Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35.408
Filter
1.
Front Immunol ; 15: 1452810, 2024.
Article in English | MEDLINE | ID: mdl-39380986

ABSTRACT

The formation of a lytic immunological synapse (IS) is crucial for cytotoxic lymphocytes to accurately target and effectively eliminate malignant cells. While significant attention has been focused on the lymphocyte side of the IS, particularly its role as a secretory domain for lytic granules, the cancer cell side of the IS has remained relatively underexplored. Recent findings have revealed that cancer cells can rapidly polarize their actin cytoskeleton toward the IS upon interaction with natural killer (NK) cells, thereby evading NK cell-mediated cytotoxicity. In this Brief Research Report, we present preliminary findings suggesting that actin cytoskeleton remodeling at the cancer cell side of the IS is associated with the targeted secretion of small extracellular vesicles towards the interacting NK cell. We observed that multivesicular bodies (MVBs) preferentially accumulate in the synaptic region in cancer cells exhibiting synaptic accumulation of F-actin, compared to those lacking actin cytoskeleton remodeling. Extracellular immunofluorescence staining revealed increased surface exposure of CD63 at the cancer cell side of the IS, suggestive of the fusion of MVBs with the plasma membrane. This hypothesis was supported by a pH-sensitive probe demonstrating dynamic trafficking of CD63 to the extracellular region of the IS. Collectively, our data support the notion that cancer cells can engage in targeted secretion of extracellular vesicles in response to NK cell attack, underscoring the need for further research into the potential role of this process in facilitating cancer cell immune evasion.


Subject(s)
Immunological Synapses , Killer Cells, Natural , Humans , Immunological Synapses/metabolism , Immunological Synapses/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Actin Cytoskeleton/metabolism , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/metabolism , Multivesicular Bodies/metabolism , Multivesicular Bodies/immunology , Cell Line, Tumor , Tetraspanin 30/metabolism , Actins/metabolism , Extracellular Vesicles/immunology , Extracellular Vesicles/metabolism , Cytotoxicity, Immunologic
2.
Cancer Immunol Immunother ; 73(12): 239, 2024 Oct 03.
Article in English | MEDLINE | ID: mdl-39358647

ABSTRACT

Natural killer (NK) cells are frontline defenders against cancer and are capable of recognizing and eliminating tumor cells without prior sensitization or antigen presentation. Due to their unique HLA mismatch tolerance, they are ideal for adoptive cell therapy (ACT) because of their ability to minimize graft-versus-host-disease risk. The therapeutic efficacy of NK cells is limited in part by inhibitory immune checkpoint receptors, which are upregulated upon interaction with cancer cells and the tumor microenvironment. Overexpression of inhibitory receptors reduces NK cell-mediated cytotoxicity by impairing the ability of NK cells to secrete effector cytokines and cytotoxic granules. T-cell immunoreceptor with immunoglobulin and ITIM domains (TIGIT), a well-known checkpoint receptor involved in T-cell exhaustion, has recently been implicated in the exhaustion of NK cells. Overcoming TIGIT-mediated inhibition of NK cells may allow for a more potent antitumor response following ACT. Here, we describe a novel approach to TIGIT inhibition using self-delivering RNAi compounds (INTASYL™) that incorporates the features of RNAi and antisense technology. INTASYL compounds demonstrate potent activity and stability, are rapidly and efficiently taken up by cells, and can be easily incorporated into cell product manufacturing. INTASYL PH-804, which targets TIGIT, suppresses TIGIT mRNA and protein expression in NK cells, resulting in increased cytotoxic capacity and enhanced tumor cell killing in vitro. Delivering PH-804 to NK cells before ACT has emerged as a promising strategy to counter TIGIT inhibition, thereby improving the antitumor response. This approach offers the potential for more potent off-the-shelf products for adoptive cell therapy, particularly for hematological malignancies.


Subject(s)
Immunotherapy, Adoptive , Killer Cells, Natural , Neoplasms , Receptors, Immunologic , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/transplantation , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Receptors, Immunologic/immunology , Immunotherapy, Adoptive/methods , Neoplasms/therapy , Neoplasms/immunology , Cytotoxicity, Immunologic , RNA Interference , Cell Line, Tumor , RNA, Small Interfering/genetics
3.
Front Immunol ; 15: 1427519, 2024.
Article in English | MEDLINE | ID: mdl-39328404

ABSTRACT

Background: The relationship between chronic hepatitis B (CHB) infection and natural killer (NK) cell dysfunction is well-established, but the specific role of HBV viral antigens in driving NK cell impairment in patients with CHB remains unclear. This study investigates the modulatory effects of hepatitis B virus subviral particles (HBVsvp, a representative model for HBsAg) on the phenotypic regulation (activating and inhibitory receptors), cytokine production and cytotoxic potential of peripheral blood mononuclear cell-derived natural killer cells (PBMCs-derived NK cell), which contributes to NK cell dysfunction in CHB infection, potentially serving as an effective HBV immune evasion strategy by the virus. Methods: NK cells were isolated from peripheral blood of patients with CHB (n=5) and healthy individuals (n=5), stimulated with HBVsvp. Subsequent flow cytometric characterization involved assessing changes in activating (NKp46 and NKG2D) and inhibitory (CD94) receptors expression, quantifying TNF-α and IFN- γ cytokine secretion, and evaluating the cytotoxic response against HepG2.2.15 cells with subsequent HBVsvp quantification. Results: In CHB patients, in vitro exposure of PBMCs-derived NK cell with HBVsvp (represent HBsAg model) significantly reduced NK cell-activating receptors expression (P = 0.022), increased expression of CD94 + NK cells (p = 0.029), accompanied with a reduced TNF-α - IFN-γ cytokine levels, and impaired cytotoxic capacity (evidenced by increased cell proliferation and elevated HBVsvp levels in co-cultures with HepG2.2.15 cells in a time-dependent), relative to healthy donors. Conclusion: These findings suggest that HBVsvp may induce dysfunctional NK cell responses characterized by phenotypic imbalance with subsequent reduction in cytokine and cytotoxic levels, indicating HBVsvp immunosuppressive effect that compromises antiviral defense in CHB patients. These data enhance our understanding of NK cell interactions with HBsAg and highlight the potential for targeting CD94 inhibitory receptors to restore NK cell function as an immunotherapeutic approach. Further clinical research is needed to validate these observations and establish their utility as reliable biomarkers.


Subject(s)
Hepatitis B virus , Hepatitis B, Chronic , Killer Cells, Natural , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Hepatitis B, Chronic/immunology , Hepatitis B, Chronic/virology , Hepatitis B virus/immunology , Adult , Male , Female , Immunologic Surveillance , Phenotype , Middle Aged , Cytokines/metabolism , Cytokines/immunology , Hep G2 Cells , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Cytotoxicity, Immunologic , Hepatitis B Surface Antigens/immunology , Hepatitis B Surface Antigens/blood , NK Cell Lectin-Like Receptor Subfamily D/immunology , NK Cell Lectin-Like Receptor Subfamily D/metabolism , Natural Cytotoxicity Triggering Receptor 1/metabolism
4.
Cancer Immunol Immunother ; 73(11): 221, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39235531

ABSTRACT

BACKGROUND: Neuroblastoma is the most common extracranial solid tumor in children and accounts for 15% of pediatric cancer related deaths. Targeting neuroblastoma with immunotherapies has proven challenging due to a paucity of immune cells in the tumor microenvironment and the release of immunosuppressive cytokines by neuroblastoma tumor cells. We hypothesized that combining an oncolytic Herpes Simplex Virus (oHSV) with natural killer (NK) cells might overcome these barriers and incite tumor cell death. METHODS: We utilized MYCN amplified and non-amplified neuroblastoma cell lines, the IL-12 expressing oHSV, M002, and the human NK cell line, NK-92 MI. We assessed the cytotoxicity of NK cells against neuroblastoma with and without M002 infection, the effects of M002 on NK cell priming, and the impact of M002 and priming on the migratory capacity and CD107a expression of NK cells. To test clinical applicability, we then investigated the effects of M002 and NK cells on neuroblastoma in vivo. RESULTS: NK cells were more attracted to neuroblastoma cells that were infected with M002. There was an increase in neuroblastoma cell death with the combination treatment of M002 and NK cells both in vitro and in vivo. Priming the NK cells enhanced their cytotoxicity, migratory capacity and CD107a expression. CONCLUSIONS: To the best of our knowledge, these investigations are the first to demonstrate the effects of an oncolytic virus combined with self-maintaining NK cells in neuroblastoma and the priming effect of neuroblastoma on NK cells. The current studies provide a deeper understanding of the relation between NK cells and neuroblastoma and these data suggest that oHSV increases NK cell cytotoxicity towards neuroblastoma.


Subject(s)
Killer Cells, Natural , Neuroblastoma , Oncolytic Virotherapy , Neuroblastoma/therapy , Neuroblastoma/immunology , Killer Cells, Natural/immunology , Humans , Oncolytic Virotherapy/methods , Animals , Mice , Cell Line, Tumor , Oncolytic Viruses/immunology , Cytotoxicity, Immunologic , Simplexvirus/immunology , Xenograft Model Antitumor Assays
5.
J Innate Immun ; 16(1): 451-469, 2024.
Article in English | MEDLINE | ID: mdl-39265537

ABSTRACT

INTRODUCTION: The serine/threonine with-no-lysine (WNK) kinase family function in blood pressure control, electrolyte homeostasis, and cellular osmoregulation. These kinases and their downstream effectors are considered promising therapeutic targets in hypertension and stroke. However, the role of WNK kinases in immune cells remains poorly understood. METHODS: Using the small-molecule WNK kinase inhibitors WNK463 and WNK-IN-11, we investigated how WNK kinase inhibition affects natural killer (NK) cell physiology. RESULTS: WNK kinase inhibition with WNK463 or WNK-IN-11 significantly decreased IL-2-activated NK cell volume, motility, and cytolytic activity. Treatment of NK cells with these inhibitors induced autophagy by activating AMPK and inhibiting mTOR signaling. Moreover, WNK kinase inhibition increased phosphorylation of Akt and c-Myc by misaligning activity of activating kinases and inhibitory phosphatases. Treatment of tumor-bearing mice with WNK463 impaired tumor metastasis control by adoptively transferred NK cells. CONCLUSION: The catalytic activity of WNK kinases has a critical role of multiple aspects of NK cell physiology and their pharmacologic inhibition negatively impacts NK cell function.


Subject(s)
Killer Cells, Natural , Osmoregulation , Killer Cells, Natural/immunology , Animals , Mice , Humans , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , WNK Lysine-Deficient Protein Kinase 1/metabolism , Neoplasm Metastasis , Proto-Oncogene Proteins c-akt/metabolism , Protein Serine-Threonine Kinases/metabolism , Mice, Inbred C57BL , Cell Line, Tumor , Cytotoxicity, Immunologic
6.
Front Biosci (Landmark Ed) ; 29(8): 293, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39206895

ABSTRACT

The tumor microenvironment plays a critical role in modulating immune responses associated with tumorigenesis, tumor progression, and metastasis. Dendritic cells (DC) play a key role in preventing and progression of metastatic neoplasia by driving and restoring dysfunctional immune systems and obliterating immunosuppression, thus obstructing tumor evasion. In this review, we will discuss the functions of tumor-infiltrating DC in anti-tumor resistance, prevention of tumor recurrence, and immunosuppression. We will also describe DC metabolism, differentiation, and plasticity, which are essential for its function. Cancers like Lymphomas may be able to corrupt immune surveillance by reducing natural killer cell numbers. Thus, interactions between lymphoma and DC with reference to cytotoxicity may be an important event, likely to be mediated via activation with interferon-γ (IFN-γ) and Toll like receptors (TLR) ligands. Mechanisms of DC-mediated cytotoxicity and the role of apoptosis and death receptors, including the role played by nitric oxide, etc., are of immense significance. We will also look into the molecular mechanisms in the tumor microenvironment, reduced drug sensitivity, and tumor relapse, as well as methods for combating drug resistance and focusing on immunosuppressive tumor networks. We will address how DC mediated cytotoxicity in combination with drugs affects tumor growth and expansion in relation to checkpoint inhibitors and regulatory T cells. Innovative approaches for therapeutic modulation of this immunosuppressive adoptive DC immunotherapy will be highlighted, which is necessary for future personalized therapeutic applications.


Subject(s)
Dendritic Cells , Neoplasms , Tumor Microenvironment , Dendritic Cells/immunology , Humans , Tumor Microenvironment/immunology , Tumor Microenvironment/drug effects , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/drug therapy , Neoplasms/pathology , Animals , Cytotoxicity, Immunologic
7.
Sci Adv ; 10(34): eadp8647, 2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39178257

ABSTRACT

Agonistic antibodies against CD137 have been demonstrated to completely regress established tumors through activating T cell immunity. Unfortunately, current CD137 antibodies failed to benefit patients with cancer. Moreover, their antitumor mechanisms in vivo remain to be determined. Here, we report the development of a small molecular CD137 agonist, JNU-0921. JNU-0921 effectively activates both human and mouse CD137 through direct binding their extracellular domains to induce oligomerization and signaling and effectively shrinks tumors in vivo. Mechanistically, JNU-0921 enhances effector and memory function of cytotoxic CD8+ T cells (CTLs) and alleviates their exhaustion. JNU-0921 also skews polarization of helper T cells toward T helper 1 type and enhances their activity to boost CTL function. Meanwhile, JNU-0921 attenuates the inhibitory function of regulatory T cells on CTLs. Our current work shows that JNU-0921 shrinks tumors by enhancing the cytotoxicity of CTLs in cis and in trans and sheds light on strategy for developing CD137 small molecular agonists.


Subject(s)
CD8-Positive T-Lymphocytes , Tumor Necrosis Factor Receptor Superfamily, Member 9 , Animals , Tumor Necrosis Factor Receptor Superfamily, Member 9/agonists , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology , Humans , Mice , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , Cell Line, Tumor , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/drug effects , Antineoplastic Agents/pharmacology , Signal Transduction/drug effects , Cytotoxicity, Immunologic/drug effects , Xenograft Model Antitumor Assays , Neoplasms/immunology , Neoplasms/drug therapy , Neoplasms/pathology
8.
Sci Adv ; 10(35): eadq2366, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39196939

ABSTRACT

Adoptive cell transfer (ACT) is a therapeutic strategy to augment antitumor immunity. Here, we report that ex vivo treatment of mouse CD8+ T cells with dimethyloxalylglycine (DMOG), a stabilizer of hypoxia-inducible factors (HIFs), induced HIF binding to the genes encoding the costimulatory receptors CD81, GITR, OX40, and 4-1BB, leading to increased expression. DMOG treatment increased T cell killing of melanoma cells, which was further augmented by agonist antibodies targeting each costimulatory receptor. In tumor-bearing mice, ACT using T cells treated ex vivo with DMOG and agonist antibodies resulted in decreased tumor growth compared to ACT using control T cells and increased intratumoral markers of CD8+ T cells (CD7, CD8A, and CD8B1), natural killer cells (NCR1 and KLRK1), and cytolytic activity (perforin-1 and tumor necrosis factor-α). Costimulatory receptor gene expression was also induced when CD8+ T cells were treated with three highly selective HIF stabilizers that are currently in clinical use.


Subject(s)
CD8-Positive T-Lymphocytes , Immunotherapy, Adoptive , Animals , Mice , Immunotherapy, Adoptive/methods , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/drug effects , Amino Acids, Dicarboxylic/pharmacology , Cell Line, Tumor , Receptors, OX40/metabolism , Glucocorticoid-Induced TNFR-Related Protein/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism , Mice, Inbred C57BL , Melanoma, Experimental/therapy , Melanoma, Experimental/immunology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Cytotoxicity, Immunologic/drug effects
9.
Front Immunol ; 15: 1412378, 2024.
Article in English | MEDLINE | ID: mdl-39114666

ABSTRACT

Production of large amounts of functional NK and CAR-NK cells represents one of the bottlenecks for NK-based immunotherapy. In this study, we developed a large-scale, reliable, and practicable NK and CAR-NK production using G-Rex 100M bioreactors, which depend on a gas-permeable membrane technology. This system holds large volumes of medium with enhanced oxygen delivery, creating conditions conducive to large-scale PBNK and CAR-NK expansions for cancer therapy. Both peripheral blood NK cells (PBNKs) and CAR-NKs expanded in these bioreactors retained similar immunophenotypes and exhibited comparable cytotoxicity towards hepatocellular carcinoma (HCC) cells akin to that of NK and CAR-NK cells expanded in G-Rex 6 well bioreactors. Importantly, cryopreservation minimally affected the cytotoxicity of NK cells expanded using the G-Rex 100M bioreactors, establishing a robust platform for scaled-up NK and CAR-NK cell production. This method is promising for the development of "off-the-shelf" NK cells, supporting the future clinical implementation of NK cell immunotherapy.


Subject(s)
Bioreactors , Immunotherapy, Adoptive , Killer Cells, Natural , Receptors, Chimeric Antigen , Killer Cells, Natural/immunology , Humans , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/genetics , Immunotherapy, Adoptive/methods , Cell Culture Techniques/methods , Cytotoxicity, Immunologic , Cell Line, Tumor , Cell Proliferation , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/therapy , Liver Neoplasms/immunology , Liver Neoplasms/therapy
10.
Hum Immunol ; 85(5): 111085, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39116667

ABSTRACT

The major histocompatibility complex (MHC) class I chain-related A (MICA) plays an important role in stress cell recognition. High polymorphisms of MICA are relevant to NKG2D binding capacity, responses of NK cells and tumor progression. In this study, MICA genotyping of 97 cholangiocarcinoma patients was performed using PCR-SSP. MICA*010 was positively associated with a corrected p-value of < 0.001 (RR=2.16 (95 % CI, 1.48-3.14)). MICA*010 was previously reported as a non-expressed allele. Thus, the expression of MICA*010 on the cell surface was studied on both MICA*010 transfected cells (HEK 293 T and L929 cells) and stimulated primary monocytes obtained from homozygous MICA*010 individuals using different clones of antibodies (1H10, 1D10, 1C3.1, 1C3.2, 6D4 and 3H5) for detection. Surprisingly, the expression of MICA*010 could be observed on both transfected cells and stimulated monocytes and effectively bound to the NKG2D-Fc fusion protein. The functional study of various MICA alleles revealed the high relative killing activity of NK cells induced by the MICA*010 transfected C1R cells, not following the previously reported rule of the M129V substitution. The structural analysis highlighted the amino acid at position 36 as another important amino acid relevant to preserving the structural integrity of the MICA protein and NKG2D binding. Our data propose a new aspect of functional MICA contributing motifs and that MICA*010 has a potential effect on NK cell functions and might be applicable to other fields of immune responses.


Subject(s)
Cytotoxicity, Immunologic , Histocompatibility Antigens Class I , Killer Cells, Natural , NK Cell Lectin-Like Receptor Subfamily K , Humans , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , NK Cell Lectin-Like Receptor Subfamily K/metabolism , NK Cell Lectin-Like Receptor Subfamily K/genetics , NK Cell Lectin-Like Receptor Subfamily K/immunology , Cholangiocarcinoma/immunology , Cholangiocarcinoma/genetics , HEK293 Cells , Alleles , Male , Female , Bile Duct Neoplasms/immunology , Bile Duct Neoplasms/genetics , Genotype , Monocytes/immunology , Monocytes/metabolism , Middle Aged , Protein Binding
11.
J Transl Med ; 22(1): 737, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39103915

ABSTRACT

BACKGROUND: Cancer stem-like cells (CSCs) play an important role in initiation and progression of aggressive cancers, including esophageal cancer. Natural killer (NK) cells are key effector lymphocytes of innate immunity that directly attack a wide variety of cancer cells. NK cell-based therapy may provide a new treatment option for targeting CSCs. In this study, we aimed to investigate the sensitivity of human esophageal CSCs to NK cell-mediated cytotoxicity. METHODS: CSCs were enriched from human esophageal squamous cell carcinoma cell lines via sphere formation culture. Human NK cells were selectively expanded from the peripheral blood of healthy donors. qRT-PCR, flow cytometry and ELISA assays were performed to examine RNA expression and protein levels, respectively. CFSE-labeled target cells were co-cultured with human activated NK cells to detect the cytotoxicity of NK cells by flow cytometry. RESULTS: We observed that esophageal CSCs were more resistant to NK cell-mediated cytotoxicity compared with adherent counterparts. Consistently, esophageal CSCs showed down-regulated expression of ULBP-1, a ligand for NK cells stimulatory receptor NKG2D. Knockdown of ULBP-1 resulted in significant inhibition of NK cell cytotoxicity against esophageal CSCs, whereas ULBP-1 overexpression led to the opposite effect. Finally, the pro-differentiation agent all-trans retinoic acid was found to enhance the sensitivity of esophageal CSCs to NK cell cytotoxicity. CONCLUSIONS: This study reveals that esophageal CSCs are more resistant to NK cells through down-regulation of ULBP-1 and provides a promising approach to promote the activity of NK cells targeting esophageal CSCs.


Subject(s)
Cytotoxicity, Immunologic , Down-Regulation , Esophageal Neoplasms , Killer Cells, Natural , Neoplastic Stem Cells , Humans , Killer Cells, Natural/immunology , Esophageal Neoplasms/pathology , Esophageal Neoplasms/immunology , Esophageal Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Down-Regulation/drug effects , Cell Line, Tumor , Cytotoxicity, Immunologic/drug effects , GPI-Linked Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Gene Expression Regulation, Neoplastic/drug effects
12.
Adv Exp Med Biol ; 1448: 129-144, 2024.
Article in English | MEDLINE | ID: mdl-39117812

ABSTRACT

Familial forms of hemophagocytic lymphohistiocytosis (HLH) are caused by loss-of-function mutations in genes encoding perforin as well as those required for release of perforin-containing cytotoxic granule constituent. Perforin is expressed by subsets of CD8+ T cells and NK cells, representing lymphocytes that share mechanism of target cell killing yet display distinct modes of target cell recognition. Here, we highlight recent findings concerning the genetics of familial HLH that implicate CD8+ T cells in the pathogenesis of HLH and discuss mechanistic insights from animal models as well as patients that reveal how CD8+ T cells may contribute to or drive disease, at least in part through release of IFN-γ. Intriguingly, CD8+ T cells and NK cells may act differentially in severe hyperinflammatory diseases such as HLH. We also discuss how CD8+ T cells may promote or drive pathology in other cytokine release syndromes (CSS). Moreover, we review the molecular mechanisms underpinning CD8+ T cell-mediated lymphocyte cytotoxicity, key to the development of familial HLH. Together, recent insights to the pathophysiology of CSS in general and HLH in particular are providing promising new therapeutic targets.


Subject(s)
CD8-Positive T-Lymphocytes , Cytokine Release Syndrome , Lymphohistiocytosis, Hemophagocytic , Humans , CD8-Positive T-Lymphocytes/immunology , Lymphohistiocytosis, Hemophagocytic/immunology , Lymphohistiocytosis, Hemophagocytic/genetics , Animals , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/genetics , Killer Cells, Natural/immunology , Perforin/genetics , Perforin/metabolism , Cytotoxicity, Immunologic/genetics , Interferon-gamma/immunology , Interferon-gamma/genetics , Interferon-gamma/metabolism
13.
Viruses ; 16(8)2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39205244

ABSTRACT

Natural killer (NK) cells can be effective immunotherapeutic anti-cancer agents due to their ability to selectively target and kill tumor cells. This activity is modulated by the interaction of NK cell receptors with inhibitory ligands on the surface of target cells. NK cell inhibitory ligands can be upregulated on tumor cell surfaces in response to interferon-gamma (IFN-γ), a cytokine which is produced by activated NK cells. We hypothesized that the resistance of tumor cells to NK cell killing could be overcome by expression of the parainfluenza virus 5 (PIV5) V protein, which has known roles in blocking IFN-γ signaling. This was tested with human PM21-NK cells produced through a previously developed particle-based method which yields superior NK cells for immunotherapeutic applications. Infection of human SK-N-SH neuroblastoma cells with PIV5 blocked IFN-γ-mediated upregulation of three NK cell inhibitory ligands and enhanced in vitro killing of these tumor cells by PM21-NK cells. SK-N-SH cells transduced to constitutively express the V protein alone were resistant to IFN-γ-mediated increases in cell surface expression of NK cell inhibitory ligands. Real-time in vitro cell viability assays demonstrated that V protein expression in SK-N-SH cells was sufficient to increase PM21-NK cell-mediated killing. Toward a potential therapeutic application, transient lentiviral delivery of the V gene also enhanced PM21-NK cell killing in vitro. Our results provide the foundation for novel therapeutic applications of V protein expression in combination with ex vivo NK cell therapy to effectively increase the killing of tumor cells.


Subject(s)
Interferon-gamma , Killer Cells, Natural , Neuroblastoma , Parainfluenza Virus 5 , Up-Regulation , Humans , Killer Cells, Natural/immunology , Interferon-gamma/metabolism , Interferon-gamma/immunology , Neuroblastoma/immunology , Cell Line, Tumor , Parainfluenza Virus 5/immunology , Parainfluenza Virus 5/genetics , Ligands , Cytotoxicity, Immunologic , Viral Proteins/genetics , Viral Proteins/metabolism
14.
Cell ; 187(16): 4373-4388.e15, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39121849

ABSTRACT

Relatlimab (rela; anti-LAG-3) plus nivolumab (nivo; anti-PD-1) is safe and effective for treatment of advanced melanoma. We designed a trial (NCT03743766) where advanced melanoma patients received rela, nivo, or rela+nivo to interrogate the immunologic mechanisms of rela+nivo. Analysis of biospecimens from this ongoing trial demonstrated that rela+nivo led to enhanced capacity for CD8+ T cell receptor signaling and altered CD8+ T cell differentiation, leading to heightened cytotoxicity despite the retention of an exhaustion profile. Co-expression of cytotoxic and exhaustion signatures was driven by PRDM1, BATF, ETV7, and TOX. Effector function was upregulated in clonally expanded CD8+ T cells that emerged after rela+nivo. A rela+nivo intratumoral CD8+ T cell signature was associated with a favorable prognosis. This intratumoral rela+nivo signature was validated in peripheral blood as an elevated frequency of CD38+TIM3+CD8+ T cells. Overall, we demonstrated that cytotoxicity can be enhanced despite the retention of exhaustion signatures, which will inform future therapeutic strategies.


Subject(s)
CD8-Positive T-Lymphocytes , Lymphocyte Activation Gene 3 Protein , Melanoma , Programmed Cell Death 1 Receptor , Humans , Antigens, CD/metabolism , Antigens, CD/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Basic-Leucine Zipper Transcription Factors/genetics , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation , Cytotoxicity, Immunologic , High Mobility Group Proteins , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Lymphocyte Activation Gene 3 Protein/antagonists & inhibitors , Melanoma/immunology , Melanoma/drug therapy , Melanoma/genetics , Nivolumab/therapeutic use , Nivolumab/pharmacology , Positive Regulatory Domain I-Binding Factor 1/metabolism , Positive Regulatory Domain I-Binding Factor 1/genetics , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Signal Transduction
15.
Mol Immunol ; 174: 1-10, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39126837

ABSTRACT

BACKGROUND: Cannabidiol (CBD), the major non-psychoactive component of cannabis, exhibits anti-inflammatory properties, but less is known about the immunomodulatory potential of CBD on activated natural killer (NK) cells and/or their targets. Many tumor cells present heat shock protein 70 (Hsp70) on their cell surface in a tumor-specific manner and although a membrane Hsp70 (mHsp70) positive phenotype serves as a target for Hsp70-activated NK cells, a high mHsp70 expression is associated with tumor aggressiveness. This study investigated the immuno-modulatory potential of CBD on NK cells stimulated with TKD Hsp70 peptide and IL-2 (TKD+IL-2) and also on HCT116 p53wt and HCT116 p53-/- colorectal cancer cells exhibiting high and low basal levels of mHsp70 expression. RESULTS: Apart from an increase in the density of NTB-A and a reduced expression of LAMP-1, the expression of all other activatory NK cell receptors including NKp30, NKG2D and CD69 which are significantly up-regulated after stimulation with TKD+IL-2 remained unaffected after a co-treatment with CBD. However, the release of major pro-inflammatory cytokines by NK cells such as interferon-γ (IFN-γ) and the effector molecule granzyme B (GrzB) was significantly reduced upon CBD treatment. With respect to the tumor target cells, CBD significantly reduced the elevated expression of mHsp70 but had no effect on the low basal mHsp70 expression. Expression of other NK cell ligands such as MICA and MICB remained unaffected, and the NK cell ligands ULBP and B7-H6 were not expressed on these target cells. Consistent with the reduced mHsp70 expression, treatment of both effector and target cells with CBD reduced the killing of high mHsp70 expressing tumor cells by TKD+IL-2+CBD pre-treated NK cells but had no effect on the killing of low mHsp70 expressing tumor cells. Concomitantly, CBD treatment reduced the TKD+IL-2 induced increased release of IFN-γ, IL-4, TNF-α and GrzB, but CBD had no effect on the release of IFN-α when NK cells were co-incubated with tumor target cells. CONCLUSION: Cannabidiol (CBD) may potentially diminish the anti-tumor effectiveness of TKD+IL-2 activated natural killer (NK) cells.


Subject(s)
Cannabidiol , HSP70 Heat-Shock Proteins , Killer Cells, Natural , Lymphocyte Activation , Humans , Cannabidiol/pharmacology , Killer Cells, Natural/immunology , Killer Cells, Natural/drug effects , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , HSP70 Heat-Shock Proteins/metabolism , HCT116 Cells , Immunologic Factors/pharmacology , Interleukin-2/metabolism , Interleukin-2/immunology , Granzymes/metabolism , Interferon-gamma/metabolism , Interferon-gamma/immunology , Cell Line, Tumor , Cytotoxicity, Immunologic/drug effects
16.
Int Immunopharmacol ; 141: 112958, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-39159564

ABSTRACT

Acute myeloid leukemia (AML) is one of the most common types of blood cancer in adults and is associated with a poor survival rate. NK cells play a crucial role in combating AML, and alterations in immune checkpoint expression can impair NK cell function against AML. Targeting certain checkpoints may restore this function. CD96, an inhibitory immune checkpoint, has unclear expression and roles on NK cells in AML patients. In this study, we initially evaluated CD96 expression and compared CD96+ NK with the inhibitory receptor and stimulatory receptors on NK cells from AML patients at initial diagnosis. We observed increased CD96 expression on NK cells with dysfunctional phenotype. Further analysis revealed that CD96+ NK cells had lower IFN-γ production than CD96- NK cells. Blocking CD96 enhanced the cytotoxicity of primary NK and cord blood-derived NK (CB-NK) cells against leukemia cells. Notably, patients with a high frequency of CD96+ NK cells at initial diagnosis exhibited poorer clinical outcomes. Additionally, TGF-ß1 was found to enhance CD96 expression on NK cells via SMAD3 signaling. These findings suggest that CD96 is invovled in NK dysfunction against AML blast, and might be a potential target for restoring NK cell function in the fight against AML.


Subject(s)
Antigens, CD , Killer Cells, Natural , Leukemia, Myeloid, Acute , Transforming Growth Factor beta1 , Humans , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/diagnosis , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Transforming Growth Factor beta1/metabolism , Antigens, CD/metabolism , Prognosis , Male , Female , Middle Aged , Adult , Aged , Interferon-gamma/metabolism , Smad3 Protein/metabolism , Cell Line, Tumor , Signal Transduction , Cytotoxicity, Immunologic , Young Adult
17.
Cells ; 13(16)2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39195212

ABSTRACT

Cholangiocarcinoma (CCA) is a rare disease characterized by malignant cells derived from the epithelial cells of the biliary duct system. Despite extensive treatments, the prognosis for CCA remains poor, emphasizing the critical need for the development of novel treatments. Considerable attention has been directed towards innate immune effector cells, which can recognize tumor cells independently of the major histocompatibility complex, laying the foundation for the development of off-the-shelf drugs. In this study, we cultured innate immune cells obtained from the peripheral blood of healthy adults and conducted a comparative analysis of the effector functions against CCA cell lines by Vδ2 γδ T cells and NK cells. This analysis was performed using standard short- and long-term cytotoxicity assays, as well as ELISA for IFN-γ. Vδ2 γδ T cells demonstrated cytotoxicity and IFN-γ production in response to CCA cells in a TCR-dependent manner, particularly in the presence of tetrakis-pivaloyloxymethyl 2-(thiazole-2-ylamino)ethylidene-1,1-bisphosphonate, a bisphosphonate prodrug. In contrast, direct killing and antibody-dependent cellular cytotoxicity were relatively slow and weak. Conversely, NK cells displayed potent, direct cytotoxicity against CCA cells. In summary, both Vδ2 γδ T cells and NK cells show promise as innate immune effector cells for adoptive transfer therapy in the context of CCA.


Subject(s)
Cholangiocarcinoma , Interferon-gamma , Killer Cells, Natural , Receptors, Antigen, T-Cell, gamma-delta , Humans , Cholangiocarcinoma/immunology , Cholangiocarcinoma/pathology , Killer Cells, Natural/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, Antigen, T-Cell, gamma-delta/immunology , Cell Line, Tumor , Interferon-gamma/metabolism , Bile Duct Neoplasms/immunology , Bile Duct Neoplasms/pathology , Cytotoxicity, Immunologic/drug effects , T-Lymphocytes/immunology , Intraepithelial Lymphocytes/immunology
18.
Anticancer Res ; 44(9): 3899-3906, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39197911

ABSTRACT

BACKGROUND/AIM: The local immune response in colorectal cancer is closely related to prognosis and therapeutic efficacy. In this study, histological analyses were performed to determine the phenotype of tumor-infiltrating lymphocytes (TILs) and their infiltration in the stromal and intratumoral regions, aiming to elucidate their interactions and prognostic effects. PATIENTS AND METHODS: Multiplex fluorescent labeling was performed using surgically resected colorectal cancer specimens to investigate the infiltration of CD45RO (+) TILs, which exhibit cytotoxicity, and subsets of CD4 (+) TILs, identified by their characteristic transcription factor expression. RESULTS: The degree of CD45RO (+) TIL infiltration in the entire observation field or stromal area was not associated with prognosis. However, a high degree of infiltration in the tumor nest (intratumoral area) was significantly associated with a favorable prognosis. CD4 (+) TILs and their subsets were not associated with prognosis. However, stratified analyses revealed that a high degree of infiltration of stromal CD4 (+) TILs and the subsets T helper (Th)1, Th2, Th17, and regulatory T cells is necessary for the association between high intratumoral CD45RO (+) TIL infiltration and favorable prognosis. CONCLUSION: A sufficient degree of infiltration of stromal CD4 (+) TIL subsets is required for intratumoral CD45RO (+) TILs to exert toxicity against cancer cells. This highlights the significance of stromal immune reactions in achieving effective cytotoxic immune responses in the intratumoral area and demonstrates the critical role of the spatial distribution pattern of TILs in exerting their functions.


Subject(s)
Colorectal Neoplasms , Lymphocytes, Tumor-Infiltrating , Humans , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Lymphocytes, Tumor-Infiltrating/immunology , Male , Female , Aged , Prognosis , Middle Aged , Leukocyte Common Antigens/metabolism , CD4-Positive T-Lymphocytes/immunology , Stromal Cells/immunology , Stromal Cells/pathology , Stromal Cells/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Aged, 80 and over , Adult , Cytotoxicity, Immunologic
20.
Adv Exp Med Biol ; 1448: 75-101, 2024.
Article in English | MEDLINE | ID: mdl-39117809

ABSTRACT

Hemophagocytic lymphohistiocytosis (HLH) constitutes a rare, potentially life-threatening hyperinflammatory immune dysregulation syndrome that can present with a variety of clinical signs and symptoms, including fever, hepatosplenomegaly, and abnormal laboratory and immunological findings such as cytopenias, hyperferritinemia, hypofibrinogenemia, hypertriglyceridemia, elevated blood levels of soluble CD25 (interleukin (IL)-2 receptor α-chain), or diminished natural killer (NK)-cell cytotoxicity (reviewed in detail in Chapter 11 of this book). While HLH can be triggered by an inciting event (e.g., infections), certain monogenic causes have been associated with a significantly elevated risk of development of HLH, or recurrence of HLH in patients who have recovered from their disease episode. These monogenic predisposition syndromes are variably referred to as "familial" (FHL) or "primary" HLH (henceforth referred to as "pHLH") and are the focus of this chapter. Conversely, secondary HLH (sHLH) often occurs in the absence of monogenic etiologies that are commonly associated with pHLH and can be triggered by infections, malignancies, or rheumatological diseases; these triggers and the genetics associated with sHLH are discussed in more detail in other chapters in this book.


Subject(s)
Lymphohistiocytosis, Hemophagocytic , Animals , Lymphohistiocytosis, Hemophagocytic/genetics , Lymphohistiocytosis, Hemophagocytic/immunology , Recurrence , Risk Factors , Genetic Predisposition to Disease , Interleukin-2 Receptor alpha Subunit/immunology , Natural Killer T-Cells/immunology , Cytotoxicity, Immunologic
SELECTION OF CITATIONS
SEARCH DETAIL