Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Am J Respir Crit Care Med ; 197(3): 325-336, 2018 02 01.
Article in English | MEDLINE | ID: mdl-28934595

ABSTRACT

RATIONALE: Chronic obstructive pulmonary disease (COPD) and non-small cell lung cancer (NSCLC) are interrelated diseases with substantial mortality, and the pathogenesis of both involves aberrant immune functioning. OBJECTIVES: To profile immune cell composition and function in patients with NSCLC and describe the effects of COPD on lung and tumor microenvironments. METHODS: We profiled resected lung and tumor tissue using flow cytometry and T-cell receptor sequencing in patients with and without COPD from a prospective cohort of patients undergoing resection of NSCLC. A murine cigarette smoke exposure model was used to evaluate the effect on pulmonary immune populations. A separate retrospective cohort of patients who received immune checkpoint inhibitors (ICIs) was analyzed, and their survival was quantified. MEASUREMENTS AND MAIN RESULTS: We observed an increased number of IFN-γ-producing CD8+ and CD4+ (T-helper cell type 1 [Th1]) lymphocytes in the lungs of patients with COPD. In both humans and mice, increased Th17 content was seen with smoke exposure, but was not associated with the development or severity of COPD. COPD-affected lung tissue displayed increased Th1 differentiation that was recapitulated in the matching tumor sample. PD-1 (programmed cell death protein 1) expression was increased in tumors of patients with COPD, and the presence of COPD was associated with progression-free survival in patients treated with ICIs. CONCLUSIONS: In patients with COPD, Th1 cell populations were expanded in both lung and tumor microenvironments, and the presence of COPD was associated with longer progression-free intervals in patients treated with ICIs. This has implications for understanding the immune mediators of COPD and developing novel therapies for NSCLC.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Carcinoma, Non-Small-Cell Lung/immunology , Lung Neoplasms/immunology , Pulmonary Disease, Chronic Obstructive/immunology , Tumor Microenvironment/immunology , Aged , Carcinoma, Non-Small-Cell Lung/physiopathology , Carcinoma, Non-Small-Cell Lung/surgery , Cohort Studies , Female , Flow Cytometry , Humans , Immunosuppressive Agents/therapeutic use , Lung Neoplasms/physiopathology , Lung Neoplasms/surgery , Male , Middle Aged , Pneumonectomy/methods , Prospective Studies , Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Disease, Chronic Obstructive/physiopathology , Sensitivity and Specificity , Signal Transduction/immunology , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/immunology
2.
Nat Commun ; 8: 14381, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28146145

ABSTRACT

The response rate to immune checkpoint inhibitor therapy for non-small-cell lung cancer (NSCLC) is just 20%. To improve this figure, several early phase clinical trials combining novel immunotherapeutics with immune checkpoint blockade have been initiated. Unfortunately, these trials have been designed without a strong foundational knowledge of the immune landscape present in NSCLC. Here, we use a flow cytometry panel capable of measuring 51 immune cell populations to comprehensively identify the immune cell composition and function in NSCLC. The results show that the immune cell composition is fundamentally different in lung adenocarcinoma as compared with lung squamous cell carcinoma, and that neutrophils are the most prevalent immune cell type. Using T-cell receptor-ß sequencing and tumour reactivity assays, we predict that tumour reactive T cells are frequently present in NSCLC. These results should help to guide the design of clinical trials and the direction of future research in this area.


Subject(s)
Carcinoma, Non-Small-Cell Lung/immunology , Immune System/immunology , Lung Neoplasms/immunology , Neutrophils/immunology , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Adenocarcinoma/therapy , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/therapy , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/therapy , Cell Count , Flow Cytometry , Humans , Immune System/pathology , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Neutrophils/pathology , T-Lymphocytes/immunology , T-Lymphocytes/pathology
3.
J Immunol ; 197(11): 4493-4503, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27799309

ABSTRACT

Lung cancer, the leading cause of cancer-related deaths worldwide, is a heterogeneous disease comprising multiple histologic subtypes that harbor disparate mutational profiles. Immune-based therapies have shown initial promise in the treatment of lung cancer patients but are limited by low overall response rates. We sought to determine whether the host immune response to lung cancer is dictated, at least in part, by histologic and genetic differences, because such correlations would have important clinical ramifications. Using mouse models of lung cancer, we show that small cell lung cancer (SCLC) and lung adenocarcinoma (ADCA) exhibit unique immune cell composition of the tumor microenvironment. The total leukocyte content was markedly reduced in SCLC compared with lung ADCA, which was validated in human lung cancer specimens. We further identified key differences in immune cell content using three models of lung ADCA driven by mutations in Kras, p53, and Egfr Although Egfr-mutant cancers displayed robust myeloid cell recruitment, they failed to mount a CD8+ immune response. In contrast, Kras-mutant tumors displayed significant expansion of multiple immune cell types, including CD8+ cells, regulatory T cells, IL-17A-producing lymphocytes, and myeloid cells. A human tissue microarray annotated for KRAS and EGFR mutations validated the finding of reduced CD8+ content in human lung ADCA. Taken together, these findings establish a strong foundational knowledge of the immune cell contexture of lung ADCA and SCLC and suggest that molecular and histological traits shape the host immune response to cancer.


Subject(s)
Adenocarcinoma/immunology , CD8-Positive T-Lymphocytes/immunology , Lung Neoplasms/immunology , Neoplasm Proteins/immunology , Small Cell Lung Carcinoma/immunology , T-Lymphocytes, Regulatory/immunology , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , CD8-Positive T-Lymphocytes/pathology , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Mice, Transgenic , Mutation , Neoplasm Proteins/genetics , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/pathology , T-Lymphocytes, Regulatory/pathology
4.
Proc Natl Acad Sci U S A ; 113(31): 8795-800, 2016 08 02.
Article in English | MEDLINE | ID: mdl-27439864

ABSTRACT

Insulin receptor substrate-1 (IRS-1) is a signaling adaptor protein that interfaces with many pathways activated in lung cancer. It has been assumed that IRS-1 promotes tumor growth through its ability to activate PI3K signaling downstream of the insulin-like growth factor receptor. Surprisingly, tumors with reduced IRS-1 staining in a human lung adenocarcinoma tissue microarray displayed a significant survival disadvantage, especially within the Kirsten rat sarcoma viral oncogene homolog (KRAS) mutant subgroup. Accordingly, adenoviral Cre recombinase (AdCre)-treated LSL-Kras/Irs-1(fl/fl) (Kras/Irs-1(-/-)) mice displayed increased tumor burden and mortality compared with controls. Mechanistically, IRS-1 deficiency promotes Janus kinase/signal transducers and activators of transcription (JAK/STAT) signaling via the IL-22 receptor, resulting in enhanced tumor-promoting inflammation. Treatment of Kras/Irs-1(+/+) and Kras/Irs-1(-/-) mice with JAK inhibitors significantly reduced tumor burden, most notably in the IRS-1-deficient group.


Subject(s)
Adenocarcinoma/metabolism , Insulin Receptor Substrate Proteins/metabolism , Lung Neoplasms/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , A549 Cells , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Animals , Cell Line, Tumor , Female , Humans , Insulin Receptor Substrate Proteins/deficiency , Insulin Receptor Substrate Proteins/genetics , Kaplan-Meier Estimate , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Mice, Knockout , Middle Aged , Mutation , Phenotype , Proto-Oncogene Proteins p21(ras)/genetics , Receptors, Interleukin/genetics , Receptors, Interleukin/metabolism , Signal Transduction/genetics
5.
Cell Rep ; 7(4): 1020-9, 2014 May 22.
Article in English | MEDLINE | ID: mdl-24794443

ABSTRACT

Epigenetic alterations, particularly in DNA methylation, are ubiquitous in cancer, yet the molecular origins and the consequences of these alterations are poorly understood. CTCF, a DNA-binding protein that regulates higher-order chromatin organization, is frequently altered by hemizygous deletion or mutation in human cancer. To date, a causal role for CTCF in cancer has not been established. Here, we show that Ctcf hemizygous knockout mice are markedly susceptible to spontaneous, radiation-, and chemically induced cancer in a broad range of tissues. Ctcf(+/-) tumors are characterized by increased aggressiveness, including invasion, metastatic dissemination, and mixed epithelial/mesenchymal differentiation. Molecular analysis of Ctcf(+/-) tumors indicates that Ctcf is haploinsufficient for tumor suppression. Tissues with hemizygous loss of CTCF exhibit increased variability in CpG methylation genome wide. These findings establish CTCF as a prominent tumor-suppressor gene and point to CTCF-mediated epigenetic stability as a major barrier to neoplastic progression.


Subject(s)
DNA Methylation , Genes, Tumor Suppressor , Neoplasms/genetics , Repressor Proteins/genetics , Animals , CCCTC-Binding Factor , Cell Line, Tumor , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Haploinsufficiency , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Neoplasms/metabolism , Protein Binding , Repressor Proteins/metabolism , Survival Analysis
6.
J Pathol ; 227(3): 298-305, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22430984

ABSTRACT

Hepatic haemangiosarcoma is a deadly malignancy whose aetiology remains poorly understood. Inactivation of the CDKN2A locus, which houses the ARF and p16(INK4a) tumour suppressor genes, is a common event in haemangiosarcoma patients, but the precise role of ARF in vascular tumourigenesis is unknown. To determine the extent to which ARF suppresses vascular neoplasia, we examined the incidence of hepatic vascular lesions in Arf-deficient mice exposed to the carcinogen urethane [intraperitoneal (i.p.), 1 mg/g]. Loss of Arf resulted in elevated morbidity and increased the incidence of both haemangiomas and incipient haemangiosarcomas. Suppression of vascular lesion development by ARF was heavily dependent on both Arf gene-dosage and the genetic strain of the mouse. Trp53-deficient mice also developed hepatic vascular lesions after exposure to urethane, suggesting that ARF signals through a p53-dependent pathway to inhibit the development of hepatic haemangiosarcoma. Our findings provide strong evidence that inactivation of Arf is a causative event in vascular neoplasia and suggest that the ARF pathway may be a novel molecular target for therapeutic intervention in haemangiosarcoma patients.


Subject(s)
Carcinogens , Cell Transformation, Neoplastic/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Hemangiosarcoma/prevention & control , Liver Neoplasms/prevention & control , Urethane , Animals , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Cyclin-Dependent Kinase Inhibitor p16/deficiency , Cyclin-Dependent Kinase Inhibitor p16/genetics , Gene Dosage , Genetic Predisposition to Disease , Hemangiosarcoma/chemically induced , Hemangiosarcoma/genetics , Hemangiosarcoma/metabolism , Hemangiosarcoma/pathology , Liver Neoplasms/chemically induced , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Signal Transduction , Time Factors , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
7.
Development ; 137(10): 1709-19, 2010 May.
Article in English | MEDLINE | ID: mdl-20430746

ABSTRACT

The metameric organization of the insect body plan is initiated with the activation of gap genes, a set of transcription-factor-encoding genes that are zygotically expressed in broad and partially overlapping domains along the anteroposterior (AP) axis of the early embryo. The spatial pattern of gap gene expression domains along the AP axis is generally conserved, but the maternal genes that regulate their expression are not. Building on the comprehensive knowledge of maternal gap gene activation in Drosophila, we used loss- and gain-of-function experiments in the hover fly Episyrphus balteatus (Syrphidae) to address the question of how the maternal regulation of gap genes evolved. We find that, in Episyrphus, a highly diverged bicoid ortholog is solely responsible for the AP polarity of the embryo. Episyrphus bicoid represses anterior zygotic expression of caudal and activates the anterior and central gap genes orthodenticle, hunchback and Krüppel. In bicoid-deficient Episyrphus embryos, nanos is insufficient to generate morphological asymmetry along the AP axis. Furthermore, we find that torso transiently regulates anterior repression of caudal and is required for the activation of orthodenticle, whereas all posterior gap gene domains of knirps, giant, hunchback, tailless and huckebein depend on caudal. We conclude that all maternal coordinate genes have altered their specific functions during the radiation of higher flies (Cyclorrhapha).


Subject(s)
Diptera/genetics , Gene Expression Regulation, Developmental , Genes, Insect , RNA, Messenger, Stored/physiology , Transcription Factors/genetics , Amino Acid Sequence , Animals , Animals, Genetically Modified , Body Patterning/genetics , Cell Polarity/genetics , Diptera/embryology , Embryo, Nonmammalian , Female , Genes, Insect/physiology , Molecular Sequence Data , Phylogeny , Sequence Homology , Transcription Factors/metabolism , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...