Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
2.
ACS Pharmacol Transl Sci ; 7(1): 137-149, 2024 Jan 12.
Article in English | MEDLINE | ID: mdl-38230292

ABSTRACT

Glioblastoma multiforme (GBM) is the most aggressive type of glioma and is often resistant to traditional therapies. Evidence suggests that glioma stem cells (GSCs) contribute to this resistance. Mithramycin (Mit-A) targets GSCs and exhibits antitumor activity in GBM by affecting transcriptional targets such as SRY-related HMG-box transcription factor 2 (SOX2), oligodendrocyte lineage transcription factor 2 (OLIG2), and zinc finger E-box binding homeobox 1 (ZEB1). However, its clinical use has been limited by toxicity. This study explored the diagnostic potential of serum extracellular vesicles (EVs) to identify Mit-A responders. Serum EVs were isolated from 70 glioma patients, and targeted gene expression was analyzed using qRT-PCR. Using chemosensitivity assay, we identified 8 Mit-A responders and 17 nonresponders among 25 glioma patients. The M-score showed a significant correlation (p = 0.045) with isocitrate dehydrogenase 1 mutation but not other clinical variables. The genes SOX2 (p = 0.005), OLIG2 (p = 0.003), and ZEB1 (p = 0.0281) were found to be upregulated in the responder EVs. SOX2 had the highest diagnostic potential (AUC = 0.875), followed by OLIG2 (AUC = 0.772) and ZEB1 (AUC = 0.632).The combined gene panel showed significant diagnostic efficacy (AUC = 0.956) through logistic regression analysis. The gene panel was further validated in the serum EVs of 45 glioma patients. These findings highlight the potential of Mit-A as a targeted therapy for high-grade glioma based on differential gene expression in serum EVs. The gene panel could serve as a diagnostic tool to predict Mit-A sensitivity, offering a promising approach for personalized treatment strategies and emphasizing the role of GSCs in therapeutic resistance.

3.
J Cell Commun Signal ; 17(3): 591-608, 2023 Sep.
Article in English | MEDLINE | ID: mdl-36063341

ABSTRACT

Vasculogenic mimicry (VM), defined as an endothelial cell independent alternative mechanism of blood and nutrient supply by dysregulated tumor cells, is associated with poor prognosis in oral squamous cell carcinoma (OSCC). Here we aim to investigate the underlying molecular mechanism of the synergistic effect of phytochemical Lupeol and standard microtubule inhibitor Paclitaxel in reversing the hypoxia induced VM formation in OSCC. The results demonstrated that the hypoxia induced upregulation of HIF-1α led to augmentation of signaling cascade associated with extracellular matrix remodeling and EMT phenotypes that are mechanistically linked to VM. Induction of HIF-1α altered the expression of EMT/CSC markers (E-Cadherin, Vimentin, Snail, Twist and CD133) and enhanced the ability of cell migration/invasion and spheroid formation. Subsequently, the targeted knockdown of HIF-1α by siRNA led to the perturbation of matrigel mediated tube formation as well as of Laminin-5γ2 expression with the down-regulation of VE-Cadherin, total and phosphorylated (S-897) EphA2, pERK1/2 and MMP2. We also observed that Lupeol in association with Paclitaxel resulted to apoptosis and the disruption of VM associated phenotypes in vitro. We further validated the impact of this novel interventional approach in a patient derived tumor explant culture model of oral malignancy. The ex vivo tumor model mimicked the in vitro anti-VM potential of Lupeol-Paclitaxel combination through down-regulating HIF-1α/EphA2/Laminin-5γ2 cascade. Together, our findings elucidated mechanistic underpinning of hypoxia induced Laminin-5γ2 driven VM formation highlighting that Lupeol-Paclitaxel combination may serve as novel therapeutic intervention in perturbation of VM in human OSCC.

4.
Nat Nanotechnol ; 17(1): 98-106, 2022 01.
Article in English | MEDLINE | ID: mdl-34795441

ABSTRACT

Cancer progresses by evading the immune system. Elucidating diverse immune evasion strategies is a critical step in the search for next-generation immunotherapies for cancer. Here we report that cancer cells can hijack the mitochondria from immune cells via physical nanotubes. Mitochondria are essential for metabolism and activation of immune cells. By using field-emission scanning electron microscopy, fluorophore-tagged mitochondrial transfer tracing and metabolic quantification, we demonstrate that the nanotube-mediated transfer of mitochondria from immune cells to cancer cells metabolically empowers the cancer cells and depletes the immune cells. Inhibiting the nanotube assembly machinery significantly reduced mitochondrial transfer and prevented the depletion of immune cells. Combining a farnesyltransferase and geranylgeranyltransferase 1 inhibitor, namely, L-778123, which partially inhibited nanotube formation and mitochondrial transfer, with a programmed cell death protein 1 immune checkpoint inhibitor improved the antitumour outcomes in an aggressive immunocompetent breast cancer model. Nanotube-mediated mitochondrial hijacking can emerge as a novel target for developing next-generation immunotherapy agents for cancer.


Subject(s)
Leukocytes/pathology , Mitochondria/metabolism , Nanotubes/chemistry , Neoplasms/pathology , Animals , Base Sequence , Cell Line, Tumor , Humans , Immunity , Mice, Inbred C57BL , Mitochondrial Proteins/metabolism , Nanotubes/ultrastructure
5.
Cancer Biol Ther ; 22(1): 66-78, 2021 01 02.
Article in English | MEDLINE | ID: mdl-33356802

ABSTRACT

The prognosis of AML is generally poor, with 5-year survival rate of 25%. There has been substantial progress in identification of new therapeutic targets, along with approval of at least three targeted therapies for AML in recent years. Nevertheless, treatment has largely remained unchanged over couple of decades, with ~40% patients not achieving remission. AML is a highly heterogenous disease and there is a need for a preclinical platform to understand the heterogeneity and tumor microenvironment that can guide therapy selection. In this study, we employed an ex vivo tumor explant model to study tumor microenvironment and to select a treatment course for AML patients. Our data reveal dysregulation of DNA methyltransferase (DNMT) and histone deacetylase (HDAC) in a subset of AML patients. Based on this observation, epigenetic modulators azacitidine and panobinostat alone and in combination, were evaluated as treatment regimens in cytarabine refractory tumors. More than 50% of the treated samples showed response to the combination therapy. In order to explore alternate treatment modalities for tumors refractory to these epigenetic modulators, TCGA data analysis was done which revealed increased expression and hypomethylation of IFNGR1/2, suggesting activation of JAK/STAT pathway in AML. This was further interrogated ex vivo, with p-STAT3 expression in patients' samples. Fedratinib, a JAK/STAT inhibitor was evaluated and 78% tumor efficacy response was achieved. Taken together, our data indicate that ex vivo platform derived from patient samples is capable in guiding optimal therapy selection for various classes of drugs including identification of novel targeted therapies.


Subject(s)
Cytarabine/therapeutic use , Epigenomics/methods , Immunosuppressive Agents/therapeutic use , Janus Kinase Inhibitors/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Cell Line, Tumor , Cytarabine/pharmacology , Humans , Immunosuppressive Agents/pharmacology , Janus Kinase Inhibitors/pharmacology , Leukemia, Myeloid, Acute/mortality , Prognosis , Survival Analysis
6.
iScience ; 23(6): 101229, 2020 Jun 26.
Article in English | MEDLINE | ID: mdl-32554190

ABSTRACT

Ex vivo human tumor models have emerged as promising, yet complex tools to study cancer immunotherapy response dynamics. Here, we present a strategy that integrates empirical data from an ex vivo human system with computational models to interpret the response dynamics of a clinically prescribed PD-1 inhibitor, nivolumab, in head and neck squamous cell carcinoma (HNSCC) biopsies (N = 50). Using biological assays, we show that drug-induced variance stratifies samples by T helper type 1 (Th1)-related pathways. We then built a systems biology network and mathematical framework of local and global sensitivity analyses to simulate and estimate antitumor phenotypes, which implicate a dynamic role for the induction of Th1-related cytokines and T cell proliferation patterns. Together, we describe a multi-disciplinary strategy to analyze and interpret the response dynamics of PD-1 blockade using heterogeneous ex vivo data and in silico simulations, which could provide researchers a powerful toolset to interrogate immune checkpoint inhibitors.

7.
Gastric Cancer ; 23(5): 796-810, 2020 09.
Article in English | MEDLINE | ID: mdl-32333232

ABSTRACT

BACKGROUND: Phosphorylation is an important regulatory mechanism of protein activity in cells. Studies in various cancers have reported perturbations in kinases resulting in aberrant phosphorylation of oncoproteins and tumor suppressor proteins. METHODS: In this study, we carried out quantitative phosphoproteomic analysis of gastric cancer tissues and corresponding xenograft samples. Using these data, we employed bioinformatics analysis to identify aberrant signaling pathways. We further performed molecular inhibition and silencing of the upstream regulatory kinase in gastric cancer cell lines and validated its effect on cellular phenotype. Through an ex vivo technology utilizing patient tumor and blood sample, we sought to understand the therapeutic potential of the kinase by recreating the tumor microenvironment. RESULTS: Using mass spectrometry-based high-throughput analysis, we identified 1,344 phosphosites and 848 phosphoproteins, including differential phosphorylation of 177 proteins (fold change cut-off ≥ 1.5). Our data showed that a subset of differentially phosphorylated proteins belonged to splicing machinery. Pathway analysis highlighted Cdc2-like kinase (CLK1) as upstream kinase. Inhibition of CLK1 using TG003 and CLK1 siRNA resulted in a decreased cell viability, proliferation, invasion and migration as well as modulation in the phosphorylation of SRSF2. Ex vivo experiments which utilizes patient's own tumor and blood to recreate the tumor microenvironment validated the use of CLK1 as a potential target for gastric cancer treatment. CONCLUSIONS: Our data indicates that CLK1 plays a crucial role in the regulation of splicing process in gastric cancer and that CLK1 can act as a novel therapeutic target in gastric cancer.


Subject(s)
Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Proteome/metabolism , Stomach Neoplasms/pathology , Animals , Apoptosis , Biomarkers, Tumor , Cell Movement , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, SCID , Neoplasm Invasiveness , Phosphorylation , Prognosis , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/genetics , Proteome/analysis , RNA, Small Interfering/genetics , Stomach Neoplasms/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Sci Signal ; 12(595)2019 08 20.
Article in English | MEDLINE | ID: mdl-31431543

ABSTRACT

Metastable phenotypic state transitions in cancer cells can lead to the development of transient adaptive resistance or tolerance to chemotherapy. Here, we report that the acquisition of a phenotype marked by increased abundance of CD44 (CD44Hi) by breast cancer cells as a tolerance response to routinely used cytotoxic drugs, such as taxanes, activated a metabolic switch that conferred tolerance against unrelated standard-of-care chemotherapeutic agents, such as anthracyclines. We characterized the sequence of molecular events that connected the induced CD44Hi phenotype to increased activity of both the glycolytic and oxidative pathways and glucose flux through the pentose phosphate pathway (PPP). When given in a specific order, a combination of taxanes, anthracyclines, and inhibitors of glucose-6-phosphate dehydrogenase (G6PD), an enzyme involved in glucose metabolism, improved survival in mouse models of breast cancer. The same sequence of the three-drug combination reduced the viability of patient breast tumor samples in an explant system. Our findings highlight a convergence between phenotypic and metabolic state transitions that confers a survival advantage to cancer cells against clinically used drug combinations. Pharmacologically targeting this convergence could overcome cross-drug tolerance and could emerge as a new paradigm in the treatment of cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Delivery Systems , Enzyme Inhibitors/pharmacology , Glucose/metabolism , Glucosephosphate Dehydrogenase/antagonists & inhibitors , Neoplasm Proteins/antagonists & inhibitors , Neoplasms , Pentose Phosphate Pathway/drug effects , Glucosephosphate Dehydrogenase/metabolism , Humans , MCF-7 Cells , Neoplasm Metastasis , Neoplasm Proteins/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology
9.
Cell Oncol (Dordr) ; 40(2): 145-155, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28039610

ABSTRACT

PURPOSE: The tumor suppressor protein p53 is known to control cell cycle arrest and apoptosis. Lupeol is a phytochemical that has been found to induce apoptosis in different cancer types through the extrinsic pathway. As yet, however, its role in the induction of cell cycle arrest and apoptosis through the intrinsic pathway in head and neck cancer has not been investigated. Here, we aimed at understanding the mechanism underlying the antitumor effect of Lupeol in head and neck cancer. METHODS: The antitumor effect of Lupeol on oral and laryngeal carcinomas was assessed using two in vitro 2D cell line models (HEp-2, UPCI:SCC-131) and, subsequently, an ex vivo 3D tumor explant culture platform that maintains key features of the native tumor microenvironment. The mechanism underlying Lupeol-mediated antitumor responses was delineated using MTT, colony formation, flow cytometry, immunofluorescence, Western blotting and immunohistochemistry assays. RESULTS: We found that Lupeol induced an enhanced expression of p53 in both cell line models tested and, subsequently, cell cycle arrest at the G1 phase. In addition we found that, following Lupeol treatment, p53 induced Bax expression and activated the intrinsic apoptotic pathway (as measured by Caspase-3 cleavage). Interestingly, Lupeol was also found to trigger G1 cell cycle arrest through up-regulation of the expression of CDKN2A, but not p21, resulting in inhibition of CyclinD1. In an ex vivo platform Lupeol was found to impart a potent antitumor response as defined by inhibition of Ki67 expression, decreased cell viability and concomitant activation (cleavage) of Caspase-3. Finally, we found that Lupeol can re-sensitize primary head and neck squamous cell carcinoma (HNSCC) tumor samples that had clinically progressed under a Cisplatin treatment regimen. CONCLUSION: Together, our data indicate that Lupeol may orchestrate a bifurcated regulation of neoplastic growth and apoptosis in head and neck cancers and may serve as a promising agent for the management of tumors that have progressed on a platinum-based treatment regimen.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Pentacyclic Triterpenes/pharmacology , Tumor Suppressor Protein p53/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , G1 Phase Cell Cycle Checkpoints/drug effects , Head and Neck Neoplasms/drug therapy , Humans , Signal Transduction/drug effects
10.
Nat Commun ; 6: 6139, 2015 Feb 11.
Article in English | MEDLINE | ID: mdl-25669750

ABSTRACT

Understanding the emerging models of adaptive resistance is key to overcoming cancer chemotherapy failure. Using human breast cancer explants, in vitro cell lines, mouse in vivo studies and mathematical modelling, here we show that exposure to a taxane induces phenotypic cell state transition towards a favoured transient CD44(Hi)CD24(Hi) chemotherapy-tolerant state. This state is associated with a clustering of CD44 and CD24 in membrane lipid rafts, leading to the activation of Src Family Kinase (SFK)/hemopoietic cell kinase (Hck) and suppression of apoptosis. The use of pharmacological inhibitors of SFK/Hck in combination with taxanes in a temporally constrained manner, where the kinase inhibitor is administered post taxane treatment, but not when co-administered, markedly sensitizes the chemotolerant cells to the chemotherapy. This approach of harnessing chemotherapy-induced phenotypic cell state transition for improving antitumour outcome could emerge as a translational strategy for the management of cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , Neoplasms/pathology , Animals , Antigens, CD/metabolism , Antineoplastic Agents/therapeutic use , Bridged-Ring Compounds/pharmacology , Carcinogenesis/drug effects , Carcinogenesis/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Evaluation, Preclinical , Female , Humans , Membrane Microdomains/metabolism , Mice, Inbred BALB C , Models, Biological , Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Phenotype , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Taxoids/pharmacology , Time Factors
11.
Nat Commun ; 6: 6169, 2015 Feb 27.
Article in English | MEDLINE | ID: mdl-25721094

ABSTRACT

Predicting clinical response to anticancer drugs remains a major challenge in cancer treatment. Emerging reports indicate that the tumour microenvironment and heterogeneity can limit the predictive power of current biomarker-guided strategies for chemotherapy. Here we report the engineering of personalized tumour ecosystems that contextually conserve the tumour heterogeneity, and phenocopy the tumour microenvironment using tumour explants maintained in defined tumour grade-matched matrix support and autologous patient serum. The functional response of tumour ecosystems, engineered from 109 patients, to anticancer drugs, together with the corresponding clinical outcomes, is used to train a machine learning algorithm; the learned model is then applied to predict the clinical response in an independent validation group of 55 patients, where we achieve 100% sensitivity in predictions while keeping specificity in a desired high range. The tumour ecosystem and algorithm, together termed the CANScript technology, can emerge as a powerful platform for enabling personalized medicine.


Subject(s)
Algorithms , Antineoplastic Agents/pharmacology , Extracellular Matrix Proteins/metabolism , Precision Medicine/methods , Tissue Engineering/methods , Tumor Microenvironment/drug effects , Analysis of Variance , Chromatography, Liquid , DNA Mutational Analysis , Gene Expression Profiling , Humans , Machine Learning , Microscopy, Electron, Scanning , Predictive Value of Tests , Tandem Mass Spectrometry
12.
Cancer Res ; 73(3): 1118-27, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23361299

ABSTRACT

The PI3K/AKT/mTOR pathway is an important signaling axis that is perturbed in majority of cancers. Biomarkers such as pS6RP, GLUT1, and tumor FDG uptake are being evaluated in patient stratification for mTOR pathway inhibitors. In the absence of a clear understanding of the underlying mechanisms in tumor signaling, the biomarker strategy for patient stratification is of limited use. Here, we show that no discernible correlation exists between FDG uptake and the corresponding Ki67, GLUT1, pS6RP expression in tumor biopsies from patients with head and neck cancer. Correlation between GLUT1 and pS6RP levels in tumors was observed but elevated pS6RP was noticed even in the absence of concomitant AKT activation, suggesting that other downstream molecules of PI3K/AKT and/or other pathways upstream of mTOR are active in these tumors. Using an ex vivo platform, we identified putative responders to rapamycin, an mTOR inhibitor in these tumors. However, rapamycin did not induce antitumor effect in the majority of tumors with activated mTOR, potentially attributable to the observation that rapamycin induces feedback activation of AKT. Accordingly, treatment of these tumors with an AKT inhibitor and rapamycin uniformly resulted in abrogation of mTOR inhibition-induced AKT activation in all tumors but failed to induce antitumor response in a subset. Phosphoproteomic profiling of tumors resistant to dual AKT/mTOR inhibitors revealed differential activation of multiple pathways involved in proliferation and survival. Collectively, our results suggest that, in addition to biomarker-based segregation, functional assessment of a patient's tumor before treatment with mTOR/AKT inhibitors may be useful for patient stratification.


Subject(s)
Head and Neck Neoplasms/drug therapy , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , TOR Serine-Threonine Kinases/antagonists & inhibitors , Adult , Apoptosis/drug effects , Cell Proliferation/drug effects , Female , Glucose Transporter Type 1/analysis , Head and Neck Neoplasms/pathology , Humans , Male , Middle Aged , Proto-Oncogene Proteins c-akt/metabolism , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/physiology
13.
Transl Oncol ; 5(6): 422-9, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23323157

ABSTRACT

Activation of the phosphoinositide 3-kinase pathway is commonly observed in human prostate cancer. Loss of function of phosphatase and tensin homolog (PTEN) is associated with the activation of AKT and mammalian target of rapamycin (mTOR) in many cancer cell lines as well as in other model systems. However, activation of mTOR is also dependent of kinases other than AKT. Here, we show that activation of mTOR is not dependent on AKT in a prostate-specific PTEN-deficient mouse model of prostate cancer. Pathway bifurcation of AKT and mTOR was noted in both mouse and human prostate tumors. We demonstrated for the first time that cotargeting mTOR and AKT with ridaforolimus/MK-8669 and M1K-2206, respectively, delivers additive antitumor effects in vivo when compared to single agents. Our preclinical data suggest that the combination of AKT and mTOR inhibitors might be more effective in treating prostate cancer patients than current treatment regimens or either treatment alone.

14.
Cancer Cell ; 19(1): 58-71, 2011 Jan 18.
Article in English | MEDLINE | ID: mdl-21215704

ABSTRACT

Activation of the PI3K-AKT pathway in tumors is modulated by negative feedback, including mTORC1-mediated inhibition of upstream signaling. We now show that AKT inhibition induces the expression and phosphorylation of multiple receptor tyrosine kinases (RTKs). In a wide spectrum of tumor types, inhibition of AKT induces a conserved set of RTKs, including HER3, IGF-1R, and insulin receptor. This is in part due to mTORC1 inhibition and in part secondary to a FOXO-dependent activation of receptor expression. PI3K-AKT inhibitors relieve this feedback and activate RTK signaling; this may attenuate their antitumor activity. Consistent with this model, we find that, in tumors in which AKT suppresses HER3 expression, combined inhibition of AKT and HER kinase activity is more effective than either alone.


Subject(s)
Feedback, Physiological/physiology , Gene Expression Regulation, Neoplastic/physiology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction/physiology , Animals , Benzylamines/pharmacology , Benzylamines/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Drug Therapy, Combination , Feedback, Physiological/drug effects , Female , Forkhead Transcription Factors/antagonists & inhibitors , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gefitinib , Gene Expression/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lapatinib , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Nude , Models, Biological , Multiprotein Complexes , Neoplasms/metabolism , Phosphorylation/drug effects , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Protein Binding/genetics , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proteins/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Quinazolines/pharmacology , Quinazolines/therapeutic use , Quinoxalines/pharmacology , Quinoxalines/therapeutic use , RNA, Small Interfering/genetics , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/genetics , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/genetics , Receptor, ErbB-3/metabolism , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Receptor, Insulin/genetics , Receptor, Insulin/metabolism
15.
Mol Cancer Ther ; 9(7): 1956-67, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20571069

ABSTRACT

The serine/threonine kinase Akt lies at a critical signaling node downstream of phosphatidylinositol-3-kinase and is important in promoting cell survival and inhibiting apoptosis. An Akt inhibitor may be particularly useful for cancers in which increased Akt signaling is associated with reduced sensitivity to cytotoxic agents or receptor tyrosine kinase inhibitors. We evaluated the effect of a novel allosteric Akt inhibitor, MK-2206, in combination with several anticancer agents. In vitro, MK-2206 synergistically inhibited cell proliferation of human cancer cell lines in combination with molecular targeted agents such as erlotinib (an epidermal growth factor receptor inhibitor) or lapatinib (a dual epidermal growth factor receptor/human epidermal growth factor receptor 2 inhibitor). Complementary inhibition of erlotinib-insensitive Akt phosphorylation by MK-2206 was one mechanism of synergism, and a synergistic effect was found even in erlotinib-insensitive cell lines. MK-2206 also showed synergistic responses in combination with cytotoxic agents such as topoisomerase inhibitors (doxorubicin, camptothecin), antimetabolites (gemcitabine, 5-fluorouracil), anti-microtubule agents (docetaxel), and DNA cross-linkers (carboplatin) in lung NCI-H460 or ovarian A2780 tumor cells. The synergy with docetaxel depended on the treatment sequence; a schedule of MK-2206 dosed before docetaxel was not effective. MK-2206 suppressed the Akt phosphorylation that is induced by carboplatin and gemcitabine. In vivo, MK-2206 in combination with these agents exerted significantly more potent tumor inhibitory activities than each agent in the monotherapy setting. These findings suggest that Akt inhibition may augment the efficacy of existing cancer therapeutics; thus, MK-2206 is a promising agent to treat cancer patients who receive these cytotoxic and/or molecular targeted agents.


Subject(s)
Antineoplastic Agents/pharmacology , Heterocyclic Compounds, 3-Ring/pharmacology , Neoplasms/drug therapy , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Allosteric Regulation/drug effects , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Blotting, Western , Camptothecin/administration & dosage , Camptothecin/pharmacology , Carboplatin/administration & dosage , Carboplatin/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Caspases/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , Enzyme Activation/drug effects , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Erlotinib Hydrochloride , Heterocyclic Compounds, 3-Ring/administration & dosage , Heterocyclic Compounds, 3-Ring/chemistry , Humans , Lapatinib , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Molecular Structure , Neoplasms/metabolism , Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , Quinazolines/administration & dosage , Quinazolines/pharmacology , Xenograft Model Antitumor Assays
16.
Cancer Res ; 70(6): 2476-84, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20197467

ABSTRACT

ERBB2/neu and Notch signaling are known to be deregulated in many human cancers. However, pathway cross-talk and dependencies are not well understood. In this study, we use an ERBB2-transgenic mouse model of breast cancer (neuT) to show that Notch signaling plays a critical role in tumor maintenance. Inhibition of the Notch pathway with a gamma-secretase inhibitor (GSI) decreased both the Notch and the mammalian target of rapamycin/AKT pathways. Antitumor activity resulting from GSI treatment was associated with decreased cell proliferation as measured by Ki67 and decreased expression of glucose transporter Glut1. Positron emission tomography (PET) imaging showed that the functional consequences of decreased Glut1 translated to reduced glucose uptake and correlated with antitumor effects as measured by micro-computed tomography imaging. The decrease of Glut1 in neuT tumors was also observed in several human breast cancer cell lines following GSI treatment. We provide evidence that approximately 27% of ERBB2-positive human breast cancer specimens display high expression of HES1, phospho-S6RP, and GLUT1. Together, these results suggest that pathways downstream of Notch signaling are, at least in part, responsible for promoting tumor growth in neuT and also active in both neuT and a subset of human breast cancers. These findings suggest that GSI may provide therapeutic benefit to a subset of ERBB2-positive breast cancers and that [(18)F]FDG-PET imaging may be useful in monitoring clinical response.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Glucose/pharmacokinetics , Intracellular Signaling Peptides and Proteins/metabolism , Oncogene Protein v-akt/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptor, ErbB-2/metabolism , Receptors, Notch/metabolism , Animals , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Cyclic S-Oxides/pharmacology , Down-Regulation/drug effects , Female , Fluorodeoxyglucose F18/pharmacokinetics , Glucose Transporter Type 1/biosynthesis , Humans , Mammary Neoplasms, Experimental , Mice , Mice, Inbred BALB C , Mice, Transgenic , Oncogene Protein v-akt/antagonists & inhibitors , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases , Thiadiazoles/pharmacology
17.
Cancer Res ; 69(23): 8949-57, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19903844

ABSTRACT

Notch pathway signaling plays a fundamental role in normal biological processes and is frequently deregulated in many cancers. Although several hypotheses regarding cancer subpopulations most likely to respond to therapies targeting the Notch pathway have been proposed, clinical utility of these predictive markers has not been shown. To understand the molecular basis of gamma-secretase inhibitor (GSI) sensitivity in breast cancer, we undertook an unbiased, de novo responder identification study using a novel genetically engineered in vivo breast cancer model. We show that tumors arising from this model are heterogeneous on the levels of gene expression, histopathology, growth rate, expression of Notch pathway markers, and response to GSI treatment. In addition, GSI treatment of this model was associated with inhibition of Hes1 and proliferation markers, indicating that GSI treatment inhibits Notch signaling. We then identified a pretreatment gene expression signature comprising 768 genes that is significantly associated with in vivo GSI efficacy across 99 tumor lines. Pathway analysis showed that the GSI responder signature is enriched for Notch pathway components and inflammation/immune-related genes. These data show the power of this novel in vivo model system for the discovery of biomarkers predictive of response to targeted therapies, and provide a basis for the identification of human breast cancers most likely to be sensitive to GSI treatment.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Cyclic S-Oxides/administration & dosage , Enzyme Inhibitors/administration & dosage , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/enzymology , Thiadiazoles/administration & dosage , Animals , Cell Growth Processes/drug effects , Drug Administration Schedule , Gene Regulatory Networks , Humans , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic
18.
Cancer Res ; 69(18): 7466-72, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19738074

ABSTRACT

Androgen receptors have been shown to play a critical role in prostate cancer. We used ultrasound imaging techniques to track tumor response to antiandrogen and rapamycin treatment in a prostate-specific Pten-deleted mouse model of cancer. Depletion of androgens by either surgical or chemical castration significantly inhibited tumor growth progression without altering the activation of Akt and mammalian target of rapamycin (mTOR). We also showed for the first time that targeting mTOR along with antiandrogen treatment exhibited additive antitumor effects in vivo when compared with single agents. Our preclinical data suggest that combination of antiandrogens with mTOR inhibitors might be more effective in treating androgen-dependent prostate cancer patients.


Subject(s)
Androgen Antagonists/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carrier Proteins/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Prostatic Neoplasms/drug therapy , Sirolimus/pharmacology , Androgen Antagonists/administration & dosage , Animals , Cell Growth Processes/drug effects , Imaging, Three-Dimensional/methods , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Oncogene Protein v-akt/metabolism , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/genetics , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/pathology , Sirolimus/administration & dosage , TOR Serine-Threonine Kinases , Ultrasonography/methods
19.
Gastroenterology ; 136(5): 1741-9.e6, 2009 May.
Article in English | MEDLINE | ID: mdl-19208345

ABSTRACT

BACKGROUND & AIMS: The Notch signaling pathway is required for the expansion of undifferentiated pancreatic progenitor cells during embryonic development and has been implicated in the progression of pancreatic ductal adenocarcinoma (PDAC). The interaction of Notch ligands with their receptors promotes a gamma-secretase-dependent cleavage of the Notch receptor and release of the Notch intracellular domain, which translocates to the nucleus and activates transcription. We investigated the role of this pathway in PDAC progression. METHODS: We tested the effects of a gamma-secretase inhibitor (GSI) that blocks Notch signaling in PDAC cell lines and a genetically engineered mouse model of PDAC (Kras p53 L/+ mice). RESULTS: Notch signaling was activated in PDAC precursors and advanced tumors. The GSI inhibited the growth of premalignant pancreatic duct-derived cells in a Notch-dependent manner. Additionally, in a panel of over 400 human solid tumor-derived cell lines, PDAC cells, as a group, were more sensitive to the GSI than any other tumor type. Finally, the GSI completely inhibited tumor development in the genetically engineered model of invasive PDAC (P < .005, chi2 test; compared with mice exposed to vehicle). CONCLUSIONS: These results suggest that Notch signaling is required for PDAC progression. Pharmacologic targeting of this pathway offers therapeutic potential in this treatment-refractory malignancy.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Pancreatic Neoplasms/metabolism , Receptors, Notch/physiology , Signal Transduction , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Animals , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/physiopathology , Cell Line , Cell Line, Tumor , Cyclic S-Oxides/pharmacology , Disease Progression , Humans , Mice , Mice, Transgenic , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/physiopathology , Thiadiazoles/pharmacology
20.
Blood ; 113(24): 6172-81, 2009 Jun 11.
Article in English | MEDLINE | ID: mdl-19246562

ABSTRACT

Mutations in NOTCH1 are frequently detected in patients with T-cell acute lymphoblastic leukemia (T-ALL) and in mouse T-ALL models. Treatment of mouse or human T-ALL cell lines in vitro with gamma-secretase inhibitors (GSIs) results in growth arrest and/or apoptosis. These studies suggest GSIs as potential therapeutic agents in the treatment of T-ALL. To determine whether GSIs have antileukemic activity in vivo, we treated near-end-stage Tal1/Ink4a/Arf+/- leukemic mice with vehicle or with a GSI developed by Merck (MRK-003). We found that GSI treatment significantly extended the survival of leukemic mice compared with vehicle-treated mice. Notch1 target gene expression was repressed and increased numbers of apoptotic cells were observed in the GSI-treated mice, demonstrating that Notch1 inhibition in vivo induces apoptosis. T-ALL cell lines also exhibit PI3K/mTOR pathway activation, indicating that rapamycin may also have therapeutic benefit. When GSIs are administered in combination with rapamycin, mTOR kinase activity is ablated and apoptosis induced. Moreover, GSI and rapamycin treatment inhibits human T-ALL growth and extends survival in a mouse xenograft model. This work supports the idea of targeting NOTCH1 in T-ALL and suggests that inhibition of the mTOR and NOTCH1 pathways may have added efficacy.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Carrier Proteins/metabolism , Cyclic S-Oxides/pharmacology , Disease Models, Animal , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Receptor, Notch1/metabolism , Thiadiazoles/pharmacology , Amyloid Precursor Protein Secretases/metabolism , Animals , Apoptosis , Basic Helix-Loop-Helix Transcription Factors/physiology , Blotting, Western , Carrier Proteins/genetics , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p16/physiology , Flow Cytometry , Humans , Immunoenzyme Techniques , Mice , Mice, Transgenic , Phosphotransferases (Alcohol Group Acceptor)/genetics , Proto-Oncogene Proteins/physiology , Receptor, Notch1/genetics , Signal Transduction , T-Cell Acute Lymphocytic Leukemia Protein 1 , TOR Serine-Threonine Kinases , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...