Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
3.
Mol Cancer Ther ; 21(3): 427-439, 2022 03 01.
Article in English | MEDLINE | ID: mdl-34965960

ABSTRACT

Targeting the programmed death 1/programmed death ligand 1 (PD-1/PD-L1) pathway with immunotherapy has revolutionized the treatment of many cancers. Somatic tumor mutational burden (TMB) and T-cell-inflamed gene expression profile (GEP) are clinically validated pan-tumor genomic biomarkers that can predict responsiveness to anti-PD-1/PD-L1 monotherapy in many tumor types. We analyzed the association between these biomarkers and the efficacy of PD-1 inhibitor in 11 commonly used preclinical syngeneic tumor mouse models using murinized rat anti-mouse PD-1 DX400 antibody muDX400, a surrogate for pembrolizumab. Response to muDX400 treatment was broadly classified into three categories: highly responsive, partially responsive, and intrinsically resistant to therapy. Molecular and cellular profiling validated differences in immune cell infiltration and activation in the tumor microenvironment of muDX400-responsive tumors. Baseline and on-treatment genomic analysis showed an association between TMB, murine T-cell-inflamed gene expression profile (murine-GEP), and response to muDX400 treatment. We extended our analysis to investigate a canonical set of cancer and immune biology-related gene signatures, including signatures of angiogenesis, myeloid-derived suppressor cells, and stromal/epithelial-to-mesenchymal transition/TGFß biology previously shown to be inversely associated with the clinical efficacy of immune checkpoint blockade. Finally, we evaluated the association between murine-GEP and preclinical efficacy with standard-of-care chemotherapy or antiangiogenic agents that previously demonstrated promising clinical activity, in combination with muDX400. Our profiling studies begin to elucidate the underlying biological mechanisms of response and resistance to PD-1/PD-L1 blockade represented by these models, thereby providing insight into which models are most appropriate for the evaluation of orthogonal combination strategies.


Subject(s)
B7-H1 Antigen , Immunotherapy , Neoplasms , Programmed Cell Death 1 Receptor , Animals , B7-H1 Antigen/antagonists & inhibitors , Biomarkers, Tumor/genetics , Cell Line, Tumor , Disease Models, Animal , Humans , Immune Checkpoint Inhibitors , Mice , Neoplasms/drug therapy , Neoplasms/genetics , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Tumor Microenvironment
4.
Clin Cancer Res ; 28(1): 57-70, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34598945

ABSTRACT

PURPOSE: In this first-in-human study (NCT03564691) in advanced solid tumors, we investigated a novel first-in-class human IgG4 monoclonal antibody targeting the immunoglobulin-like transcript 4 (ILT4) receptor, MK-4830, as monotherapy and in combination with pembrolizumab. PATIENTS AND METHODS: Patients with histologically/cytologically confirmed advanced solid tumors, measurable disease by RECIST v1.1, and evaluable baseline tumor sample received escalating doses of intravenous MK-4830 every 3 weeks as monotherapy (parts A and B) and in combination with pembrolizumab (part C). Safety and tolerability were the primary objectives. Pharmacokinetics, objective response rate per RECIST v1.1, and molecular biomarkers were also evaluated. RESULTS: Of 84 patients, 50 received monotherapy and 34 received combination therapy. No dose-limiting toxicities were observed; maximum tolerated dose was not reached. MK-4830 showed dose-related target engagement. Eleven of 34 patients in the dose-escalation phase who received combination therapy achieved objective responses; 5 previously had progressive disease on anti-PD-1/PD-L1 therapies. Exploratory evaluation of the association between response and pretreatment gene expression related to interferon-gamma signaling in tumors suggested higher sensitivity to T-cell inflammation with combination therapy than historically expected with pembrolizumab monotherapy, with greater response at more moderate levels of inflammation. CONCLUSIONS: This first-in-class MK-4830 antibody dosed as monotherapy and in combination with pembrolizumab was well tolerated with no unexpected toxicities, and demonstrated dose-related evidence of target engagement and antitumor activity. Inflammation intrinsic to the ILT4 mechanism may be facilitated by alleviating the myeloid-suppressive components of the tumor microenvironment, supporting the target of ILT4 as a potential novel immunotherapy in combination with an anti-PD-1/PD-L1 agent.


Subject(s)
Neoplasms , Programmed Cell Death 1 Receptor , Antibodies, Monoclonal , Humans , Maximum Tolerated Dose , Neoplasms/drug therapy , Neoplasms/genetics , Response Evaluation Criteria in Solid Tumors , Tumor Microenvironment
5.
Immunity ; 49(2): 342-352.e5, 2018 08 21.
Article in English | MEDLINE | ID: mdl-30097293

ABSTRACT

Interleukin-22 (IL-22)-producing group 3 innate lymphoid cells (ILC3) maintains gut homeostasis but can also promote inflammatory bowel disease (IBD). The regulation of ILC3-dependent colitis remains to be elucidated. Here we show that Foxp3+ regulatory T cells (Treg cells) prevented ILC3-mediated colitis in an IL-10-independent manner. Treg cells inhibited IL-23 and IL-1ß production from intestinal-resident CX3CR1+ macrophages but not CD103+ dendritic cells. Moreover, Treg cells restrained ILC3 production of IL-22 through suppression of CX3CR1+ macrophage production of IL-23 and IL-1ß. This suppression was contact dependent and was mediated by latent activation gene-3 (LAG-3)-an immune checkpoint receptor-expressed on Treg cells. Engagement of LAG-3 on MHC class II drove profound immunosuppression of CX3CR1+ tissue-resident macrophages. Our study reveals that the health of the intestinal mucosa is maintained by an axis driven by Treg cells communication with resident macrophages that withhold inflammatory stimuli required for ILC3 function.


Subject(s)
Antigens, CD/metabolism , CX3C Chemokine Receptor 1/metabolism , Colitis/immunology , Colitis/pathology , Interleukin-23 Subunit p19/immunology , Intestinal Mucosa/pathology , Macrophages/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cells, Cultured , Dendritic Cells/immunology , Forkhead Transcription Factors/metabolism , Histocompatibility Antigens Class II/immunology , Interleukin-10/immunology , Interleukin-1beta/immunology , Interleukins/immunology , Intestinal Mucosa/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes, Regulatory/transplantation , Lymphocyte Activation Gene 3 Protein , Interleukin-22
6.
Nat Biotechnol ; 35(10): 936-939, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28854175

ABSTRACT

We present a tool to measure gene and protein expression levels in single cells with DNA-labeled antibodies and droplet microfluidics. Using the RNA expression and protein sequencing assay (REAP-seq), we quantified proteins with 82 barcoded antibodies and >20,000 genes in a single workflow. We used REAP-seq to assess the costimulatory effects of a CD27 agonist on human CD8+ lymphocytes and to identify and characterize an unknown cell type.


Subject(s)
Proteins/metabolism , Single-Cell Analysis/methods , CD4-Positive T-Lymphocytes/metabolism , Humans , Lymphocyte Activation/immunology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Analysis, Protein , Sequence Analysis, RNA
7.
Cancer Res ; 77(16): 4378-4388, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28611044

ABSTRACT

GITR is a T-cell costimulatory receptor that enhances cellular and humoral immunity. The agonist anti-mouse GITR antibody DTA-1 has demonstrated efficacy in murine models of cancer primarily by attenuation of Treg-mediated immune suppression, but the translatability to human GITR biology has not been fully explored. Here, we report the potential utility of MK-4166, a humanized GITR mAb selected to bind to an epitope analogous to the DTA-1 epitope, which enhances the proliferation of both naïve and tumor-infiltrating T lymphocytes (TIL). We also investigated the role of GITR agonism in human antitumor immune responses and report here the preclinical characterization and toxicity assessment of MK-4166, which is currently being evaluated in a phase I clinical study. Expression of human GITR was comparable with that of mouse GITR in tumor-infiltrating Tregs despite being drastically lower in other human TILs and in many human peripheral blood populations. MK-4166 decreased induction and suppressive effects of Tregsin vitro In human TIL cultures, MK-4166 induced phosphorylation of NFκB and increased expression of dual specificity phosphatase 6 (DUSP6), indicating that MK-4166 activated downstream NFκB and Erk signaling pathways. Furthermore, MK-4166 downregulated FOXP3 mRNA in human tumor infiltrating Tregs, suggesting that, in addition to enhancing the activation of TILs, MK-4166 may attenuate the Treg-mediated suppressive tumor microenvironment. Cancer Res; 77(16); 4378-88. ©2017 AACR.


Subject(s)
Antibodies/pharmacology , Glucocorticoid-Induced TNFR-Related Protein/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antibodies/immunology , Cell Line, Tumor , Female , Glucocorticoid-Induced TNFR-Related Protein/agonists , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred DBA , Tumor Microenvironment
8.
PLoS Pathog ; 6(3): e1000815, 2010 Mar 19.
Article in English | MEDLINE | ID: mdl-20333242

ABSTRACT

Production of the pro-inflammatory cytokine IL-12 by innate phagocytes drives the differentiation of IFN-gamma-producing effector T cells during Toxoplasma gondii infection. However, the role of IL-12 in the regulation of memory CD8+ T cell differentiation and function during murine toxoplasmosis is unclear. To track memory CTL development, we identified a novel H-2K(b)-restricted CTL population specific for the Toxoplasma antigen tgd057. Tgd057-specific CTLs were induced by both vaccination and natural peroral infection, and were representative of the polyclonal CTL population. Tgd057-specific primary effector cells required IL-12 for the differentiation of KLRG1+ effector subpopulations and IFN-gamma production in response to restimulation with parasite-infected cells, but not to restimulation with cognate peptide. The effect of IL-12 deficiency during the primary response was profoundly imprinted on memory CTLs, which continued to show defects in cell numbers, KLRG1+ effector memory subpopulation differentiation, and IFN-gamma recall responses. Importantly, isolated CD62L(hi) KLRG1- CD8+ T cells differentiated in the absence of IL-12 were enhanced in their ability to generate IFN-gamma-producing secondary tgd057-specific effector cells. Our data, for the first time, demonstrate the negative impact of IL-12 signaling on the quality of the central memory CTL compartment. Thus, despite the beneficial role of IL-12 in promoting effector differentiation, excessive exposure to IL-12 during CTL priming may limit the development of long-term protective immunity through the decreased fitness of central memory CTL responses.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/parasitology , Immunologic Memory/immunology , Interleukin-12/immunology , Toxoplasma/immunology , Toxoplasmosis, Animal/immunology , Animals , Antigens, Protozoan/immunology , Cell Differentiation/immunology , Epitopes , Immunophenotyping , Interleukin-12/genetics , Interleukin-12/metabolism , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Signal Transduction/immunology
9.
Shock ; 34(2): 150-61, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20023601

ABSTRACT

Increased apoptotic cell death is believed to play a pathological role in patients with sepsis and experimental animals. Apoptosis can be induced by either a cell death receptor (extrinsic) or a mitochondrial (intrinsic) pathway. Bid, a proapoptotic member of the Bcl-2 family, is thought to mediate the cross talk between the extrinsic and intrinsic pathways of apoptosis; however, little is known about the action of Bid in the development of apoptosis and organ-specific tissue damage/cell death as seen in polymicrobial sepsis. Our results show that after the onset of sepsis, tBid (the active form of Bid) is significantly increased in mitochondrial fractions of the thymus, spleen, Peyer patches, and liver, and that Fas or FasL deficiency blocks Bid activation in various tissues after septic challenge. Increased Bid activation is correlated with increased active caspase-3, caspase-9, and apoptosis during sepsis. Bid-deficient mice exhibit significantly reduced apoptosis in the thymus, spleen, and Peyer patches compared with background mice after sepsis. Furthermore, Bid-deficient mice had significantly reduced systemic and local inflammatory cytokine levels and improved survival after sepsis. These data support not only the contribution of Bid to sepsis-induced apoptosis and the onset of septic morbidity/mortality, but also the existence of a bridge between extrinsic apoptotic signals, e.g., FasL:Fas, TNF:TNFR, and so on, and the intrinsic mitochondrial pathway via Bid-tBid activation during sepsis.


Subject(s)
Apoptosis/physiology , BH3 Interacting Domain Death Agonist Protein/deficiency , Sepsis/immunology , Alanine Transaminase/blood , Animals , BH3 Interacting Domain Death Agonist Protein/physiology , Caspase 9/metabolism , Cytokines/metabolism , Cytosol/metabolism , Fas Ligand Protein/physiology , Inflammation/immunology , Liver/immunology , Male , Mice , Mitochondria/metabolism , Protein Transport , Receptors, Tumor Necrosis Factor/physiology , Sepsis/mortality , Sepsis/pathology , Signal Transduction/physiology , Spleen/immunology
10.
J Immunol ; 182(6): 3775-81, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19265156

ABSTRACT

Cytokine-activated macrophages restrain the replication of intracellular parasites and disrupt the integrity of vacuolar pathogens. In this study, we show that inducible nitric oxide synthase and the immunity-related GTPase (IRG) family member Irgm3, respectively, are required for the ability of in vivo primed macrophages to restrain the growth of Toxoplasma gondii and to destroy the parasite's intracellular niche. Remarkably, virulent Type I strains of T. gondii evade IRG-dependent vacuolar disruption, while remaining susceptible to iNOS-dependent restriction. The ability of virulent T. gondii to escape killing by macrophages is controlled at the level of the individual vacuole and is associated with differential permissiveness for association of the IRG proteins Irga6 (IIGP1) and Irgb6 (TGTP) to the vacuolar membrane. Surprisingly, expression of the Type I ROP-18 virulence determinant in an avirulent strain did not confer the evasive phenotype. These results pinpoint evasion of vacuolar disruption by IRG proteins as a new determinant of pathogen virulence.


Subject(s)
GTP Phosphohydrolases/physiology , GTP-Binding Proteins/physiology , Macrophage Activation/immunology , Toxoplasma/immunology , Toxoplasma/pathogenicity , Vacuoles/enzymology , Vacuoles/immunology , Animals , GTP Phosphohydrolases/deficiency , GTP Phosphohydrolases/genetics , Macrophages, Peritoneal/enzymology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/parasitology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nitric Oxide Synthase Type II/deficiency , Nitric Oxide Synthase Type II/genetics , Toxoplasma/growth & development , Vacuoles/parasitology , Virulence/immunology
11.
J Immunol ; 180(9): 5935-45, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18424713

ABSTRACT

IFN-gamma-producing CD8(+) T lymphocytes are essential effector cells that mediate protective immunity during murine toxoplasmosis, and yet their effector development remains poorly characterized. Vaccination with the carbamoyl phosphate synthase (CPS) mutant strain of Toxoplasma gondii was used to examine the CD8(+) T cell response in the peritoneal effector site. Four CTL subpopulations with varying effector potentials were defined based on the expression of effector molecules and the cell surface activation markers CD62L and killer cell lectin-like receptor G1 (KLRG1). Further phenotypic analysis revealed that the acquisition of KLRG1 among effector subpopulations correlated with the down-regulation of both IL-7R and CD27, suggesting that KLRG1 marks dominant, end-stage effector cells. Using gene-targeted mice, we tested the in vivo requirements of key IL-12 signaling components for effector CTL differentiation. Contrary to established models of viral and bacterial infection, CD8(+) T cell-intrinsic IL-12 signaling was required for the generation of IFN-gamma-producing CTLs in response to T. gondii. Importantly, the development of the KLRG1(+) effector subpopulations, but not the memory precursor-containing KLRG1(-) effector subset, was critically reliant on IL-12. Furthermore, IL-12 signaling-dependent T-bet expression was also found to be important for differentiation of KLRG1(+) effectors. Our results underscore a vital role for IL-12 in not only the induction of IFN-gamma expression but also in the development of heterogeneous subpopulations of effector CD8(+) T cells generated in response to the intracellular parasite T. gondii.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Interferon-gamma/immunology , Interleukin-12/immunology , Receptors, Immunologic/immunology , Toxoplasma/immunology , Toxoplasmosis, Animal/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/parasitology , Carbon-Nitrogen Ligases/genetics , Carbon-Nitrogen Ligases/immunology , Carbon-Nitrogen Ligases/metabolism , Cell Differentiation/genetics , Down-Regulation/genetics , Down-Regulation/immunology , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interleukin-12/genetics , Interleukin-12/metabolism , L-Selectin/genetics , L-Selectin/immunology , L-Selectin/metabolism , Lectins, C-Type , Mice , Mice, Knockout , Mutation , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Protozoan Proteins/metabolism , Protozoan Vaccines/genetics , Protozoan Vaccines/immunology , Protozoan Vaccines/metabolism , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Receptors, Interleukin-7/genetics , Receptors, Interleukin-7/immunology , Receptors, Interleukin-7/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , Toxoplasma/enzymology , Toxoplasmosis, Animal/enzymology , Toxoplasmosis, Animal/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 7/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...