ganglioside--gd2 and Brain-Neoplasms

ganglioside--gd2 has been researched along with Brain-Neoplasms* in 16 studies

Reviews

2 review(s) available for ganglioside--gd2 and Brain-Neoplasms

ArticleYear
[Anti-GD2 antibodies in treatment of high-risk Neuroblastoma: present and perspectives].
    Medecine sciences : M/S, 2019, Volume: 35, Issue:12

    Neuroblastome de haut risque - Place actuelle et perspectives de l’utilisation des anticorps monoclonaux anti-GD2.. Le neuroblatome de haut risque reste un défi thérapeutique de l’oncologie pédiatrique puisque qu’il touche de très jeunes patients (90 % ont moins de 5 ans), dont les chances de survie restent inférieures à 50 % malgré des traitements très lourds. Une immunothérapie par l’anticorps monoclonal anti-GD2 dinutuximab bêta (Qarziba

    Topics: Animals; Antibodies, Monoclonal; Antineoplastic Agents, Immunological; Brain Neoplasms; Child; Child, Preschool; Gangliosides; Humans; Infant; Medical Oncology; Neuroblastoma; Risk Factors

2019
GD2-targeted immunotherapy and potential value of circulating microRNAs in neuroblastoma.
    Journal of cellular physiology, 2018, Volume: 233, Issue:2

    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.

    Topics: Animals; Antineoplastic Agents; Biomarkers, Tumor; Brain Neoplasms; Gangliosides; Humans; Immunotherapy; MicroRNAs; Molecular Diagnostic Techniques; Neuroblastoma; Predictive Value of Tests; Treatment Outcome

2018

Trials

3 trial(s) available for ganglioside--gd2 and Brain-Neoplasms

ArticleYear
Survival Impact of Anti-GD2 Antibody Response in a Phase II Ganglioside Vaccine Trial Among Patients With High-Risk Neuroblastoma With Prior Disease Progression.
    Journal of clinical oncology : official journal of the American Society of Clinical Oncology, 2021, 01-20, Volume: 39, Issue:3

    Anti-GD2 monoclonal antibody (mAb) has proven efficacy in high-risk neuroblastoma (HR-NB). A small phase I GD2/GD3 vaccine trial (n = 15) described long-term survival and a favorable safety profile among patients with a history of disease progression (PD). The kinetics of mounting antibody response to vaccine and its prognostic impact on survival are now investigated in a phase II study (ClinicalTrials.gov identifier: NCT00911560).. One hundred two patients with HR-NB who achieved remission after salvage therapies were enrolled in this trial. They received seven subcutaneous injections of GD2/GD3 vaccine spanning 1 year plus oral β-glucan starting at week 6 after the third dose of vaccine. Serum anti-vaccine antibody titers were quantified by enzyme-linked immunosorbent assay. Single nucleotide polymorphisms (SNPs) were determined by quantitative polymerase chain reaction. Kaplan-Meier and landmark Cox Regression models were used for survival estimates.. Patients had a history of one (63%), two (21%), or three to six (16%) episodes of PD. 82% of them progressed following anti-GD2 mAb (m3F8/dinutuximab/naxitamab) therapy. Vaccine-related toxicities were self-limited injection-associated local reactions and fever without any > grade 3 toxicities. The progression-free survival (PFS) was 32% ± 6%, and the overall survival (OS) was 71% ± 7% at 5 years. Serum anti-GD2 (immunoglobulin G1 [IgG1] and IgM) and anti-GD3 (IgG1) titers showed notable increases following the initiation of β-glucan at week 6. There was an association between IgG1 titer and SNP rs3901533 of dectin-1, the β-glucan receptor. Multivariable analyses showed that anti-GD2-IgG1 titer ≥ 150 ng/mL by week 8 was associated with favorable PFS and OS, while having prior episodes of PD and the time from last PD to vaccine were associated with PFS.. GD2/GD3 vaccine plus β-glucan elicited robust antibody responses in patients with HR-NB with prior PD. Higher anti-GD2-IgG1 titer was associated with improved survival.

    Topics: Adjuvants, Immunologic; beta-Glucans; Biomarkers; Brain Neoplasms; Cancer Vaccines; Child; Child, Preschool; Disease Progression; Female; Gangliosides; Glioblastoma; Humans; Immunogenicity, Vaccine; Immunoglobulin G; Infant; Lectins, C-Type; Male; Polymorphism, Single Nucleotide; Progression-Free Survival; Time Factors

2021
Ocular abnormalities in patients treated with a novel anti-GD2 monoclonal antibody, hu14.18K322A.
    Journal of AAPOS : the official publication of the American Association for Pediatric Ophthalmology and Strabismus, 2015, Volume: 19, Issue:2

    To determine the incidence of and factors associated with the development of mydriasis and impaired accommodation in patients with refractory or recurrent neuroblastoma receiving the anti-GD2 antibody hu14.18K322A.. The medical records of eligible patients with refractory or recurrent neuroblastoma who received escalating doses of hu14.18K322A, ranging from 2 to 70 mg/m(2)/dose for 4 consecutive days every 28 days, were retrospectively reviewed to identify ocular abnormalities arising during the treatment period.. A total of 38 patients (median age, 7 years; 23 males) were included. All patients underwent comprehensive eye examinations prior to each course of therapy. Mydriasis was seen in 13 patients (34%), and impaired accommodation was seen in 9 (24%), indicating a dose-related effect between hu14.18K322A and both mydriasis (P = 0.021) and impaired accommodation (P = 0.029). Age and sex were not associated with ocular abnormalities. Ocular symptoms resolved in the majority of patients after the drug was discontinued.. Side effects of mydriasis and impaired accommodation have a dose-dependent relationship with hu14.18K322A. These side effects do not warrant discontinuation of treatment, as they usually resolve after completion of therapy. Management of ocular side effects should focus on treating symptoms with manifest refraction, bifocals, or tinted spectacles.

    Topics: Accommodation, Ocular; Antibodies, Monoclonal, Humanized; Antineoplastic Agents; Brain Neoplasms; Child; Dose-Response Relationship, Drug; Female; Gangliosides; Humans; Incidence; Infusions, Intravenous; Male; Mydriasis; Neoplasm Recurrence, Local; Neuroblastoma; Retrospective Studies; Risk Factors

2015
Phase I clinical trial on adjuvant active immunotherapy of human gliomas with GD2-conjugate.
    British journal of neurosurgery, 2002, Volume: 16, Issue:3

    The objective of this study was to determine the feasibility, toxicity, and potential therapeutic benefits of an adjuvant active immunotherapy using a tumour specific ganglioside (GD2) conjugate for the adjuvant treatment of recurrent or progressive gliomas. Seven patients with proven GD2 expression in surgical specimens underwent a vaccination course with GD2-KLH/MPL-A conjugate. The follow-up was performed according to WHO guidelines regarding common toxicity criteria. Antibody titres against the ganglioside and the adjuvants were analysed. All patients developed a local type 4 reaction. Anti-GD2-antibody titres could not be detected, despite high titres against the immunoadjuvants. No tumour regression was observed. The disease remained stable for a median of 21.5 weeks (6-34 weeks). The median survival time after the first immunization was 47 weeks. The medial total survival time was 76 weeks. Adverse effects have not been observed. Active GD2-KLH/MPL-A immunization was technically feasible, but did not elicit anti-GD2 antibody generation.

    Topics: Adjuvants, Immunologic; Adult; Aged; Animals; Brain Neoplasms; Enzyme-Linked Immunosorbent Assay; Feasibility Studies; Female; Gangliosides; Glioma; Humans; Immunoglobulin G; Immunoglobulin M; Immunotherapy, Active; Male; Mice; Mice, Inbred BALB C; Middle Aged; Neoplasm Recurrence, Local; Radiography; Vaccination

2002

Other Studies

11 other study(ies) available for ganglioside--gd2 and Brain-Neoplasms

ArticleYear
Lack of GD3 synthase (St8sia1) attenuates malignant properties of gliomas in genetically engineered mouse model.
    Cancer science, 2021, Volume: 112, Issue:9

    High expression of gangliosides GD3 and GD2 is observed in human gliomas. The functions of GD3 and GD2 in malignant properties have been reported in glioma cells in vitro, but those functions have not yet been investigated in vivo. In this study, we showed that deficiency of GD3 synthase (GD3S, St8sia1) attenuated glioma progression and clinical and pathological features in a platelet-derived growth factor B-driven murine glioma model. Lack of GD3S resulted in the prolonged lifespan of glioma-bearing mice and low-grade pathology in generated gliomas. Correspondingly, they showed reduced phosphorylation levels of Akt, Erks, and Src family kinases in glioma tissues. A DNA microarray study revealed marked alteration in the expression of various genes, particularly in MMP family genes, in GD3S-deficient gliomas. Re-expression of GD3S restored expression of MMP9 in primary-cultured glioma cells. We also identified a transcription factor, Ap2α, expressed in parallel with GD3S expression, and showed that Ap2α was critical for the induction of MMP9 by transfection of its cDNA and luciferase reporter genes, and a ChIP assay. These findings suggest that GD3S enhances the progression of gliomas by enhancement of the Ap2α-MMP9 axis. This is the first report to describe the tumor-enhancing functions of GD3S in vivo.

    Topics: Animals; Astrocytes; Brain Neoplasms; Cells, Cultured; Disease Models, Animal; Disease Progression; Gangliosides; Gene Expression Regulation, Neoplastic; Glioma; Longevity; Matrix Metalloproteinase 9; Mice; Mice, Inbred BALB C; Mice, Inbred C57BL; Mice, Knockout; Sialyltransferases; Transfection

2021
Targeting GD2-positive glioblastoma by chimeric antigen receptor empowered mesenchymal progenitors.
    Cancer gene therapy, 2020, Volume: 27, Issue:7-8

    Tumor targeting by genetically modified mesenchymal stromal/stem cells (MSCs) carrying anti-cancer molecules represents a promising cell-based strategy. We previously showed that the pro-apoptotic agent tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) can be successfully delivered by MSCs to cancer sites. While the interaction between TRAIL and its receptors is clear, more obscure is the way in which MSCs can selectively target tumors and their antigens. Several neuroectoderm-derived neoplasms, including glioblastoma (GBM), sarcomas, and neuroblastoma, express high levels of the tumor-associated antigen GD2. We have already challenged this cell surface disialoganglioside by a chimeric antigen receptor (CAR)-T cell approach against neuroblastoma. With the intent to maximize the therapeutic profile of MSCs delivering TRAIL, we here originally developed a bi-functional strategy where TRAIL is delivered by MSCs that are also gene modified with the truncated form of the anti-GD2 CAR (GD2 tCAR) to mediate an immunoselective recognition of GD2-positive tumors. These bi-functional MSCs expressed high levels of TRAIL and GD2 tCAR associated with a robust anti-tumor activity against GD2-positive GBM cells. Most importantly, the anti-cancer action was reinforced by the enhanced targeting potential of such bi-functional cells. Collectively, our results suggest that a truncated anti-GD2 CAR might be a powerful new tool to redirect MSCs carrying TRAIL against GD2-expressing tumors. This affinity-based dual targeting holds the promise to combine site-specific and prolonged retention of MSCs in GD2-expressing tumors, thereby providing a more effective delivery of TRAIL for still incurable cancers.

    Topics: Antigens, Neoplasm; Brain Neoplasms; Cell Line, Tumor; Female; Gangliosides; Glioblastoma; Humans; Immunotherapy, Adoptive; Mesenchymal Stem Cells; Receptors, Chimeric Antigen

2020
Enhancement of malignant properties of human glioma cells by ganglioside GD3/GD2.
    International journal of oncology, 2018, Volume: 52, Issue:4

    Sialic acid-containing glycosphingolipids, gangliosides, are considered as cancer associated antigens in neuro-ectoderm-derived tumors such as melanomas and neuroblastomas. In particular, gangliosides GD3 and GD2 are expressed in human gliomas. It has been reported that their expression levels increase along with increased malignant properties. However, the implication of GD3/GD2 in human glioma cells has never been clarified, at least to the best of our knowledge. In this study, we introduced the cDNA of GD3 synthase (GD3S)(ST8SIA1) into a glioma cell line, U-251MG, that expresses neither GD3 nor GD2, thereby establishing transfectant cells U-251MG-GD3S(+) expressing high levels of GD3 and GD2 on the cell surface. In these U-251MG‑GD3S(+) cell lines, signaling molecules such as Erk1/2, Akt, p130Cas, paxillin and focal adhesion kinase were activated, leading to the enhancement of invasion activity and motility. It was then demonstrated that the U-251MG-GD3S(+) cells could proliferate under culture conditions with low or no serum concentrations without undergoing cell cycle arrest by escaping the accumulation of p16 and p21. All these results suggested that GD3 and GD2 highly expressed in gliomas confer increased invasion and mobility, cell growth abilities under low serum conditions, and increased ratios of the S-G2/M phase in the cell cycle.

    Topics: Brain Neoplasms; Cell Line, Tumor; Cell Movement; Cell Proliferation; Gangliosides; Glioma; Humans; Neoplasm Invasiveness; Tumor Cells, Cultured

2018
Potent antitumor efficacy of anti-GD2 CAR T cells in H3-K27M
    Nature medicine, 2018, Volume: 24, Issue:5

    Diffuse intrinsic pontine glioma (DIPG) and other diffuse midline gliomas (DMGs) with mutated histone H3 K27M (H3-K27M)

    Topics: Animals; Brain Neoplasms; Gangliosides; Glioma; Histones; Humans; Immunotherapy, Adoptive; Lysine; Methylation; Mice; Receptors, Antigen, T-Cell; Xenograft Model Antitumor Assays

2018
Anti-GD2-ch14.18/CHO coated nanoparticles mediate glioblastoma (GBM)-specific delivery of the aromatase inhibitor, Letrozole, reducing proliferation, migration and chemoresistance in patient-derived GBM tumor cells.
    Oncotarget, 2017, Mar-07, Volume: 8, Issue:10

    Aromatase is a critical enzyme in the irreversible conversion of androgens to oestrogens, with inhibition used clinically in hormone-dependent malignancies. We tested the hypothesis that targeted aromatase inhibition in an aggressive brain cancer called glioblastoma (GBM) may represent a new treatment strategy. In this study, aromatase inhibition was achieved using third generation inhibitor, Letrozole, encapsulated within the core of biodegradable poly lactic-co-glycolic acid (PLGA) nanoparticles (NPs). PLGA-NPs were conjugated to human/mouse chimeric anti-GD2 antibody ch14.18/CHO, enabling specific targeting of GD2-positive GBM cells. Treatment of primary and recurrent patient-derived GBM cells with free-Letrozole (0.1 μM) led to significant decrease in cell proliferation and migration; in addition to reduced spheroid formation. Anti-GD2-ch14.18/CHO-NPs displayed specific targeting of GBM cells in colorectal-glioblastoma co-culture, with subsequent reduction in GBM cell numbers when treated with anti-GD2-ch14.18-PLGA-Let-NPs in combination with temozolomide. As miR-191 is an estrogen responsive microRNA, its expression, fluctuation and role in Letrozole treated GBM cells was evaluated, where treatment with premiR-191 was capable of rescuing the reduced proliferative phenotype induced by aromatase inhibitor. The repurposing and targeted delivery of Letrozole for the treatment of GBM, with the potential role of miR-191 identified, provides novel avenues for target assessment in this aggressive brain cancer.

    Topics: Antibodies, Monoclonal; Antineoplastic Agents; Aromatase Inhibitors; Brain Neoplasms; Cell Line, Tumor; Cell Movement; Cell Proliferation; Gangliosides; Glioblastoma; HeLa Cells; Humans; Immunotoxins; Lactic Acid; Letrozole; Nanoparticles; Nitriles; Polyglycolic Acid; Polylactic Acid-Polyglycolic Acid Copolymer; Transfection; Triazoles

2017
The glioma-associated gangliosides 3'-isoLM1, GD3 and GM2 show selective area expression in human glioblastoma xenografts in nude rat brains.
    Neuropathology and applied neurobiology, 2001, Volume: 27, Issue:6

    This work describes the in vivo expression and distribution of glioma-associated gangliosides (GD3, GM2, 3'-isoLM1) in a novel human brain tumour nude rat xenograft model. In this model, the tumours, which are established directly from human glioblastoma biopsies, show extensive infiltrative growth within the rat brain. This model therefore provides an opportunity to study ganglioside expression not only within the macroscopic tumour, but also in brain areas with tumour cell infiltration. The ganglioside expression was studied by confocal microscopy of immunostained brain sections using antiganglioside monoclonal antibodies. Xenografts from four human glioblastoma multiformes were established in rats and the brains removed after 3-4 months. Ganglioside GD3 was expressed in the tumour parenchyma while ganglioside 3'-isoLM1 was more abundantly expressed in the periphery of the tumour associated with areas of tumour cell invasion. GM2 expression was only seen in one tumour, where it was located within the main tumour mass. Double staining with a pan antihuman monoclonal antibody (3B4) and the antiganglioside monoclonal antibodies confirmed that the ganglioside expression was associated with tumour cells. This work supports the concept of different biological roles for individual gangliosides and indicates that antibodies or ligands directed against GD3 and 3'-isoLM1 might be complementary when applied in the treatment of human glioblastomas.

    Topics: Animals; Antibodies, Monoclonal; Antigens, Tumor-Associated, Carbohydrate; Brain Injuries; Brain Neoplasms; Disease Models, Animal; Fluorescent Antibody Technique; Gangliosides; Glioblastoma; Humans; Microscopy, Confocal; Neoplasm Transplantation; Rats; Rats, Nude; Transplantation, Heterologous; Wounds, Stab

2001
Immunohistochemically visualized localisation of gangliosides Glac2 (GD3) and Gtri2 (GD2) in cells of human intracranial tumors.
    Experimental and toxicologic pathology : official journal of the Gesellschaft fur Toxikologische Pathologie, 2000, Volume: 52, Issue:4

    Antibodies against two major gangliosides detected in human brain and brain tumors--Glac2 (GD3) and Gtri2 (GD2)--were tested by immunohistochemistry in an unselected sample of intracranial tumors during the years 1986 through 1991. Two groups emerged as evaluable samples, namely gliomas of different grades and meningiomas. In a pilot series, it was shown that these gangliosides could be visualized in frozen sections of cells of gliomas and meningiomas (as well as neurinomas) and in some structures of the normal brain. It was however not possible in frozen sections to further analyze the cellular or subcellular expression pattern of the mentioned components and paraffin sections with conventional processing were only weakly and diffusely stained. A modified protocol therefore was created that replaced alcohol processing by acetone. With this protocol, interpretable results in paraffin sections were obtained. With this method, 133 single intracranial tumors were investigated as to their immunohistologically detectable ganglioside expression. The most consistent result was that the whole cytoplasm of highly fibrillary (gemistocytic) astrocytes in all grades of gliomas was stained by Gtri2 (GD2) and Glac2 (GD3) with high preponderance of Gtri2 (GD2) expression. In all meningiomas, Glac2 (GD3) had a higher expression than Gtri2. No constant pattern in the other entities emerged. By comparison with GFAP expression in gliomas and vimentin in meningiomas, the colocalisation of gangliosides and intermediary filament proteins is supposed.

    Topics: Antibodies, Monoclonal; Brain Neoplasms; Gangliosides; Glioma; Humans; Immunohistochemistry; Meningioma

2000
Enhancement of in vitro and in vivo anti-tumor activity of anti-GD2 monoclonal antibody 220-51 against human neuroblastoma by granulocyte-macrophage colony-stimulating factor and granulocyte colony-stimulating factor.
    International journal of molecular medicine, 1998, Volume: 2, Issue:4

    We have evaluated the anti-tumor effect of anti-GD2 mouse monoclonal antibody (mAb) 220-51 against human neuroblastoma cell line TGW in vitro and in vivo. The mAb 220-51 was able to mediate complement-dependent cytotoxicity (CDC) and antibody-dependent cellular cytotoxicity (ADCC) using human effector cells. In the presence of recombinant human granulocyte-macrophage colony-stimulating factor (GM-CSF), granulocyte ADCC was significantly augmented in vitro. When mAb 220-51 was administered to tumor-bearing nude mice, tumor growth was significantly inhibited as compared with untreated controls. Administration of recombinant murine GM-CSF in combination with mAb 220-51 significantly enhanced the anti-tumor effect of mAb in vivo. Recombinant human granulocyte colony-stimulating factor (G-CSF) combined with mAb 220-51 was also able to enhance it, although granulocyte ADCC was not affected by the presence of recombinant human G-CSF in vitro. Moreover, GM-CSF and G-CSF work additively to enhance the anti-tumor effect of mAb 220-51 in vivo. The GM-CSF and G-CSF may have a clinical potency in immunotherapy with anti-GD2 mAb for the treatment of neuroblastoma.

    Topics: Animals; Antibodies, Monoclonal; Antineoplastic Agents; Brain Neoplasms; Cell Line, Tumor; Drug Synergism; Drug Therapy, Combination; Female; Gangliosides; Granulocyte Colony-Stimulating Factor; Granulocyte-Macrophage Colony-Stimulating Factor; Granulocytes; Humans; Leukocyte Count; Mice; Mice, Inbred BALB C; Mice, Nude; Neoplasm Transplantation; Neuroblastoma; Recombinant Proteins; Transplantation, Heterologous

1998
Gangliosides as diagnostic markers of human astrocytomas and primitive neuroectodermal tumors.
    Cancer, 1994, Dec-01, Volume: 74, Issue:11

    Limitations of classification schemes for brain tumors based solely on morphology have stimulated searches for molecular markers of nosologic and prognostic value. Gangliosides are logical candidates because there are high concentrations of them in the nervous system, there is evidence of their roles in regulation of growth and differentiation, and data from small series suggest correlations between ganglioside composition and glioma type.. Ganglioside compositions were determined for 70 primary human brain tumors: 16 low grade astrocytomas (LG), 12 anaplastic astrocytomas (AA), 34 glioblastoma multiformes (GBM), and 8 primitive neuroectodermal tumors (PNET). This method involved identification and quantitation of specific gangliosides using chemical analysis and immunoanalysis.. Among all tumor types, histologic grade correlated with a progressive loss of 1b gangliosides (P < 0.0001). GQ1b was higher in LGs than in AAs (P < 0.001). Both GT1b and GD1b were higher in AAs than GBMs (P < 0.01 and 0.05, respectively) and lower in PNETs than in GBMs (P < 0.05). GM3 was higher in PNETs than in any astrocytoma group and higher in GBMs than in either AAs or LGs. There was a significant difference in the content of 3'-LM1 among all groups (P < 0.005), between AAs and GBMs (P < 0.05), and between low grade ordinary and juvenile pilocytic astrocyomas (P < 0.01). The lacto-series ganglioside 3'-isoLM1 was present in all groups except PNET.. These results indicate that patterns of gangliosides could be of considerable value in refining the classification and diagnosis of primary human brain tumors.

    Topics: Adult; Aged; Astrocytoma; Biomarkers, Tumor; Brain Neoplasms; Female; G(M1) Ganglioside; G(M3) Ganglioside; Gangliosides; Glioblastoma; Humans; Male; Middle Aged; Nerve Growth Factors; Neuroectodermal Tumors, Primitive, Peripheral

1994
Expression of GD2-epitopes in human intracranial tumors and normal brain.
    Experimental and toxicologic pathology : official journal of the Gesellschaft fur Toxikologische Pathologie, 1992, Volume: 44, Issue:6

    Two monoclonal antibodies (mabs) were raised against ganglioside GD2 (Gtri2) and tested on human intracranial tumors and normal brain by immunohistochemical methods both in frozen and paraffin embedded sections. In normal brain structures, astrocytes were visualized with both mabs (BW 625 and BW 704) almost exclusively in the subventricular and subpial layer. A minor amount of myelin sheaths in well defined localisation was only recognized in frozen sections. Consequently in astrocytic tumors of different grades of malignancy (WHO I-IV), astrocytes were depicted in their characteristic shape and arrangement around vessels. In addition, staining was observed in meningiomas and schwannomas, but not in pituitary adenomas or metastatic carcinomas. In meningioma und schwannoma the staining was restricted to the cellular periphery and was again present in frozen section material and absent in paraffin embedded tissue. In astrocytes, reactive and neoplastic, obviously fibrous processes and cytoplasm were distinctly stained both in frozen and paraffin embedded sections. It is concluded that some neuroectodermal derived cells as well as myelin of defined localisation express GD2 on the membrane surfaces and in the cytoplasm. The latter may primarily be the case in fibrous astrocytes, which were stained in reactive and pathologic conditions. The reaction can be used as diagnostic tool in astrocytic tumors; its positive therapeutic significance is hampered by the fact that (1) not all cells in heterogeneous tumor populations express the epitope and (2) there are normal structures which do so.

    Topics: Antibodies, Monoclonal; Brain Chemistry; Brain Neoplasms; Epitopes; Gangliosides; Glioma; Humans; Immunohistochemistry; Meningioma; Neurilemmoma

1992
Disialoganglioside GD2 in human neuroectodermal tumor cell lines and gliomas.
    Acta neuropathologica, 1991, Volume: 82, Issue:1

    Monoclonal antibodies (mAbs) recognizing the disialoganglioside II3(NeuAc)2GgOse3Cer (GD2) were produced by immunizing mice with the GD2-expressing neuroblastoma cell line LAN-1 and a prefusion boost with purified GD2 coupled to Salmonella minnesota. Two IgM mAbs were isolated which demonstrated high levels of reactivity (binding ratios in excess of 100) with GD2 by solid-phase radioimmunoassay and positivity in high-performance thin-layer chromatography (HPTLC) immunostain; only one (DMAb-20) was subsequently shown by analysis with a panel of defined ganglioside species to be specific for the minimum epitope of GD2 GalNAc beta 1-4(NeuAc alpha 2-8-NeuAc alpha 2-3)Gal-, DMAb-20 was used to evaluate the expression of GD2 by malignant glioma and medulloblastoma cell lines using cell surface radioimmunoassay. indirect membrane immunofluorescence. HPTLC immunostain, and densitometric analysis of extracted gangliosides from selected cell lines. Sixteen of 20 (80%) malignant glioma and 5 of 5 medulloblastoma cell lines reacted with DMAb-20; in agreement with previous studies, 5 of 5 neuroblastoma and 2 of 3 melanoma cell lines also reacted with DMAb-20, GD2 was proportionally increased in the glioma and medulloblastoma cell lines relative to levels in normal brain, as determined by densitometric analysis. In a phenotypic survey of malignant glioma biopsies, tumor cells in 24 of 30 (80%) cases stained positively with DMAb-20. Reactive astrocytes, both within the adjacent to tumors, were frequently intensely stained. Among the morphological variants of glioblastoma examined, the most intense staining with DMAb-20 was observed in neoplastic gemistocytes, with the weakest or absent staining in small cell glioblastomas. As GD2 is a commonly expressed surface antigen of gliomas and medulloblastomas, expression of which is retained in tissue culture. DMAb-20 will be useful in determining the functional role of GD2 in cell-cell interaction, adhesion, and invasion, and in defining altered growth control mechanisms of central nervous system neoplasms in in vitro models.

    Topics: Animals; Antibodies, Monoclonal; Antibody Specificity; Brain Neoplasms; Cell Line; Female; Fluorescent Antibody Technique; Gangliosides; Glioblastoma; Glioma; Humans; Immunoenzyme Techniques; Mice; Mice, Inbred BALB C; Radioimmunoassay

1991