Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Type of study
Publication year range
1.
Oncol Rep ; 52(4)2024 Oct.
Article in English | MEDLINE | ID: mdl-39155864

ABSTRACT

The immune system is integral to the surveillance and eradication of tumor cells. Interactions between the natural killer group 2 member D (NKG2D) receptor and its ligands (NKG2DLs) are vital for activating NKG2D receptor­positive immune cells, such as natural killer cells. This activation enables these cells to identify and destroy tumor cells presenting with NKG2DLs, which is an essential aspect of tumor immunity. However, tumor immune escape is facilitated by soluble NKG2DL (sNKG2DL) shed from the surface of tumor cells. The production of sNKG2DL is predominantly regulated by metalloproteinases [a disintegrin and metalloproteinases (ADAM) and matrix metalloproteinase (MMP) families] and exosomes. sNKG2DL not only diminish immune recognition on the tumor cell surface but also suppress the function of immune cells, such as NK cells, and reduce the expression of the NKG2D receptor. This process promotes immune evasion, progression, and metastasis of tumors. In this review, an in­depth summary of the mechanisms and factors that influence sNKG2DL production and their contribution to immune suppression within the tumor microenvironment are provided. Furthermore, due to the significant link between sNKG2DLs and tumor progression and metastasis, they have great potential as novel biomarkers. Detectable via liquid biopsies, sNKG2DLs could assess tumor malignancy and prognosis, and act as pivotal targets for immunotherapy. This could lead to the discovery of new drugs or the enhancement of existing treatments. Thus, the application of sNKG2DLs in clinical oncology was explored, offering substantial theoretical support for the development of innovative immunotherapeutic strategies for sNKG2DLs.


Subject(s)
Killer Cells, Natural , NK Cell Lectin-Like Receptor Subfamily K , Neoplasms , Tumor Escape , Tumor Microenvironment , Humans , Neoplasms/immunology , Neoplasms/pathology , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Killer Cells, Natural/immunology , Tumor Microenvironment/immunology , Ligands , GPI-Linked Proteins/immunology , GPI-Linked Proteins/metabolism , Intercellular Signaling Peptides and Proteins
2.
Sci Rep ; 14(1): 17994, 2024 08 03.
Article in English | MEDLINE | ID: mdl-39097625

ABSTRACT

CD73 is a cell-surface ectoenzyme that hydrolyzes the conversion of extracellular adenosine monophosphate to adenosine, which in turn can promote resistance to immune checkpoint blockade therapy. Immune response may therefore be improved by targeting tumor CD73, and this possibility underlines the need to non-invasively assess tumor CD73 level. In this study, we developed a cysteine site-specific 89Zr-labeled anti-CD73 (89Zr-CD73) IgG immuno-PET technique that can image tumor CD73 expression in living bodies. Anti-CD73 IgG was reduced with tris(2-carboxyethyl)phosphine, underwent sulfohydryl moiety-specific conjugation with deferoxamine-maleimide, and was radiolabeled with 89Zr. CT26 mouse colon cancer cells, CT26/CD73 cells engineered to constitutively overexpress CD73, and 4T1.2 mouse breast cancer cells underwent cell binding assays and western blotting. Balb/c nude mice bearing tumors underwent 89Zr-CD73 IgG PET imaging and biodistribution studies. 89Zr-CD73 IgG showed 20-fold higher binding to overexpressing CT26/CD73 cells compared to low-expressing CT26 cells, and moderate expressing 4T1.2 cells showed uptake that was 38.9 ± 1.51% of CT26/CD73 cells. Uptake was dramatically suppressed by excess unlabeled antibody. CD73 content proportionately increased in CT26 and CT26/CD73 cell mixtures was associated with linear increases in 89Zr-CD73 IgG uptake. 89Zr-CD73 IgG PET/CT displayed clear accumulation in CT26/CD73 tumors with greater uptake compared to CT26 tumors (3.13 ± 1.70%ID/g vs. 1.27 ± 0.31%ID/g at 8 days; P = 0.04). Specificity was further supported by low CT26/CD73 tumor-to-blood ratio of 89Zr-isotype-IgG compared to 89Zr-CD73 IgG (0.48 ± 0.08 vs. 2.68 ± 0.52 at 4 days and 0.53 ± 0.07 vs. 4.81 ± 1.02 at 8 days; both P < 0.001). Immunoblotting and immunohistochemistry confirmed strong CD73 expression in CT26/CD73 tumors and low expression in CT26 tumors. 4T1.2 tumor mice also showed clear 89Zr-CD73 IgG accumulation at 8 days (3.75 ± 0.70%ID/g) with high tumor-to-blood ratio compared to 89Zr-isotype-IgG (4.91 ± 1.74 vs. 1.20 ± 0.28; P < 0.005). 89Zr-CD73 IgG specifically targeted CD73 on high expressing cancer cells in vitro and tumors in vivo. Thus, 89Zr-CD73 IgG immuno-PET may be useful for the non-invasive monitoring of CD73 expression in tumors of living subjects.


Subject(s)
5'-Nucleotidase , Colonic Neoplasms , Cysteine , Positron-Emission Tomography , Zirconium , Animals , 5'-Nucleotidase/metabolism , Zirconium/chemistry , Colonic Neoplasms/diagnostic imaging , Colonic Neoplasms/metabolism , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Mice , Cell Line, Tumor , Positron-Emission Tomography/methods , Cysteine/metabolism , Humans , Radioisotopes , Female , Mice, Inbred BALB C , Tissue Distribution , Mice, Nude , GPI-Linked Proteins/metabolism , GPI-Linked Proteins/immunology , Immunoglobulin G/immunology , Immunoglobulin G/metabolism
3.
Front Immunol ; 15: 1418539, 2024.
Article in English | MEDLINE | ID: mdl-39131159

ABSTRACT

CD177 plays an important role in the proliferation and differentiation of myeloid lineage cells including neutrophils, myelocytes, promyelocytes, megakaryocytes, and early erythroblasts in bone marrow. CD177 deficiency is a common phenotype in humans. Our previous studies revealed genetic mechanisms of human CD177 deficiency and expression variations. Up to now, immune functions of CD177 remain undefined. In the current study, we revealed human IgG as a ligand for CD177 by using flow cytometry, bead-rosette formation, and surface plasmon resonance (SPR) assays. In addition, we show that CD177 variants affect the binding capacity of CD177 for human IgG. Furthermore, we show that the CD177 genetic variants significantly affect antibody-dependent cell-mediated cytotoxicity (ADCC) function. The demonstration of CD177 as a functional IgG Fc-receptor may provide new insights into CD177 immune function and genetic mechanism underlying CD177 as biomarkers for human diseases.


Subject(s)
GPI-Linked Proteins , Immunoglobulin G , Humans , Immunoglobulin G/immunology , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , GPI-Linked Proteins/immunology , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, Cell Surface/immunology , Antibody-Dependent Cell Cytotoxicity , Receptors, IgG/genetics , Receptors, IgG/metabolism , Isoantigens/immunology , Isoantigens/genetics , Genetic Variation , Receptors, Fc/metabolism , Receptors, Fc/genetics , Protein Binding
4.
MAbs ; 16(1): 2387240, 2024.
Article in English | MEDLINE | ID: mdl-39113562

ABSTRACT

Prostate stem cell antigen (PSCA) is expressed in all stages of prostate cancer, including in advanced androgen-independent tumors and bone metastasis. PSCA may associate with prostate carcinogenesis and lineage plasticity in prostate cancer. PSCA is also a promising theranostic marker for a variety of other solid tumors, including pancreatic adenocarcinoma and renal cell carcinoma. Here, we identified a novel fully human PSCA antibody using phage display methodology. The structure-based affinity maturation yielded a high-affinity binder, F12, which is highly specific and does not bind to 6,000 human membrane proteins based on a membrane proteome array assay. F12 targets PSCA amino acids 63-69 as tested by the peptide scanning microarray, and it cross-reacts with the murine PSCA. IgG1 F12 efficiently internalizes into PSCA-expressing tumor cells. The antimitotic reagent monomethyl auristatin E (MMAE)-conjugated IgG1 F12 (ADC, F12-MMAE) exhibits dose-dependent efficacy and specificity in a human prostate cancer PC-3-PSCA xenograft NSG mouse model. This is a first reported ADC based on a fully human PSCA antibody and MMAE that is characterized in a xenograft murine model, which warrants further optimizations and investigations in additional preclinical tumor models, including prostate and other solid tumors.


Subject(s)
Antigens, Neoplasm , GPI-Linked Proteins , Immunoconjugates , Neoplasm Proteins , Prostatic Neoplasms , Xenograft Model Antitumor Assays , Humans , Male , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Prostatic Neoplasms/immunology , Immunoconjugates/pharmacology , Animals , Antigens, Neoplasm/immunology , Mice , GPI-Linked Proteins/immunology , Neoplasm Proteins/immunology , Neoplasm Proteins/antagonists & inhibitors , Cell Line, Tumor , Oligopeptides/immunology , Oligopeptides/pharmacology , Immunoglobulin G/immunology , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology
5.
Front Immunol ; 15: 1363156, 2024.
Article in English | MEDLINE | ID: mdl-38953028

ABSTRACT

Introduction: Human Herpesvirus 6B (HHV-6B) impedes host immune responses by downregulating class I MHC molecules (MHC-I), hindering antigen presentation to CD8+ T cells. Downregulation of MHC-I disengages inhibitory receptors on natural killer (NK) cells, resulting in activation and killing of the target cell if NK cell activating receptors such as NKG2D have engaged stress ligands upregulated on the target cells. Previous work has shown that HHV-6B downregulates three MHC-like stress ligands MICB, ULBP1, and ULBP3, which are recognized by NKG2D. The U20 glycoprotein of the related virus HHV-6A has been implicated in the downregulation of ULBP1, but the precise mechanism remains undetermined. Methods: We set out to investigate the role of HHV-6B U20 in modulating NK cell activity. We used HHV-6B U20 expressed as a recombinant protein or transduced into target cells, as well as HHV-6B infection, to investigate binding interactions with NK cell ligands and receptors and to assess effects on NK cell activation. Small-angle X-ray scattering was used to align molecular models derived from machine-learning approaches. Results: We demonstrate that U20 binds directly to ULBP1 with sub-micromolar affinity. Transduction of U20 decreases NKG2D binding to ULBP1 at the cell surface but does not decrease ULBP1 protein levels, either at the cell surface or in toto. HHV-6B infection and soluble U20 have the same effect. Transduction of U20 blocks NK cell activation in response to cell-surface ULBP1. Structural modeling of the U20 - ULBP1 complex indicates some similarities to the m152-RAE1γ complex.


Subject(s)
GPI-Linked Proteins , Herpesvirus 6, Human , Killer Cells, Natural , Lymphocyte Activation , NK Cell Lectin-Like Receptor Subfamily K , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Herpesvirus 6, Human/immunology , GPI-Linked Proteins/metabolism , GPI-Linked Proteins/immunology , NK Cell Lectin-Like Receptor Subfamily K/metabolism , NK Cell Lectin-Like Receptor Subfamily K/immunology , Lymphocyte Activation/immunology , Protein Binding , Viral Proteins/immunology , Viral Proteins/metabolism , Glycoproteins/immunology , Glycoproteins/metabolism , Intracellular Signaling Peptides and Proteins
6.
Cancer Lett ; 598: 217098, 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-38969159

ABSTRACT

Immune escape is the main reason that immunotherapy is ineffective in hepatocellular carcinoma (HCC). Here, this study illustrates a pathway mediated by neutrophil extracellular traps (NETs) that can promote immune escape of HCC. Mechanistically, we demonstrated that NETs up-regulated CD73 expression through activating Notch2 mediated nuclear factor kappa B (NF-κB) pathway, promoting regulatory T cells (Tregs) infiltration to mediate immune escape of HCC. In addition, we found the similar results in mouse HCC models by hydrodynamic plasmid transfection. The treatment of deoxyribonuclease I (DNase I) could inhibit the action of NETs and improve the therapeutic effect of anti-programmed cell death protein 1 (PD-1). In summary, our results revealed that targeting of NETs was a promising treatment to improve the therapeutic effect of anti-PD-1.


Subject(s)
5'-Nucleotidase , Carcinoma, Hepatocellular , Extracellular Traps , Liver Neoplasms , Receptor, Notch2 , Tumor Escape , Up-Regulation , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Animals , Humans , Extracellular Traps/immunology , Extracellular Traps/metabolism , Mice , 5'-Nucleotidase/genetics , 5'-Nucleotidase/metabolism , 5'-Nucleotidase/immunology , Receptor, Notch2/metabolism , Receptor, Notch2/genetics , GPI-Linked Proteins/metabolism , GPI-Linked Proteins/genetics , GPI-Linked Proteins/immunology , Cell Line, Tumor , NF-kappa B/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Signal Transduction , Male , Programmed Cell Death 1 Receptor/metabolism
7.
Proc Natl Acad Sci U S A ; 121(31): e2403002121, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39047033

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) represents a challenge in oncology, with limited treatment options for advanced-stage patients. Chimeric antigen receptor T cell (CAR T) therapy targeting mesothelin (MSLN) shows promise, but challenges such as the hostile immunosuppressive tumor microenvironment (TME) hinder its efficacy. This study explores the synergistic potential of combining proton radiation therapy (RT) with MSLN-targeting CAR T therapy in a syngeneic PDAC model. Proton RT significantly increased MSLN expression in tumor cells and caused a significant increase in CAR T cell infiltration into tumors. The combination therapy reshaped the immunosuppressive TME, promoting antitumorigenic M1 polarized macrophages and reducing myeloid-derived suppressor cells (MDSC). In a flank PDAC model, the combination therapy demonstrated superior attenuation of tumor growth and improved survival compared to individual treatments alone. In an orthotopic PDAC model treated with image-guided proton RT, tumor growth was significantly reduced in the combination group compared to the RT treatment alone. Further, the combination therapy induced an abscopal effect in a dual-flank tumor model, with increased serum interferon-γ levels and enhanced proliferation of extratumoral CAR T cells. In conclusion, combining proton RT with MSLN-targeting CAR T therapy proves effective in modulating the TME, enhancing CAR T cell trafficking, and exerting systemic antitumor effects. Thus, this combinatorial approach could present a promising strategy for improving outcomes in unresectable PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal , GPI-Linked Proteins , Immunotherapy, Adoptive , Mesothelin , Pancreatic Neoplasms , Receptors, Chimeric Antigen , Tumor Microenvironment , Animals , Pancreatic Neoplasms/therapy , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/radiotherapy , Pancreatic Neoplasms/pathology , Mice , GPI-Linked Proteins/metabolism , GPI-Linked Proteins/immunology , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , Immunotherapy, Adoptive/methods , Tumor Microenvironment/immunology , Humans , Carcinoma, Pancreatic Ductal/therapy , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/radiotherapy , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Proton Therapy/methods , Combined Modality Therapy , T-Lymphocytes/immunology , Female
8.
Nat Med ; 30(6): 1636-1644, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38867077

ABSTRACT

Despite recent therapeutic advances, metastatic castration-resistant prostate cancer (mCRPC) remains lethal. Chimeric antigen receptor (CAR) T cell therapies have demonstrated durable remissions in hematological malignancies. We report results from a phase 1, first-in-human study of prostate stem cell antigen (PSCA)-directed CAR T cells in men with mCRPC. The starting dose level (DL) was 100 million (M) CAR T cells without lymphodepletion (LD), followed by incorporation of LD. The primary end points were safety and dose-limiting toxicities (DLTs). No DLTs were observed at DL1, with a DLT of grade 3 cystitis encountered at DL2, resulting in addition of a new cohort using a reduced LD regimen + 100 M CAR T cells (DL3). No DLTs were observed in DL3. Cytokine release syndrome of grade 1 or 2 occurred in 5 of 14 treated patients. Prostate-specific antigen declines (>30%) occurred in 4 of 14 patients, as well as radiographic improvements. Dynamic changes indicating activation of peripheral blood endogenous and CAR T cell subsets, TCR repertoire diversity and changes in the tumor immune microenvironment were observed in a subset of patients. Limited persistence of CAR T cells was observed beyond 28 days post-infusion. These results support future clinical studies to optimize dosing and combination strategies to improve durable therapeutic outcomes. ClinicalTrials.gov identifier NCT03873805 .


Subject(s)
Antigens, Neoplasm , GPI-Linked Proteins , Immunotherapy, Adoptive , Neoplasm Proteins , Prostatic Neoplasms, Castration-Resistant , Humans , Male , Prostatic Neoplasms, Castration-Resistant/therapy , Prostatic Neoplasms, Castration-Resistant/immunology , Prostatic Neoplasms, Castration-Resistant/pathology , Aged , Middle Aged , Antigens, Neoplasm/immunology , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , GPI-Linked Proteins/immunology , Neoplasm Proteins/immunology , Receptors, Chimeric Antigen/immunology , Neoplasm Metastasis , T-Lymphocytes/immunology , T-Lymphocytes/transplantation , Prostate-Specific Antigen/blood
10.
Int J Mol Sci ; 25(9)2024 Apr 27.
Article in English | MEDLINE | ID: mdl-38732017

ABSTRACT

Intelectins belong to a family of lectins with specific and transitory carbohydrate interaction capabilities. These interactions are related to the activity of agglutinating pathogens, as intelectins play a significant role in immunity. Despite the prominent immune defense function of intelectins, limited information about its structural characteristics and carbohydrate interaction properties is available. This study investigated an intelectin transcript identified in RNA-seq data obtained from the South American lungfish (Lepidosiren paradoxa), namely LpITLN2-B. The structural analyses predicted LpITLN2-B to be a homo-trimeric globular protein with the fibrinogen-like functional domain (FReD), exhibiting a molecular mass of 57 kDa. The quaternary structure is subdivided into three monomers, A, B, and C, and each domain comprises 11 ß-sheets: an anti-parallel ß-sheet, a ß-hairpin, and a disordered ß-sheet structure. Molecular docking demonstrates a significant interaction with disaccharides rather than monosaccharides. The preferential interaction with disaccharides highlights the potential interaction with pathogen molecules, such as LPS and Poly(I:C). The hemagglutination assay inhibited lectins activity, especially maltose and sucrose, highlighting lectin activity in L. paradoxa samples. Overall, our results show the potential relevance of LpITLN2-B in L. paradoxa immune defense against pathogens.


Subject(s)
Fish Proteins , Fishes , Immunity, Innate , Lectins , Animals , Lectins/chemistry , Lectins/metabolism , Lectins/immunology , Lectins/genetics , Fishes/immunology , Fishes/genetics , Fish Proteins/genetics , Fish Proteins/chemistry , Fish Proteins/immunology , Fish Proteins/metabolism , Molecular Docking Simulation , Amino Acid Sequence , GPI-Linked Proteins/chemistry , GPI-Linked Proteins/metabolism , GPI-Linked Proteins/genetics , GPI-Linked Proteins/immunology
11.
Adv Sci (Weinh) ; 11(29): e2403414, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38790136

ABSTRACT

The colon is the largest compartment of the immune system, with innate immune cells exposed to antigens in the environment. However, the mechanisms by which the innate immune system is instigated are poorly defined in colorectal cancer (CRC). Here, a population of CD16+ neutrophils that specifically accumulate in CRC tumor tissues by imaging mass cytometry (IMC), immune fluorescence, and flow cytometry, which demonstrated pro-tumor activity by disturbing natural killer (NK) cells are identified. It is found that these CD16+ neutrophils possess abnormal cholesterol accumulation due to activation of the CD16/TAK1/NF-κB axis, which upregulates scavenger receptors for cholesterol intake including CD36 and LRP1. Consequently, these region-specific CD16+ neutrophils not only competitively inhibit cholesterol intake of NK cells, which interrupts NK lipid raft formation and blocks their antitumor signaling but also release neutrophil extracellular traps (NETs) to induce the death of NK cells. Furthermore, CD16-knockout reverses the pro-tumor activity of neutrophils and restored NK cell cytotoxicity. Collectively, the findings suggest that CRC region-specific CD16+ neutrophils can be a diagnostic marker and potential therapeutic target for CRC.


Subject(s)
Colorectal Neoplasms , Killer Cells, Natural , Neutrophils , Receptors, IgG , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Receptors, IgG/metabolism , Receptors, IgG/immunology , Humans , Mice , Disease Progression , Animals , Disease Models, Animal , GPI-Linked Proteins/metabolism , GPI-Linked Proteins/immunology
12.
Front Immunol ; 15: 1341013, 2024.
Article in English | MEDLINE | ID: mdl-38655263

ABSTRACT

Recombinant Factor VIII-Fc fusion protein (rFVIIIFc) is an enhanced half-life therapeutic protein product used for the management of hemophilia A. Recent studies have demonstrated that rFVIIIFc interacts with Fc gamma receptors (FcγR) resulting in the activation or inhibition of various FcγR-expressing immune cells. We previously demonstrated that rFVIIIFc, unlike recombinant Factor IX-Fc (rFIXFc), activates natural killer (NK) cells via Fc-mediated interactions with FcγRIIIA (CD16). Additionally, we showed that rFVIIIFc activated CD16+ NK cells to lyse a FVIII-specific B cell clone. Here, we used human NK cell lines and primary NK cells enriched from peripheral blood leukocytes to study the role of the FVIII moiety in rFVIIIFc-mediated NK cell activation. Following overnight incubation of NK cells with rFVIIIFc, cellular activation was assessed by measuring secretion of the inflammatory cytokine IFNγ by ELISA or by cellular degranulation. We show that anti-FVIII, anti-Fc, and anti-CD16 all inhibited indicating that these molecules were involved in rFVIIIFc-mediated NK cell activation. To define which domains of FVIII were involved, we used antibodies that are FVIII domain-specific and demonstrated that blocking FVIII C1 or C2 domain-mediated membrane binding potently inhibited rFVIIIFc-mediated CD16+ NK cell activation, while targeting the FVIII heavy chain domains did not. We also show that rFVIIIFc binds CD16 with about five-fold higher affinity than rFIXFc. Based on our results we propose that FVIII light chain-mediated membrane binding results in tethering of the fusion protein to the cell surface, and this, together with increased binding affinity for CD16, allows for Fc-CD16 interactions to proceed, resulting in NK cellular activation. Our working model may explain our previous results where we observed that rFVIIIFc activated NK cells via CD16, whereas rFIXFc did not despite having identical IgG1 Fc domains.


Subject(s)
Factor VIII , GPI-Linked Proteins , Immunoglobulin Fc Fragments , Killer Cells, Natural , Lymphocyte Activation , Receptors, IgG , Recombinant Fusion Proteins , Humans , Cell Degranulation/immunology , Factor VIII/chemistry , Factor VIII/immunology , GPI-Linked Proteins/immunology , GPI-Linked Proteins/metabolism , Hemophilia A/immunology , Hemophilia A/drug therapy , Immunoglobulin Fc Fragments/immunology , Interferon-gamma/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lymphocyte Activation/immunology , Lymphocyte Activation/drug effects , Protein Binding , Receptors, IgG/metabolism , Receptors, IgG/immunology
13.
Pharmacol Res ; 203: 107186, 2024 May.
Article in English | MEDLINE | ID: mdl-38641176

ABSTRACT

Chimeric antigen receptor (CAR)-modified T cell therapy has achieved remarkable efficacy in treating hematological malignancies, but it confronts many challenges in treating solid tumors, such as the immunosuppressive microenvironment of the solid tumors. These factors reduce the antitumor activity of CAR-T cells in clinical trials. Therefore, we used the immunocytokine interleukin-12 (IL-12) to enhance the efficacy of CAR-T cell therapy. In this study, we engineered CAR-IL12R54 T cells that targeted mesothelin (MSLN) and secreted a single-chain IL-12 fused to a scFv fragment R54 that recognized a different epitope on mesothelin. The evaluation of the anti-tumor activity of the CAR-IL12R54 T cells alone or in combination with anti-PD-1 antibody in vitro and in vivo was followed by the exploration of the functional mechanism by which the immunocytokine IL-12 enhanced the antitumor activity. CAR-IL12R54 T cells had potency to lyse mesothelin positive tumor cells in vitro. In vivo studies demonstrated that CAR-IL12R54 T cells were effective in controlling the growth of established tumors in a xenograft mouse model with fewer side effects than CAR-T cells that secreted naked IL-12. Furthermore, combination of PD-1 blockade antibody with CAR-IL12R54 T cells elicited durable anti-tumor responses. Mechanistic studies showed that IL12R54 enhanced Interferon-γ (IFN-γ) production and dampened the activity of regulatory T cells (Tregs). IL12R54 also upregulated CXCR6 expression in the T cells through the NF-κB pathway, which facilitated T cell infiltration and persistence in the tumor tissues. In summary, the studies provide a good therapeutic option for the clinical treatment of solid tumors.


Subject(s)
Immunotherapy, Adoptive , Interleukin-12 , Mesothelin , Receptors, Chimeric Antigen , Animals , Interleukin-12/immunology , Interleukin-12/genetics , Humans , Immunotherapy, Adoptive/methods , Immunotherapy, Adoptive/adverse effects , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/genetics , Cell Line, Tumor , Mice , Xenograft Model Antitumor Assays , Female , GPI-Linked Proteins/immunology , GPI-Linked Proteins/genetics , GPI-Linked Proteins/antagonists & inhibitors , Tumor Microenvironment/immunology , Neoplasms/immunology , Neoplasms/therapy , Receptors, Interleukin-12/genetics , Receptors, Interleukin-12/immunology , T-Lymphocytes/immunology
14.
Clin Immunol ; 263: 110223, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38636890

ABSTRACT

Idiopathic severe aplastic anemia (SAA) is a disease of bone marrow failure caused by T-cell-induced destruction of hematopoietic stem and progenitor cells (HSPCs), however the mechanism remains unclear. We performed single-cell RNA sequencing of PBMCs and BMMCs from SAA patients and healthy donors and identified a CD8+ T cell subset with a tissue residency phenotype (Trm) in bone marrow that exhibit high IFN-γ and FasL expression and have a higher ability to induce apoptosis in HSPCs in vitro through FasL expression. CD8+ Trm cells were induced by IL-15 presented by IL-15Rα on monocytes, especially CD16+ monocytes, which were increased in SAA patients. CD16+ monocytes contributed to IL-15-induced CD38+CXCR6+ pre-Trm differentiation into CD8+ Trm cells, which can be inhibited by the CD38 inhibitor 78c. Our results demonstrate that IL-15-induced CD8+ Trm cells are pathogenic cells that mediate HSPC destruction in SAA patients and are therapeutic targets for future treatments.


Subject(s)
Anemia, Aplastic , CD8-Positive T-Lymphocytes , GPI-Linked Proteins , Hematopoietic Stem Cells , Interleukin-15 , Monocytes , Receptors, IgG , Humans , Anemia, Aplastic/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , Interleukin-15/pharmacology , Interleukin-15/immunology , Receptors, IgG/metabolism , Receptors, IgG/immunology , Monocytes/immunology , Monocytes/drug effects , Female , Male , Adult , Hematopoietic Stem Cells/immunology , GPI-Linked Proteins/metabolism , GPI-Linked Proteins/immunology , Middle Aged , Fas Ligand Protein/metabolism , Fas Ligand Protein/immunology , Young Adult , Adolescent , Interferon-gamma/immunology , Interferon-gamma/metabolism , Receptors, Interleukin-15/metabolism , Receptors, Interleukin-15/immunology , Apoptosis/drug effects , Cell Differentiation/immunology
15.
Future Oncol ; 20(19): 1367-1380, 2024.
Article in English | MEDLINE | ID: mdl-38652041

ABSTRACT

CD39 is the rate-limiting enzyme for the molecular signal cascade leading to the generation of ADP and adenosine monophosphate (AMP). In conjunction with CD73, CD39 converts adenosine triphosphate (ATP) to ADP and AMP, which leads to the accumulation of immunosuppressive adenosine in the tumor microenvironment. This review focuses on the role of CD39 and CD73 in immune response and malignant progression, including the expression of CD39 within the tumor microenvironment and its relationship to immune effector cells, and its role in antigen presentation. The role of CD39- and CD73-targeting therapeutics and cancer-directed clinical trials investigating CD39 modulation are also explored.


[Box: see text].


Subject(s)
5'-Nucleotidase , Apyrase , Neoplasms , Tumor Microenvironment , Humans , 5'-Nucleotidase/metabolism , 5'-Nucleotidase/immunology , Apyrase/metabolism , Apyrase/immunology , Neoplasms/immunology , Neoplasms/metabolism , Tumor Microenvironment/immunology , Animals , GPI-Linked Proteins/immunology , GPI-Linked Proteins/metabolism , Antigens, CD/metabolism , Antigens, CD/immunology , Antigen Presentation/immunology
16.
Vox Sang ; 119(7): 712-719, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38597364

ABSTRACT

BACKGROUND AND OBJECTIVES: The isolation of neutrophils and subsequent detection of anti-human neutrophil antigens (HNA) antibodies are crucial in clinical medicine for the diagnosis of autoimmune neutropenia, neonatal alloimmune neutropenia (NAIN) and transfusion-related acute lung injury (TRALI). This study reports two cases of maternal anti-Fc-gamma-receptor-IIIb (FcγRIIIb) isoimmunization without NAIN symptoms and compares the efficiency of immunomagnetic negative selection (IMNS) with traditional dextran/Ficoll for neutrophil isolation in HNA serological assays. MATERIALS AND METHODS: Investigating two cases of maternal anti-FcγRIIIb isoimmunization, neutrophils from three donors were isolated from 8 mL of whole blood using IMNS and dextran/Ficoll. Serological assays included the granulocyte agglutination and immunofluorescence test, monoclonal antibody immobilization of granulocyte antigens and the LABScreen Multi (One Lambda). IMNS and dextran/Ficoll were compared in terms of cell yield, viability, time, cost and purity. RESULTS: Maternal anti-FcγRIIIb isoantibodies with FCGR3B gene deletion were detected in both cases. Newborns and fathers exhibited specific gene combinations: FCGR3B*02/FCGR3B*02 (Case 1) and FCGR3B*02/FCGR3B*03 (Case 2). IMNS outperformed dextran/Ficoll, yielding four times more neutrophils (average neutrophil counts: 18.5 × 103/µL vs. 4.5 × 103/µL), efficiently removing non-neutrophil cells and reducing processing time (30-40 min vs. 70-90 min), although it incurred a higher cost (2.7 times). CONCLUSION: Two cases of maternal anti-FcγRIIIb isoantibodies, unrelated to NAIN, were identified. Although neutropenia has not been described in these cases, we emphasize the importance of identifying asymptomatic cases with the potential for severe neutropenia. Additionally, IMNS is introduced as a rapid, high-yield, high-purity neutrophil isolation technique, beneficial for serological assays detecting anti-HNA antibodies.


Subject(s)
Isoantibodies , Neutrophils , Receptors, IgG , Humans , Neutrophils/immunology , Female , Receptors, IgG/immunology , Isoantibodies/immunology , Isoantibodies/blood , Infant, Newborn , GPI-Linked Proteins/immunology , Male , Immunomagnetic Separation/methods , Adult , Pregnancy , Neutropenia/immunology , Neutropenia/blood
17.
J Clin Invest ; 134(10)2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38530357

ABSTRACT

Despite widespread utilization of immunotherapy, treating immune-cold tumors remains a challenge. Multiomic analyses and experimental validation identified the OTUD4/CD73 proteolytic axis as a promising target in treating immune-suppressive triple negative breast cancer (TNBC). Mechanistically, deubiquitylation of CD73 by OTUD4 counteracted its ubiquitylation by TRIM21, resulting in CD73 stabilization inhibiting tumor immune responses. We further demonstrated the importance of TGF-ß signaling for orchestrating the OTUD4/CD73 proteolytic axis within tumor cells. Spatial transcriptomics profiling discovered spatially resolved features of interacting malignant and immune cells pertaining to expression levels of OTUD4 and CD73. In addition, ST80, a newly developed inhibitor, specifically disrupted proteolytic interaction between CD73 and OTUD4, leading to reinvigoration of cytotoxic CD8+ T cell activities. In preclinical models of TNBC, ST80 treatment sensitized refractory tumors to anti-PD-L1 therapy. Collectively, our findings uncover what we believe to be a novel strategy for targeting the immunosuppressive OTUD4/CD73 proteolytic axis in treating immune-suppressive breast cancers with the inhibitor ST80.


Subject(s)
5'-Nucleotidase , Proteolysis , Triple Negative Breast Neoplasms , Animals , Female , Humans , Mice , 5'-Nucleotidase/genetics , 5'-Nucleotidase/immunology , 5'-Nucleotidase/antagonists & inhibitors , Cell Line, Tumor , GPI-Linked Proteins/immunology , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , GPI-Linked Proteins/antagonists & inhibitors , Neoplasm Proteins/immunology , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasm Proteins/antagonists & inhibitors , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Ubiquitination , Ubiquitin-Specific Proteases
18.
Nat Mater ; 23(6): 844-853, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38448658

ABSTRACT

Lymph nodes are crucial organs of the adaptive immune system, orchestrating T cell priming, activation and tolerance. T cell activity and function are highly regulated by lymph nodes, which have a unique structure harbouring distinct cells that work together to detect and respond to pathogen-derived antigens. Here we show that implanted patient-derived freeze-dried lymph nodes loaded with chimeric antigen receptor T cells improve delivery to solid tumours and inhibit tumour recurrence after surgery. Chimeric antigen receptor T cells can be effectively loaded into lyophilized lymph nodes, whose unaltered meshwork and cytokine and chemokine contents promote chimeric antigen receptor T cell viability and activation. In mouse models of cell-line-derived human cervical cancer and patient-derived pancreatic cancer, delivery of chimeric antigen receptor T cells targeting mesothelin via the freeze-dried lymph nodes is more effective in preventing tumour recurrence when compared to hydrogels containing T-cell-supporting cytokines. This tissue-mediated cell delivery strategy holds promise for controlled release of various cells and therapeutics with long-term activity and augmented function.


Subject(s)
Freeze Drying , Lymph Nodes , Mesothelin , Receptors, Chimeric Antigen , Animals , Humans , Mice , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , Lymph Nodes/immunology , T-Lymphocytes/immunology , T-Lymphocytes/cytology , Cell Line, Tumor , Female , GPI-Linked Proteins/immunology , GPI-Linked Proteins/metabolism , Pancreatic Neoplasms/therapy , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology
19.
Clin Cancer Res ; 30(9): 1859-1877, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38393682

ABSTRACT

PURPOSE: Targeting solid tumors with chimeric antigen receptor (CAR) T cells remains challenging due to heterogenous target antigen expression, antigen escape, and the immunosuppressive tumor microenvironment (TME). Pancreatic cancer is characterized by a thick stroma generated by cancer-associated fibroblasts (CAF), which may contribute to the limited efficacy of mesothelin-directed CAR T cells in early-phase clinical trials. To provide a more favorable TME for CAR T cells to target pancreatic ductal adenocarcinoma (PDAC), we generated T cells with an antimesothelin CAR and a secreted T-cell-engaging molecule (TEAM) that targets CAF through fibroblast activation protein (FAP) and engages T cells through CD3 (termed mesoFAP CAR-TEAM cells). EXPERIMENTAL DESIGN: Using a suite of in vitro, in vivo, and ex vivo patient-derived models containing cancer cells and CAF, we examined the ability of mesoFAP CAR-TEAM cells to target PDAC cells and CAF within the TME. We developed and used patient-derived ex vivo models, including patient-derived organoids with patient-matched CAF and patient-derived organotypic tumor spheroids. RESULTS: We demonstrated specific and significant binding of the TEAM to its respective antigens (CD3 and FAP) when released from mesothelin-targeting CAR T cells, leading to T-cell activation and cytotoxicity of the target cell. MesoFAP CAR-TEAM cells were superior in eliminating PDAC and CAF compared with T cells engineered to target either antigen alone in our ex vivo patient-derived models and in mouse models of PDAC with primary or metastatic liver tumors. CONCLUSIONS: CAR-TEAM cells enable modification of tumor stroma, leading to increased elimination of PDAC tumors. This approach represents a promising treatment option for pancreatic cancer.


Subject(s)
CD3 Complex , Endopeptidases , GPI-Linked Proteins , Immunotherapy, Adoptive , Mesothelin , Pancreatic Neoplasms , Receptors, Chimeric Antigen , Tumor Microenvironment , Xenograft Model Antitumor Assays , Humans , Animals , Mice , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/therapy , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/metabolism , Tumor Microenvironment/immunology , Immunotherapy, Adoptive/methods , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , CD3 Complex/immunology , CD3 Complex/metabolism , GPI-Linked Proteins/immunology , GPI-Linked Proteins/metabolism , Cell Line, Tumor , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/therapy , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/immunology , Membrane Proteins/immunology , Membrane Proteins/metabolism , Serine Endopeptidases/immunology , Serine Endopeptidases/metabolism , Adenocarcinoma/immunology , Adenocarcinoma/therapy , Adenocarcinoma/pathology
20.
Semin Immunopathol ; 45(4-6): 493-507, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38170255

ABSTRACT

Glycoprotein 2 (GP2) is a widely distributed protein in the digestive tract, contributing to mucosal barrier maintenance, immune homeostasis, and antigen-specific immune response, while also being linked to inflammatory bowel disease (IBD) pathogenesis. This review sheds light on the extensive distribution of GP2 within the gastrointestinal tract and its intricate interplay with the immune system. Furthermore, the significance of GP2 autoantibodies in diagnosing and categorizing IBD is underscored, alongside the promising therapeutic avenues for modulating GP2 to regulate immunity and maintain mucosal balance.


Subject(s)
GPI-Linked Proteins , Inflammatory Bowel Diseases , Intestinal Mucosa , Animals , Humans , Autoantibodies/immunology , Disease Susceptibility , GPI-Linked Proteins/metabolism , GPI-Linked Proteins/immunology , Immunity, Mucosal , Inflammation/immunology , Inflammation/metabolism , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/etiology , Inflammatory Bowel Diseases/diagnosis , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology
SELECTION OF CITATIONS
SEARCH DETAIL