Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Cancer Immunol Immunother ; 71(1): 71-83, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34023958

ABSTRACT

Immunotherapy with anti-GD2 monoclonal antibodies (mAbs) provides some benefits for patients with neuroblastoma (NB). However, the therapeutic efficacy remains limited, and treatment is associated with significant neuropathic pain. Targeting O-acetylated GD2 (OAcGD2) by 8B6 mAb has been proposed to avoid pain by more selective tumor cell targeting. Thorough understanding of its mode of action is necessary to optimize this treatment strategy. Here, we found that 8B6-mediated antibody-dependent cellular phagocytosis (ADCP) performed by macrophages is a key effector mechanism. But efficacy is limited by upregulation of CD47 expression on neuroblastoma cells in response to OAcGD2 mAb targeting, inhibiting 8B6-mediated ADCP. Antibody specific for the CD47 receptor SIRPα on macrophages restored 8B6-induced ADCP of CD47-expressing NB cells and improved the antitumor activity of 8B6 mAb therapy. These results identify ADCP as a critical mechanism for tumor cytolysis by anti-disialoganglioside mAb and support a combination with SIRPα blocking agents for effective neuroblastoma therapy.


Subject(s)
Antibodies, Monoclonal/chemistry , Antigens, Differentiation/chemistry , Neuroblastoma/immunology , Phagocytosis , Receptors, Immunologic/chemistry , Animals , Antibodies/chemistry , Antibody-Dependent Cell Cytotoxicity/immunology , Antineoplastic Agents, Immunological/pharmacology , CD47 Antigen/biosynthesis , Cell Line, Tumor , Flow Cytometry , Gangliosides/chemistry , Humans , Immunotherapy/methods , Macrophages/metabolism , Mice , Microscopy, Fluorescence , Neuroblastoma/metabolism , Up-Regulation
2.
Int J Cancer ; 146(2): 424-438, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31241171

ABSTRACT

Stem cell chemoresistance remains challenging the efficacy of the front-line temozolomide against glioblastoma. Novel therapies are urgently needed to fight those cells in order to control tumor relapse. Here, we report that anti-O-acetyl-GD2 adjuvant immunotherapy controls glioma stem-like cell-driven chemoresistance. Using patient-derived glioblastoma cells, we found that glioma stem-like cells overexpressed O-acetyl-GD2. As a result, monoclonal antibody 8B6 immunotherapy significantly increased temozolomide genotoxicity and tumor cell death in vitro by enhancing temozolomide tumor uptake. Furthermore, the combination therapy decreased the expression of the glioma stem-like cell markers CD133 and Nestin and compromised glioma stem-like cell self-renewal capabilities. When tested in vivo, adjuvant 8B6 immunotherapy prevented the extension of the temozolomide-resistant glioma stem-like cell pool within the tumor bulk in vivo and was more effective than the single agent therapies. This is the first report demonstrating that anti-O-acetyl-GD2 monoclonal antibody 8B6 targets glioblastoma in a manner that control temozolomide-resistance driven by glioma stem-like cells. Together our results offer a proof of concept for using anti-O-acetyl GD2 reagents in glioblastoma to develop more efficient combination therapies for malignant gliomas.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Gangliosides/antagonists & inhibitors , Glioblastoma/drug therapy , Neoplastic Stem Cells/drug effects , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/therapeutic use , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Cell Self Renewal/drug effects , Cell Self Renewal/immunology , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/immunology , Drug Synergism , Gangliosides/immunology , Glioblastoma/immunology , Glioblastoma/pathology , Humans , Mice , Neoplastic Stem Cells/immunology , Temozolomide/pharmacology , Temozolomide/therapeutic use , Xenograft Model Antitumor Assays
3.
J Immunol ; 189(6): 2954-64, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22896630

ABSTRACT

IFN-γ is a master regulator of the immune responses that occur in the transplanted kidney, acting both on the immune system and on the graft itself. The cellular responses to IFN-γ are complex, and emerging evidence suggests that IFN-γ may regulate autophagic functions. Conversely, autophagy modulates innate and adaptive immune functions in various contexts. In this study, we identify a novel mechanism by which IFN-γ activates autophagy in human kidney epithelial cells and provide new insights into how autophagy regulates immune functions in response to IFN-γ. Our results indicate that IFN-γ promotes tryptophan depletion, activates the eIF2α kinase general control nonderepressible-2 (GCN2), and leads to an increase in the autophagic flux. Further, tryptophan supplementation and RNA interference directed against GCN2 inhibited IFN-γ-induced autophagy. This process is of functional relevance because autophagy regulates the secretion of inflammatory cytokines and growth factors by human kidney epithelial cells in response to IFN-γ. These findings assign to IFN-γ a novel function in the regulation of autophagy, which, in turn, modulates IFN-γ-induced secretion of inflammatory cytokines.


Subject(s)
Autophagy/immunology , Epithelial Cells/enzymology , Epithelial Cells/immunology , Interferon-gamma/physiology , Protein Serine-Threonine Kinases/physiology , Tryptophan/deficiency , Autophagy/genetics , Carcinoma, Renal Cell/enzymology , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/pathology , Cytokines/antagonists & inhibitors , Cytokines/metabolism , Epithelial Cells/metabolism , Humans , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Kidney Neoplasms/enzymology , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Tryptophan/metabolism , Tryptophan/physiology , Tumor Cells, Cultured
4.
J Biol Chem ; 287(18): 14557-68, 2012 Apr 27.
Article in English | MEDLINE | ID: mdl-22403402

ABSTRACT

Ischemic injuries permanently affect kidney tissue and challenge cell viability, promoting inflammation and fibrogenesis. Ischemia results in nutrient deprivation, which triggers endoplasmic reticulum stress, ultimately resulting in the unfolded protein response (UPR). The aim of this study was to test whether the UPR could promote an angiogenic response independently of the HIF-1α pathway during ischemic stress in the human kidney epithelium. Glucose deprivation induced the secretion of vascular endothelial growth factor A (VEGFA), basic fibroblast growth factor (bFGF) and angiogenin (ANG) in human kidney epithelial cells independently of HIF-1α. Glucose deprivation, but not hypoxia, triggered endoplasmic reticulum stress and activated the UPR. RNA interference-mediated inhibition of the gene encoding the kinase PERK decreased VEGFA and bFGF expression, but neither gene was affected by the inhibition of IRE1α or ATF6. Furthermore, we show that the expression of angiogenin, which inhibits protein synthesis, is regulated by both IRE1α and PERK, which could constitute a complementary function of the UPR in the repression of translation. In a rat model of acute ischemic stress, we show that the UPR is activated in parallel with VEGFA, bFGF, and ANG expression and independently of HIF-1α.


Subject(s)
Ischemia/metabolism , Kidney/blood supply , Kidney/metabolism , Neovascularization, Pathologic/metabolism , Stress, Physiological , Unfolded Protein Response , Activating Transcription Factor 6/metabolism , Animals , Cell Line , Endoribonucleases/metabolism , Epithelium/metabolism , Epithelium/pathology , Fibroblast Growth Factor 2/metabolism , Gene Expression Regulation , Glucose/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Ischemia/pathology , Kidney/pathology , Male , Membrane Proteins/metabolism , Neovascularization, Pathologic/pathology , Protein Serine-Threonine Kinases/metabolism , Rats , Rats, Sprague-Dawley , Ribonuclease, Pancreatic/metabolism , Vascular Endothelial Growth Factor A/metabolism , eIF-2 Kinase/metabolism
5.
Am J Kidney Dis ; 59(1): 117-21, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21978600

ABSTRACT

Mutations of the UMOD gene, which encodes the uromodulin protein, are associated with tubulointerstitial nephritis and hyperuricemia. UMOD mutations impair uromodulin folding, resulting in its retention within the endoplasmic reticulum (ER) of renal tubular cells. The aim of this study was to investigate whether mutant uromodulin accumulation in epithelial tubular cells is associated with ER stress. We characterized tubular expression of uromodulin and the ER stress surrogate marker Grp78 by immunohistochemistry in kidney biopsy specimens from 7 patients with UMOD-related kidney disease. We compared this population with 5 patients with familial tubulointerstitial nephritis not related to UMOD mutation. All biopsy specimens from patients carrying the UMOD mutation showed strong heterogeneous cytoplasmic expression of uromodulin in cells of the thick ascending limb of the loop of Henle. In the same tubules, Grp78 was highly expressed in a perinuclear pattern. In contrast, in all kidney biopsy specimens from patients without UMOD mutations, uromodulin staining showed normal apical expression and Grp78 expression was not increased. Our observations support the hypothesis that ER accumulation of mutant uromodulin may cause ER stress, providing a potential mechanism for the progression of UMOD-related kidney disease.


Subject(s)
Endoplasmic Reticulum Stress , Kidney Diseases/genetics , Kidney Diseases/metabolism , Mutation , Uromodulin/genetics , Adolescent , Adult , Child , Endoplasmic Reticulum Chaperone BiP , Female , Humans , Male , Middle Aged , Retrospective Studies , Young Adult
6.
Cancers (Basel) ; 13(21)2021 Oct 29.
Article in English | MEDLINE | ID: mdl-34771608

ABSTRACT

Malignant central nervous system tumors are the leading cause of cancer death in children. Progress in high-throughput molecular techniques has increased the molecular understanding of these tumors, but the outcomes are still poor. Even when efficacious, surgery, radiation, and chemotherapy cause neurologic and neurocognitive morbidity. Adoptive cell therapy with autologous CD19 chimeric antigen receptor T cells (CAR T) has demonstrated remarkable remission rates in patients with relapsed refractory B cell malignancies. Unfortunately, tumor heterogeneity, the identification of appropriate target antigens, and location in a growing brain behind the blood-brain barrier within a specific suppressive immune microenvironment restrict the efficacy of this strategy in pediatric neuro-oncology. In addition, the vulnerability of the brain to unrepairable tissue damage raises important safety concerns. Recent preclinical findings, however, have provided a strong rationale for clinical trials of this approach in patients. Here, we examine the most important challenges associated with the development of CAR T cell immunotherapy and further present the latest preclinical strategies intending to optimize genetically engineered T cells' efficiency and safety in the field of pediatric neuro-oncology.

7.
J Vis Exp ; (143)2019 01 19.
Article in English | MEDLINE | ID: mdl-30735151

ABSTRACT

Potentiation of hostile monoclonal antibodies (mAb) by chemotherapeutic agents constitutes a valuable strategy for designing effective and safer therapy against cancer. Here we provide a protocol to identify a rational combination at the preclinical step. First, we describe a cell-based assay to assess the synergism between anticancer mAb and cytotoxic drugs, that uses the combination index equation of Chou and Talalay1. This includes the measurement of tumor cell drug- and antibody-sensitivity using an MTT assay, followed by an automated computer analysis to calculate the combination index (CI) values. CI values of <1 indicate synergism between tested mAbs and cytotoxic agents1. To corroborate the in vitro findings in vivo, we further describe a method to assess the combination regimen efficacy in a xenograft tumor model. In this model, the combined regimen significantly delays tumor growth, which results in a significant extended survival in comparison to single-agent controls. Importantly, the in vivo experimentation reveals that the combination regimen is well tolerated. This protocol allows the effective evaluation of anticancer drug combinations in preclinical models and the identification of rational combination to evaluate in clinical trials.


Subject(s)
Antibodies, Neoplasm/pharmacology , Antineoplastic Agents/pharmacology , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacology , Antibodies, Neoplasm/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Line, Tumor , Drug Interactions , Drug Synergism , Humans , Mice, Inbred NOD , Mice, SCID , Topotecan/pharmacology , Xenograft Model Antitumor Assays
8.
J Immunol Res ; 2017: 5604891, 2017.
Article in English | MEDLINE | ID: mdl-28154831

ABSTRACT

Target selection is a key feature in cancer immunotherapy, a promising field in cancer research. In this respect, gangliosides, a broad family of structurally related glycolipids, were suggested as potential targets for cancer immunotherapy based on their higher abundance in tumors when compared with the matched normal tissues. GD2 is the first ganglioside proven to be an effective target antigen for cancer immunotherapy with the regulatory approval of dinutuximab, a chimeric anti-GD2 therapeutic antibody. Although the therapeutic efficacy of anti-GD2 monoclonal antibodies is well documented, neuropathic pain may limit its application. O-Acetyl-GD2, the O-acetylated-derivative of GD2, has recently received attention as novel antigen to target GD2-positive cancers. The present paper examines the role of O-acetyl-GD2 in tumor biology as well as the available preclinical data of anti-O-acetyl-GD2 monoclonal antibodies. A discussion on the relevance of O-acetyl-GD2 in chimeric antigen receptor T cell therapy development is also included.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigens, Neoplasm/immunology , Gangliosides/immunology , Gangliosides/metabolism , Neoplasms/therapy , Acetylation , Animals , Antibodies, Anti-Idiotypic/adverse effects , Antibodies, Anti-Idiotypic/immunology , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Murine-Derived/therapeutic use , Antigens, Neoplasm/chemistry , Clinical Trials as Topic , Gangliosides/chemistry , Humans , Immunotherapy , Neoplasms/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology
9.
Oncotarget ; 7(27): 41172-41185, 2016 Jul 05.
Article in English | MEDLINE | ID: mdl-27172791

ABSTRACT

There are still unmet medical needs in the treatment of glioblastoma, the most common and the most aggressive glioma of all brain tumors. Here, we found that O-acetyl GD2 is expressed in surgically resected human glioblastoma tissue. In addition, we demonstrated that 8B6 monoclonal antibody specific for O-acetylat GD2 could effectively inhibit glioblastoma cell proliferation in vitro and in vivo. Taken together, these results indicate that O-acetylated GD2 represents a novel antigen for immunotherapeutic-based treatment of high-grade gliomas.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Cancer Vaccines/therapeutic use , Gangliosides/immunology , Glioblastoma/therapy , Adult , Aged , Animals , Apoptosis/immunology , Cell Line, Tumor , Female , Gangliosides/metabolism , Glioblastoma/immunology , Glioblastoma/metabolism , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Molecular Targeted Therapy/methods , Xenograft Model Antitumor Assays
10.
Nat Rev Nephrol ; 11(1): 34-45, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25385287

ABSTRACT

Autophagy degrades pathogens, altered organelles and protein aggregates, and is characterized by the sequestration of cytoplasmic cargos within double-membrane-limited vesicles called autophagosomes. The process is regulated by inputs from the cellular microenvironment, and is activated in response to nutrient scarcity and immune triggers, which signal through a complex molecular network. Activation of autophagy leads to the formation of an isolation membrane, recognition of cytoplasmic cargos, expansion of the autophagosomal membrane, fusion with lysosomes and degradation of the autophagosome and its contents. Autophagy maintains cellular homeostasis during stressful conditions, dampens inflammation and shapes adaptive immunity. A growing body of evidence has implicated autophagy in kidney health, ageing and disease; it modulates tissue responses during acute kidney injuries, regulates podocyte homeostasis and protects against age-related renal disorders. The renoprotective functions of autophagy in epithelial renal cells and podocytes are mostly mediated by the clearance of altered mitochondria, which can activate inflammasomes and apoptosis, and the removal of protein aggregates, which might trigger inflammation and cell death. In translational terms, autophagy is undoubtedly an attractive target for developing new renoprotective treatments and identifying markers of kidney injury.


Subject(s)
Aging/metabolism , Autophagy/physiology , Kidney Diseases/metabolism , Kidney/metabolism , Apoptosis , Homeostasis , Humans , Kidney/cytology , Lysosomes/metabolism , Mitochondria/metabolism
11.
Front Immunol ; 5: 25, 2014.
Article in English | MEDLINE | ID: mdl-24550911

ABSTRACT

Malaria is a deadly infectious disease caused by the intraerythrocytic protozoan parasite Plasmodium. The four species of Plasmodium known to affect humans all produce an inorganic crystal called hemozoin (HZ) during the heme detoxification process. HZ is released from the food vacuole into circulation during erythrocyte lysis, while the released parasites further infect additional naive red blood cells. Once in circulation, HZ is rapidly taken up by circulating monocytes and tissue macrophages, inducing the production of pro-inflammatory mediators, such as interleukin-1ß (IL-1ß). Over the last few years, it has been reported that HZ, similar to uric acid crystals, asbestos, and silica, is able to trigger IL-1ß production via the activation of the NOD-like receptor containing pyrin domain 3 (NLRP3) inflammasome complex. Additionally, recent findings have shown that host factors, such as fibrinogen, have the ability to adhere to free HZ and modify its capacity to activate host immune cells. Although much has been discovered regarding NLRP3 inflammasome induction, the mechanism through which this intracellular multimolecular complex is activated remains unclear. In the present review, the most recent discoveries regarding the capacity of HZ to trigger this innate immune complex as well as the impact of HZ on several other inflammatory signaling pathways will be discussed.

12.
PLoS One ; 8(4): e62418, 2013.
Article in English | MEDLINE | ID: mdl-23638076

ABSTRACT

Patients with Type 2 diabetes (T2D) are highly susceptible to infection and have an increased incidence of some tumors, possibly due to immune system dysfunction. In the innate cellular immune system, Natural Killer (NK) lymphocytes are important effectors responsible for controlling infections and combating tumor development. We analyzed NK cell subsets in 51 patients with long-standing T2D. Compared with healthy blood donors, diabetic patients showed a profound decrease in both NKG2D-positive NK cells (44% vs. 55.5%, P<0.01) and NKp46-positive cells (26% vs. 50%, P<0.01). Decreased expression of these receptors was associated with functional defects, such as reduced NK degranulation capacity when challenged with the tumor target cell line K562 (10.3 vs. 15.8%, P<0.05). This defect could be restored in vitro by stimulating NK cells from T2D patients with IL-15 (P<0.05). NKG2D expression was found to be negatively correlated with HBA1c level (r=-0.50; P=0.009), suggesting that sustained hyperglycemia could directly influence NK cell defects. We demonstrated that endoplasmic reticulum (ER) stress, an important mediator in diabetes-associated complications, was inducible in vitro in normal NK cells and that tunicamycin treatment resulted in a significant decrease in NKG2D expression (P<0.05). Furthermore, markers of the Unfolded Protein Response (UPR) BiP, PDI and sXBP1 mRNAs were significantly increased in NK cells from T2D patients (P<0.05, P<0.01, P<0.05, respectively), indicating that ER stress is activated in vivo through both PERK and IRE1 sensors. These results demonstrate for the first time defects in NK cell-activating receptors NKG2D and NKp46 in T2D patients, and implicate the UPR pathway as a potential mechanism. These defects may contribute to susceptibility to infections and malignancies and could be targetted therapeutically.


Subject(s)
Diabetes Mellitus, Type 2/immunology , Infections/immunology , Killer Cells, Natural/immunology , Neoplasms/immunology , Adult , Aged , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Endoplasmic Reticulum Stress/immunology , Female , Humans , Hyperglycemia/immunology , Killer Cells, Natural/metabolism , Male , Middle Aged , NK Cell Lectin-Like Receptor Subfamily K/genetics , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Natural Cytotoxicity Triggering Receptor 1/genetics , Natural Cytotoxicity Triggering Receptor 1/metabolism , Phenotype , Transcription, Genetic/immunology , Young Adult
13.
Transplantation ; 88(5): 605-13, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19741454

ABSTRACT

Endoplasmic reticulum (ER) stress is an adaptive response to the accumulation of misfolded proteins within the ER, which can trigger cell dedifferentiation and cell suicide. Increasing evidences suggest its implication in mediating allograft injury. Herein, we summarize the mechanisms of ER stress and discuss its implication in allograft injury. Increasing our understanding of the cellular and molecular mechanisms of acute and chronic allograft damages could lead to the development of new biomarkers and to the discovery of new therapeutic strategies to prevent the initiation of graft dysfunction or to promote the tissue regeneration after injury.


Subject(s)
Endoplasmic Reticulum/metabolism , Transplantation/methods , Animals , Calcineurin Inhibitors , Chronic Disease , Humans , Kidney/pathology , Models, Biological , Protein Denaturation , Protein Folding , Regeneration , Reperfusion Injury , Stress, Physiological , Transplantation, Homologous/methods
SELECTION OF CITATIONS
SEARCH DETAIL