Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.754
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 25(7): 1257-1269, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38806707

ABSTRACT

The circadian clock is a critical regulator of immunity, and this circadian control of immune modulation has an essential function in host defense and tumor immunosurveillance. Here we use a single-cell RNA sequencing approach and a genetic model of colorectal cancer to identify clock-dependent changes to the immune landscape that control the abundance of immunosuppressive cells and consequent suppression of cytotoxic CD8+ T cells. Of these immunosuppressive cell types, PD-L1-expressing myeloid-derived suppressor cells (MDSCs) peak in abundance in a rhythmic manner. Disruption of the epithelial cell clock regulates the secretion of cytokines that promote heightened inflammation, recruitment of neutrophils and the subsequent development of MDSCs. We also show that time-of-day anti-PD-L1 delivery is most effective when synchronized with the abundance of immunosuppressive MDSCs. Collectively, these data indicate that circadian gating of tumor immunosuppression informs the timing and efficacy of immune checkpoint inhibitors.


Subject(s)
B7-H1 Antigen , Circadian Clocks , Immune Checkpoint Inhibitors , Myeloid-Derived Suppressor Cells , Animals , Mice , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , Circadian Clocks/immunology , B7-H1 Antigen/metabolism , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , Mice, Inbred C57BL , Circadian Rhythm/immunology , CD8-Positive T-Lymphocytes/immunology , Colorectal Neoplasms/immunology , Colorectal Neoplasms/therapy , Colorectal Neoplasms/drug therapy , Tumor Microenvironment/immunology , Immune Tolerance , Humans , Female , Cell Line, Tumor , Single-Cell Analysis , Immunosuppression Therapy , Cytokines/metabolism , Male
2.
Nat Immunol ; 22(12): 1524-1537, 2021 12.
Article in English | MEDLINE | ID: mdl-34795443

ABSTRACT

Inhibiting PD-1:PD-L1 signaling has transformed therapeutic immune restoration. CD4+ T cells sustain immunity in chronic infections and cancer, yet little is known about how PD-1 signaling modulates CD4+ helper T (TH) cell responses or the ability to restore CD4+ TH-mediated immunity by checkpoint blockade. We demonstrate that PD-1:PD-L1 specifically suppressed CD4+ TH1 cell amplification, prevents CD4+ TH1 cytokine production and abolishes CD4+ cytotoxic killing capacity during chronic infection in mice. Inhibiting PD-L1 rapidly restored these functions, while simultaneously amplifying and activating TH1-like T regulatory cells, demonstrating a system-wide CD4-TH1 recalibration. This effect coincided with decreased T cell antigen receptor signaling, and re-directed type I interferon (IFN) signaling networks towards dominant IFN-γ-mediated responses. Mechanistically, PD-L1 blockade specifically targeted defined populations with pre-established, but actively suppressed proliferative potential, with limited impact on minimally cycling TCF-1+ follicular helper T cells, despite high PD-1 expression. Thus, CD4+ T cells require unique differentiation and functional states to be targets of PD-L1-directed suppression and therapeutic restoration.


Subject(s)
B7-H1 Antigen/antagonists & inhibitors , Immune Checkpoint Inhibitors/pharmacology , Lymphocyte Activation/drug effects , Lymphocytic Choriomeningitis/drug therapy , Lymphocytic choriomeningitis virus/immunology , Th1 Cells/drug effects , Adoptive Transfer , Animals , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Cell Proliferation/drug effects , Chronic Disease , Cytokines/metabolism , Cytotoxicity, Immunologic/drug effects , Disease Models, Animal , Female , Gene Regulatory Networks , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/metabolism , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/pathogenicity , Mice, Inbred C57BL , Phenotype , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , Th1 Cells/immunology , Th1 Cells/metabolism , Th1 Cells/virology , Transcriptome
3.
Cell ; 168(3): 487-502.e15, 2017 01 26.
Article in English | MEDLINE | ID: mdl-28111070

ABSTRACT

Immune responses involve coordination across cell types and tissues. However, studies in cancer immunotherapy have focused heavily on local immune responses in the tumor microenvironment. To investigate immune activity more broadly, we performed an organism-wide study in genetically engineered cancer models using mass cytometry. We analyzed immune responses in several tissues after immunotherapy by developing intuitive models for visualizing single-cell data with statistical inference. Immune activation was evident in the tumor and systemically shortly after effective therapy was administered. However, during tumor rejection, only peripheral immune cells sustained their proliferation. This systemic response was coordinated across tissues and required for tumor eradication in several immunotherapy models. An emergent population of peripheral CD4 T cells conferred protection against new tumors and was significantly expanded in patients responding to immunotherapy. These studies demonstrate the critical impact of systemic immune responses that drive tumor rejection.


Subject(s)
Immunotherapy , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocyte Subsets/immunology , Animals , B7-H1 Antigen/antagonists & inhibitors , Bone Marrow/immunology , Cell Proliferation , Disease Models, Animal , Female , Humans , Immune Tolerance , Killer Cells, Natural/immunology , Lymphocyte Activation , Lymphoid Tissue/immunology , Male , Melanoma/immunology , Melanoma/therapy , Mice, Inbred BALB C , Mice, Inbred C57BL , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/therapy , Tumor Microenvironment
4.
Immunity ; 55(1): 82-97.e8, 2022 01 11.
Article in English | MEDLINE | ID: mdl-34847356

ABSTRACT

CD8+ T cells responding to chronic infection adapt an altered differentiation program that provides some restraint on pathogen replication yet limits immunopathology. This adaptation is imprinted in stem-like cells and propagated to their progeny. Understanding the molecular control of CD8+ T cell differentiation in chronic infection has important therapeutic implications. Here, we find that the chemokine receptor CXCR3 is highly expressed on viral-specific stem-like CD8+ T cells and that one of its ligands, CXCL10, regulates the persistence and heterogeneity of responding CD8+ T cells in spleens of mice chronically infected with lymphocytic choriomeningitis virus. CXCL10 is produced by inflammatory monocytes and fibroblasts of the splenic red pulp, where it grants stem-like cells access to signals promoting differentiation and limits their exposure to pro-survival niches in the white pulp. Consequently, functional CD8+ T cell responses are greater in Cxcl10-/- mice and are associated with a lower viral set point.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Chemokine CXCL10/metabolism , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/physiology , Monocytes/metabolism , Receptors, CXCR3/metabolism , Spleen/pathology , Animals , B7-H1 Antigen/antagonists & inhibitors , Cell Differentiation , Cell Proliferation , Cell Self Renewal , Chemokine CXCL10/genetics , Chronic Disease , Clonal Selection, Antigen-Mediated , Female , Hepatocyte Nuclear Factor 1-alpha/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, CXCR3/genetics
5.
Cell ; 162(6): 1229-41, 2015 Sep 10.
Article in English | MEDLINE | ID: mdl-26321679

ABSTRACT

Failure of T cells to protect against cancer is thought to result from lack of antigen recognition, chronic activation, and/or suppression by other cells. Using a mouse sarcoma model, we show that glucose consumption by tumors metabolically restricts T cells, leading to their dampened mTOR activity, glycolytic capacity, and IFN-γ production, thereby allowing tumor progression. We show that enhancing glycolysis in an antigenic "regressor" tumor is sufficient to override the protective ability of T cells to control tumor growth. We also show that checkpoint blockade antibodies against CTLA-4, PD-1, and PD-L1, which are used clinically, restore glucose in tumor microenvironment, permitting T cell glycolysis and IFN-γ production. Furthermore, we found that blocking PD-L1 directly on tumors dampens glycolysis by inhibiting mTOR activity and decreasing expression of glycolysis enzymes, reflecting a role for PD-L1 in tumor glucose utilization. Our results establish that tumor-imposed metabolic restrictions can mediate T cell hyporesponsiveness during cancer.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Glycolysis , Lymphocytes, Tumor-Infiltrating/metabolism , Neoplasms/metabolism , Tumor Microenvironment , Animals , Antibodies, Monoclonal/administration & dosage , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , CD8-Positive T-Lymphocytes/immunology , CTLA-4 Antigen/antagonists & inhibitors , CTLA-4 Antigen/immunology , Interferon-gamma/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Neoplasms/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology
6.
Nature ; 627(8004): 646-655, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38418879

ABSTRACT

Tiragolumab, an anti-TIGIT antibody with an active IgG1κ Fc, demonstrated improved outcomes in the phase 2 CITYSCAPE trial (ClinicalTrials.gov: NCT03563716 ) when combined with atezolizumab (anti-PD-L1) versus atezolizumab alone1. However, there remains little consensus on the mechanism(s) of response with this combination2. Here we find that a high baseline of intratumoural macrophages and regulatory T cells is associated with better outcomes in patients treated with atezolizumab plus tiragolumab but not with atezolizumab alone. Serum sample analysis revealed that macrophage activation is associated with a clinical benefit in patients who received the combination treatment. In mouse tumour models, tiragolumab surrogate antibodies inflamed tumour-associated macrophages, monocytes and dendritic cells through Fcγ receptors (FcγR), in turn driving anti-tumour CD8+ T cells from an exhausted effector-like state to a more memory-like state. These results reveal a mechanism of action through which TIGIT checkpoint inhibitors can remodel immunosuppressive tumour microenvironments, and suggest that FcγR engagement is an important consideration in anti-TIGIT antibody development.


Subject(s)
Antibodies, Monoclonal , Antineoplastic Agents , B7-H1 Antigen , Myeloid Cells , Neoplasms , Receptors, Immunologic , T-Lymphocytes, Regulatory , Animals , Humans , Mice , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Drug Therapy, Combination , Immune Checkpoint Inhibitors/immunology , Immune Checkpoint Inhibitors/therapeutic use , Macrophage Activation , Myeloid Cells/immunology , Neoplasms/drug therapy , Neoplasms/immunology , Receptors, IgG/immunology , Receptors, Immunologic/immunology , T-Lymphocytes, Regulatory/immunology , Tumor Microenvironment/immunology , Tumor-Associated Macrophages/immunology
7.
Nature ; 625(7993): 166-174, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38057662

ABSTRACT

Myeloid cells are known to suppress antitumour immunity1. However, the molecular drivers of immunosuppressive myeloid cell states are not well defined. Here we used single-cell RNA sequencing of human and mouse non-small cell lung cancer (NSCLC) lesions, and found that in both species the type 2 cytokine interleukin-4 (IL-4) was predicted to be the primary driver of the tumour-infiltrating monocyte-derived macrophage phenotype. Using a panel of conditional knockout mice, we found that only deletion of the IL-4 receptor IL-4Rα in early myeloid progenitors in bone marrow reduced tumour burden, whereas deletion of IL-4Rα in downstream mature myeloid cells had no effect. Mechanistically, IL-4 derived from bone marrow basophils and eosinophils acted on granulocyte-monocyte progenitors to transcriptionally programme the development of immunosuppressive tumour-promoting myeloid cells. Consequentially, depletion of basophils profoundly reduced tumour burden and normalized myelopoiesis. We subsequently initiated a clinical trial of the IL-4Rα blocking antibody dupilumab2-5 given in conjunction with PD-1/PD-L1 checkpoint blockade in patients with relapsed or refractory NSCLC who had progressed on PD-1/PD-L1 blockade alone (ClinicalTrials.gov identifier NCT05013450 ). Dupilumab supplementation reduced circulating monocytes, expanded tumour-infiltrating CD8 T cells, and in one out of six patients, drove a near-complete clinical response two months after treatment. Our study defines a central role for IL-4 in controlling immunosuppressive myelopoiesis in cancer, identifies a novel combination therapy for immune checkpoint blockade in humans, and highlights cancer as a systemic malady that requires therapeutic strategies beyond the primary disease site.


Subject(s)
Bone Marrow , Carcinogenesis , Interleukin-4 , Myelopoiesis , Signal Transduction , Animals , Humans , Mice , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , Bone Marrow/drug effects , Bone Marrow/metabolism , Carcinogenesis/drug effects , Carcinogenesis/metabolism , Carcinogenesis/pathology , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/therapy , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Immune Checkpoint Inhibitors/immunology , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Interleukin-4/metabolism , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Monocytes/drug effects , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Recurrence , Signal Transduction/drug effects
8.
Immunity ; 52(1): 36-54, 2020 01 14.
Article in English | MEDLINE | ID: mdl-31940272

ABSTRACT

Therapeutics that target the T cell inhibitory checkpoint proteins CTLA-4 and PD(L)1 are efficacious across a broad range of cancers, resulting in reductions in tumor burden and increased long-term survival in subsets of patients. The significant and wide-ranging effects of these immunotherapies have prompted the clinical investigation of additional therapies that modulate anti-tumor immunity through effects on T cells, myeloid cells, and other cell types within the tumor microenvironment. The clinical activity of these newer investigational therapies has been mixed, with some therapeutics showing promise but others not exhibiting appreciable efficacy. In this review, we summarize the results of select recent clinical studies of cancer immunotherapies beyond anti-CTLA-4 and anti-PD(L)1 and discuss how these results are providing new insights into the regulation of human anti-tumor immunity.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Immunotherapy/methods , Neoplasms/therapy , T-Lymphocytes/immunology , B7-H1 Antigen/antagonists & inhibitors , CTLA-4 Antigen/antagonists & inhibitors , Humans , Lymphocyte Activation/immunology , Neoplasms/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Tumor Microenvironment/immunology
9.
Immunity ; 52(1): 55-81, 2020 01 14.
Article in English | MEDLINE | ID: mdl-31940273

ABSTRACT

Cancer is a complex disease whose outcome depends largely on the cross-talk between the tumor and its microenvironment. Here, we review the evolution of the field of tumor immunology and the advances, in lockstep, of our understanding of cancer as a disease. We discuss the involvement of different immune cells at distinct stages of tumor progression and how immune contexture determinants shaping tumor development are being exploited therapeutically. Current clinical stratification schemes focus on the tumor histopathology and the molecular characteristics of the tumor cell. We argue for the importance of revising these stratification systems to include immune parameters so as to address the immediate need for improved prognostic and/or predictive information to guide clinical decisions.


Subject(s)
Immunotherapy/methods , Neoplasms/immunology , Neoplasms/therapy , B7-H1 Antigen/antagonists & inhibitors , CTLA-4 Antigen/antagonists & inhibitors , Humans , Neoplasms/pathology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , T-Lymphocytes/immunology , Tumor Microenvironment/immunology
10.
Nature ; 611(7935): 358-364, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36323784

ABSTRACT

The accumulation of senescent cells is a major cause of age-related inflammation and predisposes to a variety of age-related diseases1. However, little is known about the molecular basis underlying this accumulation and its potential as a target to ameliorate the ageing process. Here we show that senescent cells heterogeneously express the immune checkpoint protein programmed death-ligand 1 (PD-L1) and that PD-L1+ senescent cells accumulate with age in vivo. PD-L1- cells are sensitive to T cell surveillance, whereas PD-L1+ cells are resistant, even in the presence of senescence-associated secretory phenotypes (SASP). Single-cell analysis of p16+ cells in vivo revealed that PD-L1 expression correlated with higher levels of SASP. Consistent with this, administration of programmed cell death protein 1 (PD-1) antibody to naturally ageing mice or a mouse model with normal livers or induced nonalcoholic steatohepatitis reduces the total number of p16+ cells in vivo as well as the PD-L1+ population in an activated CD8+ T cell-dependent manner, ameliorating various ageing-related phenotypes. These results suggest that the heterogeneous expression of PD-L1 has an important role in the accumulation of senescent cells and inflammation associated with ageing, and the elimination of PD-L1+ senescent cells by immune checkpoint blockade may be a promising strategy for anti-ageing therapy.


Subject(s)
Aging , B7-H1 Antigen , Phenotype , Programmed Cell Death 1 Receptor , Animals , Mice , Aging/immunology , Aging/metabolism , Aging/pathology , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Inflammation/pathology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Single-Cell Analysis , Non-alcoholic Fatty Liver Disease , Liver , Rejuvenation
11.
Nature ; 610(7930): 161-172, 2022 10.
Article in English | MEDLINE | ID: mdl-36171284

ABSTRACT

Expansion and differentiation of antigen-experienced PD-1+TCF-1+ stem-like CD8+ T cells into effector cells is critical for the success of immunotherapies based on PD-1 blockade1-4. Hashimoto et al. have shown that, in chronic infections, administration of the cytokine interleukin (IL)-2 triggers an alternative differentiation path of stem-like T cells towards a distinct population of 'better effector' CD8+ T cells similar to those generated in an acute infection5. IL-2 binding to the IL-2 receptor α-chain (CD25) was essential in triggering this alternative differentiation path and expanding better effectors with distinct transcriptional and epigenetic profiles. However, constitutive expression of CD25 on regulatory T cells and some endothelial cells also contributes to unwanted systemic effects from IL-2 therapy. Therefore, engineered IL-2 receptor ß- and γ-chain (IL-2Rßγ)-biased agonists are currently being developed6-10. Here we show that IL-2Rßγ-biased agonists are unable to preferentially expand better effector T cells in cancer models and describe PD1-IL2v, a new immunocytokine that overcomes the need for CD25 binding by docking in cis to PD-1. Cis binding of PD1-IL2v to PD-1 and IL-2Rßγ on the same cell recovers the ability to differentiate stem-like CD8+ T cells into better effectors in the absence of CD25 binding in both chronic infection and cancer models and provides superior efficacy. By contrast, PD-1- or PD-L1-blocking antibodies alone, or their combination with clinically relevant doses of non-PD-1-targeted IL2v, cannot expand this unique subset of better effector T cells and instead lead to the accumulation of terminally differentiated, exhausted T cells. These findings provide the basis for the development of a new generation of PD-1 cis-targeted IL-2R agonists with enhanced therapeutic potential for the treatment of cancer and chronic infections.


Subject(s)
CD8-Positive T-Lymphocytes , Programmed Cell Death 1 Receptor , Receptors, Interleukin-2 , Antibodies, Blocking/immunology , Antibodies, Blocking/pharmacology , Antibodies, Blocking/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Infections/drug therapy , Infections/immunology , Interleukin-2/immunology , Interleukin-2/pharmacology , Interleukin-2/therapeutic use , Interleukin-2 Receptor alpha Subunit/agonists , Neoplasms/drug therapy , Neoplasms/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Receptors, Interleukin-2/agonists
12.
Mol Cell ; 80(4): 592-606.e8, 2020 11 19.
Article in English | MEDLINE | ID: mdl-33159855

ABSTRACT

Despite its outstanding clinical success, immune checkpoint blockade remains ineffective in many patients. Accordingly, combination therapy capable of achieving greater antitumor immunity is urgently required. Here, we report that limiting glutamine metabolism in cancer cells bolsters the effectiveness of anti-programmed death ligand-1 (PD-L1) antibody. Inhibition of glutamine utilization increased PD-L1 levels in cancer cells, thereby inactivating co-cultured T cells. Under glutamine-limited conditions, reduced cellular GSH levels caused an upregulation of PD-L1 expression by impairing SERCA activity, which activates the calcium/NF-κB signaling cascade. Consequently, in tumors grown in immunocompetent mice, inhibition of glutamine metabolism decreased the antitumor activity of T cells. In combination with anti-PD-L1, however, glutamine depletion strongly promoted the antitumor efficacy of T cells in vitro and in vivo due to simultaneous increases in Fas/CD95 levels. Our results demonstrate the relevance of cancer glutamine metabolism to antitumor immunity and suggest that co-targeting of glutamine metabolism and PD-L1 represents a promising therapeutic approach.


Subject(s)
Antibodies, Monoclonal/pharmacology , B7-H1 Antigen/metabolism , Glutamine/metabolism , Glutathione/metabolism , Neoplasms/immunology , Neoplasms/prevention & control , T-Lymphocytes/immunology , Aged , Animals , Apoptosis , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , Cell Proliferation , Female , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Neoplasms/metabolism , Neoplasms/pathology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
13.
Immunity ; 48(3): 434-452, 2018 03 20.
Article in English | MEDLINE | ID: mdl-29562194

ABSTRACT

Expression of programmed death-ligand 1 (PD-L1) is frequently observed in human cancers. Binding of PD-L1 to its receptor PD-1 on activated T cells inhibits anti-tumor immunity by counteracting T cell-activating signals. Antibody-based PD-1-PD-L1 inhibitors can induce durable tumor remissions in patients with diverse advanced cancers, and thus expression of PD-L1 on tumor cells and other cells in the tumor microenviroment is of major clinical relevance. Here we review the roles of the PD-1-PD-L1 axis in cancer, focusing on recent findings on the mechanisms that regulate PD-L1 expression at the transcriptional, posttranscriptional, and protein level. We place this knowledge in the context of observations in the clinic and discuss how it may inform the design of more precise and effective cancer immune checkpoint therapies.


Subject(s)
B7-H1 Antigen/physiology , Gene Expression Regulation , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , B7-H1 Antigen/antagonists & inhibitors , Humans , Immunotherapy , Lymphocyte Activation/drug effects , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Molecular Targeted Therapy , Neoplasms/etiology , Neoplasms/metabolism , Neoplasms/therapy , Programmed Cell Death 1 Receptor/metabolism , T-Lymphocytes/drug effects
14.
Immunity ; 49(6): 1132-1147.e7, 2018 12 18.
Article in English | MEDLINE | ID: mdl-30552022

ABSTRACT

Serrated adenocarcinoma, an alternative pathway for colorectal cancer (CRC) development, accounts for 15%-30% of all CRCs and is aggressive and treatment resistant. We show that the expression of atypical protein kinase C ζ (PKCζ) and PKCλ/ι was reduced in human serrated tumors. Simultaneous inactivation of the encoding genes in the mouse intestinal epithelium resulted in spontaneous serrated tumorigenesis that progressed to advanced cancer with a strongly reactive and immunosuppressive stroma. Whereas epithelial PKCλ/ι deficiency led to immunogenic cell death and the infiltration of CD8+ T cells, which repressed tumor initiation, PKCζ loss impaired interferon and CD8+ T cell responses, which resulted in tumorigenesis. Combined treatment with a TGF-ß receptor inhibitor plus anti-PD-L1 checkpoint blockade showed synergistic curative activity. Analysis of human samples supported the relevance of these kinases in the immunosurveillance defects of human serrated CRC. These findings provide insight into avenues for the detection and treatment of this poor-prognosis subtype of CRC.


Subject(s)
Intestinal Mucosa/immunology , Intestinal Neoplasms/immunology , Isoenzymes/immunology , Protein Kinase C/immunology , Adult , Aged , Aged, 80 and over , Animals , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , Female , Humans , Immunologic Surveillance/genetics , Immunologic Surveillance/immunology , Intestinal Mucosa/enzymology , Intestinal Mucosa/pathology , Intestinal Neoplasms/enzymology , Intestinal Neoplasms/genetics , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Mice, Knockout , Mice, Transgenic , Middle Aged , Protein Kinase C/genetics , Protein Kinase C/metabolism , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism
15.
Nature ; 598(7882): 662-666, 2021 10.
Article in English | MEDLINE | ID: mdl-34616044

ABSTRACT

The availability of L-arginine in tumours is a key determinant of an efficient anti-tumour T cell response1-4. Consequently, increases of typically low L-arginine concentrations within the tumour may greatly potentiate the anti-tumour responses of immune checkpoint inhibitors, such as programmed death-ligand 1 (PD-L1)-blocking antibodies5. However, currently no means are available to locally increase intratumoural L-arginine levels. Here we used a synthetic biology approach to develop an engineered probiotic Escherichia coli Nissle 1917 strain that colonizes tumours and continuously converts ammonia, a metabolic waste product that accumulates in tumours6, to L-arginine. Colonization of tumours with these bacteria increased intratumoural L-arginine concentrations, increased the number of tumour-infiltrating T cells and had marked synergistic effects with PD-L1 blocking antibodies in the clearance of tumours. The anti-tumour effect of these bacteria was mediated by L-arginine and was dependent on T cells. These results show that engineered microbial therapies enable metabolic modulation of the tumour microenvironment leading to enhanced efficacy of immunotherapies.


Subject(s)
Immunotherapy/methods , Metabolic Engineering , Microorganisms, Genetically-Modified , Neoplasms, Experimental/therapy , Adoptive Transfer , Animals , Arginine/metabolism , B7-H1 Antigen/antagonists & inhibitors , Cell Line, Tumor , Escherichia coli , Female , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/microbiology , Probiotics , Proteome , Synthetic Biology , T-Lymphocytes/immunology , Tumor Microenvironment/immunology
16.
Mol Cell ; 76(3): 359-370, 2019 11 07.
Article in English | MEDLINE | ID: mdl-31668929

ABSTRACT

The engagement of programmed cell death protein 1 (PD-1; encoded by the PDCD1 gene) receptor expressed on activated T cells and its ligand, programmed death-ligand 1 (PD-L1; encoded by the CD274 gene), is a major co-inhibitory checkpoint signaling that controls T cell activities. Various types of cancers express high levels of PD-L1 and exploit PD-L1/PD-1 signaling to evade T cell immunity. Blocking the PD-L1/PD-1 pathway has consistently shown remarkable anti-tumor effects in patients with advanced cancers and is recognized as the gold standard for developing new immune checkpoint blockade (ICB) and combination therapies. However, the response rates of anti-PD-L1 have been limited in several solid tumors. Therefore, furthering our understanding of the regulatory mechanisms of PD-L1 can bring substantial benefits to patients with cancer by improving the efficacy of current PD-L1/PD-1 blockade or other ICBs. In this review, we provide current knowledge of PD-L1 regulatory mechanisms at the transcriptional, posttranscriptional, post-translational, and extracellular levels, and discuss the implications of these findings in cancer diagnosis and immunotherapy.


Subject(s)
B7-H1 Antigen/metabolism , Biomarkers, Tumor/metabolism , Neoplasms/metabolism , Animals , Antineoplastic Agents, Immunological/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/genetics , Biomarkers, Tumor/immunology , Gene Expression Regulation, Neoplastic , Humans , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/immunology , Protein Processing, Post-Translational , RNA Processing, Post-Transcriptional , Signal Transduction , Transcription, Genetic , Tumor Escape
17.
Mol Cell ; 73(1): 22-35.e6, 2019 01 03.
Article in English | MEDLINE | ID: mdl-30527665

ABSTRACT

Aberrant expression of programmed death ligand-1 (PD-L1) in tumor cells promotes cancer progression by suppressing cancer immunity. The retinoblastoma protein RB is a tumor suppressor known to regulate the cell cycle, DNA damage response, and differentiation. Here, we demonstrate that RB interacts with nuclear factor κB (NF-κB) protein p65 and that their interaction is primarily dependent on CDK4/6-mediated serine-249/threonine-252 (S249/T252) phosphorylation of RB. RNA-seq analysis shows a subset of NF-κB pathway genes including PD-L1 are selectively upregulated by RB knockdown or CDK4/6 inhibitor. S249/T252-phosphorylated RB inversely correlates with PD-L1 expression in patient samples. Expression of a RB-derived S249/T252 phosphorylation-mimetic peptide suppresses radiotherapy-induced upregulation of PD-L1 and augments therapeutic efficacy of radiation in vivo. Our findings reveal a previously unrecognized tumor suppressor function of hyperphosphorylated RB in suppressing NF-κB activity and PD-L1 expression and suggest that the RB-NF-κB axis can be exploited to overcome cancer immune evasion triggered by conventional or targeted therapies.


Subject(s)
B7-H1 Antigen/metabolism , Prostatic Neoplasms/metabolism , Retinoblastoma Protein/metabolism , Transcription Factor RelA/metabolism , Tumor Escape , Animals , Antineoplastic Agents, Immunological/pharmacology , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , Chemoradiotherapy/methods , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/metabolism , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Male , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , PC-3 Cells , Phosphorylation , Prostatic Neoplasms/genetics , Prostatic Neoplasms/immunology , Prostatic Neoplasms/therapy , Protein Binding , Protein Interaction Domains and Motifs , Radiation Tolerance , Retinoblastoma Protein/genetics , Retinoblastoma Protein/immunology , Signal Transduction , Transcription Factor RelA/genetics , Transcription Factor RelA/immunology , Xenograft Model Antitumor Assays
18.
Mol Cell ; 74(6): 1215-1226.e4, 2019 06 20.
Article in English | MEDLINE | ID: mdl-31053471

ABSTRACT

Programmed death ligand 1 (PD-L1, also called B7-H1) is an immune checkpoint protein that inhibits immune function through its binding of the programmed cell death protein 1 (PD-1) receptor. Clinically approved antibodies block extracellular PD-1 and PD-L1 binding, yet the role of intracellular PD-L1 in cancer remains poorly understood. Here, we discovered that intracellular PD-L1 acts as an RNA binding protein that regulates the mRNA stability of NBS1, BRCA1, and other DNA damage-related genes. Through competition with the RNA exosome, intracellular PD-L1 protects targeted RNAs from degradation, thereby increasing cellular resistance to DNA damage. RNA immunoprecipitation and RNA-seq experiments demonstrated that PD-L1 regulates RNA stability genome-wide. Furthermore, we developed a PD-L1 antibody, H1A, which abrogates the interaction of PD-L1 with CMTM6, thereby promoting PD-L1 degradation. Intracellular PD-L1 may be a potential therapeutic target to enhance the efficacy of radiotherapy and chemotherapy in cancer through the inhibition of DNA damage response and repair.


Subject(s)
B7-H1 Antigen/genetics , DNA Repair , DNA, Neoplasm/genetics , Exosome Multienzyme Ribonuclease Complex/genetics , Gene Expression Regulation, Neoplastic , Programmed Cell Death 1 Receptor/genetics , Animals , Antineoplastic Agents/pharmacology , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cisplatin/pharmacology , DNA Damage , DNA, Neoplasm/metabolism , Exosome Multienzyme Ribonuclease Complex/metabolism , Gamma Rays/therapeutic use , HCT116 Cells , HeLa Cells , Humans , MARVEL Domain-Containing Proteins , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Myelin Proteins , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Proteolysis/drug effects , Proteolysis/radiation effects , RNA Stability/drug effects , RNA Stability/radiation effects , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Xenograft Model Antitumor Assays
19.
N Engl J Med ; 389(10): 911-921, 2023 Sep 07.
Article in English | MEDLINE | ID: mdl-37672694

ABSTRACT

BACKGROUND: Alveolar soft part sarcoma (ASPS) is a rare soft-tissue sarcoma with a poor prognosis and no established therapy. Recently, encouraging responses to immune checkpoint inhibitors have been reported. METHODS: We conducted an investigator-initiated, multicenter, single-group, phase 2 study of the anti-programmed death ligand 1 (PD-L1) agent atezolizumab in adult and pediatric patients with advanced ASPS. Atezolizumab was administered intravenously at a dose of 1200 mg (in patients ≥18 years of age) or 15 mg per kilogram of body weight with a 1200-mg cap (in patients <18 years of age) once every 21 days. Study end points included objective response, duration of response, and progression-free survival according to Response Evaluation Criteria in Solid Tumors (RECIST), version 1.1, as well as pharmacodynamic biomarkers of multistep drug action. RESULTS: A total of 52 patients were evaluated. An objective response was observed in 19 of 52 patients (37%), with 1 complete response and 18 partial responses. The median time to response was 3.6 months (range, 2.1 to 19.1), the median duration of response was 24.7 months (range, 4.1 to 55.8), and the median progression-free survival was 20.8 months. Seven patients took a treatment break after 2 years of treatment, and their responses were maintained through the data-cutoff date. No treatment-related grade 4 or 5 adverse events were recorded. Responses were noted despite variable baseline expression of programmed death 1 and PD-L1. CONCLUSIONS: Atezolizumab was effective at inducing sustained responses in approximately one third of patients with advanced ASPS. (Funded by the National Cancer Institute and others; ClinicalTrials.gov number, NCT03141684.).


Subject(s)
Antibodies, Monoclonal, Humanized , B7-H1 Antigen , Sarcoma, Alveolar Soft Part , Adolescent , Adult , Child , Humans , Infant, Newborn , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , Body Weight , Sarcoma, Alveolar Soft Part/drug therapy , Administration, Intravenous
20.
Nature ; 577(7791): 561-565, 2020 01.
Article in English | MEDLINE | ID: mdl-31942071

ABSTRACT

Checkpoint blockade therapies that reactivate tumour-associated T cells can induce durable tumour control and result in the long-term survival of patients with advanced cancers1. Current predictive biomarkers for therapy response include high levels of intratumour immunological activity, a high tumour mutational burden and specific characteristics of the gut microbiota2,3. Although the role of T cells in antitumour responses has thoroughly been studied, other immune cells remain insufficiently explored. Here we use clinical samples of metastatic melanomas to investigate the role of B cells in antitumour responses, and find that the co-occurrence of tumour-associated CD8+ T cells and CD20+ B cells is associated with improved survival, independently of other clinical variables. Immunofluorescence staining of CXCR5 and CXCL13 in combination with CD20 reveals the formation of tertiary lymphoid structures in these CD8+CD20+ tumours. We derived a gene signature associated with tertiary lymphoid structures, which predicted clinical outcomes in cohorts of patients treated with immune checkpoint blockade. Furthermore, B-cell-rich tumours were accompanied by increased levels of TCF7+ naive and/or memory T cells. This was corroborated by digital spatial-profiling data, in which T cells in tumours without tertiary lymphoid structures had a dysfunctional molecular phenotype. Our results indicate that tertiary lymphoid structures have a key role in the immune microenvironment in melanoma, by conferring distinct T cell phenotypes. Therapeutic strategies to induce the formation of tertiary lymphoid structures should be explored to improve responses to cancer immunotherapy.


Subject(s)
Melanoma/immunology , Melanoma/therapy , Tertiary Lymphoid Structures/immunology , Antigens, CD20/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B7-H1 Antigen/antagonists & inhibitors , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Chemokine CXCL13/metabolism , Humans , Immunologic Memory/immunology , Melanoma/genetics , Melanoma/pathology , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Phenotype , Prognosis , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Proteomics , RNA-Seq , Receptors, CXCR5/metabolism , Single-Cell Analysis , Survival Rate , T Cell Transcription Factor 1/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tertiary Lymphoid Structures/genetics , Treatment Outcome , Tumor Microenvironment/immunology
SELECTION OF CITATIONS
SEARCH DETAIL