Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
J Exp Med ; 220(5)2023 05 01.
Article in English | MEDLINE | ID: mdl-36828390

ABSTRACT

Metastatic cancer cells adapt to thrive in secondary organs. To investigate metastatic adaptation, we performed transcriptomic analysis of metastatic and non-metastatic murine breast cancer cells. We found that pleiotrophin (PTN), a neurotrophic cytokine, is a metastasis-associated factor that is expressed highly by aggressive breast cancers. Moreover, elevated PTN in plasma correlated significantly with metastasis and reduced survival of breast cancer patients. Mechanistically, we find that PTN activates NF-κB in cancer cells leading to altered cytokine production, subsequent neutrophil recruitment, and an immune suppressive microenvironment. Consequently, inhibition of PTN, pharmacologically or genetically, reduces the accumulation of tumor-associated neutrophils and reverts local immune suppression, resulting in increased T cell activation and attenuated metastasis. Furthermore, inhibition of PTN significantly enhanced the efficacy of immune checkpoint blockade and chemotherapy in reducing metastatic burden in mice. These findings establish PTN as a previously unrecognized driver of a prometastatic immune niche and thus represents a promising therapeutic target for the treatment of metastatic breast cancer.


Subject(s)
Carrier Proteins , Neoplasms , Mice , Animals , Cytokines/metabolism , NF-kappa B , Tumor Microenvironment
2.
J Thorac Oncol ; 17(10): 1178-1191, 2022 10.
Article in English | MEDLINE | ID: mdl-35798240

ABSTRACT

INTRODUCTION: Macrophage phenotype in the tumor microenvironment correlates with prognosis in NSCLC. Immunosuppressive macrophages promote tumor progression, whereas proinflammatory macrophages may drive an antitumor immune response. How individual NSCLCs affect macrophage phenotype is a major knowledge gap. METHODS: To systematically study the impact of lung cancer cells on macrophage phenotypes, we developed an in vitro co-culture model that consisted of molecularly and clinically annotated patient-derived NSCLC lines, human cancer-associated fibroblasts, and murine macrophages. Induced macrophage phenotype was studied through quantitative real-time polymerase chain reaction and validated in vivo using NSCLC xenografts through quantitative immunohistochemistry and clinically with The Cancer Genome Atlas (TCGA)-"matched" patient tumors. RESULTS: A total of 72 NSCLC cell lines were studied. The most frequent highly induced macrophage-related gene was Arginase-1, reflecting an immunosuppressive M2-like phenotype. This was independent of multiple clinicopathologic factors, which also did not affect M2:M1 ratios in matched TCGA samples. In vivo, xenograft tumors established from high Arginase-1-inducing lines (Arghi) had a significantly elevated density of Arg1+ macrophages. Matched TCGA clinical samples to Arghi NSCLC lines had a significantly higher ratio of M2:M1 macrophages (p = 0.0361). CONCLUSIONS: In our in vitro co-culture model, a large panel of patient-derived NSCLC lines most frequently induced high-expression Arginase-1 in co-cultured mouse macrophages, independent of major clinicopathologic and oncogenotype-related factors. Arghi cluster-matched TCGA tumors contained a higher ratio of M2:M1 macrophages. Thus, this in vitro model reproducibly characterizes how individual NSCLC modulates macrophage phenotype, correlates with macrophage polarization in clinical samples, and can serve as an accessible platform for further investigation of macrophage-specific therapeutic strategies.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Animals , Arginase/genetics , Arginase/metabolism , Arginase/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Coculture Techniques , Humans , Lung Neoplasms/drug therapy , Macrophages/metabolism , Mice , Phenotype , Tumor Microenvironment
3.
Mol Cancer Ther ; 21(1): 38-47, 2022 01.
Article in English | MEDLINE | ID: mdl-34675118

ABSTRACT

Pancreatic cancer is the third leading cause of cancer-related deaths in the United States with a 5-year survival less than 5%. Resistance to standard therapy and limited response to immune checkpoint blockade due to the immunosuppressive and stroma-rich microenvironment remain major challenges in the treatment of pancreatic cancer. A key cellular program involved in therapy resistance is epithelial plasticity, which is also associated with invasion, metastasis, and evasion of immune surveillance. The receptor tyrosine kinase AXL is a key driver of tumor cell epithelial plasticity. High expression and activity of AXL is associated with poor prognosis, metastasis, and therapy resistance in multiple types of cancer including pancreatic. Here, we show that an AXL inhibitor (TP-0903), has antitumor and therapy sensitizing effects in preclinical models of pancreatic ductal adenocarcinoma (PDA). We demonstrate that TP-0903 as a single agent or in combination with gemcitabine and/or anti-programmed cell death protein 1 (PD1) antibody has anti-metastatic and anti-tumor effects in PDA tumor bearing mice, leading to increased survival. In addition, gene expression analysis of tumors demonstrated upregulation of pro-inflammatory and immune activation genes in tumors from TP-0903-treated animals compared with the vehicle, indicating pharmacologic inhibition of AXL activation leads to an immunostimulatory microenvironment. This effect was augmented when TP-0903 was combined with gemcitabine and anti-PD1 antibody. These results provide clear rationale for evaluating TP-0903 in the treatment of pancreatic cancer.


Subject(s)
Immunotherapy/methods , Pancreatic Neoplasms/drug therapy , Proto-Oncogene Proteins/therapeutic use , Pyrimidines/therapeutic use , Receptor Protein-Tyrosine Kinases/therapeutic use , Sulfonamides/therapeutic use , Animals , Cell Line, Tumor , Humans , Mice , Neoplasm Metastasis , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins/pharmacology , Pyrimidines/pharmacology , Receptor Protein-Tyrosine Kinases/pharmacology , Sulfonamides/pharmacology , Survival Analysis , Tumor Microenvironment , Axl Receptor Tyrosine Kinase
4.
Mol Cancer Res ; 19(8): 1412-1421, 2021 08.
Article in English | MEDLINE | ID: mdl-33811159

ABSTRACT

Pancreatic ductal adenocarcinoma (PDA), a leading cause of cancer-related death in the United States, has a high metastatic rate, and is associated with persistent immune suppression. AXL, a member of the TAM (TYRO3, AXL, MERTK) receptor tyrosine kinase family, is a driver of metastasis and immune suppression in multiple cancer types. Here we use single-cell RNA-sequencing to reveal that AXL is expressed highly in tumor cells that have a mesenchymal-like phenotype and that AXL expression correlates with classic markers of epithelial-to-mesenchymal transition. We demonstrate that AXL deficiency extends survival, reduces primary and metastatic burden, and enhances sensitivity to gemcitabine in an autochthonous model of PDA. PDA in AXL-deficient mice displayed a more differentiated histology, higher nucleoside transporter expression, and a more active immune microenvironment compared with PDA in wild-type mice. Finally, we demonstrate that AXL-positive poorly differentiated tumor cells are critical for PDA progression and metastasis, emphasizing the potential of AXL as a therapeutic target in PDA. IMPLICATIONS: These studies implicate AXL as a marker of undifferentiated PDA cells and a target for therapy.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Plasticity/physiology , Neoplasm Metastasis/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Animals , Biomarkers, Tumor/metabolism , Carcinoma, Pancreatic Ductal/drug therapy , Cell Line, Tumor , Cell Plasticity/drug effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/physiology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Pancreatic Neoplasms/drug therapy , Tumor Microenvironment/drug effects , Tumor Microenvironment/physiology , Gemcitabine , Axl Receptor Tyrosine Kinase
5.
J Exp Med ; 218(6)2021 06 07.
Article in English | MEDLINE | ID: mdl-33830176

ABSTRACT

Antimalarial antibody responses are essential for mediating the clearance of Plasmodium parasite-infected RBCs from infected hosts. However, the rapid appearance of large numbers of plasmablasts in Plasmodium-infected hosts can suppress the development and function of durable humoral immunity. Here, we identify that the formation of plasmablast populations in Plasmodium-infected mice is mechanistically linked to both hemolysis-induced exposure of phosphatidylserine on damaged RBCs and inflammatory cues. We also show that virus and Trypanosoma infections known to trigger hemolytic anemia and high-grade inflammation also induce exuberant plasmablast responses. The induction of hemolysis or administration of RBC membrane ghosts increases plasmablast differentiation. The phosphatidylserine receptor Axl is critical for optimal plasmablast formation, and blocking phosphatidylserine limits plasmablast expansions and reduces Plasmodium parasite burden in vivo. Our findings support that strategies aimed at modulating polyclonal B cell activation and phosphatidylserine exposure may improve immune responses against Plasmodium parasites and potentially other infectious diseases that are associated with anemia.


Subject(s)
Cell Differentiation/immunology , Hemolysis/immunology , Phosphatidylserines/immunology , Plasma Cells/immunology , Animals , Antibodies, Protozoan/immunology , Antimalarials/immunology , B-Lymphocytes/immunology , B-Lymphocytes/parasitology , Cells, Cultured , Erythrocytes/immunology , Erythrocytes/parasitology , Humans , Immunity, Humoral/immunology , Malaria/immunology , Malaria/parasitology , Mice , Mice, Inbred C57BL , Plasma Cells/parasitology , Plasmodium yoelii/immunology
6.
EMBO Mol Med ; 11(11): e10515, 2019 11 07.
Article in English | MEDLINE | ID: mdl-31609088

ABSTRACT

TGFß is important during pancreatic ductal adenocarcinoma (PDA) progression. Canonical TGFß signaling suppresses epithelial pancreatic cancer cell proliferation; as a result, inhibiting TGFß has not been successful in PDA. In contrast, we demonstrate that inhibition of stromal TGFßR2 reduces IL-6 production from cancer-associated fibroblasts, resulting in a reduction of STAT3 activation in tumor cells and reversion of the immunosuppressive landscape. Up to 7% of human PDA have tumor cell-specific deficiency in canonical TGFß signaling via loss of TGFßR2. We demonstrate that in PDA that harbors epithelial loss of TGFßR2, inhibition of TGFß signaling is selective for stromal cells and results in a therapeutic benefit. Our study highlights the potential benefit of TGFß blockade in PDA and the importance of stratifying PDA patients who might benefit from such therapy.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Cardiomegaly , Humans , Signal Transduction , Transforming Growth Factor beta
7.
Elife ; 82019 05 28.
Article in English | MEDLINE | ID: mdl-31134894

ABSTRACT

Cancer evolves through a multistep process that occurs by the temporal accumulation of genetic mutations. Tumor-derived exosomes are emerging contributors to tumorigenesis. To understand how exosomes might contribute to cell transformation, we utilized the classic two-step NIH/3T3 cell transformation assay and observed that exosomes isolated from pancreatic cancer cells, but not normal human cells, can initiate malignant cell transformation and these transformed cells formed tumors in vivo. However, cancer cell exosomes are unable to transform cells alone or to act as a promoter of cell transformation. Utilizing proteomics and exome sequencing, we discovered cancer cell exosomes act as an initiator by inducing random mutations in recipient cells. Cells from the pool of randomly mutated cells are driven to transformation by a classic promoter resulting in foci, each of which encode a unique genetic profile. Our studies describe a novel molecular understanding of how cancer cell exosomes contribute to cell transformation. Editorial note: This article has been through an editorial process in which the authors decide how to respond to the issues raised during peer review. The Reviewing Editor's assessment is that major issues remain unresolved (see decision letter).


Subject(s)
Cell Transformation, Neoplastic/pathology , Exosomes/metabolism , Pancreatic Neoplasms/pathology , Animals , Cell Line, Tumor , Disease Models, Animal , Exosomes/chemistry , Genomics , Humans , Mice , NIH 3T3 Cells , Neoplasm Transplantation , Proteomics
8.
Cancer Res ; 78(1): 246-255, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29180468

ABSTRACT

Activation of the receptor tyrosine kinase Axl is associated with poor outcomes in pancreatic cancer (PDAC), where it coordinately mediates immune evasion and drug resistance. Here, we demonstrate that the selective Axl kinase inhibitor BGB324 targets the tumor-immune interface to blunt the aggressive traits of PDAC cells in vitro and enhance gemcitibine efficacy in vivo Axl signaling stimulates the TBK1-NFκB pathway and innate immune suppression in the tumor microenvironment. In tumor cells, BGB324 treatment drove epithelial differentiation, expression of nucleoside transporters affecting gemcitabine response, and an immune stimulatory microenvironment. Our results establish a preclinical mechanistic rationale for the clinical development of Axl inhibitors to improve the treatment of PDAC patients.Significance: These results establish a preclinical mechanistic rationale for the clinical development of AXL inhibitors to improve the treatment of PDAC patients. Cancer Res; 78(1); 246-55. ©2017 AACR.


Subject(s)
Benzocycloheptenes/pharmacology , Carcinoma, Pancreatic Ductal/drug therapy , Pancreatic Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Triazoles/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzocycloheptenes/administration & dosage , Carcinoma, Pancreatic Ductal/immunology , Cell Line, Tumor , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Female , Humans , Male , Mice, Inbred C57BL , Mice, Transgenic , Molecular Targeted Therapy , Pancreatic Neoplasms/immunology , Triazoles/administration & dosage , Xenograft Model Antitumor Assays , Gemcitabine , Axl Receptor Tyrosine Kinase
9.
Nat Cell Biol ; 19(10): 1226-1236, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28945232

ABSTRACT

Direct interactions between pro- and anti-apoptotic BCL-2 family members form the basis of cell death decision-making at the outer mitochondrial membrane (OMM). Here we report that three anti-apoptotic BCL-2 proteins (MCL-1, BCL-2 and BCL-XL) found untethered from the OMM function as transcriptional regulators of a prosurvival and growth program. Anti-apoptotic BCL-2 proteins engage a BCL-2 homology (BH) domain sequence found in SUFU (suppressor of fused), a tumour suppressor and antagonist of the GLI DNA-binding proteins. BCL-2 proteins directly promote SUFU turnover, inhibit SUFU-GLI interaction, and induce the expression of the GLI target genes BCL-2, MCL-1 and BCL-XL. Anti-apoptotic BCL-2 protein/SUFU feedforward signalling promotes cancer cell survival and growth, and can be disabled with BH3 mimetics-small molecules that target anti-apoptotic BCL-2 proteins. Our findings delineate a chemical strategy for countering drug resistance in GLI-associated tumours and reveal unanticipated functions for BCL-2 proteins as transcriptional regulators.


Subject(s)
Neoplasms/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Repressor Proteins/metabolism , Transcription, Genetic , Tumor Suppressor Proteins/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis , CRISPR-Cas Systems , Cell Proliferation , Cell Survival , Female , Gene Expression Regulation, Neoplastic , Genotype , HEK293 Cells , Humans , Mice , Mice, Knockout , Mice, Nude , Molecular Mimicry , Myeloid Cell Leukemia Sequence 1 Protein/deficiency , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , NIH 3T3 Cells , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , Peptide Fragments/metabolism , Phenotype , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , RNA Interference , Repressor Proteins/genetics , Signal Transduction , Time Factors , Transcription, Genetic/drug effects , Transfection , Tumor Suppressor Proteins/genetics , Zinc Finger Protein GLI1/genetics , Zinc Finger Protein GLI1/metabolism , bcl-X Protein/genetics , bcl-X Protein/metabolism
10.
Oncotarget ; 7(32): 51569-51580, 2016 08 09.
Article in English | MEDLINE | ID: mdl-27438140

ABSTRACT

Pancreatic carcinoma ranks among the most lethal of human cancers. Besides late detection, other factors contribute to its lethality, including a high degree of chemoresistance, invasion, and distant metastases. Currently, the mainstay of therapy involves resection of local disease in a minority of patients (Whipple procedure) and systemic gemcitabine. While systemic chemotherapy has some benefit, even with optimal treatment, the five year survival after diagnosis is dismal. Thus, treatment of pancreatic carcinoma remains a tremendous unmet need.The organometallic compound tris DBA palladium is a potent inhibitor of N-myristoyltransferase 1 (NMT1), an enzyme that catalyzes the transfer of myristate to protein substrates. This compound is highly effective in vivo against murine models of melanoma with both mutant and wild type b-RAF genotypes. Based upon the signaling similarities between melanoma and pancreatic carcinoma, we evaluated the efficacy of tris DBA palladium in vitro and in vivo against pancreatic carcinoma. We found that tris DBA palladium decreased proliferation and colony formation of pancreatic cancer cells in vitro. In an orthotopic mouse model, tris DBA palladium was highly active in inhibiting growth, ascites production, and distant metastases in vivo. Furthermore, tris DBA palladium impaired chemotaxis and inhibited cilia formation in Pan02 cells in a NMT1-dependent manner. We propose that NMT1 is a novel regulator of cilia formation and tris DBA palladium a novel inhibitor of cilia formation and metastasis in pancreatic cancer. Thus, further evaluation of tris DBA palladium for the treatment of pancreatic cancer is warranted.


Subject(s)
Cell Movement/drug effects , Cell Proliferation/drug effects , Organometallic Compounds/pharmacology , Pancreatic Neoplasms/pathology , Animals , Cell Line, Tumor , Humans , Male , Mice , Mice, Inbred C57BL , Neoplasm Metastasis , Organometallic Compounds/therapeutic use , Pancreatic Neoplasms/drug therapy , Treatment Outcome , Xenograft Model Antitumor Assays
11.
Oncotarget ; 7(21): 30678-90, 2016 May 24.
Article in English | MEDLINE | ID: mdl-27120792

ABSTRACT

Phosphatidylserine (PS) is an anionic phospholipid maintained on the inner-leaflet of the cell membrane and is externalized in malignant cells. We previously launched a careful unbiased selection targeting biomolecules (e.g. protein, lipid or carbohydrate) distinct to cancer cells by exploiting HCC4017 lung cancer and HBEC30KT normal epithelial cells derived from the same patient, identifying HCC4017 specific peptide-peptoid hybrid PPS1. In this current study, we identified PS as the target of PPS1. We validated direct PPS1 binding to PS using ELISA-like assays, lipid dot blot and liposome based binding assays. In addition, PPS1 recognized other negatively charged and cancer specific lipids such as phosphatidic acid, phosphatidylinositol and phosphatidylglycerol. PPS1 did not bind to neutral lipids such as phosphatidylethanolamine found in cancer and phosphatidylcholine and sphingomyelin found in normal cells. Further we found that the dimeric version of PPS1 (PPS1D1) displayed strong cytotoxicity towards lung cancer cell lines that externalize PS, but not normal cells. PPS1D1 showed potent single agent anti-tumor activity and enhanced the efficacy of docetaxel in mice bearing H460 lung cancer xenografts. Since PS and anionic phospholipid externalization is common across many cancer types, PPS1 may be an alternative to overcome limitations of protein targeted agents.


Subject(s)
Lung Neoplasms/drug therapy , Oligopeptides/pharmacology , Phosphatidylserines/antagonists & inhibitors , Xenograft Model Antitumor Assays , Animals , Cell Line , Cell Line, Tumor , Female , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Membrane Lipids/antagonists & inhibitors , Membrane Lipids/metabolism , Mice, Inbred NOD , Mice, SCID , Molecular Targeted Therapy , Oligopeptides/chemistry , Oligopeptides/metabolism , Peptides/chemistry , Peptoids/chemistry , Phosphatidylserines/metabolism , Protein Binding , Tumor Burden/drug effects
12.
Cancer Res ; 75(23): 5058-69, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26577699

ABSTRACT

Elevated oxidative stress is an aberration seen in many solid tumors, and exploiting this biochemical difference has the potential to enhance the efficacy of anticancer agents. Homeostasis of reactive oxygen species (ROS) is important for normal cell function, but excessive production of ROS can result in cellular toxicity, and therefore ROS levels must be balanced finely. Here, we highlight the relationship between the extracellular matrix and ROS production by reporting a novel function of the matricellular protein Fibulin-5 (Fbln5). We used genetically engineered mouse models of pancreatic ductal adenocarcinoma (PDAC) and found that mutation of the integrin-binding domain of Fbln5 led to decreased tumor growth, increased survival, and enhanced chemoresponse to standard PDAC therapies. Through mechanistic investigations, we found that improved survival was due to increased levels of oxidative stress in Fbln5-mutant tumors. Furthermore, loss of the Fbln5-integrin interaction augmented fibronectin signaling, driving integrin-induced ROS production in a 5-lipooxygenase-dependent manner. These data indicate that Fbln5 promotes PDAC progression by functioning as a molecular rheostat that modulates cell-ECM interactions to reduce ROS production, and thus tip the balance in favor of tumor cell survival and treatment-refractory disease.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Extracellular Matrix Proteins/metabolism , Pancreatic Neoplasms/metabolism , Reactive Oxygen Species/metabolism , Recombinant Proteins/metabolism , Animals , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Extracellular Matrix Proteins/biosynthesis , Humans , Mice , Mice, Transgenic , Oxidative Stress/physiology , Pancreatic Neoplasms/pathology , Recombinant Proteins/biosynthesis , Tumor Microenvironment/physiology
13.
Cancer Res ; 75(18): 3699-705, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26206560

ABSTRACT

Repurposing "old" drugs can facilitate rapid clinical translation but necessitates novel mechanistic insight. Warfarin, a vitamin K "antagonist" used clinically for the prevention of thrombosis for more than 50 years, has been shown to have anticancer effects. We hypothesized that the molecular mechanism underlying its antitumor activity is unrelated to its effect on coagulation, but is due to inhibition of the Axl receptor tyrosine kinase on tumor cells. Activation of Axl by its ligand Gas6, a vitamin K-dependent protein, is inhibited at doses of warfarin that do not affect coagulation. Here, we show that inhibiting Gas6-dependent Axl activation with low-dose warfarin, or with other tumor-specific Axl-targeting agents, blocks the progression and spread of pancreatic cancer. Warfarin also inhibited Axl-dependent tumor cell migration, invasiveness, and proliferation while increasing apoptosis and sensitivity to chemotherapy. We conclude that Gas6-induced Axl signaling is a critical driver of pancreatic cancer progression and its inhibition with low-dose warfarin or other Axl-targeting agents may improve outcome in patients with Axl-expressing tumors.


Subject(s)
Carcinoma, Pancreatic Ductal/drug therapy , Epithelial-Mesenchymal Transition/drug effects , Intercellular Signaling Peptides and Proteins/physiology , Neoplasm Proteins/physiology , Pancreatic Neoplasms/drug therapy , Proto-Oncogene Proteins/physiology , Receptor Protein-Tyrosine Kinases/physiology , Warfarin/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Division/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Disease Progression , Drug Synergism , Female , Gene Knockdown Techniques , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Proteins/antagonists & inhibitors , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , RNA, Small Interfering/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/genetics , Signal Transduction/drug effects , Specific Pathogen-Free Organisms , Warfarin/administration & dosage , Warfarin/therapeutic use , Xenograft Model Antitumor Assays , Gemcitabine , Axl Receptor Tyrosine Kinase
14.
Nat Chem Biol ; 11(6): 401-8, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25867045

ABSTRACT

Modern cancer treatment employs many effective chemotherapeutic agents originally discovered from natural sources. The cyclic depsipeptide didemnin B has demonstrated impressive anticancer activity in preclinical models. Clinical use has been approved but is limited by sparse patient responses combined with toxicity risk and an unclear mechanism of action. From a broad-scale effort to match antineoplastic natural products to their cellular activities, we found that didemnin B selectively induces rapid and wholesale apoptosis through dual inhibition of PPT1 and EEF1A1. Furthermore, empirical discovery of a small panel of exceptional responders to didemnin B allowed the generation of a regularized regression model to extract a sparse-feature genetic biomarker capable of predicting sensitivity to didemnin B. This may facilitate patient selection in a fashion that could enhance and expand the therapeutic application of didemnin B against neoplastic disease.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Depsipeptides/pharmacology , Membrane Proteins/antagonists & inhibitors , Peptide Elongation Factor 1/antagonists & inhibitors , Pharmacogenetics , Apoptosis/genetics , Biomarkers/metabolism , Cell Line, Tumor , Genome-Wide Association Study , Humans , Mechanistic Target of Rapamycin Complex 1 , Membrane Proteins/genetics , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Peptide Elongation Factor 1/genetics , Protein Biosynthesis/drug effects , Protein Biosynthesis/genetics , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Thiolester Hydrolases , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics
15.
Cancer Res ; 74(4): 1032-44, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24346431

ABSTRACT

There is growing evidence that antiangiogenic therapy stimulates cancer cell invasion and metastasis. However, the underlying molecular mechanisms responsible for these changes have not been fully defined. Here, we report that anti-VEGF therapy promotes local invasion and metastasis by inducing collagen signaling in cancer cells. We show that chronic VEGF inhibition in a genetically engineered mouse model of pancreatic ductal adenocarcinoma (PDA) induces hypoxia, a less differentiated mesenchymal-like tumor cell phenotype, TGF-ß expression, and collagen deposition and signaling. In addition, we show that collagen signaling is critical for protumorigenic activity of TGF-ß in vitro. To further model the impact of collagen signaling in tumors, we evaluated PDA in mice lacking Sparc, a protein that reduces collagen binding to cell surface receptors. Importantly, we show that loss of Sparc increases collagen signaling and tumor progression. Together, these findings suggest that collagen actively promotes PDA spread and that enhanced disease progression associated with anti-VEGF therapy can arise from elevated extracellular matrix-mediated signaling.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/pathology , Collagen/physiology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Bevacizumab , Cell Line, Tumor , Disease Models, Animal , Disease Progression , Mice , Mice, Transgenic , Neoplasm Invasiveness , Signal Transduction/physiology , Treatment Failure , Vascular Endothelial Growth Factor A/immunology
16.
Cell ; 155(3): 552-66, 2013 Oct 24.
Article in English | MEDLINE | ID: mdl-24243015

ABSTRACT

Context-specific molecular vulnerabilities that arise during tumor evolution represent an attractive intervention target class. However, the frequency and diversity of somatic lesions detected among lung tumors can confound efforts to identify these targets. To confront this challenge, we have applied parallel screening of chemical and genetic perturbations within a panel of molecularly annotated NSCLC lines to identify intervention opportunities tightly linked to molecular response indicators predictive of target sensitivity. Anchoring this analysis on a matched tumor/normal cell model from a lung adenocarcinoma patient identified three distinct target/response-indicator pairings that are represented with significant frequencies (6%-16%) in the patient population. These include NLRP3 mutation/inflammasome activation-dependent FLIP addiction, co-occurring KRAS and LKB1 mutation-driven COPI addiction, and selective sensitivity to a synthetic indolotriazine that is specified by a seven-gene expression signature. Target efficacies were validated in vivo, and mechanism-of-action studies informed generalizable principles underpinning cancer cell biology.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Drug Screening Assays, Antitumor , Indoles/pharmacology , Lung Neoplasms/metabolism , Triazines/pharmacology , Animals , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Carrier Proteins , Cell Line, Tumor , Coatomer Protein/metabolism , Female , Genes, ras , Heterografts , Humans , Lung Neoplasms/pathology , Lysosomes/metabolism , Mice , Molecular Targeted Therapy , NLR Family, Pyrin Domain-Containing 3 Protein , Neoplasm Transplantation , Oxidative Phosphorylation
17.
Mol Cancer Ther ; 11(11): 2473-82, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22933705

ABSTRACT

In lung cancer, platelet-derived growth factor receptor α (PDGFRα) is expressed frequently by tumor-associated stromal cells and by cancer cells in a subset of tumors. We sought to determine the effect of targeting stromal PDGFRα in preclinical lung tumor xenograft models (human tumor, mouse stroma). Effects of anti-human (IMC-3G3) and anti-mouse (1E10) PDGFRα monoclonal antibodies (mAb) on proliferation and PDGFRα signaling were evaluated in lung cancer cell lines and mouse fibroblasts. Therapy studies were conducted using established PDGFRα-positive H1703 cells and PDGFRα-negative Calu-6, H1993, and A549 subcutaneous tumors in immunocompromised mice treated with vehicle, anti-PDGFRα mAbs, chemotherapy, or combination therapy. Tumors were analyzed for growth and levels of growth factors. IMC-3G3 inhibited PDGFRα activation and the growth of H1703 cells in vitro and tumor growth in vivo, but had no effect on PDGFRα-negative cell lines or mouse fibroblasts. 1E10 inhibited growth and PDGFRα activation of mouse fibroblasts, but had no effect on human cancer cell lines in vitro. In vivo, 1E10-targeted inhibition of murine PDGFRα reduced tumor growth as single-agent therapy in Calu-6 cells and enhanced the effect of chemotherapy in xenografts derived from A549 cells. We also identified that low expression cancer cell expression of VEGF-A and elevated expression of PDGF-AA were associated with response to stromal PDGFRα targeting. We conclude that stromal PDGFRα inhibition represents a means for enhancing control of lung cancer growth in some cases, independent of tumor cell PDGFRα expression.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , Molecular Targeted Therapy , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Xenograft Model Antitumor Assays , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Female , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mice , Platelet-Derived Growth Factor/metabolism , Receptor, Platelet-Derived Growth Factor alpha/antagonists & inhibitors , Species Specificity , Stromal Cells/drug effects , Stromal Cells/metabolism , Stromal Cells/pathology , Vascular Endothelial Growth Factor A/metabolism
18.
Clin Cancer Res ; 18(18): 5031-42, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22829202

ABSTRACT

PURPOSE: COX-2 is expressed highly in pancreatic cancer and implicated in tumor progression. COX-2 inhibition can reduce tumor growth and augment therapy. The precise function of COX-2 in tumors remains poorly understood, but it is implicated in tumor angiogenesis, evasion of apoptosis, and induction of epithelial-to-mesenchymal transition (EMT). Current therapeutic regimens for pancreatic cancer are minimally effective, highlighting the need for novel treatment strategies. Here, we report that apricoxib, a novel COX-2 inhibitor in phase II clinical trials, significantly enhances the efficacy of gemcitabine/erlotinib in preclinical models of pancreatic cancer. EXPERIMENTAL DESIGN: Human pancreatic cell lines were evaluated in vitro and in vivo for response to apricoxib ± standard-of-care therapy (gemcitabine + erlotinib). Tumor tissue underwent posttreatment analysis for cell proliferation, viability, and EMT phenotype. Vascular parameters were also determined. RESULTS: COX-2 inhibition reduced the IC(50) of gemcitabine ± erlotinib in six pancreatic cancer cell lines tested in vitro. Furthermore, apricoxib increased the antitumor efficacy of standard combination therapy in several orthotopic xenograft models. In vivo apricoxib combination therapy was only effective at reducing tumor growth and metastasis in tumors with elevated COX-2 activity. In each model examined, treatment with apricoxib resulted in vascular normalization without a decrease in microvessel density and promotion of an epithelial phenotype by tumor cells regardless of basal COX-2 expression. CONCLUSIONS: Apricoxib robustly reverses EMT and augments standard therapy without reducing microvessel density and warrants further clinical evaluation in patients with pancreatic cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Cyclooxygenase 2 Inhibitors/therapeutic use , Pancreatic Neoplasms/drug therapy , Pyrroles/therapeutic use , Sulfonamides/therapeutic use , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/administration & dosage , Cyclooxygenase 2 Inhibitors/pharmacology , Drug Synergism , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , ErbB Receptors/metabolism , Erlotinib Hydrochloride , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Metastasis , Neovascularization, Pathologic/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pyrroles/administration & dosage , Pyrroles/pharmacology , Quinazolines/pharmacology , Sulfonamides/administration & dosage , Sulfonamides/pharmacology
19.
Carcinogenesis ; 33(9): 1639-46, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22678114

ABSTRACT

Although cyclooxygenase-2 (COX-2) inhibitors, such as the late stage development drug apricoxib, exhibit antitumor activity, their mechanisms of action have not been fully defined. In this study, we characterized the mechanisms of action of apricoxib in HT29 colorectal carcinoma. Apricoxib was weakly cytotoxic toward naive HT29 cells in vitro but inhibited tumor growth markedly in vivo. Pharmacokinetic analyses revealed that in vivo drug levels peaked at 2-4 µM and remained sufficient to completely inhibit prostaglandin E(2) production, but failed to reach concentrations cytotoxic for HT29 cells in monolayer culture. Despite this, apricoxib significantly inhibited tumor cell proliferation and induced apoptosis without affecting blood vessel density, although it did promote vascular normalization. Strikingly, apricoxib treatment induced a dose-dependent reversal of epithelial-mesenchymal transition (EMT), as shown by robust upregulation of E-cadherin and the virtual disappearance of vimentin and ZEB1 protein expression. In vitro, either anchorage-independent growth conditions or forced EMT sensitized HT29 and non-small cell lung cancer cells to apricoxib by 50-fold, suggesting that the occurrence of EMT may actually increase the dependence of colon and lung carcinoma cells on COX-2. Taken together, these data suggest that acquisition of mesenchymal characteristics sensitizes carcinoma cells to apricoxib resulting in significant single-agent antitumor activity.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclooxygenase 2 Inhibitors/pharmacology , Epithelial-Mesenchymal Transition , Pyrroles/pharmacology , Sulfonamides/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Dinoprostone/biosynthesis , Female , HT29 Cells , Humans , Mice , Mice, Nude , Vascular Endothelial Growth Factor A/physiology
20.
PLoS One ; 7(2): e31384, 2012.
Article in English | MEDLINE | ID: mdl-22348081

ABSTRACT

Pancreatic adenocarcinoma, a desmoplastic disease, is the fourth leading cause of cancer-related death in the Western world due, in large part, to locally invasive primary tumor growth and ensuing metastasis. SPARC is a matricellular protein that governs extracellular matrix (ECM) deposition and maturation during tissue remodeling, particularly, during wound healing and tumorigenesis. In the present study, we sought to determine the mechanism by which lack of host SPARC alters the tumor microenvironment and enhances invasion and metastasis of an orthotopic model of pancreatic cancer. We identified that levels of active TGFß1 were increased significantly in tumors grown in SPARC-null mice. TGFß1 contributes to many aspects of tumor development including metastasis, endothelial cell permeability, inflammation and fibrosis, all of which are altered in the absence of stromal-derived SPARC. Given these results, we performed a survival study to assess the contribution of increased TGFß1 activity to tumor progression in SPARC-null mice using losartan, an angiotensin II type 1 receptor antagonist that diminishes TGFß1 expression and activation in vivo. Tumors grown in SPARC-null mice progressed more quickly than those grown in wild-type littermates leading to a significant reduction in median survival. However, median survival of SPARC-null animals treated with losartan was extended to that of losartan-treated wild-type controls. In addition, losartan abrogated TGFß induced gene expression, reduced local invasion and metastasis, decreased vascular permeability and altered the immune profile of tumors grown in SPARC-null mice. These data support the concept that aberrant TGFß1-activation in the absence of host SPARC contributes significantly to tumor progression and suggests that SPARC, by controlling ECM deposition and maturation, can regulate TGFß availability and activation.


Subject(s)
Losartan/pharmacology , Osteonectin/deficiency , Pancreatic Neoplasms/drug therapy , Transforming Growth Factor beta/antagonists & inhibitors , Angiotensin II Type 1 Receptor Blockers , Animals , Disease Progression , Extracellular Matrix/metabolism , Losartan/therapeutic use , Mice , Mice, Knockout , Neoplasm Invasiveness , Neoplasm Metastasis , Survival Rate , Transforming Growth Factor beta/metabolism , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...