Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Cancer ; 4(1): 43-61, 2023 01.
Article in English | MEDLINE | ID: mdl-36646856

ABSTRACT

Prolonged interferon (IFN) signaling in cancer cells can promote resistance to immune checkpoint blockade (ICB). How cancer cells retain effects of prolonged IFN stimulation to coordinate resistance is unclear. We show that, across human and/or mouse tumors, immune dysfunction is associated with cancer cells acquiring epigenetic features of inflammatory memory. Here, inflammatory memory domains, many of which are initiated by chronic IFN-γ, are maintained by signal transducer and activator of transcription (STAT)1 and IFN regulatory factor (IRF)3 and link histone 3 lysine 4 monomethylation (H3K4me1)-marked chromatin accessibility to increased expression of a subset of IFN-stimulated genes (ISGs). These ISGs include the RNA sensor OAS1 that amplifies type I IFN (IFN-I) and immune inhibitory genes. Abrogating cancer cell IFN-I signaling restores anti-programmed cell death protein 1 (PD1) response by increasing IFN-γ in immune cells, promoting dendritic cell and CD8+ T cell interactions, and expanding T cells toward effector-like states rather than exhausted states. Thus, cancer cells acquire inflammatory memory to augment a subset of ISGs that promote and predict IFN-driven immune dysfunction.


Subject(s)
Interferon Type I , Neoplasms , Animals , Humans , Mice , Epigenetic Memory , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Interferon Type I/metabolism , Interferon Type I/pharmacology , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interferon-gamma/pharmacology , Neoplasms/drug therapy , Neoplasms/genetics , Signal Transduction , T-Lymphocytes/immunology
2.
Genome Biol ; 21(1): 10, 2020 01 14.
Article in English | MEDLINE | ID: mdl-31937348

ABSTRACT

Although scRNA-seq is now ubiquitously adopted in studies of intratumor heterogeneity, detection of somatic mutations and inference of clonal membership from scRNA-seq is currently unreliable. We propose DENDRO, an analysis method for scRNA-seq data that clusters single cells into genetically distinct subclones and reconstructs the phylogenetic tree relating the subclones. DENDRO utilizes transcribed point mutations and accounts for technical noise and expression stochasticity. We benchmark DENDRO and demonstrate its application on simulation data and real data from three cancer types. In particular, on a mouse melanoma model in response to immunotherapy, DENDRO delineates the role of neoantigens in treatment response.


Subject(s)
Genetic Heterogeneity , Genetic Techniques , Neoplasms/genetics , Phylogeny , Software , Animals , Humans , Mice , Single-Cell Analysis
3.
Cell ; 178(4): 933-948.e14, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31398344

ABSTRACT

Interferon-gamma (IFNG) augments immune function yet promotes T cell exhaustion through PDL1. How these opposing effects are integrated to impact immune checkpoint blockade (ICB) is unclear. We show that while inhibiting tumor IFNG signaling decreases interferon-stimulated genes (ISGs) in cancer cells, it increases ISGs in immune cells by enhancing IFNG produced by exhausted T cells (TEX). In tumors with favorable antigenicity, these TEX mediate rejection. In tumors with neoantigen or MHC-I loss, TEX instead utilize IFNG to drive maturation of innate immune cells, including a PD1+TRAIL+ ILC1 population. By disabling an inhibitory circuit impacting PD1 and TRAIL, blocking tumor IFNG signaling promotes innate immune killing. Thus, interferon signaling in cancer cells and immune cells oppose each other to establish a regulatory relationship that limits both adaptive and innate immune killing. In melanoma and lung cancer patients, perturbation of this relationship is associated with ICB response independent of tumor mutational burden.


Subject(s)
Adaptive Immunity/immunology , Immunity, Innate/immunology , Interferon-gamma/genetics , Interferon-gamma/metabolism , Lung Neoplasms/immunology , Melanoma/immunology , Adoptive Transfer , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , CD8-Positive T-Lymphocytes/immunology , CTLA-4 Antigen/antagonists & inhibitors , Cell Line, Tumor , Cohort Studies , Female , Gene Knockout Techniques , Humans , Interferon-gamma/antagonists & inhibitors , Killer Cells, Natural/immunology , Lung Neoplasms/drug therapy , Melanoma/drug therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Progression-Free Survival , RNA-Seq , Transfection
4.
Cell ; 167(6): 1540-1554.e12, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27912061

ABSTRACT

Therapeutic blocking of the PD1 pathway results in significant tumor responses, but resistance is common. We demonstrate that prolonged interferon signaling orchestrates PDL1-dependent and PDL1-independent resistance to immune checkpoint blockade (ICB) and to combinations such as radiation plus anti-CTLA4. Persistent type II interferon signaling allows tumors to acquire STAT1-related epigenomic changes and augments expression of interferon-stimulated genes and ligands for multiple T cell inhibitory receptors. Both type I and II interferons maintain this resistance program. Crippling the program genetically or pharmacologically interferes with multiple inhibitory pathways and expands distinct T cell populations with improved function despite expressing markers of severe exhaustion. Consequently, tumors resistant to multi-agent ICB are rendered responsive to ICB monotherapy. Finally, we observe that biomarkers for interferon-driven resistance associate with clinical progression after anti-PD1 therapy. Thus, the duration of tumor interferon signaling augments adaptive resistance and inhibition of the interferon response bypasses requirements for combinatorial ICB therapies.


Subject(s)
CTLA-4 Antigen/antagonists & inhibitors , Melanoma/immunology , Melanoma/therapy , Radioimmunotherapy , Animals , B7-H1 Antigen/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm , Heterografts , Humans , Interferons/immunology , Melanoma/drug therapy , Melanoma/radiotherapy , Mice , Neoplasm Transplantation , STAT1 Transcription Factor , T-Lymphocytes/immunology
5.
Nature ; 520(7547): 373-7, 2015 Apr 16.
Article in English | MEDLINE | ID: mdl-25754329

ABSTRACT

Immune checkpoint inhibitors result in impressive clinical responses, but optimal results will require combination with each other and other therapies. This raises fundamental questions about mechanisms of non-redundancy and resistance. Here we report major tumour regressions in a subset of patients with metastatic melanoma treated with an anti-CTLA4 antibody (anti-CTLA4) and radiation, and reproduced this effect in mouse models. Although combined treatment improved responses in irradiated and unirradiated tumours, resistance was common. Unbiased analyses of mice revealed that resistance was due to upregulation of PD-L1 on melanoma cells and associated with T-cell exhaustion. Accordingly, optimal response in melanoma and other cancer types requires radiation, anti-CTLA4 and anti-PD-L1/PD-1. Anti-CTLA4 predominantly inhibits T-regulatory cells (Treg cells), thereby increasing the CD8 T-cell to Treg (CD8/Treg) ratio. Radiation enhances the diversity of the T-cell receptor (TCR) repertoire of intratumoral T cells. Together, anti-CTLA4 promotes expansion of T cells, while radiation shapes the TCR repertoire of the expanded peripheral clones. Addition of PD-L1 blockade reverses T-cell exhaustion to mitigate depression in the CD8/Treg ratio and further encourages oligoclonal T-cell expansion. Similarly to results from mice, patients on our clinical trial with melanoma showing high PD-L1 did not respond to radiation plus anti-CTLA4, demonstrated persistent T-cell exhaustion, and rapidly progressed. Thus, PD-L1 on melanoma cells allows tumours to escape anti-CTLA4-based therapy, and the combination of radiation, anti-CTLA4 and anti-PD-L1 promotes response and immunity through distinct mechanisms.


Subject(s)
B7-H1 Antigen/antagonists & inhibitors , CTLA-4 Antigen/antagonists & inhibitors , Cell Cycle Checkpoints/drug effects , Melanoma/drug therapy , Melanoma/immunology , Melanoma/radiotherapy , T-Lymphocytes/drug effects , T-Lymphocytes/radiation effects , Animals , B7-H1 Antigen/metabolism , Female , Humans , Melanoma/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Receptors, Antigen, T-Cell/drug effects , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/radiation effects
6.
Cell ; 159(3): 499-513, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25417103

ABSTRACT

Stromal communication with cancer cells can influence treatment response. We show that stromal and breast cancer (BrCa) cells utilize paracrine and juxtacrine signaling to drive chemotherapy and radiation resistance. Upon heterotypic interaction, exosomes are transferred from stromal to BrCa cells. RNA within exosomes, which are largely noncoding transcripts and transposable elements, stimulates the pattern recognition receptor RIG-I to activate STAT1-dependent antiviral signaling. In parallel, stromal cells also activate NOTCH3 on BrCa cells. The paracrine antiviral and juxtacrine NOTCH3 pathways converge as STAT1 facilitates transcriptional responses to NOTCH3 and expands therapy-resistant tumor-initiating cells. Primary human and/or mouse BrCa analysis support the role of antiviral/NOTCH3 pathways in NOTCH signaling and stroma-mediated resistance, which is abrogated by combination therapy with gamma secretase inhibitors. Thus, stromal cells orchestrate an intricate crosstalk with BrCa cells by utilizing exosomes to instigate antiviral signaling. This expands BrCa subpopulations adept at resisting therapy and reinitiating tumor growth.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/radiotherapy , Exosomes/metabolism , Paracrine Communication , Stromal Cells/metabolism , Animals , Breast Neoplasms/metabolism , Cell Line, Tumor , Computer Simulation , Drug Resistance, Neoplasm , Female , Humans , Interferons/metabolism , Mice, Nude , Radiation Tolerance , Receptors, Notch/metabolism , STAT1 Transcription Factor/metabolism , Signal Transduction , rab GTP-Binding Proteins/metabolism
7.
Biomaterials ; 33(28): 6833-42, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22727465

ABSTRACT

Clinical studies are underway for the aerosol delivery of plasmid DNA complexed with Genzyme Lipid GL67A to the lungs of patients with cystic fibrosis (CF). Plasmid vectors contain several functional elements all of which play a role in determining the efficacy of the final clinical product. To optimise the final plasmid, variations of CpG-free 5' enhancer elements and 3'UTR regions were inserted into a common CpG-free, plasmid backbone containing Luciferase or CFTR transgenes. Plasmids were compared in immortalised cell culture, human airway liquid interface primary cell cultures, and mouse lung models to determine which design directed optimal transgene expression. Following aerosol delivery to mouse lung, plasmids containing the murine CMV enhancer showed higher peak Luciferase activity than the human CMV enhancer, but the human version resulted in persistent expression. In cell culture, the SV40 3'UTR and a novel BGH2 3'UTR exhibited up to 20-fold higher Luciferase activity than the commonly used BGH 3'UTR, but in mouse lung aerosol studies the activity and duration was greater for BGH 3'UTR. Systematic evaluation of each functional component of the plasmid has resulted in an improved design, exhibiting superior levels and duration of lung gene expression.


Subject(s)
Cystic Fibrosis/therapy , Enhancer Elements, Genetic , Gene Transfer Techniques , Genetic Therapy/methods , Plasmids/genetics , Promoter Regions, Genetic , Aerosols/chemistry , Animals , CpG Islands/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/administration & dosage , Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , DNA/administration & dosage , Female , Gene Expression/genetics , HEK293 Cells , Humans , Luciferases/administration & dosage , Luciferases/chemistry , Lung/metabolism , Mice , Mice, Inbred BALB C , Plasmids/administration & dosage , Transgenes
SELECTION OF CITATIONS
SEARCH DETAIL
...