Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 122
Filter
1.
Immunol Rev ; 323(1): 19-39, 2024 May.
Article in English | MEDLINE | ID: mdl-38459782

ABSTRACT

Natural Killer (NK) cells are a top contender in the development of adoptive cell therapies for cancer due to their diverse antitumor functions and ability to restrict their activation against nonmalignant cells. Despite their success in hematologic malignancies, NK cell-based therapies have been limited in the context of solid tumors. Tumor cells undergo various metabolic adaptations to sustain the immense energy demands that are needed to support their rapid and uncontrolled proliferation. As a result, the tumor microenvironment (TME) is depleted of nutrients needed to fuel immune cell activity and contains several immunosuppressive metabolites that hinder NK cell antitumor functions. Further, we now know that NK cell metabolic status is a main determining factor of their effector functions. Hence, the ability of NK cells to withstand and adapt to these metabolically hostile conditions is imperative for effective and sustained antitumor activity in the TME. With this in mind, we review the consequences of metabolic hostility in the TME on NK cell metabolism and function. We also discuss tumor-like metabolic programs in NK cell induced by STAT3-mediated expansion that adapt NK cells to thrive in the TME. Finally, we examine how other approaches can be applied to enhance NK cell metabolism in tumors.


Subject(s)
Killer Cells, Natural , Neoplasms , Tumor Microenvironment , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Humans , Tumor Microenvironment/immunology , Animals , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/therapy , Immunotherapy, Adoptive/methods , Adaptation, Physiological , STAT3 Transcription Factor/metabolism , Energy Metabolism , Tumor Escape
2.
Nat Commun ; 15(1): 896, 2024 Feb 05.
Article in English | MEDLINE | ID: mdl-38316762

ABSTRACT

Although many viral infections are linked to the development of neurological disorders, the mechanism governing virus-induced neuropathology remains poorly understood, particularly when the virus is not directly neuropathic. Using a mouse model of Zika virus (ZIKV) infection, we found that the severity of neurological disease did not correlate with brain ZIKV titers, but rather with infiltration of bystander activated NKG2D+CD8+ T cells. Antibody depletion of CD8 or blockade of NKG2D prevented ZIKV-associated paralysis, suggesting that CD8+ T cells induce neurological disease independent of TCR signaling. Furthermore, spleen and brain CD8+ T cells exhibited antigen-independent cytotoxicity that correlated with NKG2D expression. Finally, viral infection and inflammation in the brain was necessary but not sufficient to induce neurological damage. We demonstrate that CD8+ T cells mediate virus-induced neuropathology via antigen-independent, NKG2D-mediated cytotoxicity, which may serve as a therapeutic target for treatment of virus-induced neurological disease.


Subject(s)
Nervous System Diseases , Virus Diseases , Zika Virus Infection , Zika Virus , Humans , Antigens, Viral/metabolism , CD8-Positive T-Lymphocytes , NK Cell Lectin-Like Receptor Subfamily K/genetics , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Nervous System Diseases/metabolism
3.
Front Immunol ; 14: 1286750, 2023.
Article in English | MEDLINE | ID: mdl-38022679

ABSTRACT

The ability to expand and activate natural Killer (NK) cells ex vivo has dramatically changed the landscape in the development of novel adoptive cell therapies for treating cancer over the last decade. NK cells have become a key player for cancer immunotherapy due to their innate ability to kill malignant cells while not harming healthy cells, allowing their potential use as an "off-the-shelf" product. Furthermore, recent advancements in NK cell genetic engineering methods have enabled the efficient generation of chimeric antigen receptor (CAR)-expressing NK cells that can exert both CAR-dependent and antigen-independent killing. Clinically, CAR-NK cells have shown promising efficacy and safety for treating CD19-expressing hematologic malignancies. While the number of pre-clinical studies using CAR-NK cells continues to expand, it is evident that solid tumors pose a unique challenge to NK cell-based adoptive cell therapies. Major barriers for efficacy include low NK cell trafficking and infiltration into solid tumor sites, low persistence, and immunosuppression by the harsh solid tumor microenvironment (TME). In this review we discuss the barriers posed by the solid tumor that prevent immune cell trafficking and NK cell effector functions. We then discuss promising strategies to enhance NK cell infiltration into solid tumor sites and activation within the TME. This includes NK cell-intrinsic and -extrinsic mechanisms such as NK cell engineering to resist TME-mediated inhibition and use of tumor-targeted agents such as oncolytic viruses expressing chemoattracting and activating payloads. We then discuss opportunities and challenges for using combination therapies to extend NK cell therapies for the treatment of solid tumors.


Subject(s)
Neoplasms , Receptors, Chimeric Antigen , Humans , Tumor Microenvironment , Killer Cells, Natural , Immunotherapy, Adoptive/methods
4.
PLoS Pathog ; 19(6): e1011485, 2023 06.
Article in English | MEDLINE | ID: mdl-37384813

ABSTRACT

Mucosa-associated invariant T (MAIT) cells are MR1-restricted, innate-like T lymphocytes with tremendous antibacterial and immunomodulatory functions. Additionally, MAIT cells sense and respond to viral infections in an MR1-independent fashion. However, whether they can be directly targeted in immunization strategies against viral pathogens is unclear. We addressed this question in multiple wild-type and genetically altered but clinically relevant mouse strains using several vaccine platforms against influenza viruses, poxviruses and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We demonstrate that 5-(2-oxopropylideneamino)-6-D-ribitylaminouracil (5-OP-RU), a riboflavin-based MR1 ligand of bacterial origin, can synergize with viral vaccines to expand MAIT cells in multiple tissues, reprogram them towards a pro-inflammatory MAIT1 phenotype, license them to bolster virus-specific CD8+ T cell responses, and potentiate heterosubtypic anti-influenza protection. Repeated 5-OP-RU administration did not render MAIT cells anergic, thus allowing for its inclusion in prime-boost immunization protocols. Mechanistically, tissue MAIT cell accumulation was due to their robust proliferation, as opposed to altered migratory behavior, and required viral vaccine replication competency and Toll-like receptor 3 and type I interferon receptor signaling. The observed phenomenon was reproducible in female and male mice, and in both young and old animals. It could also be recapitulated in a human cell culture system in which peripheral blood mononuclear cells were exposed to replicating virions and 5-OP-RU. In conclusion, although viruses and virus-based vaccines are devoid of the riboflavin biosynthesis machinery that supplies MR1 ligands, targeting MR1 enhances the efficacy of vaccine-elicited antiviral immunity. We propose 5-OP-RU as a non-classic but potent and versatile vaccine adjuvant against respiratory viruses.


Subject(s)
COVID-19 , Mucosal-Associated Invariant T Cells , Vaccines , Female , Male , Humans , Mice , Animals , Vaccine Efficacy , Leukocytes, Mononuclear , COVID-19/metabolism , SARS-CoV-2 , Riboflavin/metabolism , Histocompatibility Antigens Class I , Minor Histocompatibility Antigens
5.
J Infect Dis ; 228(7): 834-839, 2023 10 03.
Article in English | MEDLINE | ID: mdl-36994782

ABSTRACT

Regulation of immune responses during viral infection is critical to preventing the development immunopathology that impairs host survival. Natural killer (NK) cells are well known for their antiviral functions that promote viral clearance; however, their roles in limiting immune-mediated pathology are still unclear. Using a mouse model for genital herpes simplex virus type 2 infection, we find that NK cell-derived interferon-γ directly counteracts interleukin-6-mediated matrix metalloproteases (MMPs) activity in macrophages to limit MMP-mediated tissue damage. Our findings uncover a key immunoregulatory function of NK cells during host-pathogen interactions that highlight the potential of NK cell therapy for treatment of severe viral infections.


Subject(s)
Herpes Genitalis , Interferon-gamma , Humans , Killer Cells, Natural , Herpesvirus 2, Human , Macrophages
6.
J Interferon Cytokine Res ; 42(11): 568-579, 2022 11.
Article in English | MEDLINE | ID: mdl-36346324

ABSTRACT

Type I interferons (IFNs) consist of a group of structurally similar cytokines that play an integral role in regulating the immune response to combat lung infections. In certain models type I IFNs have also been associated with suppression of Th2-skewed immune and inflammatory responses. Transient pulmonary overexpression of the gp130 cytokine Oncostatin M (OSM) by Adenovirus vector (AdOSM) induces a robust Th2-skewed cytokine/inflammatory profile in C57Bl/6 murine lungs. In this study we assessed type I IFN function in OSM-mediated inflammation in vivo using Ifnar1-/- C57Bl/6 mice and Ifnar1-deficient cells in vitro. Ifnar1-/- mice showed a significant reduction in AdOSM-induced histopathology (epithelial hyperplasia, alveolar septal wall thickening, cellular infiltration), and levels of IL-6 and chemokine protein (CXCL-1/KC and CCL24/eotaxin-2) in lungs compared with wild-type. Ifnar1-/- murine fibroblasts and human type I IFN receptor (Ifnar)-knockdown fibroblasts were also less responsive to OSM in STAT3 activation and cytokine production compared with Ifnar-sufficient cells in vitro. Exogenous type I IFN induced IL-6 responses in mouse and human fibroblasts and in combination with OSM further stimulated IL-6 production, suggesting a concerted action of type I IFNs and OSM. Taken together, these results demonstrate that cross-talk between IFNAR and OSM signaling enhances cell responses and modulates OSM-driven responses in lung inflammation.


Subject(s)
Interferon Type I , Pneumonia , Mice , Humans , Animals , Oncostatin M/metabolism , Interleukin-6/metabolism , Lung/metabolism , Mice, Inbred C57BL , Pneumonia/pathology , Interferon Type I/metabolism
7.
PLoS Pathog ; 18(5): e1010471, 2022 05.
Article in English | MEDLINE | ID: mdl-35512020

ABSTRACT

The ability to treat severe viral infections is limited by our understanding of the mechanisms behind virus-induced immunopathology. While the role of type I interferons (IFNs) in early control of viral replication is clear, less is known about how IFNs can regulate the development of immunopathology and affect disease outcomes. Here, we report that absence of type I IFN receptor (IFNAR) is associated with extensive immunopathology following mucosal viral infection. This pathology occurred independent of viral load or type II immunity but required the presence of macrophages and IL-6. The depletion of macrophages and inhibition of IL-6 signaling significantly abrogated immunopathology. Tissue destruction was mediated by macrophage-derived matrix metalloproteinases (MMPs), as MMP inhibition by doxycycline and Ro 28-2653 reduced the severity of tissue pathology. Analysis of post-mortem COVID-19 patient lungs also displayed significant upregulation of the expression of MMPs and accumulation of macrophages. Overall, we demonstrate that IFNs inhibit macrophage-mediated MMP production to prevent virus-induced immunopathology and uncover MMPs as a therapeutic target towards viral infections.


Subject(s)
COVID-19 , Interferon Type I , Orthomyxoviridae Infections , Humans , Interleukin-6/metabolism , Macrophages/metabolism , Proteolysis
9.
Immunol Cell Biol ; 100(6): 468-473, 2022 07.
Article in English | MEDLINE | ID: mdl-35355318

ABSTRACT

Zika virus (ZIKV) has emerged as a significant health threat worldwide. Although typically mosquito-borne, recent evidence suggests that ZIKV is also a sexually transmitted virus. While persistent ZIKV infections in male reproductive tissues have been identified, little is understood regarding the outcomes of primary sexual transmission in females. We investigated how the route of infection affects vaginal ZIKV shedding and dissemination. In two mouse models, vaginal infection resulted in prolonged ZIKV shedding in the vaginal mucosa with delayed systemic infection. Furthermore, heightened vaginal inflammation did not influence ZIKV replication or dissemination, in contrast to previous studies of mosquito-borne infection. Thus, vaginal infection significantly alters ZIKV infection kinetics and must be considered when developing novel treatments.


Subject(s)
Zika Virus Infection , Zika Virus , Animals , Female , Male , Mice , Mucous Membrane , Vagina , Virus Shedding
10.
ACS Appl Mater Interfaces ; 14(9): 11068-11077, 2022 Mar 09.
Article in English | MEDLINE | ID: mdl-35225604

ABSTRACT

Amidst the COVID-19 pandemic, it is evident that viral spread is mediated through several different transmission pathways. Reduction of these transmission pathways is urgently needed to control the spread of viruses between infected and susceptible individuals. Herein, we report the use of pathogen-repellent plastic wraps (RepelWrap) with engineered surface structures at multiple length scales (nanoscale to microscale) as a means of reducing the indirect contact transmission of viruses through fomites. To quantify viral repellency, we developed a touch-based viral quantification assay to mimic the interaction of a contaminated human touch with a surface through the modification of traditional viral quantification methods (viral plaque and TCID50 assays). These studies demonstrate that RepelWrap reduced contamination with an enveloped DNA virus as well as the human coronavirus 229E (HuCoV-229E) by more than 4 log 10 (>99.99%) compared to a standard commercially available polyethylene plastic wrap. In addition, RepelWrap maintained its repellent properties after repeated 300 touches and did not show an accumulation in viral titer after multiple contacts with contaminated surfaces, while increases were seen on other commonly used surfaces. These findings show the potential use of repellent surfaces in reducing viral contamination on surfaces, which could, in turn, reduce the surface-based spread and transmission.


Subject(s)
COVID-19/prevention & control , Coronavirus 229E, Human/growth & development , Equipment Contamination/prevention & control , Infection Control/instrumentation , Plastics/chemistry , COVID-19/transmission , COVID-19/virology , Humans , Infection Control/methods , SARS-CoV-2/growth & development , Surface Properties
12.
Viruses ; 13(11)2021 11 08.
Article in English | MEDLINE | ID: mdl-34835050

ABSTRACT

Following the recent outbreak of Zika virus (ZIKV) infections in Latin America, ZIKV has emerged as a global health threat due to its ability to induce neurological disease in both adults and the developing fetus. ZIKV is largely mosquito-borne and is now endemic in many parts of Africa, Asia, and South America. However, several reports have demonstrated persistent ZIKV infection of the male reproductive tract and evidence of male-to-female sexual transmission of ZIKV. Sexual transmission may broaden the reach of ZIKV infections beyond its current geographical limits, presenting a significant threat worldwide. Several mouse models of ZIKV infection have been developed to investigate ZIKV pathogenesis and develop effective vaccines and therapeutics. However, the majority of these models focus on mosquito-borne infection, while few have considered the impact of sexual transmission on immunity and pathogenesis. This review will examine the advantages and disadvantages of current models of mosquito-borne and sexually transmitted ZIKV and provide recommendations for the effective use of ZIKV mouse models.


Subject(s)
Disease Models, Animal , Sexually Transmitted Diseases, Viral/transmission , Zika Virus Infection/transmission , Animals , Female , Mice , Pregnancy , Pregnancy Complications, Infectious/prevention & control , Pregnancy Complications, Infectious/virology , Sexually Transmitted Diseases, Viral/prevention & control , Vector Borne Diseases/prevention & control , Vector Borne Diseases/transmission , Vector Borne Diseases/virology , Zika Virus/physiology , Zika Virus Infection/prevention & control , Zika Virus Infection/virology
13.
STAR Protoc ; 2(4): 100956, 2021 12 17.
Article in English | MEDLINE | ID: mdl-34825217

ABSTRACT

Although natural killer (NK) cells have become a promising immune effector cell for chimeric antigen receptor (CAR)-based therapy, generating human CAR-NK cells with high transgene efficiency has been challenging. In this protocol, we describe how to generate CAR-NK cells with transduction efficiencies >15% from healthy donor ex vivo expanded NK cells using third generation lentiviral vectors (LVs). We also show how to assess CAR-NK cell anti-tumor function in vitro using a flow cytometry-based killing assay. For complete details on the use and execution of this protocol, please refer to Portillo et al. (2021).


Subject(s)
Cell Culture Techniques/methods , Genetic Vectors/genetics , Killer Cells, Natural , Lentivirus/genetics , Receptors, Chimeric Antigen/genetics , Cells, Cultured , Humans , Immunotherapy, Adoptive
14.
Front Immunol ; 12: 724618, 2021.
Article in English | MEDLINE | ID: mdl-34484233

ABSTRACT

Herpes simplex virus type 2 (HSV-2) infection is one of the most prevalent sexually transmitted infections that disproportionately impacts women worldwide. Currently, there are no vaccines or curative treatments, resulting in life-long infection. The mucosal environment of the female reproductive tract (FRT) is home to a complex array of local immune defenses that must be carefully coordinated to protect against genital HSV-2 infection, while preventing excessive inflammation to prevent disease symptoms. Crucial to the defense against HSV-2 infection in the FRT are three classes of highly related and integrated cytokines, type I, II, and III interferons (IFN). These three classes of cytokines control HSV-2 infection and reduce tissue damage through a combination of directly inhibiting viral replication, as well as regulating the function of resident immune cells. In this review, we will examine how interferons are induced and their critical role in how they shape the local immune response to HSV-2 infection in the FRT.


Subject(s)
Herpes Genitalis/immunology , Herpesvirus 2, Human/immunology , Interferons/immunology , Animals , Female , Humans , Immunity, Mucosal , Mice , Mucous Membrane/virology
15.
Nat Commun ; 12(1): 5148, 2021 08 26.
Article in English | MEDLINE | ID: mdl-34446714

ABSTRACT

Coronavirus infection in humans is usually associated to respiratory tract illnesses, ranging in severity from mild to life-threatening respiratory failure. The aryl hydrocarbon receptor (AHR) was recently identified as a host factor for Zika and dengue viruses; AHR antagonists boost antiviral immunity, decrease viral titers and ameliorate Zika-induced pathology in vivo. Here we report that AHR is activated by infection with different coronaviruses, potentially impacting antiviral immunity and lung epithelial cells. Indeed, the analysis of single-cell RNA-seq from lung tissue detected increased expression of AHR and AHR transcriptional targets, suggesting AHR signaling activation in SARS-CoV-2-infected epithelial cells from COVID-19 patients. Moreover, we detected an association between AHR expression and viral load in SARS-CoV-2 infected patients. Finally, we found that the pharmacological inhibition of AHR suppressed the replication in vitro of one of the causative agents of the common cold, HCoV-229E, and the causative agent of the COVID-19 pandemic, SARS-CoV-2. Taken together, these findings suggest that AHR activation is a common strategy used by coronaviruses to evade antiviral immunity and promote viral replication, which may also contribute to lung pathology. Future studies should further evaluate the potential of AHR as a target for host-directed antiviral therapy.


Subject(s)
Coronavirus Infections/metabolism , Coronavirus/physiology , Receptors, Aryl Hydrocarbon/metabolism , Signal Transduction , COVID-19/genetics , COVID-19/metabolism , COVID-19/virology , Coronavirus Infections/genetics , Coronavirus Infections/virology , Epithelial Cells/metabolism , Epithelial Cells/virology , Female , Humans , Male , Receptors, Aryl Hydrocarbon/genetics , SARS-CoV-2/physiology
16.
Commun Biol ; 4(1): 859, 2021 07 12.
Article in English | MEDLINE | ID: mdl-34253827

ABSTRACT

Triple negative breast cancer holds a dismal clinical outcome and as such, patients routinely undergo aggressive, highly toxic treatment regimens. Clinical trials for TNBC employing immune checkpoint blockade in combination with chemotherapy show modest prognostic benefit, but the percentage of patients that respond to treatment is low, and patients often succumb to relapsed disease. Here, we show that a combination immunotherapy platform utilizing low dose chemotherapy (FEC) combined with oncolytic virotherapy (oHSV-1) increases tumor-infiltrating lymphocytes, in otherwise immune-bare tumors, allowing 60% of mice to achieve durable tumor regression when treated with immune checkpoint blockade. Whole-tumor RNA sequencing of mice treated with FEC + oHSV-1 shows an upregulation of B cell receptor signaling pathways and depletion of B cells prior to the start of treatment in mice results in complete loss of therapeutic efficacy and expansion of myeloid-derived suppressor cells. Additionally, RNA sequencing data shows that FEC + oHSV-1 suppresses genes associated with myeloid-derived suppressor cells, a key population of cells that drive immune escape and mediate therapeutic resistance. These findings highlight the importance of tumor-infiltrating B cells as drivers of antitumor immunity and their potential role in the regulation of myeloid-derived suppressor cells.


Subject(s)
B-Lymphocytes/drug effects , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy/methods , Lymphocytes, Tumor-Infiltrating/drug effects , Myeloid-Derived Suppressor Cells/drug effects , Triple Negative Breast Neoplasms/therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Line, Tumor , Chlorocebus aethiops , Combined Modality Therapy , Female , Humans , Immune Checkpoint Inhibitors/immunology , Kaplan-Meier Estimate , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Mice, Inbred C57BL , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , Oncolytic Virotherapy/methods , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/pathology , Vero Cells
17.
iScience ; 24(6): 102619, 2021 Jun 25.
Article in English | MEDLINE | ID: mdl-34159300

ABSTRACT

Despite the remarkable success of chimeric antigen receptor (CAR)-T cells against hematologic malignancies, severe off-tumor effects have constrained their use against solid tumors. Recently, CAR-engineered natural killer (NK) cells have emerged as an effective and safe alternative. Here, we demonstrate that HER2 CAR-expression in NK cells from healthy donors and patients with breast cancer potently enhances their anti-tumor functions against various HER2-expressing cancer cells, regardless of MHC class I expression. Moreover, HER2 CAR-NK cells exert higher cytotoxicity than donor-matched HER2 CAR-T cells against tumor targets. Importantly, unlike CAR-T cells, HER2 CAR-NK cells do not elicit enhanced cytotoxicity or inflammatory cytokine production against non-malignant human lung epithelial cells with basal HER2 expression. Further, HER2 CAR-NK cells maintain high cytotoxic function in the presence of immunosuppressive factors enriched in solid tumors. These results show that CAR-NK cells may be a highly potent and safe source of immunotherapy in the context of solid tumors.

19.
Cells ; 10(3)2021 03 23.
Article in English | MEDLINE | ID: mdl-33806810

ABSTRACT

As highlighted by the COVID-19 global pandemic, elderly individuals comprise the majority of cases of severe viral infection outcomes and death. A combined inability to control viral replication and exacerbated inflammatory immune activation in elderly patients causes irreparable immune-mediated tissue pathology in response to infection. Key to these responses are type I, II, and III interferons (IFNs), which are involved in inducing an antiviral response, as well as controlling and suppressing inflammation and immunopathology. IFNs support monocyte/macrophage-stimulated immune responses that clear infection and promote their immunosuppressive functions that prevent excess inflammation and immune-mediated pathology. The timing and magnitude of IFN responses to infection are critical towards their immunoregulatory functions and ability to prevent immunopathology. Aging is associated with multiple defects in the ability of macrophages and dendritic cells to produce IFNs in response to viral infection, leading to a dysregulation of inflammatory immune responses. Understanding the implications of aging on IFN-regulated inflammation will give critical insights on how to treat and prevent severe infection in vulnerable individuals. In this review, we describe the causes of impaired IFN production in aging, and the evidence to suggest that these impairments impact the regulation of the innate and adaptive immune response to infection, thereby causing disease pathology.


Subject(s)
Adaptive Immunity , Aging/immunology , COVID-19/immunology , Immunity, Innate , Interferons/physiology , SARS-CoV-2/immunology , Virus Replication/immunology , Aged , COVID-19/virology , Humans , Inflammation/drug therapy , Inflammation/immunology , Interferon Type I/immunology , Interferon Type I/therapeutic use , Interferon-gamma/immunology , Interferon-gamma/therapeutic use , Interferons/immunology , Interferons/therapeutic use , Interferon Lambda , COVID-19 Drug Treatment
20.
Cell Metab ; 33(6): 1205-1220.e5, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33852875

ABSTRACT

NK cells are central to anti-tumor immunity and recently showed efficacy for treating hematologic malignancies. However, their dysfunction in the hostile tumor microenvironment remains a pivotal barrier for cancer immunotherapies against solid tumors. Using cancer patient samples and proteomics, we found that human NK cell dysfunction in the tumor microenvironment is due to suppression of glucose metabolism via lipid peroxidation-associated oxidative stress. Activation of the Nrf2 antioxidant pathway restored NK cell metabolism and function and resulted in greater anti-tumor activity in vivo. Strikingly, expanded NK cells reprogrammed with complete metabolic substrate flexibility not only sustained metabolic fitness but paradoxically augmented their tumor killing in the tumor microenvironment and in response to nutrient deprivation. Our results uncover that metabolic flexibility enables a cytotoxic immune cell to exploit the metabolic hostility of tumors for their advantage, addressing a critical hurdle for cancer immunotherapy.


Subject(s)
Antineoplastic Agents/immunology , Immunotherapy/methods , Killer Cells, Natural/immunology , Neoplasms/therapy , Tumor Microenvironment , Adult , Aged , Animals , Cell Line, Tumor , Female , Humans , Killer Cells, Natural/cytology , Male , Mice , Middle Aged , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...