Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Cancer Immunol Res ; 9(10): 1202-1213, 2021 10.
Article in English | MEDLINE | ID: mdl-34389558

ABSTRACT

Outcomes for patients with melanoma have improved over the past decade as a result of the development and FDA approval of immunotherapies targeting cytotoxic T lymphocyte antigen-4 (CTLA-4), programmed death-1 (PD-1), and programmed death ligand 1 (PD-L1). However, these therapies do not benefit all patients, and an area of intensive research investigation is identifying biomarkers that can predict which patients are most likely to benefit from them. Here, we report exploratory analyses of the associations of tumor mutational burden (TMB), a 4-gene inflammatory gene expression signature, and BRAF mutation status with tumor response, progression-free survival, and overall survival in patients with advanced melanoma treated as part of the CheckMate 066 and 067 phase III clinical trials evaluating immuno-oncology therapies. In patients enrolled in CheckMate 067 receiving the anti-PD-1 inhibitor nivolumab (NIVO) alone or in combination with the anti-CTLA-4 inhibitor ipilimumab (IPI) or IPI alone, longer survival appeared to associate with high (>median) versus low (≤median) TMB and with high versus low inflammatory signature scores. For NIVO-treated patients, the results regarding TMB association were confirmed in CheckMate 066. In addition, improved survival was observed with high TMB and absence of BRAF mutation. Weak correlations were observed between PD-L1, TMB, and the inflammatory signature. Combined assessment of TMB, inflammatory gene expression signature, and BRAF mutation status may be predictive for response to immune checkpoint blockade in advanced melanoma.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , B7-H1 Antigen/biosynthesis , Melanoma/drug therapy , Mutation , Skin Neoplasms/drug therapy , B7-H1 Antigen/immunology , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/immunology , Humans , Immunotherapy/methods , Ipilimumab/administration & dosage , Melanoma/genetics , Melanoma/immunology , Nivolumab/administration & dosage , Progression-Free Survival , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Treatment Outcome
2.
Cancer Cell ; 35(6): 885-900.e10, 2019 06 10.
Article in English | MEDLINE | ID: mdl-31185212

ABSTRACT

We investigated the role of chemokines in regulating T cell accumulation in solid tumors. CCL5 and CXCL9 overexpression was associated with CD8+ T cell infiltration in solid tumors. T cell infiltration required tumor cell-derived CCL5 and was amplified by IFN-γ-inducible, myeloid cell-secreted CXCL9. CCL5 and CXCL9 coexpression revealed immunoreactive tumors with prolonged survival and response to checkpoint blockade. Loss of CCL5 expression in human tumors was associated with epigenetic silencing through DNA methylation. Reduction of CCL5 expression caused tumor-infiltrating lymphocyte (TIL) desertification, whereas forced CCL5 expression prevented Cxcl9 expression and TILs loss, and attenuated tumor growth in mice through IFN-γ. The cooperation between tumor-derived CCL5 and IFN-γ-inducible CXCR3 ligands secreted by myeloid cells is key for orchestrating T cell infiltration in immunoreactive and immunoresponsive tumors.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Chemotaxis, Leukocyte , Cytokines/metabolism , Dendritic Cells/metabolism , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/metabolism , Macrophages/metabolism , Ovarian Neoplasms/metabolism , Animals , Antineoplastic Agents, Immunological/pharmacology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Chemokine CCL5/genetics , Chemokine CCL5/immunology , Chemokine CCL5/metabolism , Chemokine CXCL9/genetics , Chemokine CXCL9/immunology , Chemokine CXCL9/metabolism , Chemotaxis, Leukocyte/drug effects , Coculture Techniques , Cytokines/genetics , Cytokines/immunology , DNA Methylation , Dendritic Cells/drug effects , Dendritic Cells/immunology , Epigenesis, Genetic , Female , Gene Expression Regulation, Neoplastic , Humans , Immunotherapy/methods , Interferon-gamma/genetics , Interferon-gamma/immunology , Interferon-gamma/metabolism , Lymphocyte Activation/drug effects , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Macrophages/drug effects , Macrophages/immunology , Mice, Inbred C57BL , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Ovarian Neoplasms/therapy , Paracrine Communication , Receptors, CXCR3/genetics , Receptors, CXCR3/immunology , Receptors, CXCR3/metabolism , Signal Transduction
3.
Sci Transl Med ; 10(450)2018 07 18.
Article in English | MEDLINE | ID: mdl-30021886

ABSTRACT

Combination anti-cytotoxic T lymphocyte antigen 4 (CTLA-4) and anti-programmed cell death protein 1 (PD-1) therapy promotes antitumor immunity and provides superior benefit to patients with advanced-stage melanoma compared with either therapy alone. T cell immunity requires recognition of antigens in the context of major histocompatibility complex (MHC) class I and class II proteins by CD8+ and CD4+ T cells, respectively. We examined MHC class I and class II protein expression on tumor cells from previously untreated melanoma patients and correlated the results with transcriptional and genomic analyses and with clinical response to anti-CTLA-4, anti-PD-1, or combination therapy. Most (>50% of cells) or complete loss of melanoma MHC class I membrane expression was observed in 78 of 181 cases (43%), was associated with transcriptional repression of HLA-A, HLA-B, HLA-C, and B2M, and predicted primary resistance to anti-CTLA-4, but not anti-PD-1, therapy. Melanoma MHC class II membrane expression on >1% cells was observed in 55 of 181 cases (30%), was associated with interferon-γ (IFN-γ) and IFN-γ-mediated gene signatures, and predicted response to anti-PD-1, but not anti-CTLA-4, therapy. We conclude that primary response to anti-CTLA-4 requires robust melanoma MHC class I expression. In contrast, primary response to anti-PD-1 is associated with preexisting IFN-γ-mediated immune activation that includes tumor-specific MHC class II expression and components of innate immunity when MHC class I is compromised. The benefits of combined checkpoint blockade may be attributable, in part, to distinct requirements for melanoma-specific antigen presentation to initiate antitumor immunity.


Subject(s)
CTLA-4 Antigen/metabolism , HLA Antigens/metabolism , Immunotherapy , Melanoma/drug therapy , Melanoma/secondary , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Gene Expression Regulation, Neoplastic , Humans , Immunity, Innate , Melanoma/genetics , Melanoma/immunology , Programmed Cell Death 1 Receptor/metabolism , Transcription, Genetic
4.
PLoS One ; 6(6): e21097, 2011.
Article in English | MEDLINE | ID: mdl-21701589

ABSTRACT

It is well established that genomic alterations play an essential role in oncogenesis, disease progression, and response of tumors to therapeutic intervention. The advances of next-generation sequencing technologies (NGS) provide unprecedented capabilities to scan genomes for changes such as mutations, deletions, and alterations of chromosomal copy number. However, the cost of full-genome sequencing still prevents the routine application of NGS in many areas. Capturing and sequencing the coding exons of genes (the "exome") can be a cost-effective approach for identifying changes that result in alteration of protein sequences. We applied an exome-sequencing technology (Roche Nimblegen capture paired with 454 sequencing) to identify sequence variation and mutations in eight commonly used cancer cell lines from a variety of tissue origins (A2780, A549, Colo205, GTL16, NCI-H661, MDA-MB468, PC3, and RD). We showed that this technology can accurately identify sequence variation, providing ∼95% concordance with Affymetrix SNP Array 6.0 performed on the same cell lines. Furthermore, we detected 19 of the 21 mutations reported in Sanger COSMIC database for these cell lines. We identified an average of 2,779 potential novel sequence variations/mutations per cell line, of which 1,904 were non-synonymous. Many non-synonymous changes were identified in kinases and known cancer-related genes. In addition we confirmed that the read-depth of exome sequence data can be used to estimate high-level gene amplifications and identify homologous deletions. In summary, we demonstrate that exome sequencing can be a reliable and cost-effective way for identifying alterations in cancer genomes, and we have generated a comprehensive catalogue of genomic alterations in coding regions of eight cancer cell lines. These findings could provide important insights into cancer pathways and mechanisms of resistance to anti-cancer therapies.


Subject(s)
Exons/genetics , Genome, Human/genetics , Neoplasms/genetics , Cell Line, Tumor , Genotype , High-Throughput Nucleotide Sequencing , Humans , Mutation/genetics , Sequence Analysis, DNA
5.
Dev Biol ; 290(2): 482-94, 2006 Feb 15.
Article in English | MEDLINE | ID: mdl-16403493

ABSTRACT

decapentaplegic (dpp) is a direct target of Ultrabithorax (Ubx) in parasegment 7 (PS7) of the embryonic visceral mesoderm. We demonstrate that extradenticle (exd) and homothorax (hth) are also required for dpp expression in this location, as well as in PS3, at the site of the developing gastric caecae. A 420 bp element from dpp contains EXD binding sites necessary for expressing a reporter gene in both these locations. Using a specificity swap, we demonstrate that EXD directly activates this element in vivo. Activation does not require Ubx, demonstrating that EXD can activate transcription independently of homeotic proteins. Restoration is restricted to the domains of endogenous dpp expression, despite ubiquitous expression of altered specificity EXD. We demonstrate that nuclear EXD is more extensively phosphorylated than the cytoplasmic form, suggesting that EXD is a target of signal transduction by protein kinases.


Subject(s)
Drosophila Proteins/metabolism , Drosophila Proteins/physiology , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Homeodomain Proteins/physiology , Mesoderm/metabolism , Transcription Factors/metabolism , Transcription Factors/physiology , Transcriptional Activation , Amino Acid Sequence , Animals , Animals, Genetically Modified , Binding Sites , Blotting, Western , Body Patterning , Cell Nucleus/metabolism , Crosses, Genetic , Cytoplasm/metabolism , Drosophila Proteins/genetics , Electrophoresis, Gel, Two-Dimensional , Embryo, Nonmammalian/metabolism , Enhancer Elements, Genetic , Genes, Reporter , Homeodomain Proteins/genetics , Immunoblotting , Immunohistochemistry , Molecular Sequence Data , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Signal Transduction , Transcription Factors/genetics , Transgenes , beta-Galactosidase/metabolism
6.
Cancer Cell ; 1(2): 203-9, 2002 Mar.
Article in English | MEDLINE | ID: mdl-12086878

ABSTRACT

Prostate tumors are among the most heterogeneous of cancers, both histologically and clinically. Microarray expression analysis was used to determine whether global biological differences underlie common pathological features of prostate cancer and to identify genes that might anticipate the clinical behavior of this disease. While no expression correlates of age, serum prostate specific antigen (PSA), and measures of local invasion were found, a set of genes was identified that strongly correlated with the state of tumor differentiation as measured by Gleason score. Moreover, a model using gene expression data alone accurately predicted patient outcome following prostatectomy. These results support the notion that the clinical behavior of prostate cancer is linked to underlying gene expression differences that are detectable at the time of diagnosis.


Subject(s)
Gene Expression Profiling , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/genetics , Adult , Aged , Disease-Free Survival , Humans , Male , Middle Aged , Predictive Value of Tests , Prognosis , Prostatic Neoplasms/classification , Prostatic Neoplasms/pathology , Reproducibility of Results , Risk Factors , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...