Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Nat Immunol ; 21(8): 914-926, 2020 08.
Article in English | MEDLINE | ID: mdl-32424363

ABSTRACT

Adoptive cell therapies using genetically engineered T cell receptor or chimeric antigen receptor T cells are emerging forms of immunotherapy that redirect T cells to specifically target cancer. However, tumor antigen heterogeneity remains a key challenge limiting their efficacy against solid cancers. Here, we engineered T cells to secrete the dendritic cell (DC) growth factor Fms-like tyrosine kinase 3 ligand (Flt3L). Flt3L-secreting T cells expanded intratumoral conventional type 1 DCs and substantially increased host DC and T cell activation when combined with immune agonists poly (I:C) and anti-4-1BB. Importantly, combination therapy led to enhanced inhibition of tumor growth and the induction of epitope spreading towards antigens beyond those recognized by adoptively transferred T cells in solid tumor models of T cell receptor and chimeric antigen receptor T cell therapy. Our data suggest that augmenting endogenous DCs is a promising strategy to overcome the clinical problem of antigen-negative tumor escape following adoptive cell therapy.


Subject(s)
Dendritic Cells/immunology , Immunotherapy, Adoptive , Membrane Proteins/immunology , Neoplasms, Experimental/immunology , T-Lymphocytes/immunology , Animals , Antigens, Neoplasm/immunology , Humans , Immunologic Factors , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Antigen, T-Cell/immunology , Receptors, Chimeric Antigen/immunology
2.
Nature ; 629(8010): 201-210, 2024 May.
Article in English | MEDLINE | ID: mdl-38600376

ABSTRACT

Chimeric antigen receptor (CAR) T cell therapy has transformed the treatment of haematological malignancies such as acute lymphoblastic leukaemia, B cell lymphoma and multiple myeloma1-4, but the efficacy of CAR T cell therapy in solid tumours has been limited5. This is owing to a number of factors, including the immunosuppressive tumour microenvironment that gives rise to poorly persisting and metabolically dysfunctional T cells. Analysis of anti-CD19 CAR T cells used clinically has shown that positive treatment outcomes are associated with a more 'stem-like' phenotype and increased mitochondrial mass6-8. We therefore sought to identify transcription factors that could enhance CAR T cell fitness and efficacy against solid tumours. Here we show that overexpression of FOXO1 promotes a stem-like phenotype in CAR T cells derived from either healthy human donors or patients, which correlates with improved mitochondrial fitness, persistence and therapeutic efficacy in vivo. This work thus reveals an engineering approach to genetically enforce a favourable metabolic phenotype that has high translational potential to improve the efficacy of CAR T cells against solid tumours.


Subject(s)
Forkhead Box Protein O1 , Immunotherapy, Adoptive , Neoplasms , Receptors, Chimeric Antigen , Stem Cells , T-Lymphocytes , Humans , Mice , Cell Line, Tumor , Forkhead Box Protein O1/metabolism , Forkhead Box Protein O1/genetics , Mitochondria/metabolism , Phenotype , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/cytology , Tumor Microenvironment/immunology , Stem Cells/cytology , Stem Cells/immunology , Stem Cells/metabolism , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy
3.
Mol Ther ; 28(11): 2379-2393, 2020 11 04.
Article in English | MEDLINE | ID: mdl-32735774

ABSTRACT

Chimeric antigen receptor (CAR) T cell therapy has been highly successful in hematological malignancies leading to their US Food and Drug Administration (FDA) approval. However, the efficacy of CAR T cells in solid tumors is limited by tumor-induced immunosuppression, leading to the development of combination approaches, such as adjuvant programmed cell death 1 (PD-1) blockade. Current FDA-approved methods for generating CAR T cells utilize either anti-CD3 and interleukin (IL)-2 or anti-CD3/CD28 beads, which can generate a T cell product with an effector/exhausted phenotype. Whereas different cytokine preconditioning milieu, such as IL-7/IL-15, have been shown to promote T cell engraftment, the impact of this approach on CAR T cell responses to adjuvant immune-checkpoint blockade has not been assessed. In the current study, we reveal that the preconditioning of CAR T cells with IL-7/IL-15 increased CAR T cell responses to anti-PD-1 adjuvant therapy. This was associated with the emergence of an intratumoral CD8+CD62L+TCF7+IRF4- population that was highly responsive to anti-PD-1 therapy and mediated the vast majority of transcriptional and epigenetic changes in vivo following PD-1 blockade. Our data indicate that preservation of CAR T cells in a TCF7+ phenotype is crucial for their responsiveness to adjuvant immunotherapy approaches and should be a key consideration when designing clinical protocols.


Subject(s)
Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy, Adoptive , Interleukin-15/administration & dosage , Neoplasms/therapy , Biomarkers, Tumor , Combined Modality Therapy , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immune Checkpoint Proteins/metabolism , Immunotherapy, Adoptive/methods , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Neoplasms/etiology , Treatment Outcome
4.
Cell Rep ; 42(8): 113014, 2023 08 29.
Article in English | MEDLINE | ID: mdl-37605534

ABSTRACT

CXCL9 expression is a strong predictor of response to immune checkpoint blockade therapy. Accordingly, we sought to develop therapeutic strategies to enhance the expression of CXCL9 and augment antitumor immunity. To perform whole-genome CRISPR-Cas9 screening for regulators of CXCL9 expression, a CXCL9-GFP reporter line is generated using a CRISPR knockin strategy. This approach finds that IRF1 limits CXCL9 expression in both tumor cells and primary myeloid cells through induction of SOCS1, which subsequently limits STAT1 signaling. Thus, we identify a subset of STAT1-dependent genes that do not require IRF1 for their transcription, including CXCL9. Targeting of either IRF1 or SOCS1 potently enhances CXCL9 expression by intratumoral macrophages, which is further enhanced in the context of immune checkpoint blockade therapy. We hence show a non-canonical role for IRF1 in limiting the expression of a subset of STAT1-dependent genes through induction of SOCS1.


Subject(s)
CRISPR-Cas Systems , Immune Checkpoint Inhibitors , Feedback , Suppressor of Cytokine Signaling Proteins/genetics , Signal Transduction
5.
Nat Commun ; 14(1): 6990, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37914685

ABSTRACT

There is significant clinical interest in targeting adenosine-mediated immunosuppression, with several small molecule inhibitors having been developed for targeting the A2AR receptor. Understanding of the mechanism by which A2AR is regulated has been hindered by difficulty in identifying the cell types that express A2AR due to a lack of robust antibodies for these receptors. To overcome this limitation, here an A2AR eGFP reporter mouse is developed, enabling the expression of A2AR during ongoing anti-tumor immune responses to be assessed. This reveals that A2AR is highly expressed on all tumor-infiltrating lymphocyte subsets including Natural Killer (NK) cells, NKT cells, γδ T cells, conventional CD4+ and CD8+ T lymphocytes and on a MHCIIhiCD86hi subset of type 2 conventional dendritic cells. In response to PD-L1 blockade, the emergence of PD-1+A2AR- cells correlates with successful therapeutic responses, whilst IL-18 is identified as a cytokine that potently upregulates A2AR and synergizes with A2AR deficiency to improve anti-tumor immunity. These studies provide insight into the biology of A2AR in the context of anti-tumor immunity and reveals potential combination immunotherapy approaches.


Subject(s)
Neoplasms , Animals , Mice , Cytokines/metabolism , Immunity , Immunotherapy , Lymphocytes, Tumor-Infiltrating , Neoplasms/genetics , Neoplasms/metabolism , Tumor Microenvironment
7.
Sci Rep ; 12(1): 4034, 2022 03 08.
Article in English | MEDLINE | ID: mdl-35260653

ABSTRACT

Natural Killer T (NKT) cells and Mucosal-Associated Invariant T (MAIT) cells are innate-like T cells that express semi-invariant αß T cell receptors (TCRs) through which they recognise CD1d and MR1 molecules, respectively, in complex with specific ligands. These cells play important roles in health and disease in many organs, but their precise intra-organ location is not well established. Here, using CD1d and MR1 tetramer staining techniques, we describe the precise location of NKT and MAIT cells in lymphoid and peripheral organs. Within the thymus, NKT cells were concentrated in the medullary side of the corticomedullary junction. In spleen and lymph nodes, NKT cells were mainly localised within T cell zones, although following in vivo activation with the potent NKT-cell ligand α-GalCer, they expanded throughout the spleen. MAIT cells were clearly detectable in Vα19 TCR transgenic mice and were rare but detectable in lymphoid tissue of non-transgenic mice. In contrast to NKT cells, MAIT cells were more closely associated with the B cell zone and red pulp of the spleen. Accordingly, we have provided an extensive analysis of the in situ localisation of NKT and MAIT cells and suggest differences between the intra-organ location of these two cell types.


Subject(s)
Lymphoid Tissue , Mucosal-Associated Invariant T Cells , Natural Killer T-Cells , Animals , Lymphoid Tissue/metabolism , Mice , Mice, Transgenic , Mucosal-Associated Invariant T Cells/metabolism , Natural Killer T-Cells/metabolism , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism
8.
Nat Commun ; 12(1): 3236, 2021 05 28.
Article in English | MEDLINE | ID: mdl-34050151

ABSTRACT

Adenosine is an immunosuppressive factor that limits anti-tumor immunity through the suppression of multiple immune subsets including T cells via activation of the adenosine A2A receptor (A2AR). Using both murine and human chimeric antigen receptor (CAR) T cells, here we show that targeting A2AR with a clinically relevant CRISPR/Cas9 strategy significantly enhances their in vivo efficacy, leading to improved survival of mice. Effects evoked by CRISPR/Cas9 mediated gene deletion of A2AR are superior to shRNA mediated knockdown or pharmacological blockade of A2AR. Mechanistically, human A2AR-edited CAR T cells are significantly resistant to adenosine-mediated transcriptional changes, resulting in enhanced production of cytokines including IFNγ and TNF, and increased expression of JAK-STAT signaling pathway associated genes. A2AR deficient CAR T cells are well tolerated and do not induce overt pathologies in mice, supporting the use of CRISPR/Cas9 to target A2AR for the improvement of CAR T cell function in the clinic.


Subject(s)
Immunotherapy, Adoptive/methods , Neoplasms/therapy , Receptor, Adenosine A2A/genetics , T-Lymphocytes/transplantation , Adenosine/metabolism , Adenosine A2 Receptor Antagonists/pharmacology , Animals , CRISPR-Cas Systems/genetics , Cell Engineering/methods , Cell Line, Tumor/transplantation , Disease Models, Animal , Female , Gene Editing , Gene Expression Regulation, Neoplastic/immunology , Gene Knockdown Techniques , Gene Knockout Techniques , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Mice, Transgenic , Neoplasms/genetics , Neoplasms/immunology , RNA, Small Interfering/metabolism , RNA-Seq , Receptor, Adenosine A2A/metabolism , Receptor, ErbB-2/genetics , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Escape/drug effects , Tumor Escape/genetics
9.
Nat Commun ; 12(1): 4746, 2021 08 06.
Article in English | MEDLINE | ID: mdl-34362900

ABSTRACT

The function of MR1-restricted mucosal-associated invariant T (MAIT) cells in tumor immunity is unclear. Here we show that MAIT cell-deficient mice have enhanced NK cell-dependent control of metastatic B16F10 tumor growth relative to control mice. Analyses of this interplay in human tumor samples reveal that high expression of a MAIT cell gene signature negatively impacts the prognostic significance of NK cells. Paradoxically, pre-pulsing tumors with MAIT cell antigens, or activating MAIT cells in vivo, enhances anti-tumor immunity in B16F10 and E0771 mouse tumor models, including in the context of established metastasis. These effects are associated with enhanced NK cell responses and increased expression of both IFN-γ-dependent and inflammatory genes in NK cells. Importantly, activated human MAIT cells also promote the function of NK cells isolated from patient tumor samples. Our results thus describe an activation-dependent, MAIT cell-mediated regulation of NK cells, and suggest a potential therapeutic avenue for cancer treatment.


Subject(s)
Immunity, Cellular , Killer Cells, Natural/immunology , Mucosal-Associated Invariant T Cells/immunology , Neoplasms/immunology , Animals , Antineoplastic Agents , Cell Line, Tumor , Cytokines , Histocompatibility Antigens Class I/genetics , Humans , Immunity , Mice , Mice, Inbred C57BL , Mice, Knockout , Minor Histocompatibility Antigens/genetics , Neoplasm Metastasis , Neoplasms/pathology
10.
ChemMedChem ; 15(13): 1128-1138, 2020 07 03.
Article in English | MEDLINE | ID: mdl-32400116

ABSTRACT

Multivalent structures can provide multiple interactions at a target site and improve binding affinity. The multivalent presentation of the anti-tumour heptapeptide, SNTSESF, was investigated. This peptide's activity has been attributed to blockade of the PD-1 receptor-mediated signalling pathway. Two and four peptide units were conjugated to poly ethoxy ethyl glycinamide (PEE-G) scaffolds to prepare high-purity products. These conjugates and the peptide were examined in a mouse model implanted with GL261 tumours that indicated that presenting more than two copies of peptide SNTSESF on the dendritic scaffold does not increase anti-tumour activity per peptide. The fluorescent labelled peptide and most active multivalent peptide conjugate were therefore screened for their interaction with the human PD-L1 protein in a fluorescence polarisation assay. No indication of a specific SNTSESF peptide/PD-L1 interaction was observed. This finding was further supported by a molecular modelling binding study.


Subject(s)
Glycine/analogs & derivatives , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Animals , Cell Line, Tumor , Dose-Response Relationship, Drug , Glycine/chemical synthesis , Glycine/chemistry , Glycine/pharmacology , Humans , Mice , Mice, Inbred C57BL , Molecular Structure , Programmed Cell Death 1 Receptor/metabolism , Structure-Activity Relationship
11.
Clin Cancer Res ; 26(2): 487-504, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31636098

ABSTRACT

PURPOSE: Response rates to immune checkpoint blockade (ICB; anti-PD-1/anti-CTLA-4) correlate with the extent of tumor immune infiltrate, but the mechanisms underlying the recruitment of T cells following therapy are poorly characterized. A greater understanding of these processes may see the development of therapeutic interventions that enhance T-cell recruitment and, consequently, improved patient outcomes. We therefore investigated the chemokines essential for immune cell recruitment and subsequent therapeutic efficacy of these immunotherapies. EXPERIMENTAL DESIGN: The chemokines upregulated by dual PD-1/CTLA-4 blockade were assessed using NanoString-based analysis with results confirmed at the protein level by flow cytometry and cytometric bead array. Blocking/neutralizing antibodies confirmed the requirement for key chemokines/cytokines and immune effector cells. Results were confirmed in patients treated with immune checkpoint inhibitors using single-cell RNA-sequencing (RNA-seq) and paired survival analyses. RESULTS: The CXCR3 ligands, CXCL9 and CXCL10, were significantly upregulated following dual PD-1/CTLA-4 blockade and both CD8+ T-cell infiltration and therapeutic efficacy were CXCR3 dependent. In both murine models and patients undergoing immunotherapy, macrophages were the predominant source of CXCL9 and their depletion abrogated CD8+ T-cell infiltration and the therapeutic efficacy of dual ICB. Single-cell RNA-seq analysis of patient tumor-infiltrating lymphocytes (TIL) revealed that CXCL9/10/11 was predominantly expressed by macrophages following ICB and we identified a distinct macrophage signature that was associated with positive responses to ICB. CONCLUSIONS: These data underline the fundamental importance of macrophage-derived CXCR3 ligands for the therapeutic efficacy of ICB and highlight the potential of manipulating this axis to enhance patient responses.


Subject(s)
CTLA-4 Antigen/antagonists & inhibitors , Chemokine CXCL10/metabolism , Chemokine CXCL9/metabolism , Immunotherapy/methods , Macrophages/immunology , Neoplasms/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Receptors, CXCR3/metabolism , Tumor Microenvironment
13.
FEBS J ; 285(6): 1030-1032, 2018 03.
Article in English | MEDLINE | ID: mdl-29570251

ABSTRACT

Tumor cells can develop a variety of mechanisms to evade and subvert the immune system for their survival. Bland et al., in this edition of The FEBS Journal, make the novel finding that the tumor line B16F0 can deliver mRNA/miRNA loaded exosomes to cytotoxic T lymphocytes and alter their metabolic function and interferon gamma production.


Subject(s)
Exosomes , T-Lymphocytes, Cytotoxic , Humans , Melanoma , MicroRNAs , Transcriptome
14.
Cancer Immunol Res ; 6(9): 1069-1081, 2018 09.
Article in English | MEDLINE | ID: mdl-30018045

ABSTRACT

Immunotherapy is widely accepted as a powerful new treatment modality for the treatment of cancer. The most successful form of immunotherapy to date has been the blockade of the immune checkpoints PD-1 and CTLA-4. Combining inhibitors of both PD-1 and CTLA-4 increases the proportion of patients who respond to immunotherapy. However, most patients still do not respond to checkpoint inhibitors, and prognostic biomarkers are currently lacking. Therefore, a better understanding of the mechanism by which these checkpoint inhibitors enhance antitumor immune responses is required to more accurately predict which patients are likely to respond and further enhance this treatment modality. Our current study of two mouse tumor models revealed that CD4+Foxp3- cells activated by dual PD-1/CTLA-4 blockade modulated the myeloid compartment, including activation of conventional CD103+ dendritic cells (DC) and expansion of a myeloid subset that produces TNFα and iNOS (TIP-DCs). CD4+Foxp3- T cell-mediated activation of CD103+ DCs resulted in enhanced IL12 production by these cells and IL12 enhanced the therapeutic effect of dual PD-1/CTLA-4 blockade. Given the importance of these myeloid subsets in the antitumor immune response, our data point to a previously underappreciated role of CD4+Foxp3- cells in modulating this arm of the antitumor immune response. Cancer Immunol Res; 6(9); 1069-81. ©2018 AACR.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CTLA-4 Antigen/antagonists & inhibitors , Dendritic Cells/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Animals , Antigens, CD/genetics , CD8-Positive T-Lymphocytes/immunology , CTLA-4 Antigen/immunology , Cell Line, Tumor , Hepatocyte Nuclear Factor 3-gamma/genetics , Immunotherapy , Integrin alpha Chains/genetics , Interleukin-12/immunology , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/immunology
SELECTION OF CITATIONS
SEARCH DETAIL