Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters

Publication year range
1.
Immunity ; 53(3): 658-671.e6, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32937153

ABSTRACT

Identifying signals in the tumor microenvironment (TME) that shape CD8+ T cell phenotype can inform novel therapeutic approaches for cancer. Here, we identified a gradient of increasing glucocorticoid receptor (GR) expression and signaling from naïve to dysfunctional CD8+ tumor-infiltrating lymphocytes (TILs). Conditional deletion of the GR in CD8+ TILs improved effector differentiation, reduced expression of the transcription factor TCF-1, and inhibited the dysfunctional phenotype, culminating in tumor growth inhibition. GR signaling transactivated the expression of multiple checkpoint receptors and promoted the induction of dysfunction-associated genes upon T cell activation. In the TME, monocyte-macrophage lineage cells produced glucocorticoids and genetic ablation of steroidogenesis in these cells as well as localized pharmacologic inhibition of glucocorticoid biosynthesis improved tumor growth control. Active glucocorticoid signaling associated with failure to respond to checkpoint blockade in both preclinical models and melanoma patients. Thus, endogenous steroid hormone signaling in CD8+ TILs promotes dysfunction, with important implications for cancer immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Glucocorticoids/metabolism , Macrophages/metabolism , Melanoma, Experimental/pathology , Tumor Microenvironment/immunology , Animals , CD8-Positive T-Lymphocytes/cytology , Cell Line, Tumor , Hematopoiesis/immunology , Hepatocyte Nuclear Factor 1-alpha/biosynthesis , Immune Checkpoint Inhibitors , Lymphocyte Activation/immunology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Signal Transduction/immunology
2.
Nature ; 595(7865): 101-106, 2021 07.
Article in English | MEDLINE | ID: mdl-34108686

ABSTRACT

T cell immunoglobulin and mucin-containing molecule 3 (TIM-3), first identified as a molecule expressed on interferon-γ producing T cells1, is emerging as an important immune-checkpoint molecule, with therapeutic blockade of TIM-3 being investigated in multiple human malignancies. Expression of TIM-3 on CD8+ T cells in the tumour microenvironment is considered a cardinal sign of T cell dysfunction; however, TIM-3 is also expressed on several other types of immune cell, confounding interpretation of results following blockade using anti-TIM-3 monoclonal antibodies. Here, using conditional knockouts of TIM-3 together with single-cell RNA sequencing, we demonstrate the singular importance of TIM-3 on dendritic cells (DCs), whereby loss of TIM-3 on DCs-but not on CD4+ or CD8+ T cells-promotes strong anti-tumour immunity. Loss of TIM-3 prevented DCs from expressing a regulatory program and facilitated the maintenance of CD8+ effector and stem-like T cells. Conditional deletion of TIM-3 in DCs led to increased accumulation of reactive oxygen species resulting in NLRP3 inflammasome activation. Inhibition of inflammasome activation, or downstream effector cytokines interleukin-1ß (IL-1ß) and IL-18, completely abrogated the protective anti-tumour immunity observed with TIM-3 deletion in DCs. Together, our findings reveal an important role for TIM-3 in regulating DC function and underscore the potential of TIM-3 blockade in promoting anti-tumour immunity by regulating inflammasome activation.


Subject(s)
Hepatitis A Virus Cellular Receptor 2/metabolism , Inflammasomes/metabolism , Neoplasms/immunology , Neoplasms/metabolism , Animals , Dendritic Cells , Female , Hepatitis A Virus Cellular Receptor 2/deficiency , Hepatitis A Virus Cellular Receptor 2/genetics , Interleukin-18/immunology , Interleukin-1beta/immunology , Male , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neoplasms/pathology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
3.
J Clin Immunol ; 44(3): 63, 2024 Feb 16.
Article in English | MEDLINE | ID: mdl-38363399

ABSTRACT

Inflammatory bowel disease (IBD) occurring following allogeneic stem cell transplantation (aSCT) is a very rare condition. The underlying pathogenesis needs to be better defined. There is currently no systematic effort to exclude loss- or gain-of-function mutations in immune-related genes in stem cell donors. This is despite the fact that more than 100 inborn errors of immunity may cause or contribute to IBD. We have molecularly characterized a patient who developed fulminant inflammatory bowel disease following aSCT with stable 100% donor-derived hematopoiesis. A pathogenic c.A291G; p.I97M HAVCR2 mutation encoding the immune checkpoint protein TIM-3 was identified in the patient's blood-derived DNA, while being absent in DNA derived from the skin. TIM-3 expression was much decreased in the patient's serum, and in vitro-activated patient-derived T cells expressed reduced TIM-3 levels. In contrast, T cell-intrinsic CD25 expression and production of inflammatory cytokines were preserved. TIM-3 expression was barely detectable in the immune cells of the patient's intestinal mucosa, while being detected unambiguously in the inflamed and non-inflamed colon from unrelated individuals. In conclusion, we report the first case of acquired, "transplanted" insufficiency of the regulatory TIM-3 checkpoint linked to post-aSCT IBD.


Subject(s)
Hepatitis A Virus Cellular Receptor 2 , Inflammatory Bowel Diseases , Stem Cell Transplantation , Humans , Cytokines/metabolism , Hepatitis A Virus Cellular Receptor 2/genetics , Inflammatory Bowel Diseases/diagnosis , Inflammatory Bowel Diseases/etiology , Intestinal Mucosa , Stem Cell Transplantation/adverse effects
4.
Proc Natl Acad Sci U S A ; 118(42)2021 10 19.
Article in English | MEDLINE | ID: mdl-34654745

ABSTRACT

Information about features in the visual world is parsed by circuits in the retina and is then transmitted to the brain by distinct subtypes of retinal ganglion cells (RGCs). Axons from RGC subtypes are stratified in retinorecipient brain nuclei, such as the superior colliculus (SC), to provide a segregated relay of parallel and feature-specific visual streams. Here, we sought to identify the molecular mechanisms that direct the stereotyped laminar targeting of these axons. We focused on ipsilateral-projecting subtypes of RGCs (ipsiRGCs) whose axons target a deep SC sublamina. We identified an extracellular glycoprotein, Nephronectin (NPNT), whose expression is restricted to this ipsiRGC-targeted sublamina. SC-derived NPNT and integrin receptors expressed by ipsiRGCs are both required for the targeting of ipsiRGC axons to the deep sublamina of SC. Thus, a cell-extracellular matrix (ECM) recognition mechanism specifies precise laminar targeting of ipsiRGC axons and the assembly of eye-specific parallel visual pathways.


Subject(s)
Brain/physiology , Extracellular Matrix/physiology , Retinal Ganglion Cells/physiology , Visual Pathways , Animals , Axons/physiology , Integrins/metabolism , Mice , Signal Transduction , Superior Colliculi/cytology , Superior Colliculi/metabolism , Superior Colliculi/physiology
5.
Aust J Rural Health ; 31(5): 878-885, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37350539

ABSTRACT

INTRODUCTION: Stroke in Regional Australia may have worse outcomes due to difficulties accessing optimal care. The South Australian Regional Telestroke service aimed to improve telestroke neurologist access, supported by improved ambulance triage. OBJECTIVE: To assess stroke care quality and patient mortality pre- and postimplementation of a vascular neurologist-led Telestroke service. DESIGN: Historically controlled mixed methods cohort study comparing key quality indicators and patient mortality (6 months pre- vs. 18 months postimplementation date [4 June 2018]) at the three major South Australian regional stroke centres. The primary outcome was 13 care quality indicators as a combined composite risk-adjusted score, and the secondary outcome was risk-adjusted mortality at 12-month postadmission. FINDINGS: On an annualised basis, of 189 patients with stroke, more were admitted postintervention to the regional stroke centres than in the control period (158 [annualised rate 105.3, 95% CI 86.2-127.4] vs. 31 [annualised rate 62.0, 95% CI 47.5-79.5]) Baseline patient characteristics were similar in both periods. Post-implementation, median last-known-well time to presentation (3.5 h [IQR 1.6-17] vs. 2.0 [IQR 1-14]; p = 0.46) and door to needle times (121 min [IQR 97-144] vs. 90 [IQR 75-138]; p = 0.65) were not significantly lower but an improvement in the combined composite quality score was observed (0.069 [95% CI 0.004-0.134; p = 0.04]), reflecting individual improvements in some quality indicators. Mortality at 12-month postimplementation was substantially lower postimplementation (prechange 23% vs. postchange 13% [hazard ratio 0.58 (95% CI 0.44-0.76; p < 0.001)]). CONCLUSION: Implementation of a South Australian Regional Telestroke service was associated with improved care metrics and lower mortality.


Subject(s)
Stroke , Telemedicine , Humans , Fibrinolytic Agents/therapeutic use , Thrombolytic Therapy , South Australia , Retrospective Studies , Cohort Studies , Telemedicine/methods , Treatment Outcome , Australia , Stroke/therapy , Tissue Plasminogen Activator/therapeutic use
6.
J Neurophysiol ; 127(4): 913-927, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35294270

ABSTRACT

The superior colliculus (SC) integrates visual and other sensory information to regulate critical reflexive and innate behaviors, such as prey capture. In the mouse, the vast majority of retinal ganglion cells (RGCs) innervate the SC, including inputs from both the contralateral (contra-RGCs) and ipsilateral (ipsi-RGCs) eye. Despite this, previous studies revealed minimal neuronal responses to ipsilateral stimulation and few binocular interactions in the mouse SC. More recent work suggests that ipsi-RGC function and innervation of the SC are critical for efficient prey capture, raising the possibility that binocular interactions in the mouse SC may be more prevalent than previously thought. To explore this possibility, we investigated eye-specific and binocular influences on visual responses and tuning of SC neurons, focusing on the anteromedial region. Although the majority of SC neurons were primarily driven by contralateral eye stimulation, we observed that a substantial proportion of units were influenced or driven by ipsilateral stimulation. Clustering based on differential responses to eye-specific stimulus presentation revealed five distinct putative subpopulations and multiple modes of binocular interaction, including facilitation, summation, and suppression. Each of the putative subpopulations exhibited selectivity for orientation, and differences in spatial frequency tuning and spatial summation properties were observed between subpopulations. Further analysis of orientation tuning under different ocular conditions supported differential modes of binocular interaction between putative subtypes. Taken together, these data suggest that binocular interactions in the mouse SC may be more prevalent and diverse than previously understood.NEW & NOTEWORTHY The mouse superior colliculus (SC) receives binocular inputs, which inform complex behavioral programs. However, we know surprisingly little about binocular tuning in the rodent SC. Here, we characterize responses to eye-specific presentations of visual stimuli and reveal a previously unappreciated diversity of binocularly modulated neurons in the SC. This foundational work broadens our understanding of visual processing in the SC and sets the stage for future studies interrogating the circuit mechanisms underlying binocular tuning.


Subject(s)
Superior Colliculi , Visual Pathways , Animals , Mice , Photic Stimulation , Retinal Ganglion Cells , Superior Colliculi/physiology , Visual Pathways/physiology , Visual Perception/physiology
7.
J Immunol ; 200(8): 3000-3007, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29500245

ABSTRACT

Coinhibitory receptors, such as CTLA-4 and PD-1, play a critical role in maintaining immune homeostasis by dampening T cell responses. Recently, they have gained attention as therapeutic targets in chronic disease settings where their dysregulated expression contributes to suppressed immune responses. The novel coinhibitory receptor TIGIT (T cell Ig and ITIM domain) has been shown to play an important role in modulating immune responses in the context of autoimmunity and cancer. However, the molecular mechanisms by which TIGIT modulates immune responses are still insufficiently understood. We have generated a panel of monoclonal anti-mouse TIGIT Abs that show functional properties in mice in vivo and can serve as important tools to study the underlying mechanisms of TIGIT function. We have identified agonistic as well as blocking anti-TIGIT Ab clones that are capable of modulating T cell responses in vivo. Administration of either agonist or blocking anti-TIGIT Abs modulated autoimmune disease severity whereas administration of blocking anti-TIGIT Abs synergized with anti-PD-1 Abs to affect partial or even complete tumor regression. The Abs presented in this study can thus serve as important tools for detailed analysis of TIGIT function in different disease settings and the knowledge gained will provide valuable insight for the development of novel therapeutic approaches targeting TIGIT.


Subject(s)
Antibodies, Monoclonal/immunology , Autoimmunity/immunology , Neoplasms/immunology , Receptors, Immunologic/immunology , Animals , Mice
8.
Eur J Immunol ; 47(3): 470-480, 2017 03.
Article in English | MEDLINE | ID: mdl-28105653

ABSTRACT

Dendritic cells (DCs) and complement are both key members of the innate and adaptive immune response. Recent experimental mouse models have shown that production of alternative pathway (AP) components by DCs strongly affects their ability to activate and regulate T-cell responses. In this study we investigated the production and regulation of properdin (fP) and factor H (fH) both integral regulators of the AP, by DCs and tolerogenic DCs (tolDCs). Both fP and fH were produced by DCs, with significantly higher levels of both AP components produced by tolDCs. Upon activation with IFN-γ both cells increased fH production, while simultaneously decreasing production of fP. IL-27, a member of the IL-12 family, increased fH, but production of fP remained unaffected. The functional capacity of fP and fH produced by DCs and tolDCs was confirmed by their ability to bind C3b. Inhibition of fH production by DCs resulted in a greater ability to induce allogenic CD4+ T-cell proliferation. In contrast, inhibition of fP production led to a significantly reduced allostimulatory capacity. In summary, this study shows that production of fP and fH by DCs, differentially regulates their immunogenicity, and that the local cytokine environment can profoundly affect the production of fP and fH.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Complement Factor H/metabolism , Complement Pathway, Alternative , Dendritic Cells/physiology , Properdin/metabolism , Cell Proliferation , Cells, Cultured , Complement C3b/metabolism , Complement Factor H/genetics , Humans , Immune Tolerance , Interferon-gamma/metabolism , Interleukin-27/metabolism , Isoantigens/immunology , Lymphocyte Activation , Properdin/genetics , RNA, Small Interfering/genetics
9.
Am J Physiol Renal Physiol ; 310(11): F1308-16, 2016 06 01.
Article in English | MEDLINE | ID: mdl-26984957

ABSTRACT

Previous studies have shown that complement activation on renal tubular cells is involved in the induction of interstitial fibrosis and cellular injury. Evidence suggests that the tubular cell damage is initiated by the alternative pathway (AP) of complement with properdin having an instrumental role. Properdin is a positive regulator of the AP, which can bind necrotic cells as well as viable proximal tubular epithelial cells (PTECs), inducing complement activation. Various studies have indicated that in the circulation there is an unidentified inhibitor of properdin. We investigated the ability of C-reactive protein (CRP), both in its monomeric (mCRP) and pentameric (pCRP) form, to inhibit AP activation and injury in vitro on renal tubular cells by fluorescent microscopy, ELISA, and flow cytometry. We demonstrated that preincubation of properdin with normal human serum inhibits properdin binding to viable PTECs. We identified mCRP as a factor able to bind to properdin in solution, thereby inhibiting its binding to PTECs. In contrast, pCRP exhibited no such binding and inhibitory effect. Furthermore, mCRP was able to inhibit properdin-directed C3 and C5b-9 deposition on viable PTECs. The inhibitory ability of mCRP was not unique for viable cells but also demonstrated for binding to necrotic Jurkat cells, a target for properdin binding and complement activation. In summary, mCRP is an inhibitor of properdin in both binding to necrotic cells and viable renal cells, regulating complement activation on the cell surface. We propose that mCRP limits amplification of tissue injury by controlling properdin-directed complement activation by damaged tissue and cells.


Subject(s)
C-Reactive Protein/metabolism , Complement Activation/drug effects , Complement C3/metabolism , Complement C5/metabolism , Kidney/metabolism , Properdin/pharmacology , Cell Line , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Kidney/drug effects , Protein Binding/drug effects
10.
Eur J Immunol ; 45(6): 1736-47, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25820702

ABSTRACT

IL-35 is a cytokine of the IL-12 family, existing as a heterodimer of IL-12p35 and Ebi3. IL-35 has anti-inflammatory properties and is produced by regulatory T cells in humans and mice, where it is required for optimal suppression of immune responses. Distinct from other IL-12 cytokines, the expression of IL-35 has not been described in antigen-presenting cells. In view of the immune-regulatory properties of IL-35, we investigated the expression, regulation, and function of IL-12p35 and Ebi3 in human monocyte-derived dendritic cells and tolerogenic DCs (tolDCs). These tolDCs do not produce IL-12p70 or the homodimer IL-12p40. We demonstrate that tolDCs completely lack transcriptional expression of IL-12p40. However, tolDCs maintain mRNA expression of IL-12p35 and Ebi3. Using intracellular flow cytometry and Western blot analysis, we show that tolDCs produce Ebi3 and IL-12p35, and both can be enhanced upon stimulation with IFN-γ, LPS, or CD40L. tolDCs supernatants have the capacity to suppress T-cell activation. Using IL12A silencing, we demonstrate that IL-12p35 is required for tolDCs to reach their full suppressive potential. Taken together, our results indicate that tolDCs produce IL-35, providing an additional novel mechanism by which tolDCs elicit their tolerogenic potential.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , Immune Tolerance , Interleukins/biosynthesis , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , B7 Antigens/metabolism , B7-2 Antigen/metabolism , B7-H1 Antigen/metabolism , Dendritic Cells/drug effects , Dexamethasone/pharmacology , Gene Expression , Humans , Immune Tolerance/drug effects , Interleukin-12/biosynthesis , Interleukin-12 Subunit p35/genetics , Interleukin-12 Subunit p35/metabolism , Interleukin-27/genetics , Interleukin-27/metabolism , Interleukins/genetics , Interleukins/metabolism , Lipopolysaccharides/immunology , Minor Histocompatibility Antigens , Phenotype
11.
Immunol Cell Biol ; 92(8): 688-98, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24913322

ABSTRACT

Fibroblasts reside within the renal interstitium in close proximity to neighbouring dendritic cells (DCs). It is likely that these cells have a central role in the maintenance and function of resident and infiltrating renal DCs, though studies to confirm this have been lacking. We investigated whether renal fibroblasts influence human DC generation and function. We found that co-culture with renal fibroblasts led to the generation of monocyte-derived dendritic cells (Fibro-DCs), with significantly reduced CD80, CD83 and CD86 but elevated B7H1 and B7DC expression. In addition, these Fibro-DCs displayed a reduced capacity to produce interleukin (IL)-12p40 and IL-12p70 but maintained normal levels of IL-23 and IL-27. Furthermore, IL-10 production was elevated, which together resulted in a regulatory DC population with a reduced capacity to stimulate allogenic T-cell proliferation and interferon γ production, while preserving IL-17A. Supernatant transfer experiments suggested that a soluble mediator from the fibroblasts was sufficient to inhibit the immunogenic capability of DCs. Further experiments demonstrated that IL-6 was at least partially responsible for the modulating effect of renal fibroblasts on DC generation and subsequent function. In summary, renal fibroblasts may have a crucial decisive role in regulating local DC immune responses in vivo. Better understanding of this cell population and their mechanisms of action may have therapeutic relevance in many immune-driven renal diseases.


Subject(s)
Cell Communication , Dendritic Cells/metabolism , Fibroblasts/metabolism , Kidney/metabolism , B7-1 Antigen/metabolism , B7-2 Antigen/metabolism , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Biomarkers , Cell Line , Coculture Techniques , Dendritic Cells/immunology , Humans , Immunomodulation/genetics , Interleukins/metabolism , Kidney/cytology , Kidney/immunology
12.
Sci Immunol ; 9(93): eadf2223, 2024 Mar 08.
Article in English | MEDLINE | ID: mdl-38457514

ABSTRACT

T cell immunoglobulin and mucin domain-containing protein 3 (TIM-3) is an important immune checkpoint molecule initially identified as a marker of IFN-γ-producing CD4+ and CD8+ T cells. Since then, our understanding of its role in immune responses has significantly expanded. Here, we review emerging evidence demonstrating unexpected roles for TIM-3 as a key regulator of myeloid cell function, in addition to recent work establishing TIM-3 as a delineator of terminal T cell exhaustion, thereby positioning TIM-3 at the interface between fatigued immune responses and reinvigoration. We share our perspective on the antagonism between TIM-3 and T cell stemness, discussing both cell-intrinsic and cell-extrinsic mechanisms underlying this relationship. Looking forward, we discuss approaches to decipher the underlying mechanisms by which TIM-3 regulates stemness, which has remarkable potential for the treatment of cancer, autoimmunity, and autoinflammation.


Subject(s)
Hepatitis A Virus Cellular Receptor 2 , Neoplasms , Humans , CD8-Positive T-Lymphocytes , Myeloid Cells , T-Cell Exhaustion
13.
Front Neurol ; 15: 1428198, 2024.
Article in English | MEDLINE | ID: mdl-38957351

ABSTRACT

Background: Patients with a large vessel occlusion require a transfer from a primary stroke centre to access thrombectomy, often over significant distances in regional areas. We sought to optimise stroke care access in the regional South Australian Tele-Strokeservice (SATS) to improve patient access to thrombectomy. Methods: We undertook a 24-month interventional historically controlled cohort study comparing acute stroke care metrics in the SATS. This consisted of a 12-month control period and a 12-month intervention monitoring period. The study intervention considered of an education package provided to the regional hospitals, a stroke neurologist roster to receive consultations and the intervention of a centralised tele-stroke system to provide treatment advice and organise patient transfers where needed. The SATS services 61 rural hospitals in South Australia, and Alice Springs in the Northern Territory. Suspected acute stroke patients presenting to the participating regional hospitals in SATS network where a telehealth consultation took place. Results: Over the study period, there were 919 patient referrals, with 449 consultations in the pre-intervention phase and 470 in the post-intervention phase. Demographic features in both epochs were similar. The post-intervention phase was associated with shorter door-to-scan time (35 min, IQR: 18,70; vs. 49 min, IQR:25,102, p < 0.0001), faster door-to-thrombolysis time (58 min, IQR: 39,91, vs.83 min, IQR: 55,100, p = 0.0324) and a higher portion of patients treated with thrombectomy (54, 11.5% vs. 26, 5.8%, p = 0.002). Conclusion: An optimised implementation of a streamlined telehealth platform with ongoing education and feedback to referring sites was associated with improved stroke workflow metrics and higher thrombectomy rates.

14.
Cancer Discov ; 14(4): 669-673, 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38571430

ABSTRACT

SUMMARY: The field of cancer neuroscience has begun to define the contributions of nerves to cancer initiation and progression; here, we highlight the future directions of basic and translational cancer neuroscience for malignancies arising outside of the central nervous system.


Subject(s)
Neoplasms , Neurosciences , Humans , Central Nervous System , Forecasting , Proteomics
15.
J Clin Invest ; 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38916965

ABSTRACT

Leukemia relapse is a major cause of death after allogeneic hematopoietic cell transplantation (allo-HCT). We tested the potential of targeting TIM-3 for improving graft-versus-leukemia (GVL) effects. We observed differential expression of TIM-3 ligands when hematopoietic stem cells overexpressed certain oncogenic-driver mutations. Anti-TIM-3 Ab-treatment improved survival of mice bearing leukemia with oncogene-induced TIM-3 ligand expression. Conversely, leukemia cells with low ligand expression were anti-TIM-3 treatment-resistant. In vitro, TIM-3 blockade or genetic deletion in CD8+ T cells (Tc) enhanced Tc activation, proliferation and IFN-γ production while enhancing GVL effects, preventing Tc exhaustion and improving Tc cytotoxicity and glycolysis in vivo. Conversely, TIM-3 deletion in myeloid cells did not affect allogeneic Tc proliferation and activation in vitro, suggesting that anti-TIM-3-treatment-mediated GVL effects are Tc-induced. In contrast to anti-PD-1 and anti-CTLA-4-treatment, anti-TIM-3-treatment did not enhance acute graft-versus-host-disease (aGVHD). TIM-3 and its ligands were frequently expressed in acute myeloid leukemia (AML) cells of patients with post-allo-HCT relapse. We deciphered the connection between oncogenic mutations found in AML and TIM-3 ligands expression and identify anti-TIM-3-treatment as a strategy to enhance GVL effects via metabolic and transcriptional Tc-reprogramming, without exacerbation of aGVHD. Our findings support clinical testing of anti-TIM-3 Abs in patients with AML relapse post-allo-HCT.

16.
J Immunother Cancer ; 11(5)2023 05.
Article in English | MEDLINE | ID: mdl-37208130

ABSTRACT

BACKGROUND: Although immune checkpoint inhibitors have been a breakthrough in clinical oncology, these therapies fail to produce durable responses in a significant fraction of patients. This lack of long-term efficacy may be due to a poor pre-existing network linking innate and adaptive immunity. Here, we present an antisense oligonucleotide (ASO)-based strategy that dually targets toll-like receptor 9 (TLR9) and programmed cell death ligand 1 (PD-L1), aiming to overcome resistance to anti-PD-L1 monoclonal therapy. METHODS: We designed a high-affinity immunomodulatory IM-TLR9:PD-L1-ASO antisense oligonucleotide (hereafter, IM-T9P1-ASO) targeting mouse PD-L1 messenger RNA and activating TLR9. Then, we performed in vitro and in vivo studies to validate the IM-T9P1-ASO activity, efficacy, and biological effects in tumors and draining lymph nodes. We also performed intravital imaging to study IM-T9P1-ASO pharmacokinetics in the tumor. RESULTS: IM-T9P1-ASO therapy, unlike PD-L1 antibody therapy, results in durable antitumor responses in multiple mouse cancer models. Mechanistically, IM-T9P1-ASO activates a state of tumor-associated dendritic cells (DCs), referred to here as DC3s, which have potent antitumor potential but express the PD-L1 checkpoint. IM-T9P1-ASO has two roles: it triggers the expansion of DC3s by engaging with TLR9 and downregulates PD-L1, thereby unleashing the antitumor functions of DC3s. This dual action leads to tumor rejection by T cells. The antitumor efficacy of IM-T9P1-ASO depends on the antitumor cytokine interleukin-12 (IL-12), produced by DC3s, and Batf3, a transcription factor required for DC development. CONCLUSIONS: By simultaneously targeting TLR9 and PD-L1, IM-T9P1-ASO amplifies antitumor responses via DC activation, leading to sustained therapeutic efficacy in mice. By highlighting differences and similarities between mouse and human DCs, this study could serve to develop similar therapeutic strategies for patients with cancer.


Subject(s)
Neoplasms , Toll-Like Receptor 9 , Humans , Mice , Animals , Toll-Like Receptor 9/metabolism , Immunotherapy/methods , Neoplasms/drug therapy , Oligonucleotides, Antisense , Dendritic Cells
17.
Cancer Immunol Res ; 11(12): 1571-1577, 2023 12 01.
Article in English | MEDLINE | ID: mdl-37906619

ABSTRACT

The Arthur and Sandra Irving Cancer Immunology Symposium has been created as a platform for established cancer immunologists to mentor trainees and young investigators as they launch their research career in the field. By sharing their different paths to success, the senior faculty mentors provide an invaluable resource to support the development of the next generation of leaders in the cancer immunology community. This Commentary describes some of the key topics that were discussed during the 2022 symposium: scientific and career trajectory, leadership, mentoring, collaborations, and publishing. For each of these topics, established investigators discussed the elements that facilitate success in these areas as well as mistakes that can hinder progress. Herein, we outline the critical points raised in these discussions for establishing a successful independent research career. These points are highly relevant for the broader scientific community.


Subject(s)
Mentoring , Neoplasms , Physicians , Humans , Mentors , Research Personnel , Neoplasms/therapy
18.
Health Soc Care Community ; 30(6): e5839-e5847, 2022 11.
Article in English | MEDLINE | ID: mdl-36069171

ABSTRACT

Women should be able to access mental health services that are safe, free from harassment and abuse. Yet, research indicates that women experiencing mental health issues are often not safe in mixed gender environments, and especially in inpatient settings. This qualitative study draws on a photo-elicitation method ('photovoice') and semi-structured interviews to explore women's experiences of a sub-acute women-only prevention and recovery care (PARC) service in Australia. Twelve women experiencing mental health issues were recruited via an aftercare peer support group for recent service participants. The women took photographs guided by the central question: 'What were your experiences of a women-only prevention and recovery care service?' They then shared these photographs with the researchers and each other, and described them in detail. Four key themes were identified by thematic analysis of the photovoice visual and narrative data: (a) Only women can understand what women go through; (b) I feel safer with no men around due to my history of trauma; (c) This environment feels safe, making it easier to talk and (d) Staff are accessible and make time for me to talk about difficult topics. Woven throughout the women's narratives was the expressed desire to feel safe and supported during the process of recovering. Aspects of service delivery that contributed to these feelings and facilitated shared support were also valued in this setting. These findings indicate that access to women-only services and peer support are not only valued by women experiencing mental health issues, but need to be more widely available to support their recovery. They also underline the importance of a trauma-informed approach for improving the gender sensitivity of services.


Subject(s)
Mental Health Services , Female , Humans , Qualitative Research , Australia
19.
Sci Immunol ; 7(69): eabm0631, 2022 03 11.
Article in English | MEDLINE | ID: mdl-35275752

ABSTRACT

Dendritic cells (DCs) sense environmental cues and adopt either an immune-stimulatory or regulatory phenotype, thereby fine-tuning immune responses. Identifying endogenous regulators that determine DC function can thus inform the development of therapeutic strategies for modulating the immune response in different disease contexts. Tim-3 plays an important role in regulating immune responses by inhibiting the activation status and the T cell priming ability of DC in the setting of cancer. Bat3 is an adaptor protein that binds to the tail of Tim-3; therefore, we studied its role in regulating the functional status of DCs. In murine models of autoimmunity (experimental autoimmune encephalomyelitis) and cancer (MC38-OVA-implanted tumor), lack of Bat3 expression in DCs alters the T cell compartment-it decreases TH1, TH17 and cytotoxic effector cells, increases regulatory T cells, and exhausted CD8+ tumor-infiltrating lymphocytes, resulting in the attenuation of autoimmunity and acceleration of tumor growth. We found that Bat3 expression levels were differentially regulated by activating versus inhibitory stimuli in DCs, indicating a role for Bat3 in the functional calibration of DC phenotypes. Mechanistically, loss of Bat3 in DCs led to hyperactive unfolded protein response and redirected acetyl-coenzyme A to increase cell intrinsic steroidogenesis. The enhanced steroidogenesis in Bat3-deficient DC suppressed T cell response in a paracrine manner. Our findings identified Bat3 as an endogenous regulator of DC function, which has implications for DC-based immunotherapies.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Hepatitis A Virus Cellular Receptor 2 , Molecular Chaperones/metabolism , Nuclear Proteins/metabolism , Adaptor Proteins, Signal Transducing , Animals , Autoimmunity , Dendritic Cells , Mice , T-Lymphocytes, Regulatory
SELECTION OF CITATIONS
SEARCH DETAIL