Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
J Neuroinflammation ; 17(1): 24, 2020 Jan 17.
Article in English | MEDLINE | ID: mdl-31952519

ABSTRACT

BACKGROUND: Individuals with impaired immunity are more susceptible to infections than immunocompetent subjects. No vaccines are currently available to induce protection against E. coli meningoencephalitis. This study evaluated the potential of poly(I:C) pre-treatment to induce trained immunity. Poly(I:C) was administered as a non-specific stimulus of innate immune responses to protect immunocompetent and neutropenic wild-type mice from a subsequent challenge by the intracranial injection of E. coli K1. METHODS: Three days prior to infection, mice received an intraperitoneal injection of poly(I:C) or vehicle. Kaplan-Meier survival curves were analyzed. In short-term experiments, bacterial titers and the inflammatory response were characterized in the blood, cerebellum, and spleen homogenates. NK cell subpopulations in the brain and spleen were analyzed by flow cytometry. Numbers of microglia and activation scores were evaluated by histopathology. RESULTS: Pre-treatment with 200 µg poly(I:C) increased survival time, reduced mortality, and enhanced bacterial clearance in the blood, cerebellum, and spleen at early infection in neutropenic mice. Poly(I:C)-mediated protection correlated with an augmented number of NK cells (CD45+NK1.1+CD3-) and Iba-1+ microglial cells and a higher production of IFN-γ in the brain. In the spleen, levels of CCL5/RANTES and IFN-γ were increased and sustained in surviving poly(I:C)-treated animals for 14 days after infection. In immunocompetent animals, survival time was not significantly prolonged in poly(I:C)-treated animals although poly(I:C) priming reduced brain bacterial concentrations compared with vehicle-injected animals at early infection. CONCLUSIONS: Pre-treatment with the viral TLR3 agonist poly(I:C) modulated innate immune responses and strengthened the resistance of neutropenic mice against E. coli K1 meningoencephalitis.


Subject(s)
Immunity, Innate/drug effects , Immunocompromised Host/immunology , Meningitis, Escherichia coli/immunology , Poly I-C/pharmacology , Animals , Immunity, Innate/immunology , Male , Mice , Mice, Inbred C57BL , Neutropenia/immunology , Poly I-C/immunology , Toll-Like Receptor 3/agonists , Toll-Like Receptor 3/drug effects
2.
Nature ; 512(7514): 324-7, 2014 Aug 21.
Article in English | MEDLINE | ID: mdl-25043048

ABSTRACT

Monoallelic point mutations of isocitrate dehydrogenase type 1 (IDH1) are an early and defining event in the development of a subgroup of gliomas and other types of tumour. They almost uniformly occur in the critical arginine residue (Arg 132) in the catalytic pocket, resulting in a neomorphic enzymatic function, production of the oncometabolite 2-hydroxyglutarate (2-HG), genomic hypermethylation, genetic instability and malignant transformation. More than 70% of diffuse grade II and grade III gliomas carry the most frequent mutation, IDH1(R132H) (ref. 3). From an immunological perspective, IDH1(R132H) represents a potential target for immunotherapy as it is a tumour-specific potential neoantigen with high uniformity and penetrance expressed in all tumour cells. Here we demonstrate that IDH1(R132H) contains an immunogenic epitope suitable for mutation-specific vaccination. Peptides encompassing the mutated region are presented on major histocompatibility complexes (MHC) class II and induce mutation-specific CD4(+) T-helper-1 (TH1) responses. CD4(+) TH1 cells and antibodies spontaneously occurring in patients with IDH1(R132H)-mutated gliomas specifically recognize IDH1(R132H). Peptide vaccination of mice devoid of mouse MHC and transgenic for human MHC class I and II with IDH1(R132H) p123-142 results in an effective MHC class II-restricted mutation-specific antitumour immune response and control of pre-established syngeneic IDH1(R132H)-expressing tumours in a CD4(+) T-cell-dependent manner. As IDH1(R132H) is present in all tumour cells of these slow-growing gliomas, a mutation-specific anti-IDH1(R132H) vaccine may represent a viable novel therapeutic strategy for IDH1(R132H)-mutated tumours.


Subject(s)
Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Glioma/immunology , Glioma/therapy , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/immunology , Mutant Proteins/immunology , Animals , Antibody Specificity , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Female , Glioma/enzymology , Glioma/genetics , Histocompatibility Antigens Class II/immunology , Humans , Immunity, Humoral , Immunotherapy/methods , Male , Mice , Mutant Proteins/genetics , Mutation , T-Lymphocytes, Helper-Inducer/immunology , Xenograft Model Antitumor Assays
3.
J Neuroinflammation ; 16(1): 49, 2019 Feb 26.
Article in English | MEDLINE | ID: mdl-30808363

ABSTRACT

BACKGROUND: Quinoline-3-carboxamides, such as laquinimod, ameliorate CNS autoimmunity in patients and reduce tumor cell metastasis experimentally. Previous studies have focused on the immunomodulatory effect of laquinimod on myeloid cells. The data contained herein suggest that quinoline-3-carboxamides improve the immunomodulatory and anti-tumor effects of NK cells by upregulating the adhesion molecule DNAX accessory molecule-1 (DNAM-1). METHODS: We explored how NK cell activation by laquinimod inhibits CNS autoimmunity in experimental autoimmune encephalomyelitis (EAE), the most utilized model of MS, and improves immunosurveillance of experimental lung melanoma metastasis. Functional manipulations included in vivo NK and DC depletion experiments and in vitro assays of NK cell function. Clinical, histological, and flow cytometric read-outs were assessed. RESULTS: We demonstrate that laquinimod activates natural killer (NK) cells via the aryl hydrocarbon receptor and increases their DNAM-1 cell surface expression. This activation improves the cytotoxicity of NK cells against B16F10 melanoma cells and augments their immunoregulatory functions in EAE by interacting with CD155+ dendritic cells (DC). Noteworthy, the immunosuppressive effect of laquinimod-activated NK cells was due to decreasing MHC class II antigen presentation by DC and not by increasing DC killing. CONCLUSIONS: This study clarifies how DNAM-1 modifies the bidirectional crosstalk of NK cells with CD155+ DC, which can be exploited to suppress CNS autoimmunity and strengthen tumor surveillance.


Subject(s)
Antigens, Differentiation, T-Lymphocyte/metabolism , Autoimmunity/drug effects , Dendritic Cells/drug effects , Immunologic Surveillance/immunology , Killer Cells, Natural/drug effects , Quinolones/pharmacology , Animals , Antigens, Differentiation, T-Lymphocyte/immunology , Autoimmunity/immunology , Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Humans , Killer Cells, Natural/immunology , Lymphocyte Activation/drug effects , Melanoma, Experimental , Mice , Mice, Inbred C57BL , Mice, Transgenic , Quinolines/agonists , Receptors, Aryl Hydrocarbon/agonists , Receptors, Virus/immunology
4.
Proteomics ; 18(12): e1700284, 2018 06.
Article in English | MEDLINE | ID: mdl-29505699

ABSTRACT

Immunotherapy is revolutionizing cancer treatment and has shown success in particular for tumors with a high mutational load. These effects have been linked to neoantigens derived from patient-specific mutations. To expand efficacious immunotherapy approaches to the vast majority of tumor types and patient populations carrying only a few mutations and maybe not a single presented neoepitope, it is necessary to expand the target space to non-mutated cancer-associated antigens. Mass spectrometry enables the direct and unbiased discovery and selection of tumor-specific human leukocyte antigen (HLA) peptides that can be used to define targets for immunotherapy. Combining these targets into a warehouse allows for multi-target therapy and accelerated clinical application. For precise personalization aimed at optimally ensuring treatment efficacy and safety, it is necessary to assess the presence of the target on each individual patient's tumor. Here we show how LC-MS paired with gene expression data was used to define mRNA biomarkers currently being used as diagnostic test IMADETECT™ for patient inclusion and personalized target selection within two clinical trials (NCT02876510, NCT03247309). Thus, we present a way how to translate HLA peptide presentation into gene expression thresholds for companion diagnostics in immunotherapy considering the peptide-specific correlation to its encoding mRNA.


Subject(s)
Antigens, Neoplasm/metabolism , HLA Antigens/metabolism , Immunotherapy , Neoplasms/metabolism , Peptide Fragments/metabolism , Precision Medicine , Proteogenomics/methods , Antigen Presentation/immunology , Antigens, Neoplasm/analysis , Antigens, Neoplasm/immunology , Decision Making , Epitopes/immunology , Epitopes/metabolism , HLA Antigens/analysis , HLA Antigens/immunology , High-Throughput Nucleotide Sequencing , Humans , Mass Spectrometry/methods , Neoplasms/immunology , Peptide Fragments/analysis , Peptide Fragments/immunology , RNA, Messenger/analysis , RNA, Messenger/genetics , RNA, Messenger/immunology
5.
Acta Neuropathol ; 133(4): 629-644, 2017 04.
Article in English | MEDLINE | ID: mdl-28124097

ABSTRACT

Mutations in codon 132 of isocitrate dehydrogenase (IDH) 1 are frequent in diffuse glioma, acute myeloid leukemia, chondrosarcoma and intrahepatic cholangiocarcinoma. These mutations result in a neomorphic enzyme specificity which leads to a dramatic increase of intracellular D-2-hydroxyglutarate (2-HG) in tumor cells. Therefore, mutant IDH1 protein is a highly attractive target for inhibitory drugs. Here, we describe the development and properties of BAY 1436032, a pan-inhibitor of IDH1 protein with different codon 132 mutations. BAY 1436032 strongly reduces 2-HG levels in cells carrying IDH1-R132H, -R132C, -R132G, -R132S and -R132L mutations. Cells not carrying IDH mutations were unaffected. BAY 1436032 did not exhibit toxicity in vitro or in vivo. The pharmacokinetic properties of BAY 1436032 allow for oral administration. In two independent experiments, BAY 1436032 has been shown to significantly prolong survival of mice intracerebrally transplanted with human astrocytoma carrying the IDH1R132H mutation. In conclusion, we developed a pan-inhibitor targeting tumors with different IDH1R132 mutations.


Subject(s)
Aniline Compounds/pharmacology , Antineoplastic Agents/pharmacology , Astrocytoma/drug therapy , Benzimidazoles/pharmacology , Brain Neoplasms/drug therapy , Isocitrate Dehydrogenase/antagonists & inhibitors , Isocitrate Dehydrogenase/genetics , Aniline Compounds/chemistry , Aniline Compounds/pharmacokinetics , Aniline Compounds/toxicity , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/toxicity , Astrocytoma/enzymology , Astrocytoma/genetics , Benzimidazoles/chemistry , Benzimidazoles/pharmacokinetics , Benzimidazoles/toxicity , Brain Neoplasms/enzymology , Brain Neoplasms/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/enzymology , Colonic Neoplasms/genetics , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/toxicity , Escherichia coli , Female , Glutarates/metabolism , HEK293 Cells , Humans , Isocitrate Dehydrogenase/metabolism , Mice, Inbred BALB C , Mice, Nude , Mutation , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sarcoma/drug therapy , Sarcoma/enzymology , Sarcoma/genetics , Sf9 Cells , Xenograft Model Antitumor Assays
6.
Nature ; 478(7368): 197-203, 2011 Oct 05.
Article in English | MEDLINE | ID: mdl-21976023

ABSTRACT

Activation of the aryl hydrocarbon receptor (AHR) by environmental xenobiotic toxic chemicals, for instance 2,3,7,8-tetrachlorodibenzo-p-dioxin (dioxin), has been implicated in a variety of cellular processes such as embryogenesis, transformation, tumorigenesis and inflammation. But the identity of an endogenous ligand activating the AHR under physiological conditions in the absence of environmental toxic chemicals is still unknown. Here we identify the tryptophan (Trp) catabolite kynurenine (Kyn) as an endogenous ligand of the human AHR that is constitutively generated by human tumour cells via tryptophan-2,3-dioxygenase (TDO), a liver- and neuron-derived Trp-degrading enzyme not yet implicated in cancer biology. TDO-derived Kyn suppresses antitumour immune responses and promotes tumour-cell survival and motility through the AHR in an autocrine/paracrine fashion. The TDO-AHR pathway is active in human brain tumours and is associated with malignant progression and poor survival. Because Kyn is produced during cancer progression and inflammation in the local microenvironment in amounts sufficient for activating the human AHR, these results provide evidence for a previously unidentified pathophysiological function of the AHR with profound implications for cancer and immune biology.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Glioma/metabolism , Glioma/pathology , Kynurenine/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Animals , Autocrine Communication , Brain Neoplasms/genetics , Brain Neoplasms/immunology , Cell Line, Tumor , Cell Survival , Disease Progression , Gene Expression Regulation, Neoplastic , Glioma/genetics , Glioma/immunology , Humans , Kynurenine/immunology , Kynurenine/pharmacology , Ligands , Mice , Mice, Inbred C57BL , Mice, Nude , Neoplasm Transplantation , Paracrine Communication , Receptors, Aryl Hydrocarbon/immunology , Tryptophan/metabolism , Tryptophan Oxygenase/deficiency , Tryptophan Oxygenase/genetics , Tryptophan Oxygenase/metabolism
7.
J Neurochem ; 136(6): 1142-1154, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26708701

ABSTRACT

Malignant gliomas and other types of tumors generate a local immunosuppressive microenvironment, which prohibits an effective anti-tumor immune response and promotes tumor growth. Along with others, we have recently demonstrated that catabolism of the essential amino acid tryptophan via tryptophan-2,3-dioxygenase (TDO) is an important mechanism mediating tumor-associated immunosuppression particularly in gliomas. The pathways regulating TDO in tumors, however, are poorly understood. Here, we show that prostaglandins enhance TDO expression and enzymatic activity in malignant gliomas via activation of prostaglandin E receptor-4 (EP4). Stimulation with prostaglandin E2 (PGE2 ) up-regulated TDO-mediated kynurenine release in human glioma cell lines, whereas knockdown of the PGE2 receptor EP4 inhibited TDO expression and activity. In human malignant glioma tissue expression of the PGE2 -producing enzyme cyclooxygenase-2 (COX2) and its receptor EP4 were associated with TDO expression both on transcript and protein level. High expression of EP4 correlated with poor survival in malignant glioma patients WHO III-IV. Importantly, treatment of glioma cells with an EP4 inhibitor decreased TDO expression and activity. Moreover, TDO-over-expressing murine gliomas showed increased COX2 and EP4 expression suggesting a positive feedback mechanism in vivo. In summary, targeting EP4 may inhibit - in addition to immunosuppressive COX2 signaling - tryptophan degradation as another important immunosuppressive pathway and thus, could provide a dual clinically relevant immunotherapeutic avenue for the treatment of malignant gliomas. We proposed that in malignant gliomas prostaglandin E2 (PGE2 ) produced by cyclooxygenases (COX) up-regulates tryptophan-2,3-dioxygenase (TDO) expression and enzyme activity through binding to its Gs-coupled receptor EP4 and therefore may mediate tumor immune escape in part through aryl hydrocarbon receptor (AHR) activation. Moreover, TDO activity itself seems to induce intratumoral PGE2 metabolism suggesting an immunosuppressive loop involving COX/EP4/TDO.

8.
Proc Natl Acad Sci U S A ; 110(36): 14735-40, 2013 Sep 03.
Article in English | MEDLINE | ID: mdl-23959874

ABSTRACT

Disruption of the blood-brain barrier (BBB) is a hallmark of acute inflammatory lesions in multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis. This disruption may precede and facilitate the infiltration of encephalitogenic T cells. The signaling events that lead to this BBB disruption are incompletely understood but appear to involve dysregulation of tight-junction proteins such as claudins. Pharmacological interventions aiming at stabilizing the BBB in MS might have therapeutic potential. Here, we show that the orally available small molecule LY-317615, a synthetic bisindolylmaleimide and inhibitor of protein kinase Cß, which is clinically under investigation for the treatment of cancer, suppresses the transmigration of activated T cells through an inflamed endothelial cell barrier, where it leads to the induction of the tight-junction molecules zona occludens-1, claudin 3, and claudin 5 and other pathways critically involved in transendothelial leukocyte migration. Treatment of mice with ongoing experimental autoimmune encephalomyelitis with LY-317615 ameliorates inflammation, demyelination, axonal damage, and clinical symptoms. Although LY-317615 dose-dependently suppresses T-cell proliferation and cytokine production independent of antigen specificity, its therapeutic effect is abrogated in a mouse model requiring pertussis toxin. This abrogation indicates that the anti-inflammatory and clinical efficacy is mainly mediated by stabilization of the BBB, thus suppressing the transmigration of encephalitogenic T cells. Collectively, our data suggest the involvement of endothelial protein kinase Cß in stabilizing the BBB in autoimmune neuroinflammation and imply a therapeutic potential of BBB-targeting agents such as LY-317615 as therapeutic approaches for MS.


Subject(s)
Blood-Brain Barrier/drug effects , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Indoles/pharmacology , Protein Kinase C beta/antagonists & inhibitors , Animals , Blood-Brain Barrier/immunology , Cell Proliferation/drug effects , Claudin-3/immunology , Claudin-3/metabolism , Claudin-5/immunology , Claudin-5/metabolism , Cytokines/immunology , Cytokines/metabolism , Demyelinating Diseases/immunology , Demyelinating Diseases/prevention & control , Dose-Response Relationship, Drug , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Endothelial Cells/drug effects , Endothelial Cells/immunology , Endothelial Cells/metabolism , Female , Gene Expression Profiling , Immunohistochemistry , Indoles/immunology , Inflammation/immunology , Inflammation/metabolism , Inflammation/prevention & control , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Microscopy, Confocal , Protein Kinase C beta/immunology , Protein Kinase C beta/metabolism , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tight Junctions/drug effects , Tight Junctions/immunology , Tight Junctions/metabolism , Transendothelial and Transepithelial Migration/drug effects , Transendothelial and Transepithelial Migration/immunology , Zonula Occludens-1 Protein/immunology , Zonula Occludens-1 Protein/metabolism
9.
Glia ; 63(1): 78-90, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25132599

ABSTRACT

Tryptophan catabolism is increasingly recognized as a key and druggable molecular mechanism active in cancer, immune, and glioneural cells and involved in the modulation of antitumor immunity, autoimmunity and glioneural function. In addition to the pivotal rate limiting enzyme indoleamine-2,3-dioxygenase, expression of tryptophan-2,3-dioxygenase (TDO) has recently been described as an alternative pathway responsible for constitutive tryptophan degradation in malignant gliomas and other types of cancer. In addition, TDO has been implicated as a key regulator of neurotoxicity involved in neurodegenerative diseases and ageing. The pathways regulating TDO expression, however, are largely unknown. Here, a siRNA-based transcription factor profiling in human glioblastoma cells revealed that the expression of human TDO is suppressed by endogenous glucocorticoid signaling. Similarly, treatment of glioblastoma cells with the synthetic glucocorticoid dexamethasone led to a reduction of TDO expression and activity in vitro and in vivo. TDO inhibition was dependent on the immunophilin FKBP52, whose FK1 domain physically interacted with the glucocorticoid receptor as demonstrated by bimolecular fluorescence complementation and in situ proximity ligation assays. Accordingly, gene expression profile analyses revealed negative correlation of FKBP52 and TDO in glial and neural tumors and in normal brain. Knockdown of FKBP52 and treatment with the FK-binding immunosuppressant FK506 enhanced TDO expression and activity in glioblastoma cells. In summary, we identify a novel steroid-responsive FKBP52-dependent pathway suppressing the expression and activity of TDO, a central and rate-limiting enzyme in tryptophan metabolism, in human gliomas.


Subject(s)
Glioblastoma/metabolism , Signal Transduction/drug effects , Tacrolimus Binding Proteins/metabolism , Tryptophan Oxygenase/metabolism , Tryptophan/metabolism , Aging/drug effects , Animals , Cell Line, Tumor , Dexamethasone/pharmacology , Glioblastoma/drug therapy , Humans , Mice , Tacrolimus/pharmacology , Tryptophan Oxygenase/antagonists & inhibitors
10.
J Neuroinflammation ; 11: 14, 2014 Jan 23.
Article in English | MEDLINE | ID: mdl-24456653

ABSTRACT

BACKGROUND: Prophylaxis with unmethylated cytosine phosphate guanidine (CpG) oligodeoxynucleotides (ODN) protects against several systemic experimental infections. Escherichia coli is a major cause of Gram-negative neonatal bacterial meningitis and also causes meningitis and meningoencephalitis in older and immunocompromised patients. METHODS: Wild-type (wt) and Toll-like receptor 9 (TLR9)-deficient mice were rendered neutropenic by intraperitoneal administration of the anti-Ly-6G monoclonal antibody. Immunocompetent and neutropenic mice received intraperitoneal CpG ODN or vehicle 72 h prior to induction of E. coli K1 meningoencephalitis. RESULTS: Pre-treatment with CpG ODN significantly increased survival of neutropenic wt mice from 33% to 75% (P = 0.0003) but did not protect neutropenic TLR9-/- mice. The protective effect of CpG ODN was associated with an enhanced production of interleukin (IL)-12/IL-23p40 with sustained increased levels in serum and spleen at least for 17 days after conditioning compared to buffer-treated animals. CpG-treated neutropenic wt mice showed reduced bacterial concentrations and increased recruitment of Ly6ChighCCR2+ monocytes in brain and spleen 42 h after infection. The levels of macrophage inflammatory protein 1α (MIP-1α) and interferon gamma (IFN-γ) in spleen were higher 42 h after infection in CpG-treated compared to buffer-treated neutropenic animals. In immunocompetent mice, prophylaxis with CpG ODN did not significantly increase survival compared to the buffer group (60% vs. 45%, P = 0.2). CONCLUSIONS: These findings suggest that systemic administration of CpG ODN may help to prevent bacterial CNS infections in immunocompromised individuals.


Subject(s)
Escherichia coli Infections/prevention & control , Guanidine/chemistry , Oligodeoxyribonucleotides/therapeutic use , Animals , Antigens, CD/metabolism , Central Nervous System/drug effects , Central Nervous System/microbiology , Central Nervous System/pathology , Cytokines/metabolism , Disease Models, Animal , Drug Administration Schedule , Escherichia coli/physiology , Flow Cytometry , Meningoencephalitis/prevention & control , Mice , Mice, Knockout , Spleen/microbiology , Spleen/pathology , Toll-Like Receptor 9/deficiency
11.
J Clin Invest ; 134(12)2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38941297

ABSTRACT

STING agonists can reprogram the tumor microenvironment to induce immunological clearance within the central nervous system. Using multiplexed sequential immunofluorescence (SeqIF) and the Ivy Glioblastoma Atlas, STING expression was found in myeloid populations and in the perivascular space. The STING agonist 8803 increased median survival in multiple preclinical models of glioblastoma, including QPP8, an immune checkpoint blockade-resistant model, where 100% of mice were cured. Ex vivo flow cytometry profiling during the therapeutic window demonstrated increases in myeloid tumor trafficking and activation, alongside enhancement of CD8+ T cell and NK effector responses. Treatment with 8803 reprogrammed microglia to express costimulatory CD80/CD86 and iNOS, while decreasing immunosuppressive CD206 and arginase. In humanized mice, where tumor cell STING is epigenetically silenced, 8803 therapeutic activity was maintained, further attesting to myeloid dependency and reprogramming. Although the combination with a STAT3 inhibitor did not further enhance STING agonist activity, the addition of anti-PD-1 antibodies to 8803 treatment enhanced survival in an immune checkpoint blockade-responsive glioma model. In summary, 8803 as a monotherapy demonstrates marked in vivo therapeutic activity, meriting consideration for clinical translation.


Subject(s)
Glioblastoma , Membrane Proteins , Tumor Microenvironment , Animals , Glioblastoma/immunology , Glioblastoma/pathology , Glioblastoma/drug therapy , Glioblastoma/genetics , Tumor Microenvironment/immunology , Mice , Membrane Proteins/immunology , Membrane Proteins/genetics , Membrane Proteins/agonists , Humans , Cell Line, Tumor , Brain Neoplasms/immunology , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Brain Neoplasms/genetics
12.
Nat Commun ; 14(1): 1333, 2023 03 11.
Article in English | MEDLINE | ID: mdl-36906610

ABSTRACT

The prototypic cancer-predisposition disease Fanconi Anemia (FA) is identified by biallelic mutations in any one of twenty-three FANC genes. Puzzlingly, inactivation of one Fanc gene alone in mice fails to faithfully model the pleiotropic human disease without additional external stress. Here we find that FA patients frequently display FANC co-mutations. Combining exemplary homozygous hypomorphic Brca2/Fancd1 and Rad51c/Fanco mutations in mice phenocopies human FA with bone marrow failure, rapid death by cancer, cellular cancer-drug hypersensitivity and severe replication instability. These grave phenotypes contrast the unremarkable phenotypes seen in mice with single gene-function inactivation, revealing an unexpected synergism between Fanc mutations. Beyond FA, breast cancer-genome analysis confirms that polygenic FANC tumor-mutations correlate with lower survival, expanding our understanding of FANC genes beyond an epistatic FA-pathway. Collectively, the data establish a polygenic replication stress concept as a testable principle, whereby co-occurrence of a distinct second gene mutation amplifies and drives endogenous replication stress, genome instability and disease.


Subject(s)
Breast Neoplasms , Fanconi Anemia , Animals , Female , Humans , Mice , BRCA2 Protein/genetics , Breast Neoplasms/genetics , DNA-Binding Proteins/genetics , Fanconi Anemia/genetics , Genotype , Mutation , Phenotype
14.
Oncoimmunology ; 11(1): 2062827, 2022.
Article in English | MEDLINE | ID: mdl-35433114

ABSTRACT

Osteopontin (OPN) is produced by tumor cells as well as by myeloid cells and is enriched in the tumor microenvironment (TME) of many cancers. Given the roles of OPN in tumor progression and immune suppression, we hypothesized that targeting OPN with aptamers that have high affinity and specificity could be a promising therapeutic strategy. Bi-specific aptamers targeting ligands for cellular internalization were conjugated to siRNAs to suppress OPN were created, and therapeutic leads were selected based on target engagement and in vivo activity. Aptamers as carriers for siRNA approaches were created including a cancer targeting nucleolin aptamer Ncl-OPN siRNA and a myeloid targeting CpG oligodeoxynucleotide (ODN)-OPN siRNA conjugate. These aptamers were selected as therapeutic leads based on 70-90% OPN inhibition in cancer (GL261, 344SQ, 4T1B2b) and myeloid (DC2.4) cells relative to scramble controls. In established immune competent 344SQ lung cancer and 4T1B2b breast cancer models, these aptamers, including in combination, demonstrate therapeutic activity by inhibiting tumor growth. The Ncl-OPN siRNA aptamer demonstrated efficacy in an immune competent orthotopic glioma model administered systemically secondary to the ability of the aptamer to access the glioma TME. Therapeutic activity was demonstrated using both aptamers in a breast cancer brain metastasis model. Targeted inhibition of OPN in tumor cells and myeloid cells using bifunctional aptamers that are internalized by specific cell types and suppress OPN expression once internalized may have clinical potential in cancer treatment.


Subject(s)
Aptamers, Nucleotide , Breast Neoplasms , Glioma , Aptamers, Nucleotide/genetics , Aptamers, Nucleotide/metabolism , Aptamers, Nucleotide/therapeutic use , Central Nervous System/metabolism , Female , Humans , RNA, Small Interfering/genetics , RNA, Small Interfering/therapeutic use , Tumor Microenvironment
15.
JCI Insight ; 7(12)2022 06 22.
Article in English | MEDLINE | ID: mdl-35653194

ABSTRACT

Novel therapeutic strategies targeting glioblastoma (GBM) often fail in the clinic, partly because preclinical models in which hypotheses are being tested do not recapitulate human disease. To address this challenge, we took advantage of our previously developed spontaneous Qk/Trp53/Pten (QPP) triple-knockout model of human GBM, comparing the immune microenvironment of QPP mice with that of patient-derived tumors to determine whether this model provides opportunity for gaining insights into tumor physiopathology and preclinical evaluation of therapeutic agents. Immune profiling analyses and single-cell sequencing of implanted and spontaneous tumors from QPP mice and from patients with glioma revealed intratumoral immune components that were predominantly myeloid cells (e.g., monocytes, macrophages, and microglia), with minor populations of T, B, and NK cells. When comparing spontaneous and implanted mouse samples, we found more neutrophils and T and NK cells in the implanted model. Neutrophils and T and NK cells were increased in abundance in samples derived from human high-grade glioma compared with those derived from low-grade glioma. Overall, our data demonstrate that our implanted and spontaneous QPP models recapitulate the immunosuppressive myeloid-dominant nature of the tumor microenvironment of human gliomas. Our model provides a suitable tool for investigating the complex immune compartment of gliomas.


Subject(s)
Glioblastoma , Glioma , Animals , Disease Models, Animal , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Macrophages , Mice , Tumor Microenvironment
16.
CNS Oncol ; 11(2): CNS87, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35575067

ABSTRACT

Aim: To ascertain the maximum tolerated dose (MTD)/maximum feasible dose (MFD) of WP1066 and p-STAT3 target engagement within recurrent glioblastoma (GBM) patients. Patients & methods: In a first-in-human open-label, single-center, single-arm 3 + 3 design Phase I clinical trial, eight patients were treated with WP1066 until disease progression or unacceptable toxicities. Results: In the absence of significant toxicity, the MFD was identified to be 8 mg/kg. The most common adverse event was grade 1 nausea and diarrhea in 50% of patients. No treatment-related deaths occurred; 6 of 8 patients died from disease progression and one was lost to follow-up. Of 8 patients with radiographic follow-up, all had progressive disease. The longest response duration exceeded 3.25 months. The median progression-free survival (PFS) time was 2.3 months (95% CI: 1.7 months-NA months), and 6-month PFS (PFS6) rate was 0%. The median overall survival (OS) rate was 25 months (95% CI: 22.5 months-NA months), with an estimated 1-year OS rate of 100%. Pharmacokinetic (PK) data demonstrated that at 8 mg/kg, the T1/2 was 2-3 h with a dose dependent increase in the Cmax. Immune monitoring of the peripheral blood demonstrated that there was p-STAT3 suppression starting at a dose of 1 mg/kg. Conclusion: Immune analyses indicated that WP1066 inhibited systemic immune p-STAT3. WP1066 had an MFD identified at 8 mg/kg which is the target allometric dose based on prior preclinical modeling in combination with radiation therapy and a Phase II study is being planned for newly diagnosed MGMT promoter unmethylated glioblastoma patients.


Subject(s)
Brain Neoplasms , Glioblastoma , Glioma , Brain Neoplasms/pathology , Disease Progression , Glioblastoma/pathology , Glioma/drug therapy , Humans , Pyridines , STAT3 Transcription Factor/therapeutic use , Tyrphostins
17.
JCI Insight ; 7(9)2022 05 09.
Article in English | MEDLINE | ID: mdl-35316217

ABSTRACT

BACKGROUNDImmune cell profiling of primary and metastatic CNS tumors has been focused on the tumor, not the tumor microenvironment (TME), or has been analyzed via biopsies.METHODSEn bloc resections of gliomas (n = 10) and lung metastases (n = 10) were analyzed via tissue segmentation and high-dimension Opal 7-color multiplex imaging. Single-cell RNA analyses were used to infer immune cell functionality.RESULTSWithin gliomas, T cells were localized in the infiltrating edge and perivascular space of tumors, while residing mostly in the stroma of metastatic tumors. CD163+ macrophages were evident throughout the TME of metastatic tumors, whereas in gliomas, CD68+, CD11c+CD68+, and CD11c+CD68+CD163+ cell subtypes were commonly observed. In lung metastases, T cells interacted with CD163+ macrophages as dyads and clusters at the brain-tumor interface and within the tumor itself and as clusters within the necrotic core. In contrast, gliomas typically lacked dyad and cluster interactions, except for T cell CD68+ cell dyads within the tumor. Analysis of transcriptomic data in glioblastomas revealed that innate immune cells expressed both proinflammatory and immunosuppressive gene signatures.CONCLUSIONOur results show that immunosuppressive macrophages are abundant within the TME and that the immune cell interactome between cancer lineages is distinct. Further, these data provide information for evaluating the role of different immune cell populations in brain tumor growth and therapeutic responses.FUNDINGThis study was supported by the NIH (NS120547), a Developmental research project award (P50CA221747), ReMission Alliance, institutional funding from Northwestern University and the Lurie Comprehensive Cancer Center, and gifts from the Mosky family and Perry McKay. Performed in the Flow Cytometry & Cellular Imaging Core Facility at MD Anderson Cancer Center, this study received support in part from the NIH (CA016672) and the National Cancer Institute (NCI) Research Specialist award 1 (R50 CA243707). Additional support was provided by CCSG Bioinformatics Shared Resource 5 (P30 CA046592), a gift from Agilent Technologies, a Research Scholar Grant from the American Cancer Society (RSG-16-005-01), a Precision Health Investigator Award from University of Michigan (U-M) Precision Health, the NCI (R37-CA214955), startup institutional research funds from U-M, and a Biomedical Informatics & Data Science Training Grant (T32GM141746).


Subject(s)
Brain Neoplasms , Glioblastoma , Lung Neoplasms , Brain Neoplasms/pathology , Central Nervous System/metabolism , Glioblastoma/pathology , Humans , Lung Neoplasms/pathology , Macrophages/metabolism , STAT3 Transcription Factor/metabolism , Tumor Microenvironment , United States
18.
Sci Transl Med ; 14(660): eabo6135, 2022 08 31.
Article in English | MEDLINE | ID: mdl-36044599

ABSTRACT

T cell receptor (TCR)-based immunotherapy has emerged as a promising therapeutic approach for the treatment of patients with solid cancers. Identifying peptide-human leukocyte antigen (pHLA) complexes highly presented on tumors and rarely expressed on healthy tissue in combination with high-affinity TCRs that when introduced into T cells can redirect T cells to eliminate tumor but not healthy tissue is a key requirement for safe and efficacious TCR-based therapies. To discover promising shared tumor antigens that could be targeted via TCR-based adoptive T cell therapy, we employed population-scale immunopeptidomics using quantitative mass spectrometry across ~1500 tumor and normal tissue samples. We identified an HLA-A*02:01-restricted pan-cancer epitope within the collagen type VI α-3 (COL6A3) gene that is highly presented on tumor stroma across multiple solid cancers due to a tumor-specific alternative splicing event that rarely occurs outside the tumor microenvironment. T cells expressing natural COL6A3-specific TCRs demonstrated only modest activity against cells presenting high copy numbers of COL6A3 pHLAs. One of these TCRs was affinity-enhanced, enabling transduced T cells to specifically eliminate tumors in vivo that expressed similar copy numbers of pHLAs as primary tumor specimens. The enhanced TCR variants exhibited a favorable safety profile with no detectable off-target reactivity, paving the way to initiate clinical trials using COL6A3-specific TCRs to target an array of solid tumors.


Subject(s)
Immunotherapy, Adoptive , Receptors, Antigen, T-Cell , T-Lymphocytes , Antigens, Neoplasm , Cell Line, Tumor , Cell- and Tissue-Based Therapy , Humans , Immunotherapy, Adoptive/methods , Proteomics , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/therapeutic use
19.
Nat Rev Clin Oncol ; 18(11): 729-744, 2021 11.
Article in English | MEDLINE | ID: mdl-34117475

ABSTRACT

Immunotherapy has enabled remarkable therapeutic responses across cancers of various lineages, albeit with some notable exceptions such as glioblastoma. Several previous misconceptions, which have impaired progress in the past, including the presence and role of the blood-brain barrier and a lack of lymphatic drainage, have been refuted. Nonetheless, a subset of patients with brain metastases but, paradoxically, not the vast majority of those with gliomas are able to respond to immune-checkpoint inhibitors. Immune profiling of samples obtained from patients with central nervous system malignancies using techniques such as mass cytometry and single-cell sequencing along with experimental data from genetically engineered mouse models have revealed fundamental differences in immune composition and immunobiology that not only explain the differences in responsiveness to these agents but also lay the foundations for immunotherapeutic strategies that are applicable to gliomas. Herein, we review the emerging data on the differences in immune cell composition, function and interactions within central nervous system tumours and provide guidance on the development of novel immunotherapies for these historically difficult-to-treat cancers.


Subject(s)
Central Nervous System Neoplasms/drug therapy , Immunotherapy/methods , Humans
20.
Cancers (Basel) ; 13(3)2021 Jan 24.
Article in English | MEDLINE | ID: mdl-33498872

ABSTRACT

Glioblastoma remains one of the deadliest and treatment-refractory human malignancies in large part due to its diffusely infiltrative nature, molecular heterogeneity, and capacity for immune escape. The Janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling pathway contributes substantively to a wide variety of protumorigenic functions, including proliferation, anti-apoptosis, angiogenesis, stem cell maintenance, and immune suppression. We review the current state of knowledge regarding the biological role of JAK/STAT signaling in glioblastoma, therapeutic strategies, and future directions for the field.

SELECTION OF CITATIONS
SEARCH DETAIL