial responses in patients with breast and ovarian cancer with tumors containing AKT1(E17K) mutations. In MGH-U3 bladder cancer xenografts, which contain both AKT1(E17K) and FGFR3(Y373C) mutations, AZD5363 monotherapy did not significantly reduce tumor growth, but tumor regression was observed in combination with the FGFR inhibitor AZD4547. The data show that tumors with AKT1(E17K) mutations are rational therapeutic targets for AKT inhibitors, although combinations with other targeted agents may be required where activating oncogenic mutations of other proteins are present in the same tumor.


Subject(s)
Mutation, Missense , Neoplasms/genetics , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction/genetics , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Blotting, Western , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Doxycycline/pharmacology , Female , Heterocyclic Compounds, 3-Ring/administration & dosage , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , Male , Mice, Nude , Neoplasms/drug therapy , Neoplasms/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Pyrimidines/administration & dosage , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Pyrroles/administration & dosage , Pyrroles/pharmacology , Pyrroles/therapeutic use , Signal Transduction/drug effects , Xenograft Model Antitumor Assays/methods
9.
Cancer Chemother Pharmacol ; 76(2): 343-56, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26092323

ABSTRACT

PURPOSE: Intermittent dosing of inhibitors of the PI3K/AKT/mTOR network offers the potential to maximize the therapeutic margin. Here, we validate a predictive modeling approach to establish the relative efficacy of continuous and two intermittent dosing schedules of the AKT inhibitor AZD5363. METHODS: A mathematical model of pharmacokinetics, pharmacodynamics and anti-tumor effect was constructed based upon experimental data from dosing regimens that give constant and transient inhibition of the AKT pathway. RESULTS: Continuous and intermittent dosing of AZD5363 inhibited growth of BT474c xenografts and caused dose- and time-dependent inhibition of AKT substrate phosphorylation. Both dosing schedules inhibited proliferation, but a higher intermittent dose also induced apoptosis. The mathematical model described this pharmacodynamic and efficacy data well, for both monotherapy and combination dosing with docetaxel, and predicted that equivalent efficacy could be achieved at 1.3- and 1.7× continuous dose when AZD5363 was dosed intermittently for 4 and 2 days per week, respectively. These predictions were confirmed in two independent xenograft models. Moreover, the model also correctly predicted the relative efficacy of three different sequences of intermittent dosing of AZD5363 with docetaxel. CONCLUSIONS: Equivalent anti-tumor activity to continuous dosing can be achieved at modestly increased intermittent doses of AZD5363. These intermittent dosing regimens may potentially overcome tolerability issues seen with continuous dosing and enable greater flexibility of dosing schedule in combination with other agents, including chemotherapy.


Subject(s)
Antineoplastic Agents/administration & dosage , Models, Biological , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Pyrimidines/administration & dosage , Pyrroles/administration & dosage , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis , Biomarkers, Pharmacological/metabolism , Cell Line, Tumor , Docetaxel , Drug Administration Schedule , Drug Interactions , Female , Heterografts , Mice, Nude , Neoplasm Transplantation , Pyrimidines/pharmacokinetics , Pyrimidines/pharmacology , Pyrroles/pharmacokinetics , Pyrroles/pharmacology , Taxoids/administration & dosage , Taxoids/pharmacokinetics , Taxoids/pharmacology , Tumor Burden/drug effects
10.
Clin Cancer Res ; 21(12): 2811-9, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25779944

ABSTRACT

PURPOSE: Papillary renal cell carcinoma (PRCC) is the second most common cancer of the kidney and carries a poor prognosis for patients with nonlocalized disease. The HGF receptor MET plays a central role in PRCC and aberrations, either through mutation, copy number gain, or trisomy of chromosome 7 occurring in the majority of cases. The development of effective therapies in PRCC has been hampered in part by a lack of available preclinical models. We determined the pharmacodynamic and antitumor response of the selective MET inhibitor AZD6094 in two PRCC patient-derived xenograft (PDX) models. EXPERIMENTAL DESIGN: Two PRCC PDX models were identified and MET mutation status and copy number determined. Pharmacodynamic and antitumor activity of AZD6094 was tested using a dose response up to 25 mg/kg daily, representing clinically achievable exposures, and compared with the activity of the RCC standard-of-care sunitinib (in RCC43b) or the multikinase inhibitor crizotinib (in RCC47). RESULTS: AZD6094 treatment resulted in tumor regressions, whereas sunitinib or crizotinib resulted in unsustained growth inhibition. Pharmacodynamic analysis of tumors revealed that AZD6094 could robustly suppress pMET and the duration of target inhibition was dose related. AZD6094 inhibited multiple signaling nodes, including MAPK, PI3K, and EGFR. Finally, at doses that induced tumor regression, AZD6094 resulted in a dose- and time-dependent induction of cleaved PARP, a marker of cell death. CONCLUSIONS: Data presented provide the first report testing therapeutics in preclinical in vivo models of PRCC and support the clinical development of AZD6094 in this indication.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Papillary/metabolism , Carcinoma, Papillary/pathology , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Pyrazines/pharmacology , Triazines/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Carcinoma, Papillary/drug therapy , Carcinoma, Papillary/genetics , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/genetics , Cell Line, Tumor , Crizotinib , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Humans , Indoles/pharmacology , Protein Kinase Inhibitors/administration & dosage , Proto-Oncogene Proteins c-met/genetics , Pyrazines/administration & dosage , Pyrazoles/pharmacology , Pyridines/pharmacology , Pyrroles/pharmacology , Sunitinib , Triazines/administration & dosage , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
11.
Mol Cancer Ther ; 14(1): 48-58, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25398829

ABSTRACT

Loss of PTEN protein results in upregulation of the PI3K/AKT pathway, which appears dependent on the PI3Kß isoform. Inhibitors of PI3Kß have potential to reduce growth of tumors in which loss of PTEN drives tumor progression. We have developed a small-molecule inhibitor of PI3Kß and PI3Kδ (AZD8186) and assessed its antitumor activity across a panel of cell lines. We have then explored the antitumor effects as single agent and in combination with docetaxel in triple-negative breast (TNBC) and prostate cancer models. In vitro, AZD8186 inhibited growth of a range of cell lines. Sensitivity was associated with inhibition of the AKT pathway. Cells sensitive to AZD8186 (GI50 < 1 µmol/L) are enriched for, but not exclusively associated with, PTEN deficiency. In vivo, AZD8186 inhibits PI3K pathway biomarkers in prostate and TNBC tumors. Scheduling treatment with AZD8186 shows antitumor activity required only intermittent exposure, and that increased tumor control is achieved when AZD8186 is used in combination with docetaxel. AZD8186 is a potent inhibitor of PI3Kß with activity against PI3Kδ signaling, and has potential to reduce growth of tumors dependent on dysregulated PTEN for growth. Moreover, AZD8186 can be combined with docetaxel, a chemotherapy commonly used to treat advanced TBNC and prostate tumors. The ability to schedule AZD8186 and maintain efficacy offers opportunity to combine AZD8186 more effectively with other drugs.


Subject(s)
Aniline Compounds/administration & dosage , Antineoplastic Agents/administration & dosage , Chromones/administration & dosage , Phosphoinositide-3 Kinase Inhibitors , Prostatic Neoplasms/drug therapy , Signal Transduction/drug effects , Taxoids/administration & dosage , Triple Negative Breast Neoplasms/drug therapy , Aniline Compounds/pharmacology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Cell Line, Tumor , Cell Proliferation/drug effects , Chromones/pharmacology , Docetaxel , Drug Synergism , Female , Humans , Male , Mice , PTEN Phosphohydrolase/deficiency , Prostatic Neoplasms/metabolism , Triple Negative Breast Neoplasms/metabolism , Xenograft Model Antitumor Assays
12.
Oncotarget ; 6(4): 2407-20, 2015 Feb 10.
Article in English | MEDLINE | ID: mdl-25537515

ABSTRACT

Acquired resistance to PI3K/mTOR/Akt pathway inhibitors is often associated with compensatory feedback loops involving the activation of oncogenes. Here, we have generated everolimus resistance in ER+ breast cancer cells and in long-term estrogen deprived (LTED) models that mimic progression on anti-estrogens. This allowed us to uncover MYC as a driver of mTOR inhibitor resistance. We demonstrate that both everolimus resistance and acute treatment of everolimus can lead to the upregulation of MYC mRNA, protein expression and, consequently, the enrichment of MYC signatures as revealed by RNA sequencing data. Depletion of MYC resulted in resensitization to everolimus, confirming its functional importance in this setting. Furthermore, ChIP assays demonstrate that MYC upregulation in the everolimus resistant lines is mediated by increased association of the BRD4 transcription factor with the MYC gene. Finally, JQ1, a BRD4 inhibitor combined with everolimus exhibited increased tumor growth inhibition in 3D Matrigel models and an in vivo xenograft model. These data suggest that MYC plays an important role in mediating resistance to everolimus in ER+ and ER+/LTED models. Furthermore, given the regulation ofMYCby BRD4 in this setting, these data have implications for increased therapeutic potential of combining epigenetic agents with mTOR inhibitors to effectively downregulate otherwise difficult to target transcription factors such as MYC.


Subject(s)
Breast Neoplasms/drug therapy , Epigenesis, Genetic/drug effects , Everolimus/pharmacology , Proto-Oncogene Proteins c-myc/genetics , Animals , Antineoplastic Agents/pharmacology , Azepines/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Cycle Proteins , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/genetics , Drug Synergism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunoblotting , MCF-7 Cells , Mice, Nude , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-myc/metabolism , RNA Interference , Receptors, Estrogen/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription Factors/metabolism , Triazoles/pharmacology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
13.
Genome Biol ; 9(12): R180, 2008.
Article in English | MEDLINE | ID: mdl-19094238

ABSTRACT

The ability to detect activation of signaling pathways based solely on gene expression data represents an important goal in biological research. We tested the sensitivity of singular value decomposition-based regression by focusing on functional interactions between the Ras and transforming growth factor beta signaling pathways. Our findings demonstrate that this approach is sufficiently sensitive to detect the secondary activation of endogenous signaling pathways as it occurs through crosstalk following ectopic activation of a primary pathway.


Subject(s)
Computational Biology/methods , Gene Expression Profiling , Mammary Neoplasms, Animal/metabolism , Regression Analysis , Signal Transduction , Transforming Growth Factor beta/metabolism , ras Proteins/metabolism , Animals , Cell Line, Tumor , Female , Mammary Glands, Animal/metabolism , Mice , Mice, Transgenic
14.
Mol Cell Biol ; 27(14): 5135-46, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17485441

ABSTRACT

c-myc is frequently amplified in breast cancer; however, the mechanism of myc-induced mammary epithelial cell transformation has not been defined. We show that c-Myc induces a profound morphological transformation in human mammary epithelial cells and anchorage-independent growth. c-Myc suppresses the Wnt inhibitors DKK1 and SFRP1, and derepression of DKK1 or SFRP1 reduces Myc-dependent transforming activity. Myc-dependent repression of DKK1 and SFRP1 is accompanied by Wnt target gene activation and endogenous T-cell factor activity. Myc-induced mouse mammary tumors have repressed SFRP1 and increased expression of Wnt target genes. DKK1 and SFRP1 inhibit the transformed phenotype of breast cancer cell lines, and DKK1 inhibits tumor formation. We propose a positive feedback loop for activation of the c-myc and Wnt pathways in breast cancer.


Subject(s)
Cell Transformation, Neoplastic , Epithelial Cells/cytology , Intercellular Signaling Peptides and Proteins/metabolism , Mammary Glands, Human/cytology , Membrane Proteins/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Wnt Proteins/antagonists & inhibitors , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Genes, Neoplasm , Humans , Mice , Phenotype , Repressor Proteins/metabolism , Signal Transduction , TCF Transcription Factors/metabolism
15.
Cancer Res ; 66(12): 6421-31, 2006 Jun 15.
Article in English | MEDLINE | ID: mdl-16778221

ABSTRACT

Women who have their first child early in life have a substantially lower lifetime risk of breast cancer. The mechanism for this is unknown. Similar to humans, rats exhibit parity-induced protection against mammary tumorigenesis. To explore the basis for this phenomenon, we identified persistent pregnancy-induced changes in mammary gene expression that are tightly associated with protection against tumorigenesis in multiple inbred rat strains. Four inbred rat strains that exhibit marked differences in their intrinsic susceptibilities to carcinogen-induced mammary tumorigenesis were each shown to display significant protection against methylnitrosourea-induced mammary tumorigenesis following treatment with pregnancy levels of estradiol and progesterone. Microarray expression profiling of parous and nulliparous mammary tissue from these four strains yielded a common 70-gene signature. Examination of the genes constituting this signature implicated alterations in transforming growth factor-beta signaling, the extracellular matrix, amphiregulin expression, and the growth hormone/insulin-like growth factor I axis in pregnancy-induced alterations in breast cancer risk. Notably, related molecular changes have been associated with decreased mammographic density, which itself is strongly associated with decreased breast cancer risk. Our findings show that hormone-induced protection against mammary tumorigenesis is widely conserved among divergent rat strains and define a gene expression signature that is tightly correlated with reduced mammary tumor susceptibility as a consequence of a normal developmental event. Given the conservation of this signature, these pathways may contribute to pregnancy-induced protection against breast cancer.


Subject(s)
Hormones/genetics , Mammary Neoplasms, Experimental/genetics , Pregnancy, Animal/genetics , Amphiregulin , Animals , EGF Family of Proteins , Female , Gene Expression , Gene Expression Profiling , Glycoproteins/biosynthesis , Glycoproteins/genetics , Growth Hormone/biosynthesis , Growth Hormone/genetics , Hormones/biosynthesis , Intercellular Signaling Peptides and Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/genetics , Mammary Glands, Animal , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/prevention & control , Mice , Oligonucleotide Array Sequence Analysis , Parity , Pregnancy , Pregnancy, Animal/metabolism , Rats , Rats, Inbred F344 , Rats, Inbred Lew , Rats, Inbred WF , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta3 , Up-Regulation
16.
Development ; 132(5): 1147-60, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15689376

ABSTRACT

Epidemiological findings suggest that the consequences of a given oncogenic stimulus vary depending upon the developmental state of the target tissue at the time of exposure. This is particularly evident in the mammary gland, where both age at exposure to a carcinogenic stimulus and the timing of a first full-term pregnancy can markedly alter the risk of developing breast cancer. Analogous to this, the biological consequences of activating oncogenes, such as MYC, can be influenced by cellular context both in terms of cell lineage and cellular environment. In light of this, we hypothesized that the consequences of aberrant MYC activation in the mammary gland might be determined by the developmental state of the gland at the time of MYC exposure. To test this hypothesis directly, we have used a doxycycline-inducible transgenic mouse model to overexpress MYC during different stages of mammary gland development. Using this model, we find that the ability of MYC to inhibit postpartum lactation is due entirely to its activation within a specific 72-hour window during mid-pregnancy; by contrast, MYC activation either prior to or following this 72-hour window has little or no effect on postpartum lactation. Surprisingly, we find that MYC does not block postpartum lactation by inhibiting mammary epithelial differentiation, but rather by promoting differentiation and precocious lactation during pregnancy, which in turn leads to premature involution of the gland. We further show that this developmental stage-specific ability of MYC to promote mammary epithelial differentiation is tightly linked to its ability to downregulate caveolin 1 and activate Stat5 in a developmental stage-specific manner. Our findings provide unique in vivo molecular evidence for developmental stage-specific effects of oncogene activation, as well as the first evidence linking MYC with activation of the Jak2-Stat5 signaling pathway.


Subject(s)
Epithelium/metabolism , Gene Expression Regulation, Developmental , Mammary Glands, Animal/metabolism , Proto-Oncogene Proteins c-myc/biosynthesis , Proto-Oncogene Proteins c-myc/physiology , Animals , Apoptosis , Blotting, Northern , Blotting, Western , Caveolin 1 , Caveolins/biosynthesis , Cell Differentiation , Cell Lineage , DNA Primers/chemistry , DNA-Binding Proteins/metabolism , Down-Regulation , Immunohistochemistry , In Situ Nick-End Labeling , Lactation , Mice , Mice, Transgenic , Milk Proteins/metabolism , STAT3 Transcription Factor , STAT5 Transcription Factor , Signal Transduction , Time Factors , Trans-Activators/metabolism , Up-Regulation
17.
Mol Endocrinol ; 16(9): 2034-51, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12198241

ABSTRACT

Epidemiological studies have repeatedly demonstrated that women who undergo an early first full-term pregnancy have a significantly reduced lifetime risk of breast cancer. Similarly, rodents that have previously undergone a full-term pregnancy are highly resistant to carcinogen-induced breast cancer compared with age-matched nulliparous controls. Little progress has been made, however, toward understanding the biological basis of this phenomenon. We have used DNA microarrays to identify a panel of 38 differentially expressed genes that reproducibly distinguishes, in a blinded manner, between the nulliparous and parous states of the mammary gland in multiple strains of mice and rats. We find that parity results in the persistent down-regulation of multiple genes encoding growth factors, such as amphiregulin, pleiotrophin, and IGF-1, as well as the persistent up-regulation of the growth-inhibitory molecule, TGF-beta3, and several of its transcriptional targets. Our studies further indicate that parity results in a persistent increase in the differentiated state of the mammary gland as well as lifelong changes in the hematopoietic cell types resident within the gland. These findings define a developmental state of the mammary gland that is refractory to carcinogenesis and suggest novel hypotheses for the mechanisms by which parity may modulate breast cancer risk.


Subject(s)
Cell Differentiation , Gene Expression Regulation, Developmental , Growth Substances/genetics , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/metabolism , Parity/genetics , Transforming Growth Factor beta/genetics , Animals , Female , Gene Expression Profiling , Mammary Glands, Animal/cytology , Mice , Morphogenesis , Oligonucleotide Array Sequence Analysis , Pregnancy , Rats , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta3
18.
Mol Endocrinol ; 16(6): 1185-203, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12040007

ABSTRACT

The use of DNA microarrays to study vertebrate organogenesis presents unique analytical challenges compared with expression profiling of homogeneous cell populations. We have used a general approach that permits the automated, unbiased identification of biologically relevant patterns of gene expression to study murine mammary gland development. Our studies confirm the utility of this approach by demonstrating the ready identification of cellular processes and pathways of known functional importance in mammary development. Additionally, this approach permitted the identification of genetic pathways with unpredicted patterns of developmental regulation, including those involved in angiogenesis, extracellular matrix synthesis, and the beta-oxidation of fatty acids. Surprisingly, our findings demonstrate that the coordinate regulation of genes involved in the beta-oxidation of fatty acids reflects the presence of an abundant, yet previously unrecognized stromal compartment within the mammary gland that is composed of brown adipose tissue. Our data demonstrate that the amount of brown adipose tissue present in the mammary gland is developmentally regulated; that PPARalpha, Ucp1, and genes involved in fatty acid oxidation are spatially and temporally coregulated during development; that the mammary gland plays a functional role in adaptive thermogenesis; and that the transcriptional control of this adaptive response to cold is itself developmentally regulated.


Subject(s)
Breast/growth & development , Oligonucleotide Array Sequence Analysis/methods , Organogenesis , Thermogenesis , Adipose Tissue, Brown/metabolism , Animals , Breast/metabolism , Carrier Proteins/metabolism , Fatty Acids/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation , Ion Channels , Membrane Proteins/metabolism , Mice , Mitochondrial Proteins , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , Uncoupling Protein 1
SELECTION OF CITATIONS
SEARCH DETAIL
...