Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Cancer Immunol Immunother ; 71(1): 71-83, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34023958

ABSTRACT

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


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

ABSTRACT

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


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Gangliosides/antagonists & inhibitors , Glioblastoma/drug therapy , Neoplastic Stem Cells/drug effects , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/therapeutic use , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Cell Self Renewal/drug effects , Cell Self Renewal/immunology , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/immunology , Drug Synergism , Gangliosides/immunology , Glioblastoma/immunology , Glioblastoma/pathology , Humans , Mice , Neoplastic Stem Cells/immunology , Temozolomide/pharmacology , Temozolomide/therapeutic use , Xenograft Model Antitumor Assays
3.
Int J Cancer ; 133(3): 757-65, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23354868

ABSTRACT

Immunocytokines (ICKs) targeting cytokines to the tumor environment using antibodies directed against a tumor-associated antigen often have a higher therapeutic index than the corresponding unconjugated cytokines. Various ICKs displaying significant antitumoral effects in several murine tumor models have already been developed, and some of them, in particular interleukin (IL)-2-based ICKs, are in Phase II clinical trials. Although sharing common biological activities with IL-2 in vitro, IL-15 is now considered as having a better potential in antitumor immunotherapeutical strategies and has been shown to be less toxic than IL-2 in preclinical studies. We previously developed the fusion protein RLI, linking a soluble form of human IL-15Rα-sushi+ domain to human IL-15. RLI showed better biological activities than IL-15 in vitro as well as higher antitumoral effects in vivo in murine and human cancer models. Here, we investigated, in the context of an ICK, the effect of associating RLI with an antibody targeting the GD2 ganglioside, a validated tumoral target expressed on many neurectodermal tumors. Anti-GD2-RLI fully retained the cytokine potential of RLI and the antibody effector functions (antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity). It displayed strong antitumor activities in two syngeneic cancer models in immunocompetent mice (subcutaneous EL4 and metastatic NXS2). Its therapeutic potency was higher than those of RLI and anti-GD2 alone or in combination. We suggest that this is related to its bifunctional (cytokine and antibody) nature.


Subject(s)
Gangliosides/immunology , Lymphoma, T-Cell/drug therapy , Neuroblastoma/drug therapy , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/therapeutic use , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacology , Antibodies, Neoplasm/immunology , Antibodies, Neoplasm/metabolism , Antibodies, Neoplasm/pharmacology , Antibody-Dependent Cell Cytotoxicity/immunology , Antigens, Neoplasm/immunology , Cell Line, Tumor , Cell Proliferation , Female , Gangliosides/metabolism , Humans , Immunotherapy , Interleukin-15/agonists , Interleukin-15/immunology , Interleukin-15/therapeutic use , Interleukin-15 Receptor alpha Subunit , Liver Neoplasms/drug therapy , Liver Neoplasms/secondary , Male , Mice , Mice, Inbred A , Mice, Inbred C57BL , Neoplasm Metastasis/drug therapy , Protein Binding/immunology , Recombinant Fusion Proteins/pharmacology
4.
Front Pharmacol ; 14: 1249929, 2023.
Article in English | MEDLINE | ID: mdl-37670947

ABSTRACT

Part of the broader glycosphingolipid family, gangliosides are composed of a ceramide bound to a sialic acid-containing glycan chain, and locate at the plasma membrane. Gangliosides are produced through sequential steps of glycosylation and sialylation. This diversity of composition is reflected in differences in expression patterns and functions of the various gangliosides. Ganglioside GD2 designates different subspecies following a basic structure containing three carbohydrate residues and two sialic acids. GD2 expression, usually restrained to limited tissues, is frequently altered in various neuroectoderm-derived cancers. While GD2 is of evident interest, its glycolipid nature has rendered research challenging. Physiological GD2 expression has been linked to developmental processes. Passing this stage, varying levels of GD2, physiologically expressed mainly in the central nervous system, affect composition and formation of membrane microdomains involved in surface receptor signaling. Overexpressed in cancer, GD2 has been shown to enhance cell survival and invasion. Furthermore, binding of antibodies leads to immune-independent cell death mechanisms. In addition, GD2 contributes to T-cell dysfunction, and functions as an immune checkpoint. Given the cancer-associated functions, GD2 has been a source of interest for immunotherapy. As a potential biomarker, methods are being developed to quantify GD2 from patients' samples. In addition, various therapeutic strategies are tested. Based on initial success with antibodies, derivates such as bispecific antibodies and immunocytokines have been developed, engaging patient immune system. Cytotoxic effectors or payloads may be redirected based on anti-GD2 antibodies. Finally, vaccines can be used to mount an immune response in patients. We review here the pertinent biological information on GD2 which may be of use for optimizing current immunotherapeutic strategies.

5.
Cancers (Basel) ; 13(21)2021 Oct 29.
Article in English | MEDLINE | ID: mdl-34771608

ABSTRACT

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

6.
Article in English | MEDLINE | ID: mdl-33383867

ABSTRACT

Biologic reference drugs and their copies, biosimilars, have a complex structure. Biosimilars need to demonstrate their biosimilarity during development but unpredictable variations can remain, such as micro-heterogeneity. The healthcare community may raise questions regarding the clinical outcomes induced by this micro-heterogeneity. Indeed, unwanted immune reactions may be induced for numerous reasons, including product variations. However, it is challenging to assess these unwanted immune reactions because of the multiplicity of causes and potential delays before any reaction. Moreover, safety assessments as part of preclinical studies and clinical trials may be of limited value with respect to immunogenicity assessments because they are performed on a standardised population during a limited period. Real-life data could therefore supplement the assessments of clinical trials by including data on the real-life use of biosimilars, such as switches. Furthermore, real-life data also include any economic incentives to prescribe or use biosimilars. This article raises the question of relevance of automating real life data processing regarding Biosimilars. The objective is to initiate a discussion about different approaches involving Machine Learning. So, the discussion is established regarding implementation of Neural Network model to ensure safety of biosimilars subject to economic incentives. Nevertheless, the application of Machine Learning in the healthcare field raises ethical, legal and technical issues that require further discussion.


Subject(s)
Biosimilar Pharmaceuticals/standards , Public Health , Follow-Up Studies , Humans , Machine Learning , Neural Networks, Computer
7.
J Vis Exp ; (143)2019 01 19.
Article in English | MEDLINE | ID: mdl-30735151

ABSTRACT

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


Subject(s)
Antibodies, Neoplasm/pharmacology , Antineoplastic Agents/pharmacology , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacology , Antibodies, Neoplasm/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Line, Tumor , Drug Interactions , Drug Synergism , Humans , Mice, Inbred NOD , Mice, SCID , Topotecan/pharmacology , Xenograft Model Antitumor Assays
9.
Clin Cancer Res ; 13(18 Pt 2): 5613s-5620s, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17875797

ABSTRACT

PURPOSE: We previously generated a mouse monoclonal antibody (mAb) specific for the tumor-associated GD2 ganglioside antigen. Here, we describe the development of a chimeric anti-GD2 mAb for more effective tumor immunotherapy. EXPERIMENTAL DESIGN: We cloned the cDNA encoding the immunoglobulin light and heavy chains of the 60C3 anti-GD2 mAb, and constructed chimeric genes by linking the cDNA fragments of the variable regions of the murine light and heavy chains to cDNA fragments of the human kappa and gamma1 constant regions, respectively. RESULTS: The resultant chimeric anti-GD2 mAb, c.60C3, showed identical binding affinity and specificity to that of its murine counterpart. Both c.60C3 and 60C3 were rapidly internalized by tumor cells at 37 degrees C. When human serum and human natural killer cells were used as effectors in complement-mediated cytotoxicity and antibody-dependent cell cytotoxicity, respectively, c.60C3 was more effective in killing GD2-expressing tumor cells. However, c.60C3 was ineffective at inducing cell death by apoptosis, although binding of 60C3 induced apoptotic death in vitro. In an in vivo, GD2-expressing, syngeneic tumor model, i.v. injection of c.60C3, but not of 60C3, significantly suppressed tumor growth in mice (P<0.0005). CONCLUSION: Immune effector functions mediated by this antibody and its potentially reduced immunogenicity make chimeric c.60C3 a promising therapeutic agent against neuroectodermic tumors.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibody-Dependent Cell Cytotoxicity , Antineoplastic Agents/pharmacology , G(M2) Ganglioside/immunology , Recombinant Fusion Proteins/pharmacology , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Apoptosis/immunology , Complement System Proteins/metabolism , Cytotoxicity, Immunologic , DNA, Complementary/genetics , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression/genetics , Genetic Vectors/genetics , Humans , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Light Chains/genetics , Immunoglobulin Light Chains/immunology , Immunoglobulin M/genetics , Immunoglobulin Variable Region/genetics , Immunoglobulin gamma-Chains/genetics , Immunoglobulin gamma-Chains/immunology , Immunoglobulin kappa-Chains/genetics , Immunoglobulin kappa-Chains/immunology , Mice , Mice, Inbred C57BL , Recombinant Fusion Proteins/genetics , Survival Rate , Transfection , Transplantation, Heterologous
10.
Mol Immunol ; 44(7): 1680-90, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17011035

ABSTRACT

Single-domain antibodies specific to methotrexate (MTX) were obtained after immunization of one llama (Llama glama). Specific VHH domains (V-D-J-REGION) were selected by panning from an immune-llama library using phage display technology. The antibody fragments specific to MTX were purified from Escherichia coli (C41 strain) periplasm by immobilized metal affinity chromatography with an expression level of around 10mg/L. A single band around 16,000Da corresponding to VHH fragments was found after analysis by SDS-PAGE and Western blotting, while competition ELISA demonstrated selective binding to soluble MTX. Surface plasmon resonance (SPR) analysis showed that anti-MTX VHH domains had affinities in the nanomolar range (29-515nM) to MTX-serum albumin conjugates. The genes encoding anti-MTX VHH were found by IMGT/V-QUEST to be similar to the previously reported llama and human IGHV germline genes. The V-D and D-J junction rearrangements in the seven anti-MTX CDR3 sequences indicate that they were originated from three distinct progenitor B cells. Our results demonstrate that camelid single-domain antibodies are capable of high affinity binding to low molecular weight hydrosoluble haptens. Furthermore, these anti-MTX VHH give new insights on how the antigen binding repertoire of llama single-domain antibody can provide combining sites to haptens in the absence of a VL. This type of single-domain antibodies offers advantages compared to murine recombinant antibodies in terms of production rate and sequence similarity to the human IGHV3 subgroup genes.


Subject(s)
Antibodies/metabolism , Camelids, New World/immunology , Methotrexate/immunology , Recombinant Proteins/biosynthesis , Amino Acid Sequence , Animals , Antibodies/genetics , Antibodies/isolation & purification , Base Sequence , Escherichia coli/genetics , Female , Haptens/immunology , Molecular Sequence Data , Peptide Library , Protein Structure, Tertiary , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Sequence Alignment , Surface Plasmon Resonance
11.
J Immunol Res ; 2017: 5604891, 2017.
Article in English | MEDLINE | ID: mdl-28154831

ABSTRACT

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


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigens, Neoplasm/immunology , Gangliosides/immunology , Gangliosides/metabolism , Neoplasms/therapy , Acetylation , Animals , Antibodies, Anti-Idiotypic/adverse effects , Antibodies, Anti-Idiotypic/immunology , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Murine-Derived/therapeutic use , Antigens, Neoplasm/chemistry , Clinical Trials as Topic , Gangliosides/chemistry , Humans , Immunotherapy , Neoplasms/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology
12.
Oncotarget ; 7(27): 41172-41185, 2016 Jul 05.
Article in English | MEDLINE | ID: mdl-27172791

ABSTRACT

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


Subject(s)
Antibodies, Monoclonal/therapeutic use , Cancer Vaccines/therapeutic use , Gangliosides/immunology , Glioblastoma/therapy , Adult , Aged , Animals , Apoptosis/immunology , Cell Line, Tumor , Female , Gangliosides/metabolism , Glioblastoma/immunology , Glioblastoma/metabolism , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Molecular Targeted Therapy/methods , Xenograft Model Antitumor Assays
13.
PLoS One ; 9(2): e87210, 2014.
Article in English | MEDLINE | ID: mdl-24520328

ABSTRACT

BACKGROUND: Anti-GD2 antibody is a proven therapy for GD2-positive neuroblastoma. Monoclonal antibodies against GD2, such as chimeric mAb ch14.18, have become benchmarks for neuroblastoma therapies. Pain, however, can limit immunotherapy with anti-GD2 therapeutic antibodies like ch14.18. This adverse effect is attributed to acute inflammation via complement activation on GD2-expressing nerves. Thus, new strategies are needed for the development of treatment intensification strategies to improve the outcome of these patients. METHODOLOGY/PRINCIPAL FINDINGS: We established the mouse-human chimeric antibody c.8B6 specific to OAcGD2 in order to reduce potential immunogenicity in patients and to fill the need for a selective agent that can kill neuroblastoma cells without inducing adverse neurological side effects caused by anti-GD2 antibody immunotherapy. We further analyzed some of its functional properties compared with anti-GD2 ch14.18 therapeutic antibody. With the exception of allodynic activity, we found that antibody c.8B6 shares the same anti-neuroblastoma attributes as therapeutic ch14.18 anti-GD2 mAb when tested in cell-based assay and in vivo in an animal model. CONCLUSION/SIGNIFICANCE: The absence of OAcGD2 expression on nerve fibers and the lack of allodynic properties of c.8B6-which are believed to play a major role in mediating anti-GD2 mAb dose-limiting side effects-provide an important rationale for the clinical application of c.8B6 in patients with high-risk neuroblastoma.


Subject(s)
Antibodies, Monoclonal/immunology , Gangliosides/immunology , Hyperalgesia/chemically induced , Neuroblastoma/immunology , Neuroblastoma/therapy , Acetylation , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antibody Specificity/immunology , Flow Cytometry , Humans , Hyperalgesia/pathology , Injections, Intravenous , Mice , Neuroblastoma/pathology , Protein Binding , Rats , Rats, Sprague-Dawley
14.
Cancer Lett ; 333(2): 194-204, 2013 Jun 10.
Article in English | MEDLINE | ID: mdl-23370223

ABSTRACT

O-Acetyl-GD2 ganglioside is suitable antigen for tumor immunotherapy with specific therapeutic antibody. Here, we investigate the anti-tumor activity of O-acetyl-GD2-specific monoclonal antibody 8B6 on O-acetyl-GD2-positive tumor cells. The results indicated that mAb 8B6 induced growth inhibition of O-acetyl-GD2-expressing tumor cell lines in vitro with features of cell cycle arrest and apoptosis. Monoclonal antibody 8B6 treatment was also very effective in suppression of tumor growth in mice by reducing the proliferation index and increasing the apoptotic index. Such a study represents a useful framework to optimize immunotherapy with O-acetyl-GD2-specific antibody in combination with chemotherapeutic agents.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Gangliosides/immunology , Animals , Antibodies, Monoclonal/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Humans , Mice , Mice, SCID , Xenograft Model Antitumor Assays
15.
Oncoimmunology ; 2(4): e23700, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23734323

ABSTRACT

Current antiangiogenic immunotherapeutic strategies mainly focus on the blockade of circulating cytokines or receptors that are overexpressed by endothelial cells. We proposed globotriaosylceramide (Gb3) as a viable alternative target for antiangiogenic therapies. In this setting, we developed an anti-Gb3 antibody and validated its therapeutic efficacy in metastatic tumor models.

16.
PLoS One ; 7(11): e45423, 2012.
Article in English | MEDLINE | ID: mdl-23189121

ABSTRACT

Inhibiting the growth of tumor vasculature represents one of the relevant strategies against tumor progression. Between all the different pro-angiogenic molecular targets, plasma membrane glycosphingolipids have been under-investigated. In this present study, we explore the anti-angiogenic therapeutic advantage of a tumor immunotherapy targeting the globotriaosylceramide Gb3. In this purpose, a monoclonal antibody against Gb3, named 3E2 was developed and characterized. We first demonstrate that Gb3 is over-expressed in proliferative endothelial cells relative to quiescent cells. Then, we demonstrate that 3E2 inhibits endothelial cell proliferation in vitro by slowing endothelial cell proliferation and by increasing mitosis duration. Antibody 3E2 is further effective in inhibiting ex vivo angiogenesis in aorta ring assays. Moreover, 3E2 treatment inhibits NXS2 neuroblastoma development and liver metastases spreading in A/J mice. Immunohistology examination of the NXS2 metastases shows that only endothelial cells, but not cancer cells express Gb3. Finally, 3E2 treatment diminishes tumor vessels density, proving a specific therapeutic action of our monoclonal antibody to tumor vasculature. Our study demonstrates that Gb3 is a viable alternative target for immunotherapy and angiogenesis inhibition.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antigens, Tumor-Associated, Carbohydrate/immunology , Antineoplastic Agents/pharmacology , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/immunology , Neovascularization, Pathologic/immunology , Angiogenesis Inhibitors/administration & dosage , Animals , Antibodies, Monoclonal/administration & dosage , Antigens, Tumor-Associated, Carbohydrate/metabolism , Antineoplastic Agents/administration & dosage , Cell Line , Cell Line, Tumor , Cell Membrane/immunology , Cell Membrane/metabolism , Cell Proliferation , Disease Models, Animal , Human Umbilical Vein Endothelial Cells/immunology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Male , Mice , Neoplasm Metastasis , Neovascularization, Pathologic/drug therapy , Neovascularization, Physiologic/drug effects , Neuroblastoma/drug therapy , Neuroblastoma/immunology , Neuroblastoma/pathology , Tumor Burden/drug effects
17.
PLoS One ; 6(9): e25220, 2011.
Article in English | MEDLINE | ID: mdl-21966461

ABSTRACT

BACKGROUND: Monoclonal antibodies (mAb) against GD2 ganglioside have been shown to be effective for the treatment of neuroblastoma. Beneficial actions are, however, associated with generalized pain due to the binding of anti- GD2 mAbs to peripheral nerve fibers followed by complement activation. Neuroblastoma cells that express GD2 also express its O-acetyl derivative, O-acetyl- GD2 ganglioside (OAcGD2). Hence, we investigated the distribution of OAcGD2 in human tissues using mAb 8B6 to study the cross-reactivity of mAb 8B6 with human tissues. METHODOLOGY/PRINCIPAL FINDINGS: The distribution of OAcGD2 was performed in normal and malignant tissues using an immunoperoxydase technique. Anti-tumor properties of mAb 8B6 were studied in vitro and in vivo in a transplanted tumor model in mice. We found that OAcGD2 is not expressed by peripheral nerve fibers. Furthermore, we demonstrated that mAb 8B6 was very effective in the in vitro and in vivo suppression of the growth of tumor cells. Importantly, mAb 8B6 anti-tumor efficacy was comparable to that of mAb 14G2a specific to GD2. CONCLUSION/SIGNIFICANCE: Development of therapeutic antibodies specific to OAcGD2 may offer treatment options with reduced adverse side effects, thereby allowing dose escalation of antibodies.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Gangliosides/immunology , Peripheral Nervous System/drug effects , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Humans , In Vitro Techniques , Neuroblastoma/metabolism , Peripheral Nervous System/pathology
SELECTION OF CITATIONS
SEARCH DETAIL