Ganglioside GD2

14,000,000 Leading Edge Experts on the ideXlab platform

Scan Science and Technology

Contact Leading Edge Experts & Companies

Scan Science and Technology

Contact Leading Edge Experts & Companies

The Experts below are selected from a list of 3324 Experts worldwide ranked by ideXlab platform

Nai-kong V. Cheung - One of the best experts on this subject based on the ideXlab platform.

  • abstract b38 tetravalent bispecific antibodies specific for her2 and disialoGanglioside GD2 to engage polyclonal t cells for osteosarcoma therapy
    Cancer Research, 2018
    Co-Authors: Jeong A Park, Irene Cheung, Nai-kong V. Cheung
    Abstract:

    Introduction: Osteosarcoma is the most common pediatric high-grade bone tumor. Although multimodal therapeutic approaches have significantly improved patient survival to more than 60%, the prognosis for patients with metastatic or relapsed disease remains dismal—an urgent unmet need. Its genomic complexity and higher mutational burden compared to other pediatric cancers should provide neoantigens as potential targets for T cell-based immunotherapy; however, these tumors are in general “cold,” with insufficient or inactive tumor-infiltrating lymphocytes. Ganglioside GD2 is a tumor-associated surface antigen expressed in a broad spectrum of pediatric malignancies, including neuroblastoma, brain tumors, Ewing’s sarcoma, rhabdomyosarcoma, and osteosarcoma, while being restricted in normal tissues. Another promising target, HER2 or ErbB2, is overexpressed in many aggressive malignancies, including pediatric medulloblastoma, nephroblastoma, osteosarcoma, and desmoplastic small round cell tumor (DSRT). Fully humanized tetravalent bispecific antibodies (BsAb) specific for human GD2 and CD3 (hu3F8-BsAb) or HER2 and CD3 (HER2-BsAb), built on an IgG(L)-scFv platform, have been developed (Oncoimmunology 2017; Can Immunol Res 2015). They induce rapid and quantitative T-cell homing to tumors, mediating potent T-cell dependent cytotoxicity (TDCC) against GD2 or HER2 expressing tumor cells, effecting cures of xenografts in SCID mice. In this study, we evaluated the in vitro and in vivo antitumor properties of these BsAbs in the treatment of osteosarcoma. Methods: Hu3F8-BsAb and HER2-BsAb were tested both in vitro and in vivo against a panel of osteosarcoma cell lines (RG143B, U2OS, CRL1427, HOS, and SaOS2). Using these cell lines, FACS analysis for surface antigen (GD2 and HER2) expression (MFI), sensitivity to TDCC (EC50), and antitumor activity in vivo were evaluated. Two in vivo tumor models with different effector routes were used to simulate different clinical situations: 1) subcutaneous (sc) tumor cells/sc effector peripheral blood mononuclear cells (PBMCs), and 2) sc tumor cells/intravenous (iv) PBMCs. In vivo tumor responses were measured by Peira TM900 imaging device or by bioluminescence. Results: The majority of osteosarcoma cell lines express GD2 and HER2 on their surface, and their in vitro sensitivity to TDCC (EC50) was inversely correlated with MFI of their respective antigen. Both hu3F8-BsAb and HER2-BsAb mediated potent TDCC against osteosarcoma cell lines. In vivo, both hu3F8-BsAb and HER2-BsAb exerted a significant antitumor effect compared to control BsAb (P=0.001). Conclusions: Hu3F8-BsAb and HER2-BsAb induced strong TDCC and had significant antitumor effect against osteosarcoma cell lines both in vitro and in vivo. Considering the limited therapeutic options currently available in advanced osteosarcomas, these results support their further clinical development as potential T cell-based immunotherapeutics. Citation Format: Jeong A. Park, Hong Xu, Irene Cheung, Nai-Kong V. Cheung. Tetravalent bispecific antibodies specific for HER2 and disialoGanglioside GD2 to engage polyclonal T cells for osteosarcoma therapy [abstract]. In: Proceedings of the AACR Special Conference: Pediatric Cancer Research: From Basic Science to the Clinic; 2017 Dec 3-6; Atlanta, Georgia. Philadelphia (PA): AACR; Cancer Res 2018;78(19 Suppl):Abstract nr B38.

  • alteration of electrostatic surface potential enhances affinity and tumor killing properties of anti Ganglioside GD2 monoclonal antibody hu3f8
    Journal of Biological Chemistry, 2015
    Co-Authors: Qi Zhao, Mahiuddin Ahmed, Irene Y Cheung, Nai-kong V. Cheung
    Abstract:

    Abstract Ganglioside GD2 is highly expressed on neuroectodermal tumors and an attractive therapeutic target for antibodies that have already shown some clinical efficacy. To further improve the current antibodies, which have modest affinity, we sought to improve affinity by using a combined method of random mutagenesis and in silico assisted design to affinity-mature the anti-GD2 monoclonal antibody hu3F8. Using yeast display, mutants in the Fv with enhanced binding over the parental clone were FACS sorted and cloned. In silico modeling identified the minimal key interacting residues involved in the important charged interactions with the sialic acid groups of GD2. Two mutations, D32H (L-CDR2) and E1K (L-FR1) altered the electrostatic surface potential of the antigen binding site, allowing for an increase in positive charge to enhance the interaction with the negatively charged GD2-pentasaccharide head group. Purified scFv and IgG mutant forms were then tested for antigen specificity by ELISA, tissue specificity by immunohistochemistry, affinity by BIACORE, antibody-dependent cell-mediated cytotoxicity (ADCC) and complement mediated cytotoxicity (CMC) in vitro, and anti-tumor efficacy in xenografted humanized mice. The near 7-fold improvement in affinity of hu3F8 with single D32H (L-CDR2) mutation translated into a ~12-fold improvement in NK92MI-transfected CD16-mediated ADCC, 6-fold in CD32-mediated ADCC, and 2.5-fold in CMC, while maintaining restricted normal tissue cross-reactivity, and achieving substantial improvement in tumor ablation in vivo. Despite increasing GD2 affinity, the double mutation D32H (L-CDR2) and E1K (L-FR1) did not further improve anti-tumor efficacy.

  • alteration of electrostatic surface potential enhances affinity and tumor killing properties of anti Ganglioside GD2 monoclonal antibody hu3f8
    Journal of Biological Chemistry, 2015
    Co-Authors: Qi Zhao, Mahiuddin Ahmed, Irene Y Cheung, Hongfen Guo, Nai-kong V. Cheung
    Abstract:

    Ganglioside GD2 is highly expressed on neuroectodermal tumors and an attractive therapeutic target for antibodies that have already shown some clinical efficacy. To further improve the current antibodies, which have modest affinity, we sought to improve affinity by using a combined method of random mutagenesis and in silico assisted design to affinity-mature the anti-GD2 monoclonal antibody hu3F8. Using yeast display, mutants in the Fv with enhanced binding over the parental clone were FACS-sorted and cloned. In silico modeling identified the minimal key interacting residues involved in the important charged interactions with the sialic acid groups of GD2. Two mutations, D32H (L-CDR1) and E1K (L-FR1) altered the electrostatic surface potential of the antigen binding site, allowing for an increase in positive charge to enhance the interaction with the negatively charged GD2-pentasaccharide headgroup. Purified scFv and IgG mutant forms were then tested for antigen specificity by ELISA, for tissue specificity by immunohistochemistry, for affinity by BIACORE, for antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-mediated cytotoxicity in vitro, and for anti-tumor efficacy in xenografted humanized mice. The nearly 7-fold improvement in affinity of hu3F8 with a single D32H (L-CDR1) mutation translated into a ∼12-fold improvement in NK92MI-transfected CD16-mediated ADCC, a 6-fold improvement in CD32-mediated ADCC, and a 2.5-fold improvement in complement-mediated cytotoxicity while maintaining restricted normal tissue cross-reactivity and achieving substantial improvement in tumor ablation in vivo. Despite increasing GD2 affinity, the double mutation D32H (L-CDR1) and E1K (L-FR1) did not further improve anti-tumor efficacy.

  • DisialoGanglioside GD2 as a therapeutic target for human diseases.
    Expert opinion on therapeutic targets, 2015
    Co-Authors: Maya Suzuki, Nai-kong V. Cheung
    Abstract:

    Introduction: Ganglioside GD2 is found in vertebrates and invertebrates, overexpressed among pediatric and adult solid tumors, including neuroblastoma, glioma, retinoblastoma, Ewing’s family of tumors, rhabdomyosarcoma, osteosarcoma, leiomyosarcoma, liposarcoma, fibrosarcoma, small cell lung cancer and melanoma. It is also found on stem cells, neurons, some nerve fibers and basal layer of the skin.Areas covered: GD2 provides a promising clinical target for radiolabeled antibodies, bispecific antibodies, chimeric antigen receptor (CAR)-modified T cells, drug conjugates, nanoparticles and vaccines. Here, we review its biochemistry, normal physiology, role in tumorigenesis, important characteristics as a target, as well as anti-GD2-targeted strategies.Expert opinion: Bridging the knowledge gaps in understanding the interactions of GD2 with signaling molecules within the glycosynapses, and the regulation of its cellular expression should improve therapeutic strategies targeting this Ganglioside. In addition t...

  • GD2-Targeted Immunotherapy and Radioimmunotherapy
    Seminars in oncology, 2014
    Co-Authors: Konstantin Dobrenkov, Nai-kong V. Cheung
    Abstract:

    Ganglioside GD2 is a tumor-associated surface antigen found in a broad spectrum of human cancers and stem cells. They include pediatric embryonal tumors (neuroblastoma, retinoblastoma, brain tumors, osteosarcoma, Ewing sarcoma, rhabdomyosarcoma), as well as adult cancers (small cell lung cancer, melanoma, soft tissue sarcomas). Because of its restricted normal tissue distribution, GD2 has been proven safe for antibody targeting. Anti-GD2 antibody is now incorporated into the standard of care for the treatment of high-risk metastatic neuroblastoma. Building on this experience, novel combinations of antibodies, cytokines, cells, and genetically engineered products all directed at GD2 are rapidly moving into the clinic. In this review, past and present immunotherapy trials directed at GD2 will be summarized, highlighting the lessons learned and the future directions.

Mahiuddin Ahmed - One of the best experts on this subject based on the ideXlab platform.

  • a self assembling and disassembling sada bispecific antibody bsab platform for curative two step pretargeted radioimmunotherapy
    Clinical Cancer Research, 2021
    Co-Authors: Brian H Santich, Sarah M Cheal, Mahiuddin Ahmed, Michael R Mcdevitt, Ouathek Ouerfelli, Guangbin Yang, Darren R Veach, Edward K Fung, Mitesh Patel, Daniela Burnes Vargas
    Abstract:

    Purpose Many cancer treatments suffer from dose-limiting toxicities to vital organs due to poor therapeutic indices (TI). To overcome these challenges we developed a novel multimerization platform that rapidly removed tumor targeting proteins from the blood to substantially improve TI. Experimental design The platform was designed as a fusion of a Self-Assembling and DisAssembling (SADA) domain to a tandem single-chain bispecific antibody (BsAb, anti-Ganglioside GD2 x anti-DOTA). SADA-BsAbs were assessed with multiple in vivo tumor models using 2-step pretargeted radioimmunotherapy (PRIT) to evaluate tumor uptake, dosimetry and anti-tumor responses. Results SADA-BsAbs self-assembled into stable tetramers (220 kDa) but could also disassemble into dimers or monomers (55 kDa) that rapidly cleared via renal filtration and substantially reduced immunogenicity in mice. When used with rapidly clearing DOTA-caged PET isotopes, SADA-BsAbs demonstrated accurate tumor localization, dosimetry, and improved imaging contrast by PET/CT. When combined with therapeutic isotopes, 2-step SADA-PRIT safely delivered massive doses of alpha-emitting (225Ac, 1.48 MBq/kg) or beta-emitting (177Lu, 6,660 MBq/kg) DOTA payloads to tumors, ablating them without any short-term or long-term toxicities to the bone marrow, kidneys, or liver. Conclusions The SADA-BsAb platform safely delivered large doses of radioisotopes to tumors and demonstrated no toxicities to the bone marrow, kidneys or liver. Due to its modularity, SADA-BsAbs can be easily adapted to most tumor antigens, tumor types, or drug delivery approaches to improve TI and maximize the delivered dose.

  • a self assembling and disassembling sada bispecific antibody bsab platform for curative 2 step pre targeted radioimmunotherapy
    Clinical Cancer Research, 2020
    Co-Authors: Brian H Santich, Sarah M Cheal, Mahiuddin Ahmed, Michael R Mcdevitt, Ouathek Ouerfelli, Guangbin Yang, Darren R Veach, Edward K Fung, Mitesh Patel, Daniela Burnes Vargas
    Abstract:

    Purpose Many cancer treatments suffer from dose-limiting toxicities to vital organs due to poor therapeutic indices (TI). To overcome these challenges we developed a novel multimerization platform that rapidly removed tumor targeting proteins from the blood to substantially improve TI. Experimental design The platform was designed as a fusion of a Self-Assembling and DisAssembling (SADA) domain to a tandem single-chain bispecific antibody (BsAb, anti-Ganglioside GD2 x anti-DOTA). SADA-BsAbs were assessed with multiple in vivo tumor models using 2-step pretargeted radioimmunotherapy (PRIT) to evaluate tumor uptake, dosimetry and anti-tumor responses. Results SADA-BsAbs self-assembled into stable tetramers (220 kDa) but could also disassemble into dimers or monomers (55 kDa) that rapidly cleared via renal filtration and substantially reduced immunogenicity in mice. When used with rapidly clearing DOTA-caged PET isotopes, SADA-BsAbs demonstrated accurate tumor localization, dosimetry, and improved imaging contrast by PET/CT. When combined with therapeutic isotopes, 2-step SADA-PRIT safely delivered massive doses of alpha-emitting (225Ac, 1.48 MBq/kg) or beta-emitting (177Lu, 6,660 MBq/kg) DOTA payloads to tumors, ablating them without any short-term or long-term toxicities to the bone marrow, kidneys, or liver. Conclusions The SADA-BsAb platform safely delivered large doses of radioisotopes to tumors and demonstrated no toxicities to the bone marrow, kidneys or liver. Due to its modularity, SADA-BsAbs can be easily adapted to most tumor antigens, tumor types, or drug delivery approaches to improve TI and maximize the delivered dose.

  • alteration of electrostatic surface potential enhances affinity and tumor killing properties of anti Ganglioside GD2 monoclonal antibody hu3f8
    Journal of Biological Chemistry, 2015
    Co-Authors: Qi Zhao, Mahiuddin Ahmed, Irene Y Cheung, Nai-kong V. Cheung
    Abstract:

    Abstract Ganglioside GD2 is highly expressed on neuroectodermal tumors and an attractive therapeutic target for antibodies that have already shown some clinical efficacy. To further improve the current antibodies, which have modest affinity, we sought to improve affinity by using a combined method of random mutagenesis and in silico assisted design to affinity-mature the anti-GD2 monoclonal antibody hu3F8. Using yeast display, mutants in the Fv with enhanced binding over the parental clone were FACS sorted and cloned. In silico modeling identified the minimal key interacting residues involved in the important charged interactions with the sialic acid groups of GD2. Two mutations, D32H (L-CDR2) and E1K (L-FR1) altered the electrostatic surface potential of the antigen binding site, allowing for an increase in positive charge to enhance the interaction with the negatively charged GD2-pentasaccharide head group. Purified scFv and IgG mutant forms were then tested for antigen specificity by ELISA, tissue specificity by immunohistochemistry, affinity by BIACORE, antibody-dependent cell-mediated cytotoxicity (ADCC) and complement mediated cytotoxicity (CMC) in vitro, and anti-tumor efficacy in xenografted humanized mice. The near 7-fold improvement in affinity of hu3F8 with single D32H (L-CDR2) mutation translated into a ~12-fold improvement in NK92MI-transfected CD16-mediated ADCC, 6-fold in CD32-mediated ADCC, and 2.5-fold in CMC, while maintaining restricted normal tissue cross-reactivity, and achieving substantial improvement in tumor ablation in vivo. Despite increasing GD2 affinity, the double mutation D32H (L-CDR2) and E1K (L-FR1) did not further improve anti-tumor efficacy.

  • alteration of electrostatic surface potential enhances affinity and tumor killing properties of anti Ganglioside GD2 monoclonal antibody hu3f8
    Journal of Biological Chemistry, 2015
    Co-Authors: Qi Zhao, Mahiuddin Ahmed, Irene Y Cheung, Hongfen Guo, Nai-kong V. Cheung
    Abstract:

    Ganglioside GD2 is highly expressed on neuroectodermal tumors and an attractive therapeutic target for antibodies that have already shown some clinical efficacy. To further improve the current antibodies, which have modest affinity, we sought to improve affinity by using a combined method of random mutagenesis and in silico assisted design to affinity-mature the anti-GD2 monoclonal antibody hu3F8. Using yeast display, mutants in the Fv with enhanced binding over the parental clone were FACS-sorted and cloned. In silico modeling identified the minimal key interacting residues involved in the important charged interactions with the sialic acid groups of GD2. Two mutations, D32H (L-CDR1) and E1K (L-FR1) altered the electrostatic surface potential of the antigen binding site, allowing for an increase in positive charge to enhance the interaction with the negatively charged GD2-pentasaccharide headgroup. Purified scFv and IgG mutant forms were then tested for antigen specificity by ELISA, for tissue specificity by immunohistochemistry, for affinity by BIACORE, for antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-mediated cytotoxicity in vitro, and for anti-tumor efficacy in xenografted humanized mice. The nearly 7-fold improvement in affinity of hu3F8 with a single D32H (L-CDR1) mutation translated into a ∼12-fold improvement in NK92MI-transfected CD16-mediated ADCC, a 6-fold improvement in CD32-mediated ADCC, and a 2.5-fold improvement in complement-mediated cytotoxicity while maintaining restricted normal tissue cross-reactivity and achieving substantial improvement in tumor ablation in vivo. Despite increasing GD2 affinity, the double mutation D32H (L-CDR1) and E1K (L-FR1) did not further improve anti-tumor efficacy.

  • acute pain relief after mantram meditation in children with neuroblastoma undergoing anti GD2 monoclonal antibody therapy
    Journal of Pediatric Hematology Oncology, 2014
    Co-Authors: Mahiuddin Ahmed, Shakeel Modak, Sonia Sequeira
    Abstract:

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

Daniela Burnes Vargas - One of the best experts on this subject based on the ideXlab platform.

  • a self assembling and disassembling sada bispecific antibody bsab platform for curative two step pretargeted radioimmunotherapy
    Clinical Cancer Research, 2021
    Co-Authors: Brian H Santich, Sarah M Cheal, Mahiuddin Ahmed, Michael R Mcdevitt, Ouathek Ouerfelli, Guangbin Yang, Darren R Veach, Edward K Fung, Mitesh Patel, Daniela Burnes Vargas
    Abstract:

    Purpose Many cancer treatments suffer from dose-limiting toxicities to vital organs due to poor therapeutic indices (TI). To overcome these challenges we developed a novel multimerization platform that rapidly removed tumor targeting proteins from the blood to substantially improve TI. Experimental design The platform was designed as a fusion of a Self-Assembling and DisAssembling (SADA) domain to a tandem single-chain bispecific antibody (BsAb, anti-Ganglioside GD2 x anti-DOTA). SADA-BsAbs were assessed with multiple in vivo tumor models using 2-step pretargeted radioimmunotherapy (PRIT) to evaluate tumor uptake, dosimetry and anti-tumor responses. Results SADA-BsAbs self-assembled into stable tetramers (220 kDa) but could also disassemble into dimers or monomers (55 kDa) that rapidly cleared via renal filtration and substantially reduced immunogenicity in mice. When used with rapidly clearing DOTA-caged PET isotopes, SADA-BsAbs demonstrated accurate tumor localization, dosimetry, and improved imaging contrast by PET/CT. When combined with therapeutic isotopes, 2-step SADA-PRIT safely delivered massive doses of alpha-emitting (225Ac, 1.48 MBq/kg) or beta-emitting (177Lu, 6,660 MBq/kg) DOTA payloads to tumors, ablating them without any short-term or long-term toxicities to the bone marrow, kidneys, or liver. Conclusions The SADA-BsAb platform safely delivered large doses of radioisotopes to tumors and demonstrated no toxicities to the bone marrow, kidneys or liver. Due to its modularity, SADA-BsAbs can be easily adapted to most tumor antigens, tumor types, or drug delivery approaches to improve TI and maximize the delivered dose.

  • a self assembling and disassembling sada bispecific antibody bsab platform for curative 2 step pre targeted radioimmunotherapy
    Clinical Cancer Research, 2020
    Co-Authors: Brian H Santich, Sarah M Cheal, Mahiuddin Ahmed, Michael R Mcdevitt, Ouathek Ouerfelli, Guangbin Yang, Darren R Veach, Edward K Fung, Mitesh Patel, Daniela Burnes Vargas
    Abstract:

    Purpose Many cancer treatments suffer from dose-limiting toxicities to vital organs due to poor therapeutic indices (TI). To overcome these challenges we developed a novel multimerization platform that rapidly removed tumor targeting proteins from the blood to substantially improve TI. Experimental design The platform was designed as a fusion of a Self-Assembling and DisAssembling (SADA) domain to a tandem single-chain bispecific antibody (BsAb, anti-Ganglioside GD2 x anti-DOTA). SADA-BsAbs were assessed with multiple in vivo tumor models using 2-step pretargeted radioimmunotherapy (PRIT) to evaluate tumor uptake, dosimetry and anti-tumor responses. Results SADA-BsAbs self-assembled into stable tetramers (220 kDa) but could also disassemble into dimers or monomers (55 kDa) that rapidly cleared via renal filtration and substantially reduced immunogenicity in mice. When used with rapidly clearing DOTA-caged PET isotopes, SADA-BsAbs demonstrated accurate tumor localization, dosimetry, and improved imaging contrast by PET/CT. When combined with therapeutic isotopes, 2-step SADA-PRIT safely delivered massive doses of alpha-emitting (225Ac, 1.48 MBq/kg) or beta-emitting (177Lu, 6,660 MBq/kg) DOTA payloads to tumors, ablating them without any short-term or long-term toxicities to the bone marrow, kidneys, or liver. Conclusions The SADA-BsAb platform safely delivered large doses of radioisotopes to tumors and demonstrated no toxicities to the bone marrow, kidneys or liver. Due to its modularity, SADA-BsAbs can be easily adapted to most tumor antigens, tumor types, or drug delivery approaches to improve TI and maximize the delivered dose.

Ralph A Reisfeld - One of the best experts on this subject based on the ideXlab platform.

  • phase i study of chimeric human murine anti Ganglioside GD2 monoclonal antibody ch14 18 with granulocyte macrophage colony stimulating factor in children with neuroblastoma immediately after hematopoietic stem cell transplantation a children s cancer group study
    Journal of Clinical Oncology, 2000
    Co-Authors: Fevzi M Ozkaynak, Paul M Sondel, Ralph A Reisfeld, Mark Krailo, Brad Javorsky, Katherine K Matthay, Gregory H Reaman, Robert C Seeger
    Abstract:

    PURPOSE: Ganglioside GD2 is strongly expressed on the surface of human neuroblastoma cells. It has been shown that the chimeric human/murine anti-GD2 monoclonal antibody (ch14.18) can induce lysis of neuroblastoma cells by antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity. The purposes of the study were (1) to determine the maximum-tolerated dose (MTD) of ch14.18 in combination with standard dose granulocyte-macrophage colony-stimulating factor (GM-CSF) for patients with neuroblastoma who recently completed hematopoietic stem-cell transplantation (HSCT), and (2) to determine the toxicities of ch14.18 with GM-CSF in this setting. PATIENTS AND METHODS: Patients became eligible when the total absolute phagocyte count (APC) was greater than 1,000/μL after HSCT. ch14.18 was infused intravenously over 5 hours daily for 4 consecutive days. Patients received GM-CSF 250 μg/m2/d starting at least 3 days before ch14.18 and continued for 3 days after the completion of ch14.18. The ch14.18...

  • activation of human effector cells by a tumor reactive recombinant anti Ganglioside GD2 interleukin 2 fusion protein ch14 18 il2
    Clinical Cancer Research, 1996
    Co-Authors: Jacquelyn A Hank, Ralph A Reisfeld, Jean E Surfus, Stephen D Gillies, Peter Jaeger, Paul M Sondel
    Abstract:

    Cytotoxic effector cells interact with target cells through various mechanisms. CTLs use the antigen-specific T cell receptor, whereas Fc receptor-positive natural killer cells use this receptor to interact with antibody-coated target cells. We evaluated the tumor-binding and lymphocyte-activating capability of a recombinant fusion protein consisting of a tumor-selective human/mouse chimeric anti-Ganglioside GD2 antibody (ch14.18) and recombinant human interleukin-2 (IL2) (ch14.18-IL2). This fusion protein bound specifically to GD2-positive melanoma and neuroblastoma tumor cell lines, and its IL2 component stimulated in vitro proliferation of an IL2-dependent cell line, as well as peripheral blood mononuclear cells, in healthy control individuals and in cancer patients receiving continuous infusion of IL2. The IL2 presented by the fusion protein, when bound to tumor cells, induced proliferation of IL2-responsive cells as well as a comparable amount of soluble IL2 did. This suggests that localization of IL2 at the site of contact between tumor and effector cells is an effective way of presenting this cytokine to IL2-responsive cells. The ch14.18-IL2 fusion protein also mediated antibody-dependent cellular cytotoxicity with Fc receptor-positive effector cells to an extent similar to ch14.18. These results, together with those of previous studies documenting antitumor efficacy against human tumor xenografts in SCID mice and GD2-positive murine tumors in immunocompetent syngeneic mice, suggest that the ch14.18-IL2 fusion protein should be tested in Phase I and II trials in patients with GD2-positive tumors.

  • a recombinant antibody interleukin 2 fusion protein suppresses growth of hepatic human neuroblastoma metastases in severe combined immunodeficiency mice
    Proceedings of the National Academy of Sciences of the United States of America, 1994
    Co-Authors: Helen Sabzevari, Stephen D Gillies, Barbara M Mueller, James D Pancook, Ralph A Reisfeld
    Abstract:

    A genetically engineered fusion protein consisting of a human/mouse chimeric anti-Ganglioside GD2 antibody (ch14.18) and recombinant human interleukin 2 (rhIL-2) was tested for its ability to target rhIL-2 to tumor sites and stimulate immune effector cells sufficiently to achieve effective tumor cell lysis in vivo. The ch14.18-IL-2 fusion protein proved more effective than equivalent doses of rhIL-2 in suppressing dissemination and growth of human neuroblastoma in an experimental hepatic metastases model of scid (severe combined immunodeficiency) mice reconstituted with human lymphokine-activated killer cells. The ch14.18-IL-2 fusion protein was also more proficient than equivalent doses of rhIL-2 in prolonging the life-span of these animals. This recombinant antibody-cytokine fusion protein may prove useful for future treatment of GD2-expressing human tumors in an adjuvant setting.

  • A phase I study of neuroblastoma with the anti-Ganglioside GD2 antibody 14.G2a
    Cancer Immunology Immunotherapy, 1992
    Co-Authors: Rupert Handgretinger, Ralph A Reisfeld, J Treuner, Peter Baader, Roland Dopfer, Thomas Klingebiel, Peter Reuland, D Niethammer
    Abstract:

    Nine patients with neuroblastoma stage IV were treated with the murine monoclonal antibody 14.G2a, directed against disialoGanglioside GD2. The antibody was injected daily for 5–10 days and the total applied dosage ranged between 100 mg/m^2 and 400 mg/m^2. The peak serum levels of mAb 14.G2a ranged from 28 µg/ml to 61 µg/ml. Pharmacokinetic data obtained in three patients indicated that the serum elimination of mAb 14.G2a fits a two-compartment model, with an α-half-time ( t _ 1/2α ) between 0.66 h and 1.98 h and a β-half-time ( t _ 1/2β ) between 30.13 h and 53.33 h. All patients presented with a human anti-(mouse IgG) antibody response either during or shortly after therapy. Eight patients showed a continuous decrease in complement component C4 during therapy, as well as an initial decrease in C3c and an initial increase in C3a, all suggesting an activation of the complement cascade. Side-effects consisted of allergic reactions like pruritus, exanthema, urticaria and of severe pain, predominantly located in the abdomen and lower extremities, which required the use of continuous intravenous morphine. Four patients additionally developed a transient hypertension and one patient experienced a transient nephrotic syndrome. Three patients were treated in an adjuvant setting and are not evaluable for tumor response. Of the remaining six patients, two had a complete remission, two showed a partial remission, and two patients did not respond to treatment.

  • antibody targeted interleukin 2 stimulates t cell killing of autologous tumor cells
    Proceedings of the National Academy of Sciences of the United States of America, 1992
    Co-Authors: Stephen D Gillies, E B Reilly, Ralph A Reisfeld
    Abstract:

    A genetically engineered fusion protein consisting of a chimeric anti-Ganglioside GD2 antibody (ch14.18) and interleukin 2 (IL2) was tested for its ability to enhance the killing of autologous GD2-expressing melanoma target cells by a tumor-infiltrating lymphocyte line (660 TIL). The fusion of IL2 to the carboxyl terminus of the immunoglobulin heavy chain did not reduce IL2 activity as measured in a standard proliferation assay using either mouse or human T-cell lines. Antigen-binding activity was greater than that of the native chimeric antibody. The ability of resting 660 TIL cells to kill their autologous GD2-positive target cells was enhanced if the target cells were first coated with the fusion protein. This stimulation of killing was greater than that of uncoated cells in the presence of equivalent or higher concentrations of free IL2. Such antibody-cytokine fusion proteins may prove useful in targeting the biological effect of IL2 and other cytokines to tumor cells and in this way stimulate their immune destruction.

Venkata Lokesh Battula - One of the best experts on this subject based on the ideXlab platform.

  • anti GD2 antibody dinutuximab inhibits triple negative breast tumor growth by targeting GD2 breast cancer stem like cells
    Journal for ImmunoTherapy of Cancer, 2021
    Co-Authors: Vivek Anand, Fouad Eldana, Khoa Nguyen, Yiming Cai, Shirong Cai, Helen Piwnicaworms, Debasish Tripathy, A Sahin, Michael Andreeff, Venkata Lokesh Battula
    Abstract:

    Background Triple-negative breast cancer (TNBC) is the most aggressive breast cancer subtype with no effective standard therapy. Breast cancer stem-like cells (BCSCs) in primary TNBCs are reported to be responsible for metastatic spread of the disease and resistance to chemotherapy, but no available therapeutic tools target BCSCs. We previously reported that the Ganglioside GD2 is highly expressed on BCSCs and that inhibition of its expression hampers TNBC growth. We therefore hypothesized that the anti-GD2 antibody dinutuximab (ch14.18) targets GD2+ BCSCs and inhibits TNBC growth. Method To test our hypothesis, we first determined GD2 expression via immunohistochemistry in frozen primary tumor samples from patients with TNBC (n=89). Then, we examined the effects of dinutuximab on TNBC cell adhesion, migration, and mammosphere formation in vitro and on tumor growth in vivo using TNBC cell-line and patient-derived xenograft (PDX) models. Results We found that GD2 was expressed in around 60% of primary TNBC tumors at variable levels and was associated with worse overall survival of patients with TNBC (p=0.002). GD2 was found to be expressed in tumors and stroma, but normal ducts and lobules in adjacent tissues have shown low or no GD2 staining, indicating that GD2 is potentially a novel biomarker for tumor and its microenvironment. Treatment with dinutuximab significantly decreased adhesion and migration of MDA-MB-231 and SUM159 TNBC cells. Moreover, dinutuximab treatment inhibited mTOR signaling, which has been shown to be regulated by GD2 in BCSCs. Dinutuximab also reduced tumor growth in nude mice bearing TNBC cell-line xenografts. Finally, dinutuximab in combination with activated natural killer cells inhibited tumor growth in a TNBC PDX model and improved overall survival of tumor-bearing mice. Conclusions Dinutuximab successfully eliminated GD2+ cells and reduced tumor growth in both in vivo models. Our data provide proof-of-concept for the criticality of GD2 in BCSCs and demonstrate the potential of dinutuximab as a novel therapeutic approach for TNBC.

  • expression of Ganglioside GD2 reprogram the lipid metabolism and emt phenotype in bladder cancer
    Oncotarget, 2017
    Co-Authors: Venkatrao Vantaku, Khoa Nguyen, Venkata Lokesh Battula, Sri Ramya Donepudi, Chandrashekar R Ambati, Feng Jin, Vasanta Putluri, Kimal Rajapakshe, Cristian Coarfa, Yair Lotan
    Abstract:

    // Venkatrao Vantaku 1, * , Sri Ramya Donepudi 2, * , Chandrashekar R. Ambati 2, * , Feng Jin 2 , Vasanta Putluri 2 , Khoa Nguyen 3 , Kimal Rajapakshe 1 , Cristian Coarfa 1, 2 , Venkata Lokesh Battula 3 , Yair Lotan 4 and Nagireddy Putluri 1, 2 1 Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, TX, USA 2 Dan L. Duncan Cancer Center, Advanced Technology Core, Alkek Center for Molecular Discovery, Baylor College of Medicine, Houston, TX, USA 3 Section of Molecular Hematology and Therapy, Department of Leukemia, and Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA 4 Department of Urology, University of Texas Southwestern, Dallas, TX, USA * These authors have contributed equally to this work Correspondence to: Nagireddy Putluri, email: putluri@bcm.edu Keywords: bladder cancer; Ganglioside GD2; lipid metabolism; EMT Received: June 27, 2017      Accepted: August 04, 2017      Published: September 16, 2017 ABSTRACT High-grade Bladder Cancer (BLCA) represents the most aggressive and treatment-resistant cancer that renders the patients with poor survival. However, only a few biomarkers have been identified for the detection and treatment of BLCA. Recent studies show that Ganglioside GD2 can be used as cancer biomarker and/or therapeutic target for various cancers. Despite its potential relevance in cancer diagnosis and therapeutics, the role of GD2 is unknown in BLCA. Here, we report for the first time that high-grade BLCA tissues and cell lines have higher expression of GD2 compared to low-grade by high-resolution Mass Spectrometry. The muscle invasive UMUC3 cell line showed high GD2, mesenchymal phenotype, and cell proliferation. Besides, we have shown the cancer stem cells (CSC) property (CD44hiCD24lo) of GD2+ UMUC3 and J82 cells. Also, the evaluation of lipid metabolism in GD2+ BLCA cell lines revealed higher levels of Phosphatidylinositol (PI), Phosphatidic acid (PA), Cardiolipin (CL) and lower levels of Phosphatidylserine (PS), plasmenyl-phosphatidylethanolamines (pPE), plasmenyl-phosphocholines (pPC), sphingomyelins (SM), triglycerides (TGs) and N-Acetylneuraminic acid. These findings are significantly correlated with the tissues of BLCA patients. Based on this evidence, we propose that GD2 may be used as an effective diagnostic and therapeutic target for aggressive BLCA.

  • abstract p5 07 03 GD2 mediated fak signaling regulates breast cancer stem cell function in triple negative breast cancer
    Cancer Research, 2017
    Co-Authors: K Nguyen, Michael Andreeff, J C Sun, Gabriel N Hortobagyi, Venkata Lokesh Battula
    Abstract:

    Ganglioside GD2 identifies breast cancer stem cells (BCSCs, Battula et al., JCI, 2012) and expression of GD2 is tightly regulated by GD3 synthase (GD3S). GD3S is highly expressed in GD2 + cells and inhibition of GD3S inhibits tumor formation and metastasis of breast cancer cells. However, the mechanism of GD2-mediated regulation of BCSC function is not known. Here we hypothesize that GD2 regulates signaling pathways involved in cell adhesion, migration and invasion of breast cancer cells. To identify these signaling pathways, antibody micro-arrays were used with 850 validated antibodies specific to total or phosphorylated proteins. Interestingly, focal adhesion kinase (FAK) was the most significantly phosphorylated protein in GD2 + compared to GD2 - cells (S910 and S722). In addition, expression of FAK downstream mediators including Csk, PKCq, PKCl/I, Pyk2, and p38MAPK, was up-regulated in GD2 + compared to GD2 - cells. Western blot analysis of FACS sorted SUM159 cells also revealed increased phosphorylation of FAK >80% at Y397 and >25% at Y861 in GD2 + compared to GD2 - cells. FAK downstream targets including paxillin, p130 Cas, pERK were also up-regulated in GD2 + cells compared to GD2 - cells indicating definitive activation of FAK signaling in GD2 + BCSCs. To investigate the functional role of GD2 in FAK mediated functions, we genetically deleted GD3S using the CRISPR knock-out system in SUM159 cells. only 20% in parental cells was observed. GD3S-KO cells grew 5-10% slower in cell culture mostly because of the reduction (15±5%) in adherence. Trans-well assays revealed 3-5 fold reduction in migration and invasion of GD3S-KO compared to parental cells. These data indicate that GD2 and GD3S are not only the markers of BCSCs but also regulators of their function. Finally, we tested the effect of FAK inhibitor (PF-573228) against GD2 + BCSCs and GD3S-KO SUM159 cells. PF-573228 treatment decreased the number of mammospheres generated by GD2 + cells 3-4 fold in a dose dependent manner (100nM-1µm). In addition, treatment of PF-573228, inhibited migration and invasion of GD2 + cells 2 and 3 fold, respectively. However, treatment with PF-573228 on GD3S-KO cells further reduced their ability to migrate and invade by over 70% compared to untreated cells. In addition, GD3S-KO cells failed to form any mammospheres when cultured under low adherence conditions (p + cells but that FAK inhibition alone may not be sufficient to inhibit BCSC function. Combined FAK and GD3S inhibition may exert highly synergistic effects against BCSCs. Citation Format: Nguyen K, Sun JC, Hortobagyi GN, Andreeff M, Battula VL. GD2-mediated FAK signaling regulates breast cancer stem cell function in triple negative breast cancer [abstract]. In: Proceedings of the 2016 San Antonio Breast Cancer Symposium; 2016 Dec 6-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2017;77(4 Suppl):Abstract nr P5-07-03.

  • Ganglioside GD2 identifies breast cancer stem cells and promotes tumorigenesis
    The Journal of clinical investigation, 2012
    Co-Authors: Venkata Lokesh Battula, Yuexi Shi, Kurt W. Evans, Rui-yu Wang, Rodrigo Jacamo, Rudy Guerra, Aysegul A. Sahin, Frank C. Marini, Erika L. Spaeth, Gabriel N Hortobagyi
    Abstract:

    Cancer stem cells (CSCs) are a small subpopulation of cancer cells that have increased resistance to conventional therapies and are capable of establishing metastasis. However, only a few biomarkers of CSCs have been identified. Here, we report that Ganglioside GD2 (a glycosphingolipid) identifies a small fraction of cells in human breast cancer cell lines and patient samples that are capable of forming mammospheres and initiating tumors with as few as 10 GD2+ cells. In addition, the majority of GD2+ cells are also CD44hiCD24lo, the previously established CSC-associated cell surface phenotype. Gene expression analysis revealed that GD3 synthase (GD3S) is highly expressed in GD2+ as well as in CD44hiCD24lo cells and that interference with GD3S expression, either by shRNA or using a pharmacological inhibitor, reduced the CSC population and CSC-associated properties. GD3S knockdown completely abrogated tumor formation in vivo. Also, induction of epithelial-mesenchymal transition (EMT) in transformed human mammary epithelial cells (HMLER cells) dramatically increased GD2 as well as GD3S expression in these cells, suggesting a role of EMT in the origin of GD2+ breast CSCs. In summary, we identified GD2 as a new CSC-specific cell surface marker and GD3S as a potential therapeutic target for CSCs, with the possibility of improving survival and cure rates in patients with breast cancer.

  • Abstract LB-193: Ganglioside GD2 identifies cancer stem cells and inhibition of GD2 biosynthesis by targeting GD3 synthase exerts antitumor effects
    Tumor Biology, 2012
    Co-Authors: Venkata Lokesh Battula, Yuexi Shi, Kurt W. Evans, Rui-yu Wang, Erika L. Speath, Rodrigo Jacamo, Rudy Guerra, Aysegul A. Sahin, Frank C. Marini, Sendurai A. Mani
    Abstract:

    Proceedings: AACR 103rd Annual Meeting 2012‐‐ Mar 31‐Apr 4, 2012; Chicago, IL Currently, only very few markers either as single or in combination are available to identify cancer stem cells (CSCs). In this report, we identified that the Ganglioside GD2, a marker known to express on mesenchymal stromal cells (MSCs) is expressed on a small fraction (5.5% ± 3.4%) of transformed human mammary epithelial cells (HMLER) and breast cancer patient tumors. FACS sorted GD2+ cells appear spindle shaped and proliferate 5 fold slower compared to GD2- cells in-vitro. Analysis of breast cancer cell lines (n=12) indicated that GD2 expression varies and that the basal breast cancer cell lines have a higher percentage of GD2+ cells (median 9%, range 1.2-17%, n=6), as compared to their luminal counterparts (median 0.2%, range 0-3%, n=6, p 10 fold. Moreover, we observed spontaneous generation of GD2+ from GD2- cells and vice versa in both in-vitro and in-vivo, suggesting a role of EMT in this process. Stable knock-down of GD3S in MDA-MB-231 cells using shRNA impaired in-vitro matrigel invasion by more than 10-fold and completely abolished tumor growth in-vivo. Importantly, Triptolide, an anti-inflammatory and anti-cancer drug, which was recently shown to inhibit GD3S expression in melanoma cells, also inhibited GD3S expression in MDA-MB-231 and SUM159 cells by >95% in a dose dependent manner and thereby inhibited growth of GD2+ cells in a time dependent manner. Intra-peritoneal administration of Triptolide (0.15mg/Kg/day) in NOD/SCID mice bearing MDA-MB-231 breast tumors completely eliminated tumors in 50% and reduced the tumor volume 7- to 8-fold in 25% of the mice. In conclusion, we identified GD2 as a new CSC specific cell surface marker and GD3 synthase as a potential therapeutic target for CSCs, with the potential of improving survival and cure rates of patients with breast cancer. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr LB-193. doi:1538-7445.AM2012-LB-193