Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
JCI Insight ; 2(21)2017 11 02.
Article in English | MEDLINE | ID: mdl-29093275

ABSTRACT

The complex signaling networks of the tumor microenvironment that facilitate tumor growth and progression toward metastatic disease are becoming a focus of potential therapeutic options. The chemokine IL-8 is overexpressed in multiple cancer types, including triple-negative breast cancer (TNBC), where it promotes the acquisition of mesenchymal features, stemness, resistance to therapies, and the recruitment of immune-suppressive cells to the tumor site. The present study explores the utility of a clinical-stage monoclonal antibody that neutralizes IL-8 (HuMax-IL8) as a potential therapeutic option for TNBC. HuMax-IL8 was shown to revert mesenchymalization in claudin-low TNBC models both in vitro and in vivo as well as to significantly decrease the recruitment of polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) at the tumor site, an effect substantiated when used in combination with docetaxel. In addition, HuMax-IL8 enhanced the susceptibility of claudin-low breast cancer cells to immune-mediated lysis with NK and antigen-specific T cells in vitro. These results demonstrate the multifaceted way in which neutralizing this single chemokine reverts mesenchymalization, decreases recruitment of MDSCs at the tumor site, assists in immune-mediated killing, and forms the rationale for using HuMax-IL8 in combination with chemotherapy or immune-based therapies for the treatment of TNBC.


Subject(s)
Claudins/metabolism , Interleukin-8/metabolism , Myeloid-Derived Suppressor Cells/metabolism , Triple Negative Breast Neoplasms/metabolism , Animals , Antibodies, Monoclonal , Antibodies, Neutralizing/therapeutic use , Cell Line, Tumor , Cell Survival , Chemokines , Docetaxel/pharmacology , Drug Therapy , Female , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Interleukin-8/genetics , Mice , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/pathology , Signal Transduction , T-Lymphocytes , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/pathology , Tumor Microenvironment
2.
Oncoimmunology ; 6(10): e1349589, 2017.
Article in English | MEDLINE | ID: mdl-29123964

ABSTRACT

Mesenchymalization is a cellular and molecular program in which epithelial cells progressively lose their well-differentiated phenotype and adopt mesenchymal characteristics. Tumor mesenchymalization occurs during the progression of cancer to metastatic disease, and is also associated with resistance to multiple therapeutics, including killing by cytotoxic immune cells. Furthermore, tumor cells can evade immune destruction by upregulating the checkpoint molecule PD-L1, and emerging research has found higher PD-L1 expression in mesenchymalized tumors. Here, the association between TGF-ß1-mediated mesenchymalization and PD-L1 was investigated in non-small cell lung cancer cells (NSCLC). TGF-ß1 was found to upregulate PD-L1 gene transcription in a Smad2-dependent manner, and a positive association between PD-L1 and phosphorylated Smad2 was found in NSCLC tumors. The potential to target these 2 negative immune regulators with a single agent was investigated using M7824, a novel clinical-stage bifunctional agent that targets both PD-L1 and TGF-ß. Treatment of NSCLC cells with M7824 in vitro and in vivo attenuated features of TGF-ß1-mediated mesenchymalization, including mesenchymal marker expression, proliferation suppression, and chemoresistance. These findings demonstrate that upregulation of tumor cell PD-L1 is a novel mechanism of TGF-ß1-induced immunosuppression in NSCLC, and that treatment with M7824 has the potential to simultaneously block both tumor mesenchymalization and PD-L1-dependent immunosuppression.

3.
Semin Cancer Biol ; 47: 177-184, 2017 12.
Article in English | MEDLINE | ID: mdl-28823497

ABSTRACT

Tumor growth and progression are the products of complex signaling networks between different cell types within the tumor and its surrounding stroma. In particular, established tumors are known to stimulate an inflammatory reaction via the secretion of cytokines, chemokines, and growth factors that favor the recruitment of a range of infiltrating immune cell populations into the tumor microenvironment. While potentially able to exert tumor control, this inflammatory reaction is typically seized upon by the tumor to promote its own growth and progression towards metastasis. This review focuses on recent advances in understanding how an established tumor can initiate an inflammatory response via the release of pro-inflammatory mediators, such as IL-6 and IL-8, and their roles in cancer metastasis. In particular, the role of the epithelial-mesenchymal transition (EMT), a phenotypic switch observed in carcinomas that promotes progression towards metastasis, is discussed here in relation to cancer inflammation.


Subject(s)
Epithelial-Mesenchymal Transition , Inflammation/pathology , Neoplasms/pathology , Tumor Microenvironment , Animals , Cytokines/metabolism , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Epithelial-Mesenchymal Transition/immunology , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Inflammation Mediators , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Signal Transduction/drug effects , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
4.
Cells Tissues Organs ; 203(2): 128-138, 2017.
Article in English | MEDLINE | ID: mdl-28214895

ABSTRACT

Epithelial-mesenchymal transition (EMT) is recognized as a relevant process during the progression of carcinomas towards metastatic disease. Epithelial cancer cells undergoing an EMT program may acquire mesenchymal features, motility, invasiveness, and resistance to a variety of anticancer therapeutics. Preventing or reverting the EMT process in carcinomas has the potential to minimize tumor dissemination and the emergence of therapeutic resistance. One of the strategies currently under investigation to target tumor cells undergoing EMT is the generation of a sustained immune response directed against an essential molecular driver of the process. This review focuses on the current development of immune-mediated anticancer interventions aimed at targeting a transcription factor, brachyury, associated with human tumor EMT. Also presented here is a summary of recent studies demonstrating a role for EMT in tumor resistance to immune effector cytotoxicity, and the study of novel strategies aimed at reverting the EMT to be used in combination with immune-mediated anticancer interventions.


Subject(s)
Cancer Vaccines/immunology , Epithelial-Mesenchymal Transition , Fetal Proteins/immunology , Neoplasms/pathology , T-Box Domain Proteins/immunology , Animals , Drug Resistance, Neoplasm , Humans , Immunotherapy
5.
Pharmacol Ther ; 170: 212-225, 2017 02.
Article in English | MEDLINE | ID: mdl-27916651

ABSTRACT

Controlling the spread of carcinoma cells to distant organs is the foremost challenge in cancer treatment, as metastatic disease is generally resistant to therapy and is ultimately incurable for the majority of patients. The plasticity of tumor cell phenotype, in which the behaviors and functions of individual tumor cells differ markedly depending upon intrinsic and extrinsic factors, is now known to be a central mechanism in cancer progression. Our expanding knowledge of epithelial and mesenchymal phenotypic states in tumor cells, and the dynamic nature of the transitions between these phenotypes has created new opportunities to intervene to better control the behavior of tumor cells. There are now a variety of innovative pharmacological approaches to preferentially target tumor cells that have acquired mesenchymal features, including cytotoxic agents that directly kill these cells, and inhibitors that block or revert the process of mesenchymalization. Furthermore, novel immunological strategies have been developed to engage the immune system in seeking out and destroying mesenchymalized tumor cells. This review highlights the relevance of phenotypic plasticity in tumor biology, and discusses recently developed pharmacological and immunological means of targeting this phenomenon.


Subject(s)
Antineoplastic Agents/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Neoplasms/drug therapy , Animals , Disease Progression , Epithelial-Mesenchymal Transition/immunology , Humans , Neoplasm Metastasis , Neoplasms/immunology , Neoplasms/pathology , Phenotype
6.
Cell Death Dis ; 7(9): e2380, 2016 09 29.
Article in English | MEDLINE | ID: mdl-27685624

ABSTRACT

The epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI) erlotinib has been approved for years as a first-line therapy for patients harboring EGFR-sensitizing mutations. With the promising implementation of immunotherapeutic strategies for the treatment of lung cancer, there is a growing interest in developing combinatorial therapies that could utilize immune approaches in the context of conventional or targeted therapies. Tumor cells are known to evade immune attack by multiple strategies, including undergoing phenotypic plasticity via a process designated as the epithelial-mesenchymal transition (EMT). As signaling through EGFR is a major inducer of EMT in epithelial cells, we have investigated the effect of EGFR inhibition with erlotinib on tumor phenotype and susceptibility to immune attack. Our data shows that short-term exposure of tumor cells to low-dose erlotinib modulates tumor plasticity and immune-mediated cytotoxicity in lung cancer cells harboring a sensitizing EGFR mutation, leading to a remarkable enhancement of tumor lysis mediated by innate NK cells and antigen-specific T cells. This effect positively correlated with the ability of short-term EGFR blockade to modulate tumor phenotype towards a more epithelial one, as well as to increase susceptibility to caspase-mediated apoptosis. The effect, however, was lost when erlotinib was utilized for long periods of time in vitro or in vivo, which resulted in gain of mesenchymal features and decreased (rather than increased) tumor lysis in response to immune effector mechanisms. Our data provides rationale for potential combinations of erlotinib and immunotherapies for the treatment of lung carcinomas in the early setting, before the establishment of tumor relapse with long-term EGFR inhibition.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cytotoxicity, Immunologic , ErbB Receptors/antagonists & inhibitors , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Mutation/genetics , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/pathology , Caspases/metabolism , Cell Line, Tumor , Cytotoxicity, Immunologic/drug effects , Drug Resistance, Neoplasm/drug effects , Epithelial-Mesenchymal Transition/drug effects , ErbB Receptors/metabolism , Erlotinib Hydrochloride/pharmacology , Erlotinib Hydrochloride/therapeutic use , Humans , Interleukin-8/metabolism , Lung Neoplasms/genetics , Phenotype , Receptors, Death Domain/metabolism , Signal Transduction/drug effects , Time Factors
7.
Oncotarget ; 7(27): 42031-42044, 2016 Jul 05.
Article in English | MEDLINE | ID: mdl-27248176

ABSTRACT

A signaling pathway that is frequently deregulated in human carcinomas and has been explored as a therapeutic target involves the activation of the epidermal growth factor receptor (EGFR). Inhibition of EGFR via the small molecule inhibitors erlotinib and gefitinib commonly results in tumor resistance, even in patients with EGFR-mutant tumors that initially show substantial clinical responses. This study was designed to broaden our understanding of the molecular mechanisms of acquired resistance to erlotinib in lung cancer cells bearing wild type or mutated EGFR. We report here that generation of erlotinib-resistant lung cancer cells in vitro resulted in a phenotypic alteration reminiscent of an epithelial-mesenchymal transition (EMT) concomitant with a robust upregulation of the IL-8/IL-8R axis. Our results also demonstrate that upregulation of p38 MAPK signaling is responsible for the enhanced IL-8 secretion in the erlotinib-resistant tumor cells. Blockade of IL-8 signaling effectively reduced mesenchymal features of the resistant cells and also markedly enhanced their susceptibility to erlotinib. These results provide a rationale for the development of new therapeutic approaches involving blockade of IL-8 signaling for the management of acquired resistance to EGFR inhibition in patients with lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Drug Resistance, Neoplasm , Erlotinib Hydrochloride/therapeutic use , Interleukin-8/metabolism , Lung Neoplasms/metabolism , A549 Cells , Animals , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Epithelial-Mesenchymal Transition , ErbB Receptors/metabolism , Female , Gefitinib , Humans , Lung Neoplasms/drug therapy , Mice , Mice, Nude , Mutation , Neoplasm Transplantation , Phenotype , Quinazolines/therapeutic use , Signal Transduction , Up-Regulation , p38 Mitogen-Activated Protein Kinases/metabolism
8.
Vaccines (Basel) ; 4(3)2016 Jun 24.
Article in English | MEDLINE | ID: mdl-27348007

ABSTRACT

Interleukin-8 (IL-8, CXCL8) is a pro-inflammatory chemokine produced by various cell types to recruit leukocytes to sites of infection or tissue injury. Acquisition of IL-8 and/or its receptors CXCR1 and CXCR2 are known to be a relatively common occurrence during tumor progression. Emerging research now indicates that paracrine signaling by tumor-derived IL-8 promotes the trafficking of neutrophils and myeloid-derived suppressor cells (MDSCs) into the tumor microenvironment, which have the ability to dampen anti-tumor immune responses. Furthermore, recent studies have also shown that IL-8 produced by the tumor mass can induce tumor cells to undergo the transdifferentiation process epithelial-to-mesenchymal transition (EMT) in which tumor cells shed their epithelial characteristics and acquire mesenchymal characteristics. EMT can increase metastatic dissemination, stemness, and intrinsic resistance, including to killing by cytotoxic immune cells. This review highlights the dual potential roles that the inflammatory cytokine IL-8 plays in promoting tumor resistance by enhancing the immunosuppressive microenvironment and activating EMT, and then discusses the potential for targeting the IL-8/IL-8 receptor axis to combat these various resistance mechanisms.

9.
Neuro Oncol ; 15(12): 1652-63, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24158111

ABSTRACT

BACKGROUND: Despite advances in the treatment of the most aggressive form of brain tumor, glioblastoma, patient prognosis remains disappointing. This failure in treatment has been attributed to dysregulated oncogenic pathways, as observed in other tumors. We and others have suggested the use of microRNAs (miRs) as therapeutic tools able to target multiple pathways in glioblastoma. METHODS: This work features PCR quantification of miRs and transient transfection of many glioblastoma cell lines with miRs, followed by cell number analysis, trypan blue staining, alamarBlue assay of cell viability, caspase-3/-7 activity assay, immunoblot of cleaved poly(ADP-ribose) polymerase and fluorescence activated cell sorting and imaging of apoptotic nuclei, cell invasion assays, MRIs of glioblastoma xenografts in mice using transiently transfected cells as well as posttumor treatment with lentiviral vector encoding miR-297, and analysis of miR-297 target diacylglycerol kinase (DGK)-α including immunoblot, 3'UTR luciferase activity, and rescue with DGK-α overexpression. Cell counts and DGK-α immunoblot were also analyzed in the context of hypoxia and with overexpression of heterogeneous ribonucleoprotein L (hnRNPL). RESULTS: We identified miR-297 as a highly cytotoxic microRNA in glioblastoma, with minimal cytotoxicity to normal astrocytes. miR-297 overexpression reduced in vitro invasiveness and in vivo tumor formation. DGK-α is shown to be a miR-297 target with a critical role in miR-297 toxicity. In addition, hypoxia and its mediator hnRNPL upregulated DGK-α and buffered the cytotoxic effects of miR-297. CONCLUSION: This work shows miR-297 as a novel and physiologic regulator of cancer cell survival, largely through targeting of DGK-α, and also indicates that hypoxia ameliorates miR-297 toxicity to cancer cells.


Subject(s)
Brain Neoplasms/mortality , Diacylglycerol Kinase/metabolism , Glioblastoma/mortality , Hypoxia/genetics , MicroRNAs/genetics , 3' Untranslated Regions/genetics , Animals , Apoptosis , Blotting, Western , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Movement , Cell Proliferation , Diacylglycerol Kinase/genetics , Flow Cytometry , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Male , Mice , Mice, Inbred BALB C , Mice, SCID , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
10.
Cancer Discov ; 3(7): 782-97, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23558954

ABSTRACT

Although diacylglycerol kinase α (DGKα) has been linked to several signaling pathways related to cancer cell biology, it has been neglected as a target for cancer therapy. The attenuation of DGKα activity via DGKα-targeting siRNA and small-molecule inhibitors R59022 and R59949 induced caspase-mediated apoptosis in glioblastoma cells and in other cancers, but lacked toxicity in noncancerous cells. We determined that mTOR and hypoxia-inducible factor-1α (HIF-1α) are key targets of DGKα inhibition, in addition to its regulation of other oncogenes. DGKα regulates mTOR transcription via a unique pathway involving cyclic AMP. Finally, we showed the efficacy of DGKα inhibition with short hairpin RNA or a small-molecule agent in glioblastoma and melanoma xenograft treatment models, with growth delay and decreased vascularity. This study establishes DGKα as a central signaling hub and a promising therapeutic target in the treatment of cancer.


Subject(s)
Brain Neoplasms/genetics , Diacylglycerol Kinase/genetics , Glioblastoma/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Apoptosis/drug effects , Brain Neoplasms/pathology , Cell Line, Tumor , Diacylglycerol Kinase/antagonists & inhibitors , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Molecular Targeted Therapy , Piperidines/administration & dosage , Pyrimidinones/administration & dosage , Quinazolinones/administration & dosage , RNA, Small Interfering , Thiazoles/administration & dosage
11.
Neuro Oncol ; 14(10): 1215-26, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22962413

ABSTRACT

The Notch pathway is dysregulated and a potential target in glioblastoma multiforme (GBM). Currently available Notch inhibitors block γ-secretase, which is necessary for Notch processing. However, Notch is first cleaved by α-secretase outside the plasma membrane, via a disintegrin and metalloproteinase-10 and -17. In this work, we used a potent α-secretase inhibitor (ASI) to test inhibition of glioblastoma growth and inhibition of Notch and of both novel and known Notch targets. Featured in this study are luciferase reporter assays and immunoblot, microarray analysis, chromatin immunoprecipitation (ChIP), quantitative real-time PCR, cell number assay, bromodeoxyuridine incorporation, plasmid rescue, orthotopic xenograft model, and local delivery of treatment with convection-enhanced delivery using nanoparticles, as well as survival, MRI, and ex vivo luciferase assay. A CBF1-luciferase reporter assay as well as an immunoblot of endogenous Notch revealed Notch inhibition by the ASI. Microarray analysis, quantitative real-time PCR, and ChIP of ASI and γ-secretase inhibitor (GSI) treatment of GBM cells identified known Notch pathway targets, as well as novel Notch targets, including YKL-40 and leukemia inhibitory factor. Finally, we found that local nanoparticle delivery of ASIs but not GSIs increased survival time significantly in a GBM stem cell xenograft treatment model, and ASI treatment resulted in decreased tumor size and Notch activity. This work indicates α-secretase as an alternative to γ-secretase for inhibition of Notch in GBM and possibly other cancers as well, and it identifies novel Notch targets with biologic relevance and potential as biomarkers.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Brain Neoplasms/pathology , Cell Proliferation , Glioblastoma/pathology , Neoplastic Stem Cells/metabolism , Receptors, Notch/metabolism , Amyloid Precursor Protein Secretases/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Cell Cycle , Chromatin Immunoprecipitation , Gene Expression Profiling , Glioblastoma/drug therapy , Glioblastoma/metabolism , Humans , In Vitro Techniques , Luciferases/metabolism , Magnetics , Mice , Mice, Inbred BALB C , Nanoparticles , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Oligonucleotide Array Sequence Analysis , Piperidines/pharmacology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, Notch/genetics , Reverse Transcriptase Polymerase Chain Reaction , Spiro Compounds/pharmacology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...