Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Nat Commun ; 9(1): 1048, 2018 03 13.
Article in English | MEDLINE | ID: mdl-29535388

ABSTRACT

Pulmonary large-cell neuroendocrine carcinomas (LCNECs) have similarities with other lung cancers, but their precise relationship has remained unclear. Here we perform a comprehensive genomic (n = 60) and transcriptomic (n = 69) analysis of 75 LCNECs and identify two molecular subgroups: "type I LCNECs" with bi-allelic TP53 and STK11/KEAP1 alterations (37%), and "type II LCNECs" enriched for bi-allelic inactivation of TP53 and RB1 (42%). Despite sharing genomic alterations with adenocarcinomas and squamous cell carcinomas, no transcriptional relationship was found; instead LCNECs form distinct transcriptional subgroups with closest similarity to SCLC. While type I LCNECs and SCLCs exhibit a neuroendocrine profile with ASCL1high/DLL3high/NOTCHlow, type II LCNECs bear TP53 and RB1 alterations and differ from most SCLC tumors with reduced neuroendocrine markers, a pattern of ASCL1low/DLL3low/NOTCHhigh, and an upregulation of immune-related pathways. In conclusion, LCNECs comprise two molecularly defined subgroups, and distinguishing them from SCLC may allow stratified targeted treatment of high-grade neuroendocrine lung tumors.


Subject(s)
Carcinoma, Neuroendocrine/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Neuroendocrine Tumors/genetics , Small Cell Lung Carcinoma/genetics , DNA Mutational Analysis , Genomics/methods , High-Throughput Nucleotide Sequencing , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , In Vitro Techniques , Lung Neoplasms/genetics
2.
Cell Rep ; 20(12): 2833-2845, 2017 Sep 19.
Article in English | MEDLINE | ID: mdl-28930680

ABSTRACT

Kinase inhibitors represent the backbone of targeted cancer therapy, yet only a limited number of oncogenic drivers are directly druggable. By interrogating the activity of 1,505 kinase inhibitors, we found that BRD4-NUT-rearranged NUT midline carcinoma (NMC) cells are specifically killed by CDK9 inhibition (CDK9i) and depend on CDK9 and Cyclin-T1 expression. We show that CDK9i leads to robust induction of apoptosis and of markers of DNA damage response in NMC cells. While both CDK9i and bromodomain inhibition over time result in reduced Myc protein expression, only bromodomain inhibition induces cell differentiation and a p21-induced cell-cycle arrest in these cells. Finally, RNA-seq and ChIP-based analyses reveal a BRD4-NUT-specific CDK9i-induced perturbation of transcriptional elongation. Thus, our data provide a mechanistic basis for the genotype-dependent vulnerability of NMC cells to CDK9i that may be of relevance for the development of targeted therapies for NMC patients.


Subject(s)
Molecular Targeted Therapy , Neoplasms/enzymology , Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Cell Cycle Proteins , Cell Line, Tumor , Cyclin T/metabolism , Cyclin-Dependent Kinase 9/antagonists & inhibitors , Cyclin-Dependent Kinase 9/metabolism , HEK293 Cells , High-Throughput Screening Assays , Humans , Neoplasms/genetics , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Protein Kinase Inhibitors/chemistry , RNA Polymerase II/metabolism , Transcription Elongation, Genetic/drug effects , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Transcription, Genetic/drug effects
3.
Clin Cancer Res ; 23(18): 5527-5536, 2017 Sep 15.
Article in English | MEDLINE | ID: mdl-28630215

ABSTRACT

Purpose: The 8p12-p11 locus is frequently amplified in squamous cell lung cancer (SQLC); the receptor tyrosine kinase fibroblast growth factor receptor 1 (FGFR1) being one of the most prominent targets of this amplification. Thus, small molecules inhibiting FGFRs have been employed to treat FGFR1-amplified SQLC. However, only about 11% of such FGFR1-amplified tumors respond to single-agent FGFR inhibition and several tumors exhibited insufficient tumor shrinkage, compatible with the existence of drug-resistant tumor cells.Experimental Design: To investigate possible mechanisms of resistance to FGFR inhibition, we studied the lung cancer cell lines DMS114 and H1581. Both cell lines are highly sensitive to three different FGFR inhibitors, but exhibit sustained residual cellular viability under treatment, indicating a subpopulation of existing drug-resistant cells. We isolated these subpopulations by treating the cells with constant high doses of FGFR inhibitors.Results: The FGFR inhibitor-resistant cells were cross-resistant and characterized by sustained MAPK pathway activation. In drug-resistant H1581 cells, we identified NRAS amplification and DUSP6 deletion, leading to MAPK pathway reactivation. Furthermore, we detected subclonal NRAS amplifications in 3 of 20 (15%) primary human FGFR1-amplified SQLC specimens. In contrast, drug-resistant DMS114 cells exhibited transcriptional upregulation of MET that drove MAPK pathway reactivation. As a consequence, we demonstrate that rational combination therapies resensitize resistant cells to treatment with FGFR inhibitors.Conclusions: We provide evidence for the existence of diverse mechanisms of primary drug resistance in FGFR1-amplified lung cancer and provide a rational strategy to improve FGFR inhibitor therapies by combination treatment. Clin Cancer Res; 23(18); 5527-36. ©2017 AACR.


Subject(s)
Drug Resistance, Neoplasm/genetics , Gene Amplification , Lung Neoplasms/genetics , Receptor, Fibroblast Growth Factor, Type 1/genetics , Animals , Antineoplastic Agents/pharmacology , Biomarkers, Tumor , Cell Line, Tumor , Disease Models, Animal , GTP Phosphohydrolases/genetics , Humans , Lung Neoplasms/diagnosis , Lung Neoplasms/drug therapy , Membrane Proteins/genetics , Mice , Protein Binding , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Tomography, X-Ray Computed , Translocation, Genetic , Xenograft Model Antitumor Assays
4.
Oncoimmunology ; 5(5): e1131379, 2016 May.
Article in English | MEDLINE | ID: mdl-27467949

ABSTRACT

Inhibition of the PD-1/PD-L1 pathway may induce anticancer immune responses in non-small cell lung cancer (NSCLC). Two PD-L1 immunohistochemistry (IHC) assays have been approved as companion diagnostic tests for therapeutic anti-PD-1 antibodies. However, many aspects of PD-L1 prevalence and association with genetically defined subtypes have not been addressed systematically. Here, we analyzed PD-L1 expression in 436 genetically annotated NSCLC specimens enriched for early stages using PD-L1 antibody 5H1. Expression of PD-L1 was detected in the tumor cells (TC) (34% of cases) and in associated immune cells (IC) (49%) across all stages of NSCLC, either alone or in combination. PD-L1 IHC-positive TC, but not IC showed significantly higher PD-L1 RNA expression levels. Expression in TC was associated with TP53, KRAS and STK11 mutational status in adenocarcinomas (AD) and with NFE2L2 mutations in squamous cell carcinomas (SQ). No correlations with histological subtype, clinical characteristics and overall survival were found. The presence of PD-L1-positive IC was significantly associated with patients' smoking status in AD. The findings are in agreement with the emerging concept that tumors with high mutational burden are more likely to benefit from immunotherapy, since TP53 and KRAS mutations are linked to smoking, increased numbers of somatic mutations and expression of neoantigens. Current clinical studies focus on stage IIIB and IV NSCLC; however, PD-L1 expression occurs in earlier stages and might be a predictive biomarker in clinical trials testing (neo-) adjuvant strategies.

5.
Clin Cancer Res ; 22(19): 4837-4847, 2016 Oct 01.
Article in English | MEDLINE | ID: mdl-27252416

ABSTRACT

PURPOSE: To identify novel mechanisms of resistance to third-generation EGFR inhibitors in patients with lung adenocarcinoma that progressed under therapy with either AZD9291 or rociletinib (CO-1686). EXPERIMENTAL DESIGN: We analyzed tumor biopsies from seven patients obtained before, during, and/or after treatment with AZD9291 or rociletinib (CO-1686). Targeted sequencing and FISH analyses were performed, and the relevance of candidate genes was functionally assessed in in vitro models. RESULTS: We found recurrent amplification of either MET or ERBB2 in tumors that were resistant or developed resistance to third-generation EGFR inhibitors and show that ERBB2 and MET activation can confer resistance to these compounds. Furthermore, we identified a KRASG12S mutation in a patient with acquired resistance to AZD9291 as a potential driver of acquired resistance. Finally, we show that dual inhibition of EGFR/MEK might be a viable strategy to overcome resistance in EGFR-mutant cells expressing mutant KRAS CONCLUSIONS: Our data suggest that heterogeneous mechanisms of resistance can drive primary and acquired resistance to third-generation EGFR inhibitors and provide a rationale for potential combination strategies. Clin Cancer Res; 22(19); 4837-47. ©2016 AACR.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm/genetics , Lung Neoplasms/drug therapy , Acrylamides/therapeutic use , Adenocarcinoma of Lung , Aged , Aniline Compounds/therapeutic use , ErbB Receptors/antagonists & inhibitors , Female , Humans , Male , Middle Aged , Pyrimidines/therapeutic use
6.
Genome Biol ; 16: 7, 2015 Jan 05.
Article in English | MEDLINE | ID: mdl-25650807

ABSTRACT

Genomic translocation events frequently underlie cancer development through generation of gene fusions with oncogenic properties. Identification of such fusion transcripts by transcriptome sequencing might help to discover new potential therapeutic targets. We developed TRUP (Tumor-specimen suited RNA-seq Unified Pipeline) (https://github.com/ruping/TRUP), a computational approach that combines split-read and read-pair analysis with de novo assembly for the identification of chimeric transcripts in cancer specimens. We apply TRUP to RNA-seq data of different tumor types, and find it to be more sensitive than alternative tools in detecting chimeric transcripts, such as secondary rearrangements in EML4-ALK-positive lung tumors, or recurrent inactivating rearrangements affecting RASSF8.


Subject(s)
Chromosome Breakpoints , Computational Biology , High-Throughput Nucleotide Sequencing , Oncogene Fusion , Transcriptome , Translocation, Genetic , Base Sequence , Cell Line, Tumor , Cluster Analysis , Computational Biology/methods , Gene Silencing , Genomics , Humans , In Situ Hybridization, Fluorescence , Lung Neoplasms/genetics , Oncogene Proteins, Fusion/genetics , Tumor Suppressor Proteins/genetics
7.
Nat Med ; 20(9): 1027-34, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25173427

ABSTRACT

Crizotinib, a selective tyrosine kinase inhibitor (TKI), shows marked activity in patients whose lung cancers harbor fusions in the gene encoding anaplastic lymphoma receptor tyrosine kinase (ALK), but its efficacy is limited by variable primary responses and acquired resistance. In work arising from the clinical observation of a patient with ALK fusion-positive lung cancer who had an exceptional response to an insulin-like growth factor 1 receptor (IGF-1R)-specific antibody, we define a therapeutic synergism between ALK and IGF-1R inhibitors. Similar to IGF-1R, ALK fusion proteins bind to the adaptor insulin receptor substrate 1 (IRS-1), and IRS-1 knockdown enhances the antitumor effects of ALK inhibitors. In models of ALK TKI resistance, the IGF-1R pathway is activated, and combined ALK and IGF-1R inhibition improves therapeutic efficacy. Consistent with this finding, the levels of IGF-1R and IRS-1 are increased in biopsy samples from patients progressing on crizotinib monotherapy. Collectively these data support a role for the IGF-1R-IRS-1 pathway in both ALK TKI-sensitive and ALK TKI-resistant states and provide a biological rationale for further clinical development of dual ALK and IGF-1R inhibitors.


Subject(s)
Lung Neoplasms/drug therapy , Pyrazoles/therapeutic use , Pyridines/therapeutic use , Receptor Protein-Tyrosine Kinases/drug effects , Receptor, IGF Type 1/drug effects , Anaplastic Lymphoma Kinase , Crizotinib , Female , Gene Knockdown Techniques , Humans , Insulin Receptor Substrate Proteins/genetics , Lung Neoplasms/enzymology , Lung Neoplasms/metabolism , Middle Aged , Pyrazoles/pharmacology , Pyridines/pharmacology , Receptor, IGF Type 1/metabolism , Up-Regulation
8.
Nat Commun ; 5: 3518, 2014 Mar 27.
Article in English | MEDLINE | ID: mdl-24670920

ABSTRACT

Pulmonary carcinoids are rare neuroendocrine tumours of the lung. The molecular alterations underlying the pathogenesis of these tumours have not been systematically studied so far. Here we perform gene copy number analysis (n=54), genome/exome (n=44) and transcriptome (n=69) sequencing of pulmonary carcinoids and observe frequent mutations in chromatin-remodelling genes. Covalent histone modifiers and subunits of the SWI/SNF complex are mutated in 40 and 22.2% of the cases, respectively, with MEN1, PSIP1 and ARID1A being recurrently affected. In contrast to small-cell lung cancer and large-cell neuroendocrine lung tumours, TP53 and RB1 mutations are rare events, suggesting that pulmonary carcinoids are not early progenitor lesions of the highly aggressive lung neuroendocrine tumours but arise through independent cellular mechanisms. These data also suggest that inactivation of chromatin-remodelling genes is sufficient to drive transformation in pulmonary carcinoids.


Subject(s)
Carcinoid Tumor/genetics , Chromatin Assembly and Disassembly/genetics , Lung Neoplasms/genetics , Mutation , Adolescent , Adult , Aged , Base Sequence , Carcinoid Tumor/pathology , Chromosome Mapping , DNA Copy Number Variations , Exome/genetics , Female , Gene Dosage , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genome, Human/genetics , Humans , Lung Neoplasms/pathology , Male , Middle Aged , Molecular Sequence Data , Polymorphism, Single Nucleotide , Sequence Analysis, DNA , Young Adult
9.
Cancer Discov ; 4(4): 415-22, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24469108

ABSTRACT

UNLABELLED: We discovered a novel somatic gene fusion, CD74-NRG1, by transcriptome sequencing of 25 lung adenocarcinomas of never smokers. By screening 102 lung adenocarcinomas negative for known oncogenic alterations, we found four additional fusion-positive tumors, all of which were of the invasive mucinous subtype. Mechanistically, CD74-NRG1 leads to extracellular expression of the EGF-like domain of NRG1 III-ß3, thereby providing the ligand for ERBB2-ERBB3 receptor complexes. Accordingly, ERBB2 and ERBB3 expression was high in the index case, and expression of phospho-ERBB3 was specifically found in tumors bearing the fusion (P < 0.0001). Ectopic expression of CD74-NRG1 in lung cancer cell lines expressing ERBB2 and ERBB3 activated ERBB3 and the PI3K-AKT pathway, and led to increased colony formation in soft agar. Thus, CD74-NRG1 gene fusions are activating genomic alterations in invasive mucinous adenocarcinomas and may offer a therapeutic opportunity for a lung tumor subtype with, so far, no effective treatment. SIGNIFICANCE: CD74­NRG1 fusions may represent a therapeutic opportunity for invasive mucinous lung adenocarcinomas, a tumor with no effective treatment that frequently presents with multifocal unresectable disease.


Subject(s)
Adenocarcinoma, Mucinous/genetics , Adenocarcinoma/genetics , Antigens, Differentiation, B-Lymphocyte/genetics , Histocompatibility Antigens Class II/genetics , Lung Neoplasms/genetics , Neuregulin-1/genetics , Oncogene Proteins, Fusion/genetics , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Adenocarcinoma, Mucinous/pathology , Adult , Aged , Aged, 80 and over , Animals , Base Sequence , Cell Line, Tumor , Female , Gene Expression Profiling , Humans , Lung Neoplasms/pathology , Male , Mice , Middle Aged , Molecular Sequence Data , NIH 3T3 Cells , Oncogene Proteins, Fusion/metabolism , Sequence Analysis, DNA , Signal Transduction/genetics
10.
Eur J Haematol ; 92(6): 537-40, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24354760

ABSTRACT

Isolated myeloid sarcoma is a rare presentation of acute myeloid leukemia. There are limited data available concerning the prognostic relevance and the right treatment strategy for this clinical scenario. Here, we report a case of acute myeloid leukemia with extensive lesions and fractures in multiple bones in a 64-yr-old male patient. Remarkably, treatment with a high-dose cytarabine regimen led to rapid remineralization of all bone lesions and recovery of the patient's mobility within a few weeks. Thereby, surgical treatment and radiotherapy could be avoided, supporting the role of intensive induction and standard consolidation chemotherapy as first-line treatment for myeloid sarcoma.


Subject(s)
Antimetabolites, Antineoplastic/administration & dosage , Bone Diseases/etiology , Calcinosis/etiology , Cytarabine/administration & dosage , Sarcoma, Myeloid/complications , Sarcoma, Myeloid/drug therapy , Antimetabolites, Antineoplastic/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biopsy , Bone Diseases/diagnosis , Bone Marrow/pathology , Calcinosis/diagnosis , Cytarabine/adverse effects , Humans , Immunophenotyping , Male , Middle Aged , Sarcoma, Myeloid/diagnosis , Tomography, X-Ray Computed , Treatment Outcome
11.
Nat Genet ; 44(10): 1104-10, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22941188

ABSTRACT

Small-cell lung cancer (SCLC) is an aggressive lung tumor subtype with poor prognosis. We sequenced 29 SCLC exomes, 2 genomes and 15 transcriptomes and found an extremely high mutation rate of 7.4±1 protein-changing mutations per million base pairs. Therefore, we conducted integrated analyses of the various data sets to identify pathogenetically relevant mutated genes. In all cases, we found evidence for inactivation of TP53 and RB1 and identified recurrent mutations in the CREBBP, EP300 and MLL genes that encode histone modifiers. Furthermore, we observed mutations in PTEN, SLIT2 and EPHA7, as well as focal amplifications of the FGFR1 tyrosine kinase gene. Finally, we detected many of the alterations found in humans in SCLC tumors from Tp53 and Rb1 double knockout mice. Our study implicates histone modification as a major feature of SCLC, reveals potentially therapeutically tractable genomic alterations and provides a generalizable framework for the identification of biologically relevant genes in the context of high mutational background.


Subject(s)
Genome, Human , Lung Neoplasms/genetics , Small Cell Lung Carcinoma/genetics , Amino Acid Substitution , Animals , CREB-Binding Protein/genetics , Cell Line, Tumor , DNA Copy Number Variations , DNA Mutational Analysis , E1A-Associated p300 Protein/genetics , Gene Expression Profiling , Gene Regulatory Networks , Genome-Wide Association Study , Histone-Lysine N-Methyltransferase , Humans , Intercellular Signaling Peptides and Proteins/genetics , Mice , Mice, Knockout , Models, Molecular , Mutation , Myeloid-Lymphoid Leukemia Protein/genetics , Nerve Tissue Proteins/genetics , Oligonucleotide Array Sequence Analysis , PTEN Phosphohydrolase/genetics , Polymorphism, Single Nucleotide , Protein Processing, Post-Translational/genetics , Retinoblastoma Protein/genetics , Tumor Suppressor Protein p53/genetics
12.
Cell ; 150(6): 1107-20, 2012 Sep 14.
Article in English | MEDLINE | ID: mdl-22980975

ABSTRACT

Lung adenocarcinoma, the most common subtype of non-small cell lung cancer, is responsible for more than 500,000 deaths per year worldwide. Here, we report exome and genome sequences of 183 lung adenocarcinoma tumor/normal DNA pairs. These analyses revealed a mean exonic somatic mutation rate of 12.0 events/megabase and identified the majority of genes previously reported as significantly mutated in lung adenocarcinoma. In addition, we identified statistically recurrent somatic mutations in the splicing factor gene U2AF1 and truncating mutations affecting RBM10 and ARID1A. Analysis of nucleotide context-specific mutation signatures grouped the sample set into distinct clusters that correlated with smoking history and alterations of reported lung adenocarcinoma genes. Whole-genome sequence analysis revealed frequent structural rearrangements, including in-frame exonic alterations within EGFR and SIK2 kinases. The candidate genes identified in this study are attractive targets for biological characterization and therapeutic targeting of lung adenocarcinoma.


Subject(s)
Adenocarcinoma/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Genes, Neoplasm , High-Throughput Nucleotide Sequencing , Lung Neoplasms/genetics , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Adult , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/pathology , Cohort Studies , Exome , Female , Genome-Wide Association Study , Humans , Lung Neoplasms/pathology , Male , Middle Aged , Mutation , Mutation Rate
13.
Mod Pathol ; 25(11): 1473-80, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22684217

ABSTRACT

We recently reported fibroblast growth factor receptor-type 1 (FGFR1) amplification to be associated with therapeutically tractable FGFR1 dependency in squamous cell lung cancer. This makes FGFR1 a novel target for directed therapy in these tumors. To reproducibly identify patients for clinical studies, we developed a standardized reading and evaluation strategy for FGFR1 fluorescence in-situ hybridization (FISH) and propose evaluation criteria, describe different patterns of low- and high-level amplifications and report on the prevalence of FGFR1 amplifications in pulmonary carcinomas. A total of 420 lung cancer patients including 307 squamous carcinomas, 100 adenocarcinomas of the lung and 13 carcinomas of other types were analyzed for FGFR1 amplification using a dual color FISH. We found heterogeneous and different patterns of gene copy numbers. FGFR1 amplifications were observed in 20% of pulmonary squamous carcinomas but not in adenocarcinomas. High-level amplification (as defined by an FGFR1/centromer 8 (CEN8) ratio ≥2.0, or average number of FGFR1 signals per tumor cell nucleus ≥6, or the percentage of tumor cells containing ≥15 FGFR1 signals or large clusters ≥10%) was detected at a frequency of 16% and low-level amplification (as defined by ≥5 FGFR1 signals in ≥50% of tumor cells) at a frequency of 4%. We conclude that FGFR1 amplification is one of the most frequent therapeutically tractable genetic lesions in pulmonary carcinomas. Standardized reporting of FGFR1 amplification in squamous carcinomas of the lung will become increasingly important to correlate therapeutic responses with FGFR1 inhibitors in clinical studies. Thus, our reading and evaluation strategy might serve as a basis for identifying patients for ongoing and upcoming clinical trials.


Subject(s)
Adenocarcinoma/genetics , Carcinoma, Squamous Cell/genetics , Gene Amplification , In Situ Hybridization, Fluorescence , Lung Neoplasms/genetics , Receptor, Fibroblast Growth Factor, Type 1/genetics , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Carcinoma, Squamous Cell/pathology , Fixatives , Formaldehyde , Gene Dosage , Genetic Predisposition to Disease , Germany , Humans , Lung Neoplasms/pathology , Paraffin Embedding , Phenotype , Predictive Value of Tests , Reproducibility of Results , Tissue Fixation
14.
PLoS One ; 7(1): e30229, 2012.
Article in English | MEDLINE | ID: mdl-22279573

ABSTRACT

BACKGROUND: Using in vivo mouse models, the mechanisms of CD4+ T cell help have been intensively investigated. However, a mechanistic analysis of human CD4+ T cell help is largely lacking. Our goal was to elucidate the mechanisms of human CD4+ T cell help of CD8+ T cell proliferation using a novel in vitro model. METHODS/PRINCIPAL FINDINGS: We developed a genetically engineered novel human cell-based artificial APC, aAPC/mOKT3, which expresses a membranous form of the anti-CD3 monoclonal antibody OKT3 as well as other immune accessory molecules. Without requiring the addition of allogeneic feeder cells, aAPC/mOKT3 enabled the expansion of both peripheral and tumor-infiltrating T cells, regardless of HLA-restriction. Stimulation with aAPC/mOKT3 did not expand Foxp3+ regulatory T cells, and expanded tumor infiltrating lymphocytes predominantly secreted Th1-type cytokines, interferon-γ and IL-2. In this aAPC-based system, the presence of autologous CD4+ T cells was associated with significantly improved CD8+ T cell expansion in vitro. The CD4+ T cell derived cytokines IL-2 and IL-21 were necessary but not sufficient for this effect. However, CD4+ T cell help of CD8+ T cell proliferation was partially recapitulated by both adding IL-2/IL-21 and by upregulation of IL-21 receptor on CD8+ T cells. CONCLUSIONS: We have developed an in vitro model that advances our understanding of the immunobiology of human CD4+ T cell help of CD8+ T cells. Our data suggests that human CD4+ T cell help can be leveraged to expand CD8+ T cells in vitro.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , CD3 Complex/immunology , CD3 Complex/metabolism , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Coculture Techniques , Cytokines/immunology , Cytokines/metabolism , Flow Cytometry , Humans , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-2/immunology , Interleukin-2/metabolism , Interleukin-2/pharmacology , Interleukins/immunology , Interleukins/metabolism , Interleukins/pharmacology , K562 Cells , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Muromonab-CD3/immunology , Muromonab-CD3/metabolism , Muromonab-CD3/pharmacology , Receptors, Interleukin-21/immunology , Receptors, Interleukin-21/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Up-Regulation/drug effects
15.
J Immunol ; 188(4): 1609-19, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22238455

ABSTRACT

Although both MHC class II/CD8α double-knockout and CD8ß null mice show a defect in the development of MHC class I-restricted CD8(+) T cells in the thymus, they possess low numbers of high-avidity peripheral CTL with limited clonality and are able to contain acute and chronic infections. These in vivo data suggest that the CD8 coreceptor is not absolutely necessary for the generation of Ag-specific CTL. Lack of CD8 association causes partial TCR signaling because of the absence of CD8/Lck recruitment to the proximity of the MHC/TCR complex, resulting in suboptimal MAPK activation. Therefore, there should exist a signaling mechanism that can supplement partial TCR activation caused by the lack of CD8 association. In this human study, we have shown that CD8-independent stimulation of Ag-specific CTL previously primed in the presence of CD8 coligation, either in vivo or in vitro, induced severely impaired in vitro proliferation. When naive CD8(+) T cells were primed in the absence of CD8 binding and subsequently restimulated in the presence of CD8 coligation, the proliferation of Ag-specific CTL was also severely hampered. However, when CD8-independent T cell priming and restimulation were supplemented with IL-21, Ag-specific CD8(+) CTL expanded in two of six individuals tested. We found that IL-21 rescued partial MAPK activation in a STAT3- but not STAT1-dependent manner. These results suggest that CD8 coligation is critical for the expansion of postthymic peripheral Ag-specific CTL in humans. However, STAT3-mediated IL-21 signaling can supplement partial TCR signaling caused by the lack of CD8 association.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Interleukins/metabolism , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Mitogen-Activated Protein Kinases/metabolism , STAT3 Transcription Factor/metabolism , T-Lymphocytes, Cytotoxic/immunology , CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Humans , Interleukins/immunology , Lymphocyte Activation , Major Histocompatibility Complex , Signal Transduction , T-Lymphocytes, Cytotoxic/metabolism
16.
Clin Cancer Res ; 17(16): 5392-401, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21705450

ABSTRACT

PURPOSE: In previous cancer vaccine clinical trials targeting survivin, induction of specific CD8(+) T-cell responses did not consistently lead to clinical responses. Considering the critical role of CD4(+) T-cell help in generating antitumor immunity, integration of anti-survivin CD4(+) T-cell responses may enhance the efficacy of anti-survivin cancer immunotherapy. Human leukocyte antigen (HLA)-DP4 is emerging as an attractive MHC target allele of CD4(+) T cell-mediated immunotherapy, because it is one of the most frequent HLA alleles in many ethnic groups. In this article, we aimed to elucidate DP4-restricted CD4(+) T-cell responses against survivin in cancer patients. EXPERIMENTAL DESIGN: We generated a human cell-based artificial antigen-presenting cell (aAPC) expressing HLA-DP4, CD80, and CD83 and induced DP4-restricted antigen-specific CD4(+) T cells. The number, phenotype, effector function, and in vitro longevity of generated CD4(+) T cells were determined. RESULTS: We first determined previously unknown DP4-restricted CD4(+) T-cell epitopes derived from cytomegalovirus pp65, to which sustained Th1-biased recall responses were induced in vitro by using DP4-aAPC. In contrast, DP4-aAPC induced in vitro both Th1 and Th2 long-lived anti-survivin CD4(+) T cells from cancer patients. Both survivin-specific Th1 and Th2 cells were able to recognize survivin-expressing tumors in a DP4-restricted manner. Neither survivin-specific interleukin 10 secreting Tr1 cells nor Th17 cells were induced by DP4-aAPC. CONCLUSIONS: DP4-restricted anti-survivin Th1 and Th2 immunity with sufficient functional avidity can be induced from cancer patients. The development of strategies to concurrently induce both CD4(+) and CD8(+) T-cell responses against survivin is warranted for optimal anti-survivin cancer immunotherapy.


Subject(s)
Antigen-Presenting Cells/immunology , HLA-DP beta-Chains/immunology , Inhibitor of Apoptosis Proteins/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Alleles , Amino Acid Sequence , Antigen-Presenting Cells/metabolism , Antigens, CD/immunology , Antigens, CD/metabolism , B7-1 Antigen/immunology , B7-1 Antigen/metabolism , Blotting, Western , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/metabolism , Flow Cytometry , HLA-DP beta-Chains/genetics , HLA-DP beta-Chains/metabolism , Humans , Immunoglobulins/immunology , Immunoglobulins/metabolism , Immunotherapy/methods , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-4/immunology , Interleukin-4/metabolism , K562 Cells , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Molecular Sequence Data , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/therapy , Phosphoproteins/immunology , Survivin , Th1 Cells/metabolism , Th2 Cells/metabolism , Viral Matrix Proteins/immunology , CD83 Antigen
17.
Clin Cancer Res ; 17(10): 3360-7, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21558400

ABSTRACT

PURPOSE: Blood-based surrogate markers would be attractive biomarkers for early detection, diagnosis, prognosis, and prediction of therapeutic outcome in cancer. Disease-associated gene expression signatures in peripheral blood mononuclear cells (PBMC) have been described for several cancer types. However, RNA-stabilized whole blood-based technologies would be clinically more applicable and robust. We evaluated the applicability of whole blood-based gene expression profiling for the detection of non-small cell lung cancer (NSCLC). EXPERIMENTAL DESIGN: Expression profiles were generated from PAXgene-stabilized blood samples from three independent groups consisting of NSCLC cases and controls (n = 77, 54, and 102), using the Illumina WG6-VS2 system. RESULTS: Several genes are consistently differentially expressed in whole blood of NSCLC patients and controls. These expression profiles were used to build a diagnostic classifier for NSCLC, which was validated in an independent validation set of NSCLC patients (stages I-IV) and hospital-based controls. The area under the receiver operator curve was calculated to be 0.824 (P < 0.001). In a further independent dataset of stage I NSCLC patients and healthy controls the AUC was 0.977 (P < 0.001). Specificity of the classifier was validated by permutation analysis in both validation cohorts. Genes within the classifier are enriched in immune-associated genes and show specificity for NSCLC. CONCLUSIONS: Our results show that gene expression profiles of whole blood allow for detection of manifest NSCLC. These results prompt further development of gene expression-based biomarker tests in peripheral blood for the diagnosis and early detection of NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Gene Expression Profiling , Leukocytes, Mononuclear/metabolism , Lung Neoplasms/genetics , Adult , Aged , Algorithms , Carcinoma, Non-Small-Cell Lung/blood , Carcinoma, Non-Small-Cell Lung/classification , Case-Control Studies , Female , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/blood , Lung Neoplasms/classification , Male , Middle Aged , Oligonucleotide Array Sequence Analysis
18.
PLoS One ; 6(5): e19601, 2011 May 05.
Article in English | MEDLINE | ID: mdl-21573178

ABSTRACT

Treatment of EGFR-mutant non-small cell lung cancer patients with the tyrosine kinase inhibitors erlotinib or gefitinib results in high response rates and prolonged progression-free survival. Despite the development of sensitive mutation detection approaches, a thorough validation of these in a clinical setting has so far been lacking. We performed, in a clinical setting, a systematic validation of dideoxy 'Sanger' sequencing and pyrosequencing against massively parallel sequencing as one of the most sensitive mutation detection technologies available. Mutational annotation of clinical lung tumor samples revealed that of all patients with a confirmed response to EGFR inhibition, only massively parallel sequencing detected all relevant mutations. By contrast, dideoxy sequencing missed four responders and pyrosequencing missed two responders, indicating a dramatic lack of sensitivity of dideoxy sequencing, which is widely applied for this purpose. Furthermore, precise quantification of mutant alleles revealed a low correlation (r(2) = 0.27) of histopathological estimates of tumor content and frequency of mutant alleles, thereby questioning the use of histopathology for stratification of specimens for individual analytical procedures. Our results suggest that enhanced analytical sensitivity is critically required to correctly identify patients responding to EGFR inhibition. More broadly, our results emphasize the need for thorough evaluation of all mutation detection approaches against massively parallel sequencing as a prerequisite for any clinical implementation.


Subject(s)
Benchmarking , DNA Mutational Analysis/methods , DNA Mutational Analysis/standards , Lung Neoplasms/genetics , Mutation/genetics , Adult , Aged , Base Sequence , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , ErbB Receptors/genetics , Exons/genetics , Female , Genome, Human/genetics , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Male , Middle Aged , Molecular Sequence Data , Treatment Outcome
19.
Cancer Discov ; 1(1): 78-89, 2011 Jun.
Article in English | MEDLINE | ID: mdl-22328973

ABSTRACT

UNLABELLED: While genomically targeted therapies have improved outcomes for patients with lung adenocarcinoma, little is known about the genomic alterations which drive squamous cell lung cancer. Sanger sequencing of the tyrosine kinome identified mutations in the DDR2 kinase gene in 3.8% of squamous cell lung cancers and cell lines. Squamous lung cancer cell lines harboring DDR2 mutations were selectively killed by knock-down of DDR2 by RNAi or by treatment with the multi-targeted kinase inhibitor dasatinib. Tumors established from a DDR2 mutant cell line were sensitive to dasatinib in xenograft models. Expression of mutated DDR2 led to cellular transformation which was blocked by dasatinib. A squamous cell lung cancer patient with a response to dasatinib and erlotinib treatment harbored a DDR2 kinase domain mutation. These data suggest that gain-of-function mutations in DDR2 are important oncogenic events and are amenable to therapy with dasatinib. As dasatinib is already approved for use, these findings could be rapidly translated into clinical trials. SIGNIFICANCE: DDR2 mutations are present in 4% of lung SCCs, and DDR2 mutations are associated with sensitivity to dasatinib. These findings provide a rationale for designing clinical trials with the FDA-approved drug dasatinib in patients with lung SCCs.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Mitogen/genetics , Animals , Carcinoma, Non-Small-Cell Lung/enzymology , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Dasatinib , Discoidin Domain Receptors , Erlotinib Hydrochloride , Humans , Lung Neoplasms/enzymology , Mice , Mice, Nude , Mutation , NIH 3T3 Cells , Phosphorylation/drug effects , Phosphorylation/genetics , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Quinazolines/pharmacology , Quinazolines/therapeutic use , Thiazoles/pharmacology , Thiazoles/therapeutic use , src-Family Kinases/genetics
20.
Sci Transl Med ; 2(62): 62ra93, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21160078

ABSTRACT

Lung cancer remains one of the leading causes of cancer-related death in developed countries. Although lung adenocarcinomas with EGFR mutations or EML4-ALK fusions respond to treatment by epidermal growth factor receptor (EGFR) and anaplastic lymphoma kinase (ALK) inhibition, respectively, squamous cell lung cancer currently lacks therapeutically exploitable genetic alterations. We conducted a systematic search in a set of 232 lung cancer specimens for genetic alterations that were therapeutically amenable and then performed high-resolution gene copy number analyses. We identified frequent and focal fibroblast growth factor receptor 1 (FGFR1) amplification in squamous cell lung cancer (n = 155), but not in other lung cancer subtypes, and, by fluorescence in situ hybridization, confirmed the presence of FGFR1 amplifications in an independent cohort of squamous cell lung cancer samples (22% of cases). Using cell-based screening with the FGFR inhibitor PD173074 in a large (n = 83) panel of lung cancer cell lines, we demonstrated that this compound inhibited growth and induced apoptosis specifically in those lung cancer cells carrying amplified FGFR1. We validated the FGFR1 dependence of FGFR1-amplified cell lines by FGFR1 knockdown and by ectopic expression of an FGFR1-resistant allele (FGFR1(V561M)), which rescued FGFR1-amplified cells from PD173074-mediated cytotoxicity. Finally, we showed that inhibition of FGFR1 with a small molecule led to significant tumor shrinkage in vivo. Thus, focal FGFR1 amplification is common in squamous cell lung cancer and associated with tumor growth and survival, suggesting that FGFR inhibitors may be a viable therapeutic option in this cohort of patients.


Subject(s)
Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma of Lung , Animals , Apoptosis/genetics , Apoptosis/physiology , Blotting, Western , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line , Enzyme Inhibitors/therapeutic use , Gene Expression Regulation, Neoplastic/genetics , Humans , Lung Neoplasms/drug therapy , Male , Mice , Mice, Nude , Pyrimidines/therapeutic use , RNA Interference , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 1/genetics , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL