Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
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
2.
Oncogene ; 37(27): 3686-3697, 2018 07.
Article in English | MEDLINE | ID: mdl-29622795

ABSTRACT

The CD56 antigen (NCAM-1) is highly expressed on several malignancies with neuronal or neuroendocrine differentiation, including small-cell lung cancer and neuroblastoma, tumor types for which new therapeutic options are needed. We hypothesized that CD56-specific chimeric antigen receptor (CAR) T cells could target and eliminate CD56-positive malignancies. Sleeping Beauty transposon-generated CD56R-CAR T cells exhibited αßT-cell receptors, released antitumor cytokines upon co-culture with CD56+ tumor targets, demonstrated a lack of fratricide, and expression of cytolytic function in the presence of CD56+ stimulation. The CD56R-CAR+ T cells are capable of killing CD56+ neuroblastoma, glioma, and SCLC tumor cells in in vitro co-cultures and when tested against CD56+ human xenograft neuroblastoma models and SCLC models, CD56R-CAR+ T cells were able to inhibit tumor growth in vivo. These results indicate that CD56-CARs merit further investigation as a potential treatment for CD56+ malignancies.


Subject(s)
CD56 Antigen/metabolism , Glioma/therapy , Lung Neoplasms/therapy , Neuroblastoma/therapy , Receptors, Chimeric Antigen/metabolism , Small Cell Lung Carcinoma/therapy , T-Lymphocytes/immunology , T-Lymphocytes/transplantation , Animals , CD56 Antigen/genetics , Cell Line, Tumor , Glioma/pathology , Humans , Lung Neoplasms/pathology , Lymphocyte Activation/immunology , Mice , Mice, Inbred NOD , Mice, Nude , Neuroblastoma/pathology , Small Cell Lung Carcinoma/pathology , Transposases/metabolism , Xenograft Model Antitumor Assays
3.
PLoS Pathog ; 13(1): e1006087, 2017 01.
Article in English | MEDLINE | ID: mdl-28125732

ABSTRACT

HIV-1 infection is associated with an early and profound depletion of mucosal memory CD4+ T cells, a population that plays an indispensable role in the regulation of isotype switching and transepithelial transport of antibodies. In this study, we addressed whether the depletion of CD4+ T cell in HIV-1-infected individuals results in altered humoral responses specific to antigens encountered at mucosal surfaces. Comprehensive protein microarray of systemic humoral responses to intestinal microbiota demonstrated reduced IgG responses to antigens derived from Proteobacteria and Firmicutes but not Bacteroidetes. Importantly, intestinal secretions of antiretroviral therapy-treated HIV-1-infected individuals exhibited a significant elevation of IgM levels and decreased IgA/IgM and IgG/IgM ratios of antibodies specific to a variety of microbial and food antigens. The presented findings indicate reduced competence of mucosal B cells for class switch recombination from IgM to other isotypes limiting their capacity to react to changing antigenic variety in the gut lumen. Decreased availability of microbiota-specific IgA and IgG may be an important factor contributing to the translocation of microbial antigens across the intestinal mucosal barrier and their systemic dissemination that drives chronic inflammation in HIV-1-infected individuals.


Subject(s)
Antigens/immunology , HIV Infections/immunology , HIV-1/immunology , Microbiota/immunology , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Chronic Disease , Food , Gene Expression Regulation , HIV Infections/virology , Humans , Immunity, Humoral , Immunoglobulin A/immunology , Immunoglobulin Class Switching , Immunoglobulin G/immunology , Inflammation , Intestinal Mucosa/immunology , Intestinal Mucosa/virology
4.
Clin Cancer Res ; 22(14): 3630-42, 2016 07 15.
Article in English | MEDLINE | ID: mdl-26851185

ABSTRACT

PURPOSE: Promising results in the treatment of non-small cell lung cancer (NSCLC) have been seen with agents targeting immune checkpoints, such as programmed cell death 1 (PD-1) or programmed death ligand-1 (PD-L1). However, only a select group of patients respond to these interventions. The identification of biomarkers that predict clinical benefit to immune checkpoint blockade is critical to successful clinical translation of these agents. METHODS: We conducted an integrated analysis of three independent large datasets, including The Cancer Genome Atlas of lung adenocarcinoma and two datasets from MD Anderson Cancer Center (Houston, TX), Profiling of Resistance Patterns and Oncogenic Signaling Pathways in Evaluation of Cancers of the Thorax (named PROSPECT) and Biomarker-Integrated Approaches of Targeted Therapy for Lung Cancer Elimination (named BATTLE-1). Comprehensive analysis of mRNA gene expression, reverse-phase protein array, IHC, and correlation with clinical data were performed. RESULTS: Epithelial-mesenchymal transition (EMT) is highly associated with an inflammatory tumor microenvironment in lung adenocarcinoma, independent of tumor mutational burden. We found immune activation coexistent with elevation of multiple targetable immune checkpoint molecules, including PD-L1, PD-L2, PD-1, TIM-3, B7-H3, BTLA, and CTLA-4, along with increases in tumor infiltration by CD4(+)Foxp3(+) regulatory T cells in lung adenocarcinomas that displayed an EMT phenotype. Furthermore, we identify B7-H3 as a prognostic marker for NSCLC. CONCLUSIONS: The strong association between EMT status and an inflammatory tumor microenvironment with elevation of multiple targetable immune checkpoint molecules warrants further investigation of using EMT as a predictive biomarker for immune checkpoint blockade agents and other immunotherapies in NSCLC and possibly a broad range of other cancers. Clin Cancer Res; 22(14); 3630-42. ©2016 AACRSee related commentary by Datar and Schalper, p. 3422.


Subject(s)
Adenocarcinoma/pathology , Epithelial-Mesenchymal Transition/physiology , Inflammation/pathology , Lung Neoplasms/pathology , Tumor Microenvironment/physiology , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Adenocarcinoma of Lung , Antineoplastic Agents/therapeutic use , B7 Antigens/metabolism , Biomarkers, Tumor/metabolism , Epithelial-Mesenchymal Transition/drug effects , Humans , Immunotherapy/methods , Inflammation/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Mutation/drug effects , Signal Transduction/drug effects , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology , Tumor Microenvironment/drug effects
5.
Cancer Res ; 76(5): 999-1008, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26833127

ABSTRACT

STK11/LKB1 is among the most commonly inactivated tumor suppressors in non-small cell lung cancer (NSCLC), especially in tumors harboring KRAS mutations. Many oncogenes promote immune escape, undermining the effectiveness of immunotherapies, but it is unclear whether the inactivation of tumor suppressor genes, such as STK11/LKB1, exerts similar effects. In this study, we investigated the consequences of STK11/LKB1 loss on the immune microenvironment in a mouse model of KRAS-driven NSCLC. Genetic ablation of STK11/LKB1 resulted in accumulation of neutrophils with T-cell-suppressive effects, along with a corresponding increase in the expression of T-cell exhaustion markers and tumor-promoting cytokines. The number of tumor-infiltrating lymphocytes was also reduced in LKB1-deficient mouse and human tumors. Furthermore, STK11/LKB1-inactivating mutations were associated with reduced expression of PD-1 ligand PD-L1 in mouse and patient tumors as well as in tumor-derived cell lines. Consistent with these results, PD-1-targeting antibodies were ineffective against Lkb1-deficient tumors. In contrast, treating Lkb1-deficient mice with an IL6-neutralizing antibody or a neutrophil-depleting antibody yielded therapeutic benefits associated with reduced neutrophil accumulation and proinflammatory cytokine expression. Our findings illustrate how tumor suppressor mutations can modulate the immune milieu of the tumor microenvironment, and they offer specific implications for addressing STK11/LKB1-mutated tumors with PD-1-targeting antibody therapies.


Subject(s)
Cytokines/biosynthesis , Lung Neoplasms/immunology , Neutrophil Infiltration , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/physiology , T-Lymphocytes/immunology , Tumor Microenvironment , AMP-Activated Protein Kinase Kinases , AMP-Activated Protein Kinases , Animals , B7-H1 Antigen/analysis , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Mutation , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics
6.
J Mol Med (Berl) ; 92(1): 13-30, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24311119

ABSTRACT

For the most part, normal epithelial cells do not disseminate to other parts of the body and proliferate, as do metastatic cells. Presumably, a class of molecules-termed metastasis suppressors-are involved in this homeostatic control. Metastasis suppressors are, by definition, cellular factors that, when re-expressed in metastatic cells, functionally inhibit metastasis without significantly inhibiting tumor growth. In this brief review, we catalog known metastasis suppressors, what is known about their mechanism(s) of action, and experimental and clinical associations to date.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Tumor Suppressor Proteins/metabolism , Animals , Breast Neoplasms/genetics , Female , Humans , Neoplasm Metastasis , Signal Transduction , Tumor Suppressor Proteins/genetics
7.
Cancer Res ; 74(3): 954-63, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24351292

ABSTRACT

Cancer cells tend to utilize aerobic glycolysis even under normoxic conditions, commonly called the "Warburg effect." Aerobic glycolysis often directly correlates with malignancy, but its purpose, if any, in metastasis remains unclear. When wild-type KISS1 metastasis suppressor is expressed, aerobic glycolysis decreases and oxidative phosphorylation predominates. However, when KISS1 is missing the secretion signal peptide (ΔSS), invasion and metastasis are no longer suppressed and cells continue to metabolize using aerobic glycolysis. KISS1-expressing cells have 30% to 50% more mitochondrial mass than ΔSS-expressing cells, which are accompanied by correspondingly increased mitochondrial gene expression and higher expression of PGC1α, a master coactivator that regulates mitochondrial mass and metabolism. PGC1α-mediated downstream pathways (i.e., fatty acid synthesis and ß-oxidation) are differentially regulated by KISS1, apparently reliant upon direct KISS1 interaction with NRF1, a major transcription factor involved in mitochondrial biogenesis. Since the downstream effects could be reversed using short hairpin RNA to KISS1 or PGC1α, these data appear to directly connect changes in mitochondria mass, cellular glucose metabolism, and metastasis.


Subject(s)
Kisspeptins/genetics , Mitochondria/genetics , Mitochondria/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Animals , Cell Line, Tumor , Disease Models, Animal , Extracellular Space/metabolism , Female , Gene Expression , Glucose/metabolism , Glycolysis , Humans , Hydrogen-Ion Concentration , Kisspeptins/metabolism , Lactic Acid/metabolism , Mice , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasms/pathology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Transcription Factors/genetics , Transcription Factors/metabolism
8.
PLoS One ; 6(2): e16897, 2011 Feb 17.
Article in English | MEDLINE | ID: mdl-21379572

ABSTRACT

Harnessing the ability of cytotoxic T lymphocytes (CTLs) to recognize and eradicate tumor or pathogen-infected cells is a critical goal of modern immune-based therapies. Although multiple immunization strategies efficiently induce high levels of antigen-specific CTLs, the initial increase is typically followed by a rapid contraction phase resulting in a sharp decline in the frequency of functional CTLs. We describe a novel approach to immunotherapy based on a transplantation of low numbers of antigen-expressing hematopoietic stem cells (HSCs) following nonmyeloablative or partially myeloablative conditioning. Continuous antigen presentation by a limited number of differentiated transgenic hematopoietic cells results in an induction and prolonged maintenance of fully functional effector T cell responses in a mouse model. Recipient animals display high levels of antigen-specific CTLs four months following transplantation in contrast to dendritic cell-immunized animals in which the response typically declines at 4-6 weeks post-immunization. Majority of HSC-induced antigen-specific CD8+ T cells display central memory phenotype, efficiently kill target cells in vivo, and protect recipients against tumor growth in a preventive setting. Furthermore, we confirm previously published observation that high level engraftment of antigen-expressing HSCs following myeloablative conditioning results in tolerance and an absence of specific cytotoxic activity in vivo. In conclusion, the data presented here supports potential application of immunization by limited transplantation of antigen-expressing HSCs for the prevention and treatment of cancer and therapeutic immunization of chronic infectious diseases such as HIV-1/AIDS.


Subject(s)
Antigens/metabolism , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/metabolism , Immunologic Memory/genetics , T-Lymphocytes, Cytotoxic/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/transplantation , Cells, Cultured , Chickens/genetics , Communicable Diseases/immunology , Communicable Diseases/therapy , Genetic Therapy/methods , Hematopoietic Stem Cells/immunology , Immunologic Memory/physiology , Immunotherapy, Adoptive/methods , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms/immunology , Neoplasms/therapy , Ovalbumin/genetics , Ovalbumin/metabolism , T-Lymphocytes, Cytotoxic/metabolism , Time Factors
9.
J Immunol ; 177(9): 5956-67, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-17056520

ABSTRACT

The fate of dendritic cells (DCs) after Ag presentation may be DC subset-specific and controlled by many factors. The role of activation-induced apoptosis in regulating DC function is not clear. We investigated the fate of cutaneous DCs (cDCs), specifically Langerhans cells (LCs), and observed that they undergo apoptosis after successful Ag presentation to CD4 T cells. Caspase-specific inhibitors revealed that LC lines use a type II apoptosis pathway in response to CD4 T cells. In support of this, BH3-interacting domain (Bid) protein was present at high levels and specifically cleaved in the presence of Ag-specific T cells. Significant resistance to apoptosis by OT-2 CD4 cells was also observed for Bid knockout (KO) LCs in vitro. To test whether Bid was required to regulate LC function in vivo, we measured contact sensitization and topical immunization responses in Bid KO mice and observed markedly enhanced ear swelling and proliferation responses compared with wild-type mice. Furthermore, when Ag-pulsed Bid KO migratory cDCs were inoculated into wild-type recipients, an increase in both the rate and percentage of expanded OT-2 T cells expressing IFN-gamma was observed. Thus, enhanced Ag presentation function was intrinsic to Bid KO cDCs. Therefore, Bid is an important regulator of LC viability and Ag presentation function.


Subject(s)
Antigen Presentation , Apoptosis/immunology , BH3 Interacting Domain Death Agonist Protein/physiology , CD4-Positive T-Lymphocytes/immunology , Langerhans Cells/immunology , Animals , Antigen Presentation/genetics , Apoptosis/drug effects , BH3 Interacting Domain Death Agonist Protein/analysis , BH3 Interacting Domain Death Agonist Protein/genetics , CD4-Positive T-Lymphocytes/enzymology , CD4-Positive T-Lymphocytes/transplantation , Caspase Inhibitors , Cell Line , Cysteine Proteinase Inhibitors/pharmacology , Dendritic Cells/immunology , Langerhans Cells/chemistry , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Mice , Mice, Knockout , Skin/cytology , Skin/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...