Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.117
Filter
1.
Front Immunol ; 15: 1382931, 2024.
Article in English | MEDLINE | ID: mdl-38736882

ABSTRACT

Background: Neuroblastoma (NB) is characterized by both adrenergic (ADRN) and undifferentiated mesenchymal (MES) subsets. The ganglioside sialic acid-containing glycosphingolipid (GD2) is widely overexpressed on tumors of neuroectodermal origin promoting malignant phenotypes. MES cells are greatly enriched in post-therapy and relapsing tumors and are characterized by decreased expression of GD2. This event may cause failure of GD2-based immunotherapy. NK cells represent a key innate cell subset able to efficiently kill tumors. However, the tumor microenvironment (TME) that includes tumor cells and tumor-associated (TA) cells could inhibit their effector function. Methods: We studied eight NB primary cultures that, in comparison with commercial cell lines, more faithfully reflect the tumor cell characteristics. We studied four primary NB-MES cell cultures and two pairs of MES/ADRN (691 and 717) primary cultures, derived from the same patient. In particular, in the six human NB primary cultures, we assessed their phenotype, the expression of GD2, and the enzymes that control its expression, as well as their interactions with NK cells, using flow cytometry, RT-qPCR, and cytotoxicity assays. Results: We identified mature (CD105+/CD133-) and undifferentiated (CD133+/CD105-) NB subsets that express high levels of the MES transcripts WWTR1 and SIX4. In addition, undifferentiated MES cells display a strong resistance to NK-mediated killing. On the contrary, mature NB-MES cells display an intermediate resistance to NK-mediated killing and exhibit some immunomodulatory capacities on NK cells but do not inhibit their cytolytic activity. Notably, independent from their undifferentiated or mature phenotype, NB-MES cells express GD2 that can be further upregulated in undifferentiated NB-MES cells upon co-culture with NK cells, leading to the generation of mature mesenchymal GD2bright neuroblasts. Concerning 691 and 717, they show high levels of GD2 and resistance to NK cell-mediated killing that can be overcome by the administration of dinutuximab beta, the anti-GD2 monoclonal antibody applied in the clinic. Conclusions: NB is a heterogeneous tumor representing a further hurdle in NB immunotherapy. However, different from what was reported with NB commercial cells and independent of their MES/ADRN phenotype, the expression of GD2 and its displayed sensitivity to anti-GD2 mAb ADCC indicated the possible effectiveness of anti-GD2 immunotherapy.


Subject(s)
Gangliosides , Killer Cells, Natural , Neuroblastoma , Tumor Escape , Tumor Microenvironment , Humans , Neuroblastoma/immunology , Neuroblastoma/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Gangliosides/immunology , Gangliosides/metabolism , Tumor Microenvironment/immunology , Cell Line, Tumor , Cytotoxicity, Immunologic , Tumor Cells, Cultured , Mesenchymal Stem Cells/immunology , Mesenchymal Stem Cells/metabolism
2.
Brain Nerve ; 76(5): 508-514, 2024 May.
Article in Japanese | MEDLINE | ID: mdl-38741489

ABSTRACT

Fisher syndrome is recognized as a variant of Guillain-Barré syndrome, encompassing acute onset immune-mediated neuropathies marked by the classical triad of ataxia, areflexia, and ophthalmoplegia. Generally, Fisher syndrome follows a self-limited course with a good prognosis. Ophthalmoplegia, typically bilateral, progresses to complete external ophthalmoplegia within 1-2 weeks. Ataxia, often very severe, may cause an inability to walk without support despite normal strength. Fisher syndrome is also frequently concomitant with additional clinical features, including ptosis, internal ophthalmoplegia, facial nerve palsy, sensory deficits, and bulbar palsy. The confirmation of an antecedent infection is often established. Among the ganglioside antibodies, anti-GQ1b antibodies exhibit positivity in over 80% of patients. The syndrome manifests in three distinct types: a partial subtype exhibiting only a subset of the triad symptoms, Bickerstaff's brainstem encephalitis marked by impaired consciousness and pyramidal tract signs, and an overlapping subtype with Guillain-Barré syndrome, characterized by weakness in the extremities.


Subject(s)
Miller Fisher Syndrome , Humans , Miller Fisher Syndrome/diagnosis , Miller Fisher Syndrome/immunology , Miller Fisher Syndrome/therapy , Miller Fisher Syndrome/physiopathology , Gangliosides/immunology , Prognosis , Guillain-Barre Syndrome/diagnosis , Guillain-Barre Syndrome/therapy
3.
Brain Behav ; 14(5): e3514, 2024 May.
Article in English | MEDLINE | ID: mdl-38698593

ABSTRACT

BACKGROUND: There have been multiple reports about the occurrence of dysphagia after the contraction of coronavirus disease 2019 (COVID-19). However, a detailed pathology and epidemiologic relation between COVID-19 infection and dysphagia have yet to be established. Here, we report three cases of unexplained dysphagia after COVID-19 diagnosis, with atypical clinical presentations. CASE REPORT: All patients showed severe isolated lower cranial nerve involvement with dysphagia and aspiration, which required full tube feeding but showed no evidence of limb weakness or sensory symptoms. All tested positive for anti-ganglioside antibody tests, which all commonly (GD1b, GM1, and GQ1b) are known to have terminal NeuNAc(α2-3)Gal epitope. DISCUSSION: We report a series of cases featuring severe, isolated dysphagia post-COVID-19 infection, concomitant with positive anti-ganglioside antibodies. One potential etiology is a variant of Guillain-Barré syndrome. Because only isolated dysphagia with sparing of the facial and extraocular muscles was evident in these cases, we explore the association between anti-ganglioside antibodies specific to NeuNAc(α2-3)Gal, which has been frequently associated with the development of bulbar dysfunction. Given that NeuNAc(α2-3)Gal exhibits an affinity for the spike glycoprotein of SARS-CoV-2, a cross-reaction against NeuNAc(α2-3)Gal may possibly contribute to isolated dysphagia following COVID-19 infection.


Subject(s)
COVID-19 , Deglutition Disorders , Gangliosides , Humans , COVID-19/complications , COVID-19/immunology , Deglutition Disorders/etiology , Male , Gangliosides/immunology , Middle Aged , Female , Aged , SARS-CoV-2/immunology , Autoantibodies/blood , Guillain-Barre Syndrome/immunology
4.
J Immunother Cancer ; 12(5)2024 May 15.
Article in English | MEDLINE | ID: mdl-38754916

ABSTRACT

BACKGROUND: Chimeric antigen receptor (CAR) T cell therapies specific for the CD19 and B-cell maturation antigen have become an approved standard of care worldwide for relapsed and refractory B-cell malignancies. If CAR-T cell therapy for non-hematological malignancies is to achieve the same stage of clinical development, then iterative early-phase clinical testing can add value to the clinical development process for evaluating CAR-T cell products containing different CAR designs and manufactured under differing conditions. METHODS: We conducted a phase 1 trial of third-generation GD2-specific CAR-T cell therapy, which has previously been tested in neuroblastoma patients. In this study, the GD2-CAR-T therapy was evaluated for the first time in metastatic melanoma patients in combination with BRAF/MEK inhibitor therapy, and as a monotherapy in patients with colorectal cancer and a patient with fibromyxoid sarcoma. Feasibility and safety were determined and persistence studies, multiplex cytokine arrays on sera and detailed immune phenotyping of the original CAR-T products, the circulating CAR-T cells, and, in select patients, the tumor-infiltrating CAR-T cells were performed. RESULTS: We demonstrate the feasibility of manufacturing CAR-T products at point of care for patients with solid cancer and show that a single intravenous infusion was well tolerated with no dose-limiting toxicities or severe adverse events. In addition, we note significant improvements in CAR-T cell immune phenotype, and expansion when a modified manufacturing procedure was adopted for the latter 6 patients recruited to this 12-patient trial. We also show evidence of CAR-T cell-mediated immune activity and in some patients expanded subsets of circulating myeloid cells after CAR-T cell therapy. CONCLUSIONS: This is the first report of third-generation GD2-targeting CAR-T cells in patients with metastatic melanoma and other solid cancers such as colorectal cancer, showing feasibility, safety and immune activity, but limited clinical effect. TRIAL REGISTRATION NUMBER: ACTRN12613000198729.


Subject(s)
Immunotherapy, Adoptive , Melanoma , Receptors, Chimeric Antigen , Humans , Melanoma/immunology , Melanoma/therapy , Immunotherapy, Adoptive/methods , Immunotherapy, Adoptive/adverse effects , Receptors, Chimeric Antigen/immunology , Male , Female , Middle Aged , Gangliosides/immunology , Adult , Aged , T-Lymphocytes/immunology , Treatment Outcome
6.
JAMA Oncol ; 9(2): 242-250, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36547975

ABSTRACT

Importance: Among patients with high-risk relapsed metastatic neuroblastoma, oral ß-glucan adjuvant during GD2/GD3 ganglioside vaccine boost has stimulated IgG antibody response, which was associated with improved survival; however, the effectiveness of oral ß-glucan during the vaccine priming phase remains unproven. Objective: To isolate the adjuvant effect of oral ß-glucan on antibody response to GD2/GD3 ganglioside vaccine in patients with high-risk neuroblastoma. Design, Setting, and Participants: In this phase 2 randomized clinical trial, enrolled patients with high-risk neuroblastoma were randomized to 2 groups to receive the GD2/GD3 vaccine at a large cancer center in a major metropolitan area from October 2018 to September 2020. Data were analyzed from October 7, 2021, to February 28, 2022. Interventions: Eligible patients receiving GD2/GD3 vaccine were randomly assigned to group 1 (n = 54) to receive no ß-glucan or group 2 (n = 53) to receive an oral ß-glucan regimen during the first 5 weeks of vaccine priming. From week 6 onwards, all 107 patients received oral ß-glucan during vaccine boost for 1 year or until disease progression. Main Outcomes and Measures: Primary end point was comparison of anti-GD2 IgG1 response before vaccine injection 6 (week 32) in group 1 vs group 2. Seroconversion rate and the association of antibody titer with ß-glucan receptor dectin-1 single nucleotide polymorphism (SNP) rs3901533 were also assessed. Results: In all, 107 patients with high-risk neuroblastoma were randomized to the 2 groups: 54 patients (median [range] age, 5.2 [1.0-17.3] years; 28 [52%] male and 26 [48%] female) in group 1; and 53 patients (median [range] age, 6.2 [1.9-18.4] years; 25 [47%] male and 28 [53%] female) in group 2; both groups were also comparable in their first remission status at study entry (70% vs 70%). Adding oral ß-glucan during the first 5 weeks of vaccine priming elicited a higher anti-GD2 IgG1 antibody response in group 2 (1.80; 90% CI, 0.12-3.39; P = .08; planned type I error, 0.10). Anti-GD2 IgG1 titer of 230 ng/mL or greater by week 8 was associated with statistically favorable PFS. Antibody titer correlated significantly with dectin-1 SNP. The genotype frequency, seroconversion rates, and vaccine-related toxic effects were similar in the 2 groups. Conclusions and Relevance: This phase 2 randomized clinical trial found that adding oral ß-glucan during vaccine priming increased anti-GD2 IgG1 titer among genetic responders without added toxic effects. Because responder dectin-1 SNP was identical in the 2 randomized groups, no difference was detected in seroconversion rates. Alternative or additional adjuvants may be needed to enhance seroconversion. Trial Registration: ClinicalTrials.gov Identifier: NCT00911560.


Subject(s)
Cancer Vaccines , Gangliosides , Neuroblastoma , beta-Glucans , Child , Child, Preschool , Female , Humans , Male , Antibody Formation , beta-Glucans/pharmacology , beta-Glucans/therapeutic use , Gangliosides/immunology , Gangliosides/therapeutic use , Immunoglobulin G , Neuroblastoma/therapy , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use
7.
Nature ; 603(7903): 934-941, 2022 03.
Article in English | MEDLINE | ID: mdl-35130560

ABSTRACT

Diffuse intrinsic pontine glioma (DIPG) and other H3K27M-mutated diffuse midline gliomas (DMGs) are universally lethal paediatric tumours of the central nervous system1. We have previously shown that the disialoganglioside GD2 is highly expressed on H3K27M-mutated glioma cells and have demonstrated promising preclinical efficacy of GD2-directed chimeric antigen receptor (CAR) T cells2, providing the rationale for a first-in-human phase I clinical trial (NCT04196413). Because CAR T cell-induced brainstem inflammation can result in obstructive hydrocephalus, increased intracranial pressure and dangerous tissue shifts, neurocritical care precautions were incorporated. Here we present the clinical experience from the first four patients with H3K27M-mutated DIPG or spinal cord DMG treated with GD2-CAR T cells at dose level 1 (1 × 106 GD2-CAR T cells per kg administered intravenously). Patients who exhibited clinical benefit were eligible for subsequent GD2-CAR T cell infusions administered intracerebroventricularly3. Toxicity was largely related to the location of the tumour and was reversible with intensive supportive care. On-target, off-tumour toxicity was not observed. Three of four patients exhibited clinical and radiographic improvement. Pro-inflammatory cytokine levels were increased in the plasma and cerebrospinal fluid. Transcriptomic analyses of 65,598 single cells from CAR T cell products and cerebrospinal fluid elucidate heterogeneity in response between participants and administration routes. These early results underscore the promise of this therapeutic approach for patients with H3K27M-mutated DIPG or spinal cord DMG.


Subject(s)
Astrocytoma , Brain Stem Neoplasms , Gangliosides , Glioma , Histones , Immunotherapy, Adoptive , Mutation , Receptors, Chimeric Antigen , Astrocytoma/genetics , Astrocytoma/immunology , Astrocytoma/pathology , Astrocytoma/therapy , Brain Stem Neoplasms/genetics , Brain Stem Neoplasms/immunology , Brain Stem Neoplasms/pathology , Brain Stem Neoplasms/therapy , Child , Gangliosides/immunology , Gene Expression Profiling , Glioma/genetics , Glioma/immunology , Glioma/pathology , Glioma/therapy , Histones/genetics , Humans , Immunotherapy, Adoptive/methods , Receptors, Chimeric Antigen/immunology , Spinal Cord Neoplasms/genetics , Spinal Cord Neoplasms/immunology , Spinal Cord Neoplasms/pathology , Spinal Cord Neoplasms/therapy
8.
J Immunother Cancer ; 10(1)2022 01.
Article in English | MEDLINE | ID: mdl-35086947

ABSTRACT

BACKGROUND: Tumorous heterogeneity is a hallmark of tumor evolution and cancer progression, being a longstanding challenge to targeted immunotherapy. Ex vivo armed T cells (EATs) using IgG-(L)-scFv bispecific antibodies (BsAbs) are potent tumor-specific cytotoxic effectors. To improve the anti-tumor efficacy of EATs against heterogeneous solid tumors, we explored multi-antigen targeting approaches. METHODS: Ex vivo expanded T cells were armed with BsAbs built on the IgG-(L)-scFv platform, where an anti-CD3 (huOKT3) scFv was attached to the carboxyl end of both light chains of a tumor specific IgG. Multispecificity was created by combining monospecific EATs, combining BsAbs on the same T cell, or combining specificities on the same antibody. Three multi-antigens targeting EAT strategies were tested: (1) pooled-EATs (EATs each with unique specificity administered simultaneously) or alternate-EATs (EATs each with unique specificity administered in an alternating schedule), (2) dual-EATs or multi-EATs (T cells simultaneously armed with ≥2 BsAbs), and (3) TriAb-EATs (T cells armed with BsAb specific for two targets besides CD3 (TriAb)). The properties and efficiencies of these three strategies were evaluated by flow cytometry, in vitro cytotoxicity, cytokine release assays, and in vivo studies performed in BALB-Rag2-/-IL-2R-γc-KO (BRG) mice xenografted with cancer cell line (CDX) or patient-derived tumor (PDX). RESULTS: Multi-EATs retained target antigen specificity and anti-tumor potency. Cytokine release with multi-EATs in the presence of tumor cells was substantially less than when multiple BsAbs were mixed with unarmed T cells. When tested against CDXs or PDXs, dual-EATs or multi-EATs effectively suppressed tumor growth without clinical toxicities. Most importantly, dual-EATs or multi-EATs were highly efficient in preventing clonal escape while mono-EATs or TriAb- EATs were not as effective. CONCLUSIONS: Multi-EATs have the potential to increase potency, reduce toxicity, and overcome tumor heterogeneity without excessive cytokine release. Arming T cells with multiple BsAbs deserves further exploration to prevent or to treat cancer resistance.


Subject(s)
Antibodies, Bispecific/therapeutic use , Gangliosides/immunology , Immunotherapy/methods , Neoplasms/therapy , Receptor, ErbB-2/immunology , T-Lymphocytes/immunology , Animals , Cell Line, Tumor , Cytokines/biosynthesis , Female , Humans , Mice , Mice, Inbred BALB C , Neoplasms/immunology , Neoplasms/pathology , Xenograft Model Antitumor Assays
9.
Cancer Immunol Immunother ; 71(1): 153-164, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34043024

ABSTRACT

Neuroblastoma (NBL) accounts for a disproportionate number of deaths among childhood malignancies despite intensive multimodal therapy that includes antibody targeting disialoganglioside GD2, a NBL antigen. Unfortunately, resistance to anti-GD2 immunotherapy is frequent and we aimed to investigate mechanisms of resistance in NBL. GD2 expression was quantified by flow cytometry and anti-GD2 antibody internalization was measured using real-time microscopy in 20 human NBL cell lines. Neutrophil-mediated antibody-dependent cellular cytotoxicity (ADCC) assays were performed on a subset of the cell lines (n = 12), and results were correlated with GD2 expression and antibody internalization. GD2 was expressed on 19 of 20 NBL cell lines at variable levels, and neutrophil-mediated ADCC was observed only in GD2-expressing cell lines. We found no correlation between level of GD2 expression and sensitivity to neutrophil-mediated ADCC, suggesting that GD2 expression of many cell lines was above a threshold required for maximal ADCC, such that expression level could not be used to predict subsequent cytotoxicity. Instead, anti-GD2 antibody internalization, a process that occurred universally but differentially across GD2-expressing NBL cell lines, was inversely correlated with ADCC. Treatment with endocytosis inhibitors EIPA, chlorpromazine, MBCD, and cytochalasin-D showed potential to inhibit antibody internalization; however, only MBCD resulted in significantly increased sensitivity to neutrophil-mediated ADCC in 4 of 4 cell lines in vitro. Our data suggest that antibody internalization may represent a novel mechanism of immunotherapy escape by NBL and provide proof-of-principle that targeting pathways involved in antibody internalization may improve the efficacy of anti-GD2 immunotherapies.


Subject(s)
Antibodies/chemistry , Drug Resistance , Gangliosides/chemistry , Immunotherapy/methods , Neuroblastoma/immunology , Neuroblastoma/therapy , Antibodies, Monoclonal/pharmacology , Antibody-Dependent Cell Cytotoxicity/immunology , Cell Line, Tumor , Endocytosis , Flow Cytometry , Gangliosides/immunology , Humans , Immunologic Factors , Killer Cells, Natural/immunology , Neutrophils/metabolism
11.
Front Immunol ; 12: 668307, 2021.
Article in English | MEDLINE | ID: mdl-34489927

ABSTRACT

Management for high-risk neuroblastoma (NBL) has included autologous hematopoietic stem cell transplant (HSCT) and anti-GD2 immunotherapy, but survival remains around 50%. The aim of this study was to determine if allogeneic HSCT could serve as a platform for inducing a graft-versus-tumor (GVT) effect against NBL with combination immunocytokine and NK cells in a murine model. Lethally irradiated C57BL/6 (B6) x A/J recipients were transplanted with B6 bone marrow on Day +0. On day +10, allogeneic HSCT recipients were challenged with NXS2, a GD2+ NBL. On days +14-16, mice were treated with the anti-GD2 immunocytokine hu14.18-IL2. In select groups, hu14.18-IL2 was combined with infusions of B6 NK cells activated with IL-15/IL-15Rα and CD137L ex vivo. Allogeneic HSCT alone was insufficient to control NXS2 tumor growth, but the addition of hu14.18-IL2 controlled tumor growth and improved survival. Adoptive transfer of ex vivo CD137L/IL-15/IL-15Rα activated NK cells with or without hu14.18-IL2 exacerbated lethality. CD137L/IL-15/IL-15Rα activated NK cells showed enhanced cytotoxicity and produced high levels of TNF-α in vitro, but induced cytokine release syndrome (CRS) in vivo. Infusing Perforin-/- CD137L/IL-15/IL-15Rα activated NK cells had no impact on GVT, whereas TNF-α-/- CD137L/IL-15/IL-15Rα activated NK cells improved GVT by decreasing peripheral effector cell subsets while preserving tumor-infiltrating lymphocytes. Depletion of Ly49H+ NK cells also improved GVT. Using allogeneic HSCT for NBL is a viable platform for immunocytokines and ex vivo activated NK cell infusions, but must be balanced with induction of CRS. Regulation of TNFα or activating NK subsets may be needed to improve GVT effects.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Cytokines/pharmacology , Gangliosides/antagonists & inhibitors , Graft vs Tumor Effect , Hematopoietic Stem Cell Transplantation , Immunotherapy, Adoptive , Killer Cells, Natural/drug effects , Killer Cells, Natural/transplantation , Lymphocyte Activation/drug effects , Neuroblastoma/therapy , Animals , Cell Line, Tumor , Combined Modality Therapy , Female , Gangliosides/immunology , Gangliosides/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Neuroblastoma/immunology , Neuroblastoma/metabolism , Neuroblastoma/pathology
12.
Angew Chem Int Ed Engl ; 60(45): 24179-24188, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34469031

ABSTRACT

Ganglioside GD2 is an attractive tumor-associated carbohydrate antigen for anti-cancer vaccine development. However, its low immunogenicity and the significant side effects observed with anti-GD2 antibodies present significant obstacles for vaccines. To overcome these, a new GD2 derivative bearing an N-acetamide (NHAc) at its non-reducing end neuraminic acid (9NHAc-GD2) has been designed to mimic the 9-O-acetylated-GD2 (9OAc-GD2), a GD2 based antigen with a restricted expression on tumor cells. 9NHAc-GD2 was synthesized efficiently via a chemoenzymatic method and subsequently conjugated with a powerful carrier bacteriophage Qß. Mouse immunization with the Qß-9NHAc-GD2 conjugate elicited strong and long-lasting IgG antibodies, which were highly selective toward 9NHAc-GD2 with little cross-recognition of GD2. Immunization of canines with Qß-9NHAc-GD2 showed the construct was immunogenic in canines with little adverse effects, paving the way for future clinical translation to humans.


Subject(s)
Cancer Vaccines/chemistry , Gangliosides/chemical synthesis , Vaccines, Conjugate/chemistry , Acetamides/chemistry , Acetamides/immunology , Acetylation , Animals , Cancer Vaccines/immunology , Carbohydrate Conformation , Gangliosides/chemistry , Gangliosides/immunology , Hydrolysis , Mice , Neuraminic Acids/chemistry , Neuraminic Acids/immunology , Vaccine Development , Vaccines, Conjugate/immunology
13.
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
14.
BMC Neurol ; 21(1): 212, 2021 May 28.
Article in English | MEDLINE | ID: mdl-34049504

ABSTRACT

BACKGROUND: Graves' disease and anti-GQ1b antibody syndrome are both autoimmune diseases, and there have been few reports on whether there is a correlation between the two. In this study, we present the case of a woman who was diagnosed with Graves' disease and anti-GQ1b antibody syndrome in succession. CASE PRESENTATION: The chief complaints of this patient were limb weakness and blurred vision. Graves' disease was diagnosed by examination of thyroid function and thyroid autoantibodies, but the clinical symptoms were not relieved after antihyperthyroidism treatment. Finally, it was found that Graves' disease was complicated by anti-GQ1b antibody syndrome, and the symptoms were relieved after treatment with glucocorticoids and intravenous immunoglobulin. We also explored the possible mechanism of these diseases through a literature review. CONCLUSIONS: We report a rare case of the cooccurrence of Graves' disease and anti-GQ1b antibody syndrome. Immune dysregulation might be the pathogenesis of the association, but there is no precise supporting evidence, and more research is needed.


Subject(s)
Autoantibodies , Autoimmune Diseases , Gangliosides/immunology , Graves Disease , Autoantibodies/blood , Autoantibodies/immunology , Female , Humans
15.
Clin Neurol Neurosurg ; 206: 106691, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34022690

ABSTRACT

Miller-Fisher syndrome (MFS) together with Guillan-Barré syndrome (GBS) and Bickerstaff brainstem encephalitis (BBE) are considered to form a continuous clinical spectrum of the same disease, possibly affecting the peripheral and/or central nervous systems, with monophasic symptoms. The frequency of overlapping clinical signs and the risk of recurrence are independent and very low, but no cases of GQ1b-seropositive recurrent MFS overlapping with GBS and BBE have been described so far. Here, we describe for the first time an atypical case of recurrent GQ1b-seropositive MFS overlapping GBS and BBE, 12 years after a previous GQ1b-seronegative typical MFS episode. Our case expands the clinical spectrum of recurrent MFS, and it should prompt clinicians to investigate the presence of anti-ganglioside antibodies in recurrent MFS even when these were negative in the previous episode, especially in those presenting with overlapping spectrum symptoms and a critically ill picture during the second episode.


Subject(s)
Encephalitis , Guillain-Barre Syndrome , Miller Fisher Syndrome , Adult , Autoantibodies/immunology , Autoantigens/immunology , Brain Stem , Encephalitis/immunology , Encephalitis/physiopathology , Female , Gangliosides/immunology , Guillain-Barre Syndrome/immunology , Guillain-Barre Syndrome/physiopathology , Humans , Miller Fisher Syndrome/immunology , Miller Fisher Syndrome/physiopathology , Recurrence
16.
J Immunother Cancer ; 9(4)2021 04.
Article in English | MEDLINE | ID: mdl-33858849

ABSTRACT

An important component of research using animal models is ensuring rigor and reproducibility. This study was prompted after two experimenters performing virtually identical studies obtained different results when syngeneic B78 murine melanoma cells were implanted into the skin overlying the flank and treated with an in situ vaccine (ISV) immunotherapy. Although both experimenters thought they were using identical technique, we determined that one was implanting the tumors intradermally (ID) and the other was implanting them subcutaneously (SC). Though the baseline in vivo immunogenicity of tumors can depend on depth of their implantation, the response to immunotherapy as a function of tumor depth, particularly in immunologically 'cold' tumors, has not been well studied. The goal of this study was to evaluate the difference in growth kinetics and response to immunotherapy between identically sized melanoma tumors following ID versus SC implantation. We injected C57BL/6 mice with syngeneic B78 melanoma cells either ID or SC in the flank. When tumors reached 190-230 mm3, they were grouped into a 'wave' and treated with our previously published ISV regimen (12 Gy local external beam radiation and intratumoral hu14.18-IL2 immunocytokine). Physical examination demonstrated that ID-implanted tumors were mobile on palpation, while SC-implanted tumors became fixed to the underlying fascia. Histologic examination identified a critical fascial layer, the panniculus carnosus, which separated ID and SC tumors. SC tumors reached the target tumor volume significantly faster compared with ID tumors. Most ID tumors exhibited either partial or complete response to this immunotherapy, whereas most SC tumors did not. Further, the 'mobile' or 'fixed' phenotype of tumors predicted response to therapy, regardless of intended implantation depth. These findings were then extended to additional immunotherapy regimens in four separate tumor models. These data indicate that the physical 'fixed' versus 'mobile' characterization of the tumors may be one simple method of ensuring homogeneity among implanted tumors prior to initiation of treatment. Overall, this short report demonstrates that small differences in depth of tumor implantation can translate to differences in response to immunotherapy, and proposes a simple physical examination technique to ensure consistent tumor depth when conducting implantable tumor immunotherapy experiments.


Subject(s)
Antibodies/administration & dosage , Cancer Vaccines/administration & dosage , Immunotherapy , Interleukin-2/administration & dosage , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Soft Tissue Neoplasms/drug therapy , Animals , Antibodies/immunology , Cancer Vaccines/immunology , Cell Line, Tumor , Female , Gangliosides/immunology , Injections, Intralesional , Interleukin-2/immunology , Kinetics , Melanoma/genetics , Melanoma/immunology , Melanoma/pathology , Mice, Inbred C57BL , Neoplasm Transplantation , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/immunology , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Soft Tissue Neoplasms/genetics , Soft Tissue Neoplasms/immunology , Soft Tissue Neoplasms/pathology , Transplantation, Isogeneic , Tumor Burden/drug effects , Vaccination
17.
Rinsho Shinkeigaku ; 61(5): 305-309, 2021 May 19.
Article in Japanese | MEDLINE | ID: mdl-33867411

ABSTRACT

A 35-year-old male developed sensory abnormality of peripheral limbs and oral cavity after prior infection with diarrhea and cold symptoms. Hyperrhinolalia, nasopharyngeal reflux, double vision, and wobbling in walking rapidly progressed. Neurological examination revealed palatoplegia, omnidirectional ophthalmoplegia, hyperreflexia, sensory disturbance of extremities, and truncal and limb ataxia due to decreased deep sensation. A peripheral nerve conduction study found a slight decrease in sensory nerve action potential of the median nerve and a decrease in F wave frequency of the median nerve. Serum IgM-CMV antibody was positive on admission. After IVIg therapy, palatoplegia and ataxia markedly improved. In this case, GalNAc-GD1a and GM2 antibodies, which are often detected after CMV infection, were positive in addition to the GT1a and GQ1b antibodies, and it was assumed that these findings were associated with the palatoplegia, which is included in cranial nerve palsy. Pathophysiologically, the present case is considered to be an overlap with acute oropharyngeal palsy (AOP), which is a rare subtype of Guillain-Barre syndrome, and Fisher syndrome (FS). The clinical aspects of the present case suggest a continuous spectrum between AOP and FS.


Subject(s)
Cranial Nerve Diseases/etiology , Cytomegalovirus Infections/complications , Miller Fisher Syndrome/etiology , Adult , Antibodies, Viral/blood , Autoantibodies/blood , Biomarkers/blood , Cranial Nerve Diseases/diagnosis , Cranial Nerve Diseases/therapy , Cytomegalovirus/immunology , Cytomegalovirus Infections/diagnosis , Diagnostic Techniques, Neurological , Disease Progression , Gangliosides/immunology , Humans , Immunoglobulin M/blood , Immunoglobulins, Intravenous/administration & dosage , Male , Median Nerve/physiopathology , Miller Fisher Syndrome/diagnosis , Neural Conduction
18.
Mol Pharm ; 18(6): 2406-2415, 2021 06 07.
Article in English | MEDLINE | ID: mdl-33896187

ABSTRACT

Gangliosides (glycosphingolipids) reduce antibody production by inhibiting B-cell receptor (BCR) signaling. We have shown that a copresentation of gangliosides and polyethylene glycol (PEG) on the same liposomes suppresses anti-PEG IgM production in mice. In addition, we recently observed that pDNA incorporated in PEGylated cationic liposomes (PCLs) induces anti-DNA IgM, which could be a hurdle to the development of efficient gene delivery systems. Therefore, the focus of this study was to determine if the copresentation of gangliosides and DNA on the same PCL would suppress antibody production against DNA. PCLs including DNA induced both anti-PEG IgM production and anti-DNA IgM production. The extent of anti-PEG and anti-DNA IgM production was likely dependent on the immunogenicity of the complexed DNA. Treatment of clodronate-containing liposomes, which causes a depletion of phagocytic cells, suppressed anti-PEG IgM production from PCLs that did not include DNA but failed to suppress anti-PEG IgM production from PCLs that complexed DNA (PCLD). Both anti-PEG IgM and anti-DNA IgM was induced in T-cell-deficient nude mice as well as in normal mice following treatment with PCLs and PCLD, respectively. These results indicate that phagocytic cells contribute to anti-PEG IgM production but not to anti-DNA IgM production, while T-cells do not contribute to any form of antibody production. The copresentation of gangliosides and DNA significantly reduced anti-PEG IgM production but unfortunately did not reduce anti-DNA IgM production. It appears that the immunosuppressive effect of gangliosides, presumably via the CD22 signaling pathway, is limited only to anti-PEG immunity.


Subject(s)
Clodronic Acid/administration & dosage , DNA/immunology , Gangliosides/immunology , Gene Transfer Techniques/adverse effects , Immunoglobulin M/metabolism , Animals , Antibody Formation , Cations , Gangliosides/chemistry , Genetic Therapy/methods , Liposomes , Male , Mice , Phagocytes/drug effects , Phagocytes/immunology , Phagocytes/metabolism , Plasmids/administration & dosage , Plasmids/genetics , Polyethylene Glycols/chemistry
19.
Rev Neurol ; 72(6): 203-212, 2021 03 16.
Article in English, Spanish | MEDLINE | ID: mdl-33710610

ABSTRACT

INTRODUCTION: The coronavirus disease 2019 (COVID-19) pandemic is a major worldwide health disorder. There is an increasing number of neurological complications recognized with COVID-19 including patients with GBS and its variants. DEVELOPMENT: A review of the clinical cases of GBS associated to COVID-19 infection published in the last months has been developed. We included 48 patients (31 men, mean age 56.4 years). The most common COVID-19 symptoms were cough (60.4%) and fever (56.3%). Mean time from COVID-19 symptoms to neurologic manifestations was 12.1 days, but in nine patients (18.8%) developed GBS within seven days. Eleven patients (22.9%) presented cranial nerve involvement in the absence of muscle weakness; 36 presented the classic sensory motor variant (75%) and one had a pure motor variant (2.1%). The electrodiagnostic pattern was considered demyelinating in 82.4% of the generalized variants. The presence of hyposmia/dysgeusia was associated with a latency shorter than seven days to GBS onset of symptoms (30% vs 15.6%), and cranial nerve involvement in the absence of weakness (30.8% vs 17.1%). Most patients (87.5%) were treated with intravenous immunoglobulin. Neurological outcome was favorable in 64.6%; 29.2% had respiratory failure and 4.2% died shortly after being admitted. CONCLUSIONS: GBS in patients with SARS-CoV-2 infection resembles clinically and electrophysiology the classical forms. Further studies are necessary to understand whether GBS frequency is actually increased due to SARS-CoV-2 infection and explore pathogenic mechanisms.


TITLE: Síndrome de Guillain-Barré asociado a infección por COVID-19: revisión de casos publicados.Introducción. La pandemia por la enfermedad por coronavirus 2019 (COVID-19) es un importante problema para la salud mundial. Hay un incremento en las complicaciones neurológicas reconocidas por la COVID-19, incluyendo el síndrome de Guillain-Barré (SGB) y sus variantes. Desarrollo. Se realizó una revisión de los casos publicados en los últimos meses de SGB asociado a infección por COVID-19. Incluimos a 48 pacientes (31 hombres; edad media: 56,4 años). Los síntomas de COVID-19 más comunes fueron tos (60,4%) y fiebre (56,3%). El tiempo promedio entre los síntomas de COVID-19 y el SGB fue de 12,1 días, pero nueve pacientes (18,8%) desarrollaron SGB en menos de siete días. Once pacientes (22,9%) presentaron afectación de los nervios craneales en ausencia de debilidad muscular, 36 presentaron la variante clásica sensitivomotora (75%) y uno tuvo una variante motora pura (2,1%). El patrón electrofisiológico se consideró desmielinizante en el 82,4% de las variantes generalizadas. La presencia de hiposmia/disgeusia estuvo asociada con una latencia menor a los siete días hasta el inicio de los síntomas del SGB (30 frente a 15,6%) y a la afectación de los nervios craneales en ausencia de debilidad (30,8 frente a 17,1%). La mayoría de los pacientes (87,5%) fueron tratados con inmunoglobulina endovenosa. La evolución neurológica fue favorable en el 64,6%, el 29,2% tuvo insuficiencia respiratoria y hubo un 4,2% de muertes. Conclusiones. El SGB en pacientes con infección por SARS-CoV-2 es similar clínica y electrofisiológicamente a las formas clásicas. Se requieren más estudios para comprender si la frecuencia del SGB realmente aumentó debido a la pandemia por COVID-19 y explorar los mecanismos patógenos involucrados.


Subject(s)
COVID-19/complications , Guillain-Barre Syndrome/etiology , Pandemics , SARS-CoV-2 , Adolescent , Adult , Aged , Anosmia/etiology , Autoantibodies/blood , Autoantibodies/immunology , Autoantigens/immunology , Cranial Nerve Diseases/etiology , Dysgeusia/etiology , Female , Gangliosides/immunology , Guillain-Barre Syndrome/cerebrospinal fluid , Guillain-Barre Syndrome/immunology , Guillain-Barre Syndrome/therapy , Humans , Immunoglobulins, Intravenous/therapeutic use , Male , Middle Aged , Plasmapheresis , Respiratory Insufficiency/etiology , Retrospective Studies , Symptom Assessment , Treatment Outcome , Young Adult
20.
J Immunother Cancer ; 9(3)2021 03.
Article in English | MEDLINE | ID: mdl-33737337

ABSTRACT

Immune escape mechanisms employed by neuroblastoma (NB) cells include secretion of immunosuppressive factors disrupting effective antitumor immunity. The use of cellular therapy to treat solid tumors needs to be implemented. Killing activity of anti-GD2 Chimeric Antigen Receptor (CAR) T or natural killer (NK) cells against target NB cells was assessed through coculture experiments and quantified by FACS analysis. ELISA assay was used to quantify interferon-γ (IFNγ) secreted by NK and CAR T cells. Real Time PCR and Western Blot were performed to analyze gene and protein levels modifications. Transcriptional study was performed by chromatin immunoprecipitation and luciferase reporter assays on experiments of mutagenesis on the promoter sequence. NB tissue sample were analyzed by IHC and Real Time PCR to perform correlation study. We demonstrate that Indoleamine-pyrrole 2,3-dioxygenase1 (IDO1), due to its ability to convert tryptophan into kynurenines, is involved in NB resistance to activity of immune cells. In NB, IDO1 is able to inhibit the anti-tumor effect displayed by of both anti-GD2 CAR (GD2.CAR) T-cell and NK cells, mainly by impairing their IFNγ production. Furthermore, inhibition of MYCN expression in NB results into accumulation of IDO1 and consequently of kynurenines, which negatively affect the immune surveillance. Inverse correlation between IDO1 and MYCN expression has been observed in a wide cohort of NB samples. This finding was supported by the identification of a transcriptional repressive role of MYCN on IDO1 promoter. The evidence of IDO1 involvement in NB immune escape and its ability to impair NK and GD2.CAR T-cell activity contribute to clarify one of the possible mechanisms responsible for the limited efficacy of these immunotherapeutic approaches. A combined therapy of NK or GD2.CAR T-cells with IDO1 inhibitors, a class of compounds already in phase I/II clinical studies, could represent a new and still unexplored strategy capable to improve long-term efficacy of these immunotherapeutic approaches.


Subject(s)
Gangliosides/metabolism , Immunotherapy, Adoptive , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Interferon-gamma/metabolism , Killer Cells, Natural/transplantation , Lymphocyte Activation , N-Myc Proto-Oncogene Protein/metabolism , Neuroblastoma/therapy , Receptors, Chimeric Antigen/genetics , T-Lymphocytes/transplantation , Cell Line, Tumor , Coculture Techniques , Gangliosides/immunology , Gene Expression Regulation, Neoplastic , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/enzymology , Neuroblastoma/genetics , Neuroblastoma/immunology , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Escape , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL
...