Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Cell ; 185(7): 1189-1207.e25, 2022 03 31.
Article in English | MEDLINE | ID: mdl-35325594

ABSTRACT

Macrophage infiltration is a hallmark of solid cancers, and overall macrophage infiltration correlates with lower patient survival and resistance to therapy. Tumor-associated macrophages, however, are phenotypically and functionally heterogeneous. Specific subsets of tumor-associated macrophage might be endowed with distinct roles on cancer progression and antitumor immunity. Here, we identify a discrete population of FOLR2+ tissue-resident macrophages in healthy mammary gland and breast cancer primary tumors. FOLR2+ macrophages localize in perivascular areas in the tumor stroma, where they interact with CD8+ T cells. FOLR2+ macrophages efficiently prime effector CD8+ T cells ex vivo. The density of FOLR2+ macrophages in tumors positively correlates with better patient survival. This study highlights specific roles for tumor-associated macrophage subsets and paves the way for subset-targeted therapeutic interventions in macrophages-based cancer therapies.


Subject(s)
Breast Neoplasms , Macrophages , Breast/immunology , Breast Neoplasms/epidemiology , Breast Neoplasms/immunology , CD8-Positive T-Lymphocytes , Female , Folate Receptor 2 , Humans , Lymphocytes, Tumor-Infiltrating , Prognosis
2.
Immunity ; 38(4): 729-41, 2013 Apr 18.
Article in English | MEDLINE | ID: mdl-23562161

ABSTRACT

The therapeutic efficacy of anthracyclines relies on antitumor immune responses elicited by dying cancer cells. How chemotherapy-induced cell death leads to efficient antigen presentation to T cells, however, remains a conundrum. We found that intratumoral CD11c(+)CD11b(+)Ly6C(hi) cells, which displayed some characteristics of inflammatory dendritic cells and included granulomonocytic precursors, were crucial for anthracycline-induced anticancer immune responses. ATP released by dying cancer cells recruited myeloid cells into tumors and stimulated the local differentiation of CD11c(+)CD11b(+)Ly6C(hi) cells. Such cells efficiently engulfed tumor antigens in situ and presented them to T lymphocytes, thus vaccinating mice, upon adoptive transfer, against a challenge with cancer cells. Manipulations preventing tumor infiltration by CD11c(+)CD11b(+)Ly6C(hi) cells, such as the local overexpression of ectonucleotidases, the blockade of purinergic receptors, or the neutralization of CD11b, abolished the immune system-dependent antitumor activity of anthracyclines. Our results identify a subset of tumor-infiltrating leukocytes as therapy-relevant antigen-presenting cells.


Subject(s)
Anthracyclines/administration & dosage , Antigen-Presenting Cells/immunology , Antineoplastic Agents/administration & dosage , Dendritic Cells/immunology , Neoplasms, Experimental/immunology , Adoptive Transfer , Animals , Anthracyclines/adverse effects , Antigens, Ly/metabolism , Antigens, Neoplasm/immunology , Antineoplastic Agents/adverse effects , Apoptosis , CD11b Antigen/metabolism , CD11c Antigen/metabolism , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Granulocyte Precursor Cells/immunology , Immunity, Cellular , Mice , Mice, Inbred C57BL , Monocyte-Macrophage Precursor Cells/immunology , Neoplasms, Experimental/drug therapy , Nucleotidases/metabolism , Receptors, Purinergic/metabolism
3.
Blood ; 126(26): 2781-9, 2015 Dec 24.
Article in English | MEDLINE | ID: mdl-26508783

ABSTRACT

Cytomegalovirus (CMV) infection is responsible for substantial morbidity and mortality after allogeneic hematopoietic stem cell transplant. T-cell immunity is critical for control of CMV infection, and correction of the immune deficiency induced by transplant is now clinically achievable by the adoptive transfer of donor-derived CMV-specific T cells. It is notable, however, that most clinical studies of adoptive T- cell therapy exclude patients with graft-versus-host disease (GVHD) from receiving systemic corticosteroid therapy, which impairs cellular immunity. This group of patients remains the highest clinical risk group for recurrent and problematic infections. Here, we address this unmet clinical need by genetic disruption of the glucocorticoid receptor (GR) gene using electroporation of transcription activator-like effector nuclease (TALEN) messenger RNA. We demonstrate efficient inactivation of the GR gene without off-target activity in Streptamer-selected CMV-specific CD8(+) T cells (HLA-A02/NLV peptide), conferring resistance to glucocorticoids. TALEN-modified CMV-specific T cells retained specific killing of target cells pulsed with the CMV peptide NLV in the presence of dexamethasone (DEX). Inactivation of the GR gene also conferred resistance to DEX in a xenogeneic GVHD model in sublethally irradiated NOD-scid IL2rγ(null) mice. This proof of concept provides the rationale for the development of clinical protocols for producing and administering high-purity genetically engineered virus-specific T cells that are resistant to the suppressive effects of corticosteroids.


Subject(s)
Adoptive Transfer/methods , CD8-Positive T-Lymphocytes/immunology , Cytomegalovirus Infections/immunology , Gene Knockdown Techniques/methods , Hematopoietic Stem Cell Transplantation/adverse effects , Receptors, Glucocorticoid/genetics , Animals , Cytomegalovirus Infections/prevention & control , Electroporation , Endonucleases/genetics , Graft vs Host Disease , Heterografts , Humans , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , RNA, Messenger , Transfection
4.
STAR Protoc ; 4(1): 101922, 2023 03 17.
Article in English | MEDLINE | ID: mdl-36516053

ABSTRACT

In vivo genome-wide CRISPR screens in primary T cells allow the systematic and unbiased identification of non-redundant regulatory mechanisms shaping immune responses. Here, we present an optimized protocol for efficient generation of a pool of genome-wide inactivated Cas9-expressing T cells using a retroviral library of sgRNA. We detail the process of large-scale viral production and library integration in activated murine T cells as well as the two-step PCR approach for sgRNA recovery and abundance evaluation. For complete details on the use and execution of this protocol, please refer to Sutra Del Galy et al. (2020).


Subject(s)
CRISPR-Cas Systems , RNA, Guide, CRISPR-Cas Systems , Animals , Mice , CRISPR-Cas Systems/genetics , T-Lymphocytes , Gene Library , Retroviridae/genetics
5.
Nat Cancer ; 4(7): 968-983, 2023 07.
Article in English | MEDLINE | ID: mdl-37248395

ABSTRACT

CD4+ T cells and CD4+ chimeric antigen receptor (CAR) T cells display highly variable antitumor activity in preclinical models and in patients; however, the mechanisms dictating how and when CD4+ T cells promote tumor regression are incompletely understood. With the help of functional intravital imaging, we report that interferon (IFN)-γ production but not perforin-mediated cytotoxicity was the dominant mechanism for tumor elimination by anti-CD19 CD4+ CAR T cells. Mechanistically, mouse or human CD4+ CAR T-cell-derived IFN-γ diffused extensively to act on tumor cells at distance selectively killing tumors sensitive to cytokine-induced apoptosis, including antigen-negative variants. In anti-CD19 CAR T-cell-treated patients exhibiting elevated CAR CD4:CD8 ratios, strong induction of serum IFN-γ was associated with increased survival. We propose that the sensitivity of tumor cells to the pro-apoptotic activity of IFN-γ is a major determinant of CD4+ CAR T-cell efficacy and may be considered to guide the use of CD4+ T cells during immunotherapy.


Subject(s)
Neoplasms , T-Lymphocytes , Humans , Animals , Mice , Receptors, Antigen, T-Cell , Cytokines , Interferon-gamma , CD4-Positive T-Lymphocytes
6.
Cancer Immunol Res ; 11(4): 530-545, 2023 04 03.
Article in English | MEDLINE | ID: mdl-36883368

ABSTRACT

One billion people worldwide get flu every year, including patients with non-small cell lung cancer (NSCLC). However, the impact of acute influenza A virus (IAV) infection on the composition of the tumor microenvironment (TME) and the clinical outcome of patients with NSCLC is largely unknown. We set out to understand how IAV load impacts cancer growth and modifies cellular and molecular players in the TME. Herein, we report that IAV can infect both tumor and immune cells, resulting in a long-term protumoral effect in tumor-bearing mice. Mechanistically, IAV impaired tumor-specific T-cell responses, led to the exhaustion of memory CD8+ T cells and induced PD-L1 expression on tumor cells. IAV infection modulated the transcriptomic profile of the TME, fine-tuning it toward immunosuppression, carcinogenesis, and lipid and drug metabolism. Consistent with these data, the transcriptional module induced by IAV infection in tumor cells in tumor-bearing mice was also found in human patients with lung adenocarcinoma and correlated with poor overall survival. In conclusion, we found that IAV infection worsened lung tumor progression by reprogramming the TME toward a more aggressive state.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Influenza A virus , Influenza, Human , Lung Neoplasms , Orthomyxoviridae Infections , Humans , Animals , Mice , Lung Neoplasms/pathology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Tumor Microenvironment , CD8-Positive T-Lymphocytes , Lung , Orthomyxoviridae Infections/pathology
7.
Cancer Metastasis Rev ; 30(1): 61-9, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21249425

ABSTRACT

The success of some chemo- and radiotherapeutic regimens relies on the induction of immunogenic tumor cell death and on the induction of an anticancer immune response. Cells succumbing to immunogenic cell death undergo specific changes in their surface characteristics and release pro-immunogenic factors according to a defined spatiotemporal pattern. This stimulates antigen presenting cells such as dendritic cells to efficiently take up tumor antigens, process them, and cross-prime cytotoxic T lymphocytes, thus eliciting a tumor-specific cognate immune response. Such a response can also target therapy-resistant tumor (stem) cells, thereby leading, at least in some instances, to tumor eradication. In this review, we shed some light on the molecular identity of the factors that are required for cell death to be perceived as immunogenic. We discuss the intriguing observations that the most abundant endoplasmic reticulum protein, calreticulin, the most abundant intracellular metabolite, ATP, and the most abundant non-histone chromatin-binding protein, HMGB1, can determine whether cell death is immunogenic as they appear on the surface or in the microenvironment of dying cells.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Death/drug effects , Animals , Antineoplastic Agents/therapeutic use , Cell Death/immunology , Humans , Neoplasms/drug therapy , Neoplasms/immunology , Signal Transduction/drug effects , Signal Transduction/immunology
8.
J Immunother Cancer ; 10(8)2022 08.
Article in English | MEDLINE | ID: mdl-36054452

ABSTRACT

BACKGROUND: High-risk neuroblastoma is a pediatric cancer with still a dismal prognosis, despite multimodal and intensive therapies. Tumor microenvironment represents a key component of the tumor ecosystem the complexity of which has to be accurately understood to define selective targeting opportunities, including immune-based therapies. METHODS: We combined various approaches including single-cell transcriptomics to dissect the tumor microenvironment of both a transgenic mouse neuroblastoma model and a cohort of 10 biopsies from neuroblastoma patients, either at diagnosis or at relapse. Features of related cells were validated by multicolor flow cytometry and functional assays. RESULTS: We show that the immune microenvironment of MYCN-driven mouse neuroblastoma is characterized by a low content of T cells, several phenotypes of macrophages and a population of cells expressing signatures of myeloid-derived suppressor cells (MDSCs) that are molecularly distinct from the various macrophage subsets. We document two cancer-associated fibroblasts (CAFs) subsets, one of which corresponding to CAF-S1, known to have immunosuppressive functions. Our data unravel a complex content in myeloid cells in patient tumors and further document a striking correspondence of the microenvironment populations between both mouse and human tumors. We show that mouse intratumor T cells exhibit increased expression of inhibitory receptors at the protein level. Consistently, T cells from patients are characterized by features of exhaustion, expressing inhibitory receptors and showing low expression of effector cytokines. We further functionally demonstrate that MDSCs isolated from mouse neuroblastoma have immunosuppressive properties, impairing the proliferation of T lymphocytes. CONCLUSIONS: Our study demonstrates that neuroblastoma tumors have an immunocompromised microenvironment characterized by dysfunctional T cells and accumulation of immunosuppressive cells. Our work provides a new and precious data resource to better understand the neuroblastoma ecosystem and suggest novel therapeutic strategies, targeting both tumor cells and components of the microenvironment.


Subject(s)
Neuroblastoma , Transcriptome , Animals , Child , Ecosystem , Humans , Mice , Neoplasm Recurrence, Local , Neuroblastoma/pathology , Tumor Microenvironment/genetics
9.
Bull Mem Acad R Med Belg ; 166(3-4): 130-8; discussion 139-40, 2011.
Article in French | MEDLINE | ID: mdl-22375493

ABSTRACT

Immunogenic cell death, characterized by calreticulin exposure on the surface of the dying cell, release of the nuclear protein high mobility group box 1 (HMGB1), and release of ATP, enables stimulation of the immune system. We outlined the importance of this kind of cell death for the success of some anticancer chemotherapies. However, defects in the immunogenic cell death signalling pathway can lead to therapeutic failure, apparently because anticancer immune responses must contribute to the efficacy of chemotherapeutic regimens. These defects can be related to the therapy, the tumour cell, the host or the tumour-host interface. It is necessary to characterize these defects to restore and improve the efficacy of anticancer chemotherapies.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Death/immunology , Drug Resistance, Neoplasm/immunology , Neoplasms/drug therapy , Neoplasms/immunology , Antineoplastic Agents/therapeutic use , Humans
10.
Sci Immunol ; 6(66): eabe8219, 2021 Dec 03.
Article in English | MEDLINE | ID: mdl-34860579

ABSTRACT

Although CD8+ T cells undergo autonomous clonal proliferation after antigen stimulation in vivo, the expansion of activated CD4+ T cells is limited by intrinsic factors that are poorly characterized. Using genome-wide CRISPR-Cas9 screens and an in vivo system modeling of antigen-experienced CD4+ T cell recruitment and proliferation during a localized immune response, we identified suppressor of cytokine signaling 1 (SOCS1) as a major nonredundant checkpoint imposing a brake on CD4+ T cell proliferation. Using anti­interleukin-2 receptor (IL-2R) blocking antibodies, interferon-γ receptor (IFN-γR) knockout mice, and transcriptomic analysis, we show that SOCS1 is a critical node integrating both IL-2 and IFN-γ signals to block multiple downstream signaling pathways abrogating CD4+ T helper 1 (TH1) cell response. Inactivation of SOCS1 in both murine and human CD4+ T cell antitumor adoptive therapies restored intratumor accumulation, proliferation/survival, persistence, and polyfunctionality and promoted rejection of established tumors. However, in CD8+ T cells, SOCS1 deletion did not affect the proliferation but rather improved survival and effector functions, which allowed for optimal therapeutic outcome when associated with SOCS1 inactivation in CD4+ T cells. Together, these findings identify SOCS1 as a major intracellular negative checkpoint of adoptive T cell response, opening new possibilities to optimize CAR-T cell therapy composition and efficacy.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Clustered Regularly Interspaced Short Palindromic Repeats/immunology , Suppressor of Cytokine Signaling 1 Protein/immunology , Th1 Cells/immunology , Animals , Female , Male , Mice , Mice, Knockout , Mice, Transgenic
11.
Methods Enzymol ; 631: 107-135, 2020.
Article in English | MEDLINE | ID: mdl-31948544

ABSTRACT

This chapter summarizes the recent findings regarding the development of gene-editing technologies applied to adoptive T cell immunotherapy for cancer. The emerging tools and strategies described below enable the generation of a tailored system, establishing T cells as an advanced biomaterial with redirected specificities that are both programmable and inducible and establish long-term antitumor potential. These reviewed techniques and strategies are implemented with an optimized protocol for the generation of CAR-T cells using lentiviral gene transfer.


Subject(s)
Gene Editing/methods , Genetic Therapy/methods , Immunotherapy, Adoptive/methods , Neoplasms/therapy , T-Lymphocytes , Animals , Humans , Lentivirus , Receptors, Chimeric Antigen/genetics
12.
J Exp Med ; 216(1): 133-151, 2019 01 07.
Article in English | MEDLINE | ID: mdl-30518599

ABSTRACT

Mucosal-associated invariant T (MAIT) cells are abundant T cells with unique specificity for microbial metabolites. MAIT conservation along evolution indicates important functions, but their low frequency in mice has hampered their detailed characterization. Here, we performed the first transcriptomic analysis of murine MAIT cells. MAIT1 (RORγtneg) and MAIT17 (RORγt+) subsets were markedly distinct from mainstream T cells, but quasi-identical to NKT1 and NKT17 subsets. The expression of similar programs was further supported by strong correlations of MAIT and NKT frequencies in various organs. In both mice and humans, MAIT subsets expressed gene signatures associated with tissue residency. Accordingly, parabiosis experiments demonstrated that MAIT and NKT cells are resident in the spleen, liver, and lungs, with LFA1/ICAM1 interactions controlling MAIT1 and NKT1 retention in spleen and liver. The transcriptional program associated with tissue residency was already expressed in thymus, as confirmed by adoptive transfer experiments. Altogether, shared thymic differentiation processes generate "preset" NKT and MAIT subsets with defined effector functions, associated with specific positioning into tissues.


Subject(s)
Natural Killer T-Cells/immunology , Thymus Gland/immunology , Transcriptome/immunology , Animals , Female , Humans , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/immunology , Liver/immunology , Liver/pathology , Lung/immunology , Lung/pathology , Lymphocyte Function-Associated Antigen-1/genetics , Lymphocyte Function-Associated Antigen-1/immunology , Male , Mice , Mice, Transgenic , Natural Killer T-Cells/pathology , Organ Specificity , Spleen/immunology , Spleen/pathology , Thymus Gland/pathology
13.
Cancer Res ; 76(8): 2087-93, 2016 04 15.
Article in English | MEDLINE | ID: mdl-27197251

ABSTRACT

Despite the promising efficacy of adoptive cell therapies (ACT) in melanoma, complete response rates remain relatively low and outcomes in other cancers are less impressive. The immunosuppressive nature of the tumor microenvironment and the expression of immune-inhibitory ligands, such as PD-L1/CD274 by the tumor and stroma are considered key factors limiting efficacy. The addition of checkpoint inhibitors (CPI) to ACT protocols bypasses some mechanisms of immunosuppression, but associated toxicities remain a significant concern. To overcome PD-L1-mediated immunosuppression and reduce CPI-associated toxicities, we used TALEN technology to render tumor-reactive T cells resistant to PD-1 signaling. Here, we demonstrate that inactivation of the PD-1 gene in melanoma-reactive CD8(+) T cells and in fibrosarcoma-reactive polyclonal T cells enhanced the persistence of PD-1 gene-modified T cells at the tumor site and increased tumor control. These results illustrate the feasibility and potency of approaches incorporating advanced gene-editing technologies into ACT protocols to silence immune checkpoints as a strategy to overcome locally active immune escape pathways. Cancer Res; 76(8); 2087-93. ©2016 AACR.


Subject(s)
Lymphocytes, Tumor-Infiltrating/immunology , Programmed Cell Death 1 Receptor/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Fibrosarcoma/immunology , Gene Silencing , Lymphocyte Activation , Melanoma, Experimental/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Programmed Cell Death 1 Receptor/genetics , Signal Transduction , Transfection , Tumor Microenvironment/immunology
14.
Mol Oncol ; 9(10): 1936-65, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26578451

ABSTRACT

The term 'inhibitory checkpoint' refers to the broad spectrum of co-receptors expressed by T cells that negatively regulate T cell activation thus playing a crucial role in maintaining peripheral self-tolerance. Co-inhibitory receptor ligands are highly expressed by a variety of malignancies allowing evasion of anti-tumour immunity. Recent studies demonstrate that manipulation of these co-inhibitory pathways can remove the immunological brakes that impede endogenous immune responses against tumours. Antibodies that block the interactions between co-inhibitory receptors and their ligands have delivered very promising clinical responses, as has been shown by recent successful trials targeting the CTLA-4 and PD-1 pathways. In this review, we discuss the mechanisms of action and expression pattern of co-inhibitory receptors on different T cells subsets, emphasising differences between CD4(+) and CD8(+) T cells. We also summarise recent clinical findings utilising immune checkpoint blockade.


Subject(s)
Immunotherapy , Neoplasms/therapy , T-Lymphocytes/immunology , Animals , Humans
15.
Oncoimmunology ; 2(2): e23082, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23525565

ABSTRACT

Cardiac glycosides (CGs) are natural compounds sharing the ability to operate as potent inhibitors of the plasma membrane Na+/K+-ATPase, hence promoting-via an indirect mechanism-the intracellular accumulation of Ca2+ ions. In cardiomyocytes, increased intracellular Ca2+ concentrations exert prominent positive inotropic effects, that is, they increase myocardial contractility. Owing to this feature, two CGs, namely digoxin and digitoxin, have extensively been used in the past for the treatment of several cardiac conditions, including distinct types of arrhythmia as well as contractility disorders. Nowadays, digoxin is approved by the FDA and indicated for the treatment of congestive heart failure, atrial fibrillation and atrial flutter with rapid ventricular response, whereas the use of digitoxin has been discontinued in several Western countries. Recently, CGs have been suggested to exert potent antineoplastic effects, notably as they appear to increase the immunogenicity of dying cancer cells. In this Trial Watch, we summarize the mechanisms that underpin the unsuspected anticancer potential of CGs and discuss the progress of clinical studies that have evaluated/are evaluating the safety and efficacy of CGs for oncological indications.

16.
Methods Mol Biol ; 965: 121-42, 2013.
Article in English | MEDLINE | ID: mdl-23296654

ABSTRACT

Cellular senescence, which can be defined as a stress response preventing the propagation of cells that have accumulated potentially oncogenic alterations, is invariably associated with a permanent cell cycle arrest. Such an irreversible blockage is mainly mediated by the persistent upregulation of one or more cyclin-dependent kinase inhibitors (CKIs), including (though not limited to) p16( INK4A ) and p21( CIP1 ) and p27( KIP1 ). CKIs operate by binding to cyclin-dependent kinases (CDKs), de facto inhibiting their enzymatic activity. Here, we provide an immunoblotting-based method for the detection and quantification of CKIs in vitro and ex vivo, together with a set of guidelines for the interpretation of results.


Subject(s)
Cell Cycle Checkpoints , Cyclin-Dependent Kinase Inhibitor Proteins/metabolism , Immunoblotting/methods , Cellular Senescence , Cyclin-Dependent Kinase Inhibitor Proteins/isolation & purification , Electrophoresis , Gene Expression Regulation , HeLa Cells , Humans
17.
Methods Mol Biol ; 1004: 43-56, 2013.
Article in English | MEDLINE | ID: mdl-23733568

ABSTRACT

During necrosis and following some instances of apoptosis (in particular in the absence of a proficient phagocytic system), the nonhistone chromatin component high-mobility group box 1 (HMGB1) is released in the extracellular space. In vivo, extracellular HMGB1 can bind Toll-like receptor 4 on the surface of dendritic cells, de facto operating as a danger-associated molecular pattern and alarming the organism to the presence of stressful conditions. Recent results indicate that the release of HMGB1 is one of the key features for cell death to be perceived as immunogenic, i.e., to be capable of triggering a cognate immune response in vivo. Thus, only anticancer agents that-among other features-allow for the release of HMGB1 as they induce cell death are expected to stimulate anticancer immune responses. To investigate the immunogenic potential of conventional anticancer agents and novel cell death inducers on a high-throughput scale, we engineered human osteosarcoma U2OS cells to express HMGB1 fused at the N-terminus of the green fluorescent protein (GFP). Coupled to fluorescence microscopy workstations for automated image acquisition and analysis, this HMGB1-GFP-based biosensor is amenable for the identification of potential inducers of immunogenic cell death among large chemical libraries.


Subject(s)
Biosensing Techniques/methods , HMGB1 Protein/analysis , HMGB1 Protein/metabolism , Cell Death , Cells, Cultured , Green Fluorescent Proteins/metabolism , Humans , Image Processing, Computer-Assisted , Microscopy, Fluorescence , Recombinant Fusion Proteins/metabolism
18.
Cytokine Growth Factor Rev ; 24(4): 311-8, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23787159

ABSTRACT

Preclinical and clinical findings suggest that tumor-specific immune responses may be responsible--at least in part--for the clinical success of therapeutic regimens that rely on immunogenic cell death (ICD) inducers, including anthracyclines and oxaliplatin. The molecular pathways whereby some, but not all, cytotoxic agents promote bona fide ICD remain to be fully elucidated. Nevertheless, a central role for the endoplasmic reticulum (ER) stress response has been revealed in all scenarios of ICD described thus far. Hence, components of the ER stress machinery may constitute clinically relevant druggable targets for the induction of ICD. In this review, we will summarize recent findings in the field of ICD research with a special focus on ER stress mechanisms and their implication for cancer therapy.


Subject(s)
Cell Death/immunology , Endoplasmic Reticulum Stress/immunology , Endoplasmic Reticulum/immunology , Neoplasms/immunology , Animals , Anthracyclines/therapeutic use , Antineoplastic Agents/therapeutic use , Humans , Mice , Neoplasms/drug therapy , Neoplasms/radiotherapy , Organoplatinum Compounds/therapeutic use , Oxaliplatin
19.
Autophagy ; 8(3): 413-5, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22361584

ABSTRACT

One particular strategy to render anticancer therapies efficient consists of converting the patient's own tumor cells into therapeutic vaccines, via the induction of immunogenic cell death (ICD). One of the hallmarks of ICD dwells in the active release of ATP by cells committed to undergo, but not yet having succumbed to, apoptosis. We observed that the knockdown of essential autophagy-related genes (ATG3, ATG5, ATG7 and BECN1) abolishes the pre-apoptotic secretion of ATP by several human and murine cancer cell lines undergoing ICD. Accordingly, autophagy-competent, but not autophagy-deficient, tumor cells treated with ICD inducers in vitro could induce a tumor-specific immune response in vivo. Cancer cell lines stably depleted of ATG5 or ATG7 normally generate tumors in vivo, and such autophagy-deficient neoplasms, upon systemic treatment with ICD inducers, exhibit the same levels of apoptosis (as monitored by nuclear shrinkage and caspase-3 activation) and necrosis (as determined by following the kinetics of HMGB1 release) as their autophagy-proficient counterparts. However, autophagy-incompetent cancers fail to release ATP, to recruit immune effectors into the tumor bed and to respond to chemotherapy in conditions in which autophagy-competent tumors do so. The intratumoral administration of ecto-ATPase inhibitors increases extracellular ATP concentrations, re-establishes the therapy-induced recruitment of dendritic cells and T cells into the tumor bed, and restores the chemotherapeutic response of autophagy-deficient cancers. Altogether, these results suggest that autophagy-incompetent tumor cells escape from chemotherapy-induced (and perhaps natural?) immunosurveillance because they are unable to release ATP.


Subject(s)
Antineoplastic Agents/therapeutic use , Autophagy/immunology , Neoplasms/immunology , Neoplasms/pathology , Adenosine Triphosphate/metabolism , Animals , Antineoplastic Agents/pharmacology , Autophagy/drug effects , Dendritic Cells/drug effects , Dendritic Cells/immunology , Gene Knockdown Techniques , Humans , Mice , Models, Biological , Neoplasms/drug therapy , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
20.
Oncoimmunology ; 1(9): 1640-1642, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-23264921

ABSTRACT

Retrospective clinical data indicate that cardiac glycosides (CGs), notably digoxin, prolong the survival of carcinoma patients treated with conventional chemotherapy. CGs are known to influence the immune response at multiple levels. In addition, recent results suggest that CGs trigger the immunogenic demise of cancer cells, an effect that most likely contributes to their clinical anticancer activity.

SELECTION OF CITATIONS
SEARCH DETAIL