Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
PLoS Genet ; 12(4): e1005895, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27093186

ABSTRACT

Small cell lung cancer (SCLC) is an aggressive disease with poor survival. A few sequencing studies performed on limited number of samples have revealed potential disease-driving genes in SCLC, however, much still remains unknown, particularly in the Asian patient population. Here we conducted whole exome sequencing (WES) and transcriptomic sequencing of primary tumors from 99 Chinese SCLC patients. Dysregulation of tumor suppressor genes TP53 and RB1 was observed in 82% and 62% of SCLC patients, respectively, and more than half of the SCLC patients (62%) harbored TP53 and RB1 mutation and/or copy number loss. Additionally, Serine/Arginine Splicing Factor 1 (SRSF1) DNA copy number gain and mRNA over-expression was strongly associated with poor survival using both discovery and validation patient cohorts. Functional studies in vitro and in vivo demonstrate that SRSF1 is important for tumorigenicity of SCLC and may play a key role in DNA repair and chemo-sensitivity. These results strongly support SRSF1 as a prognostic biomarker in SCLC and provide a rationale for personalized therapy in SCLC.


Subject(s)
Carcinoma, Small Cell/genetics , Lung Neoplasms/genetics , Oncogene Proteins/genetics , Serine-Arginine Splicing Factors/genetics , Adult , Aged , DNA Copy Number Variations , DNA Damage , Female , Gene Silencing , Humans , Male , Middle Aged , Mutation
2.
Gastroenterology ; 153(1): 77-86.e6, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28390867

ABSTRACT

BACKGROUND & AIMS: MEDI2070 is a human monoclonal antibody that selectively inhibits interleukin 23 (IL23), a cytokine implicated in the pathogenesis of Crohn's disease (CD). We analyzed its safety and efficacy in treatment of CD in a phase 2a study. METHODS: We conducted a double-blind, placebo-controlled study of 119 adults with moderate to severe CD failed by treatment with tumor necrosis factor antagonists. Patients were randomly assigned (1:1) to groups given MEDI2070 (700 mg) or placebo intravenously at weeks 0 and 4. Patients received open-label MEDI2070 (210 mg) subcutaneously every 4 weeks from weeks 12 to 112. The CD Activity Index was used to measure disease activity. RESULTS: The primary outcome, clinical response (either a 100-point decrease in CD Activity Index score from baseline or clinical remission, defined as CD Activity Index score <150) at week 8 occurred in 49.2% of patients receiving MEDI2070 (n = 59) compared with 26.7% receiving placebo (n = 60; absolute difference, 22.5%; 95% confidence interval, 5.6%-39.5%; P = .010). Clinical response at week 24 occurred in 53.8% of patients who continued to receive open-label MEDI2070 and in 57.7% of patients who had received placebo during the double-blind period and open-label MEDI2070 thereafter. The most common adverse events were headache and nasopharyngitis. Higher baseline serum concentrations of IL22, a cytokine whose expression is induced by IL23, were associated with greater likelihood of response to MEDI2070 compared with placebo. CONCLUSIONS: In a phase 2a trial of patients with moderate to severe Crohn's disease who had failed treatment with tumor necrosis factor antagonists, 8 and 24 weeks of treatment with MEDI2070 were associated with clinical improvement. ClinicalTrials.gov ID: NCT01714726.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Crohn Disease/drug therapy , Adult , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Humanized , Double-Blind Method , Female , Headache/chemically induced , Humans , Interleukin-23/antagonists & inhibitors , Interleukins/blood , Male , Middle Aged , Nasopharyngitis/chemically induced , Retreatment , Severity of Illness Index , Treatment Outcome , Young Adult , Interleukin-22
3.
J Immunol ; 197(1): 42-50, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27226090

ABSTRACT

Type I IFNs play a critical role in the immune response to viral infection and may also drive autoimmunity through modulation of monocyte maturation and promotion of autoreactive lymphocyte survival. Recent demonstrations of type I IFN gene signatures in autoimmune diseases, including scleroderma, led us to investigate the pathological role of IFNs in a preclinical model of sclerodermatous graft-versus-host disease. Using a neutralizing Ab against the type I IFN receptor IFNAR1, we observed a marked reduction in dermal inflammation, vasculopathy, and fibrosis compared with that seen in the presence of intact IFNAR1 signaling. The ameliorative effects of IFNAR1 blockade were restricted to the skin and were highly associated with inhibition of chronic vascular injury responses and not due to the inhibition of the T or B cell alloresponse. Inhibition of IFNAR1 normalized the overexpression of IFN-inducible genes in graft-versus-host disease skin and markedly reduced dermal IFN-α levels. Depletion of plasmacytoid dendritic cells, a major cellular source of type I IFNs, did not reduce the severity of fibrosis or type I IFN gene signature in the skin. Taken together, these studies demonstrate an important role for type I IFN in skin fibrosis, and they provide a rationale for IFNAR1 inhibition in scleroderma.


Subject(s)
Dendritic Cells/immunology , Graft vs Host Disease/immunology , Inflammation/immunology , Interferon-alpha/metabolism , Scleroderma, Systemic/immunology , Skin/pathology , Vascular Diseases/immunology , Animals , Antibodies, Blocking/administration & dosage , Autoantibodies/blood , Disease Models, Animal , Female , Fibrosis , Gene Expression Regulation , Humans , Interferon-alpha/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptor, Interferon alpha-beta/immunology , Signal Transduction
4.
Transgenic Res ; 26(6): 791-798, 2017 12.
Article in English | MEDLINE | ID: mdl-28819706

ABSTRACT

Humanized mouse models are important tools in many areas of biological drug development including, within oncology research, the development of antagonistic antibodies that have the potential to block tumor growth by controlling vascularization and are key to the generation of in vivo proof-of-concept efficacy data. However, due to cross reactivity between human antibodies and mouse target such studies regularly require mouse models expressing only the human version of the target molecule. Such humanized knock-in/knock-out, KIKO, models are dependent upon the generation of homozygous mice expressing only the human molecule, compensating for loss of the mouse form. However, KIKO strategies can fail to generate homozygous mice, even though the human form is expressed and the endogenous mouse locus is correctly targeted. A typical strategy for generating KIKO mice is by ATG fusion where the human cDNA is inserted downstream of the endogenous mouse promoter elements. However, when adopting this strategy it is possible that the mouse promoter fails to express the human form in a manner compensating for loss of the mouse form or alternatively the human protein is incompatible in the context of the mouse pathway being investigated. So to understand more around the biology of KIKO models, and to overcome our failure with a number of ATG fusion strategies, we developed a range of humanized models focused on Delta-like 4 (Dll4), a target where we initially failed to generate a humanized model. By adopting a broader biologic strategy, we successfully generated a humanized DLL4 KIKO which led to a greater understanding of critical biological aspects for consideration when developing humanized models.


Subject(s)
Antineoplastic Agents/pharmacology , Intercellular Signaling Peptides and Proteins/genetics , Mice, Transgenic/genetics , Adaptor Proteins, Signal Transducing , Animals , Antibodies, Monoclonal/pharmacology , Calcium-Binding Proteins , Female , Humans , Intercellular Signaling Peptides and Proteins/immunology , Intercellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Male , Membrane Proteins/genetics , Mice, Knockout , Mice, SCID , Xenograft Model Antitumor Assays
5.
J Transl Med ; 13: 379, 2015 Dec 11.
Article in English | MEDLINE | ID: mdl-26653219

ABSTRACT

BACKGROUND AND AIMS: Orthotopic liver transplantation (OLT) can be an effective treatment option for certain patients with early stage hepatocellular carcinoma (HCC) meeting Milan, UCSF, or Hangzhou criteria. However, HCC recurrence rates post-OLT range from 20 to 40 %, with limited follow-up options. Elucidating genetic drivers common to primary and post-OLT recurrent tumors may further our understanding and help identify predictive biomarkers of recurrence-both to ultimately help manage clinical decisions for patients undergoing OLT. METHODS: Whole exome and RNA sequencing in matched primary and recurrent tumors, normal adjacent tissues, and blood from four Chinese HCC patients was conducted. SiRNA knockdown and both qRT-PCR and Western assays were performed on PLCPRF5, SNU449 and HEPG2 cell lines; immunohistochemistry and RNA Sequencing were conducted on the primary tumors of Chinese HCC patients who experienced tumor recurrence post-OLT (n = 9) or did not experience tumor recurrence (n = 12). RESULTS: In three independent HCC studies of patients undergoing transplantation (n = 21) or surgical resection (n = 242, n = 44) of primary tumors (total n = 307), HERC5 mRNA under-expression correlated with shorter: time to tumor recurrence (p = 0.007 and 0.02) and overall survival (p = 0.0063 and 0.023), even after adjustment for relevant clinical variables. HERC5 loss drives CCL20 mRNA and protein over-expression and associates with regulatory T cell infiltration as measured by FOXP3 expression. Further, matched primary and recurrent tumors from the 4 HCC patients indicated clonal selection advantage of Wnt signaling activation and CDKN2A inactivation. CONCLUSIONS: HERC5 plays a crucial role in HCC immune evasion and has clinical relevance as a reproducible prognostic marker for risk of tumor recurrence and survival in patients.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/surgery , Intracellular Signaling Peptides and Proteins/genetics , Liver Neoplasms/surgery , Liver Transplantation , DNA Copy Number Variations , Humans , Prognosis , Recurrence
6.
Ann Rheum Dis ; 73(1): 256-62, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23434567

ABSTRACT

OBJECTIVE: To assess the pharmacodynamic effects of sifalimumab, an investigational anti-IFN-α monoclonal antibody, in the blood and muscle of adult dermatomyositis and polymyositis patients by measuring neutralisation of a type I IFN gene signature (IFNGS) following drug exposure. METHODS: A phase 1b randomised, double-blinded, placebo controlled, dose-escalation, multicentre clinical trial was conducted to evaluate sifalimumab in dermatomyositis or polymyositis patients. Blood and muscle biopsies were procured before and after sifalimumab administration. Selected proteins were measured in patient serum with a multiplex assay, in the muscle using immunohistochemistry, and transcripts were profiled with microarray and quantitative reverse transcriptase PCR assays. A 13-gene IFNGS was used to measure the pharmacological effect of sifalimumab. RESULTS: The IFNGS was suppressed by a median of 53-66% across three time points (days 28, 56 and 98) in blood (p=0.019) and 47% at day 98 in muscle specimens post-sifalimumab administration. Both IFN-inducible transcripts and proteins were prevalently suppressed following sifalimumab administration. Patients with 15% or greater improvement from baseline manual muscle testing scores showed greater neutralisation of the IFNGS than patients with less than 15% improvement in both blood and muscle. Pathway/functional analysis of transcripts suppressed by sifalimumab showed that leucocyte infiltration, antigen presentation and immunoglobulin categories were most suppressed by sifalimumab and highly correlated with IFNGS neutralisation in muscle. CONCLUSIONS: Sifalimumab suppressed the IFNGS in blood and muscle tissue in myositis patients, consistent with this molecule's mechanism of action with a positive correlative trend between target neutralisation and clinical improvement. These observations will require confirmation in a larger trial powered to evaluate efficacy.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Dermatomyositis/drug therapy , Dermatomyositis/immunology , Immunosuppressive Agents/administration & dosage , Polymyositis/drug therapy , Polymyositis/immunology , Adult , Aged , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Humanized , Double-Blind Method , Female , Gene Expression/drug effects , Gene Expression/immunology , Humans , Immunosuppressive Agents/adverse effects , Interferon Type I/blood , Interferon Type I/genetics , Interferon Type I/immunology , Interferon-alpha/blood , Interferon-alpha/genetics , Interferon-alpha/immunology , Leukocytes/drug effects , Leukocytes/immunology , Male , Middle Aged , Muscle, Skeletal/immunology , Placebos , Treatment Outcome , Young Adult
7.
BMC Cancer ; 14: 131, 2014 Feb 26.
Article in English | MEDLINE | ID: mdl-24571613

ABSTRACT

BACKGROUND: To evaluate the insulin receptor isoform mRNA expression status in non-small cell lung cancer (NSCLC) patients. METHODS: RNA-seq data from 614 NSCLC [355 adenocarcinomas (LUAD) and 259 squamous cell carcinomas (LUSC)] and 92 normal lung specimens were obtained from The Cancer Genome Atlas (TCGA) to evaluate the mRNA expression of insulin receptor isoform A (IR-A) and insulin receptor isoform B (IR-B). The differential expression status of the insulin receptor isoforms in NSCLC patients was confirmed using qRT-PCR assays with lung cancer cDNA arrays and primary tumor samples. RESULTS: The mRNA expression levels of IR-B were significantly lower in some NSCLC samples compared to normal lung specimens, including both LUAD and LUSC. Notably, no IR-B transcripts were detected - only the IR-A isoform was expressed in 11% of NSCLC patients. This decrease in IR-B expression contributed to an elevated IR-A/IR-B ratio, which was also associated with lower epithelial-mesenchymal transition gene signatures in NSCLC and longer patient survival under standard of care in LUSC. In addition to NSCLC, RNA-seq data from TCGA revealed a similar increase in IR-A/IR-B ratio in many other cancer types, with high prevalence in acute myeloid leukemia, glioblastoma multiforme, and brain lower grade glioma. CONCLUSIONS: Our results indicate a common reduction of the mRNA expression level of IR-B and an increased IR-A/IR-B mRNA ratio in NSCLC and other tumor types. The relationship of altered IR-A/IR-B ratios with cancer progression and patient survival should be prospectively explored in future studies.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Epithelial-Mesenchymal Transition/genetics , Lung Neoplasms/genetics , Lung Neoplasms/pathology , RNA Isoforms , Receptor, Insulin/genetics , Alternative Splicing , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/pathology , Computational Biology/methods , Databases, Nucleic Acid , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/mortality , Outcome Assessment, Health Care , Prognosis , RNA, Messenger/genetics
8.
J Immunol ; 187(7): 3603-12, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21873529

ABSTRACT

Production of pathogenic Abs contributes to disease progression in many autoimmune disorders. The immunosuppressant agent mycophenolic acid (MPA) has shown clinical efficacy for patients with autoimmunity. The goal of these studies was to elucidate the mechanisms of action of MPA on B cells isolated from healthy individuals and autoimmune patients. In this study, we show that MPA significantly inhibited both proliferation and differentiation of primary human B cells stimulated under various conditions. Importantly, MPA did not globally suppress B cell responsiveness or simply induce cell death, but rather selectively inhibited early activation events and arrested cells in the G0/G1 phase of the cell cycle. Furthermore, MPA blocked expansion of both naive and memory B cells and prevented plasma cell (PC) differentiation and Ab production from healthy controls and individuals with rheumatoid arthritis. Finally, whereas MPA potently suppressed Ig secretion from activated primary B cells, terminally differentiated PCs were not susceptible to inhibition by MPA. The target of MPA, IMPDH2, was found to be downregulated in PCs, likely explaining the resistance of these cells to MPA. These results suggest that MPA provides benefit in settings of autoimmunity by directly preventing activation and PC differentiation of B cells; however, MPA is unlikely to impact autoantibody production by preexisting, long-lived PCs.


Subject(s)
B-Lymphocytes/drug effects , Cell Differentiation/drug effects , Immunosuppressive Agents/pharmacology , Lymphocyte Activation/drug effects , Mycophenolic Acid/pharmacology , Apoptosis/drug effects , Apoptosis/immunology , B-Lymphocytes/cytology , Cell Differentiation/immunology , Cell Proliferation/drug effects , Cell Separation , Coculture Techniques , Flow Cytometry , Humans , Lymphocyte Activation/immunology , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/immunology
9.
Ann Rheum Dis ; 70(11): 2029-36, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21803750

ABSTRACT

OBJECTIVE: To characterise activation of the type I interferon (IFN) pathway in patients with systemic lupus erythematosus (SLE), dermatomyositis (DM), polymyositis (PM), rheumatoid arthritis (RA) and systemic scleroderma (SSc) and to evaluate the potential to develop a molecular diagnostic tool from the peripheral blood that reflects this activation in disease-affected tissues. METHODS: Overexpressed transcripts were identified in the whole blood (WB) of 262 patients with SLE, 44 with DM, 33 with PM, 28 with SSc and 89 with RA and compared with 24 healthy subjects using Affymetrix microarrays. A five gene type I IFN signature was assessed in these subjects to identify subpopulations showing both activation and concordance of the type I IFN pathway in the peripheral blood and disease-affected tissues of each disease and to correlate activation of this pathway in the WB with clinical measurements. RESULTS: A common set of 36 type I IFN inducible transcripts were identified among the most overexpressed in the WB of all subjects. Significant activation of the type I IFN pathway in subgroups of each of the five diseases studied was observed. Baseline disease activity measurements correlated with a type I IFN gene signature in the WB of subjects with SLE, PM and SSc, as did various serum autoantibody levels in subjects with SLE and DM. This signature was also well correlated between disease-affected tissue and WB in subjects with SLE, DM, PM and SSc. CONCLUSIONS: The results indicate that the type I IFN pathway is activated in patient subsets of five rheumatic diseases and suggest that these subsets may benefit from anti-IFN therapy.


Subject(s)
Interferon Type I/biosynthesis , Rheumatic Diseases/immunology , Adult , Arthritis, Rheumatoid/immunology , Biomarkers/blood , Blood Proteins/metabolism , Female , Gene Expression Profiling/methods , Humans , Interferon Type I/genetics , Interferon-alpha/antagonists & inhibitors , Lupus Erythematosus, Systemic/immunology , Male , Middle Aged , Myositis/immunology , Oligonucleotide Array Sequence Analysis/methods , Scleroderma, Systemic/immunology , Severity of Illness Index , Signal Transduction/immunology
10.
Ann Neurol ; 67(1): 53-63, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20186858

ABSTRACT

OBJECTIVE: We investigated interferon-stimulated gene 15 (ISG15), a poorly understood ubiquitin-like modifier, and its enzymatic pathway in dermatomyositis (DM), an autoimmune disease primarily involving muscle and skin. METHODS: We generated microarray data measuring transcript abundance for approximately 18,000 genes in each of 113 human muscle biopsy specimens, and studied biopsy specimens and cultured skeletal muscle using immunohistochemistry, immunoblotting proteomics, real-time quantitative polymerase chain reaction, and laser-capture microdissection. RESULTS: Transcripts encoding ISG15-conjugation pathway proteins were markedly upregulated in DM with perifascicular atrophy (DM-PFA) muscle (ISG15 339-fold, HERC5 62-fold, and USP18 68-fold) compared with 99 non-DM samples. Combined analysis with publicly available microarray datasets showed that >50-fold ISG15 transcript elevation had 100% sensitivity and specificity for 28 biopsies from adult DM-PFA and juvenile DM patients compared with 199 muscle samples from other muscle diseases. Free ISG15 and ISG15-conjugated proteins were only found on immunoblots from DM-PFA muscle. Cultured human skeletal muscle exposed to type 1 interferons produced similar transcripts and ISG15 protein and conjugates. Laser-capture microdissection followed by proteomic analysis showed deficiency of titin in DM perifascicular atrophic myofibers. INTERPRETATION: A large-scale microarray study of muscle samples demonstrated that among a diverse group of muscle diseases DM was uniquely associated with upregulation of the ISG15 conjugation pathway. Exposure of human skeletal muscle cell culture to type 1 interferons produced a molecular picture highly similar to that seen in human DM muscle. Perifascicular atrophic myofibers in DM were deficient in a number of skeletal muscle proteins including titin.


Subject(s)
Cytokines/metabolism , Dermatomyositis/metabolism , Muscle, Skeletal/metabolism , Ubiquitins/metabolism , Cells, Cultured , Connectin , Cytokines/genetics , Databases, Genetic , Dermatomyositis/diagnosis , Dermatomyositis/genetics , Humans , Immunoblotting , Immunohistochemistry , Interferon Type I/metabolism , Lasers , Microdissection/methods , Muscle Proteins/deficiency , Muscular Diseases/diagnosis , Muscular Diseases/genetics , Muscular Diseases/metabolism , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Protein Kinases/deficiency , Proteomics/methods , Sensitivity and Specificity , Ubiquitins/genetics , Up-Regulation
11.
Clin Immunol ; 136(1): 130-8, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20346735

ABSTRACT

To determine the potential consequences of plasmacytoid dendritic cell (pDC) accumulation in tissue sites observed in several autoimmune diseases, we measured type 1 interferon production from circulating human pDCs as a function of pDC concentration. The effects of interferon-alpha and blockade of the type 1 interferon receptor (IFNAR) on human pDC type 1 interferon and interferon-inducible transcription and protein production were measured. Human pDCs became far more efficient producers of interferon-alpha at concentrations beyond those normally present in blood, through an IFNAR-dependent mechanism. Extracellular interferon-alpha increased pDC production of type 1 interferons. The accumulation of pDCs in diseased tissue sites allows marked non-linear amplification of type 1 interferon production locally. The role of the IFNAR-dependent mechanism of interferon production by human pDCs is greater than previously suggested. IFNAR blockade has potential for diminishing type 1 interferon production by all human cells.


Subject(s)
Dendritic Cells/cytology , Dendritic Cells/metabolism , Interferon Type I/metabolism , Adaptor Proteins, Signal Transducing , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Autocrine Communication/drug effects , Autocrine Communication/immunology , Carrier Proteins/genetics , Cell Count , Cytokines/genetics , Cytokines/metabolism , Dendritic Cells/drug effects , Endopeptidases/genetics , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Gene Expression/drug effects , Gene Expression/genetics , Humans , Interferon Type I/genetics , Interferon alpha-2 , Interferon-alpha/genetics , Interferon-alpha/metabolism , Interferon-alpha/pharmacology , Interferon-beta/genetics , Intracellular Signaling Peptides and Proteins/genetics , Kinetics , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Myxovirus Resistance Proteins , Oligodeoxyribonucleotides/pharmacology , Oxidoreductases Acting on CH-CH Group Donors , Protein Isoforms/genetics , Proteins/genetics , RNA-Binding Proteins , Receptor, Interferon alpha-beta/antagonists & inhibitors , Receptor, Interferon alpha-beta/immunology , Recombinant Proteins , Toll-Like Receptor 9/agonists , Transcription, Genetic/drug effects , Transcription, Genetic/genetics , Ubiquitin Thiolesterase , Ubiquitins/genetics , Ubiquitins/metabolism
12.
Muscle Nerve ; 42(4): 576-83, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20740627

ABSTRACT

Myositis muscle contains antigen-matured B-cells and plasma cells. Myositis muscle biopsy specimens were examined for nodular collections of T-cells, B-cells, myeloid dendritic cells, plasma cells, and follicular dendritic cells. Immunoglobulin and B-cell-activating factor (BAFF) transcripts were quantitated. Laser-capture microdissection was used to isolate single plasma cells, and their immunoglobulin transcripts were sequenced. Dense inflammatory infiltrates contained histological elements of ectopic lymphoid tissue but not B-cell follicles. Immunoglobulin transcript sequence analysis demonstrated spatially distributed, clonally related B-cells and plasma cells, suggesting local maturation of B-cells into plasma cells in myositis muscle. Regions of dense cellular infiltrates in myositis muscle are sometimes areas of B-cell maturation into antibody-producing plasma cells. An atypical lymphoid histology, lacking concentrated collections of germinal-center-like B-cell follicles, is capable of antigen-stimulated clonal maturation of antibody-producing plasma cells.


Subject(s)
B-Lymphocytes , Cellular Senescence , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Myositis/physiopathology , Antigen Presentation , B-Cell Activating Factor/genetics , B-Cell Maturation Antigen/pharmacology , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Cellular Senescence/drug effects , Choristoma/pathology , Dendritic Cells/immunology , Dendritic Cells/pathology , Germinal Center , Humans , Immunoglobulins/genetics , In Vitro Techniques , Lymphoid Tissue , Microdissection/methods , Muscular Diseases/pathology , Plasma Cells/metabolism , Plasma Cells/pathology , RNA, Messenger/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/pathology
13.
Clin Cancer Res ; 26(23): 6284-6298, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32817076

ABSTRACT

PURPOSE: While immune checkpoint inhibitors such as anti-PD-L1 are rapidly becoming the standard of care in the treatment of many cancers, only a subset of treated patients have long-term responses. IL12 promotes antitumor immunity in mouse models; however, systemic recombinant IL12 had significant toxicity and limited efficacy in early clinical trials. EXPERIMENTAL DESIGN: We therefore designed a novel intratumoral IL12 mRNA therapy to promote local IL12 tumor production while mitigating systemic effects. RESULTS: A single intratumoral dose of mouse (m)IL12 mRNA induced IFNγ and CD8+ T-cell-dependent tumor regression in multiple syngeneic mouse models, and animals with a complete response demonstrated immunity to rechallenge. Antitumor activity of mIL12 mRNA did not require NK and NKT cells. mIL12 mRNA antitumor activity correlated with TH1 tumor microenvironment (TME) transformation. In a PD-L1 blockade monotherapy-resistant model, antitumor immunity induced by mIL12 mRNA was enhanced by anti-PD-L1. mIL12 mRNA also drove regression of uninjected distal lesions, and anti-PD-L1 potentiated this response. Importantly, intratumoral delivery of mRNA encoding membrane-tethered mIL12 also drove rejection of uninjected lesions with very limited circulating IL12p70, supporting the hypothesis that local IL12 could induce a systemic antitumor immune response against distal lesions. Furthermore, in ex vivo patient tumor slice cultures, human IL12 mRNA (MEDI1191) induced dose-dependent IL12 production, downstream IFNγ expression and TH1 gene expression. CONCLUSIONS: These data demonstrate the potential for intratumorally delivered IL12 mRNA to promote TH1 TME transformation and robust antitumor immunity.See related commentary by Cirella et al., p. 6080.


Subject(s)
Colorectal Neoplasms/prevention & control , Interleukin-12/administration & dosage , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma/prevention & control , RNA, Messenger/administration & dosage , Th1 Cells/immunology , Tumor Microenvironment/immunology , Animals , Antibodies, Monoclonal/pharmacology , Apoptosis , B7-H1 Antigen/antagonists & inhibitors , CD8-Positive T-Lymphocytes , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm , Female , Humans , Interleukin-12/genetics , Melanoma/genetics , Melanoma/immunology , Melanoma/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Mice, SCID , RNA, Messenger/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
14.
Clin Lung Cancer ; 20(6): e601-e608, 2019 11.
Article in English | MEDLINE | ID: mdl-31327642

ABSTRACT

INTRODUCTION: Two clinical studies (Study 1108 and ATLANTIC) were analyzed to evaluate the prognostic value of baseline liver metastases (LMs) in advanced/metastatic non-small-cell lung cancer patients treated with durvalumab 10 mg/kg every 2 weeks. PATIENTS AND METHODS: A multivariate Cox proportional hazards analysis was conducted; covariates included performance status, tumor stage, histology, sex, age, smoking status, and programmed cell death ligand 1 (PD-L1) status. RESULTS: In all, 569 patients were included. LMs were present in 31.6% (96/304) of Study 1108 patients and 17.9% (47/263) of ATLANTIC patients. Median overall survival (OS) was shorter in patients with LMs than in those without in both studies. In both studies, LMs were an independent negative prognostic factor for OS and progression-free survival. Objective response rates were also significantly lower. PD-L1 independently predicted benefit across all patients. CONCLUSION: Liver metastases were associated with worse outcomes irrespective of PD-L1 status, but PD-L1 status predicted benefit from durvalumab irrespective of LMs.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Carcinoma, Non-Small-Cell Lung/diagnosis , Immunotherapy/methods , Liver Neoplasms/diagnosis , Lung Neoplasms/diagnosis , Aged , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/secondary , Female , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/mortality , Liver Neoplasms/secondary , Lung Neoplasms/drug therapy , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Neoplasm Metastasis , Neoplasm Staging , Patient Selection , Prognosis , Survival Analysis , Treatment Outcome
15.
J Immunother Cancer ; 7(1): 225, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31439037

ABSTRACT

BACKGROUND: The safety, efficacy, pharmacokinetics, and pharmacodynamics of the anti-programmed cell death-1 antibody MEDI0680 were evaluated in a phase I, multicenter, dose-escalation study in advanced solid malignancies. METHODS: MEDI0680 was administered intravenously once every 2 weeks (Q2W) or once every 3 weeks at 0.1, 0.5, 2.5, 10 or 20 mg/kg. Two cohorts received 20 mg/kg once a week for 2 or 4 weeks, then 20 mg/kg Q2W. All were treated for 12 months or until progression. The primary endpoint was safety. Secondary endpoints were efficacy and pharmacokinetics. Exploratory endpoints included pharmacodynamics. RESULTS: Fifty-eight patients were treated. Median age was 62.5 years and 81% were male. Most had kidney cancer (n = 36) or melanoma (n = 9). There were no dose-limiting toxicities. Treatment-related adverse events occurred in 83% and were grade ≥ 3 in 21%. Objective clinical responses occurred in 8/58 patients (14%): 5 with kidney cancer, including 1 with a complete response, and 3 with melanoma. The relationship between dose and serum levels was predictable and linear, with apparent receptor saturation at 10 mg/kg Q2W and all 20 mg/kg cohorts. CONCLUSIONS: MEDI0680 induced peripheral T-cell proliferation and increased plasma IFNγ and associated chemokines regardless of clinical response. CD8+ T-cell tumor infiltration and tumoral gene expression of IFNG, CD8A, CXCL9, and granzyme K (GZMK) were also increased following MEDI0680 administration. TRIAL REGISTRATION: NCT02013804 ; date of registration December 12, 2013.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Melanoma/drug therapy , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Disease Progression , Female , Humans , Male , Middle Aged , Treatment Outcome
16.
Lupus Sci Med ; 5(1): e000261, 2018.
Article in English | MEDLINE | ID: mdl-29644082

ABSTRACT

OBJECTIVE: We investigated the mechanistic and pharmacological properties of anifrolumab, a fully human, effector-null, anti-type I interferon (IFN) alpha receptor 1 (IFNAR1) monoclonal antibody in development for SLE. METHODS: IFNAR1 surface expression and internalisation on human monocytes before and after exposure to anifrolumab were assessed using confocal microscopy and flow cytometry. The effects of anifrolumab on type I IFN pathway activation were assessed using signal transducer and activator of transcription 1 (STAT1) phosphorylation, IFN-stimulated response element-luciferase reporter cell assays and type I IFN gene signature induction. The ability of anifrolumab to inhibit plasmacytoid dendritic cell (pDC) function and plasma cell differentiation was assessed by flow cytometry and ELISA. Effector-null properties of anifrolumab were assessed in antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) assays with B cells. RESULTS: Anifrolumab reduced cell surface IFNAR1 by eliciting IFNAR1 internalisation. Anifrolumab blocked type I IFN-dependent STAT1 phosphorylation and IFN-dependent signalling induced by recombinant and pDC-derived type I IFNs and serum of patients with SLE. Anifrolumab suppressed type I IFN production by blocking the type I IFN autoamplification loop and inhibited proinflammatory cytokine induction and the upregulation of costimulatory molecules on stimulated pDCs. Blockade of IFNAR1 suppressed plasma cell differentiation in pDC/B cell co-cultures. Anifrolumab did not exhibit CDC or ADCC activity. CONCLUSIONS: Anifrolumab potently inhibits type I IFN-dependent signalling, including the type I IFN autoamplification loop, and is a promising therapeutic for patients with SLE and other diseases that exhibit chronic dysfunctional type I IFN signalling.

17.
Sci Rep ; 7(1): 17567, 2017 12 14.
Article in English | MEDLINE | ID: mdl-29242501

ABSTRACT

We aimed to characterize the molecular differences and effects from prednisone treatment among IgG4-related disease with salivary gland lesions (RD-SG), without SG lesions (RD-nonSG), and IgG4-related retroperitoneal fibrosis (RF). RNA sequencing was conducted on blood from 25 RD-SG, 11 RD-nonSG, 3 RF and 10 control subjects. Among these, 8 RD-nonSG and 12 RD-SG patients were subjected to treatment with prednisone and/or glucocorticoid-sparing agents. Six RD patients had a longitudinal time point. The mRNA levels of IgG4 and IgE, genes specific for Th2 cells, eosinophils, and neutrophils were over-expressed in RD-SG and RD-nonSG. A B-cell signature was suppressed in patients group versus controls, while Th1, Th2, Treg, and eosinophil gene signatures were increased in patients without treatment. Interestingly, Tfh genes and B cell signature were decreased at flare disease state. Prednisone treatment led to increased neutrophil, but decreased Treg signatures. Serum IgG4 levels correlated with the eosinophil and neutrophil gene signatures in RD-SG patients, and with a B cell signature in only RD-nonSG patients. IgG4, IgE, and cell-specific signatures are regulated in patients, suggesting the imbalance of immune and inflammatory cells in IgG4-related disease. Prednisone treatment selectively modulates Treg, eosinophil, and neutrophil signatures.


Subject(s)
High-Throughput Nucleotide Sequencing , Immunoglobulin G4-Related Disease/genetics , Sequence Analysis, RNA , Aged , Case-Control Studies , Eosinophils/metabolism , Female , Gene Expression Profiling , Humans , Immunoglobulin E/genetics , Immunoglobulin G/genetics , Immunoglobulin G4-Related Disease/complications , Immunoglobulin G4-Related Disease/immunology , Male , Middle Aged , Neutrophils/metabolism , Principal Component Analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retroperitoneal Fibrosis/complications , Salivary Gland Diseases/complications , T-Lymphocytes, Regulatory/metabolism , Th2 Cells/metabolism
18.
Sci Transl Med ; 8(361): 361ra137, 2016 10 19.
Article in English | MEDLINE | ID: mdl-27798262

ABSTRACT

Autoantibodies can be present years to decades before the onset of disease manifestations in autoimmunity. This finding suggests that the initial autoimmune trigger involves a peripheral lymphoid component, which ultimately drives disease pathology in local tissues later in life. We show that Sjögren's syndrome manifestations that develop in aged NOD.H-2h4 mice were driven by and dependent on peripheral dysregulation that arose in early life. Specifically, elimination of spontaneous germinal centers in spleens of young NOD.H-2h4 mice by transient blockade of CD40 ligand (CD40L) or splenectomy abolished Sjögren's pathology of aged mice. Strikingly, a single injection of anti-CD40L at 4 weeks of age prevented tertiary follicle neogenesis and greatly blunted the formation of key autoantibodies implicated in glandular pathology, including anti-muscarinic receptor antibodies. Microarray profiling of the salivary gland characterized the expression pattern of genes that increased with disease progression and showed that early anti-CD40L greatly repressed B cell function while having a broader effect on multiple biological pathways, including interleukin-12 and interferon signaling. A single prophylactic treatment with anti-CD40L also inhibited the development of autoimmune thyroiditis and diabetes in NOD.H-2h4 and nonobese diabetic mice, respectively, supporting a key role for CD40L in the pathophysiology of several autoimmune models. These results strongly suggest that early peripheral immune dysregulation gives rise to autoimmune manifestations later in life, and for diseases predated by autoantibodies, early prophylactic intervention with biologics may prove efficacious.


Subject(s)
Autoimmunity , CD40 Antigens/metabolism , Immune System/pathology , Aging , Animals , Antibodies, Monoclonal/pharmacology , Autoantibodies/immunology , Bone Marrow Cells/metabolism , CD40 Antigens/genetics , CD40 Ligand/antagonists & inhibitors , CD40 Ligand/immunology , Cytokines/metabolism , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/immunology , Disease Models, Animal , Female , Ligands , Lymphocytes/metabolism , Mice , Mice, Inbred NOD , Salivary Glands/metabolism , Sjogren's Syndrome/genetics , Sjogren's Syndrome/immunology , Spleen/metabolism , Thyroiditis, Autoimmune/genetics , Thyroiditis, Autoimmune/immunology
19.
Arthritis Rheumatol ; 67(12): 3209-18, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26315540

ABSTRACT

OBJECTIVE: Systemic lupus erythematosus (SLE) is characterized by the uncontrolled production of inflammatory cytokines, among which type I interferon (IFN) is recognized as a crucial pathogenic factor. The expression of microRNA-146a (miR-146a) is reduced in the white blood cells of SLE patients and accounts for their overactivated inflammatory responses. However, the mechanism of the reduction of miR-146a is still not fully understood. This study was undertaken to test whether the key pathogenic cytokine, type I IFN, is responsible for the dysregulation of miR-146a in SLE. METHODS: Gene and protein expression was measured in all cells by reverse transcription-quantitative polymerase chain reaction, Northern blotting, or Western blotting. In THP-1 cells, expression of monocyte chemotactic protein-induced protein 1 (MCPIP-1) was knocked down with a lentivirus encoding a short hairpin RNA targeting MCPIP1. The cells were pretreated with type I IFN and assessed for gene expression levels of miR-146a. White blood cells from patients with SLE were analyzed for the expression of the IFN-inducible genes MCPIP1 and miR-146a, and the gene expression data were compared for correlation. RESULTS: Pretreatment of THP-1 cells with type I IFN attenuated the induction of miR-146a posttranscriptionally, by down-regulating the expression of pre-miR-146a but not pri-miR-146a or its original unspliced transcript. Expression of MCPIP-1, which was enhanced by type I IFN, was found to be responsible for the inhibition of miR-146a. In white blood cells from patients with SLE, MCPIP1 expression was elevated, and its expression correlated positively with the IFN score and negatively with the miR-146a transcript level. CONCLUSION: Type I IFN inhibits the maturation of miR-146a through the up-regulation of MCPIP-1, and thus contributes to the uncontrolled inflammation and excessive inflammatory gene expression in SLE.


Subject(s)
Interferon Type I/pharmacology , Lupus Erythematosus, Systemic/genetics , MicroRNAs/drug effects , Monocytes/drug effects , RNA, Messenger/drug effects , Ribonucleases/drug effects , Transcription Factors/drug effects , 2',5'-Oligoadenylate Synthetase/genetics , 2',5'-Oligoadenylate Synthetase/immunology , Antigens, Surface/genetics , Antigens, Surface/immunology , Cell Line , GPI-Linked Proteins/genetics , GPI-Linked Proteins/immunology , Gene Expression , Gene Knockdown Techniques , Humans , Interferon Type I/genetics , Interferon Type I/immunology , Leukocytes/immunology , Lipopolysaccharides/pharmacology , Lupus Erythematosus, Systemic/immunology , MicroRNAs/genetics , MicroRNAs/immunology , Monocytes/immunology , Myxovirus Resistance Proteins/genetics , Myxovirus Resistance Proteins/immunology , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Ribonucleases/genetics , Ribonucleases/immunology , Transcription Factors/genetics , Transcription Factors/immunology , Up-Regulation/drug effects
20.
Stem Cells Transl Med ; 2(1): 43-52, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23283488

ABSTRACT

Although cancer is largely seen as a disease stemming from genetic mutations, evidence has implicated epigenetic regulation of gene expression as a driving force for tumorigenesis. Epigenetic regulation by histone modification, specifically through polycomb group (PcG) proteins such as EZH2 and BMI-1, is a major driver in stem cell biology and is found to be correlated with poor prognosis in many tumor types. This suggests a role for PcG proteins in cancer stem cells (CSCs). We hypothesized that epigenetic modification by EZH2, specifically, helps maintain the CSC phenotype and that in turn this epigenetic modifier can be used as a reporter for CSC activity in an in vitro high-throughput screening assay. CSCs isolated from pancreatic and breast cancer lines had elevated EZH2 levels over non-CSCs. Moreover, EZH2 knockdown by RNA interference significantly reduced the frequency of CSCs in all models tested, confirming the role of EZH2 in maintenance of the CSC population. Interestingly, genes affected by EZH2 loss, and therefore CSC loss, were inversely correlated with genes identified by CSC enrichment, further supporting the function of EZH2 CSC regulation. We translated these results into a novel assay whereby elevated EZH2 staining was used as a reporter for CSCs. Data confirmed that this assay could effectively measure changes, both inhibition and enrichment, in the CSC population, providing a novel approach to look at CSC activity. This assay provides a unique, rapid way to facilitate CSC screening across several tumor types to aid in further CSC-related research.


Subject(s)
Biomarkers, Tumor/metabolism , Neoplastic Stem Cells/metabolism , Polycomb Repressive Complex 2/metabolism , Biomarkers, Tumor/genetics , Breast Neoplasms , Cell Line, Tumor , Chromatin Immunoprecipitation , Enhancer of Zeste Homolog 2 Protein , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Histones/metabolism , Humans , Neoplastic Stem Cells/physiology , Oligonucleotide Array Sequence Analysis , Pancreatic Neoplasms , Polycomb Repressive Complex 2/genetics , RNA Interference , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL