Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Transl Med ; 11(483)2019 03 13.
Article in English | MEDLINE | ID: mdl-30867319

ABSTRACT

Mitogen-activated protein kinase (MAPK) kinase (MEK) inhibitors have failed to show clinical benefit in Kirsten rat sarcoma (KRAS) mutant lung cancer due to various resistance mechanisms. To identify differential therapeutic sensitivities between epithelial and mesenchymal lung tumors, we performed in vivo small hairpin RNA screens, proteomic profiling, and analysis of patient tumor datasets, which revealed an inverse correlation between mitogen-activated protein kinase (MAPK) signaling dependency and a zinc finger E-box binding homeobox 1 (ZEB1)-regulated epithelial-to-mesenchymal transition. Mechanistic studies determined that MAPK signaling dependency in epithelial lung cancer cells is due to the scaffold protein interleukin-17 receptor D (IL17RD), which is directly repressed by ZEB1. Lung tumors in multiple Kras mutant murine models with increased ZEB1 displayed low IL17RD expression, accompanied by MAPK-independent tumor growth and therapeutic resistance to MEK inhibition. Suppression of ZEB1 function with miR-200 expression or the histone deacetylase inhibitor mocetinostat sensitized resistant cancer cells to MEK inhibition and markedly reduced in vivo tumor growth, showing a promising combinatorial treatment strategy for KRAS mutant cancers. In human lung tumor samples, high ZEB1 and low IL17RD expression correlated with low MAPK signaling, presenting potential markers that predict patient response to MEK inhibitors.


Subject(s)
Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mutation/genetics , Neoplasms/genetics , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins p21(ras)/genetics , Receptors, Interleukin-17/metabolism , Zinc Finger E-box-Binding Homeobox 1/metabolism , Animals , Benzamides/pharmacology , Benzamides/therapeutic use , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Drug Resistance, Neoplasm , Epithelial Cells/pathology , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , MAP Kinase Signaling System , Mesoderm/pathology , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Neoplasms/drug therapy , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins p21(ras)/metabolism , Pyrimidines/pharmacology , Pyrimidines/therapeutic use
2.
Cancer Res ; 78(21): 6196-6208, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30185546

ABSTRACT

Cancer testis antigens (CTA) are expressed in testis and placenta and anomalously activated in a variety of tumors. The mechanistic contribution of CTAs to neoplastic phenotypes remains largely unknown. Using a chemigenomics approach, we find that the CTA HORMAD1 correlates with resistance to the mitochondrial complex I inhibitor piericidin A in non-small cell lung cancer (NSCLC). Resistance was due to a reductive intracellular environment that attenuated the accumulation of free radicals. In human lung adenocarcinoma (LUAD) tumors, patients expressing high HORMAD1 exhibited elevated mutational burden and reduced survival. HORMAD1 tumors were enriched for genes essential for homologous recombination (HR), and HORMAD1 promoted RAD51-filament formation, but not DNA resection, during HR. Accordingly, HORMAD1 loss enhanced sensitivity to γ-irradiation and PARP inhibition, and HORMAD1 depletion significantly reduced tumor growth in vivo These results suggest that HORMAD1 expression specifies a novel subtype of LUAD, which has adapted to mitigate DNA damage. In this setting, HORMAD1 could represent a direct target for intervention to enhance sensitivity to DNA-damaging agents or as an immunotherapeutic target in patients.Significance: This study uses a chemigenomics approach to demonstrate that anomalous expression of the CTA HORMAD1 specifies resistance to oxidative stress and promotes HR to support tumor cell survival in NSCLC. Cancer Res; 78(21); 6196-208. ©2018 AACR.


Subject(s)
Adenocarcinoma of Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/diagnosis , Cell Cycle Proteins/metabolism , Gene Expression Regulation, Neoplastic , Lung Neoplasms/diagnosis , A549 Cells , Adenocarcinoma of Lung/metabolism , Animals , Antigens, Neoplasm/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Survival , DNA Damage , DNA Repair , Female , Free Radicals , Gene Expression Profiling , Humans , Lung Neoplasms/metabolism , Mice , Mice, Inbred NOD , Mutagens , Neoplasm Transplantation , Oxidative Stress , Prognosis , Recombination, Genetic
3.
Cancer Discov ; 8(9): 1156-1175, 2018 09.
Article in English | MEDLINE | ID: mdl-30012853

ABSTRACT

Although treatment with immune checkpoint inhibitors provides promising benefit for patients with cancer, optimal use is encumbered by high resistance rates and requires a thorough understanding of resistance mechanisms. We observed that tumors treated with PD-1/PD-L1 blocking antibodies develop resistance through the upregulation of CD38, which is induced by all-trans retinoic acid and IFNß in the tumor microenvironment. In vitro and in vivo studies demonstrate that CD38 inhibits CD8+ T-cell function via adenosine receptor signaling and that CD38 or adenosine receptor blockade are effective strategies to overcome the resistance. Large data sets of human tumors reveal expression of CD38 in a subset of tumors with high levels of basal or treatment-induced T-cell infiltration, where immune checkpoint therapies are thought to be most effective. These findings provide a novel mechanism of acquired resistance to immune checkpoint therapy and an opportunity to expand their efficacy in cancer treatment.Significance: CD38 is a major mechanism of acquired resistance to PD-1/PD-L1 blockade, causing CD8+ T-cell suppression. Coinhibition of CD38 and PD-L1 improves antitumor immune response. Biomarker assessment in patient cohorts suggests that a combination strategy is applicable to a large percentage of patients in whom PD-1/PD-L1 blockade is currently indicated. Cancer Discov; 8(9); 1156-75. ©2018 AACR.See related commentary by Mittal et al., p. 1066This article is highlighted in the In This Issue feature, p. 1047.


Subject(s)
ADP-ribosyl Cyclase 1/metabolism , Antineoplastic Agents, Immunological/administration & dosage , Carcinoma, Non-Small-Cell Lung/drug therapy , Drug Resistance, Neoplasm/drug effects , Lung Neoplasms/drug therapy , Melanoma/drug therapy , Membrane Glycoproteins/metabolism , ADP-ribosyl Cyclase 1/antagonists & inhibitors , Animals , Antineoplastic Agents, Immunological/pharmacology , B7-H1 Antigen/antagonists & inhibitors , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Non-Small-Cell Lung/immunology , Cell Line, Tumor , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Interferon-gamma/metabolism , Lung Neoplasms/immunology , Melanoma/genetics , Melanoma/immunology , Membrane Glycoproteins/antagonists & inhibitors , Mice , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Receptors, Purinergic P1/metabolism , Signal Transduction/drug effects , Tretinoin/metabolism , Tumor Microenvironment/drug effects , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
4.
Cancer Discov ; 8(7): 822-835, 2018 07.
Article in English | MEDLINE | ID: mdl-29773717

ABSTRACT

KRAS is the most common oncogenic driver in lung adenocarcinoma (LUAC). We previously reported that STK11/LKB1 (KL) or TP53 (KP) comutations define distinct subgroups of KRAS-mutant LUAC. Here, we examine the efficacy of PD-1 inhibitors in these subgroups. Objective response rates to PD-1 blockade differed significantly among KL (7.4%), KP (35.7%), and K-only (28.6%) subgroups (P < 0.001) in the Stand Up To Cancer (SU2C) cohort (174 patients) with KRAS-mutant LUAC and in patients treated with nivolumab in the CheckMate-057 phase III trial (0% vs. 57.1% vs. 18.2%; P = 0.047). In the SU2C cohort, KL LUAC exhibited shorter progression-free (P < 0.001) and overall (P = 0.0015) survival compared with KRASMUT;STK11/LKB1WT LUAC. Among 924 LUACs, STK11/LKB1 alterations were the only marker significantly associated with PD-L1 negativity in TMBIntermediate/High LUAC. The impact of STK11/LKB1 alterations on clinical outcomes with PD-1/PD-L1 inhibitors extended to PD-L1-positive non-small cell lung cancer. In Kras-mutant murine LUAC models, Stk11/Lkb1 loss promoted PD-1/PD-L1 inhibitor resistance, suggesting a causal role. Our results identify STK11/LKB1 alterations as a major driver of primary resistance to PD-1 blockade in KRAS-mutant LUAC.Significance: This work identifies STK11/LKB1 alterations as the most prevalent genomic driver of primary resistance to PD-1 axis inhibitors in KRAS-mutant lung adenocarcinoma. Genomic profiling may enhance the predictive utility of PD-L1 expression and tumor mutation burden and facilitate establishment of personalized combination immunotherapy approaches for genomically defined LUAC subsets. Cancer Discov; 8(7); 822-35. ©2018 AACR.See related commentary by Etxeberria et al., p. 794This article is highlighted in the In This Issue feature, p. 781.


Subject(s)
Adenocarcinoma of Lung/drug therapy , Drug Resistance, Neoplasm/genetics , Lung Neoplasms/drug therapy , Mutation , Nivolumab/therapeutic use , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins p21(ras)/genetics , AMP-Activated Protein Kinase Kinases , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/therapy , Adult , Aged , Aged, 80 and over , Animals , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Disease Models, Animal , Humans , Immunotherapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/therapy , Male , Mice , Middle Aged , Nivolumab/pharmacology , Prognosis , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Progression-Free Survival
5.
Cancer Res ; 78(11): 2911-2924, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29514796

ABSTRACT

Bone metastasis from prostate cancer can occur years after prostatectomy, due to reactivation of dormant disseminated tumor cells (DTC) in the bone, yet the mechanism by which DTCs are initially induced into a dormant state in the bone remains to be elucidated. We show here that the bone microenvironment confers dormancy to C4-2B4 prostate cancer cells, as they become dormant when injected into mouse femurs but not under the skin. Live-cell imaging of dormant cells at the single-cell level revealed that conditioned medium from differentiated, but not undifferentiated, osteoblasts induced C4-2B4 cellular quiescence, suggesting that differentiated osteoblasts present locally around the tumor cells in the bone conferred dormancy to prostate cancer cells. Gene array analyses identified GDF10 and TGFß2 among osteoblast-secreted proteins that induced quiescence of C4-2B4, C4-2b, and PC3-mm2, but not 22RV1 or BPH-1 cells, indicating prostate cancer tumor cells differ in their dormancy response. TGFß2 and GDF10 induced dormancy through TGFßRIII to activate phospho-p38MAPK, which phosphorylates retinoblastoma (RB) at the novel N-terminal S249/T252 sites to block prostate cancer cell proliferation. Consistently, expression of dominant-negative p38MAPK in C4-2b and C4-2B4 prostate cancer cell lines abolished tumor cell dormancy both in vitro and in vivo Lower TGFßRIII expression in patients with prostate cancer correlated with increased metastatic potential and decreased survival rates. Together, our results identify a dormancy mechanism by which DTCs are induced into a dormant state through TGFßRIII-p38MAPK-pS249/pT252-RB signaling and offer a rationale for developing strategies to prevent prostate cancer recurrence in the bone.Significance: These findings provide mechanistic insights into the dormancy of metastatic prostate cancer in the bone and offer a rationale for developing strategies to prevent prostate cancer recurrence in the bone. Cancer Res; 78(11); 2911-24. ©2018 AACR.


Subject(s)
Bone Neoplasms/metabolism , Osteoblasts/metabolism , Prostatic Neoplasms/metabolism , Proteoglycans/metabolism , Receptors, Transforming Growth Factor beta/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , A549 Cells , Animals , Bone and Bones/metabolism , Cell Differentiation/physiology , Cell Line, Tumor , Cell Proliferation/physiology , Humans , Male , Mice , Neoplasm Proteins/metabolism , PC-3 Cells , Prostate/metabolism , Signal Transduction/physiology
6.
J Clin Oncol ; 36(14): 1419-1427, 2018 05 10.
Article in English | MEDLINE | ID: mdl-29432077

ABSTRACT

Purpose DNX-2401 (Delta-24-RGD; tasadenoturev) is a tumor-selective, replication-competent oncolytic adenovirus. Preclinical studies demonstrated antiglioma efficacy, but the effects and mechanisms of action have not been evaluated in patients. Methods A phase I, dose-escalation, biologic-end-point clinical trial of DNX-2401 was conducted in 37 patients with recurrent malignant glioma. Patients received a single intratumoral injection of DNX-2401 into biopsy-confirmed recurrent tumor to evaluate safety and response across eight dose levels (group A). To investigate the mechanism of action, a second group of patients (group B) underwent intratumoral injection through a permanently implanted catheter, followed 14 days later by en bloc resection to acquire post-treatment specimens. Results In group A (n = 25), 20% of patients survived > 3 years from treatment, and three patients had a ≥ 95% reduction in the enhancing tumor (12%), with all three of these dramatic responses resulting in > 3 years of progression-free survival from the time of treatment. Analyses of post-treatment surgical specimens (group B, n = 12) showed that DNX-2401 replicates and spreads within the tumor, documenting direct virus-induced oncolysis in patients. In addition to radiographic signs of inflammation, histopathologic examination of immune markers in post-treatment specimens showed tumor infiltration by CD8+ and T-bet+ cells, and transmembrane immunoglobulin mucin-3 downregulation after treatment. Analyses of patient-derived cell lines for damage-associated molecular patterns revealed induction of immunogenic cell death in tumor cells after DNX-2401 administration. Conclusion Treatment with DNX-2401 resulted in dramatic responses with long-term survival in recurrent high-grade gliomas that are probably due to direct oncolytic effects of the virus followed by elicitation of an immune-mediated antiglioma response.


Subject(s)
Adenoviridae , Brain Neoplasms/drug therapy , Glioma/drug therapy , Neoplasm Recurrence, Local/drug therapy , Oncolytic Virotherapy/methods , Oncolytic Viruses , Adult , Biopsy , Female , Humans , Injections, Intralesional , Male , Middle Aged , Survival Rate , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...