Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
Sci Rep ; 14(1): 16721, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39030304

ABSTRACT

Antigen-specific cytotoxic CD8 T cells are extremely effective in controlling tumor growth and have been the focus of immunotherapy approaches. We leverage in silico tools to investigate whether the occurrence of mutations in proteins previously described as immunogenic and highly expressed by glioblastoma multiforme (GBM), such as Epidermal Growth Factor Receptor (EGFR), Isocitrate Dehydrogenase 1 (IDH1), Phosphatase and Tensin homolog (PTEN) and Tumor Protein 53 (TP53), may be contributing to the differential presentation of immunogenic epitopes. We recovered Class I MHC binding information from wild-type and mutated proteins using the Immune Epitope Database (IEDB). After that, we built peptide-MHC (pMHC-I) models in HLA-arena, followed by hierarchical clustering analysis based on electrostatic surface features from each complex. We identified point mutations that are determinants for the presentation of a set of peptides from TP53 protein. We point to structural features in the pMHC-I complexes of wild-type and mutated peptides, which may play a role in the recognition of CD8 T cells. To further explore these features, we performed 100 ns molecular dynamics simulations for the peptide pairs (wt/mut) selected. In pursuit of novel therapeutic targets for GBM treatment, we selected peptides where our predictive results indicated that mutations would not disrupt epitope presentation, thereby maintaining a specific CD8 T cell immune response. These peptides hold potential for future GBM interventions, including peptide-based or mRNA vaccine development applications.


Subject(s)
Antigen Presentation , CD8-Positive T-Lymphocytes , Glioblastoma , Isocitrate Dehydrogenase , Tumor Suppressor Protein p53 , Glioblastoma/immunology , Glioblastoma/genetics , Glioblastoma/therapy , Humans , CD8-Positive T-Lymphocytes/immunology , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/immunology , Isocitrate Dehydrogenase/chemistry , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/immunology , Antigen Presentation/immunology , Mutation , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/immunology , PTEN Phosphohydrolase/chemistry , ErbB Receptors/immunology , ErbB Receptors/genetics , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/genetics , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/genetics , Brain Neoplasms/immunology , Brain Neoplasms/genetics , Brain Neoplasms/therapy
2.
Blood ; 143(17): 1726-1737, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38241630

ABSTRACT

ABSTRACT: For patients with high-risk or relapsed/refractory acute myeloid leukemia (AML), allogeneic stem cell transplantation (allo-HSCT) and the graft-versus-leukemia effect mediated by donor T cells, offer the best chance of long-term remission. However, the concurrent transfer of alloreactive T cells can lead to graft-versus-host disease that is associated with transplant-related morbidity and mortality. Furthermore, ∼60% of patients will ultimately relapse after allo-HSCT, thus, underscoring the need for novel therapeutic strategies that are safe and effective. In this study, we explored the feasibility of immunotherapeutically targeting neoantigens, which arise from recurrent nonsynonymous mutations in AML and thus represent attractive targets because they are exclusively present on the tumor. Focusing on 14 recurrent driver mutations across 8 genes found in AML, we investigated their immunogenicity in 23 individuals with diverse HLA profiles. We demonstrate the immunogenicity of AML neoantigens, with 17 of 23 (74%) reactive donors screened mounting a response. The most immunodominant neoantigens were IDH2R140Q (n = 11 of 17 responders), IDH1R132H (n = 7 of 17), and FLT3D835Y (n = 6 of 17). In-depth studies of IDH2R140Q-specific T cells revealed the presence of reactive CD4+ and CD8+ T cells capable of recognizing distinct mutant-specific epitopes restricted to different HLA alleles. These neo-T cells could selectively recognize and kill HLA-matched AML targets endogenously expressing IDH2R140Q both in vitro and in vivo. Overall, our findings support the clinical translation of neoantigen-specific T cells to treat relapsed/refractory AML.


Subject(s)
Antigens, Neoplasm , Isocitrate Dehydrogenase , Leukemia, Myeloid, Acute , Humans , Antigens, Neoplasm/immunology , Antigens, Neoplasm/genetics , Hematopoietic Stem Cell Transplantation , Immunotherapy/methods , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/immunology , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/therapy , Mutation
3.
Front Immunol ; 13: 939523, 2022.
Article in English | MEDLINE | ID: mdl-36091049

ABSTRACT

Background: Glioblastoma (GBM) is the most prominent and aggressive primary brain tumor in adults. Anoikis is a specific form of programmed cell death that plays a key role in tumor invasion and metastasis. The presence of anti-anoikis factors is associated with tumor aggressiveness and drug resistance. Methods: The non-negative matrix factorization algorithm was used for effective dimension reduction for integrated datasets. Differences in the tumor microenvironment (TME), stemness indices, and clinical characteristics between the two clusters were analyzed. Difference analysis, weighted gene coexpression network analysis (WGCNA), univariate Cox regression, and least absolute shrinkage and selection operator regression were leveraged to screen prognosis-related genes and construct a risk score model. Immunohistochemistry was performed to evaluate the expression of representative genes in clinical specimens. The relationship between the risk score and the TME, stemness, clinical traits, and immunotherapy response was assessed in GBM and pancancer. Results: Two definite clusters were identified on the basis of anoikis-related gene expression. Patients with GBM assigned to C1 were characterized by shortened overall survival, higher suppressive immune infiltration levels, and lower stemness indices. We further constructed a risk scoring model to quantify the regulatory patterns of anoikis-related genes. The higher risk score group was characterized by a poor prognosis, the infiltration of suppressive immune cells and a differentiated phenotype, whereas the lower risk score group exhibited the opposite effects. In addition, patients in the lower risk score group exhibited a higher frequency of isocitrate dehydrogenase (IDH) mutations and a more sensitive response to immunotherapy. Drug sensitivity analysis was performed, revealing that the higher risk group may benefit more from drugs targeting the PI3K/mTOR signaling pathway. Conclusion: We revealed potential relationships between anoikis-related genes and clinical features, TME, stemness, IDH mutation, and immunotherapy and elucidated their therapeutic value.


Subject(s)
Anoikis , Brain Neoplasms , Glioblastoma , Isocitrate Dehydrogenase , Tumor Microenvironment , Algorithms , Anoikis/genetics , Anoikis/immunology , Brain Neoplasms/diagnosis , Brain Neoplasms/genetics , Brain Neoplasms/immunology , Brain Neoplasms/therapy , Glioblastoma/diagnosis , Glioblastoma/genetics , Glioblastoma/immunology , Glioblastoma/therapy , Humans , Immunotherapy , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/immunology , Mutation , Neoplastic Stem Cells/physiology , Prognosis , Risk Assessment , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
4.
Nat Commun ; 12(1): 5271, 2021 09 06.
Article in English | MEDLINE | ID: mdl-34489470

ABSTRACT

Chimeric antigen receptor (CAR) T cells have emerged as a promising class of therapeutic agents, generating remarkable responses in the clinic for a subset of human cancers. One major challenge precluding the wider implementation of CAR therapy is the paucity of tumor-specific antigens. Here, we describe the development of a CAR targeting the tumor-specific isocitrate dehydrogenase 2 (IDH2) with R140Q mutation presented on the cell surface in complex with a common human leukocyte antigen allele, HLA-B*07:02. Engineering of the hinge domain of the CAR, as well as crystal structure-guided optimization of the IDH2R140Q-HLA-B*07:02-targeting moiety, enhances the sensitivity and specificity of CARs to enable targeting of this HLA-restricted neoantigen. This approach thus holds promise for the development and optimization of immunotherapies specific to other cancer driver mutations that are difficult to target by conventional means.


Subject(s)
HLA-B7 Antigen/chemistry , Isocitrate Dehydrogenase/metabolism , Protein Engineering/methods , Receptors, Chimeric Antigen/chemistry , Animals , Antigens, Neoplasm/metabolism , COS Cells , Cell Line , Chlorocebus aethiops , Epitopes , HLA-B7 Antigen/metabolism , Humans , Immunoglobulin Fab Fragments/chemistry , Isocitrate Dehydrogenase/chemistry , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/immunology , Mutation , Peptide Library , Protein Conformation , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/physiology
5.
Cells ; 10(8)2021 08 09.
Article in English | MEDLINE | ID: mdl-34440802

ABSTRACT

Despite the important evolution of immunotherapeutic agents, brain tumors remain, in general, refractory to immune therapeutics. Recent discoveries have revealed that the glioma microenvironment includes a wide variety of immune cells in various states that play an important role in the process of tumorigenesis. Anti-tumor immune activity may be occurring or induced in immunogenic hot spots or at the invasive edge of central nervous system (CNS) tumors. Understanding the complex heterogeneity of the immune microenvironment in gliomas will likely be the key to unlocking the full potential of immunotherapeutic strategies. An essential consideration will be the induction of immunological effector responses in the setting of the numerous aspects of immunosuppression and evasion. As such, immune therapeutic combinations are a fundamental objective for clinical studies in gliomas. Through immune profiling conducted on immune competent murine models of glioma and ex vivo human glioma tissue, we will discuss how the frequency, distribution of immune cells within the microenvironment, and immune modulatory processes, may be therapeutically modulated to lead to clinical benefits.


Subject(s)
Brain Neoplasms/therapy , Glioma/therapy , Immunotherapy/methods , Tumor Microenvironment/immunology , Animals , Brain Neoplasms/genetics , Brain Neoplasms/immunology , Glioma/genetics , Glioma/immunology , Humans , Immune Tolerance/genetics , Immune Tolerance/immunology , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/immunology , Isocitrate Dehydrogenase/metabolism , Macrophages/immunology , Macrophages/metabolism , Mutation/genetics , Mutation/immunology , Tumor Microenvironment/genetics
6.
Nature ; 592(7854): 463-468, 2021 04.
Article in English | MEDLINE | ID: mdl-33762734

ABSTRACT

Mutated isocitrate dehydrogenase 1 (IDH1) defines a molecularly distinct subtype of diffuse glioma1-3. The most common IDH1 mutation in gliomas affects codon 132 and encodes IDH1(R132H), which harbours a shared clonal neoepitope that is presented on major histocompatibility complex (MHC) class II4,5. An IDH1(R132H)-specific peptide vaccine (IDH1-vac) induces specific therapeutic T helper cell responses that are effective against IDH1(R132H)+ tumours in syngeneic MHC-humanized mice4,6-8. Here we describe a multicentre, single-arm, open-label, first-in-humans phase I trial that we carried out in 33 patients with newly diagnosed World Health Organization grade 3 and 4 IDH1(R132H)+ astrocytomas (Neurooncology Working Group of the German Cancer Society trial 16 (NOA16), ClinicalTrials.gov identifier NCT02454634). The trial met its primary safety endpoint, with vaccine-related adverse events restricted to grade 1. Vaccine-induced immune responses were observed in 93.3% of patients across multiple MHC alleles. Three-year progression-free and death-free rates were 0.63 and 0.84, respectively. Patients with immune responses showed a two-year progression-free rate of 0.82. Two patients without an immune response showed tumour progression within two years of first diagnosis. A mutation-specificity score that incorporates the duration and level of vaccine-induced IDH1(R132H)-specific T cell responses was associated with intratumoral presentation of the IDH1(R132H) neoantigen in pre-treatment tumour tissue. There was a high frequency of pseudoprogression, which indicates intratumoral inflammatory reactions. Pseudoprogression was associated with increased vaccine-induced peripheral T cell responses. Combined single-cell RNA and T cell receptor sequencing showed that tumour-infiltrating CD40LG+ and CXCL13+ T helper cell clusters in a patient with pseudoprogression were dominated by a single IDH1(R132H)-reactive T cell receptor.


Subject(s)
Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Glioma/diagnosis , Glioma/therapy , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/immunology , Mutation , Adult , Cells, Cultured , Disease Progression , Female , Glioma/genetics , Glioma/immunology , Humans , Male , Mutant Proteins/genetics , Mutant Proteins/immunology , Phenotype , Receptors, Antigen, T-Cell/immunology , Survival Rate , T-Lymphocytes/immunology
7.
J Clin Invest ; 131(4)2021 02 15.
Article in English | MEDLINE | ID: mdl-33332283

ABSTRACT

Mutant isocitrate dehydrogenase 1 (IDH1-R132H; mIDH1) is a hallmark of adult gliomas. Lower grade mIDH1 gliomas are classified into 2 molecular subgroups: 1p/19q codeletion/TERT-promoter mutations or inactivating mutations in α-thalassemia/mental retardation syndrome X-linked (ATRX) and TP53. This work focuses on glioma subtypes harboring mIDH1, TP53, and ATRX inactivation. IDH1-R132H is a gain-of-function mutation that converts α-ketoglutarate into 2-hydroxyglutarate (D-2HG). The role of D-2HG within the tumor microenvironment of mIDH1/mATRX/mTP53 gliomas remains unexplored. Inhibition of D-2HG, when used as monotherapy or in combination with radiation and temozolomide (IR/TMZ), led to increased median survival (MS) of mIDH1 glioma-bearing mice. Also, D-2HG inhibition elicited anti-mIDH1 glioma immunological memory. In response to D-2HG inhibition, PD-L1 expression levels on mIDH1-glioma cells increased to similar levels as observed in WT-IDH gliomas. Thus, we combined D-2HG inhibition/IR/TMZ with anti-PDL1 immune checkpoint blockade and observed complete tumor regression in 60% of mIDH1 glioma-bearing mice. This combination strategy reduced T cell exhaustion and favored the generation of memory CD8+ T cells. Our findings demonstrate that metabolic reprogramming elicits anti-mIDH1 glioma immunity, leading to increased MS and immunological memory. Our preclinical data support the testing of IDH-R132H inhibitors in combination with IR/TMZ and anti-PDL1 as targeted therapy for mIDH1/mATRX/mTP53 glioma patients.


Subject(s)
Cellular Reprogramming , Glioma/therapy , Glutarates/pharmacology , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Line, Tumor , Cellular Reprogramming/drug effects , Cellular Reprogramming/genetics , Cellular Reprogramming/immunology , Chemoradiotherapy , Gain of Function Mutation , Glioma/genetics , Glioma/immunology , Glioma/pathology , Humans , Immunologic Memory/drug effects , Immunologic Memory/genetics , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/immunology , Mice , Temozolomide/pharmacology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/immunology , X-linked Nuclear Protein/genetics , X-linked Nuclear Protein/immunology
8.
JCI Insight ; 4(13)2019 07 11.
Article in English | MEDLINE | ID: mdl-31292299

ABSTRACT

Oncolytic viruses induce local tumor destruction and inflammation. Whether virotherapy can also overcome immunosuppression in noninfected tumor areas is under debate. To address this question, we have explored immunologic effects of oncolytic herpes simplex viruses (oHSVs) in a genetically engineered mouse model of isocitrate dehydrogenase (IDH) wild-type glioblastoma, the most common and most malignant primary brain tumor in adults. Our model recapitulates the genomics, the diffuse infiltrative growth pattern, and the extensive macrophage-dominant immunosuppression of human glioblastoma. Infection with an oHSV that was armed with a UL16-binding protein 3 (ULBP3) expression cassette inhibited distant tumor growth in the absence of viral spreading (abscopal effect) and yielded accumulation of activated macrophages and T cells. There was also abscopal synergism of oHSVULBP3 with anti-programmed cell death 1 (anti-PD-1) against distant, uninfected tumor areas; albeit consistent with clinical trials in patients with glioblastoma, monotherapy with anti-PD-1 was ineffective in our model. Arming oHSV with ULBP3 led to upregulation of antigen processing and presentation gene sets in myeloid cells. The cognate ULBP3 receptor NKG2D, however, is not present on myeloid cells, suggesting a noncanonical mechanism of action of ULBP3. Overall, the myeloid-dominant, anti-PD-1-sensitive abscopal effect of oHSVULBP3 warrants further investigation in patients with IDH wild-type glioblastoma.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Brain Neoplasms/therapy , Glioblastoma/therapy , Intercellular Signaling Peptides and Proteins/immunology , Oncolytic Virotherapy/methods , Oncolytic Viruses/immunology , Simplexvirus/immunology , Animals , Antigen Presentation/genetics , Antineoplastic Agents, Immunological/pharmacology , Brain/pathology , Brain Neoplasms/genetics , Brain Neoplasms/immunology , Brain Neoplasms/mortality , Cell Line, Tumor , Combined Modality Therapy/methods , Disease Models, Animal , Female , GPI-Linked Proteins/genetics , GPI-Linked Proteins/immunology , Gene Expression Regulation, Neoplastic/immunology , Glioblastoma/genetics , Glioblastoma/immunology , Glioblastoma/mortality , Humans , Intercellular Signaling Peptides and Proteins/genetics , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/immunology , Kaplan-Meier Estimate , Male , Mice , Mice, Transgenic , Oncolytic Viruses/genetics , Primary Cell Culture , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Simplexvirus/genetics , Up-Regulation
9.
Immunol Cell Biol ; 97(5): 457-469, 2019 05.
Article in English | MEDLINE | ID: mdl-30575118

ABSTRACT

Mutations in the isocitrate dehydrogenase (IDH) 1 gene, especially the R132H mutation, have been reported to be associated with a better prognosis in glioma patients. However, the underlying molecular mechanisms are not yet well understood. Many factors may contribute to differences in the survival of IDH1 wild-type and IDH1 mutant glioma patients, in which immune components play a potentially important role. In this study, we analyzed The Cancer Genome Atlas (TCGA), and the Chinese Glioma Genome Atlas (CGGA) databases, as well as glioma patient-derived tumor samples. We found that there was a higher infiltration of natural killer (NK) cells in IDH1 mutant glioma patients, and this was correlated with a better prognosis. We also showed that IDH1-R132 tumor cells had higher expression levels of the chemokine CX3CL1. This arises as a result of the conversion of α-ketoglutarate to R(-)-2-hydroxyglutarate by the IDH1 mutant and the resultant phosphorylation of nuclear factor kappa B. Knockdown of CX3CL1 decreased the migration of NK cells. In addition, the high levels of expression of CX3CL1 were positively correlated with glioma patient survival in the TCGA and CGGA databases, and in the clinical samples. Overall, our data have identified a novel mechanism in which R132H mutation of the IDH1 gene serves as a tumor suppressor by promoting the recruitment of NK cells through CX3CL1/CX3CR1 chemotaxis.


Subject(s)
CX3C Chemokine Receptor 1/immunology , Chemokine CX3CL1/immunology , Chemotaxis , Gene Expression Regulation, Neoplastic/immunology , Glioma , Isocitrate Dehydrogenase , Killer Cells, Natural/immunology , Mutation, Missense , Tumor Suppressor Proteins , Amino Acid Substitution , CX3C Chemokine Receptor 1/genetics , Cell Line, Tumor , Chemokine CX3CL1/genetics , Chemotaxis/genetics , Chemotaxis/immunology , Female , Glioma/genetics , Glioma/immunology , Glioma/pathology , Humans , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/immunology , Killer Cells, Natural/pathology , Male , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/immunology
10.
Appl Immunohistochem Mol Morphol ; 27(10): 722-725, 2019.
Article in English | MEDLINE | ID: mdl-30358614

ABSTRACT

Determination of the isocitrate dehydrogenase (IDH) mutation status, presence or absence of mutation in IDH genes (IDH1 or IDH2), has become one of the most important molecular features taken into account in the management of patients with diffuse gliomas. Tumors that are IDH-mutant have a better prognosis than their counterparts with similar histologic grade and IDH-wildtype phenotype. IDH1-R132H is the most common IDH mutation, present in ~90% of IDH-mutant cases. This mutation yields an altered protein that can be detected by immunohistochemistry. We evaluated the IDH1-R132H antibody (clone H09) to determine IDH mutation status as the first line test and compared with the results of polymerase chain reaction (PCR) testing that can detect more types of mutations in IDH1 or IDH2. A total of 62 gliomas were evaluated: 30 glioblastomas (including 3 gliosarcomas), 11 grade III diffuse gliomas, 17 grade II diffuse gliomas, and 4 circumscribed gliomas. Twelve of 62 cases were IDH-mutant by immunohistochemistry and 15 of 62 by PCR. PCR detected the following mutations: IDH1-R132H (11 cases), IDH1-R132C (1 case), IDH2 R172, NOS (1 case), IDH1 R132, NOS (1 case), and IDH2-R172K (1 case). The R132H antibody had high specificity (100%) and sensitivity (80%) to detect IDH mutation status; the discordant results were 3 false-negatives. IDH-R132H immunostain is suitable as a first line test. Nonimmunoreactive cases could be studied by PCR following recommendations of the 2016 World Health Organization guidelines.


Subject(s)
Brain Neoplasms/diagnosis , Glioma/diagnosis , Gliosarcoma/diagnosis , Immunohistochemistry/methods , Isocitrate Dehydrogenase/metabolism , Polymerase Chain Reaction/methods , Antibodies/metabolism , False Negative Reactions , Humans , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/immunology , Mutation/genetics , Prognosis , Reproducibility of Results , Sensitivity and Specificity
11.
Curr Opin Oncol ; 30(6): 368-374, 2018 11.
Article in English | MEDLINE | ID: mdl-30102604

ABSTRACT

PURPOSE OF REVIEW: The present review introduces recent progress in eliciting the role of mutant isocitrate dehydrogenase (IDH) in gliomas, especially regarding its mode of action as a modulator of antitumor immune response, and provides rationales for targeting mutant IDH in glioma immunotherapy. Both the development of small molecule inhibitors repressing the enzymatic activity of mutant IDH and novel, mechanism-led combination immunotherapies are discussed. RECENT FINDINGS: Since the discovery of highly frequent IDH mutations in low-grade gliomas and nonsolid malignancies, its tumor cell-intrinsic effects have been intensively investigated. Tumor cells expressing mutant IDH display profound alterations of redox control capacity, phospholipid profile, and ATP supply. Recent findings suggest that IDH mutations - via intricate, yet druggable pathways - cause immunological alterations, highlighting the importance of oncogenic drivers as modulators of antitumor immunity and targets for immunotherapy. SUMMARY: Mutant IDH is not only a disease-defining biomarker and oncogenic driver in glioma, but is also a neoantigen and a regulator of glioma immune evasion. Effective and specific strategies targeting the immunomodulatory properties of mutant IDH may complement current (immuno-)therapeutic strategies and approved antiglioma treatments to improve outcome.


Subject(s)
Brain Neoplasms/enzymology , Brain Neoplasms/therapy , Glioma/enzymology , Glioma/therapy , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/immunology , Animals , Brain Neoplasms/genetics , Brain Neoplasms/immunology , Glioma/genetics , Glioma/immunology , Humans , Immunotherapy/methods , Mutation , Tumor Escape/genetics
12.
Biochem Biophys Res Commun ; 503(2): 798-802, 2018 09 05.
Article in English | MEDLINE | ID: mdl-29913148

ABSTRACT

Acute lung injury (ALI) is an acute failure of the respiratory system with unacceptably high mortality, for which effective treatment is urgently necessary. Infiltrations by immune cells, such as leukocytes and macrophages, are responsible for the inflammatory response in ALI, which is characterized by excessive production of pro-inflammatory mediators in lung tissues exposed to various pathogen-associated molecules such as lipopolysaccharide (LPS) from microbial organisms. α-Ketoglutarate (α-KG) is a key metabolic intermediate and acts as a pro-inflammatory metabolite, which is responsible for LPS-induced proinflammatory cytokine production through NF-κB signaling pathway. Mitochondrial NADP+-dependent isocitrate dehydrogenase (IDH2) has been reported as an essential enzyme catalyzing the conversion of isocitrate to α-KG with concurrent production of NAPDH. Therefore, we evaluated the role of IDH2 in LPS-induced ALI using IDH2-deficient mice. We observed that LPS-induced inflammation and lung injury is attenuated in IDH2-deficient mice, leading to a lengthened life span of the mice. Our results also suggest that IDH2 disruption suppresses LPS-induced proinflammatory cytokine production, resulting from an inhibition of the NF-κB signaling axis in an α-KG-dependent manner. In conclusion, disruption of IDH2 leads to a decrease in α-KG levels, and the activation of NF-κB in response to LPS is attenuated by reduction of α-KG levels, which eventually reduces the inflammatory response in the lung during LPS-induced ALI. The present study supports the rationale for targeting IDH2 as an important therapeutic strategy for the treatment of systemic inflammatory response syndromes, particularly ALI.


Subject(s)
Acute Lung Injury/immunology , Isocitrate Dehydrogenase/immunology , Ketoglutaric Acids/immunology , Lipopolysaccharides/immunology , Pneumonia/immunology , Acute Lung Injury/genetics , Animals , Gene Knockout Techniques , Isocitrate Dehydrogenase/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/immunology , Pneumonia/genetics
13.
Am J Surg Pathol ; 42(5): 569-577, 2018 05.
Article in English | MEDLINE | ID: mdl-29635257

ABSTRACT

Isocitrate dehydrogenase 1 (IDH1) and IDH2 mutations occur in a variety of myeloid neoplasms. Immunohistochemistry (IHC)-based direct visualization of mutant clones of hematopoietic cells can be useful for rapid diagnostic screening and for monitoring treatment response. In this study, we first evaluated the sensitivity and specificity of the IDH1 p.R132H mutation-specific antibody by IHC. All IDH1 wild type cases (n=11) and IDH1 mutant cases with a non-p.R132H mutation (n=30) were negative by IHC, demonstrating 100% antibody specificity. All the initial diagnostic specimens with IDH1 p.R132H mutation including acute myeloid leukemia (n=30), myelodysplastic syndromes (MDS) (n=10), MDS/myeloproliferative neoplasms (MPN) (n=4), and MPN (n=5) were positive by IHC, demonstrating 100% antibody sensitivity. Both immature and mature myeloid cells showed immunoreactivity. Erythroid precursors, lymphoid cells, endothelial cells, and osteoblasts were consistently negative by IHC. We then evaluated the follow-up specimens with a known IDH1 mutation status including acute myeloid leukemia (n=23), MDS (n=2), MDS/MPN (n=2), and MPN (n=2). Thirty-three IDH1 p.R132H mutant cases were positive by IHC and 12 IDH1 mutation negative cases were negative by IHC. However, IHC reactivity in up to 25% of bone marrow cells was noted in 8 of 20 polymerase chain reaction-negative cases, all from patients with a known history of IDH1 p.R132H mutation indicating sampling error or a sensitivity issue with molecular tests. These data indicate that IHC is a highly specific and sensitive tool to detect IDH1 p.R132H mutation in bone marrow involved by myeloid neoplasms. In addition, IDH1 p.R132H IHC also allows localization and assessment of the maturation stage of the clones carrying the mutation.


Subject(s)
Antibodies/immunology , Biomarkers, Tumor/genetics , DNA Mutational Analysis/methods , Immunohistochemistry/methods , Isocitrate Dehydrogenase/genetics , Leukemia, Myeloid, Acute/genetics , Mutation , Myelodysplastic Syndromes/genetics , Myeloproliferative Disorders/genetics , Antibody Specificity , Biomarkers, Tumor/immunology , Genetic Predisposition to Disease , Humans , Isocitrate Dehydrogenase/immunology , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/pathology , Myelodysplastic Syndromes/enzymology , Myelodysplastic Syndromes/immunology , Myelodysplastic Syndromes/pathology , Myeloid Cells/enzymology , Myeloid Cells/immunology , Myeloid Cells/pathology , Myeloproliferative Disorders/enzymology , Myeloproliferative Disorders/immunology , Myeloproliferative Disorders/pathology , Phenotype , Predictive Value of Tests , Reproducibility of Results
14.
Vaccine ; 36(17): 2337-2345, 2018 04 19.
Article in English | MEDLINE | ID: mdl-29567033

ABSTRACT

Edwardsiella tarda has become one of the most severe fish pathogens throughout the world. Thus, studies on the design and production of highly protective vaccines against this pathogen, as well as the mechanisms of vaccine-induced disease resistance are urgently needed. In this study, the NADP-dependent isocitrate dehydrogenase (IDH) of E. tarda was recombinantly expressed and flounder (Paralichthys olivaceus) anti-rIDH serum was prepared. Also, the outer membrane proteins (OMPs) of E. tarda were extracted and analyzed by western blotting. The results showed that flounder anti-rIDH serum could specifically recognize a 44 kDa protein of E. tarda OMPs, which was identified to be the native IDH of E. tarda by mass spectrometric. Consistently, immunogold electron microscopy showed that IDH could be detected on the membrane of E. tarda. Then, the vaccine potential of rIDH was tested in a flounder model, and the results showed that rIDH produced a relative percent survival (RPS) of 73.3%, which was significantly higher than that produced by formalin killed E. tarda cells. Immunological analysis showed that rIDH could induce the proliferation of rIDH-specific sIg+ lymphocytes, which resulted in the production of anti-E. tarda antibodies. Accordingly, serum bactericidal activity assay showed that the serum of rIDH vaccinated fish exhibited the highest bactericidal activity compared with other groups. qRT-PCR analysis showed that rIDH could enhance the expressions of IFN-γ, NKEF, IL-6, MHCIα, CD4-1 and CD8α. Moreover, the bacterial burden was also detected in vaccinated fish after challenge, which showed that the number of E. tarda cells in spleen of rIDH group was significantly lower than other groups. All these results suggested that rIDH is a promising vaccine candidate against E. tarda infection.


Subject(s)
Edwardsiella tarda/immunology , Enterobacteriaceae Infections/immunology , Flounder/immunology , Isocitrate Dehydrogenase/immunology , Recombinant Fusion Proteins/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Animals , Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , Bacterial Vaccines/immunology , Fish Diseases/immunology , Fish Diseases/microbiology , Flounder/microbiology , Vaccines, DNA/immunology
15.
BMC Microbiol ; 17(1): 217, 2017 Nov 14.
Article in English | MEDLINE | ID: mdl-29137620

ABSTRACT

BACKGROUND: Vibrio anguillarum is an extracellular bacterial pathogen that is a causative agent of vibriosis in finfish and crustaceans with mortality rates ranging from 30% to 100%. Mutations in central metabolism (glycolysis and the TCA cycle) of intracellular pathogens often result in attenuated virulence due to depletion of required metabolic intermediates; however, it was not known whether mutations in central metabolism would affect virulence in an extracellular pathogen such as V. anguillarum. RESULTS: Seven central metabolism mutants were created and characterized with regard to growth in minimal and complex media, expression of virulence genes, and virulence in juvenile rainbow trout (Oncorhynchus mykiss). Only the isocitrate dehydrogenase (icd) mutant was attenuated in virulence against rainbow trout challenged by either intraperitoneal injection or immersion. Further, the icd mutant was shown to be immunoprotective against wild type V. anguillarum infection. There was no significant decrease in the expression of the three hemolysin genes detected by qRT-PCR. Additionally, only the icd mutant exhibited a significantly decreased growth yield in complex media. Growth yield was directly related to the abundance of glutamate. A strain with a restored wild type icd gene was created and shown to restore growth to a wild type cell density in complex media and pathogenicity in rainbow trout. CONCLUSIONS: The data strongly suggest that a decreased growth yield, resulting from the inability to synthesize α-ketoglutarate, caused the attenuation despite normal levels of expression of virulence genes. Therefore, the ability of an extracellular pathogen to cause disease is dependent upon the availability of host-supplied nutrients for growth. Additionally, a live vaccine strain could be created from an icd deletion strain.


Subject(s)
Fish Diseases/immunology , Fish Diseases/microbiology , Isocitrate Dehydrogenase/genetics , Oncorhynchus mykiss/immunology , Vibrio Infections/veterinary , Vibrio , Virulence/genetics , Adjuvants, Immunologic/genetics , Animals , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Gene Expression/genetics , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Isocitrate Dehydrogenase/immunology , Mutation , Oncorhynchus mykiss/microbiology , Vaccines, Attenuated/immunology , Vibrio/enzymology , Vibrio/genetics , Vibrio/immunology , Vibrio/pathogenicity , Vibrio Infections/immunology , Vibrio Infections/microbiology
16.
J Clin Invest ; 127(4): 1218-1220, 2017 Apr 03.
Article in English | MEDLINE | ID: mdl-28319049

ABSTRACT

Immunotherapy has emerged as a potent approach for treating aggressive cancers, such as non-small-cell lung tumors and metastatic melanoma. Clinical trials are now in progress for patients with malignant gliomas; however, a better understanding of how these tumors escape immune surveillance is required to enhance antitumor immune responses. With gliomas, the recruitment of CD8+ T cells to the tumor is impaired, in part preventing containment or elimination of the tumor. In this issue of the JCI, Kohanbash and colleagues present an elegant dissection of how gliomas exploit an enzymatic activity acquired through a common mutation to abrogate the migration of CD8+ T cells to the tumor. They show that the oncometabolite 2-hydroxyglutarate (2HG), generated by mutated forms of isocitrate dehydrogenase (IDH1 and IDH2), reduces the expression of STAT1, thereby limiting the production of the chemokines CXCL9 and CXCL10. As a result, IDH1-mutated tumors are less effectively infiltrated by CD8+ T cells, contributing to tumor escape. Finally, in mice harboring syngeneic gliomas, an inhibitor of 2HG synthesis complemented vaccination to ameliorate tumor control. Understanding how to increase immune infiltration of gliomas represents a key first step in achieving tumor destruction through immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Glioma/immunology , Glutarates/immunology , Tumor Escape , Animals , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Chemokine CXCL10/genetics , Chemokine CXCL10/immunology , Chemokine CXCL9/genetics , Chemokine CXCL9/immunology , Chemokine CXCL9/metabolism , Glioma/genetics , Glioma/metabolism , Glioma/pathology , Glutarates/metabolism , Humans , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/immunology , Isocitrate Dehydrogenase/metabolism , Mutation , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Neoplasm Proteins/metabolism , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/immunology , STAT1 Transcription Factor/metabolism
17.
Ann Pathol ; 37(1): 117-126, 2017 Feb.
Article in French | MEDLINE | ID: mdl-28111040

ABSTRACT

Diffuse gliomas represent the most common primary central nervous system (CNS) tumors in adults and children alike. Glioblastoma is the most frequent and malignant form of diffuse glioma with a median overall survival of 15 months despite aggressive treatments. New therapeutic approaches are needed to prolong survival in this always fatal disease. The CNS has been considered for a long time as an immune privileged organ, in part because of the existence of the blood-brain barrier. Nonetheless, immunotherapy is a novel approach in the therapeutic management of glioma patients, which has shown promising results in several clinical trials, especially in the adult population. Vaccination, with or without dendritic cells, blockade of the immune checkpoints, and adoptive T cell transfer are the most studied modalities of diffuse glioma immunotherapy. The future most likely resides in combinatorial approaches, with administration of conventional treatments (surgery, radiochemotherapy) and immunotherapy following yet to determine schedules.


Subject(s)
Brain Neoplasms/therapy , Glioblastoma/therapy , Glioma/therapy , Immunotherapy/methods , Adult , Animals , Brain Neoplasms/drug therapy , Brain Neoplasms/immunology , Cancer Vaccines/therapeutic use , Child , Combined Modality Therapy , Drug Screening Assays, Antitumor , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , ErbB Receptors/immunology , Genetic Therapy , Glioblastoma/drug therapy , Glioblastoma/immunology , Glioma/drug therapy , Glioma/immunology , Humans , Immunotherapy, Adoptive , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Mice, Transgenic , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , T-Lymphocyte Subsets/immunology , Tumor Escape
18.
Biochem Biophys Res Commun ; 478(3): 1274-9, 2016 09 23.
Article in English | MEDLINE | ID: mdl-27553275

ABSTRACT

A point mutation in isocitrate dehydrogenase 1 (IDH1) and IDH2 is directly linked to the pathogenesis of certain types of tumors. To detect this mutation, several antibodies that can distinguish between mutant and wild-type enzymes have been established. One of which, MsMab-1, has a unique multi-specific character against several types of mutated IDH1/2. This promiscuous character is in remarkable contrast to the highly specific antigen recognition typically observed with a monoclonal antibody. We solved the crystal structure of MsMab-1 Fab fragment in complex with either IDH1 or IDH2-derived peptides. Based on the structure, it became clear that the peptide-binding pocket of the antibody is highly complementary to the core determinant shared between the IDH1 and IDH2, while leaving just enough space for the side chain of the pathogenic but not the wild-type amino acids located in the mutation position. Clarification of the molecular basis for the peculiar binding characteristics of MsMab-1 in atomic detail will help facilitating its diagnostic application, and may be used to develop better diagnostic reagents through structure-guided protein engineering.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antibody Specificity/immunology , Isocitrate Dehydrogenase/immunology , Peptides/chemistry , Peptides/immunology , Amino Acid Sequence , Amino Acid Substitution , Animals , CHO Cells , Cricetinae , Cricetulus , Crystallography, X-Ray , Immunoglobulin Fab Fragments/chemistry , Ligands , Mice , Molecular Docking Simulation , Mutant Proteins/chemistry , Mutant Proteins/immunology
19.
Biochem Biophys Res Commun ; 466(4): 733-9, 2015 Oct 30.
Article in English | MEDLINE | ID: mdl-26381180

ABSTRACT

Isocitrate dehydrogenase 1 (IDH1) mutations have been detected in gliomas and other tumors. Although IDH1 catalyzes the oxidative carboxylation of isocitrate to α-ketoglutarate (α-KG) in cytosol, mutated IDH1 proteins possess the ability to change α-KG into the oncometabolite D-2-hydroxyglutarate (D-2HG). Several monoclonal antibodies (mAbs) specific for IDH1 mutations have been established, such as H09, IMab-1, and HMab-1 against IDH1-R132H, which is the most frequent IDH1 mutation in gliomas. In this study, we established a novel high-sensitive mAb HMab-2, which reacts with IDH1-R132H but not with wild type IDH1 in ELISA. HMab-2 reacted only with IDH1-R132H, not with wild type IDH1/2 and other IDH1/2 mutants in Western-blot analysis. Furthermore, HMab-2 recognized IDH1-R132H more sensitively compared with our previously established HMab-1. HMab-2 detected endogenous IDH1-R132H protein expressed in glioblastoma in immunohistochemical analysis. HMab-2 is expected to be useful for the diagnosis of IDH1-R132H-bearing tumors.


Subject(s)
Antibodies, Monoclonal/immunology , Central Nervous System Neoplasms/enzymology , Central Nervous System Neoplasms/genetics , Glioma/enzymology , Glioma/genetics , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/immunology , Mutant Proteins/genetics , Mutant Proteins/immunology , Amino Acid Substitution , Animals , Antibodies, Monoclonal/biosynthesis , Antibody Specificity , Cell Line, Tumor , Central Nervous System Neoplasms/diagnosis , Glioma/diagnosis , Humans , Hybridomas/immunology , Immunohistochemistry , Mice , Mice, Inbred BALB C , Recombinant Proteins/genetics , Recombinant Proteins/immunology
20.
Brain Tumor Pathol ; 32(4): 237-44, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26006098

ABSTRACT

Glioma cells with the isocitrate dehydrogenase (IDH) 1 G395A mutation are strongly immunopositive for mIDH1(R132H), an antibody against mutant IDH1(R132H) (clone H09). However, we encountered some gliomas which were ambiguously positive for mIDH1(R132H) despite having the IDH1 G395A mutation. The aim of this study was to establish an evaluation procedure of IDH1 status by immunohistochemistry. Forty-three diffusely infiltrating gliomas were studied, and four of eight anaplastic oligoastrocytomas with the IDH1 G395A mutation were modestly or weakly positive for both the mIDH1(R132H) and an antibody against wild type IDH1, RcMab-1. Based on our staining results, the IDH1 expression of both wild and mutated types seemed to be codominant and also to be evenly suppressed under a certain condition. We propose a procedure for determining IDH1 status. If a glioma is weakly positive for mIDH1(R132H), immunohistochemistry for RcMab-1 should be performed. If the tumor cells are strongly positive for RcMab-1, the IDH1 G395A mutation is judged to be absent on the grounds that IDH1 expression is not suppressed. If the tumor cells are weakly positive for both mIDH1(R132H) and RcMab-1, then a conclusion should be made after DNA sequencing. This procedure is useful for practical evaluation of IDH1 status.


Subject(s)
Antibodies, Monoclonal , Brain Neoplasms/genetics , Gene Expression Regulation, Neoplastic/genetics , Gene Expression/genetics , Glioma/genetics , Immunohistochemistry/methods , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/immunology , Mutation , Adolescent , Adult , Female , Humans , Male , Middle Aged , Sequence Analysis, DNA , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...