Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Nat Immunol ; 23(6): 904-915, 2022 06.
Article in English | MEDLINE | ID: mdl-35618834

ABSTRACT

Malignancy can be suppressed by the immune system. However, the classes of immunosurveillance responses and their mode of tumor sensing remain incompletely understood. Here, we show that although clear cell renal cell carcinoma (ccRCC) was infiltrated by exhaustion-phenotype CD8+ T cells that negatively correlated with patient prognosis, chromophobe RCC (chRCC) had abundant infiltration of granzyme A-expressing intraepithelial type 1 innate lymphoid cells (ILC1s) that positively associated with patient survival. Interleukin-15 (IL-15) promoted ILC1 granzyme A expression and cytotoxicity, and IL-15 expression in chRCC tumor tissue positively tracked with the ILC1 response. An ILC1 gene signature also predicted survival of a subset of breast cancer patients in association with IL-15 expression. Notably, ILC1s directly interacted with cancer cells, and IL-15 produced by cancer cells supported the expansion and anti-tumor function of ILC1s in a murine breast cancer model. Thus, ILC1 sensing of cancer cell IL-15 defines an immunosurveillance mechanism of epithelial malignancies.


Subject(s)
Breast Neoplasms , Interleukin-15/metabolism , Animals , Breast Neoplasms/genetics , CD8-Positive T-Lymphocytes , Female , Granzymes , Humans , Immunity, Innate , Lymphocytes , Mice
2.
Immunity ; 56(11): 2555-2569.e5, 2023 Nov 14.
Article in English | MEDLINE | ID: mdl-37967531

ABSTRACT

Tumors develop by invoking a supportive environment characterized by aberrant angiogenesis and infiltration of tumor-associated macrophages (TAMs). In a transgenic model of breast cancer, we found that TAMs localized to the tumor parenchyma and were smaller than mammary tissue macrophages. TAMs had low activity of the metabolic regulator mammalian/mechanistic target of rapamycin complex 1 (mTORC1), and depletion of negative regulator of mTORC1 signaling, tuberous sclerosis complex 1 (TSC1), in TAMs inhibited tumor growth in a manner independent of adaptive lymphocytes. Whereas wild-type TAMs exhibited inflammatory and angiogenic gene expression profiles, TSC1-deficient TAMs had a pro-resolving phenotype. TSC1-deficient TAMs relocated to a perivascular niche, depleted protein C receptor (PROCR)-expressing endovascular endothelial progenitor cells, and rectified the hyperpermeable blood vasculature, causing tumor tissue hypoxia and cancer cell death. TSC1-deficient TAMs were metabolically active and effectively eliminated PROCR-expressing endothelial cells in cell competition experiments. Thus, TAMs exhibit a TSC1-dependent mTORC1-low state, and increasing mTORC1 signaling promotes a pro-resolving state that suppresses tumor growth, defining an innate immune tumor suppression pathway that may be exploited for cancer immunotherapy.


Subject(s)
Endothelial Progenitor Cells , Tumor Suppressor Proteins , Animals , Humans , TOR Serine-Threonine Kinases/metabolism , Tuberous Sclerosis Complex 1 Protein/genetics , Tumor-Associated Macrophages/metabolism , Endothelial Progenitor Cells/metabolism , Endothelial Protein C Receptor , Mechanistic Target of Rapamycin Complex 1 , Neovascularization, Pathologic , Mammals
3.
Immunity ; 55(11): 2044-2058.e5, 2022 11 08.
Article in English | MEDLINE | ID: mdl-36288724

ABSTRACT

Tumors are populated by antigen-presenting cells (APCs) including macrophage subsets with distinct origins and functions. Here, we examined how cancer impacts mononuclear phagocytic APCs in a murine model of breast cancer. Tumors induced the expansion of monocyte-derived tumor-associated macrophages (TAMs) and the activation of type 1 dendritic cells (DC1s), both of which expressed and required the transcription factor interferon regulatory factor-8 (IRF8). Although DC1s mediated cytotoxic T lymphocyte (CTL) priming in tumor-draining lymph nodes, TAMs promoted CTL exhaustion in the tumor, and IRF8 was required for TAMs' ability to present cancer cell antigens. TAM-specific IRF8 deletion prevented exhaustion of cancer-cell-reactive CTLs and suppressed tumor growth. Tumors from patients with immune-infiltrated renal cell carcinoma had abundant TAMs that expressed IRF8 and were enriched for an IRF8 gene expression signature. Furthermore, the TAM-IRF8 signature co-segregated with CTL exhaustion signatures across multiple cancer types. Thus, CTL exhaustion is promoted by TAMs via IRF8.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Humans , Animals , Mice , Tumor-Associated Macrophages , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , T-Lymphocytes, Cytotoxic , Dendritic Cells
5.
Nature ; 619(7970): 616-623, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37380769

ABSTRACT

In metazoan organisms, cell competition acts as a quality control mechanism to eliminate unfit cells in favour of their more robust neighbours1,2. This mechanism has the potential to be maladapted, promoting the selection of aggressive cancer cells3-6. Tumours are metabolically active and are populated by stroma cells7,8, but how environmental factors affect cancer cell competition remains largely unknown. Here we show that tumour-associated macrophages (TAMs) can be dietarily or genetically reprogrammed to outcompete MYC-overexpressing cancer cells. In a mouse model of breast cancer, MYC overexpression resulted in an mTORC1-dependent 'winner' cancer cell state. A low-protein diet inhibited mTORC1 signalling in cancer cells and reduced tumour growth, owing unexpectedly to activation of the transcription factors TFEB and TFE3 and mTORC1 in TAMs. Diet-derived cytosolic amino acids are sensed by Rag GTPases through the GTPase-activating proteins GATOR1 and FLCN to control Rag GTPase effectors including TFEB and TFE39-14. Depletion of GATOR1 in TAMs suppressed the activation of TFEB, TFE3 and mTORC1 under the low-protein diet condition, causing accelerated tumour growth; conversely, depletion of FLCN or Rag GTPases in TAMs activated TFEB, TFE3 and mTORC1 under the normal protein diet condition, causing decelerated tumour growth. Furthermore, mTORC1 hyperactivation in TAMs and cancer cells and their competitive fitness were dependent on the endolysosomal engulfment regulator PIKfyve. Thus, noncanonical engulfment-mediated Rag GTPase-independent mTORC1 signalling in TAMs controls competition between TAMs and cancer cells, which defines a novel innate immune tumour suppression pathway that could be targeted for cancer therapy.


Subject(s)
Cell Competition , Cellular Reprogramming Techniques , Immunity, Innate , Neoplasms , Tumor-Associated Macrophages , Animals , Mice , Amino Acids/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Competition/genetics , Cell Competition/immunology , Dietary Proteins/pharmacology , Disease Models, Animal , GTP Phosphohydrolases/metabolism , Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism
6.
Nature ; 605(7908): 139-145, 2022 05.
Article in English | MEDLINE | ID: mdl-35444279

ABSTRACT

Cellular transformation induces phenotypically diverse populations of tumour-infiltrating T cells1-5, and immune checkpoint blockade therapies preferentially target T cells that recognize cancer cell neoantigens6,7. Yet, how other classes of tumour-infiltrating T cells contribute to cancer immunosurveillance remains elusive. Here, in a survey of T cells in mouse and human malignancies, we identified a population of αß T cell receptor (TCR)-positive FCER1G-expressing innate-like T cells with high cytotoxic potential8 (ILTCKs). These cells were broadly reactive to unmutated self-antigens, arose from distinct thymic progenitors following early encounter with cognate antigens, and were continuously replenished by thymic progenitors during tumour progression. Notably, expansion and effector differentiation of intratumoural ILTCKs depended on interleukin-15 (IL-15) expression in cancer cells, and inducible activation of IL-15 signalling in adoptively transferred ILTCK progenitors suppressed tumour growth. Thus, the antigen receptor self-reactivity, unique ontogeny, and distinct cancer cell-sensing mechanism distinguish ILTCKs from conventional cytotoxic T cells, and define a new class of tumour-elicited immune response.


Subject(s)
Immunity, Innate , Interleukin-15 , Neoplasms , Animals , Cell Differentiation , Mice , Neoplasms/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes, Cytotoxic/metabolism
7.
Immunity ; 46(1): 3-5, 2017 01 17.
Article in English | MEDLINE | ID: mdl-28099863

ABSTRACT

Mechanisms that govern PD1 expression and exhaustion in T cells are not fully understood. In this issue of Immunity, Stephen et al. (2017) uncover a key role for the genome organizer Satb1 in restraining PD1 expression and promoting tumor immunity.


Subject(s)
Matrix Attachment Region Binding Proteins/genetics , T-Lymphocytes , Genome , Humans
8.
Nature ; 587(7832): 121-125, 2020 11.
Article in English | MEDLINE | ID: mdl-33087933

ABSTRACT

Cancer arises from malignant cells that exist in dynamic multilevel interactions with the host tissue. Cancer therapies aiming to directly kill cancer cells, including oncogene-targeted therapy and immune-checkpoint therapy that revives tumour-reactive cytotoxic T lymphocytes, are effective in some patients1,2, but acquired resistance frequently develops3,4. An alternative therapeutic strategy aims to rectify the host tissue pathology, including abnormalities in the vasculature that foster cancer progression5,6; however, neutralization of proangiogenic factors such as vascular endothelial growth factor A (VEGFA) has had limited clinical benefits7,8. Here, following the finding that transforming growth factor-ß (TGF-ß) suppresses T helper 2 (TH2)-cell-mediated cancer immunity9, we show that blocking TGF-ß signalling in CD4+ T cells remodels the tumour microenvironment and restrains cancer progression. In a mouse model of breast cancer resistant to immune-checkpoint or anti-VEGF therapies10,11, inducible genetic deletion of the TGF-ß receptor II (TGFBR2) in CD4+ T cells suppressed tumour growth. For pharmacological blockade, we engineered a bispecific receptor decoy by attaching the TGF-ß-neutralizing TGFBR2 extracellular domain to ibalizumab, a non-immunosuppressive CD4 antibody12,13, and named it CD4 TGF-ß Trap (4T-Trap). Compared with a non-targeted TGF-ß-Trap, 4T-Trap selectively inhibited TH cell TGF-ß signalling in tumour-draining lymph nodes, causing reorganization of tumour vasculature and cancer cell death, a process dependent on the TH2 cytokine interleukin-4 (IL-4). Notably, the 4T-Trap-induced tumour tissue hypoxia led to increased VEGFA expression. VEGF inhibition enhanced the starvation-triggered cancer cell death and amplified the antitumour effect of 4T-Trap. Thus, targeted TGF-ß signalling blockade in helper T cells elicits an effective tissue-level cancer defence response that can provide a basis for therapies directed towards the cancer environment.


Subject(s)
Breast Neoplasms/therapy , Immunotherapy , Signal Transduction/drug effects , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/immunology , Transforming Growth Factor beta/antagonists & inhibitors , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Breast Neoplasms/blood supply , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Death/drug effects , Cell Hypoxia , Cell Line, Tumor , Female , HEK293 Cells , Humans , Interleukin-4/immunology , Lymph Nodes/cytology , Lymph Nodes/drug effects , Lymph Nodes/immunology , Male , Mice , Receptor, Transforming Growth Factor-beta Type II/chemistry , Receptor, Transforming Growth Factor-beta Type II/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Transforming Growth Factor beta/immunology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism
9.
Nature ; 587(7832): 115-120, 2020 11.
Article in English | MEDLINE | ID: mdl-33087928

ABSTRACT

The immune system uses two distinct defence strategies against infections: microbe-directed pathogen destruction characterized by type 1 immunity1, and host-directed pathogen containment exemplified by type 2 immunity in induction of tissue repair2. Similar to infectious diseases, cancer progresses with self-propagating cancer cells inflicting host-tissue damage. The immunological mechanisms of cancer cell destruction are well defined3-5, but whether immune-mediated cancer cell containment can be induced remains poorly understood. Here we show that depletion of transforming growth factor-ß receptor 2 (TGFBR2) in CD4+ T cells, but not CD8+ T cells, halts cancer progression as a result of tissue healing and remodelling of the blood vasculature, causing cancer cell hypoxia and death in distant avascular regions. Notably, the host-directed protective response is dependent on the T helper 2 cytokine interleukin-4 (IL-4), but not the T helper 1 cytokine interferon-γ (IFN-γ). Thus, type 2 immunity can be mobilized as an effective tissue-level defence mechanism against cancer.


Subject(s)
Neoplasms/immunology , Neoplasms/pathology , Signal Transduction/immunology , Th2 Cells/immunology , Transforming Growth Factor beta/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Death/drug effects , Cell Hypoxia , Cell Line , Disease Progression , Female , Gene Expression Regulation/immunology , Humans , Interferon-gamma/immunology , Interleukin-4/immunology , Male , Mice , Mice, Inbred C57BL , Neoplasms/blood supply , Neoplasms/metabolism , Receptor, Transforming Growth Factor-beta Type II/deficiency , Signal Transduction/drug effects , Stromal Cells/cytology , Stromal Cells/immunology , Th2 Cells/metabolism , Transforming Growth Factor beta/antagonists & inhibitors
10.
J Immunol ; 200(2): 408-414, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29311382

ABSTRACT

Innate lymphoid cells (ILCs) are critical components of tissues in the body, providing a first line of defense against challenges to host integrity. In contrast to strictly cytokine-producing helper ILCs, resident innate lymphocyte populations with cytolytic potential have been identified in multiple tissues in both mouse and human. These cells express the transcription factor Tbet, NK cell receptors, granzymes, perforin, and death receptors, and can directly kill tumor cells. Signals in the tumor microenvironment may promote this response, including the cytokine IL-15 and stress-associated ligands for activating NK receptors. Although there is evidence that these cells are tissue and tumor resident, their lineage remains unclear. Whether they are derived from the NK or helper ILC lineages or represent a third differentiation pathway remains to be determined. A better understanding of their lineage will help clarify their regulation and function in the context of antitumor immunity.


Subject(s)
Cytotoxicity, Immunologic , Immunity, Innate , Lymphocyte Subsets/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Neoplasms/immunology , Animals , Cytokines/metabolism , Humans , Lymphocyte Activation/immunology , Lymphocyte Subsets/metabolism , Lymphocytes, Tumor-Infiltrating/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Tumor Microenvironment
11.
Proc Natl Acad Sci U S A ; 114(36): E7536-E7544, 2017 09 05.
Article in English | MEDLINE | ID: mdl-28827353

ABSTRACT

Peripheral T cell tolerance is promoted by the regulatory cytokine TGF-ß and Foxp3-expressing Treg cells. However, whether TGF-ß and Treg cells are part of the same regulatory module, or exist largely as distinct pathways to repress self-reactive T cells remains incompletely understood. Using a transgenic model of autoimmune diabetes, here we show that ablation of TGF-ß receptor II (TßRII) in T cells, but not Foxp3 deficiency, resulted in early-onset diabetes with complete penetrance. The rampant autoimmune disease was associated with enhanced T cell priming and elevated T cell expression of the inflammatory cytokine GM-CSF, concomitant with pancreatic infiltration of inflammatory monocytes that triggered immunopathology. Ablation of the GM-CSF receptor alleviated the monocyte response and inhibited disease development. These findings reveal that TGF-ß promotes T cell tolerance primarily via Foxp3-independent mechanisms and prevents autoimmunity in this model by repressing the cross talk between adaptive and innate immune systems.


Subject(s)
Forkhead Transcription Factors/metabolism , Peripheral Tolerance/immunology , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta/metabolism , Adaptive Immunity/immunology , Animals , Autoimmunity/immunology , Cytokines/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Immunity, Innate/immunology , Inflammation/metabolism , Mice , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , T-Lymphocytes, Regulatory/immunology
12.
STAR Protoc ; 4(1): 102136, 2023 03 17.
Article in English | MEDLINE | ID: mdl-36861825

ABSTRACT

Group 1 innate lymphocytes are heterogeneous, and their ontogeny and function remain ambiguous. Here, we describe a protocol to measure cell ontogeny and effector functions of natural killer (NK) and ILC1 subsets based on current understanding of their differentiation pathways. We use cre drivers to genetically fate-map cells, tracking plasticity between mature NK and ILC1. We describe innate lymphoid cell precursor transfer studies that determine ontogeny of granzyme-C-expressing ILC1. Additionally, we detail in vitro killing assays that test cytolytic potential of ILC1s. For complete details on the use and execution of this protocol, please refer to Nixon et al. (2022).1.


Subject(s)
Immunity, Innate , Killer Cells, Natural , Animals , Mice , Cell Differentiation
13.
STAR Protoc ; 4(2): 102185, 2023 Mar 22.
Article in English | MEDLINE | ID: mdl-36952337

ABSTRACT

Here, we focus on tumor-associated macrophages (TAMs) in the PyMT model of breast cancer, detailing a protocol for assessing antigen presentation capabilities of immune populations of interest. We describe a stringent bone marrow chimera system to demonstrate presentation of exogenous antigen that is acquired and processed in the tumor microenvironment. We describe steps for testing antigen presentation activity of TAMs to CD8+ T cells in vivo and ex vivo and the requirement for the transcription factor IRF8 in this function. For complete details on the use and execution of this protocol, please refer to Nixon et al. (2022).1.

14.
Nat Rev Immunol ; 23(6): 346-362, 2023 06.
Article in English | MEDLINE | ID: mdl-36380023

ABSTRACT

The immune system responds to cancer in two main ways. First, there are prewired responses involving myeloid cells, innate lymphocytes and innate-like adaptive lymphocytes that either reside in premalignant tissues or migrate directly to tumours, and second, there are antigen priming-dependent responses, in which adaptive lymphocytes are primed in secondary lymphoid organs before homing to tumours. Transforming growth factor-ß (TGFß) - one of the most potent and pleiotropic regulatory cytokines - controls almost every stage of the tumour-elicited immune response, from leukocyte development in primary lymphoid organs to their priming in secondary lymphoid organs and their effector functions in the tumour itself. The complexity of TGFß-regulated immune cell circuitries, as well as the contextual roles of TGFß signalling in cancer cells and tumour stromal cells, necessitates the use of rigorous experimental systems that closely recapitulate human cancer, such as autochthonous tumour models, to uncover the underlying immunobiology. The diverse functions of TGFß in healthy tissues further complicate the search for effective and safe cancer therapeutics targeting the TGFß pathway. Here we discuss the contextual complexity of TGFß signalling in tumour-elicited immune responses and explain how understanding this may guide the development of mechanism-based cancer immunotherapy.


Subject(s)
Neoplasms , Transforming Growth Factor beta , Humans , Transforming Growth Factor beta/metabolism , Neoplasms/pathology , Cytokines , Immunotherapy , Immunity
15.
Sci Immunol ; 7(70): eabi8642, 2022 04 08.
Article in English | MEDLINE | ID: mdl-35394814

ABSTRACT

Innate lymphocytes are integral components of the cellular immune system that can coordinate host defense against a multitude of challenges and trigger immunopathology when dysregulated. Natural killer (NK) cells and innate lymphoid cells (ILCs) are innate immune effectors postulated to functionally mirror conventional cytotoxic T lymphocytes and helper T cells, respectively. Here, we showed that the cytolytic molecule granzyme C was expressed in cells with the phenotype of type 1 ILCs (ILC1s) in mouse liver and salivary gland. Cell fate-mapping and transfer studies revealed that granzyme C-expressing innate lymphocytes could be derived from ILC progenitors and did not interconvert with NK cells, ILC2s, or ILC3s. Granzyme C defined a maturation state of ILC1s. These granzyme C-expressing ILC1s required the transcription factors T-bet and, to a lesser extent, Eomes and support from transforming growth factor-ß (TGF-ß) signaling for their maintenance in the salivary gland. In a transgenic mouse breast cancer model, depleting ILC1s caused accelerated tumor growth. ILC1s gained granzyme C expression following interleukin-15 (IL-15) stimulation, which enabled perforin-mediated cytotoxicity. Constitutive activation of STAT5, a transcription factor regulated by IL-15, in granzyme C-expressing ILC1s triggered lethal perforin-dependent autoimmunity in neonatal mice. Thus, granzyme C marks a cytotoxic effector state of ILC1s, broadening their function beyond "helper-like" lymphocytes.


Subject(s)
Immunity, Innate , Interleukin-15 , Animals , Autoimmunity , Granzymes , Killer Cells, Natural , Mice , Perforin
16.
J Clin Invest ; 132(17)2022 09 01.
Article in English | MEDLINE | ID: mdl-35852856

ABSTRACT

Immune checkpoint blockade (ICB) has demonstrated clinical success in "inflamed" tumors with substantial T cell infiltrates, but tumors with an immune-desert tumor microenvironment (TME) fail to benefit. The tumor cell-intrinsic molecular mechanisms of the immune-desert phenotype remain poorly understood. Here, we demonstrated that inactivation of the polycomb-repressive complex 2 (PRC2) core components embryonic ectoderm development (EED) or suppressor of zeste 12 homolog (SUZ12), a prevalent genetic event in malignant peripheral nerve sheath tumors (MPNSTs) and sporadically in other cancers, drove a context-dependent immune-desert TME. PRC2 inactivation reprogramed the chromatin landscape that led to a cell-autonomous shift from primed baseline signaling-dependent cellular responses (e.g., IFN-γ signaling) to PRC2-regulated developmental and cellular differentiation transcriptional programs. Further, PRC2 inactivation led to diminished tumor immune infiltrates through reduced chemokine production and impaired antigen presentation and T cell priming, resulting in primary resistance to ICB. Intratumoral delivery of inactivated modified vaccinia virus Ankara (MVA) enhanced tumor immune infiltrates and sensitized PRC2-loss tumors to ICB. Our results identify molecular mechanisms of PRC2 inactivation-mediated, context-dependent epigenetic reprogramming that underline the immune-desert phenotype in cancer. Our studies also point to intratumoral delivery of immunogenic viruses as an initial therapeutic strategy to modulate the immune-desert TME and capitalize on the clinical benefit of ICB.


Subject(s)
Neoplasms , Viruses , Chromatin , Humans , Polycomb Repressive Complex 2/genetics , Tumor Microenvironment , Viruses/genetics
17.
Cancer Discov ; 12(10): 2308-2329, 2022 10 05.
Article in English | MEDLINE | ID: mdl-35758895

ABSTRACT

It is poorly understood how the tumor immune microenvironment influences disease recurrence in localized clear-cell renal cell carcinoma (ccRCC). Here we performed whole-transcriptomic profiling of 236 tumors from patients assigned to the placebo-only arm of a randomized, adjuvant clinical trial for high-risk localized ccRCC. Unbiased pathway analysis identified myeloid-derived IL6 as a key mediator. Furthermore, a novel myeloid gene signature strongly correlated with disease recurrence and overall survival on uni- and multivariate analyses and is linked to TP53 inactivation across multiple data sets. Strikingly, effector T-cell gene signatures, infiltration patterns, and exhaustion markers were not associated with disease recurrence. Targeting immunosuppressive myeloid inflammation with an adenosine A2A receptor antagonist in a novel, immunocompetent, Tp53-inactivated mouse model significantly reduced metastatic development. Our findings suggest that myeloid inflammation promotes disease recurrence in ccRCC and is targetable as well as provide a potential biomarker-based framework for the design of future immuno-oncology trials in ccRCC. SIGNIFICANCE: Improved understanding of factors that influence metastatic development in localized ccRCC is greatly needed to aid accurate prediction of disease recurrence, clinical decision-making, and future adjuvant clinical trial design. Our analysis implicates intratumoral myeloid inflammation as a key driver of metastasis in patients and a novel immunocompetent mouse model. This article is highlighted in the In This Issue feature, p. 2221.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Animals , Mice , Adenosine A2 Receptor Antagonists , Biomarkers, Tumor/genetics , Carcinoma, Renal Cell/pathology , Inflammation , Interleukin-6 , Kidney Neoplasms/pathology , Neoplasm Recurrence, Local/pathology , Prognosis , Tumor Microenvironment/genetics , Humans
18.
Cancer Cell ; 39(5): 662-677.e6, 2021 05 10.
Article in English | MEDLINE | ID: mdl-33861994

ABSTRACT

Clear cell renal cell carcinomas (ccRCCs) are highly immune infiltrated, but the effect of immune heterogeneity on clinical outcome in ccRCC has not been fully characterized. Here we perform paired single-cell RNA (scRNA) and T cell receptor (TCR) sequencing of 167,283 cells from multiple tumor regions, lymph node, normal kidney, and peripheral blood of two immune checkpoint blockade (ICB)-naïve and four ICB-treated patients to map the ccRCC immune landscape. We detect extensive heterogeneity within and between patients, with enrichment of CD8A+ tissue-resident T cells in a patient responsive to ICB and tumor-associated macrophages (TAMs) in a resistant patient. A TCR trajectory framework suggests distinct T cell differentiation pathways between patients responding and resistant to ICB. Finally, scRNA-derived signatures of tissue-resident T cells and TAMs are associated with response to ICB and targeted therapies across multiple independent cohorts. Our study establishes a multimodal interrogation of the cellular programs underlying therapeutic efficacy in ccRCC.


Subject(s)
Carcinoma, Renal Cell/genetics , Kidney Neoplasms/genetics , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , Humans , Kidney Neoplasms/immunology , Lymphocyte Activation/genetics , Programmed Cell Death 1 Receptor/genetics , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology
19.
J Exp Med ; 217(1)2020 01 06.
Article in English | MEDLINE | ID: mdl-31649036

ABSTRACT

Foxp3+ regulatory T (T reg) cells are pivotal regulators of immune tolerance, with T cell receptor (TCR)-driven activated T reg (aT reg) cells playing a central role; yet how TCR signaling propagates to control aT reg cell responses remains poorly understood. Here we show that TCR signaling induces expression of amino acid transporters, and renders amino acid-induced activation of mTORC1 in aT reg cells. T reg cell-specific ablation of the Rag family small GTPases RagA and RagB impairs amino acid-induced mTORC1 signaling, causing defective amino acid anabolism, reduced T reg cell proliferation, and a rampant autoimmune disorder similar in severity to that triggered by T reg cell-specific TCR deficiency. Notably, T reg cells in peripheral tissues, including tumors, are more sensitive to Rag GTPase-dependent nutrient sensing. Ablation of RagA alone impairs T reg cell accumulation in the tumor, resulting in enhanced antitumor immunity. Thus, nutrient mTORC1 signaling is an essential component of TCR-initiated T reg cell reprogramming, and Rag GTPase activities may be titrated to break tumor immune tolerance.


Subject(s)
Immune Tolerance/immunology , Mechanistic Target of Rapamycin Complex 1/immunology , Nutrients/immunology , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology , Amino Acid Transport Systems/immunology , Animals , Female , Male , Mice , Mice, Inbred C57BL , Monomeric GTP-Binding Proteins/immunology , Receptors, Antigen, T-Cell/immunology , TOR Serine-Threonine Kinases/immunology
20.
Cancer Discov ; 9(4): 510-525, 2019 04.
Article in English | MEDLINE | ID: mdl-30622105

ABSTRACT

Metastasis remains the main reason for renal cell carcinoma (RCC)-associated mortality. Tyrosine kinase inhibitors (TKI) impart clinical benefit for most patients with RCC, but the determinants of response are poorly understood. We report an integrated genomic and transcriptomic analysis of patients with metastatic clear cell RCC (ccRCC) treated with TKI therapy and identify predictors of response. Patients in the COMPARZ phase III trial received first-line sunitinib or pazopanib with comparable efficacy. RNA-based analyses revealed four distinct molecular subgroups associated with response and survival. Characterization of these subgroups identified mutation profiles, angiogenesis, and macrophage infiltration programs to be powerful predictors of outcome with TKI therapy. Notably, predictors differed by the type of TKI received. Our study emphasizes the clinical significance of angiogenesis and immune tumor microenvironment and suggests that the critical effects its various aspects have on TKI efficacy vary by agent. This has broad implications for optimizing precision treatment of RCC. SIGNIFICANCE: The determinants of response to TKI therapy in metastatic ccRCC remain unknown. Our study demonstrates that key angiogenic and immune profiles of the tumor microenvironment may affect TKI response. These findings have the potential to inform treatment personalization in patients with RCC.This article is highlighted in the In This Issue feature, p. 453.


Subject(s)
Carcinoma, Renal Cell/genetics , Gene Expression Profiling/methods , Kidney Neoplasms/genetics , Female , Humans , Male , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL