Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
1.
Cell ; 184(19): 4981-4995.e14, 2021 09 16.
Article in English | MEDLINE | ID: mdl-34464586

ABSTRACT

Poor tumor infiltration, development of exhaustion, and antigen insufficiency are common mechanisms that limit chimeric antigen receptor (CAR)-T cell efficacy. Delivery of pattern recognition receptor agonists is one strategy to improve immune function; however, targeting these agonists to immune cells is challenging, and off-target signaling in cancer cells can be detrimental. Here, we engineer CAR-T cells to deliver RN7SL1, an endogenous RNA that activates RIG-I/MDA5 signaling. RN7SL1 promotes expansion and effector-memory differentiation of CAR-T cells. Moreover, RN7SL1 is deployed in extracellular vesicles and selectively transferred to immune cells. Unlike other RNA agonists, transferred RN7SL1 restricts myeloid-derived suppressor cell (MDSC) development, decreases TGFB in myeloid cells, and fosters dendritic cell (DC) subsets with costimulatory features. Consequently, endogenous effector-memory and tumor-specific T cells also expand, allowing rejection of solid tumors with CAR antigen loss. Supported by improved endogenous immunity, CAR-T cells can now co-deploy peptide antigens with RN7SL1 to enhance efficacy, even when heterogenous CAR antigen tumors lack adequate neoantigens.


Subject(s)
Immunologic Factors/pharmacology , RNA/pharmacology , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , Animals , Antigens/metabolism , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , DEAD Box Protein 58/metabolism , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Extracellular Vesicles/metabolism , Humans , Immunity/drug effects , Immunocompetence , Immunologic Memory , Immunotherapy , Interferons/metabolism , Melanoma, Experimental/pathology , Mice, Inbred C57BL , Myeloid Cells/drug effects , Myeloid Cells/metabolism , Peptides/metabolism , Receptors, Pattern Recognition/metabolism , T-Lymphocytes/drug effects
2.
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
3.
Cell ; 170(2): 352-366.e13, 2017 Jul 13.
Article in English | MEDLINE | ID: mdl-28709002

ABSTRACT

Interactions between stromal fibroblasts and cancer cells generate signals for cancer progression, therapy resistance, and inflammatory responses. Although endogenous RNAs acting as damage-associated molecular patterns (DAMPs) for pattern recognition receptors (PRRs) may represent one such signal, these RNAs must remain unrecognized under non-pathological conditions. We show that triggering of stromal NOTCH-MYC by breast cancer cells results in a POL3-driven increase in RN7SL1, an endogenous RNA normally shielded by RNA binding proteins SRP9/14. This increase in RN7SL1 alters its stoichiometry with SRP9/14 and generates unshielded RN7SL1 in stromal exosomes. After exosome transfer to immune cells, unshielded RN7SL1 drives an inflammatory response. Upon transfer to breast cancer cells, unshielded RN7SL1 activates the PRR RIG-I to enhance tumor growth, metastasis, and therapy resistance. Corroborated by evidence from patient tumors and blood, these results demonstrate that regulation of RNA unshielding couples stromal activation with deployment of RNA DAMPs that promote aggressive features of cancer. VIDEO ABSTRACT.


Subject(s)
Breast Neoplasms/pathology , Exosomes/pathology , RNA, Untranslated/metabolism , Stromal Cells/pathology , Tumor Microenvironment , Breast Neoplasms/metabolism , DEAD Box Protein 58/metabolism , Exosomes/metabolism , Humans , Interferon Regulatory Factors/metabolism , MCF-7 Cells , Neoplasm Metastasis , RNA Polymerase III/genetics , RNA Polymerase III/metabolism , Receptors, Immunologic , Receptors, Pattern Recognition/metabolism , Signal Recognition Particle/metabolism , Stromal Cells/metabolism , Virus Diseases/metabolism
4.
Immunity ; 55(4): 671-685.e10, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35417675

ABSTRACT

Interferon-gamma (IFN-γ) has pleiotropic effects on cancer immune checkpoint blockade (ICB), including roles in ICB resistance. We analyzed gene expression in ICB-sensitive versus ICB-resistant tumor cells and identified a strong association between interferon-mediated resistance and expression of Ripk1, a regulator of tumor necrosis factor (TNF) superfamily receptors. Genetic interaction screening revealed that in cancer cells, RIPK1 diverted TNF signaling through NF-κB and away from its role in cell death. This promoted an immunosuppressive chemokine program by cancer cells, enhanced cancer cell survival, and decreased infiltration of T and NK cells expressing TNF superfamily ligands. Deletion of RIPK1 in cancer cells compromised chemokine secretion, decreased ARG1+ suppressive myeloid cells linked to ICB failure in mice and humans, and improved ICB response driven by CASP8-killing and dependent on T and NK cells. RIPK1-mediated resistance required its ubiquitin scaffolding but not kinase function. Thus, cancer cells co-opt RIPK1 to promote cell-intrinsic and cell-extrinsic resistance to immunotherapy.


Subject(s)
Drug Resistance, Neoplasm , Immune Checkpoint Inhibitors , Interferons , Neoplasms , Receptor-Interacting Protein Serine-Threonine Kinases , Animals , Immunotherapy , Interferon-gamma/metabolism , Interferons/metabolism , Mice , NF-kappa B/metabolism , Neoplasms/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
5.
Cell ; 165(2): 272-5, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-27058661

ABSTRACT

Improving efficacy of immune checkpoint blockade for cancer can be facilitated by combining these agents with each other and/or with other conventional or targeted therapies. Interferon and innate immune signaling pathways in immune and tumor cells have emerged as intriguing determinants of response and resistance, often in complex and seemingly paradoxical ways.


Subject(s)
Interferons/metabolism , Neoplasms/immunology , Neoplasms/therapy , Signal Transduction , Animals , Humans , Immune Tolerance , Immunity, Innate , Lymphocytes/immunology , Neoplasms/metabolism
6.
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
7.
Genes Dev ; 37(13-14): 605-620, 2023 07 01.
Article in English | MEDLINE | ID: mdl-37536952

ABSTRACT

The transcription factor RUNX1 is mutated in familial platelet disorder with associated myeloid malignancy (FPDMM) and in sporadic myelodysplastic syndrome and leukemia. RUNX1 was shown to regulate inflammation in multiple cell types. Here we show that RUNX1 is required in granulocyte-monocyte progenitors (GMPs) to epigenetically repress two inflammatory signaling pathways in neutrophils: Toll-like receptor 4 (TLR4) and type I interferon (IFN) signaling. RUNX1 loss in GMPs augments neutrophils' inflammatory response to the TLR4 ligand lipopolysaccharide through increased expression of the TLR4 coreceptor CD14. RUNX1 binds Cd14 and other genes encoding proteins in the TLR4 and type I IFN signaling pathways whose chromatin accessibility increases when RUNX1 is deleted. Transcription factor footprints for the effectors of type I IFN signaling-the signal transducer and activator of transcription (STAT1::STAT2) and interferon regulatory factors (IRFs)-were enriched in chromatin that gained accessibility in both GMPs and neutrophils when RUNX1 was lost. STAT1::STAT2 and IRF motifs were also enriched in the chromatin of retrotransposons that were derepressed in RUNX1-deficient GMPs and neutrophils. We conclude that a major direct effect of RUNX1 loss in GMPs is the derepression of type I IFN and TLR4 signaling, resulting in a state of fixed maladaptive innate immunity.


Subject(s)
Neutrophils , Toll-Like Receptor 4 , Toll-Like Receptor 4/metabolism , Monocytes/metabolism , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Cytokines/metabolism , Chromatin/metabolism , STAT1 Transcription Factor/metabolism
8.
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
9.
Immunity ; 48(3): 417-433, 2018 03 20.
Article in English | MEDLINE | ID: mdl-29562193

ABSTRACT

The success of immune checkpoint blockade in patients with a wide variety of malignancies has changed the treatment paradigm in oncology. However, combination therapies with immune checkpoint blockade will be needed to overcome resistance and broaden the clinical utility of immunotherapy. Here we discuss a framework for rationally designing combination therapy strategies based on enhancing major discriminatory functions of the immune system that are corrupted by cancer-namely, antigenicity, adjuvanticity, and homeostatic feedback inhibition. We review recent advances on how conventional genotoxic cancer therapies, molecularly targeted therapies, epigenetic agents, and immune checkpoint inhibitors can restore these discriminatory functions. Potential barriers that can impede response despite combination therapy are also discussed.


Subject(s)
Immunotherapy , Molecular Targeted Therapy , Neoplasms/immunology , Neoplasms/therapy , Animals , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Biomarkers, Tumor , Combined Modality Therapy , Humans , Immune System/cytology , Immune System/drug effects , Immune System/immunology , Immune System/metabolism , Immunomodulation/drug effects , Neoplasms/metabolism , Neoplasms/pathology , Receptors, Pattern Recognition/metabolism , Signal Transduction/drug effects , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
10.
Mol Cell ; 67(2): 252-265.e6, 2017 Jul 20.
Article in English | MEDLINE | ID: mdl-28689661

ABSTRACT

While maintaining the integrity of the genome and sustaining bioenergetics are both fundamental functions of the cell, potential crosstalk between metabolic and DNA repair pathways is poorly understood. Since histone acetylation plays important roles in DNA repair and is sensitive to the availability of acetyl coenzyme A (acetyl-CoA), we investigated a role for metabolic regulation of histone acetylation during the DNA damage response. In this study, we report that nuclear ATP-citrate lyase (ACLY) is phosphorylated at S455 downstream of ataxia telangiectasia mutated (ATM) and AKT following DNA damage. ACLY facilitates histone acetylation at double-strand break (DSB) sites, impairing 53BP1 localization and enabling BRCA1 recruitment and DNA repair by homologous recombination. ACLY phosphorylation and nuclear localization are necessary for its role in promoting BRCA1 recruitment. Upon PARP inhibition, ACLY silencing promotes genomic instability and cell death. Thus, the spatial and temporal control of acetyl-CoA production by ACLY participates in the mechanism of DNA repair pathway choice.


Subject(s)
ATP Citrate (pro-S)-Lyase/metabolism , Acetyl Coenzyme A/metabolism , BRCA1 Protein/metabolism , Cell Nucleus/enzymology , DNA Breaks, Double-Stranded , Recombinational DNA Repair , A549 Cells , ATP Citrate (pro-S)-Lyase/genetics , Acetylation , Animals , BRCA1 Protein/genetics , Cell Nucleus/drug effects , Female , G2 Phase Cell Cycle Checkpoints , Genomic Instability , Glucose/metabolism , HCT116 Cells , HeLa Cells , Histones/metabolism , Humans , Melanoma, Experimental/enzymology , Melanoma, Experimental/genetics , Melanoma, Experimental/pathology , Mice, Inbred C57BL , Phosphorylation , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Protein Binding , Protein Processing, Post-Translational , RNA Interference , Recombinational DNA Repair/drug effects , S Phase Cell Cycle Checkpoints , Serine , Time Factors , Transfection , Tumor Suppressor p53-Binding Protein 1/metabolism
11.
Nature ; 548(7668): 466-470, 2017 08 24.
Article in English | MEDLINE | ID: mdl-28759889

ABSTRACT

Inflammatory gene expression following genotoxic cancer therapy is well documented, yet the events underlying its induction remain poorly understood. Inflammatory cytokines modify the tumour microenvironment by recruiting immune cells and are critical for both local and systemic (abscopal) tumour responses to radiotherapy. A poorly understood feature of these responses is the delayed onset (days), in contrast to the acute DNA-damage responses that occur in minutes to hours. Such dichotomous kinetics implicate additional rate-limiting steps that are essential for DNA-damage-induced inflammation. Here we show that cell cycle progression through mitosis following double-stranded DNA breaks leads to the formation of micronuclei, which precede activation of inflammatory signalling and are a repository for the pattern-recognition receptor cyclic GMP-AMP synthase (cGAS). Inhibiting progression through mitosis or loss of pattern recognition by stimulator of interferon genes (STING)-cGAS impaired interferon signalling. Moreover, STING loss prevented the regression of abscopal tumours in the context of ionizing radiation and immune checkpoint blockade in vivo. These findings implicate temporal modulation of the cell cycle as an important consideration in the context of therapeutic strategies that combine genotoxic agents with immune checkpoint blockade.


Subject(s)
DNA Damage , Inflammation/metabolism , Micronuclei, Chromosome-Defective , Mitosis , Receptors, Pattern Recognition/metabolism , Signal Transduction , Animals , CTLA-4 Antigen/antagonists & inhibitors , Cell Cycle Checkpoints , Cell Line, Tumor , DNA Breaks, Double-Stranded , Disease Models, Animal , Female , Humans , Inflammation/pathology , Interferons/metabolism , Melanoma, Experimental/drug therapy , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Nucleotidyltransferases/metabolism
12.
Proc Natl Acad Sci U S A ; 116(14): 7005-7014, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30877256

ABSTRACT

p27 shifts from CDK inhibitor to oncogene when phosphorylated by PI3K effector kinases. Here, we show that p27 is a cJun coregulator, whose assembly and chromatin association is governed by p27 phosphorylation. In breast and bladder cancer cells with high p27pT157pT198 or expressing a CDK-binding defective p27pT157pT198 phosphomimetic (p27CK-DD), cJun is activated and interacts with p27, and p27/cJun complexes localize to the nucleus. p27/cJun up-regulates TGFB2 to drive metastasis in vivo. Global analysis of p27 and cJun chromatin binding and gene expression shows that cJun recruitment to many target genes is p27 dependent, increased by p27 phosphorylation, and activates programs of epithelial-mesenchymal transformation and metastasis. Finally, human breast cancers with high p27pT157 differentially express p27/cJun-regulated genes of prognostic relevance, supporting the biological significance of the work.


Subject(s)
Cell Movement , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Neoplasms/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Cell Adhesion , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p27/genetics , Humans , Neoplasms/genetics , Neoplasms/pathology , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-jun/genetics
13.
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
14.
Nature ; 527(7578): 329-35, 2015 Nov 19.
Article in English | MEDLINE | ID: mdl-26524530

ABSTRACT

Ever since Stephen Paget's 1889 hypothesis, metastatic organotropism has remained one of cancer's greatest mysteries. Here we demonstrate that exosomes from mouse and human lung-, liver- and brain-tropic tumour cells fuse preferentially with resident cells at their predicted destination, namely lung fibroblasts and epithelial cells, liver Kupffer cells and brain endothelial cells. We show that tumour-derived exosomes uptaken by organ-specific cells prepare the pre-metastatic niche. Treatment with exosomes from lung-tropic models redirected the metastasis of bone-tropic tumour cells. Exosome proteomics revealed distinct integrin expression patterns, in which the exosomal integrins α6ß4 and α6ß1 were associated with lung metastasis, while exosomal integrin αvß5 was linked to liver metastasis. Targeting the integrins α6ß4 and αvß5 decreased exosome uptake, as well as lung and liver metastasis, respectively. We demonstrate that exosome integrin uptake by resident cells activates Src phosphorylation and pro-inflammatory S100 gene expression. Finally, our clinical data indicate that exosomal integrins could be used to predict organ-specific metastasis.


Subject(s)
Brain/metabolism , Exosomes/metabolism , Integrins/metabolism , Liver/metabolism , Lung/metabolism , Neoplasm Metastasis/pathology , Neoplasm Metastasis/prevention & control , Tropism , Animals , Biomarkers/metabolism , Brain/cytology , Cell Line, Tumor , Endothelial Cells/cytology , Endothelial Cells/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Genes, src , Humans , Integrin alpha6beta1/metabolism , Integrin alpha6beta4/antagonists & inhibitors , Integrin alpha6beta4/metabolism , Integrin beta Chains/metabolism , Integrin beta4/metabolism , Integrins/antagonists & inhibitors , Kupffer Cells/cytology , Kupffer Cells/metabolism , Liver/cytology , Lung/cytology , Mice , Mice, Inbred C57BL , Organ Specificity , Phosphorylation , Receptors, Vitronectin/antagonists & inhibitors , Receptors, Vitronectin/metabolism , S100 Proteins/genetics
15.
Proc Natl Acad Sci U S A ; 113(37): E5528-37, 2016 09 13.
Article in English | MEDLINE | ID: mdl-27573852

ABSTRACT

Cancer is a disease driven by evolutionary selection on somatic genetic and epigenetic alterations. Here, we propose Canopy, a method for inferring the evolutionary phylogeny of a tumor using both somatic copy number alterations and single-nucleotide alterations from one or more samples derived from a single patient. Canopy is applied to bulk sequencing datasets of both longitudinal and spatial experimental designs and to a transplantable metastasis model derived from human cancer cell line MDA-MB-231. Canopy successfully identifies cell populations and infers phylogenies that are in concordance with existing knowledge and ground truth. Through simulations, we explore the effects of key parameters on deconvolution accuracy and compare against existing methods. Canopy is an open-source R package available at https://cran.r-project.org/web/packages/Canopy/.


Subject(s)
Clonal Evolution/genetics , Evolution, Molecular , High-Throughput Nucleotide Sequencing/methods , Neoplasms/genetics , Computational Biology/methods , DNA Copy Number Variations/genetics , Exome/genetics , Genetic Heterogeneity , Humans , Internet , Mutation , Neoplasms/pathology , Phylogeny , Polymorphism, Single Nucleotide , Software
16.
Cancer ; 124(1): 183-191, 2018 Jan 01.
Article in English | MEDLINE | ID: mdl-28885696

ABSTRACT

BACKGROUND: Nearly 1 in 5 Americans will develop skin cancer, and as a result, survivors of skin cancer compose one of the largest groups of cancer survivors. Survivorship care plans (SCPs) are an important tool for improving patient outcomes and provide critical information to both survivors and health care professionals. Recent efforts have been made to expand SCP utilization; however, which patients currently receive SCPs is poorly understood. METHODS: This study used 596 individuals with a diagnosis of melanoma (n = 391) or nonmelanoma skin cancer (n = 205) who had used an Internet-based SCP tool from May 2010 to December 2016 to model the patient and provider characteristics that determine SCP utilization. RESULTS: Survivors were predominantly white (95.3%) and female (56.5%). Survivors who received a treatment summary were more likely to also receive an SCP. University and nonuniversity cancer centers used SCPs at a higher rate than other care settings. Survivors whose care was managed by a team rather than just an individual physician were also more likely to receive an SCP. Survivors older than 70 years at diagnosis were almost twice as likely to receive a plan as survivors who were diagnosed at a younger age. CONCLUSIONS: With a convenience sample of skin cancer survivors, it is possible to model factors that predict the receipt of SCPs. Important variables include the diagnosis age, treatment setting, physician type, and treatment-summary utilization. A closer examination of these variables identified several disparities in care-plan use and, therefore, opportunities to improve the distribution of SCPs. Further validation in additional cohorts of survivors is necessary to confirm these conclusions. Cancer 2018;124:183-91. © 2017 American Cancer Society.


Subject(s)
Aftercare/methods , Cancer Survivors , Melanoma/therapy , Patient Care Planning/statistics & numerical data , Skin Neoplasms/therapy , Survivorship , Academic Medical Centers , Adult , Age Factors , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Oncologists , Physicians, Primary Care , Supervised Machine Learning , United States
17.
Br J Cancer ; 119(10): 1200-1207, 2018 11.
Article in English | MEDLINE | ID: mdl-30318516

ABSTRACT

BACKGROUND: We conducted a phase I trial evaluating pembrolizumab+hypofractionated radiotherapy (HFRT) for patients with metastatic cancers. METHODS: There were two strata (12 patients each): (i) NSCLC/melanoma progressing on prior anti-PD-1 therapy, (ii) other cancer types; anti-PD-1-naive. Patients received 6 cycles of pembrolizumab, starting 1 week before HFRT. Patients had ≥2 lesions; only one was irradiated (8 Gy × 3 for first half; 17 Gy × 1 for second half in each stratum) and the other(s) followed for response. RESULTS: Of the 24 patients, 20 (83%) had treatment-related adverse events (AEs) (all grade 1 or 2). There were eight grade 3 AEs, none treatment related. There were no dose-limiting toxicities or grade 4/5 AEs. Stratum 1: two patients (of 12) with progression on prior PD-1 blockade experienced prolonged responses (9.2 and 28.1 months). Stratum 2: one patient experienced a complete response and two had prolonged stable disease (7.4 and 7.0 months). Immune profiling demonstrated that anti-PD-1 therapy and radiation induced a consistent increase in the proliferation marker Ki67 in PD-1-expressing CD8 T cells. CONCLUSIONS: HFRT was well tolerated with pembrolizumab, and in some patients with metastatic NSCLC or melanoma, it reinvigorated a systemic response despite previous progression on anti-PD-1 therapy. CLINICAL TRIAL REGISTRATION: NCT02303990 ( www.clinicaltrials.gov ).


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/radiotherapy , Chemoradiotherapy , Lung Neoplasms/drug therapy , Lung Neoplasms/radiotherapy , Melanoma/drug therapy , Melanoma/radiotherapy , Radiation Dose Hypofractionation , Skin Neoplasms/drug therapy , Skin Neoplasms/radiotherapy , Adult , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/pathology , Disease-Free Survival , Female , Humans , Lung Neoplasms/pathology , Male , Melanoma/pathology , Middle Aged , Neoplasm Metastasis/drug therapy , Neoplasm Metastasis/radiotherapy , Skin Neoplasms/pathology
18.
Trends Immunol ; 36(11): 725-737, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26604042

ABSTRACT

Much of our understanding on resistance mechanisms to conventional cancer therapies such as chemotherapy and radiation has focused on cell intrinsic properties that antagonize the detrimental effects of DNA and other cellular damage. However, it is becoming clear that the immune system and/or innate immune signaling pathways can integrate with these intrinsic mechanisms to profoundly influence treatment efficacy. In this context, recent evidence indicates that interferon (IFN) signaling has an important role in this integration by influencing immune and intrinsic/non-immune determinants of therapy response. However, IFN signaling can be both immunostimulatory and immunosuppressive, and the factors determining these outcomes in different disease settings are unclear. Here I discuss the regulation and molecular events in cancer that are associated with these dichotomous functions.


Subject(s)
Interferons/immunology , Neoplasms/drug therapy , Neoplasms/immunology , Animals , Humans , Neoplasms/pathology
19.
Proc Natl Acad Sci U S A ; 111(1): 291-6, 2014 Jan 07.
Article in English | MEDLINE | ID: mdl-24368849

ABSTRACT

Glioblastoma multiforme (GBM) and the mesenchymal GBM subtype in particular are highly malignant tumors that frequently exhibit regions of severe hypoxia and necrosis. Because these features correlate with poor prognosis, we investigated microRNAs whose expression might regulate hypoxic GBM cell survival and growth. We determined that the expression of microRNA-218 (miR-218) is decreased significantly in highly necrotic mesenchymal GBM, and orthotopic tumor studies revealed that reduced miR-218 levels confer GBM resistance to chemotherapy. Importantly, miR-218 targets multiple components of receptor tyrosine kinase (RTK) signaling pathways, and miR-218 repression increases the abundance and activity of multiple RTK effectors. This elevated RTK signaling also promotes the activation of hypoxia-inducible factor (HIF), most notably HIF2α. We further show that RTK-mediated HIF2α regulation is JNK dependent, via jun proto-oncogene. Collectively, our results identify an miR-218-RTK-HIF2α signaling axis that promotes GBM cell survival and tumor angiogenesis, particularly in necrotic mesenchymal tumors.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain Neoplasms/metabolism , Glioblastoma/metabolism , Mesoderm/metabolism , MicroRNAs/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Adult , Aged , Aged, 80 and over , Animals , Antineoplastic Agents/pharmacology , Cell Survival , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Hypoxia , Mice , Mice, Nude , Middle Aged , Necrosis , Neoplasm Transplantation , Neovascularization, Pathologic , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Mas , Signal Transduction , Young Adult
20.
Breast Cancer Res Treat ; 156(2): 405-6, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26993130

ABSTRACT

Erratum to: Breast Cancer Res Treat (2013),138:369­381,DOI 10.1007/s10549-012-2389-6. In the original publication of the article, the Fig. 4c and d were published erroneously. The revised Fig. 4 is given in this erratum.

SELECTION OF CITATIONS
SEARCH DETAIL