Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 188
1.
Science ; 383(6679): eadf6493, 2024 01 12.
Article En | MEDLINE | ID: mdl-38207030

Neutrophils are increasingly recognized as key players in the tumor immune response and are associated with poor clinical outcomes. Despite recent advances characterizing the diversity of neutrophil states in cancer, common trajectories and mechanisms governing the ontogeny and relationship between these neutrophil states remain undefined. Here, we demonstrate that immature and mature neutrophils that enter tumors undergo irreversible epigenetic, transcriptional, and proteomic modifications to converge into a distinct, terminally differentiated dcTRAIL-R1+ state. Reprogrammed dcTRAIL-R1+ neutrophils predominantly localize to a glycolytic and hypoxic niche at the tumor core and exert pro-angiogenic function that favors tumor growth. We found similar trajectories in neutrophils across multiple tumor types and in humans, suggesting that targeting this program may provide a means of enhancing certain cancer immunotherapies.


Cellular Reprogramming , Neoplasms , Neovascularization, Pathologic , Neutrophils , Humans , Neoplasms/blood supply , Neoplasms/immunology , Neutrophils/immunology , Proteomics , Cellular Reprogramming/genetics , Cellular Reprogramming/immunology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/immunology , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , Epigenesis, Genetic , Hypoxia , Transcription, Genetic
2.
Clin Cancer Res ; 29(16): 2988-3003, 2023 08 15.
Article En | MEDLINE | ID: mdl-37265425

PURPOSE: Patients with unresectable/metastatic chondrosarcoma have poor prognoses; conventional chondrosarcoma is associated with a median progression-free survival (PFS) of <4 months after first-line chemotherapy. No standard targeted therapies are available. We present the preclinical characterization of INBRX-109, a third-generation death receptor 5 (DR5) agonist, and clinical findings from a phase I trial of INBRX-109 in unresectable/metastatic chondrosarcoma (NCT03715933). PATIENTS AND METHODS: INBRX-109 was first characterized preclinically as a DR5 agonist, with binding specificity and hepatotoxicity evaluated in vitro and antitumor activity evaluated both in vitro and in vivo. INBRX-109 (3 mg/kg every 3 weeks) was then evaluated in a phase I study of solid tumors, which included a cohort with any subtype of chondrosarcoma and a cohort with IDH1/IDH2-mutant conventional chondrosarcoma. The primary endpoint was safety. Efficacy was an exploratory endpoint, with measures including objective response, disease control rate, and PFS. RESULTS: In preclinical studies, INBRX-109 led to antitumor activity in vitro and in patient-derived xenograft models, with minimal hepatotoxicity. In the phase I study, INBRX-109 was well tolerated and demonstrated antitumor activity in unresectable/metastatic chondrosarcoma. INBRX-109 led to a disease control rate of 87.1% [27/31; durable clinical benefit, 40.7% (11/27)], including two partial responses, and median PFS of 7.6 months. Most treatment-related adverse events, including liver-related events, were low grade (grade ≥3 events in chondrosarcoma cohorts, 5.7%). CONCLUSIONS: INBRX-109 demonstrated encouraging antitumor activity with a favorable safety profile in patients with unresectable/metastatic chondrosarcoma. A randomized, placebo-controlled, phase II trial (ChonDRAgon, NCT04950075) will further evaluate INBRX-109 in conventional chondrosarcoma.


Bone Neoplasms , Chondrosarcoma , Receptors, TNF-Related Apoptosis-Inducing Ligand , Humans , Antibodies, Monoclonal/therapeutic use , Bone Neoplasms/drug therapy , Chemical and Drug Induced Liver Injury , Chondrosarcoma/therapy , Receptors, TNF-Related Apoptosis-Inducing Ligand/agonists , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology
3.
J Med Virol ; 94(11): 5574-5581, 2022 11.
Article En | MEDLINE | ID: mdl-35869417

Mortality in coronavirus disease 2019 (COVID-19) patients has been linked to the presence of a "cytokine storm" induced by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, which involves elevated levels of circulating cytokines and immune-cell hyperactivation. Targeting cytokines during the management of COVID-19 patients has the potential to improve survival rates and reduce mortality. Although cytokine blockers and immune-host modulators are currently being tested in severely ill COVID-19 patients to cope with the overwhelming systemic inflammation, there is not too many successful cases, thus finding new cytokine blockers to attenuate the cytokine storm syndrome is meaningful. In this paper, we significantly attenuated the inflammatory responses induced by mouse hepatitis viruses A59 and SARS-CoV-2 through a soluble DR5-Fc (sDR5-Fc) chimeric protein that blocked the TNF-related apoptosis-inducing ligand-death receptor 5 (TRAIL-DR5) interaction. Our findings indicates that blocking the TRAIL-DR5 pathway through the sDR5-Fc chimeric protein is a promising strategy to treat COVID-19 severe patients requiring intensive care unit  admission or with chronic metabolic diseases.


COVID-19 Drug Treatment , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , SARS-CoV-2 , Animals , Cytokine Release Syndrome/drug therapy , Cytokine Release Syndrome/prevention & control , Cytokines/metabolism , Mice , Recombinant Fusion Proteins/genetics
4.
Cell Rep ; 37(5): 109953, 2021 11 02.
Article En | MEDLINE | ID: mdl-34731630

Receptor clustering is the first and critical step to activate apoptosis by death receptor-5 (DR5). The recent discovery of the autoinhibitory DR5 ectodomain has challenged the long-standing view of its mechanistic activation by the natural ligand Apo2L. Because the autoinhibitory residues have remained unknown, here we characterize a crucial patch of positively charged residues (PPCR) in the highly variable domain of DR5. The PPCR electrostatically separates DR5 receptors to autoinhibit their clustering in the absence of ligand and antibody binding. Mutational substitution and antibody-mediated PPCR interference resulted in increased apoptotic cytotoxic function. A dually specific antibody that enables sustained tampering with PPCR function exceptionally enhanced DR5 clustering and apoptotic activation and distinctively improved the survival of animals bearing aggressive metastatic and recurrent tumors, whereas clinically tested DR5 antibodies without PPCR blockade function were largely ineffective. Our study provides mechanistic insights into DR5 activation and a therapeutic analytical design for potential clinical success.


Antibodies, Monoclonal/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Apoptosis/drug effects , Neoplasms/drug therapy , Receptors, TNF-Related Apoptosis-Inducing Ligand/antagonists & inhibitors , A549 Cells , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/metabolism , Antibodies, Monoclonal, Humanized/pharmacology , Antibody Specificity , Antineoplastic Agents, Immunological/immunology , Antineoplastic Agents, Immunological/metabolism , Epitopes , Humans , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Neoplasms/immunology , Neoplasms/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Signal Transduction , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
5.
Science ; 372(6537)2021 04 02.
Article En | MEDLINE | ID: mdl-33795432

Multivalent display of receptor-engaging antibodies or ligands can enhance their activity. Instead of achieving multivalency by attachment to preexisting scaffolds, here we unite form and function by the computational design of nanocages in which one structural component is an antibody or Fc-ligand fusion and the second is a designed antibody-binding homo-oligomer that drives nanocage assembly. Structures of eight nanocages determined by electron microscopy spanning dihedral, tetrahedral, octahedral, and icosahedral architectures with 2, 6, 12, and 30 antibodies per nanocage, respectively, closely match the corresponding computational models. Antibody nanocages targeting cell surface receptors enhance signaling compared with free antibodies or Fc-fusions in death receptor 5 (DR5)-mediated apoptosis, angiopoietin-1 receptor (Tie2)-mediated angiogenesis, CD40 activation, and T cell proliferation. Nanocage assembly also increases severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pseudovirus neutralization by α-SARS-CoV-2 monoclonal antibodies and Fc-angiotensin-converting enzyme 2 (ACE2) fusion proteins.


Antibodies/chemistry , Antibodies/immunology , Nanostructures , Protein Engineering , Signal Transduction , Angiopoietins/chemistry , Angiopoietins/immunology , Angiopoietins/metabolism , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/immunology , Antibodies, Viral/chemistry , Antibodies, Viral/immunology , B-Lymphocytes/immunology , CD40 Antigens/chemistry , CD40 Antigens/immunology , CD40 Antigens/metabolism , Cell Line, Tumor , Cell Proliferation , Computer Simulation , Genes, Synthetic , Humans , Immunoglobulin Fc Fragments/chemistry , Lymphocyte Activation , Models, Molecular , Protein Binding , Receptor, TIE-2/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , SARS-CoV-2/immunology , T-Lymphocytes/immunology , T-Lymphocytes/physiology
6.
Proc Natl Acad Sci U S A ; 118(4)2021 01 26.
Article En | MEDLINE | ID: mdl-33483421

MYC is a powerful transcription factor overexpressed in many human cancers including B cell and prostate cancers. Antibody therapeutics are exciting opportunities to attack cancers but require knowledge of surface proteins that change due to oncogene expression. To identify how MYC overexpression remodels the cell surface proteome in a cell autologous fashion and in different cell types, we investigated the impact of MYC overexpression on 800 surface proteins in three isogenic model cell lines either of B cell or prostate cell origin engineered to have high or low MYC levels. We found that MYC overexpression resulted in dramatic remodeling (both up- and down-regulation) of the cell surfaceome in a cell type-dependent fashion. We found systematic and large increases in distinct sets of >80 transporters including nucleoside transporters and nutrient transporters making cells more sensitive to toxic nucleoside analogs like cytarabine, commonly used for treating hematological cancers. Paradoxically, MYC overexpression also increased expression of surface proteins driving cell turnover such as TNFRSF10B, also known as death receptor 5, and immune cell attacking signals such as the natural killer cell activating ligand NCR3LG1, also known as B7-H6. We generated recombinant antibodies to these two targets and verified their up-regulation in MYC overexpression cell lines and showed they were sensitive to bispecific T cell engagers (BiTEs). Our studies demonstrate how MYC overexpression leads to dramatic bidirectional remodeling of the surfaceome in a cell type-dependent but functionally convergent fashion and identify surface targets or combinations thereof as possible candidates for cytotoxic metabolite or immunotherapy.


Antibodies, Bispecific/pharmacology , B-Lymphocytes/drug effects , B7 Antigens/genetics , Epithelial Cells/drug effects , Proto-Oncogene Proteins c-myc/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Antibodies, Bispecific/biosynthesis , B-Lymphocytes/immunology , B-Lymphocytes/pathology , B7 Antigens/antagonists & inhibitors , B7 Antigens/immunology , Cell Engineering/methods , Cell Line, Tumor , Cytarabine/pharmacology , Epithelial Cells/immunology , Epithelial Cells/pathology , Gene Expression Regulation, Neoplastic , Humans , Immunosuppressive Agents/pharmacology , Immunotherapy/methods , Male , Molecular Targeted Therapy/methods , Plasmids/chemistry , Plasmids/metabolism , Prostate/immunology , Prostate/pathology , Protein Binding , Proto-Oncogene Proteins c-myc/immunology , Receptors, TNF-Related Apoptosis-Inducing Ligand/antagonists & inhibitors , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , Signal Transduction , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Transfection
7.
Eur J Immunol ; 51(3): 721-733, 2021 03.
Article En | MEDLINE | ID: mdl-33180337

Costimulatory signals potently promote T-cell proliferation and effector function. Agonistic antibodies targeting costimulatory receptors of the TNFR family, such as 4-1BB and CD27, have entered clinical trials in cancer patients. Currently there is limited information how costimulatory signals regulate antigen-specific but also bystander activation of human CD8 T cells. Engineered antigen presenting cells (eAPC) efficiently presenting several common viral epitopes on HLA-A2 in combination with MHC class I tetramer staining were used to investigate the impact of costimulatory signals on human CD8 T-cell responses. CD28 costimulation potently augmented the percentage and number of antigen-reactive CD8 T cells, whereas eAPC expressing 4-1BB-ligand induced bystander proliferation of CD8 T cells and massive expansion of NK cells. Moreover, the 4-1BB agonist urelumab similarly induced bystander proliferation of CD8 T cells and NK cells in a dose-dependent manner. However, the promotion of bystander CD8 T-cell responses is not a general attribute of costimulatory TNF receptor superfamily (TNFRSF) members, since CD27 signals enhanced antigen-specific CD8 T cells responses without promoting significant bystander activation. Thus, the differential effects of costimulatory signals on the activation of human bystander CD8 T cells should be taken into account when costimulatory pathways are harnessed for cancer immunotherapy.


CD8-Positive T-Lymphocytes/immunology , Lymphocyte Activation/immunology , Antigen-Presenting Cells/immunology , Cell Line , Cell Line, Tumor , Cell Proliferation/physiology , Genes, MHC Class I/immunology , Humans , K562 Cells , Killer Cells, Natural/immunology , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology
8.
Sci Rep ; 10(1): 6294, 2020 04 14.
Article En | MEDLINE | ID: mdl-32286343

Dengue virus (DENV) infections may cause life-threatening dengue hemorrhagic fever (DHF). Suppressed protective immunity was shown in these patients. Although several hypotheses have been formulated, the mechanism of DENV-induced immunosuppression remains unclear. Previously, we found that cross-reactive antibodies against tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) receptor 1 (death receptor 4 [DR4]) were elicited in DHF patients, and that anti-DR4 autoantibody fractions were elicited by nonstructural protein 1 (NS1) immunizations in experimental mice. In this study, we found that anti-DR4 antibodies could suppress B lymphocyte function in vitro and in vivo. Treatment with the anti-DR4 immunoglobulin (Ig) induced caspase-dependent cell death in immortalized B lymphocyte Raji cells in vitro. Anti-DR4 Igs elicited by NS1 and DR4 immunizations markedly suppressed mouse spleen transitional T2 B (IgM+IgD+), bone marrow pre-pro-B (B220+CD43+), pre-B (B220+CD43-), and mature B cell (B220+IgD+) subsets in mice. Furthermore, functional analysis revealed that the pre-elicitation of anti-NS1 and anti-DR4 Ig titers suppressed subsequently neutralizing antibody production by immunization with DENV envelop protein. Our data suggest that the elicitation of anti-DR4 titers through DENV NS1 immunization plays a suppressive role in humoral immunity in mice.


Antibodies, Viral/immunology , Immunity, Humoral , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , Severe Dengue/immunology , Viral Nonstructural Proteins/immunology , Animals , Autoantibodies/blood , Cells, Cultured , Dengue Virus/immunology , Humans , Immunization , Mice , Mice, Inbred C57BL
9.
Front Immunol ; 10: 2514, 2019.
Article En | MEDLINE | ID: mdl-31708930

T-cell-based immunotherapy strategies have profoundly improved the clinical management of several solid tumors and hematological malignancies. A recently developed and promising immunotherapy approach is to redirect polyclonal MHC-unrestricted T lymphocytes toward cancer cells by bispecific antibodies (bsAbs) that engage the CD3 complex and a tumor-associated antigen (TAA). The TNF-related apoptosis-inducing ligand receptor 2 (TRAIL-R2) is an attractive immunotherapy target, frequently expressed by neoplastic cells, that we decided to exploit as a TAA. We found that a TRAIL-R2xCD3 bsAb efficiently activates T cells and specifically redirect their cytotoxicity against cancer cells of different origins in vitro, thereby demonstrating its potential as a pan-carcinoma reagent. Moreover, to mimic in vivo conditions, we assessed its ability to retarget T-cell activity in an ex vivo model of ovarian cancer patients' ascitic fluids containing both effector and target cells-albeit with a suboptimal effector-to-target ratio-with remarkable results.


Antibodies, Bispecific/therapeutic use , Antigens, Neoplasm/immunology , CD3 Complex/immunology , Neoplasms/therapy , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , Cell Line, Tumor , Female , Humans , Immunotherapy , Lymphocyte Activation/immunology , Male , Neoplasms/immunology , T-Lymphocytes/immunology
10.
Biomater Sci ; 8(1): 256-265, 2019 Dec 17.
Article En | MEDLINE | ID: mdl-31687671

Chemotherapy is a dominant treatment modality for different types and stages of cancer. However, hypoxia is one of the undesirable limitations of chemotherapy, which reduces the therapeutic efficiency in cancer treatment, ultimately leading to failure of the treatment. Herein, an ideal chemosensitization system capable of attenuating the tumor hypoxia microenvironment and enhancing chemotherapy effects in tumors was designed. This system (designated as the RA/RX Liposome) uses for the first time a pH-sensitive liposome to co-deliver cyclopeptide RA-V as chemotherapeutic drugs and antisense oligonucleotides as HIF-1α inhibitors (RX-0047) for attenuating tumor hypoxia, as well as a caspase-8 activation probe for therapeutic self-monitoring. After modification with death receptor 5-specific antibodies (anti-DR5) on the surface of the liposome, the RA/RX Liposome can successfully deliver components targeting colon tumors in vivo. This work should synergistically enhance the therapeutic effects of the treatment by successfully down-regulating HIF-1α expression against tumor hypoxia during the RA-V-induced apoptotic process. More importantly, the RA/RX Liposome can be precisely applied for therapeutic self-monitoring with the light-up fluorescence of the caspase-8 probe.


Antibodies/administration & dosage , Colonic Neoplasms/drug therapy , Oligonucleotides/administration & dosage , Peptides, Cyclic/administration & dosage , Tumor Hypoxia/drug effects , Animals , Antibodies/pharmacology , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Caspase 8/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Colonic Neoplasms/metabolism , Drug Synergism , Female , HCT116 Cells , HT29 Cells , Humans , Liposomes , Mice , Oligonucleotides/pharmacology , Peptides, Cyclic/pharmacology , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , Xenograft Model Antitumor Assays
11.
Front Immunol ; 10: 1530, 2019.
Article En | MEDLINE | ID: mdl-31333662

Tumor necrosis factor (TNF) related apoptosis-inducing ligand (TRAIL) signaling is far more complex than initially anticipated and can lead to either anti- or protumorigenic effects, hampering the successful clinical use of therapeutic TRAIL receptor agonists. Cell autonomous resistance mechanisms have been identified in addition to paracrine factors that can modulate apoptosis sensitivity. The tumor microenvironment (TME), consisting of cellular and non-cellular components, is a source for multiple signals that are able to modulate TRAIL signaling in tumor and stromal cells. Particularly immune effector cells, also part of the TME, employ the TRAIL/TRAIL-R system whereby cell surface expressed TRAIL can activate apoptosis via TRAIL receptors on tumor cells, which is part of tumor immune surveillance. In this review we aim to dissect the impact of the TME on signaling induced by endogenous and exogenous/therapeutic TRAIL, thereby distinguishing different components of the TME such as immune effector cells, neutrophils, macrophages, and non-hematopoietic stromal cells. In addition, also non-cellular biochemical and biophysical properties of the TME are considered including mechanical stress, acidity, hypoxia, and glucose deprivation. Available literature thus far indicates that tumor-TME interactions are complex and often bidirectional leading to tumor-enhancing or tumor-reducing effects in a tumor model- and tumor type-dependent fashion. Multiple signals originating from different components of the TME simultaneously affect TRAIL receptor signaling. We conclude that in order to unleash the full clinical potential of TRAIL receptor agonists it will be necessary to increase our understanding of the contribution of different TME components on outcome of therapeutic TRAIL receptor activation in order to identify the most critical mechanism responsible for resistance, allowing the design of effective combination treatments.


Cell Communication , Neoplasms , Signal Transduction , TNF-Related Apoptosis-Inducing Ligand , Tumor Microenvironment/immunology , Cell Communication/drug effects , Cell Communication/immunology , Humans , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/pathology , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , Signal Transduction/drug effects , Signal Transduction/immunology , TNF-Related Apoptosis-Inducing Ligand/immunology , TNF-Related Apoptosis-Inducing Ligand/therapeutic use
12.
MAbs ; 11(6): 996-1011, 2019.
Article En | MEDLINE | ID: mdl-31156033

Agonism of members of the tumor necrosis factor receptor superfamily (TNFRSF) with monoclonal antibodies is of high therapeutic interest due to their role in immune regulation and cell proliferation. A major hurdle for pharmacologic activation of this receptor class is the requirement for high-order clustering, a mechanism that imposes a reliance in vivo on Fc receptor-mediated crosslinking. This extrinsic dependence represents a potential limitation of virtually the entire pipeline of agonist TNFRSF antibody drugs, of which none have thus far been approved or reached late-stage clinical trials. We show that tetravalent biepitopic targeting enables robust intrinsic antibody agonism for two members of this family, OX40 and DR5, that is superior to extrinsically crosslinked native parental antibodies. Tetravalent biepitopic anti-OX40 engagement co-stimulated OX40low cells, obviated the requirement for CD28 co-signal for T cell activation, and enabled superior pharmacodynamic activity relative to native IgG in a murine vaccination model. This work establishes a proof of concept for an engineering approach that addresses a major gap for the therapeutic activation of this important receptor class.


Antibodies, Monoclonal/immunology , Immunologic Capping , OX40 Ligand/agonists , Receptors, TNF-Related Apoptosis-Inducing Ligand/agonists , Signal Transduction/immunology , T-Lymphocytes/immunology , Animals , CD28 Antigens/immunology , CHO Cells , Cricetulus , Humans , Jurkat Cells , Mice , Mice, SCID , Mice, Transgenic , OX40 Ligand/immunology , Receptors, Fc/immunology , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , T-Lymphocytes/cytology
13.
Front Immunol ; 10: 951, 2019.
Article En | MEDLINE | ID: mdl-31114586

The maintenance of B cell homeostasis requires a tight control of B cell generation, survival, activation, and maturation. In lymphocytes upon activation, increased sensitivity to apoptotic signals helps controlling differentiation and proliferation. The death receptor Fas is important in this context because genetic Fas mutations in humans lead to an autoimmune lymphoproliferative syndrome that is similar to lymphoproliferation observed in Fas-deficient mice. In contrast, the physiological role of TNF-related apoptosis-inducing ligand receptors (TRAIL-Rs) in humans has been poorly studied so far. Indeed, most studies have focused on tumor cell lines and on mouse models whose results are difficult to transpose to primary human B cells. In the present work, the expression of apoptosis-inducing TRAIL-R1 and TRAIL-R2 and of the decoy receptors TRAIL-R3 and TRAIL-R4 was systematically studied in all developmental stages of peripheral B cells isolated from the blood and secondary lymphoid organs. Expression of TRAIL-Rs is modulated along development, with highest levels observed in germinal center B cells. In addition, T-dependent and T-independent signals elicited induction of TRAIL-Rs with distinct kinetics, which differed among B cell subpopulations: switched memory cells rapidly upregulated TRAIL-R1 and -2 upon activation while naïve B cells only reached similar expression levels at later time points in culture. Increased expression of TRAIL-R1 and -2 coincided with a caspase-3-dependent sensitivity to TRAIL-induced apoptosis in activated B cells but not in freshly isolated resting B cells. Finally, both TRAIL-R1 and TRAIL-R2 could signal actively and both contributed to TRAIL-induced apoptosis. In conclusion, this study provides a systematic analysis of the expression of TRAIL-Rs in human primary B cells and of their capacity to signal and induce apoptosis. This dataset forms a basis to further study and understand the dysregulation of TRAIL-Rs and TRAIL expression observed in autoimmune diseases. Additionally, it will be important to foresee potential bystander immunomodulation when TRAIL-R agonists are used in cancer treatment.


Apoptosis/immunology , B-Lymphocytes/immunology , Intercellular Signaling Peptides and Proteins/immunology , Lymphocyte Activation , Membrane Proteins/immunology , Receptor Activator of Nuclear Factor-kappa B/immunology , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , B-Lymphocytes/cytology , Caspase 3/immunology , Female , Gene Expression Regulation/immunology , Humans , Male , Signal Transduction/immunology
14.
Cell Death Dis ; 10(2): 101, 2019 02 04.
Article En | MEDLINE | ID: mdl-30718507

Development of therapeutic antibodies in oncology has attracted much interest in the past decades. More than 30 of them have been approved and are being used to treat patients suffering from cancer. Despite encouraging results, and albeit most clinical trials aiming at evaluating monoclonal antibodies directed against TRAIL agonist receptors have been discontinued, DR4 or DR5 remain interesting targets, since these receptors are overexpressed by tumour cells and are able to trigger their death. In an effort to develop novel and specific anti-DR4 and anti-DR5 antibodies with improved properties, we used genetic immunization to express native proteins in vivo. Injection of DR4 and DR5 cDNA into the tail veins of mice elicited significant humoral anti-DR4 and anti-DR5 responses and fusions of the corresponding spleens resulted in numerous hybridomas secreting antibodies that could specifically recognize DR4 or DR5 in their native forms. All antibodies bound specifically to their targets with a very high affinity, from picomolar to nanomolar range. Among the 21 anti-DR4 and anti-DR5 monoclonal antibodies that we have produced and purified, two displayed proapoptotic properties alone, five induced apoptosis after cross-linking, four were found to potentiate TRAIL-induced apoptosis and three displayed antiapoptotic potential. The most potent anti-DR4 antibody, C#16, was assessed in vivo and was found, alone, to inhibit tumour growth in animal models. This is the first demonstration that DNA-based immunization method can be used to generate novel monoclonal antibodies targeting receptors of the TNF superfamily that may constitute new therapeutic agents.


Antibodies, Monoclonal/pharmacology , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Female , HEK293 Cells , Humans , Immunization , Mice , Mice, Inbred BALB C , Mice, Nude , Receptors, TNF-Related Apoptosis-Inducing Ligand/agonists , Receptors, TNF-Related Apoptosis-Inducing Ligand/antagonists & inhibitors , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Transplantation, Heterologous
15.
J Immunother Cancer ; 6(1): 71, 2018 07 13.
Article En | MEDLINE | ID: mdl-30005714

Adoptive transfer of T cells transduced with Chimeric Antigen Receptors (CAR) are now FDA-approved for the treatment of B-cell malignancies. Yet, the functionality of the endogenous TCR in CART cells has not been fully assessed. Here, we demonstrate that CART cells progressively upregulate Fas, FasL, DR5 and TRAIL, which result in their programmed cell death, independently of antigen-mediated TCR or CAR activation. CART cell apoptosis occurs even when the CAR contains a single (co-)activatory domain such as CD3ζ, CD28 or 4-1BB. Importantly, the dominant role of the Fas and DR5 pathways in CART cell apoptosis is demonstrated by the significant rescue of CART cells upon in vivo blockade by combined Fas-Fc and DR5-Fc recombinant proteins. These observations are of crucial importance for the long-term persistence of CART cells and for the development of new applications including the combined TCR and CAR activation against solid tumors.


Immunotherapy, Adoptive , Melanoma, Experimental/therapy , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , Skin Neoplasms/therapy , fas Receptor/immunology , Animals , Cell Death , Fas Ligand Protein/immunology , Female , Melanoma, Experimental/pathology , Mice, Inbred C57BL , Receptors, Chimeric Antigen/immunology , Skin Neoplasms/pathology , TNF-Related Apoptosis-Inducing Ligand/immunology , Tumor Burden
16.
MAbs ; 10(7): 1084-1097, 2018 10.
Article En | MEDLINE | ID: mdl-29993310

Recombinant human tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) or TRAIL-receptor agonistic monoclonal antibodies promote apoptosis in most cancer cells, and the differential expression of TRAIL-R2 between tumor and normal tissues allows its exploitation as a tumor-associated antigen. The use of these antibodies as anticancer agents has been extensively studied, but the results of clinical trials were disappointing. The observed lack of anticancer activity could be attributed to intrinsic or acquired resistance of tumor cells to this type of treatment. A possible strategy to circumvent drug resistance would be to strike tumor cells with a second modality based on a different mechanism of action. We therefore set out to generate and optimize a bispecific antibody targeting TRAIL-R2 and CD3. After the construction of different bispecific antibodies in tandem-scFv or single-chain diabody formats to reduce possible immunogenicity, we selected a humanized bispecific antibody with very low aggregates and long-term high stability and functionality. This antibody triggered TRAIL-R2 in an agonistic manner and its anticancer activity proved dramatically potentiated by the redirection of cytotoxic T cells against both sensitive and resistant melanoma cells. The results of our study show that combining the TRAIL-based antitumor strategy with an immunotherapeutic approach in a single molecule could be an effective addition to the anticancer armamentarium.


Antibodies, Bispecific/chemistry , Immunotherapy/methods , Neoplasms/therapy , Single-Chain Antibodies/chemistry , T-Lymphocytes/immunology , Antibodies, Bispecific/therapeutic use , CD3 Complex/immunology , Cell Differentiation , Cell Line, Tumor , Cell Surface Display Techniques , Cytotoxicity, Immunologic , Drug Design , Drug Discovery , Humans , Lymphocyte Activation , Neoplasms/immunology , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology
17.
IET Nanobiotechnol ; 12(4): 436-440, 2018 Jun.
Article En | MEDLINE | ID: mdl-29768226

Cancer treatment with several kinds of drugs, especially targets the apoptotic pathways nowadays. TNF-related apoptosis-inducing ligand (TRAIL) as one of the important members of death receptors, significantly trigger induction of apoptosis in cancer cells. Three conserved domains of Death receptor (DR5) protein extracellular domain, which are fortified cysteine, were chosen and chemically synthesised. Hens were immunised with nano-liposomal peptides, and as a result the purified Immunoglobulin (IgYs) remarkably killed the cancerous MCF7 cells. The flow cytometric assay, confirmed the apoptotic death. Among several kinds of carriers that were used in this research, the nano-liposomal and nanoparticle conjugated, both were acceptable choices for drug delivery. Furthermore, the IgY against DR5's small peptides with such carriers successfully reached the target and significantly killed the cancer cells via apoptosis.


Antineoplastic Agents , Apoptosis/drug effects , Immunoglobulins , Liposomes/chemistry , Nanoconjugates/chemistry , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/immunology , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Biotechnology , Chickens , Drug Carriers/chemistry , Drug Carriers/pharmacology , Egg Yolk , Female , Humans , Immunoglobulins/chemistry , Immunoglobulins/immunology , Immunoglobulins/metabolism , Immunoglobulins/pharmacology , Nanoparticles/chemistry , Nanotechnology
18.
Radiother Oncol ; 124(3): 418-426, 2017 09.
Article En | MEDLINE | ID: mdl-28893414

BACKGROUND AND PURPOSE: We recently reported a time-sensitive, cooperative, anti-tumor effect elicited by radiation (RT) and intra-tumoral-immunocytokine injection in vivo. We hypothesized that RT triggers transcriptional-mediated changes in tumor expression of immune susceptibility markers at delayed time points, which may explain these previously observed time-dependent effects. MATERIALS AND METHODS: We examined the time course of changes in expression of immune susceptibility markers following in vitro or in vivo RT in B78 murine melanoma and A375 human melanoma using flow cytometry, immunoblotting, and qPCR. RESULTS: Flow cytometry and immunoblot revealed time-dependent increases in expression of death receptors and T cell co-stimulatory/co-inhibitory ligands following RT in murine and human melanoma. Using high-throughput qPCR, we observed comparable time courses of RT-induced transcriptional upregulation for multiple immune susceptibility markers. We confirmed analogous changes in B78 tumors irradiated in vivo. We observed upregulated expression of DNA damage response markers days prior to changes in immune markers, whereas phosphorylation of the STAT1 transcription factor occurred concurrently with changes following RT. CONCLUSION: This study highlights time-dependent, transcription-mediated changes in tumor immune susceptibility marker expression following RT. These findings may help in the design of strategies to optimize sequencing of RT and immunotherapy in translational and clinical studies.


Melanoma/radiotherapy , Animals , B7-1 Antigen/biosynthesis , B7-1 Antigen/immunology , B7-H1 Antigen/biosynthesis , B7-H1 Antigen/immunology , Cell Line, Tumor , Female , Flow Cytometry , Gene Knockdown Techniques , Humans , Immunoblotting , Melanoma/genetics , Melanoma/immunology , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/radiotherapy , Mice , Mice, Inbred C57BL , Phosphorylation , Receptors, TNF-Related Apoptosis-Inducing Ligand/biosynthesis , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/immunology , Transcription, Genetic , Up-Regulation
19.
Bull Exp Biol Med ; 163(3): 381-384, 2017 Jul.
Article En | MEDLINE | ID: mdl-28744632

Death receptors, in particular DR5, are highly attractive targets of antitumor therapy. The major limitation to application of natural death receptor ligands (TRAIL) is their non-specific cytotoxicity against normal cells. Since TRAIL can also bind decoy receptors (DcR) and prevent induction of apoptosis, the search for new DR-specific ligands is a topical issue. In the present study, we used combinatorial phage display peptide libraries to select a panel of DR5-binding amino acid sequences. A comparative analysis of the selected peptides enabled identification of the consensus sequence responsible for binding to DR5. Integration of this motif into polypeptide cytotoxic agents may provide targeted elimination of malignantly transformed cells.


Immunoglobulin Fc Fragments/chemistry , Peptide Library , Peptides/chemistry , Receptors, TNF-Related Apoptosis-Inducing Ligand/chemistry , Recombinant Fusion Proteins/chemistry , Amino Acid Motifs , Animals , Binding Sites , Gene Expression , HEK293 Cells , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/immunology , Immunomagnetic Separation/methods , Ligands , Mice , Models, Molecular , Peptides/genetics , Peptides/immunology , Plasmids/chemistry , Plasmids/metabolism , Protein Binding , Protein Structure, Secondary , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Transfection
20.
Sci Rep ; 7(1): 5514, 2017 07 17.
Article En | MEDLINE | ID: mdl-28717244

The TRAIL pathway can mediate apoptosis of hepatic stellate cells to promote the resolution of liver fibrosis. However, TRAIL has the capacity to bind to regulatory receptors in addition to death-inducing receptors; their differential roles in liver fibrosis have not been investigated. Here we have dissected the contribution of regulatory TRAIL receptors to apoptosis resistance in primary human hepatic stellate cells (hHSC). hHSC isolated from healthy margins of liver resections from different donors expressed variable levels of TRAIL-R2/3/4 (but negligible TRAIL-R1) ex vivo and after activation. The apoptotic potential of TRAIL-R2 on hHSC was confirmed by lentiviral-mediated knockdown. A functional inhibitory role for TRAIL-R3/4 was revealed by shRNA knockdown and mAb blockade, showing that these regulatory receptors limit apoptosis of hHSC in response to both oligomerised TRAIL and NK cells. A close inverse ex vivo correlation between hHSC TRAIL-R4 expression and susceptibility to apoptosis underscored its central regulatory role. Our data provide the first demonstration of non-redundant functional roles for the regulatory TRAIL receptors (TRAIL-R3/4) in a physiological setting. The potential for these inhibitory TRAIL receptors to protect hHSC from apoptosis opens new avenues for prognostic and therapeutic approaches to the management of liver fibrosis.


Apoptosis , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Antibodies, Monoclonal/immunology , Apoptosis/drug effects , Cells, Cultured , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Hepatic Stellate Cells/cytology , Hepatic Stellate Cells/metabolism , Humans , Killer Cells, Natural/immunology , Liver/cytology , RNA Interference , RNA, Small Interfering/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/antagonists & inhibitors , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , Receptors, Tumor Necrosis Factor, Member 10c/antagonists & inhibitors , Receptors, Tumor Necrosis Factor, Member 10c/genetics , Receptors, Tumor Necrosis Factor, Member 10c/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Tumor Necrosis Factor Decoy Receptors/antagonists & inhibitors , Tumor Necrosis Factor Decoy Receptors/genetics , Tumor Necrosis Factor Decoy Receptors/metabolism
...