Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Cell ; 186(7): 1448-1464.e20, 2023 03 30.
Article in English | MEDLINE | ID: mdl-37001504

ABSTRACT

Neutrophils accumulate in solid tumors, and their abundance correlates with poor prognosis. Neutrophils are not homogeneous, however, and could play different roles in cancer therapy. Here, we investigate the role of neutrophils in immunotherapy, leading to tumor control. We show that successful therapies acutely expanded tumor neutrophil numbers. This expansion could be attributed to a Sellhi state rather than to other neutrophils that accelerate tumor progression. Therapy-elicited neutrophils acquired an interferon gene signature, also seen in human patients, and appeared essential for successful therapy, as loss of the interferon-responsive transcription factor IRF1 in neutrophils led to failure of immunotherapy. The neutrophil response depended on key components of anti-tumor immunity, including BATF3-dependent DCs, IL-12, and IFNγ. In addition, we found that a therapy-elicited systemic neutrophil response positively correlated with disease outcome in lung cancer patients. Thus, we establish a crucial role of a neutrophil state in mediating effective cancer therapy.


Subject(s)
Lung Neoplasms , Neutrophils , Humans , Lung Neoplasms/genetics , Signal Transduction/genetics , Immunotherapy , Interferons
2.
Immunity ; 50(5): 1317-1334.e10, 2019 05 21.
Article in English | MEDLINE | ID: mdl-30979687

ABSTRACT

Tumor-infiltrating myeloid cells (TIMs) comprise monocytes, macrophages, dendritic cells, and neutrophils, and have emerged as key regulators of cancer growth. These cells can diversify into a spectrum of states, which might promote or limit tumor outgrowth but remain poorly understood. Here, we used single-cell RNA sequencing (scRNA-seq) to map TIMs in non-small-cell lung cancer patients. We uncovered 25 TIM states, most of which were reproducibly found across patients. To facilitate translational research of these populations, we also profiled TIMs in mice. In comparing TIMs across species, we identified a near-complete congruence of population structures among dendritic cells and monocytes; conserved neutrophil subsets; and species differences among macrophages. By contrast, myeloid cell population structures in patients' blood showed limited overlap with those of TIMs. This study determines the lung TIM landscape and sets the stage for future investigations into the potential of TIMs as immunotherapy targets.


Subject(s)
Carcinoma, Non-Small-Cell Lung/immunology , Dendritic Cells/immunology , Lung Neoplasms/immunology , Macrophages/immunology , Monocytes/immunology , Neutrophils/immunology , Animals , Base Sequence , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Gene Expression Profiling , Humans , Lung/immunology , Lung/pathology , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Sequence Analysis, RNA
3.
Immunity ; 49(6): 1148-1161.e7, 2018 12 18.
Article in English | MEDLINE | ID: mdl-30552023

ABSTRACT

Anti-PD-1 immune checkpoint blockers can induce sustained clinical responses in cancer but how they function in vivo remains incompletely understood. Here, we combined intravital real-time imaging with single-cell RNA sequencing analysis and mouse models to uncover anti-PD-1 pharmacodynamics directly within tumors. We showed that effective antitumor responses required a subset of tumor-infiltrating dendritic cells (DCs), which produced interleukin 12 (IL-12). These DCs did not bind anti-PD-1 but produced IL-12 upon sensing interferon γ (IFN-γ) that was released from neighboring T cells. In turn, DC-derived IL-12 stimulated antitumor T cell immunity. These findings suggest that full-fledged activation of antitumor T cells by anti-PD-1 is not direct, but rather involves T cell:DC crosstalk and is licensed by IFN-γ and IL-12. Furthermore, we found that activating the non-canonical NF-κB transcription factor pathway amplified IL-12-producing DCs and sensitized tumors to anti-PD-1 treatment, suggesting a therapeutic strategy to improve responses to checkpoint blockade.


Subject(s)
Dendritic Cells/immunology , Interferon-gamma/immunology , Interleukin-12/immunology , Neoplasms/immunology , Programmed Cell Death 1 Receptor/immunology , T-Lymphocytes/immunology , Adult , Aged , Aged, 80 and over , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Dendritic Cells/metabolism , Female , Humans , Immunotherapy/methods , Interferon-gamma/metabolism , Interleukin-12/administration & dosage , Interleukin-12/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , NF-kappa B/immunology , NF-kappa B/metabolism , Neoplasms/metabolism , Neoplasms/therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
5.
Immunity ; 44(2): 343-54, 2016 Feb 16.
Article in English | MEDLINE | ID: mdl-26872698

ABSTRACT

Checkpoint blockade immunotherapies can be extraordinarily effective, but might benefit only the minority of patients whose tumors are pre-infiltrated by T cells. Here, using lung adenocarcinoma mouse models, including genetic models, we show that autochthonous tumors that lacked T cell infiltration and resisted current treatment options could be successfully sensitized to host antitumor T cell immunity when appropriately selected immunogenic drugs (e.g., oxaliplatin combined with cyclophosphamide for treatment against tumors expressing oncogenic Kras and lacking Trp53) were used. The antitumor response was triggered by direct drug actions on tumor cells, relied on innate immune sensing through toll-like receptor 4 signaling, and ultimately depended on CD8(+) T cell antitumor immunity. Furthermore, instigating tumor infiltration by T cells sensitized tumors to checkpoint inhibition and controlled cancer durably. These findings indicate that the proportion of cancers responding to checkpoint therapy can be feasibly and substantially expanded by combining checkpoint blockade with immunogenic drugs.


Subject(s)
Adenocarcinoma/therapy , CD8-Positive T-Lymphocytes/drug effects , Immunotherapy/methods , Lung Neoplasms/therapy , Lymphocytes, Tumor-Infiltrating/drug effects , Adenocarcinoma/immunology , Animals , Cell Line, Tumor , Central Nervous System Sensitization/drug effects , Cyclophosphamide/administration & dosage , Disease Models, Animal , Drug Therapy/methods , Genes, cdc/drug effects , Humans , Immunity, Innate , Lung Neoplasms/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Organoplatinum Compounds/administration & dosage , Oxaliplatin , Toll-Like Receptor 4/metabolism
6.
Cancer Immunol Immunother ; 64(11): 1369-81, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26160687

ABSTRACT

Endogenous tumor-specific T cells are detectable in patients with different tumor types including malignant melanoma (MM). They can control tumor growth, have impact on patient survival and correlate with improved clinical response to immune checkpoint therapy. Thus, they may represent a potent biomarker for respective treatment decisions. So far, major target antigens of endogenous MM-reactive T cells have not been determined systematically. Instead, autoantibodies are discussed as surrogate parameter for MM-specific T cells. Throughout a period of more than 60 days after tumor resection, we therefore determined in 38 non-metastasized primary MM patients and in healthy individuals by IFNγ ELISpot and bead-based fluorescent multiplex assay major target antigens of spontaneous T cell and humoral responses using a broad panel of MM antigens and assessed the presence and suppressive impact of MM-reactive regulatory T cells (Tregs). We show that MM-reactive T cells are frequent in MM patients, transiently increase after tumor removal and are mostly directed against Melan-A/MART-1, Tyrosinase, NA17-A and p53. MM-specific Tregs were only detected in few patients and inhibited MM-reactive T cells particularly early after tumor resection. Tumor-specific autoantibodies occurred in most patients, but did not correlate with T cell responses. Thus, endogenous antibodies may not be reliable surrogate parameters of MM-reactive T cells.


Subject(s)
Antigens, Neoplasm/immunology , Melanoma/immunology , T-Lymphocytes/immunology , Amino Acid Sequence , Humans , MART-1 Antigen/immunology , Melanoma/pathology , Molecular Sequence Data , Monophenol Monooxygenase/immunology , Neoplasm Staging , Tumor Suppressor Protein p53/immunology
7.
Blood ; 121(22): 4493-503, 2013 May 30.
Article in English | MEDLINE | ID: mdl-23603913

ABSTRACT

Although functionally competent cytotoxic, T cells are frequently observed in malignant diseases, they possess little ability to react against tumor cells. This phenomenon is particularly apparent in multiple myeloma. We here demonstrate that cytotoxic T cells reacted against myeloma antigens when presented by autologous dendritic cells, but not by myeloma cells. We further show by gene expression profiling and flow cytometry that, similar to many other malignant tumors, freshly isolated myeloma cells expressed several carcinoembryonic antigen-related cell adhesion molecules (CEACAMs) at varying proportions. Binding and crosslinking of CEACAM-6 by cytotoxic T cells inhibited their activation and resulted in T-cell unresponsiveness. Blocking of CEACAM-6 on the surface of myeloma cells by specific monoclonal antibodies or CEACAM-6 gene knock down by short interfering RNA restored T-cell reactivity against malignant plasma cells. These findings suggest that CEACAM-6 plays an important role in the regulation of CD8+ T-cell responses against multiple myeloma; therefore, therapeutic targeting of CEACAM-6 may be a promising strategy to improve myeloma immunotherapy.


Subject(s)
Antigens, CD/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Adhesion Molecules/immunology , Multiple Myeloma/immunology , T-Lymphocytes, Cytotoxic/immunology , Antigens, CD/genetics , Antigens, CD/metabolism , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Coculture Techniques , Cytotoxicity, Immunologic/genetics , Cytotoxicity, Immunologic/immunology , GPI-Linked Proteins/genetics , GPI-Linked Proteins/immunology , GPI-Linked Proteins/metabolism , Humans , Immunotherapy/methods , MCF-7 Cells , Multiple Myeloma/pathology , Multiple Myeloma/therapy , Plasma Cells/immunology , Plasma Cells/metabolism , Plasma Cells/pathology , RNA, Small Interfering/genetics , Signal Transduction/immunology , T-Lymphocytes, Cytotoxic/metabolism , Tumor Cells, Cultured , U937 Cells
8.
Blood ; 115(25): 5154-63, 2010 Jun 24.
Article in English | MEDLINE | ID: mdl-20378753

ABSTRACT

Transfer of tumor antigen-specific T-cell receptors (TCRs) into human T cells aims at redirecting their cytotoxicity toward tumors. Efficacy and safety may be affected by pairing of natural and introduced TCRalpha/beta chains potentially leading to autoimmunity. We hypothesized that a novel single-chain (sc)TCR framework relying on the coexpression of the TCRalpha constant alpha (Calpha) domain would prevent undesired pairing while preserving structural and functional similarity to a fully assembled double-chain (dc)TCR/CD3 complex. We confirmed this hypothesis for a murine p53-specific scTCR. Substantial effector function was observed only in the presence of a murine Calpha domain preceded by a TCRalpha signal peptide for shuttling to the cell membrane. The generalization to a human gp100-specific TCR required the murinization of both C domains. Structural and functional T-cell avidities of an accessory disulfide-linked scTCR gp100/Calpha were higher than those of a dcTCR. Antigen-dependent phosphorylation of the proximal effector zeta-chain-associated protein kinase 70 at tyrosine 319 was not impaired, reflecting its molecular integrity in signaling. In melanoma-engrafted nonobese diabetic/severe combined immunodeficient mice, adoptive transfer of scTCR gp100/Calpha transduced T cells conferred superior delay in tumor growth among primary and long-term secondary tumor challenges. We conclude that the novel scTCR constitutes a reliable means to immunotherapeutically target hematologic malignancies.


Subject(s)
Adoptive Transfer , Antigens, Neoplasm/immunology , Melanoma/therapy , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocytes/immunology , Tumor Suppressor Protein p53/immunology , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Cell Line, Tumor , Humans , Immunity, Cellular , Melanoma/genetics , Melanoma/immunology , Melanoma/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Phosphorylation/genetics , Protein Structure, Tertiary , Receptors, Antigen, T-Cell, alpha-beta/biosynthesis , Receptors, Antigen, T-Cell, alpha-beta/genetics , T-Lymphocytes/metabolism , T-Lymphocytes/transplantation , Transduction, Genetic , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
9.
Cancer Immunol Res ; 10(1): 40-55, 2022 01.
Article in English | MEDLINE | ID: mdl-34795032

ABSTRACT

Macrophages often abound within tumors, express colony-stimulating factor 1 receptor (CSF1R), and are linked to adverse patient survival. Drugs blocking CSF1R signaling have been used to suppress tumor-promoting macrophage responses; however, their mechanisms of action remain incompletely understood. Here, we assessed the lung tumor immune microenvironment in mice treated with BLZ945, a prototypical small-molecule CSF1R inhibitor, using single-cell RNA sequencing and mechanistic validation approaches. We showed that tumor control was not caused by CSF1R+ cell depletion; instead, CSF1R targeting reshaped the CSF1R+ cell landscape, which unlocked cross-talk between antitumoral CSF1R- cells. These cells included IFNγ-producing natural killer and T cells, and an IL12-producing dendritic cell subset, denoted as DC3, which were all necessary for CSF1R inhibitor-mediated lung tumor control. These data indicate that CSF1R targeting can activate a cardinal cross-talk between cells that are not macrophages and that are essential to mediate the effects of T cell-targeted immunotherapies and promote antitumor immunity.See related Spotlight by Burrello and de Visser, p. 4.


Subject(s)
Dendritic Cells/immunology , Immunotherapy/methods , Interferon-gamma/metabolism , Interleukin-12/metabolism , Lung Neoplasms/therapy , Animals , Benzothiazoles/pharmacology , Cell Line, Tumor , Female , Lung Neoplasms/immunology , Mice , Mice, Inbred C57BL , Picolinic Acids/pharmacology , Tumor Microenvironment/drug effects , Tumor-Associated Macrophages/drug effects , Tumor-Associated Macrophages/metabolism , Xenograft Model Antitumor Assays
10.
Sci Immunol ; 6(61)2021 07 02.
Article in English | MEDLINE | ID: mdl-34215680

ABSTRACT

Immunotherapy is revolutionizing cancer treatment but is often restricted by toxicities. What distinguishes adverse events from concomitant antitumor reactions is poorly understood. Here, using anti-CD40 treatment in mice as a model of TH1-promoting immunotherapy, we showed that liver macrophages promoted local immune-related adverse events. Mechanistically, tissue-resident Kupffer cells mediated liver toxicity by sensing lymphocyte-derived IFN-γ and subsequently producing IL-12. Conversely, dendritic cells were dispensable for toxicity but drove tumor control. IL-12 and IFN-γ were not toxic themselves but prompted a neutrophil response that determined the severity of tissue damage. We observed activation of similar inflammatory pathways after anti-PD-1 and anti-CTLA-4 immunotherapies in mice and humans. These findings implicated macrophages and neutrophils as mediators and effectors of aberrant inflammation in TH1-promoting immunotherapy, suggesting distinct mechanisms of toxicity and antitumor immunity.


Subject(s)
Immune Checkpoint Inhibitors/adverse effects , Immunotherapy/adverse effects , Kupffer Cells/drug effects , Liver/drug effects , Neoplasms/therapy , Neutrophils/drug effects , Animals , CD40 Antigens/antagonists & inhibitors , CD40 Antigens/immunology , CTLA-4 Antigen/antagonists & inhibitors , CTLA-4 Antigen/immunology , Cytokines/immunology , Humans , Kupffer Cells/immunology , Liver/immunology , Mice, Transgenic , Neoplasms/immunology , Neutrophils/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology
11.
Cell Rep ; 32(12): 108164, 2020 09 22.
Article in English | MEDLINE | ID: mdl-32966785

ABSTRACT

Myeloid cells co-expressing the markers CD11b, Ly-6G, and SiglecF can be found in large numbers in murine lung adenocarcinomas and accelerate cancer growth by fostering tumor cell invasion, angiogenesis, and immunosuppression; however, some of these cells' fundamental features remain unexplored. Here, we show that tumor-infiltrating CD11b+ Ly-6G+ SiglecFhigh cells are bona fide mature neutrophils and therefore differ from other myeloid cells, including SiglecFhigh eosinophils, SiglecFhigh macrophages, and CD11b+ Ly-6G+ myeloid-derived suppressor cells. We further show that SiglecFhigh neutrophils gradually accumulate in growing tumors, where they can live for several days; this lifespan is in marked contrast to that of their SiglecFlow counterparts and neutrophils in general, which live for several hours only. Together, these findings reveal distinct attributes for tumor-promoting SiglecFhigh neutrophils and help explain their deleterious accumulation in the tumor bed.


Subject(s)
Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/pathology , Antigens, Ly/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Neutrophils/pathology , Sialic Acid Binding Immunoglobulin-like Lectins/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Lung/pathology , Male , Mice, Inbred C57BL
12.
Nat Commun ; 11(1): 2762, 2020 06 02.
Article in English | MEDLINE | ID: mdl-32488020

ABSTRACT

Neutrophils are an essential part of the innate immune system. To study their importance, experimental studies often aim to deplete these cells, generally by injecting anti-Ly6G or anti-Gr1 antibodies. However, these approaches are only partially effective, transient or lack specificity. Here we report that neutrophils remaining after anti-Ly6G treatment are newly derived from the bone marrow, instead of depletion escapees. Mechanistically, newly generated, circulating neutrophils have lower Ly6G membrane expression, and consequently reduced targets for anti-Ly6G-mediated depletion. To overcome this limitation, we develop a double antibody-based depletion strategy that enhances neutrophil elimination by anti-Ly6G treatment. This approach achieves specific, durable and controlled reduction of neutrophils in vivo, and may be suitable for studying neutrophil function in experimental models.


Subject(s)
Antigens, Ly/immunology , Neutrophils/immunology , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/therapeutic use , Antigens, Ly/genetics , Bone Marrow/immunology , Cell Death , Disease Models, Animal , Gene Expression , Immunity, Innate , Mice , Mice, Inbred C57BL , RNA, Messenger/metabolism
13.
Clin Cancer Res ; 26(6): 1277-1287, 2020 03 15.
Article in English | MEDLINE | ID: mdl-31852835

ABSTRACT

PURPOSE: Sessile serrated lesions (SSL) are precursors to colon carcinoma, and their distinction from other polyps, in particular hyperplastic polyps (HP), presents significant diagnostic challenges. We evaluated expression patterns in colonic polyps of previously identified colon carcinoma-associated extracellular matrix (ECM) proteins to identify markers distinguishing SSLs from other polyps. EXPERIMENTAL DESIGN: Gene-expression analyses of ECM proteins were performed using publicly available data on preneoplastic colonic polyps. In parallel, we evaluated by IHC the expression of agrin (AGRN) in over 400 colonic polyps, including HP, SSL with and without dysplasia, traditional serrated adenomas (TSA), and tubular adenomas (TA), and compared the consistency of standard histologic diagnosis of SSLs by experienced gastrointestinal pathologists with that of AGRN IHC. RESULTS: Differential gene expression analysis and IHC identified AGRN, serine peptidase inhibitor (SERPINE2), and TIMP metallopeptidase inhibitor 1 (TIMP1) elevated in SSLs and HPs but decreased in TAs and absent in normal colon. AGRN-positive basal laminae were noted in all TA, TSA, HP, and SSL in distinguishable patterns, whereas other polyps and normal mucosa were negative. SSL with or without dysplasia consistently showed IHC staining for AGRN in the muscularis mucosae, which was absent in HP, TSA, TA, and other polyps. In contrast, histologic evaluation showed only weak interobserver agreement (kappa value = 0.493) in distinguishing SSLs. CONCLUSIONS: Muscularis mucosae-based AGRN immunostaining is a novel biomarker to distinguish SSL from HP, TSA, and TA, with a specificity of 97.1% and sensitivity of 98.9% and can assist in diagnosis of morphologically challenging colonic polyps.


Subject(s)
Agrin/metabolism , Biomarkers, Tumor/metabolism , Colonic Polyps/diagnosis , Colorectal Neoplasms/diagnosis , Gene Expression Regulation, Neoplastic , Hyperplasia/diagnosis , Mucous Membrane/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Agrin/genetics , Child , Child, Preschool , Colonic Polyps/genetics , Colonic Polyps/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Diagnosis, Differential , Humans , Hyperplasia/genetics , Hyperplasia/metabolism , Middle Aged , Mucous Membrane/pathology , Young Adult
14.
Int J Oncol ; 34(4): 951-62, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19287952

ABSTRACT

Oncolytic virotherapy, a new type of cancer therapy involving viruses with oncolytic and immunostimulatory potential, is based on tumor selective viral replication, resulting in a specific lysis of tumor cells. Effective tumor targeting of oncolytic viruses remains a major problem because only a fraction of systemically applied viruses can reach the tumor tissue. We describe for the first time in an in vitro co-culture system that T lymphocytes can be loaded with Newcastle disease virus (NDV) in such a way that the virus load will be transferred to the tumor target cells upon contact of the T cells with tumor cells. The effectiveness of this NDV 'hitchhiking' on T cells can be influenced by the amount of virus, the ratio of T cells to tumor cells, the activation status of the T cells and by the virulence of the virus as shown by flow cytometry, quantitative real-time PCR and fluorescence microscopy. In a tumor neutralization assay in vitro, monolayers of human tumor cells could be completely and effectively destroyed by the addition of polyclonally activated human T cells loaded with oncolytic NDV. This process involves the formation of large T cell clusters as revealed by phase-contrast microscopy. Loading of oncolytic NDV onto activated T cells and adoptive transfer into a tumor-bearing host might enhance the efficacy of adoptive T cell therapy of tumors as well as tumor targeting of oncolytic viruses.


Subject(s)
Newcastle disease virus/metabolism , Oncolytic Virotherapy/methods , T-Lymphocytes/metabolism , T-Lymphocytes/virology , Animals , Cell Line, Tumor , Coculture Techniques , Flow Cytometry/methods , Humans , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Microscopy, Fluorescence/methods , Microscopy, Phase-Contrast/methods , Models, Biological , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/cytology
15.
Cell Stress ; 3(11): 348-360, 2019 Oct 14.
Article in English | MEDLINE | ID: mdl-31799501

ABSTRACT

LTX-315 is an oncolytic peptide that has antitumor efficacy in mice grafted with various tumor cell lines and is currently being tested in phase II clinical trials. Here we aimed to further evaluate LTX-315 in conditional genetic mouse models of cancer that typically resist current treatment options and to better understand the drug's mode of action in vivo. We report LTX-315 mediates profound antitumor effects against Braf- and Pten-driven melanoma and delays the progression of Kras- and P53-driven soft tissue sarcoma in mice. Additionally, we show in melanoma that LTX-315 triggers two sequential phases of antitumor response. The first phase of response, which begins within minutes of drug delivery into tumors, is defined by disrupted tumor vasculature and decreased tumor burden and occurs independently of lymphocytes. The second phase of response, which continues over weeks, is defined by long-term alteration of the tumor microenvironment; the changes induced by LTX-315 are most notably characterized by CD8+ T cell infiltration. We further show that these CD8+ T cells are involved in suppressing melanoma outgrowth in mice and report similar CD8+ T cell infiltration following LTX-315 treatment in melanoma and sarcoma patients. Taken together, these findings reveal LTX-315's multiple antitumor effects, including disrupting the tumor vasculature and promoting the conversion of poorly immunogenic tumors into ones that display antitumor T cell immunity.

16.
Nat Commun ; 10(1): 1486, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30940805

ABSTRACT

Immunogenic cell death (ICD) converts dying cancer cells into a therapeutic vaccine and stimulates antitumor immune responses. Here we unravel the results of an unbiased screen identifying high-dose (10 µM) crizotinib as an ICD-inducing tyrosine kinase inhibitor that has exceptional antineoplastic activity when combined with non-ICD inducing chemotherapeutics like cisplatin. The combination of cisplatin and high-dose crizotinib induces ICD in non-small cell lung carcinoma (NSCLC) cells and effectively controls the growth of distinct (transplantable, carcinogen- or oncogene induced) orthotopic NSCLC models. These anticancer effects are linked to increased T lymphocyte infiltration and are abolished by T cell depletion or interferon-γ neutralization. Crizotinib plus cisplatin leads to an increase in the expression of PD-1 and PD-L1 in tumors, coupled to a strong sensitization of NSCLC to immunotherapy with PD-1 antibodies. Hence, a sequential combination treatment consisting in conventional chemotherapy together with crizotinib, followed by immune checkpoint blockade may be active against NSCLC.


Subject(s)
Antineoplastic Agents/administration & dosage , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Death/drug effects , Crizotinib/administration & dosage , Lung Neoplasms/drug therapy , Animals , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/physiopathology , Cell Line, Tumor , Female , Humans , Interferon-gamma/immunology , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Lung Neoplasms/physiopathology , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , T-Lymphocytes/immunology
18.
Cancer Cell ; 31(5): 614-615, 2017 05 08.
Article in English | MEDLINE | ID: mdl-28486102

ABSTRACT

Successful antitumor immunity is thought to require T cell entry into tumors, though mechanisms regulating this process remain unclear. In this issue of Cancer Cell, Spranger et al. indicate that chemokines produced by intratumoral Batf3 dendritic cells are critical for effector T cell recruitment. The findings have implications for immunotherapy.


Subject(s)
T-Lymphocytes/immunology , Tumor Microenvironment/immunology , Chemokines/immunology , Dendritic Cells/immunology , Humans , Immunotherapy
19.
Sci Transl Med ; 9(392)2017 05 31.
Article in English | MEDLINE | ID: mdl-28566423

ABSTRACT

Efficient delivery of therapeutic nanoparticles (TNPs) to tumors is critical in improving efficacy, yet strategies that universally maximize tumoral targeting by TNP modification have been difficult to achieve in the clinic. Instead of focusing on TNP optimization, we show that the tumor microenvironment itself can be therapeutically primed to facilitate accumulation of multiple clinically relevant TNPs. Building on the recent finding that tumor-associated macrophages (TAM) can serve as nanoparticle drug depots, we demonstrate that local tumor irradiation substantially increases TAM relative to tumor cells and, thus, TNP delivery. High-resolution intravital imaging reveals that after radiation, TAM primarily accumulate adjacent to microvasculature, elicit dynamic bursts of extravasation, and subsequently enhance drug uptake in neighboring tumor cells. TAM depletion eliminates otherwise beneficial radiation effects on TNP accumulation and efficacy, and controls with unencapsulated drug show that radiation effects are more pronounced with TNPs. Priming with combined radiation and cyclophosphamide enhances vascular bursting and tumoral TNP concentration, in some cases leading to a sixfold increase of TNP accumulation in the tumor, reaching 6% of the injected dose per gram of tissue. Radiation therapy alters tumors for enhanced TNP delivery in a TAM-dependent fashion, and these observations have implications for the design of next-generation tumor-targeted nanomaterials and clinical trials for adjuvant strategies.


Subject(s)
Drug Delivery Systems , Macrophages/pathology , Nanoparticles/chemistry , Neoplasms/blood supply , Neoplasms/radiotherapy , Animals , Cell Count , Cell Line, Tumor , Chemistry, Pharmaceutical , Combined Modality Therapy , Cyclophosphamide/pharmacology , Cyclophosphamide/therapeutic use , Extravasation of Diagnostic and Therapeutic Materials/pathology , Humans , Intravital Microscopy , Macrophages/drug effects , Macrophages/radiation effects , Mice, Nude , Neoplasms/drug therapy , Permeability , Phagocytes/drug effects , Phagocytes/pathology , Phagocytes/radiation effects , Tumor Microenvironment/drug effects , Tumor Microenvironment/radiation effects , Xenograft Model Antitumor Assays
20.
Nat Commun ; 8: 14293, 2017 02 08.
Article in English | MEDLINE | ID: mdl-28176769

ABSTRACT

Involvement of the immune system in tumour progression is at the forefront of cancer research. Analysis of the tumour immune microenvironment has yielded a wealth of information on tumour biology, and alterations in some immune subtypes, such as tumour-associated macrophages (TAM), can be strong prognostic indicators. Here, we use optical tissue clearing and a TAM-targeting injectable fluorescent nanoparticle (NP) to examine three-dimensional TAM composition, tumour-to-tumour heterogeneity, response to colony-stimulating factor 1 receptor (CSF-1R) blockade and nanoparticle-based drug delivery in murine pulmonary carcinoma. The method allows for rapid tumour volume assessment and spatial information on TAM infiltration at the cellular level in entire lungs. This method reveals that TAM density was heterogeneous across tumours in the same animal, overall TAM density is different among separate pulmonary tumour models, nanotherapeutic drug delivery correlated with TAM heterogeneity, and successful response to CSF-1R blockade is characterized by enhanced TAM penetration throughout and within tumours.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Lewis Lung/diagnostic imaging , Imaging, Three-Dimensional/methods , Lung Neoplasms/diagnostic imaging , Macrophages/immunology , Aminopyridines/pharmacology , Aminopyridines/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Carcinoma, Lewis Lung/drug therapy , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/pathology , Cell Line, Tumor , Female , Humans , Lung/diagnostic imaging , Lung/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Macrophage Colony-Stimulating Factor/immunology , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Microscopy, Confocal/methods , Microscopy, Fluorescence/methods , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Perfusion/methods , Pyrroles/pharmacology , Pyrroles/therapeutic use , RAW 264.7 Cells , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Signal Transduction/drug effects , Tomography, X-Ray Computed , Treatment Outcome , Tumor Burden/drug effects , Tumor Burden/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL