1
|
Galili U. Self-Tumor Antigens in Solid Tumors Turned into Vaccines by α-gal Micelle Immunotherapy. Pharmaceutics 2024; 16:1263. [PMID: 39458595 PMCID: PMC11510312 DOI: 10.3390/pharmaceutics16101263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 09/02/2024] [Accepted: 09/23/2024] [Indexed: 10/28/2024] Open
Abstract
A major reason for the failure of the immune system to detect tumor antigens (TAs) is the insufficient uptake, processing, and presentation of TAs by antigen-presenting cells (APCs). The immunogenicity of TAs in the individual patient can be markedly increased by the in situ targeting of tumor cells for robust uptake by APCs, without the need to identify and characterize the TAs. This is feasible by the intra-tumoral injection of α-gal micelles comprised of glycolipids presenting the carbohydrate-antigen "α-gal epitope" (Galα1-3Galβ1-4GlcNAc-R). Humans produce a natural antibody called "anti-Gal" (constituting ~1% of immunoglobulins), which binds to α-gal epitopes. Tumor-injected α-gal micelles spontaneously insert into tumor cell membranes, so that multiple α-gal epitopes are presented on tumor cells. Anti-Gal binding to these epitopes activates the complement system, resulting in the killing of tumor cells, and the recruitment of multiple APCs (dendritic cells and macrophages) into treated tumors by the chemotactic complement cleavage peptides C5a and C3a. In this process of converting the treated tumor into a personalized TA vaccine, the recruited APC phagocytose anti-Gal opsonized tumor cells and cell membranes, process the internalized TAs and transport them to regional lymph-nodes. TA peptides presented on APCs activate TA-specific T cells to proliferate and destroy the metastatic tumor cells presenting the TAs. Studies in anti-Gal-producing mice demonstrated the induction of effective protection against distant metastases of the highly tumorigenic B16 melanoma following injection of natural and synthetic α-gal micelles into primary tumors. This treatment was further found to synergize with checkpoint inhibitor therapy by the anti-PD1 antibody. Phase-1 clinical trials indicated that α-gal micelle immunotherapy is safe and can induce the infiltration of CD4+ and CD8+ T cells into untreated distant metastases. It is suggested that, in addition to converting treated metastases into an autologous TA vaccine, this treatment should be considered as a neoadjuvant therapy, administering α-gal micelles into primary tumors immediately following their detection. Such an immunotherapy will convert tumors into a personalized anti-TA vaccine for the period prior to their resection.
Collapse
Affiliation(s)
- Uri Galili
- Department of Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
2
|
Shyr CR, Liu LC, Chien HS, Huang CP. Immunotherapeutic Agents for Intratumoral Immunotherapy. Vaccines (Basel) 2023; 11:1717. [PMID: 38006049 PMCID: PMC10674963 DOI: 10.3390/vaccines11111717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/22/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
Immunotherapy using systemic immune checkpoint inhibitors (ICI) and chimeric antigen receptor (CAR) T cells has revolutionized cancer treatment, but it only benefits a subset of patients. Systemic immunotherapies cause severe autoimmune toxicities and cytokine storms. Immune-related adverse events (irAEs) plus the immunosuppressive tumor microenvironment (TME) have been linked to the inefficacy of systemic immunotherapy. Intratumoral immunotherapy that increases immunotherapeutic agent bioavailability inside tumors could enhance the efficacy of immunotherapies and reduce systemic toxicities. In preclinical and clinical studies, intratumoral administration of immunostimulatory agents from small molecules to xenogeneic cells has demonstrated antitumor effects not only on the injected tumors but also against noninjected lesions. Herein, we review and discuss the results of these approaches in preclinical models and clinical trials to build the landscape of intratumoral immunotherapeutic agents and we describe how they stimulate the body's immune system to trigger antitumor immunity as well as the challenges in clinical practice. Systemic and intratumoral combination immunotherapy would make the best use of the body's immune system to treat cancers. Combining precision medicine and immunotherapy in cancer treatment would treat both the mutated targets in tumors and the weakened body's immune system simultaneously, exerting maximum effects of the medical intervention.
Collapse
Affiliation(s)
- Chih-Rong Shyr
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 404328, Taiwan; (C.-R.S.); (H.-S.C.)
- eXCELL Biotherapeutics Inc., Taichung 404328, Taiwan
| | - Lang-Chi Liu
- Department of Medicine, Department of Surgery, College of Medicine, China Medical University and Hospital, Taichung 404328, Taiwan;
| | - Hui-Shan Chien
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 404328, Taiwan; (C.-R.S.); (H.-S.C.)
| | - Chi-Ping Huang
- Department of Medicine, Urology Division, China Medical University and Hospital, Taichung 404328, Taiwan
| |
Collapse
|
3
|
Uvyn A, Vleugels MEJ, de Waal B, Hamouda AEI, Dhiman S, Louage B, Albertazzi L, Laoui D, Meijer EW, De Geest BG. Hapten/Myristoyl Functionalized Poly(propyleneimine) Dendrimers as Potent Cell Surface Recruiters of Antibodies for Mediating Innate Immune Killing. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2303909. [PMID: 37572294 DOI: 10.1002/adma.202303909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 08/09/2023] [Indexed: 08/14/2023]
Abstract
Recruiting endogenous antibodies to the surface of cancer cells using antibody-recruiting molecules has the potential to unleash innate immune effector killing mechanisms against antibody-bound cancer cells. The affinity of endogenous antibodies is relatively low, and many currently explored antibody-recruiting strategies rely on targeting over-expressed receptors, which have not yet been identified in most solid tumors. Here, both challenges are addressed by functionalizing poly(propyleneimine) (PPI) dendrimers with both multiple dinitrophenyl (DNP) motifs, as anti-hapten antibody-recruiting motifs, and myristoyl motifs, as universal phospholipid cell membrane anchoring motifs, to recruit anti-hapten antibodies to cell surfaces. By exploiting the multivalency of the ligand exposure on the dendrimer scaffold, it is demonstrated that dendrimers featuring ten myristoyl and six DNP motifs exhibit the highest antibody-recruiting capacity in vitro. Furthermore, it is shown that treating cancer cells with these dendrimers in vitro marks them for phagocytosis by macrophages in the presence of anti-hapten antibodies. As a proof-of-concept, it is shown that intratumoral injection of these dendrimers in vivo in tumor-bearing mice results in the recruitment of anti-DNP antibodies to the cell surface in the tumor microenvironment. These findings highlight the potential of dendrimers as a promising class of novel antibody-recruiting molecules for use in cancer immunotherapy.
Collapse
Affiliation(s)
- Annemiek Uvyn
- Department of Pharmaceutics, Ghent University, Ghent, 9000, Belgium
| | - Marle Elisabeth Jacqueline Vleugels
- Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology, MB 5600, P.O. Box 513, Eindhoven, The Netherlands
| | - Bas de Waal
- Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology, MB 5600, P.O. Box 513, Eindhoven, The Netherlands
| | - Ahmed Emad Ibrahim Hamouda
- Laboratory of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, 1050, Belgium
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, 1050, Belgium
| | - Shikha Dhiman
- Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology, MB 5600, P.O. Box 513, Eindhoven, The Netherlands
| | - Benoit Louage
- Department of Pharmaceutics, Ghent University, Ghent, 9000, Belgium
| | - Lorenzo Albertazzi
- Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology, MB 5600, P.O. Box 513, Eindhoven, The Netherlands
| | - Damya Laoui
- Laboratory of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, 1050, Belgium
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, 1050, Belgium
| | - E W Meijer
- Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology, MB 5600, P.O. Box 513, Eindhoven, The Netherlands
- School of Chemistry, RNA Institute, University of new South Wales, Sydney, NSW, 1050, Australia
| | - Bruno G De Geest
- Department of Pharmaceutics, Ghent University, Ghent, 9000, Belgium
| |
Collapse
|
4
|
Galili U. Antibody production and tolerance to the α-gal epitope as models for understanding and preventing the immune response to incompatible ABO carbohydrate antigens and for α-gal therapies. Front Mol Biosci 2023; 10:1209974. [PMID: 37449060 PMCID: PMC10338101 DOI: 10.3389/fmolb.2023.1209974] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 06/09/2023] [Indexed: 07/18/2023] Open
Abstract
This review describes the significance of the α-gal epitope (Galα-3Galβ1-4GlcNAc-R) as the core of human blood-group A and B antigens (A and B antigens), determines in mouse models the principles underlying the immune response to these antigens, and suggests future strategies for the induction of immune tolerance to incompatible A and B antigens in human allografts. Carbohydrate antigens, such as ABO antigens and the α-gal epitope, differ from protein antigens in that they do not interact with T cells, but B cells interacting with them require T-cell help for their activation. The α-gal epitope is the core of both A and B antigens and is the ligand of the natural anti-Gal antibody, which is abundant in all humans. In A and O individuals, anti-Gal clones (called anti-Gal/B) comprise >85% of the so-called anti-B activity and bind to the B antigen in facets that do not include fucose-linked α1-2 to the core α-gal. As many as 1% of B cells are anti-Gal B cells. Activation of quiescent anti-Gal B cells upon exposure to α-gal epitopes on xenografts and some protozoa can increase the titer of anti-Gal by 100-fold. α1,3-Galactosyltransferase knockout (GT-KO) mice lack α-gal epitopes and can produce anti-Gal. These mice simulate human recipients of ABO-incompatible human allografts. Exposure for 2-4 weeks of naïve and memory mouse anti-Gal B cells to α-gal epitopes in the heterotopically grafted wild-type (WT) mouse heart results in the elimination of these cells and immune tolerance to this epitope. Shorter exposures of 7 days of anti-Gal B cells to α-gal epitopes in the WT heart result in the production of accommodating anti-Gal antibodies that bind to α-gal epitopes but do not lyse cells or reject the graft. Tolerance to α-gal epitopes due to the elimination of naïve and memory anti-Gal B cells can be further induced by 2 weeks in vivo exposure to WT lymphocytes or autologous lymphocytes engineered to present α-gal epitopes by transduction of the α1,3-galactosyltransferase gene. These mouse studies suggest that autologous human lymphocytes similarly engineered to present the A or B antigen may induce corresponding tolerance in recipients of ABO-incompatible allografts. The review further summarizes experimental works demonstrating the efficacy of α-gal therapies in amplifying anti-viral and anti-tumor immune-protection and regeneration of injured tissues.
Collapse
Affiliation(s)
- Uri Galili
- Department of Medicine, Rush University Medical College, Chicago, IL, United States
| |
Collapse
|
5
|
Kreft L, Schepers A, Hils M, Swiontek K, Flatley A, Janowski R, Mirzaei MK, Dittmar M, Chakrapani N, Desai MS, Eyerich S, Deng L, Niessing D, Fischer K, Feederle R, Blank S, Schmidt-Weber CB, Hilger C, Biedermann T, Ohnmacht C. A novel monoclonal IgG1 antibody specific for Galactose-alpha-1,3-galactose questions alpha-Gal epitope expression by bacteria. Front Immunol 2022; 13:958952. [PMID: 35990627 PMCID: PMC9391071 DOI: 10.3389/fimmu.2022.958952] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 07/18/2022] [Indexed: 11/23/2022] Open
Abstract
The alpha-Gal epitope (α-Gal) with the determining element galactose-α1,3-galactose can lead to clinically relevant allergic reactions and rejections in xenotransplantation. These immune reactions can develop because humans are devoid of this carbohydrate due to evolutionary loss of the enzyme α1,3-galactosyltransferase (GGTA1). In addition, up to 1% of human IgG antibodies are directed against α-Gal, but the stimulus for the induction of anti-α-Gal antibodies is still unclear. Commensal bacteria have been suggested as a causal factor for this induction as α-Gal binding tools such as lectins were found to stain cultivated bacteria isolated from the intestinal tract. Currently available tools for the detection of the definite α-Gal epitope, however, are cross-reactive, or have limited affinity and, hence, offer restricted possibilities for application. In this study, we describe a novel monoclonal IgG1 antibody (27H8) specific for the α-Gal epitope. The 27H8 antibody was generated by immunization of Ggta1 knockout mice and displays a high affinity towards synthetic and naturally occurring α-Gal in various applications. Using this novel tool, we found that intestinal bacteria reported to be α-Gal positive cannot be stained with 27H8 questioning whether commensal bacteria express the native α-Gal epitope at all.
Collapse
Affiliation(s)
- Luisa Kreft
- Center of Allergy and Environment (ZAUM) and Institute of Allergy Research, Technical University of Munich, School of Medicine, and Helmholtz Center Munich, Research Center for Environmental Health, Neuherberg, Germany
| | - Aloys Schepers
- Monoclonal Antibody Core Facility, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Miriam Hils
- Department of Dermatology and Allergy Biederstein, School of Medicine, Technical University of Munich, Munich, Germany
| | - Kyra Swiontek
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| | - Andrew Flatley
- Monoclonal Antibody Core Facility, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Robert Janowski
- Institute of Structural Biology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Mohammadali Khan Mirzaei
- Institute of Virology, Technical University of Munich and Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
- Chair of Prevention of Microbial Diseases, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Michael Dittmar
- Center of Allergy and Environment (ZAUM) and Institute of Allergy Research, Technical University of Munich, School of Medicine, and Helmholtz Center Munich, Research Center for Environmental Health, Neuherberg, Germany
| | - Neera Chakrapani
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| | - Mahesh S. Desai
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
- Odense Research Center for Anaphylaxis, Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, Odense, Denmark
| | - Stefanie Eyerich
- Center of Allergy and Environment (ZAUM) and Institute of Allergy Research, Technical University of Munich, School of Medicine, and Helmholtz Center Munich, Research Center for Environmental Health, Neuherberg, Germany
| | - Li Deng
- Institute of Virology, Technical University of Munich and Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
- Chair of Prevention of Microbial Diseases, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Dierk Niessing
- Institute of Structural Biology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Konrad Fischer
- Chair of Livestock Biotechnology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Regina Feederle
- Monoclonal Antibody Core Facility, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Simon Blank
- Center of Allergy and Environment (ZAUM) and Institute of Allergy Research, Technical University of Munich, School of Medicine, and Helmholtz Center Munich, Research Center for Environmental Health, Neuherberg, Germany
| | - Carsten B. Schmidt-Weber
- Center of Allergy and Environment (ZAUM) and Institute of Allergy Research, Technical University of Munich, School of Medicine, and Helmholtz Center Munich, Research Center for Environmental Health, Neuherberg, Germany
- German Center of Lung Research (DZL), Munich, Germany
| | - Christiane Hilger
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| | - Tilo Biedermann
- Department of Dermatology and Allergy Biederstein, School of Medicine, Technical University of Munich, Munich, Germany
| | - Caspar Ohnmacht
- Center of Allergy and Environment (ZAUM) and Institute of Allergy Research, Technical University of Munich, School of Medicine, and Helmholtz Center Munich, Research Center for Environmental Health, Neuherberg, Germany
- *Correspondence: Caspar Ohnmacht,
| |
Collapse
|
6
|
Galili U. Biosynthesis of α-Gal Epitopes (Galα1-3Galβ1-4GlcNAc-R) and Their Unique Potential in Future α-Gal Therapies. Front Mol Biosci 2021; 8:746883. [PMID: 34805272 PMCID: PMC8601398 DOI: 10.3389/fmolb.2021.746883] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 09/21/2021] [Indexed: 11/19/2022] Open
Abstract
The α-gal epitope is a carbohydrate antigen which appeared early in mammalian evolution and is synthesized in large amounts by the glycosylation enzyme α1,3galactosyltransferase (α1,3GT) in non-primate mammals, lemurs, and New-World monkeys. Ancestral Old-World monkeys and apes synthesizing α-gal epitopes underwent complete extinction 20–30 million years ago, and their mutated progeny lacking α-gal epitopes survived. Humans, apes, and Old-World monkeys which evolved from the surviving progeny lack α-gal epitopes and produce the natural anti-Gal antibody which binds specifically to α-gal epitopes. Because of this reciprocal distribution of the α-gal epitope and anti-Gal in mammals, transplantation of organs from non-primate mammals (e.g., pig xenografts) into Old-World monkeys or humans results in hyperacute rejection following anti-Gal binding to α-gal epitopes on xenograft cells. The in vivo immunocomplexing between anti-Gal and α-gal epitopes on molecules, pathogens, cells, or nanoparticles may be harnessed for development of novel immunotherapies (referred to as “α-gal therapies”) in various clinical settings because such immune complexes induce several beneficial immune processes. These immune processes include localized activation of the complement system which can destroy pathogens and generate chemotactic peptides that recruit antigen-presenting cells (APCs) such as macrophages and dendritic cells, targeting of antigens presenting α-gal epitopes for extensive uptake by APCs, and activation of recruited macrophages into pro-reparative macrophages. Some of the suggested α-gal therapies associated with these immune processes are as follows: 1. Increasing efficacy of enveloped-virus vaccines by synthesizing α-gal epitopes on vaccinating inactivated viruses, thereby targeting them for extensive uptake by APCs. 2. Conversion of autologous tumors into antitumor vaccines by expression of α-gal epitopes on tumor cell membranes. 3. Accelerating healing of external and internal injuries by α-gal nanoparticles which decrease the healing time and diminish scar formation. 4. Increasing anti-Gal–mediated protection against zoonotic viruses presenting α-gal epitopes and against protozoa, such as Trypanosoma, Leishmania, and Plasmodium, by vaccination for elevating production of the anti-Gal antibody. The efficacy and safety of these therapies were demonstrated in transgenic mice and pigs lacking α-gal epitopes and producing anti-Gal, raising the possibility that these α-gal therapies may be considered for further evaluation in clinical trials.
Collapse
Affiliation(s)
- Uri Galili
- Department of Medicine, Rush University Medical Center, Chicago, IL, United States
| |
Collapse
|
7
|
PD-1 blockade synergizes with intratumoral vaccination of a therapeutic HPV protein vaccine and elicits regression of tumor in a preclinical model. Cancer Immunol Immunother 2020; 70:1049-1062. [PMID: 33108473 DOI: 10.1007/s00262-020-02754-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 10/14/2020] [Indexed: 12/19/2022]
Abstract
INTRODUCTION The human papillomavirus (HPV) encoded oncoproteins E6 and E7 are constitutively expressed in HPV-associated cancers, making them logical therapeutic targets. Intramuscular immunization of patients with HPV16 L2E7E6 fusion protein vaccine (TA-CIN) is well tolerated and induces HPV-specific cellular immune responses. Efficacy of PD-1 immune checkpoint blockade correlates with the level of tumor-infiltrating CD8 + T cells, yet most patients lack significant tumor infiltration of immune cells making immune checkpoint blockade suboptimal. We hypothesized that intratumoral vaccination with TA-CIN could increase the number of tumor-infiltrating CD8 + T cells, synergize with PD-1 blockade and result in better control of tumors compared with either PD-1 blockade or vaccination alone. METHODS We examined the immunogenicity and antitumor effects of intratumoral vaccination with TA-CIN alone or in combination with PD-1 blockade in the TC-1 syngeneic murine tumor model expressing HPV16 E6/E7. RESULTS Intratumoral vaccination with TA-CIN induced stronger antigen-specific CD8 + T cell responses and antitumor effects. Intratumoral TA-CIN vaccination generated a systemic immune response that was able to control distal TC-1 tumors. Furthermore, intratumoral TA-CIN vaccination induced tumor infiltration of antigen-specific CD8 + T cells. Knockout of Batf3 abolished antigen-specific CD8 + T cell responses and antitumor effects of intratumoral TA-CIN vaccination. Finally, PD-1 blockade synergizes with intratumoral TA-CIN vaccination resulting in significantly enhanced antigen-specific CD8 + T cell responses and complete regression of tumors, whereas either alone failed to control established TC-1 tumor. CONCLUSIONS Our results provide rationale for future clinical testing of intratumoral TA-CIN vaccination in combination with PD-1 blockade for the control of HPV16-associated tumors.
Collapse
|
8
|
Ayala EV, Rodrigues da Cunha G, Azevedo MA, Calderon M, Jimenez J, Venuto AP, Gazzinelli R, Lavalle RJY, Riva AGV, Hincapie R, Finn MG, Marques AF. C57BL/6 α-1,3-Galactosyltransferase Knockout Mouse as an Animal Model for Experimental Chagas Disease. ACS Infect Dis 2020; 6:1807-1815. [PMID: 32374586 DOI: 10.1021/acsinfecdis.0c00061] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The leading animal model of experimental Chagas disease, the mouse, plays a significant role in studies for vaccine development, diagnosis, and human therapies. Humans, along with Old World primates, alone among mammals, cannot make the terminal carbohydrate linkage of the α-Gal trisaccharide. It has been established that the anti-α-Gal immune response is likely to be a critical factor for protection against Trypanosoma cruzi (T. cruzi) infection in humans. However, the mice customarily employed for the study of T. cruzi infection naturally express the α-Gal epitope and therefore do not produce anti-α-Gal antibodies. Here, we used the C57BL/6 α-1,3-galactosyltransferase knockout (α-GalT-KO) mouse, which does not express the α-Gal epitope as a model for experimental Chagas disease. We found the anti-α-Gal IgG antibody response to an increase in α-GalT-KO mice infected with Arequipa and Colombiana strains of T. cruzi, leading to fewer parasite nests, lower parasitemia, and an increase of INF-γ, TNF-α, and IL-12 cytokines in the heart of α-GalT-KO mice compared with α-GalT-WT mice on days 60 and 120 postinfection. We therefore agree that the C57BL/6 α-GalT-KO mouse represents a useful model for initial testing of therapeutic and immunological approaches against different strains of T. cruzi.
Collapse
Affiliation(s)
- Edward Valencia Ayala
- Laboratório de Imunologia e Genômica de Parasitos, Departamento de Parasitologia, Instituto de Ciências Biológicas/ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270901, Brazil
- Instituto de Investigación, Centro de Investigación en Inmunología e Infectología, Facultad de Medicina Humana, Universidad de San Martin de Porres, Lima 15000, Perú
| | - Gisele Rodrigues da Cunha
- Laboratório de Imuno-Proteômica e Biologia de Parasitos, Departamento de Parasitologia, Instituto de Ciências Biológicas/ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270901, Brazil
| | - Maira Araujo Azevedo
- Laboratório de Imuno-Proteômica e Biologia de Parasitos, Departamento de Parasitologia, Instituto de Ciências Biológicas/ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270901, Brazil
| | - Maritza Calderon
- Laboratorio de Investigación en Enfermedades Infecciosas and Laboratorio de Biología Molecular, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima 15000, Perú
| | - Juan Jimenez
- Laboratorio de Parasitología en Fauna Silvestre y Zoonosis, Facultad de Ciencias Biológicas, Universidad Nacional Mayor de San Marcos, Lima 15000, Perú
| | - Ana Paula Venuto
- Laboratório de Imuno-Proteômica e Biologia de Parasitos, Departamento de Parasitologia, Instituto de Ciências Biológicas/ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270901, Brazil
| | - Ricardo Gazzinelli
- Instituto de Pesquisa Rene Rachou, Fundacao Oswaldo Cruz, Belo Horizonte, Minas Gerais 30190-009, Brazil
- Plataforma de Medicina Translacional, Fundacao Oswaldo Cruz, Belo Horizonte, Minas Gerais 30190-009, Brazil
| | - Raúl Jesus Ynocente Lavalle
- Laboratorio de Parasitología en Fauna Silvestre y Zoonosis, Facultad de Ciencias Biológicas, Universidad Nacional Mayor de San Marcos, Lima 15000, Perú
| | - Angela Giovana Vidal Riva
- Instituto de Investigación, Centro de Investigación en Inmunología e Infectología, Facultad de Medicina Humana, Universidad de San Martin de Porres, Lima 15000, Perú
- Laboratorio de Investigación en Enfermedades Infecciosas and Laboratorio de Biología Molecular, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima 15000, Perú
| | - Robert Hincapie
- School of Chemistry and Biochemistry, School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia 30332 United States
| | - M. G. Finn
- School of Chemistry and Biochemistry, School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia 30332 United States
| | - Alexandre F. Marques
- Laboratório de Imuno-Proteômica e Biologia de Parasitos, Departamento de Parasitologia, Instituto de Ciências Biológicas/ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270901, Brazil
| |
Collapse
|
9
|
Saleh FM, Chandra PK, Lin D, Robinson JE, Izadpanah R, Mondal D, Bollensdorff C, Alt EU, Zhu Q, Marasco WA, Braun SE, Abdel-Motal UM. A New Humanized Mouse Model Mimics Humans in Lacking α-Gal Epitopes and Secreting Anti-Gal Antibodies. THE JOURNAL OF IMMUNOLOGY 2020; 204:1998-2005. [PMID: 32144163 DOI: 10.4049/jimmunol.1901385] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 01/14/2020] [Indexed: 11/19/2022]
Abstract
Mice have been used as accepted tools for investigating complex human diseases and new drug therapies because of their shared genetics and anatomical characteristics with humans. However, the tissues in mice are different from humans in that human cells have a natural mutation in the α1,3 galactosyltransferase (α1,3GT) gene and lack α-Gal epitopes on glycosylated proteins, whereas mice and other nonprimate mammals express this epitope. The lack of α-Gal epitopes in humans results in the loss of immune tolerance to this epitope and production of abundant natural anti-Gal Abs. These natural anti-Gal Abs can be used as an adjuvant to enhance processing of vaccine epitopes to APCs. However, wild-type mice and all existing humanized mouse models cannot be used to test the efficacy of vaccines expressing α-Gal epitopes because they express α-Gal epitopes and lack anti-Gal Abs. Therefore, in an effort to bridge the gap between the mouse models and humans, we developed a new humanized mouse model that mimics humans in that it lacks α-Gal epitopes and secretes human anti-Gal Abs. The new humanized mouse model (Hu-NSG/α-Galnull) is designed to be used for preclinical evaluations of viral and tumor vaccines based on α-Gal epitopes, human-specific immune responses, xenotransplantation studies, and in vivo biomaterials evaluation. To our knowledge, our new Hu-NSG/α-Galnull is the first available humanized mouse model with such features.
Collapse
Affiliation(s)
- Fayez M Saleh
- Division of Immunology, Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA 70433.,Department of Medical Microbiology, Faculty of Medicine, University of Tabuk, Tabuk 71491, Kingdom of Saudi Arabia
| | - Partha K Chandra
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA 70112
| | - Dong Lin
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112
| | - James E Robinson
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, LA 70112
| | - Reza Izadpanah
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112
| | - Debasis Mondal
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA 70112.,Department of Microbiology, Lincoln Memorial University-Debusk College of Osteopathic Medicine, Knoxville, TN 37932
| | | | - Eckhard U Alt
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112
| | - Quan Zhu
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215
| | - Wayne A Marasco
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215
| | - Stephen E Braun
- Division of Immunology, Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA 70433.,Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA 70112
| | - Ussama M Abdel-Motal
- Precision Medicine, Research Branch, Sidra Medicine, Doha, Qatar; and .,Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215
| |
Collapse
|
10
|
Shaw SM, Middleton J, Wigglesworth K, Charlemagne A, Schulz O, Glossop MS, Whalen GF, Old R, Westby M, Pickford C, Tabakman R, Carmi-Levy I, Vainstein A, Sorani E, Zur AA, Kristian SA. AGI-134: a fully synthetic α-Gal glycolipid that converts tumors into in situ autologous vaccines, induces anti-tumor immunity and is synergistic with an anti-PD-1 antibody in mouse melanoma models. Cancer Cell Int 2019; 19:346. [PMID: 31889898 PMCID: PMC6923872 DOI: 10.1186/s12935-019-1059-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 12/05/2019] [Indexed: 02/04/2023] Open
Abstract
Background Treatments that generate T cell-mediated immunity to a patient’s unique neoantigens are the current holy grail of cancer immunotherapy. In particular, treatments that do not require cumbersome and individualized ex vivo processing or manufacturing processes are especially sought after. Here we report that AGI-134, a glycolipid-like small molecule, can be used for coating tumor cells with the xenoantigen Galα1-3Galβ1-4GlcNAc (α-Gal) in situ leading to opsonization with pre-existing natural anti-α-Gal antibodies (in short anti-Gal), which triggers immune cascades resulting in T cell mediated anti-tumor immunity. Methods Various immunological effects of coating tumor cells with α-Gal via AGI-134 in vitro were measured by flow cytometry: (1) opsonization with anti-Gal and complement, (2) antibody-dependent cell-mediated cytotoxicity (ADCC) by NK cells, and (3) phagocytosis and antigen cross-presentation by antigen presenting cells (APCs). A viability kit was used to test AGI-134 mediated complement dependent cytotoxicity (CDC) in cancer cells. The anti-tumoral activity of AGI-134 alone or in combination with an anti-programmed death-1 (anti-PD-1) antibody was tested in melanoma models in anti-Gal expressing galactosyltransferase knockout (α1,3GT−/−) mice. CDC and phagocytosis data were analyzed by one-way ANOVA, ADCC results by paired t-test, distal tumor growth by Mantel–Cox test, C5a data by Mann–Whitney test, and single tumor regression by repeated measures analysis. Results In vitro, α-Gal labelling of tumor cells via AGI-134 incorporation into the cell membrane leads to anti-Gal binding and complement activation. Through the effects of complement and ADCC, tumor cells are lysed and tumor antigen uptake by APCs increased. Antigen associated with lysed cells is cross-presented by CD8α+ dendritic cells leading to activation of antigen-specific CD8+ T cells. In B16-F10 or JB/RH melanoma models in α1,3GT−/− mice, intratumoral AGI-134 administration leads to primary tumor regression and has a robust abscopal effect, i.e., it protects from the development of distal, uninjected lesions. Combinations of AGI-134 and anti-PD-1 antibody shows a synergistic benefit in protection from secondary tumor growth. Conclusions We have identified AGI-134 as an immunotherapeutic drug candidate, which could be an excellent combination partner for anti-PD-1 therapy, by facilitating tumor antigen processing and increasing the repertoire of tumor-specific T cells prior to anti-PD-1 treatment.
Collapse
Affiliation(s)
- Stephen M Shaw
- Agalimmune Ltd., Sandwich, Kent, UK.,BioLineRx Ltd, Modi'in-Maccabim-Re'ut, Israel
| | - Jenny Middleton
- Agalimmune Ltd., Sandwich, Kent, UK.,BioLineRx Ltd, Modi'in-Maccabim-Re'ut, Israel
| | - Kim Wigglesworth
- 3Department of Surgery, University of Massachusetts Medical School, Worcester, MA USA
| | | | - Oliver Schulz
- 4Immunobiology Laboratory, The Francis Crick Institute, London, UK
| | | | - Giles F Whalen
- 3Department of Surgery, University of Massachusetts Medical School, Worcester, MA USA.,5Department of Cancer Biology, University of Massachusetts Medical School, Worcester, MA USA
| | - Robert Old
- 6Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, UK
| | | | | | | | | | | | - Ella Sorani
- BioLineRx Ltd, Modi'in-Maccabim-Re'ut, Israel
| | - Arik A Zur
- BioLineRx Ltd, Modi'in-Maccabim-Re'ut, Israel
| | | |
Collapse
|
11
|
Zhang Y, Xu J, Zhang N, Chen M, Wang H, Zhu D. Targeting the tumour immune microenvironment for cancer therapy in human gastrointestinal malignancies. Cancer Lett 2019; 458:123-135. [DOI: 10.1016/j.canlet.2019.05.017] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 05/11/2019] [Accepted: 05/14/2019] [Indexed: 12/12/2022]
|
12
|
Mo F, Xue D, Duan S, Liu A, Yang X, Hou X, Lu X. Novel fusion cells derived from tumor cells expressing the heterologous α-galactose epitope and dendritic cells effectively target cancer. Vaccine 2019; 37:926-936. [PMID: 30661833 DOI: 10.1016/j.vaccine.2019.01.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 11/17/2018] [Accepted: 01/04/2019] [Indexed: 11/18/2022]
Abstract
Tumor cells/dendritic cells (DCs) fusion cells (tumor/DC) represent a promising immunotherapeutic strategy but are still under performed in clinical trials for cancer treatment. To further boost their anticancer efficacy, here we developed a novel design for fusing dendritic cells with MDA-MB-231 cells expressing the heterologous α-galactose (α-gal) epitope and assessed its anticancer activities both in vitro and in vivo. The high expression of α-gal in MDA-MB-231 (Gal+)/DC correlated with enhanced DC activation. When applied to T cells, MDA-MB-231 (Gal+)/DC significantly stimulated T-cell proliferation and activation, promoted productions of cytokines IL-2 and IFN-γ, and enhanced T-cell-mediated cytotoxicity against MDA-MB-231 cells. MDA-MB-231 (Gal+)/DC inhibited proliferation and promoted apoptosis of tumor cells in vivo, prolonged mouse survival, and significantly boosted anticancer immunity by increasing CD4+ and CD8+ T cells systemically and elevating serum levels of cytokines and IgG. These results suggested that fusing dendritic cells with tumor cells expressing the heterologous α-gal epitope provides a novel therapeutic strategy for cancer treatment.
Collapse
Affiliation(s)
- Fengzhen Mo
- School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi 530021, China; Intenational Nanobody Research Center of Guangxi, Guangxi Medical University, Nanning, Guangxi 530021, China; Pharmaceutical College, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Dabing Xue
- School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi 530021, China; Intenational Nanobody Research Center of Guangxi, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Siliang Duan
- School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi 530021, China; Intenational Nanobody Research Center of Guangxi, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Aiqun Liu
- Intenational Nanobody Research Center of Guangxi, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Xiaomei Yang
- School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi 530021, China; Intenational Nanobody Research Center of Guangxi, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Xiaoqiong Hou
- School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi 530021, China; Intenational Nanobody Research Center of Guangxi, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Xiaoling Lu
- School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi 530021, China; Intenational Nanobody Research Center of Guangxi, Guangxi Medical University, Nanning, Guangxi 530021, China; National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, China.
| |
Collapse
|
13
|
Affiliation(s)
- Stephen M. Henry
- Center for Kode Technology Innovation, School of Engineering, Computer & Mathematical Sciences, Auckland University of Technology, Auckland, New Zealand
| | - Nicolai V. Bovin
- Center for Kode Technology Innovation, School of Engineering, Computer & Mathematical Sciences, Auckland University of Technology, Auckland, New Zealand
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russian Federation
| |
Collapse
|
14
|
Hapten-mediated recruitment of polyclonal antibodies to tumors engenders antitumor immunity. Nat Commun 2018; 9:3348. [PMID: 30135425 PMCID: PMC6105580 DOI: 10.1038/s41467-018-05566-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 07/11/2018] [Indexed: 12/30/2022] Open
Abstract
Uptake of tumor antigens by tumor-infiltrating dendritic cells is limiting step in the induction of tumor immunity, which can be mediated through Fc receptor (FcR) triggering by antibody-coated tumor cells. Here we describe an approach to potentiate tumor immunity whereby hapten-specific polyclonal antibodies are recruited to tumors by coating tumor cells with the hapten. Vaccination of mice against dinitrophenol (DNP) followed by systemic administration of DNP targeted to tumors by conjugation to a VEGF or osteopontin aptamer elicits potent FcR dependent, T cell mediated, antitumor immunity. Recruitment of αGal-specific antibodies, the most abundant naturally occurring antibodies in human serum, inhibits tumor growth in mice treated with a VEGF aptamer-αGal hapten conjugate, and recruits antibodies from human serum to human tumor biopsies of distinct origin. Thus, treatment with αGal hapten conjugated to broad-spectrum tumor targeting ligands could enhance the susceptibility of a broad range of tumors to immune elimination.
Collapse
|
15
|
Galili U. Natural anti-carbohydrate antibodies contributing to evolutionary survival of primates in viral epidemics? Glycobiology 2016; 26:1140-1150. [PMID: 27567275 DOI: 10.1093/glycob/cww088] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Revised: 08/09/2016] [Accepted: 08/16/2016] [Indexed: 12/17/2022] Open
Abstract
Humans produce multiple natural antibodies against carbohydrate antigens on gastrointestinal bacteria. Two such antibodies appeared in primates in recent geological times. Anti-Gal, abundant in humans, apes and Old-World monkeys, appeared 20-30 million years ago (mya) following inactivation of the α1,3GT gene (GGTA1). This gene encodes in other mammals the enzyme α1,3galactosyltransferase (α1,3GT) that synthesizes α-gal epitopes (Galα1-3Galβ1-4GlcNAc-R) which bind anti-Gal. Anti-Neu5Gc, found only in humans, appeared in hominins <6 mya, following elimination of N-glycolylneuraminic-acid (Neu5Gc) because of inactivation of CMAH, the gene encoding hydroxylase that converts N-acetylneuraminic-acid (Neu5Ac) into Neu5Gc. These antibodies, were initially produced in few individuals that acquired random mutations inactivating the corresponding genes and eliminating α-gal epitopes or Neu5Gc, which became nonself antigens. It is suggested that these evolutionary selection events were induced by epidemics of enveloped viruses, lethal to ancestral Old World primates or hominins. Such viruses presented α-gal epitopes or Neu5Gc, synthesized in primates that conserved active GGTA1 or CMAH, respectively, and were lethal to their hosts. The natural anti-Gal or anti-Neu5Gc antibodies, produced in offspring lacking the corresponding carbohydrate antigens, neutralized and destroyed viruses presenting α-gal epitopes or Neu5Gc. These antibodies further induced rapid, effective immune responses against virus antigens, thus preventing infections from reaching lethal stages. These epidemics ultimately resulted in extinction of primate populations synthesizing these carbohydrate antigens and their replacement with offspring populations lacking the antigens and producing protective antibodies against them. Similar events could mediate the elimination of various carbohydrate antigens, thus preventing the complete extinction of other vertebrate species.
Collapse
Affiliation(s)
- Uri Galili
- University of Massachusetts Medical School, Worcester, MA, USA
| |
Collapse
|
16
|
Albertini MR, Ranheim EA, Zuleger CL, Sondel PM, Hank JA, Bridges A, Newton MA, McFarland T, Collins J, Clements E, Henry MB, Neuman HB, Weber S, Whalen G, Galili U. Phase I study to evaluate toxicity and feasibility of intratumoral injection of α-gal glycolipids in patients with advanced melanoma. Cancer Immunol Immunother 2016; 65:897-907. [PMID: 27207605 DOI: 10.1007/s00262-016-1846-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 05/08/2016] [Indexed: 12/14/2022]
Abstract
Effective uptake of tumor cell-derived antigens by antigen-presenting cells is achieved pre-clinically by in situ labeling of tumor with α-gal glycolipids that bind the naturally occurring anti-Gal antibody. We evaluated toxicity and feasibility of intratumoral injections of α-gal glycolipids as an autologous tumor antigen-targeted immunotherapy in melanoma patients (pts). Pts with unresectable metastatic melanoma, at least one cutaneous, subcutaneous, or palpable lymph node metastasis, and serum anti-Gal titer ≥1:50 were eligible for two intratumoral α-gal glycolipid injections given 4 weeks apart (cohort I: 0.1 mg/injection; cohort II: 1.0 mg/injection; cohort III: 10 mg/injection). Monitoring included blood for clinical, autoimmune, and immunological analyses and core tumor biopsies. Treatment outcome was determined 8 weeks after the first α-gal glycolipid injection. Nine pts received two intratumoral injections of α-gal glycolipids (3 pts/cohort). Injection-site toxicity was mild, and no systemic toxicity or autoimmunity could be attributed to the therapy. Two pts had stable disease by RECIST lasting 8 and 7 months. Tumor nodule biopsies revealed minimal to no change in inflammatory infiltrate between pre- and post-treatment biopsies except for 1 pt (cohort III) with a post-treatment inflammatory infiltrate. Two and four weeks post-injection, treated nodules in 5 of 9 pts exhibited tumor cell necrosis without neutrophilic or lymphocytic inflammatory response. Non-treated tumor nodules in 2 of 4 evaluable pts also showed necrosis. Repeated intratumoral injections of α-gal glycolipids are well tolerated, and tumor necrosis was seen in some tumor nodule biopsies after tumor injection with α-gal glycolipids.
Collapse
Affiliation(s)
- Mark R Albertini
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA.
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
- Medical Service, William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.
- University of Wisconsin Clinical Sciences Center, Room K6/530, 600 Highland Avenue, Madison, WI, 53792, USA.
| | - Erik A Ranheim
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
- Department of Pathology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Cindy L Zuleger
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Paul M Sondel
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Jacquelyn A Hank
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Alan Bridges
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Medical Service, William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Michael A Newton
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
- Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Thomas McFarland
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | | | - Erin Clements
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Mary Beth Henry
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Heather B Neuman
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Sharon Weber
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Giles Whalen
- Department of Surgery, University of Massachusetts Medical School, Worcester, MA, USA
| | - Uri Galili
- Department of Surgery, University of Massachusetts Medical School, Worcester, MA, USA
| |
Collapse
|
17
|
Araujo RN, Franco PF, Rodrigues H, Santos LCB, McKay CS, Sanhueza CA, Brito CRN, Azevedo MA, Venuto AP, Cowan PJ, Almeida IC, Finn MG, Marques AF. Amblyomma sculptum tick saliva: α-Gal identification, antibody response and possible association with red meat allergy in Brazil. Int J Parasitol 2016; 46:213-220. [PMID: 26812026 DOI: 10.1016/j.ijpara.2015.12.005] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Revised: 12/04/2015] [Accepted: 12/10/2015] [Indexed: 01/08/2023]
Abstract
The anaphylaxis response is frequently associated with food allergies, representing a significant public health hazard. Recently, exposure to tick bites and production of specific IgE against α-galactosyl (α-Gal)-containing epitopes has been correlated to red meat allergy. However, this association and the source of terminal, non-reducing α-Gal-containing epitopes have not previously been established in Brazil. Here, we employed the α-1,3-galactosyltransferase knockout mouse (α1,3-GalT-KO) model and bacteriophage Qβ-virus like particles (Qβ-VLPs) displaying Galα1,3Galβ1,4GlcNAc (Galα3LN) epitopes to investigate the presence of α-Gal-containing epitopes in the saliva of Amblyomma sculptum, a species of the Amblyomma cajennense complex, which represents the main tick that infests humans in Brazil. We confirmed that the α-1,3-galactosyltransferase knockout animals produce significant levels of anti-α-Gal antibodies against the Galα1,3Galβ1,4GlcNAc epitopes displayed on Qβ-virus like particles. The injection of A. sculptum saliva or exposure to feeding ticks was also found to induce both IgG and IgE anti-α-Gal antibodies in α-1,3-galactosyltransferase knockout mice, thus indicating the presence of α-Gal-containing epitopes in the tick saliva. The presence of α-Gal-containing epitopes was confirmed by ELISA and immunoblotting following removal of terminal α-Gal epitopes by α-galactosidase treatment. These results suggest for the first known time that bites from the A. sculptum tick may be associated with the unknown etiology of allergic reactions to red meat in Brazil.
Collapse
Affiliation(s)
- Ricardo Nascimento Araujo
- Universidade Federal de Minas Gerais, Instituto de Ciencias Biologicas, Departamento de Parasitologia, Pampulha, Belo Horizonte, Minas Gerais, Brazil
| | - Paula Ferreira Franco
- Universidade Federal de Minas Gerais, Instituto de Ciencias Biologicas, Departamento de Parasitologia, Pampulha, Belo Horizonte, Minas Gerais, Brazil
| | - Henrique Rodrigues
- Universidade Federal de Minas Gerais, Instituto de Ciencias Biologicas, Departamento de Parasitologia, Pampulha, Belo Horizonte, Minas Gerais, Brazil
| | - Luiza C B Santos
- Universidade Federal de Minas Gerais, Instituto de Ciencias Biologicas, Departamento de Parasitologia, Pampulha, Belo Horizonte, Minas Gerais, Brazil
| | - Craig S McKay
- Georgia Institute of Technology, School of Chemistry and Biochemistry, Atlanta, GA, USA
| | - Carlos A Sanhueza
- Georgia Institute of Technology, School of Chemistry and Biochemistry, Atlanta, GA, USA
| | - Carlos Ramon Nascimento Brito
- Universidade Federal de Minas Gerais, Instituto de Ciencias Biologicas, Departamento de Parasitologia, Pampulha, Belo Horizonte, Minas Gerais, Brazil
| | - Maíra Araújo Azevedo
- Universidade Federal de Minas Gerais, Instituto de Ciencias Biologicas, Departamento de Parasitologia, Pampulha, Belo Horizonte, Minas Gerais, Brazil
| | - Ana Paula Venuto
- Universidade Federal de Minas Gerais, Instituto de Ciencias Biologicas, Departamento de Parasitologia, Pampulha, Belo Horizonte, Minas Gerais, Brazil
| | - Peter J Cowan
- Immunology Research Centre, St. Vincent's Hospital, Fitzroy, Melbourne, VIC 3065, Australia; Department of Medicine, University of Melbourne, Parkville, VIC 3052, Australia
| | - Igor C Almeida
- Border Biomedical Research Center, Department of Biological Sciences, University of Texas at El Paso, El Paso, TX 79912, USA
| | - M G Finn
- Georgia Institute of Technology, School of Chemistry and Biochemistry, Atlanta, GA, USA
| | - Alexandre F Marques
- Universidade Federal de Minas Gerais, Instituto de Ciencias Biologicas, Departamento de Parasitologia, Pampulha, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
18
|
Huai G, Qi P, Yang H, Wang Y. Characteristics of α-Gal epitope, anti-Gal antibody, α1,3 galactosyltransferase and its clinical exploitation (Review). Int J Mol Med 2015; 37:11-20. [PMID: 26531137 PMCID: PMC4687435 DOI: 10.3892/ijmm.2015.2397] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2015] [Accepted: 10/08/2015] [Indexed: 12/15/2022] Open
Abstract
The α-Gal epitope (Galα1,3Galα1,4GlcNAc-R) is ubiquitously presented in non-primate mammals, marsupials and New World Monkeys, but it is absent in humans, apes and Old World monkeys. However, the anti-Gal antibody (~1% of immunoglobulins) is naturally generated in human, and is found as the immunoglobulin G (IgG), IgM and IgA isotypes. Owing to the specific binding of the anti-Gal antibody with the α-Gal epitope, humans have a distinct anti-α-gal reactivity, which is responsible for hyperacute rejection of organs transplanted from α-gal donors. In addition, the α1,3 galactosyltransferases (α1,3GT) can catalyze the synthesis of the α-Gal epitope. Therefore, the α1,3GT gene, which encodes the α1,3GT, is developed profoundly. The distributions of the α-Gal epitope and anti-Gal antibody, and the activation of α1,3GT, reveal that the enzyme of α1,3GT in ancestral primates is ineffective. Comparison of the nucleotide sequence of the human α1,3-GT pseudogene to the corresponding different species sequence, and according to the evolutionary tree of different species, the results of evolutionary inactivation of the α1,3GT gene in ancestral primates attribute to the mutations under a stronger selective pressure. However, on the basis of the structure, the mechanism and the specificity of the α-Gal epitope and anti-Gal antibody, they can be applied to clinical exploitation. Knocking out the α1,3GT gene will eliminate the xenoantigen, Gal(α1,3)Gal, so that the transplantation of α1,3GT gene knockout pig organ into human becomes a potential clinically acceptable treatment for solving the problem of organ shortage. By contrast, the α-Gal epitope expressed through the application of chemical, biochemical and genetic engineering can be exploited for the clinical use. Targeting anti-Gal-mediated autologous tumor vaccines, which express α-Gal epitope to antigen-presenting cells, would increase their immunogenicity and elicit an immune response, which will be potent enough to eradicate the residual tumor cells. For tumor vaccines, the way of increasing immunogenicity of certain viral vaccines, including flu vaccines and human immunodeficiency virus vaccines, can also be used in the elderly. Recently, α-Gal epitope nanoparticles have been applied to accelerate wound healing and further directions on regeneration of internally injured tissues.
Collapse
Affiliation(s)
- Guoli Huai
- Department of Biomedical Engineering, Medical School of University of Electronic Science and Technology of China, Chengdu, Sichuan 610054, P.R. China
| | - Ping Qi
- Department of Pediatrics, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| | - Hongji Yang
- Department of Biomedical Engineering, Medical School of University of Electronic Science and Technology of China, Chengdu, Sichuan 610054, P.R. China
| | - Yi Wang
- Department of Pharmacy, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| |
Collapse
|
19
|
Tanemura M, Miyoshi E, Nagano H, Eguchi H, Matsunami K, Taniyama K, Hatanaka N, Akamatsu H, Mori M, Doki Y. Cancer immunotherapy for pancreatic cancer utilizing α-gal epitope/natural anti-Gal antibody reaction. World J Gastroenterol 2015; 21:11396-11410. [PMID: 26523105 PMCID: PMC4616216 DOI: 10.3748/wjg.v21.i40.11396] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 07/02/2015] [Accepted: 08/30/2015] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has the poorest prognosis of all malignancies and is largely resistant to standard therapy. Novel treatments against PDAC are desperately needed. Anti-Gal is the most abundant natural antibody in humans, comprising about 1% of immunoglobulins and is also naturally produced in apes and Old World monkeys. The anti-Gal ligand is a carbohydrate antigen called "α-gal epitopes" with the structure Galα1-3Galβ1-4GlcNAc-R. These epitopes are expressed as major carbohydrate antigens in non-primate mammals, prosimians, and New World monkeys. Anti-Gal is exploited in cancer vaccines to increase the immunogenicity of antigen-presenting cells (APCs). Cancer cells or PDAC tumor lysates are processed to express α-gal epitopes. Vaccination with these components results in in vivo opsonization by anti-Gal IgG in PDAC patients. The Fc portion of the vaccine-bound anti-Gal interacts with Fcγ receptors of APCs, inducing uptake of the vaccine components, transport of the vaccine tumor membranes to draining lymph nodes, and processing and presentation of tumor-associated antigens (TAAs). Cancer vaccines expressing α-gal epitopes elicit strong antibody production against multiple TAAs contained in PDAC cells and induce activation of multiple tumor-specific T cells. Here, we review new areas of clinical importance related to the α-gal epitope/anti-Gal antibody reaction and the advantages in immunotherapy against PDAC.
Collapse
|
20
|
Galili U. In situ conversion of tumors into autologous tumor-associated antigen vaccines by intratumoral injection of α-gal glycolipids. Oncoimmunology 2014; 2:e22449. [PMID: 23483105 PMCID: PMC3583920 DOI: 10.4161/onci.22449] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The immunogenicity of autologous tumor-associated antigens (TAAs) is markedly increased upon the intratumoral injection of α-gal glycolipids, which insert into tumor cell membranes. The binding of natural anti-Gal antibodies to these glycolipids activates the complement system and recruits antigen-presenting cells (APCs), which internalize anti-Gal-coated tumor cells upon Fc/FcγR interactions. Eventually, TAA-derived peptides presented by APCs activate a T cell-mediated tumor-specific immune response.
Collapse
Affiliation(s)
- Uri Galili
- Departments of Surgery; University of Massachusetts Medical School; Worcester, MA USA
| |
Collapse
|
21
|
Galili U. Anti-Gal: an abundant human natural antibody of multiple pathogeneses and clinical benefits. Immunology 2013; 140:1-11. [PMID: 23578170 DOI: 10.1111/imm.12110] [Citation(s) in RCA: 178] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Revised: 04/03/2013] [Accepted: 04/08/2013] [Indexed: 12/26/2022] Open
Abstract
Anti-Gal is the most abundant natural antibody in humans, constituting ~ 1% of immunoglobulins. Anti-Gal is naturally produced also in apes and Old World monkeys. The ligand of anti-Gal is a carbohydrate antigen called the 'α-gal epitope' with the structure Galα1-3Galβ1-4GlcNAc-R. The α-gal epitope is present as a major carbohydrate antigen in non-primate mammals, prosimians and New World monkeys. Anti-Gal can contributes to several immunological pathogeneses. Anti-Gal IgE produced in some individuals causes allergies to meat and to the therapeutic monoclonal antibody cetuximab, all presenting α-gal epitopes. Aberrant expression of the α-gal epitope or of antigens mimicking it in humans may result in autoimmune processes, as in Graves' disease. α-Gal epitopes produced by Trypanosoma cruzi interact with anti-Gal and induce 'autoimmune like' inflammatory reactions in Chagas' disease. Anti-Gal IgM and IgG further mediate rejection of xenografts expressing α-gal epitopes. Because of its abundance, anti-Gal may be exploited for various clinical uses. It increases immunogenicity of microbial vaccines (e.g. influenza vaccine) presenting α-gal epitopes by targeting them for effective uptake by antigen-presenting cells. Tumour lesions are converted into vaccines against autologous tumour-associated antigens by intra-tumoral injection of α-gal glycolipids, which insert into tumour cell membranes. Anti-Gal binding to α-gal epitopes on tumour cells targets them for uptake by antigen-presenting cells. Accelerated wound healing is achieved by application of α-gal nanoparticles, which bind anti-Gal, activate complement, and recruit and activate macrophages that induce tissue regeneration. This therapy may be of further significance in regeneration of internally injured tissues such as ischaemic myocardium and injured nerves.
Collapse
Affiliation(s)
- Uri Galili
- Department of Surgery, University of Massachusetts Medical School, Worcester, MA 01655, USA.
| |
Collapse
|
22
|
Blockade of invariant TCR-CD1d interaction specifically inhibits antibody production against blood group A carbohydrates. Blood 2013; 122:2582-90. [PMID: 23943651 DOI: 10.1182/blood-2012-02-407452] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Previously, we detected B cells expressing receptors for blood group A carbohydrates in the CD11b(+)CD5(+) B-1a subpopulation in mice, similar to that in blood group O or B in humans. In the present study, we demonstrate that CD1d-restricted natural killer T (NKT) cells are required to produce anti-A antibodies (Abs), probably through collaboration with B-1a cells. After immunization of wild-type (WT) mice with human blood group A red blood cells (A-RBCs), interleukin (IL)-5 exclusively and transiently increased and the anti-A Abs were elevated in sera. However, these reactions were not observed in CD1d(-/-) mice, which lack NKT cells. Administration of anti-mouse CD1d blocking monoclonal Abs (mAb) prior to immunization abolished IL-5 production by NKT cells and anti-A Ab production in WT mice. Administration of anti-IL-5 neutralizing mAb also diminished anti-A Ab production in WT mice, suggesting that IL-5 secreted from NKT cells critically regulates anti-A Ab production by B-1a cells. In nonobese diabetic/severe combined immunodeficient (NOD/SCID/γc(null)) mice, into which peripheral blood mononuclear cells from type O human volunteers were engrafted, administration of anti-human CD1d mAb prior to A-RBC immunization completely inhibited anti-A Ab production. Thus, anti-CD1d treatment might constitute a novel approach that could help in evading Ab-mediated rejection in ABO-incompatible transplant recipients.
Collapse
|
23
|
Galili U. Discovery of the natural anti-Gal antibody and its past and future relevance to medicine. Xenotransplantation 2013; 20:138-47. [PMID: 23577774 DOI: 10.1111/xen.12034] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Accepted: 03/15/2013] [Indexed: 12/11/2022]
Abstract
This is a personal account of the discovery of the natural anti-Gal antibody, the most abundant natural antibody in humans, the reciprocal distribution of this antibody and its ligand the α-gal epitope in mammals and the immunological barrier this antibody has formed in porcine to human xenotransplantation. This barrier has been overcome in the recent decade with the generation of α1,3-galactosyltransferase gene-knockout pigs. However, anti-Gal continues to be relevant in medicine as it can be harnessed for various therapeutic effects. Anti-Gal converts tumor lesions injected with α-gal glycolipids into vaccines that elicit a protective anti-tumor immune response by in situ targeting of tumor cells for uptake by antigen-presenting cells. This antibody further accelerates wound and burn healing by interaction with α-gal nanoparticles applied to injured areas and induction of rapid recruitment and activation of macrophages. Anti-Gal/α-gal nanoparticle immune complexes may further induce rapid recruitment and activation of macrophages in ischemic myocardium and injured nerves, thereby inducing tissue regeneration and prevention of fibrosis.
Collapse
Affiliation(s)
- Uri Galili
- Department of Surgery, University of Massachusetts Medical School, Worcester, MA, USA
| |
Collapse
|
24
|
Tanemura M, Miyoshi E, Nagano H, Eguchi H, Taniyama K, Kamiike W, Mori M, Doki Y. Role of α-gal epitope/anti-Gal antibody reaction in immunotherapy and its clinical application in pancreatic cancer. Cancer Sci 2013; 104:282-290. [PMID: 23240661 PMCID: PMC7657191 DOI: 10.1111/cas.12084] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Revised: 12/04/2012] [Accepted: 12/10/2012] [Indexed: 12/25/2022] Open
Abstract
Pancreatic cancer is one of the most common causes of death from cancer. Despite the availability of various treatment modalities, such as surgery, chemotherapy and radiotherapy, the 5-year survival remains poor. Although gemcitabine-based chemotherapy is typically offered as the standard care, most patients do not survive longer than 6 months. Therefore, new therapeutic approaches are needed. The α-gal epitope (Galα1-3Galβ1-4GlcNAc-R) is abundantly synthesized from glycoproteins and glycolipids in non-primate mammals and New World monkeys, but is absent in humans, apes and Old World monkeys. Instead, they produce anti-Gal antibody (Ab) (forming approximately 1% of circulating immunoglobulins), which specifically interacts with α-gal epitopes. Anti-Gal Ab can be exploited in cancer immunotherapy as vaccines that target antigen-presenting cells (APC) to increase their immunogenicity. Tumor cells or tumor cell membranes from pancreatic cancer are processed to express α-gal epitopes. Subsequent vaccination with such processed cell membranes results in in vivo opsonization by anti-Gal IgG in cancer patients. The interaction of the Fc portion of the vaccine-bound anti-Gal with Fcγ receptors of APC induces effective uptake of the vaccinating tumor cell membranes by the APC, followed by effective transport of the vaccinating tumor membranes to the regional lymph nodes, and processing and presentation of the tumor-associated antigens. Activation of tumor-specific B and T cells could elicit an immune response that in some patients is potent enough to eradicate the residual cancer cells that remain after completion of standard therapy. This review addresses these topics and new avenues of clinical importance related to this unique antigen/antibody system (α-gal epitope/anti-Gal Ab) and advances in immunotherapy in pancreatic cancer.
Collapse
Affiliation(s)
- Masahiro Tanemura
- Department of Surgery and Institute for Clinical Research, National Hospital Organization Kure Medical Center and Chugoku Cancer Center, Hiroshima, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Koevary SB. Selective toxicity of rose bengal to ovarian cancer cells in vitro. INTERNATIONAL JOURNAL OF PHYSIOLOGY, PATHOPHYSIOLOGY AND PHARMACOLOGY 2012; 4:99-107. [PMID: 22837809 PMCID: PMC3403562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Accepted: 06/23/2012] [Indexed: 06/01/2023]
Abstract
Rose bengal (RB) has been utilized as a photodynamic agent for the targeted killing of cancer cells. Recent data suggest that intralesional RB alone may be effective in chemoablating locoregional and metastatic melanomas. The ability of RB to induce direct and bystander melanoma cell death led to the speculation that it may be similarly effective in the treatment of other neoplasms. The objective of this study was to determine whether RB can limit the growth, or kill, ovarian cancer cells in vitro. Ovarian carcinoma cells with or without a germline BRCA1 mutation were cultured with up to 800 μM RB for one hour or four days, after which their ability to proliferate was assessed using the MTT assay. Control cells included an embryonic kidney cell line transformed with adenovirus, and normal human fibroblasts. Ovarian cancer cells exhibited significant dose-dependent suppression of growth in response to RB; this suppression was similar to that seen with carboplatin. RB treated ovarian cancer cells appeared rounded, shrunken, and damaged. RB also inhibited the growth of kidney tumor cells but was much less effective in slowing the growth of normal human fibroblasts suggesting that RB-mediated growth suppression might be tumor cell specific. Ovarian cancer cells treated with RB displayed a significant increase in apoptosis that peaked at approximately four times the levels seen in untreated control cells. Furthermore, RB exposure resulted in the intracellular generation of reactive oxygen species (ROS) at levels that were significantly greater than in untreated cells and similar to levels seen in cells treated short term with H(2)O(2). These data suggest that RB may not only suppress ovarian cancer cell growth but also induce their apoptotic cell death, justifying the further investigation of the effects of RB in an animal model of ovarian cancer.
Collapse
Affiliation(s)
- Steven B Koevary
- Department of Biomedical Sciences and Disease, New England College of Optometry424 Beacon Street, Boston, MA 02115
- Department of Cell Biology, University of Massachusetts Medical School55 Lake Avenue North, Worcester, MA 01655, USA
| |
Collapse
|
26
|
Conversion of tumors into autologous vaccines by intratumoral injection of α-Gal glycolipids that induce anti-Gal/α-Gal epitope interaction. Clin Dev Immunol 2011; 2011:134020. [PMID: 22162709 PMCID: PMC3226304 DOI: 10.1155/2011/134020] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2011] [Accepted: 09/05/2011] [Indexed: 02/06/2023]
Abstract
Anti-Gal is the most abundant antibody in humans, constituting 1% of immunoglobulins. Anti-Gal binds specifically α-gal epitopes (Galα1-3Galβ1-4GlcNAc-R). Immunogenicity of autologous tumor associated antigens (TAA) is greatly increased by manipulating tumor cells to express α-gal epitopes and bind anti-Gal. Glycolipids with αgal epitopes (α-gal glycolipids) injected into tumors insert into the tumor cell membrane. Anti-Gal binding to the multiple α-gal epitopes de novo presented on the tumor cells results in targeting of these cells to APC via the interaction between the Fc portion of the bound anti-Gal and Fcγ; receptors on APC. The APC process and present immunogenic TAA peptides and thus, effectively activate tumor specific CD4+ helper T cells and CD8+ cytotoxic T cells which destroy tumor cells in micrometastases. The induced immune response is potent enough to overcome immunosuppression by Treg cells. A phase I clinical trial indicated that α-gal glycolipid treatment has no adverse effects. In addition to achieving destruction of micrometastases in cancer patients with advance disease, α-gal glycolipid treatment may be effective as neo-adjuvant immunotherapy. Injection of α-gal glycolipids into primary tumors few weeks prior to resection can induce a protective immune response capable of destroying micrometastases expressing autologous TAA, long after primary tumor resection.
Collapse
|
27
|
Rahyab AS, Alam A, Kapoor A, Zhang M. Natural antibody - Biochemistry and functions. GLOBAL JOURNAL OF BIOCHEMISTRY 2011; 2:283-288. [PMID: 25309852 PMCID: PMC4189112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Natural antibodies have been common knowledge in the scientific community for more than half a century. Initially disregarded, their functions have garnered a newfound interest recently. Natural antibodies are usually polyreactive IgM antibodies and are implicated in numerous physiologic and pathologic processes. Current research demonstrates they play a role in adaptive and innate immune responses, autoimmunity, and apoptosis. Evidence exists that they are involved in the modulation of neurodegenerative disorders and malignancy. Furthermore, natural antibodies have been implicated in ischemia reperfusion injury and atherosclerosis. As such the study of natural antibodies may provide new insight into normal physiologic processes whilst concurrently paving the road for a wide-range of possible therapeutic options.
Collapse
Affiliation(s)
- Ali Seyar Rahyab
- Department of Anesthesiology, State University of NewYork Downstate Medical Center, Brooklyn, NY 11203
| | - Amit Alam
- Department of Anesthesiology, State University of NewYork Downstate Medical Center, Brooklyn, NY 11203
| | | | - Ming Zhang
- Department of Anesthesiology, State University of NewYork Downstate Medical Center, Brooklyn, NY 11203
- Department of Cell Biology, State University of NewYork Downstate Medical Center, Brooklyn, NY 11203
| |
Collapse
|
28
|
Deguchi T, Tanemura M, Miyoshi E, Nagano H, Machida T, Ohmura Y, Kobayashi S, Marubashi S, Eguchi H, Takeda Y, Ito T, Mori M, Doki Y, Sawa Y. Increased immunogenicity of tumor-associated antigen, mucin 1, engineered to express alpha-gal epitopes: a novel approach to immunotherapy in pancreatic cancer. Cancer Res 2010; 70:5259-5269. [PMID: 20530670 DOI: 10.1158/0008-5472.can-09-4313] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mucin 1 (MUC1), a bound mucin glycoprotein, is overexpressed and aberrantly glycosylated in >80% of human ductal pancreatic carcinoma. Evidence suggests that MUC1 can be used as a tumor marker and is a potential target for immunotherapy of pancreatic cancer. However, vaccination with MUC1 peptides fails to stimulate the immune response against cancer cells because immunity toward tumor-associated antigens (TAA), including MUC1, in cancer patients is relatively weak, and the presentation of these TAAs to the immune system is poor due to their low immunogenicity. We investigated whether vaccination with immunogenetically enhanced MUC1 (by expressing alpha-gal epitopes; Galalpha1-3Galbeta1-4GlcNAc-R) can elicit effective antibody production for MUC1 itself as well as certain TAAs derived from pancreatic cancer cells and induced tumor-specific T-cell responses. We also used alpha1,3galactosyltransferase (alpha1,3GT) knockout mice that were preimmunized with pig kidney and transplanted with B16F10 melanoma cells transfected with MUC1 expression vector. Vaccination of these mice with alpha-gal MUC1 resulted in marked inhibition of tumor growth and significant improvement of overall survival time compared with mice vaccinated with MUC1 alone (P = 0.003). Furthermore, vaccination with pancreatic cancer cells expressing alpha-gal epitopes induced immune responses against not only differentiated cancer cells but also cancer stem cells. The results suggested that vaccination using cells engineered to express alpha-gal epitopes is a novel strategy for treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Takashi Deguchi
- Department of Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
In Situ Conversion of Melanoma Lesions into Autologous Vaccine by Intratumoral Injections of α-gal Glycolipids. Cancers (Basel) 2010; 2:773-93. [PMID: 23087817 PMCID: PMC3475649 DOI: 10.3390/cancers2020773] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Autologous melanoma associated antigens (MAA) on murine melanoma cells can elicit a protective anti-tumor immune response following a variety of vaccine strategies. Most require effective uptake by antigen presenting cells (APC). APC transport and process internalized MAA for activation of anti-tumor T cells. One potential problem with clinical melanoma vaccines against autologous tumors may be that often tumor cells do not express surface markers that label them for uptake by APC. Effective uptake of melanoma cells by APC might be achieved by exploiting the natural anti-Gal antibody which constitutes ~1% of immunoglobulins in humans. This approach has been developed in a syngeneic mouse model using mice capable of producing anti-Gal. Anti-Gal binds specifically to α-gal epitopes (Galα1-3Galβ1-4GlcNAc-R). Injection of glycolipids carrying α-gal epitopes (α-gal glycolipids) into melanoma lesions results in glycolipid insertion into melanoma cell membranes, expression of α-gal epitopes on the tumor cells and binding of anti-Gal to these epitopes. Interaction between the Fc portions of bound anti-Gal and Fcγ receptors on APC induces effective uptake of tumor cells by APC. The resulting anti-MAA immune response can be potent enough to destroy distant micrometastases. A clinical trial is now open testing effects of intratumoral α-gal glycolipid injections in melanoma patients.
Collapse
|