Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Cell Rep ; 43(5): 114141, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38656869

ABSTRACT

The cellular source of positive signals that reinvigorate T cells within the tumor microenvironment (TME) for the therapeutic efficacy of programmed death-1 (PD-1)/programmed death-ligand 1 (PD-L1) blockade has not been clearly defined. We now show that Batf3-lineage dendritic cells (DCs) are essential in this process. Flow cytometric analysis, gene-targeted mice, and blocking antibody studies revealed that 4-1BBL is a major positive co-stimulatory signal provided by these DCs within the TME that translates to CD8+ T cell functional reinvigoration and tumor regression. Immunofluorescence and spatial transcriptomics on human tumor samples revealed clustering of Batf3+ DCs and CD8+ T cells, which correlates with anti-PD-1 efficacy. In addition, proximity to Batf3+ DCs within the TME is associated with CD8+ T cell transcriptional states linked to anti-PD-1 response. Our results demonstrate that Batf3+ DCs within the TME are critical for PD-1/PD-L1 blockade efficacy and indicate a major role for the 4-1BB/4-1BB ligand (4-1BBL) axis during this process.


Subject(s)
B7-H1 Antigen , Basic-Leucine Zipper Transcription Factors , CD8-Positive T-Lymphocytes , Dendritic Cells , Programmed Cell Death 1 Receptor , Repressor Proteins , Tumor Microenvironment , Animals , Humans , Mice , 4-1BB Ligand/metabolism , 4-1BB Ligand/genetics , B7-H1 Antigen/metabolism , Basic-Leucine Zipper Transcription Factors/metabolism , Basic-Leucine Zipper Transcription Factors/genetics , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Dendritic Cells/metabolism , Dendritic Cells/immunology , Immune Checkpoint Inhibitors/pharmacology , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Repressor Proteins/metabolism , Signal Transduction , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism
2.
Nat Biotechnol ; 42(3): 424-436, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37169967

ABSTRACT

Genetically engineered mouse models only capture a small fraction of the genetic lesions that drive human cancer. Current CRISPR-Cas9 models can expand this fraction but are limited by their reliance on error-prone DNA repair. Here we develop a system for in vivo prime editing by encoding a Cre-inducible prime editor in the mouse germline. This model allows rapid, precise engineering of a wide range of mutations in cell lines and organoids derived from primary tissues, including a clinically relevant Kras mutation associated with drug resistance and Trp53 hotspot mutations commonly observed in pancreatic cancer. With this system, we demonstrate somatic prime editing in vivo using lipid nanoparticles, and we model lung and pancreatic cancer through viral delivery of prime editing guide RNAs or orthotopic transplantation of prime-edited organoids. We believe that this approach will accelerate functional studies of cancer-associated mutations and complex genetic combinations that are challenging to construct with traditional models.


Subject(s)
Pancreatic Neoplasms , RNA, Guide, CRISPR-Cas Systems , Mice , Humans , Animals , Mice, Transgenic , Mutation/genetics , Pancreatic Neoplasms/genetics , Cell Line , Gene Editing , CRISPR-Cas Systems/genetics
3.
JCI Insight ; 8(19)2023 10 09.
Article in English | MEDLINE | ID: mdl-37669107

ABSTRACT

Engineered cytokine-based approaches for immunotherapy of cancer are poised to enter the clinic, with IL-12 being at the forefront. However, little is known about potential mechanisms of resistance to cytokine therapies. We found that orthotopic murine lung tumors were resistant to systemically delivered IL-12 fused to murine serum albumin (MSA, IL12-MSA) because of low IL-12 receptor (IL-12R) expression on tumor-reactive CD8+ T cells. IL2-MSA increased binding of IL12-MSA by tumor-reactive CD8+ T cells, and combined administration of IL12-MSA and IL2-MSA led to enhanced tumor-reactive CD8+ T cell effector differentiation, decreased numbers of tumor-infiltrating CD4+ regulatory T cells, and increased survival of lung tumor-bearing mice. Predictably, the combination of IL-2 and IL-12 at therapeutic doses led to significant dose-limiting toxicity. Administering IL-12 and IL-2 analogs with preferential binding to cells expressing Il12rb1 and CD25, respectively, led to a significant extension of survival in mice with lung tumors while abrogating dose-limiting toxicity. These findings suggest that IL-12 and IL-2 represent a rational approach to combination cytokine therapy whose dose-limiting toxicity can be overcome with engineered cytokine variants.


Subject(s)
Interleukin-12 , Lung Neoplasms , Mice , Animals , Interleukin-12/genetics , Interleukin-2/genetics , Immunotherapy , Cytokines , Lung Neoplasms/genetics , Lung Neoplasms/therapy
4.
Immunity ; 56(2): 386-405.e10, 2023 02 14.
Article in English | MEDLINE | ID: mdl-36736322

ABSTRACT

Local environmental factors influence CD8+ T cell priming in lymph nodes (LNs). Here, we sought to understand how factors unique to the tumor-draining mediastinal LN (mLN) impact CD8+ T cell responses toward lung cancer. Type 1 conventional dendritic cells (DC1s) showed a mLN-specific failure to induce robust cytotoxic T cells responses. Using regulatory T (Treg) cell depletion strategies, we found that Treg cells suppressed DC1s in a spatially coordinated manner within tissue-specific microniches within the mLN. Treg cell suppression required MHC II-dependent contact between DC1s and Treg cells. Elevated levels of IFN-γ drove differentiation Treg cells into Th1-like effector Treg cells in the mLN. In patients with cancer, Treg cell Th1 polarization, but not CD8+/Treg cell ratios, correlated with poor responses to checkpoint blockade immunotherapy. Thus, IFN-γ in the mLN skews Treg cells to be Th1-like effector Treg cells, driving their close interaction with DC1s and subsequent suppression of cytotoxic T cell responses.


Subject(s)
Lung Neoplasms , T-Lymphocytes, Regulatory , Humans , CD8-Positive T-Lymphocytes , Interferon-gamma , T-Lymphocytes, Cytotoxic
5.
Immunity ; 56(1): 8-10, 2023 01 10.
Article in English | MEDLINE | ID: mdl-36630920

ABSTRACT

IL-2 remains a promising candidate for immunotherapy of cancer, but its use is hampered by systemic toxicities. In this issue of Immunity, Tichet, Hanahan, and colleagues demonstrate that an IL-2 variant fused to an anti-PD-1 antibody overcomes these limitations to promote impressive tumor control. This approach may be a path to treat tumors that do not respond to anti-PD-1 monotherapy.


Subject(s)
Interleukin-2 , Neoplasms , Programmed Cell Death 1 Receptor , Humans , Immunotherapy , Interleukin-2/therapeutic use , Neoplasms/therapy
6.
Sci Immunol ; 6(64): eabi8800, 2021 Oct 29.
Article in English | MEDLINE | ID: mdl-34714687

ABSTRACT

In non­small cell lung cancer (NSCLC), response to immune checkpoint blockade (ICB) is associated with programmed cell death ligand 1 expression that is induced by interferon-γ­producing, tumor-infiltrating CD8+ T cells. However, not all tumors with a CD8+ T cell infiltrate respond to ICB, and little is known about the mechanisms governing ICB resistance in T cell­infiltrated NSCLC. We used an orthotopic NSCLC mouse model to study ICB-refractory CD8+ T cell responses. Single-cell RNA sequencing of the NSCLC mouse tumors revealed that lung cancer­specific tumor-infiltrating CD8+ T cells exhibited clonal expansion but lacked expression of genes associated with effector and exhausted T cell responses, indicating that they underwent a differentiation program distinct from conventional T cell exhaustion. This lung cancer­specific T cell dysfunction program was established early during priming in the mediastinal lymph node and was characterized by robust proliferation but a failed up-regulation of effector and exhausted T cell characteristics. Intriguingly, CD8+ T cells from patients with NSCLC expressed an analogous gene expression program, which appeared distinct from conventional T cell exhaustion. Administration of recombinant interleukin-2 (IL-2) and IL-12 was sufficient to restore effector T cell differentiation and induce control of KP lung tumors. These findings imply that a CD8+ T cell differentiation trajectory, activated during T cell priming in the mediastinal lymph node, limits the response of CD8+ T cells to ICB and thereby may contribute to failure of ICB in a subset T cell­infiltrated NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/immunology , Immune Checkpoint Inhibitors/immunology , Lung Neoplasms/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Cell Line, Tumor , Humans , Mice
8.
Trends Cancer ; 5(10): 593-603, 2019 10.
Article in English | MEDLINE | ID: mdl-31706507

ABSTRACT

Checkpoint blockade immunotherapy (CBT) has revolutionized cancer treatment; however, the cellular and molecular factors that govern responsiveness to immunotherapy remain poorly understood. One emerging area of clinical importance is differential responsiveness to CBT across different tissue sites of tumor growth. Each tissue site in the body can contain unique tissue-resident immune cells from both the lymphoid and the myeloid compartment and differences in tissue-specific immune cell composition might predispose tumors in certain tissue sites to be more or less responsive to immunotherapy. Understanding the interplay between tissue-resident and systemic immune responses against tumors will help to determine how to better therapeutically target the immune system to fight cancer. This review summarizes clinical and preclinical investigations of tissue-specific antitumor immune responses and how they influence the tumor immune microenvironment and the efficacy of immunotherapy.


Subject(s)
Immunity , Neoplasms/immunology , Neoplasms/therapy , Animals , Biomarkers, Tumor , Dendritic Cells/immunology , Dendritic Cells/metabolism , Energy Metabolism , Humans , Immunity, Innate , Immunotherapy , Molecular Targeted Therapy , Myeloid Cells/immunology , Myeloid Cells/metabolism , Neoplasm Staging , Neoplasms/metabolism , Neoplasms/pathology , Organ Specificity/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Tumor Microenvironment/immunology
10.
Cancer Immunol Res ; 6(1): 14-24, 2018 01.
Article in English | MEDLINE | ID: mdl-29097422

ABSTRACT

Subsets of human tumors are infiltrated with tumor antigen-specific CD8+ T cells [tumor-infiltrating lymphocytes (TILs)] despite tumor progression. These TILs are thought to be inactivated by the immunosuppressive tumor microenvironment, through the engagement of inhibitory receptors such as CTLA-4 and PD-1. However, antigen-specific CD8+ TILs are not functionally inert but are undergoing activation in situ Here, we show that antigen-specific CD8+ TILs are actively proliferating, yet also undergo high rates of apoptosis, leading to a vicious cycle of activation and death that limits immune efficacy. Preventing CD8+ TIL apoptosis by Bcl-xL overexpression enabled accumulation and improved tumor control. Effective combination immunotherapy with an agonist 4-1BB mAb plus either CTLA-4 or PD-L1 neutralization led to a marked accumulation of specific CD8+ TILs through decreased apoptosis rather than increased T-cell entry or proliferation. Our data suggest that antigen-driven apoptosis of CD8+ TILs is a barrier to effective spontaneous antitumor immunity and should be considered as a critical factor in the development of cancer immunotherapies. Cancer Immunol Res; 6(1); 14-24. ©2017 AACR.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Neoplasms/immunology , Neoplasms/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Animals , Antigens, Neoplasm/immunology , Antineoplastic Agents, Immunological/pharmacology , Apoptosis/genetics , Apoptosis/immunology , Biomarkers , Cell Line, Tumor , DNA Damage , Disease Models, Animal , Disease Progression , Gene Expression Profiling , Humans , Lymphocytes, Tumor-Infiltrating/pathology , Melanoma, Experimental , Mice , Mice, Knockout , Molecular Targeted Therapy , Neoplasms/genetics , Neoplasms/pathology , T-Cell Antigen Receptor Specificity , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/antagonists & inhibitors
11.
J Exp Med ; 214(2): 381-400, 2017 02.
Article in English | MEDLINE | ID: mdl-28115575

ABSTRACT

Although the presence of tumor-infiltrating lymphocytes (TILs) indicates an endogenous antitumor response, immune regulatory pathways can subvert the effector phase and enable tumor escape. Negative regulatory pathways include extrinsic suppression mechanisms, but also a T cell-intrinsic dysfunctional state. A more detailed study has been hampered by a lack of cell surface markers defining tumor-specific dysfunctional TILs, and PD-1 alone is not sufficient. Recently, we identified the transcription factor Egr2 as a critical component in controlling the anergic state in vitro. In this study, we show that the Egr2-driven cell surface proteins LAG-3 and 4-1BB can identify dysfunctional tumor antigen-specific CD8+ TIL. Co-expression of 4-1BB and LAG-3 was seen on a majority of CD8+ TILs, but not in lymphoid organs. Functional analysis revealed defective IL-2 and TNF production yet retained expression of IFN-γ and regulatory T cell-recruiting chemokines. Transcriptional and phenotypic characterization revealed coexpression of multiple additional co-stimulatory and co-inhibitory receptors. Administration of anti-LAG-3 plus anti-4-1BB mAbs was therapeutic against tumors in vivo, which correlated with phenotypic normalization. Our results indicate that coexpression of LAG-3 and 4-1BB characterize dysfunctional T cells within tumors, and that targeting these receptors has therapeutic utility.


Subject(s)
Antigens, CD/physiology , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Early Growth Response Protein 2/physiology , Lymphocytes, Tumor-Infiltrating/immunology , Tumor Microenvironment , Tumor Necrosis Factor Receptor Superfamily, Member 9/physiology , Animals , Antibodies, Monoclonal/therapeutic use , Female , Fingolimod Hydrochloride/pharmacology , Interferon-gamma/biosynthesis , Interleukin-2/biosynthesis , Mice , Mice, Inbred C57BL , Tumor Necrosis Factor Receptor Superfamily, Member 9/antagonists & inhibitors , Lymphocyte Activation Gene 3 Protein
12.
Emerg Top Life Sci ; 1(5): 447-456, 2017 Dec 12.
Article in English | MEDLINE | ID: mdl-33525802

ABSTRACT

The recent successes of cancer immunotherapy, first and foremost checkpoint blockade therapy, illustrate the power of the immune system to control cancer. As the number of patients receiving this therapy is increasing, the number of patients being resistant or establishing resistance toward immunotherapy is also increasing. We, therefore, need to further understand the mechanisms mediate resistance in order to prevent or overcome those mechanisms. Increasing evidence is being reported that alterations in tumor cell-intrinsic signaling pathways, including the activation of the WNT/ß-catenin pathway, are associated with blunted T-cell infiltration. Infiltration of tumor by CD8 T cells is one of the most predictive biomarkers for the response toward immunotherapy and therefore the notion that alterations of certain tumor cell-intrinsic signaling pathways might mediate resistance should be considered. Understanding the molecular and immunological mechanisms mediating resistance will ultimately facilitate the development of effective treatment strategies counteracting immune evasion.

SELECTION OF CITATIONS
SEARCH DETAIL
...