Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 92
Filter
Add more filters

Country/Region as subject
Publication year range
1.
EMBO J ; 39(6): e102214, 2020 03 16.
Article in English | MEDLINE | ID: mdl-32030804

ABSTRACT

Spinal cord microglia contribute to nerve injury-induced neuropathic pain. We have previously demonstrated that toll-like receptor 2 (TLR2) signaling is critical for nerve injury-induced activation of spinal cord microglia, but the responsible endogenous TLR2 agonist has not been identified. Here, we show that nerve injury-induced upregulation of sialyltransferase St3gal2 in sensory neurons leads to an increase in expression of the sialylated glycosphingolipid, GT1b. GT1b ganglioside is axonally transported to the spinal cord dorsal horn and contributes to characteristics of neuropathic pain such as mechanical and thermal hypersensitivity. Spinal cord GT1b functions as an TLR2 agonist and induces proinflammatory microglia activation and central sensitization. Pharmacological inhibition of GT1b synthesis attenuates nerve injury-induced spinal cord microglia activation and pain hypersensitivity. Thus, the St3gal2-GT1b-TLR2 axis may offer a novel therapeutic target for the treatment of neuropathic pain.


Subject(s)
Gangliosides/metabolism , Neuralgia/therapy , Peripheral Nerve Injuries/therapy , Signal Transduction , Toll-Like Receptor 2/agonists , Animals , Gangliosides/antagonists & inhibitors , Gene Expression Regulation , Inflammation , Male , Mice , Mice, Inbred C57BL , Microglia/metabolism , Neuralgia/etiology , Peripheral Nerve Injuries/etiology , Rats , Rats, Sprague-Dawley , Sensory Receptor Cells , Sialyltransferases/genetics , Sialyltransferases/metabolism , Spinal Cord/metabolism , Toll-Like Receptor 2/metabolism
2.
Semin Immunol ; 47: 101390, 2020 02.
Article in English | MEDLINE | ID: mdl-31982247

ABSTRACT

Tumor associated carbohydrate antigens (TACAs) are a class of attractive antigens for the development of anti-cancer immunotherapy. Besides monoclonal antibodies and vaccines, chimeric antigen receptor (CAR) T cells and bispecific antibodies (BsAbs) targeting TACA are exciting directions to harness the power of the immune system to fight cancer. In this review, we focus on two TACAs, i.e., the GD2 ganglioside and the mucin-1 (MUC1) protein. The latest advances in CAR T cells and bispecific antibodies targeting these two antigens are presented. The roles of co-stimulatory molecules, structures of the sequences for antigen binding, methods for CAR and antibody construction, as well as strategies to enhance solid tumor penetration and reduce T cell exhaustion and death are discussed. Furthermore, approaches to reduce "on target, off tumor" side effects are introduced. With further development, CAR T cells and BsAbs targeting GD2 and MUC1 can become powerful agents to effectively treat solid tumor.


Subject(s)
Antigens, Tumor-Associated, Carbohydrate/immunology , Immunotherapy, Adoptive , Neoplasms/immunology , Neoplasms/therapy , Receptors, Antigen, T-Cell/metabolism , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Antibodies, Bispecific/genetics , Antibodies, Bispecific/immunology , Antibodies, Bispecific/metabolism , Epitopes/genetics , Epitopes/immunology , Gangliosides/antagonists & inhibitors , Gangliosides/chemistry , Gangliosides/immunology , Humans , Mucin-1/immunology , Neoplasms/metabolism , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/genetics , Receptors, Chimeric Antigen/chemistry , Receptors, Chimeric Antigen/genetics
3.
J Neurochem ; 156(6): 819-833, 2021 03.
Article in English | MEDLINE | ID: mdl-32743804

ABSTRACT

Ganglioside GD3, a major ganglioside species in neural stem cells, plays a crucial role in maintenance of the self-renewal capacity of these cells. However, its bioactivity in postnatally differentiated neurons in the neurogenic regions of adult brains has not been elucidated. Here, we describe for the first time that deletion of GD3 not only impairs neurotrophin-induced stem cell proliferation, but also alters the dendritic structure as well as the number of synapses of nascent neurons in the dentate gyrus of adult brain. When examining the behavioral phenotypes, GD3 synthase-knockout (GD3S-KO) mice displayed impairment in hippocampus-dependent memory function. To further gain insight into its cellular function, we examined GD3-binding partners from mouse brain extract using a GD3-specific monoclonal antibody, R24, followed by LC-MS/MS analysis and identified a mitochondrial fission protein, the dynamin-related protein-1 (Drp1), as a novel GD3-binding protein. Biochemical and imaging analyses revealed mitochondrial fragmentation in GD3-depleted dentate gyrus neurons, suggesting that GD3 is essential for the mitochondrial Drp1 turnover that is required for efficient mitochondrial fission. These results suggest that GD3 is required for proper dendritic and spine maturation of newborn neurons in adult brain through the regulation of mitochondrial dynamics.


Subject(s)
Dendrites/physiology , Gangliosides/physiology , Hippocampus/growth & development , Hippocampus/physiology , Mitochondria/physiology , Neural Stem Cells/physiology , Neurons/physiology , Animals , Antibodies, Blocking/pharmacology , Antibodies, Monoclonal , Behavior, Animal , Cognition , Dendritic Spines/physiology , Dynamins/genetics , Dynamins/physiology , Gangliosides/antagonists & inhibitors , Gangliosides/genetics , Memory Disorders/genetics , Memory Disorders/psychology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/ultrastructure , Mitochondrial Dynamics
4.
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
5.
Scand J Immunol ; 89(3): e12741, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30549299

ABSTRACT

Novel therapies to treat patients with solid cancers that have developed resistance to chemotherapy represent unmet needs of considerable dimensions. In the present review, we will address the attempts to develop chimeric antigen receptor (CAR) targeted immunotherapy against osteosarcoma (OS). This aggressive cancer displays its peak incidence in children and young adults. The main cause of patient death is lung metastases with a 5-year survival as low as 5%-10% in the primary metastatic setting and 30% in the relapse situation, respectively. Effective adjuvant combination chemotherapy introduced more than 40 years ago improved the survival rates from below 20% to around 60% in patients; however, since then, no major breakthroughs have been made. The use of immune checkpoint inhibitors has been disappointing in OS, while other types of immunotherapies such as CAR T cells remain largely unexplored. Indeed, for CAR T-cell therapy to be efficacious, two main criteria need to be fulfilled: (a) CAR T cells should target an epitope selectively expressed on the cell surface of OS in order to prevent toxicities in normal tissues and (b) the target should also be widely expressed on OS metastases. These challenges have already been undertaken in OS and illustrate the difficulties in developing tomorrow's CAR-T treatment in a solid tumour. We will discuss the experiences with CAR-T therapy development and efficacy to combat the clinical challenges in OS.


Subject(s)
Bone Neoplasms/therapy , Immunotherapy, Adoptive/methods , Osteosarcoma/therapy , Bone Neoplasms/immunology , Bone Neoplasms/mortality , Cancer-Associated Fibroblasts/physiology , Endopeptidases , Gangliosides/antagonists & inhibitors , Gelatinases/physiology , Humans , Membrane Proteins/physiology , Osteosarcoma/immunology , Osteosarcoma/mortality , Receptor, ErbB-2/analysis , Receptor, IGF Type 1/physiology , Receptors, Interleukin-11/antagonists & inhibitors , Serine Endopeptidases/physiology , Tumor Microenvironment
6.
Molecules ; 24(21)2019 Oct 24.
Article in English | MEDLINE | ID: mdl-31653037

ABSTRACT

Antigen-binding fragments of antibodies specific to the tumor-associated ganglioside GD2 are well poised to play a substantial role in modern GD2-targeted cancer therapies, however, rapid elimination from the body and reduced affinity compared to full-length antibodies limit their therapeutic potential. In this study, scFv fragments of GD2-specific antibodies 14.18 were produced in a mammalian expression system that specifically bind to ganglioside GD2, followed by site-directed pegylation to generate mono-, di-, and tetra-scFv fragments. Fractionated pegylated dimers and tetramers of scFv fragments showed significant increase of the binding to GD2 which was not accompanied by cross-reactivity with other gangliosides. Pegylated multimeric di-scFvs and tetra-scFvs exhibited cytotoxic effects in GD2-positive tumor cells, while their circulation time in blood significantly increased compared with monomeric antibody fragments. We also demonstrated a more efficient tumor uptake of the multimers in a syngeneic GD2-positive mouse cancer model. The findings of this study provide the rationale for improving therapeutic characteristics of GD2-specific antibody fragments by multimerization and propose a strategy to generate such molecules. On the basis of multimeric antibody fragments, bispecific antibodies and conjugates with cytotoxic drugs or radioactive isotopes may be developed that will possess improved pharmacokinetic and pharmacodynamic properties.


Subject(s)
Antineoplastic Agents, Immunological , Gangliosides/antagonists & inhibitors , Neoplasms, Experimental , Polyethylene Glycols/chemistry , Single-Chain Antibodies , Animals , Antineoplastic Agents, Immunological/chemistry , Antineoplastic Agents, Immunological/pharmacokinetics , Antineoplastic Agents, Immunological/pharmacology , Cell Line, Tumor , Humans , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/blood , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/pharmacokinetics , Single-Chain Antibodies/pharmacology
7.
Bull Exp Biol Med ; 166(4): 541-547, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30783840

ABSTRACT

Induction of direct cell death is one of the mechanisms of the antitumor effect of GD2-specific antibodies used for the therapy of high-risk neuroblastoma. The mechanisms of the cytotoxic signal triggered by antibody binding to GD2 ganglioside on the surface of the tumor cell remain insufficiently studied. Using inhibitor analysis we demonstrated that actin microfilaments are involved in the cell death induced by GD2-specific antibodies. Specifically, a strong antagonistic influence of cytochalasin D on the cytotoxic effect induced by GD2-specific antibodies was demonstrated in GD2+ tumor cell lines, which was expressed in at least 20% increase in cell survival and a significant decrease of the fraction of cells with fragmented DNA.


Subject(s)
Actin Cytoskeleton/metabolism , Antibodies/pharmacology , Gangliosides/immunology , Animals , Antibodies/immunology , Apoptosis/drug effects , Apoptosis/immunology , Cell Line, Tumor , Cytochalasin D/pharmacology , Gangliosides/antagonists & inhibitors , Humans , Mice , Signal Transduction/drug effects , Signal Transduction/immunology
8.
J Cell Physiol ; 233(2): 866-879, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28145567

ABSTRACT

Neuroblastoma (NB) with various clinical presentation is a known childhood malignancy. Despite significant progress in treatment of NB afflicted patients, high risk disease is usually associated with poor outcome, resulting in long-term survival of less that 50%. Known as a disease most commonly originated form the nerve roots, the variants involved in NB imitation and progression remain to be elucidated. The outcome of low to intermediate risk disease is favorable whereas the high risk NB disease with dismal prognosis, positing the necessity of novel approaches for early detection and prognostication of advanced disease. Tailored immunotherapy approaches have shown significant improvement in high-risk NB patients. It has found a link between Gangliosides and progression of NB. The vast majority of neuroblastoma tumors express elevated levels of GD2, opening new insight into using anti-GD2 drugs as potential treatments for NBs. Implication of anti-GD2 monoclonal antibodies for treatment of high risk NBs triggers further investigation to unearth novel biomarkers as prognostic and response biomarker to guide additional multimodal tailored treatment approaches. A growing body of evidence supports the usefulness of miRNAs to evaluate high risk NBs response to anti-GD2 drugs and further prevent drug-related toxicities in refractory or recurrent NBs. miRNAs and circulating proteins in body fluids (plasma and serum) present as potential biomarkers in early detection of NBs. Here, we summarize various biomarkers involved in diagnosis, prognosis and response to treatment in patients with NB. We further attempted to overview prognostic biomarkers in response to treatment with anti-GD2 drugs.


Subject(s)
Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/blood , Brain Neoplasms/blood , Brain Neoplasms/drug therapy , Gangliosides/antagonists & inhibitors , Immunotherapy/methods , MicroRNAs/blood , Neuroblastoma/blood , Neuroblastoma/drug therapy , Animals , Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , Brain Neoplasms/immunology , Gangliosides/immunology , Humans , MicroRNAs/genetics , Molecular Diagnostic Techniques , Neuroblastoma/genetics , Neuroblastoma/immunology , Predictive Value of Tests , Treatment Outcome
9.
Apoptosis ; 23(9-10): 492-511, 2018 10.
Article in English | MEDLINE | ID: mdl-30027525

ABSTRACT

The process of autophagy and its role in survival of human neuroblastoma cell cultures was studied upon addition of an anti-GD2 ganglioside (GD2) 14G2a mouse monoclonal antibody (14G2a mAb) and an aurora A kinase specific inhibitor, MK-5108. It was recently shown that combination of these agents significantly potentiates cytotoxicity against IMR-32 and CHP-134 neuroblastoma cells in vitro, as compared to the inhibitor used alone. In this study we gained mechanistic insights on autophagy in the observed cytotoxic effects exerted by both agents using cytotoxicity assays, RT-qPCR, immunoblotting, and autophagy detection methods. Enhancement of the autophagy process in the 14G2a mAb- and MK-5108-treated IMR-32 cells was documented by assessing autophagic flux. Application of a lysosomotropic agent-chloroquine (CQ) affected the 14G2a mAb- and MK-5108-stimulated autophagic flux. It is our conclusion that the 14G2a mAb (40 µg/ml) and MK-5108 inhibitor (0.1 µM) induce autophagy in IMR-32 cells. Moreover, the combinatorial treatment of IMR-32 cells with the 14G2a mAb and CQ significantly potentiates cytotoxic effect, as compared to CQ used alone. Most importantly, we showed that interfering with autophagy at its early and late step augments the 14G2a mAb-induced apoptosis, therefore we can conclude that inhibition of autophagy is the primary mechanism of the CQ-mediated sensitization to the 14G2a mAb-induced apoptosis. Although, there was no virtual stimulation of autophagy in the 14G2a mAb-treated CHP-134 neuroblastoma cells, we were able to show that PHLDA1 protein positively regulates autophagy and this process exists in a mutually exclusive manner with apoptosis in PHLDA1-silenced CHP-134 cells.


Subject(s)
Apoptosis/genetics , Aurora Kinase A/genetics , Autophagy/genetics , Neuroblastoma/genetics , Transcription Factors/genetics , Animals , Antibodies, Monoclonal/pharmacology , Apoptosis/drug effects , Aurora Kinase A/antagonists & inhibitors , Autophagy/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclohexanecarboxylic Acids/pharmacology , Gangliosides/antagonists & inhibitors , Gangliosides/genetics , Humans , Mice , Neuroblastoma/pathology , Thiazoles/pharmacology , Transcription Factors/antagonists & inhibitors
10.
Pediatr Blood Cancer ; 65(6): e26967, 2018 06.
Article in English | MEDLINE | ID: mdl-29350486

ABSTRACT

BACKGROUND: The monoclonal anti-GD2 antibody ch14.18/CHO in combination with IL-2 is active and effective in high-risk neuroblastoma (NB) patients. Here, we investigated the inflammatory response and treatment tolerance of long-term infusion (LTI) of ch14.18/CHO (10 × 10 mg/m2 ; 24 hr) in combination with subcutaneous (s.c.) IL-2 in a single center program. METHODS: Fifty-three NB patients received up to six cycles of 100 mg/m2 ch14.18/CHO (d8-18, where d represents day(s)) as LTI combined with 6 × 106 IU/m2 s.c. IL-2 (d1-5; 8-12) and 160 mg/m2 oral 13-cis retinoic acid (RA) (d19-32). Side effects of ch14.18/CHO and IL-2 treatment require hospitalization of patients on d8. Treatment tolerance was evaluated daily with clinical parameters (body temperature, vital signs, Lansky performance status, requirement of i.v. concomitant medication) to define an outpatient candidate status. sIL-2-R and C-reactive protein values were determined to assess the inflammatory response. RESULTS: LTI of ch14.18/CHO (d8-18) in combination with s.c.IL-2 (d8-12) showed an acceptable treatment tolerance that allowed all patients to receive part of the treatment as an outpatient (median time point of discharge: d15 for all cycles). The treatment tolerance improved from cycle to cycle and the time to become an outpatient candidate decreased from d15 to d13 in subsequent cycles. Clinical and laboratory parameters indicate a maximum inflammatory response at d11 of each cycle. Interestingly, the soluble IL-2 receptor remained increased at baseline of the next cycle indicating immune activation over the entire treatment period of 6 months. CONCLUSIONS: LTI of ch14.18/CHO combined with s.c.IL-2 shows an improved tolerance in subsequent cycles allowing outpatient treatment.


Subject(s)
Antibodies, Monoclonal/adverse effects , Gangliosides/antagonists & inhibitors , Immunotherapy/adverse effects , Inflammation/pathology , Interleukin-2/adverse effects , Neuroblastoma/therapy , Adolescent , Adult , Antibodies, Monoclonal/administration & dosage , Child , Child, Preschool , Drug Therapy, Combination , Female , Follow-Up Studies , Gangliosides/immunology , Humans , Immune Tolerance , Infant , Inflammation/chemically induced , Infusions, Intravenous , Interleukin-2/administration & dosage , Male , Neuroblastoma/immunology , Neuroblastoma/pathology , Prognosis , Young Adult
11.
Chembiochem ; 18(13): 1146-1154, 2017 07 04.
Article in English | MEDLINE | ID: mdl-28295942

ABSTRACT

Gangliosides are acidic glycosphingolipids containing one or more sialic acid residues. They are essential compounds at the outer leaflet of the plasma membrane, where they interact with phospholipids, cholesterol, and transmembrane proteins, forming lipid rafts. They are involved in cell adhesion, proliferation, and recognition processes, as well as in the modulation of signal transduction pathways. These functions are mainly governed by the glycan moiety, and changes in the structures of gangliosides occur under pathological conditions, particularly in neuro-ectoderm-derived cancers. With the progress in mass spectrometry analysis of gangliosides, their role in cancer progression can be now investigated in more detail. In this review we summarize the current knowledge on the biosynthesis of gangliosides and their role in cancers, together with the recent development of cancer immunotherapy targeting gangliosides.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Gangliosides/antagonists & inhibitors , Membrane Microdomains/drug effects , Neoplasms/immunology , Animals , Carbohydrate Conformation , Carbohydrate Sequence , Cell Adhesion/drug effects , Cell Proliferation/drug effects , Disease Progression , Gangliosides/biosynthesis , Gangliosides/chemistry , Humans , Immunotherapy , Membrane Microdomains/chemistry , Membrane Microdomains/metabolism , Molecular Targeted Therapy , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/therapy , Sialic Acids/chemistry , Sialic Acids/metabolism , Sialyltransferases/antagonists & inhibitors , Sialyltransferases/genetics , Sialyltransferases/metabolism , Signal Transduction
12.
Chembiochem ; 18(13): 1194-1203, 2017 07 04.
Article in English | MEDLINE | ID: mdl-28374980

ABSTRACT

A new sialic acid (Sia)-containing glycopolymer-a fluorescent probe with high-density disialic acid (diSia) on the surface of polysaccharide dextran (diSia-Dex)-was synthesized as a key molecule to regulate the Sia recognition lectins, Siglecs, that are involved in the immune system. According to our original methods, diSia was synthesized by α-selective sialylation, and a dextran template possessing terminal acetylenes and amino groups was prepared. A diSia and a fluorescent molecule were subsequently introduced to surface-modified dextran by Hüisgen reaction and amidation, respectively. The modulatory activity of Siglec7 was evaluated by using synthetic probes. DiSia-Dex showed high binding avidity toward Siglec7, with a KD value of 5.87×10-10 m, and a high inhibitory activity for the interaction between Siglec7 and a ligand (GD3), with a IC50 value of 1.0 nm. Notably, diSia-Dex was able to release Siglec7 from the pre-existing Siglec7-GD3 complex, possibly due to its unique properties of a slow dissociation rate and a high association rate. Together, these data show that diSia-Dex can be widely applicable as a modulator of Siglec7 functions.


Subject(s)
Antigens, Differentiation, Myelomonocytic/chemistry , Dextrans/chemistry , Gangliosides/chemistry , Lectins/chemistry , Recombinant Fusion Proteins/chemistry , Sialic Acids/chemistry , Animals , Antigens, Differentiation, Myelomonocytic/genetics , Antigens, Differentiation, Myelomonocytic/metabolism , Binding Sites , CHO Cells , Carbohydrate Conformation , Carbohydrate Sequence , Chemistry Techniques, Synthetic , Cricetulus , Dextrans/chemical synthesis , Dextrans/metabolism , Dextrans/pharmacology , Fluorescent Dyes/chemistry , Gangliosides/antagonists & inhibitors , Gangliosides/metabolism , Gene Expression , Humans , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/metabolism , Immunoglobulin G/chemistry , Immunoglobulin G/genetics , Immunoglobulin G/metabolism , Kinetics , Lectins/antagonists & inhibitors , Lectins/genetics , Lectins/metabolism , Ligands , Molecular Docking Simulation , Protein Binding/drug effects , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sialic Acids/metabolism
13.
Pediatr Blood Cancer ; 62(2): 224-228, 2015 02.
Article in English | MEDLINE | ID: mdl-25382742

ABSTRACT

BACKGROUND: Addition of anti-GD2 antibody ch14.18 to the treatment of neuroblastoma has improved outcomes. The most common side effect of ch14.18 is neuropathic pain, which may in part be complement-mediated. Hu14.18K322A is a humanized anti-GD2 antibody designed to diminish complement activation and induce less pain. We compare the pain outcomes in patients treated with ch14.18 and those treated with hu14.18K322A, and explore dose-dependent relationships between pain scores, opioid requirements, and complement levels in patients treated with hu14.18K322A. PROCEDURE: Opioid (morphine equivalent mg/kg) and anxiolytic requirements during course 1 (4 days) in patients treated with hu14.18K322A and ch14.18 were reviewed. Correlations between antibody dose and pain scores, opioid requirements, and complement levels were examined for patients receiving hu14.18K322A. RESULTS: Patients treated with hu14.18K322A (n = 19) had lower opioid requirements than those who received ch14.18 (n = 9). The differences in median opioid requirements (mg/kg) were statistically significant for the overall course (1.57 vs. 2.41, P = 0.019) as well as for Days 3 (0.34 vs. 0.65, P = 0.005), and 4 (0.32 vs. 0.64, P = 0.010). No difference in anxiolytic use was observed between the two groups. In the group treated with hu14.18K322A, we found a positive correlation between antibody dose administered and pain scores, but no correlation between antibody dose and opioid requirements or changes in complement levels. CONCLUSIONS: In this retrospective analysis, hu14.18K322A induced less pain than ch14.18 based on opioid requirements. Pediatr Blood Cancer 2015;62:224-228. © 2014 Wiley Periodicals, Inc.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal/therapeutic use , Complement Inactivator Proteins/therapeutic use , Gangliosides/antagonists & inhibitors , Immunotherapy/methods , Neuralgia/drug therapy , Neuroblastoma/therapy , Adolescent , Child , Child, Preschool , Complement Activation/immunology , Complement System Proteins/drug effects , Female , Gangliosides/immunology , Humans , Infant , Male , Retrospective Studies , Treatment Outcome
14.
BMC Cancer ; 14: 295, 2014 Apr 28.
Article in English | MEDLINE | ID: mdl-24773917

ABSTRACT

BACKGROUND: Ganglioside GD2 is expressed on plasma membranes of various types of malignant cells. One of the most promising approaches for cancer immunotherapy is the treatment with monoclonal antibodies recognizing tumor-associated markers such as ganglioside GD2. It is considered that major mechanisms of anticancer activity of anti-GD2 antibodies are complement-dependent cytotoxicity and/or antibody-mediated cellular cytotoxicity. At the same time, several studies suggested that anti-GD2 antibodies are capable of direct induction of cell death of number of tumor cell lines, but it has not been investigated in details. In this study we investigated the functional role of ganglioside GD2 in the induction of cell death of multiple tumor cell lines by using GD2-specific monoclonal antibodies. METHODS: Expression of GD2 on different tumor cell lines was analyzed by flow cytometry using anti-GD2 antibodies. By using HPTLC followed by densitometric analysis we measured the amount of ganglioside GD2 in total ganglioside fractions isolated from tumor cell lines. An MTT assay was performed to assess viability of GD2-positive and -negative tumor cell lines treated with anti-GD2 mAbs. Cross-reactivity of anti-GD2 mAbs with other gangliosides or other surface molecules was investigated by ELISA and flow cytometry. Inhibition of GD2 expression was achieved by using of inhibitor for ganglioside synthesis PDMP and/or siRNA for GM2/GD2 and GD3 synthases. RESULTS: Anti-GD2 mAbs effectively induced non-classical cell death that combined features of both apoptosis and necrosis in GD2-positive tumor cells and did not affect GD2-negative tumors. Anti-GD2 mAbs directly induced cell death, which included alteration of mitochondrial membrane potential, induction of apoptotic volume decrease and cell membrane permeability. This cytotoxic effect was mediated exclusively by specific binding of anti-GD2 antibodies with ganglioside GD2 but not with other molecules. Moreover, the level of GD2 expression correlated with susceptibility of tumor cell lines to cytotoxic effect of anti-GD2 antibodies. CONCLUSIONS: Results of this study demonstrate that anti-GD2 antibodies not only passively bind to the surface of tumor cells but also directly induce rapid cell death after the incubation with GD2-positive tumor cells. These results suggest a new role of GD2 as a receptor that actively transduces death signal in malignant cells.


Subject(s)
Gangliosides/biosynthesis , Immunotherapy , Melanoma/immunology , Signal Transduction/genetics , Antibodies, Monoclonal/therapeutic use , Apoptosis/immunology , Cell Line, Tumor , Gangliosides/antagonists & inhibitors , Gangliosides/metabolism , Gene Expression Regulation, Neoplastic/immunology , Humans , Melanoma/metabolism , Melanoma/pathology , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/immunology , Signal Transduction/immunology
15.
J Pediatr Hematol Oncol ; 36(2): 152-5, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24065045

ABSTRACT

Nonpharmacologic, mind-body interventions are used to reduce anxiety in pediatric patients. Anti-ganglioside GD2 monoclonal antibody (anti-GD2 MoAb 3F8) therapy is the standard of care for high-risk neuroblastoma and pain is its major side effect. We performed a retrospective analysis of children undergoing anti-GD2 MoAb 3F8 treatment who received guided meditation. Meditation involved concentrating on the repetition of rhythmic, melodic sounds purported to slow breathing and induce a relaxation response. A total of 71% patients completed a session at first (n=19) or second attempt (n=5). Patients received fewer analgesic doses to manage anti-GD2 MoAb 3F8-induced pain when participating in meditation (n=17, mean=-0.4 dose, P<0.01). Mantram meditation is a feasible outpatient intervention associated with reduced analgesic requirements.


Subject(s)
Antineoplastic Agents/adverse effects , Meditation , Neuroblastoma/drug therapy , Pain/prevention & control , Pain/psychology , Adolescent , Antibodies, Monoclonal/adverse effects , Child , Child, Preschool , Female , Gangliosides/antagonists & inhibitors , Humans , Male , Meditation/methods , Meditation/psychology , Pain/etiology , Retrospective Studies
16.
Ann Pharmacother ; 47(2): 210-8, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23386066

ABSTRACT

OBJECTIVE: To systematically review clinical trials evaluating anti-disialoganglioside (GD2) antibodies in treating high-risk neuroblastoma in children. DATA SOURCES: A literature search was conducted in PubMed/MEDLINE, International Pharmaceutical Abstracts, and Cumulative Index of Nursing and Allied Health Literature (all searches 1990-August 2012) using the terms neuroblastoma, immunotherapy, 3F8, ch14.18, and hu14.18. Meeting abstracts presented between 1990 and 2012 from the American Society of Clinical Oncology, European Society for Medical Oncology, the American Society of Pediatric Hematology Oncology, Society of Surgical Oncology, and the American Society of Hematology were also evaluated. STUDY SELECTION AND DATA EXTRACTION: All completed and ongoing clinical trials of anti-GD2 antibodies in neuroblastoma were included. References from selected articles were also reviewed to identify additional citations. DATA SYNTHESIS: In 1999, the results of a Children's Cancer Group trial established that consolidation therapy after induction, surgery, and radiation should include purged autologous stem cell rescue followed by maintenance with isotretinoin. Overall survival at 7 years with this regimen remains below 30%. Over the following decade, antibodies targeting GD2, a surface antigen found on the surface of neuroblastoma cells, have emerged as a major therapeutic development for high-risk neuroblastoma. Anti-GD2 antibodies incite immune-mediated cytotoxicity toward neuroblastoma cells when given as monotherapy or in combination with cytokines such as sargramostim (granulocyte-macrophage colony-stimulating factor) or aldesleukin (interleukin-2). Responses to anti-GD2 agents appear most notable in patients with minimal residual disease following standard therapy. A chimeric preparation, ch14.18, is the only anti-GD2 antibody to be evaluated in a large controlled clinical trial, in which it demonstrated overall survival of 86% at 2 years in patients with high-risk neuroblastoma. Older nonrandomized studies of ch14.18 monotherapy and 3F8, a murine antibody, suggest this survival rate remains between 50 and 85% at 5 years posttreatment. CONCLUSIONS: Multiple GD2-specific monoclonal antibodies have been researched over the last decade in patients diagnosed with high-risk neuroblastoma. One anti-GD2 antibody, ch14.18, was found to significantly improve event-free and overall survival of high-risk neuroblastoma. Therefore, the standard approach to treating high-risk neuroblastoma is likely to undergo a major shift once an anti-GD2 antibody becomes commercially available.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Drugs, Investigational/therapeutic use , Gangliosides/antagonists & inhibitors , Neuroblastoma/drug therapy , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Child , Clinical Trials as Topic , Drugs, Investigational/administration & dosage , Drugs, Investigational/adverse effects , Drugs, Investigational/pharmacology , Gangliosides/metabolism , Humans , Neuroblastoma/metabolism , Risk , United States
18.
Pharm Res ; 28(9): 2261-72, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21594716

ABSTRACT

PURPOSE: To optimise and simplify preparation of targeted liposomes for efficient siRNA delivery to neuroblastoma, the most common solid tumour in early childhood. METHODS: Liposomes containing siRNA were prepared by combining the novel dual asymmetric centrifugation (DAC) method and the recently optimised sterol-based post-insertion technique (SPIT) to couple anti-GD2 antibody for selective interaction with neuroblastoma cells. Cultured human neuroblastoma cell lines were used to evaluate the efficiency of siRNA delivery. RESULTS: The size of liposomes prepared by DAC ranged from 190 to 240 nm; siRNA encapsulation efficiency was up to 50%. An average of 70 and 100 molecules of anti-GD2 antibody per particle were coupled. A significant association of liposomes with neuroblastoma cells as well as effective siRNA delivery was observed only when anti-GD2 antibody was coupled. Preliminary data suggest delivery of siRNA using anti-GD2-liposomes occurs via GD2-mediated endocytosis. Vascular endothelial growth factor A (VEGF-A) was down-regulated using siRNA delivered by anti-GD2-liposomes. CONCLUSIONS: DAC and SPIT allow for the straightforward preparation of liposomes for the targeted delivery of siRNA. Anti-GD2-liposomes thus produced can serve as versatile carriers of siRNA to neuroblastoma cells.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Gangliosides/antagonists & inhibitors , Gene Transfer Techniques , Neuroblastoma/therapy , RNA, Small Interfering/administration & dosage , Sterols/chemistry , Cell Line, Tumor , Centrifugation/methods , Drug Compounding/methods , Gangliosides/biosynthesis , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Neuroblastoma/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction
19.
Handb Exp Pharmacol ; (203): 165-78, 2011.
Article in English | MEDLINE | ID: mdl-21484572

ABSTRACT

A new concept "Life style-related diseases, such as type 2 diabetes, are a membrane microdomain disorder caused by aberrant expression of gangliosides" has arisen. By examining this working hypothesis, we demonstrate the molecular pathogenesis of type 2 diabetes and insulin resistance focusing on the interaction between insulin receptor and gangliosides in microdomains microdomains and propose the new therapeutic strategy "membrane microdomain ortho-signaling therapy".


Subject(s)
Diabetes Mellitus/drug therapy , Gangliosides/antagonists & inhibitors , Gangliosides/biosynthesis , Hypoglycemic Agents/pharmacology , Insulin Resistance/physiology , Animals , Biomarkers , Caveolae/physiology , Diabetes Mellitus, Type 2/etiology , G(M3) Ganglioside/metabolism , G(M3) Ganglioside/pharmacology , G(M3) Ganglioside/physiology , Gangliosides/metabolism , Humans , Hypoglycemic Agents/therapeutic use , Metabolic Syndrome/blood , Metabolic Syndrome/metabolism , Signal Transduction/physiology
20.
Front Immunol ; 12: 690467, 2021.
Article in English | MEDLINE | ID: mdl-34367149

ABSTRACT

Haploidentical stem cell transplantation (haplo SCT) in Stage IV neuroblastoma relapsed patients has been proven efficacious, while immunotherapy utilizing the anti-GD2 antibody dinutuximab beta has become a standard treatment for neuroblastoma. The combinatorial therapy of haplo SCT and dinutuximab may potentiate the efficacy of the immunotherapy. To gain further understanding of the synergistic effects, functional immunomonitoring was assessed during the clinical trial CH14.18 1021 Antibody and IL2 After haplo SCT in Children with Relapsed Neuroblastoma (NCT02258815). Rapid immune reconstitution of the lymphoid compartment was confirmed, with clinically relevant dinutuximab serum levels found in all patients over the course of treatment. Only one patient developed human anti-chimeric antibodies (HACAs). In-patient monitoring revealed highly functional NK cell posttransplant capable of antibody-dependent cellular cytotoxicity (ADCC). Degranulation of NK cell subsets revealed a significant response increased by dinutuximab. This was irrespective of the KIR receptor-ligand constellation within the NK subsets, defined by the major KIR receptors CD158a, CD158b, and CD158e. Moreover, complement-dependent cytotoxicity (CDC) was shown to be an extremely potent effector-cell independent mechanism of tumor cell lysis, with a clear positive correlation to GD2 expression on the cancer cells as well as to the dinutuximab concentrations. The ex vivo testing of patient-derived effector cells and the sera collected during dinutuximab therapy demonstrated both high functionality of the newly established lymphoid immune compartment and provided confidence that the antibody dosing regimen was sufficient over the duration of the dinutuximab therapy (up to nine cycles in a 9-month period). During the course of the dinutuximab therapy, proinflammatory cytokines and markers (sIL2R, TNFa, IL6, and C reactive protein) were significantly elevated indicating a strong anti-GD2 immune response. No impact of FcGR polymorphism on event-free and overall survival was found. Collectively, this study has shown that in-patient functional immunomonitoring is feasible and valuable in contributing to the understanding of anti-cancer combinatorial treatments such as haplo SCT and antibody immunotherapy.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Gangliosides/antagonists & inhibitors , Hematopoietic Stem Cell Transplantation , Monitoring, Immunologic , Neuroblastoma/therapy , Antibodies, Monoclonal/adverse effects , Antineoplastic Agents, Immunological/adverse effects , Cytokines/blood , Feasibility Studies , Gangliosides/immunology , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Inflammation Mediators/blood , Neoplasm Recurrence, Local , Neoplasm Staging , Neuroblastoma/blood , Neuroblastoma/immunology , Neuroblastoma/pathology , Predictive Value of Tests , Prospective Studies , Time Factors , Transplantation, Haploidentical , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL