1
|
Honda N, Watanabe Y, Tokuoka Y, Hanajima R. Roles of microglia/macrophage and antibody in cell sheet transplantation in the central nervous system. Stem Cell Res Ther 2022; 13:470. [PMID: 36089602 PMCID: PMC9465875 DOI: 10.1186/s13287-022-03168-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 08/28/2022] [Indexed: 11/16/2022] Open
Abstract
Background We previously established a human mesenchymal stem cell (MSC) line that was modified to express trophic factors. Transplanting a cell sheet produced from this line in an amyotrophic lateral sclerosis mouse model showed a beneficial trend for mouse life spans. However, the sheet survived for less than 14 days, and numerous microglia and macrophages were observed within and adjacent to the sheet. Here, we examined the roles of microglia and macrophages as well as acquired antibodies in cell sheet transplantation. Methods We observed the effects of several MSC lines on macrophages in vitro, that is, phenotype polarization (M1 or M2) and migration. We then investigated how phenotypic polarization affected MSC survival using antibody-dependent cellular cytotoxicity (ADCC) and phagocytosis (ADCP). We also confirmed the role of complement on cytotoxicity. Lastly, we selectively eliminated microglia and macrophages in vivo to determine whether these cells were cytoprotective to the donor sheet. Results In vitro co-culture with MSCs induced M2 polarization in macrophages and facilitated their migration toward MSCs in vitro. There was no difference between M1 and M2 phenotypes on ADCC and ADCP. Cytotoxicity was observed even in the absence of complement. Eliminating microglia/macrophage populations in vivo resulted in increased survival of donor cells after transplantation. Conclusions Acquired antibodies played a role in ADCC and ADCP. MSCs induced M2 polarization in macrophages and facilitated their migration toward MSCs in vitro. Despite these favorable characteristics of microglia and macrophages, deletion of these cells was advantageous for the survival of donor cells in vivo. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-03168-5.
Collapse
|
2
|
Sewnath CA, Behrens LM, van Egmond M. Targeting myeloid cells with bispecific antibodies as novel immunotherapies of cancer. Expert Opin Biol Ther 2022; 22:983-995. [PMID: 35854649 DOI: 10.1080/14712598.2022.2098675] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Most bispecific antibody (BsAb) therapies focus on stimulating the adaptive immune system, in particular T cells, to promote tumor cell killing. Another method to promote tumor eradication is through the engagement of myeloid cells, including macrophages and neutrophils, which are abundantly present and possess intrinsic cytotoxic mechanisms for tumor cell killing, making them interesting effector cells to recruit for BsAb therapy. AREAS COVERED In this review, we describe the evolving knowledge of the role of macrophages and neutrophils in cancer in scientific literature. Moreover, we address the BsAbs that have been developed over the years to recruit these cell types as effector cells in immunotherapy of cancer. This includes the discussion of BsAbs that target Fc receptors (i.e. FcγR and FcαRI) to induce antibody-dependent cellular phagocytosis (ADCP) by macrophages or trogoptosis via neutrophils, as well as BsAbs that interfere with checkpoint inhibition, including the SIRPα-CD47 pathway. EXPERT OPINION Elucidating the complexity of macrophage and neutrophil heterogeneity in cancer may help to specifically enlist the cytotoxic ability of these cells through targeting Fc receptors and checkpoint pathways, which may further enhance anti-cancer immunity.
Collapse
Affiliation(s)
- Celine An Sewnath
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam De Boelelaan, Amsterdam, The Netherlands.,Cancer Biology and Immunology Program, Cancer Centre Amsterdam, Amsterdam, The Netherlands.,Cancer Immunology Program, Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
| | - Leonie M Behrens
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam De Boelelaan, Amsterdam, The Netherlands.,Cancer Biology and Immunology Program, Cancer Centre Amsterdam, Amsterdam, The Netherlands.,Cancer Immunology Program, Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
| | - Marjolein van Egmond
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam De Boelelaan, Amsterdam, The Netherlands.,Cancer Biology and Immunology Program, Cancer Centre Amsterdam, Amsterdam, The Netherlands.,Cancer Immunology Program, Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands.,Department of Surgery, Amsterdam UMC, Vrije Universiteit Amsterdam De Boelelaan, Amsterdam, The Netherlands
| |
Collapse
|
3
|
Yamaguchi S, Takagi R, Hosogane T, Ohashi Y, Sakai Y, Sakakihara S, Iino R, Tabata KV, Noji H, Okamoto A. Single Cell Array Enclosed with a Photodegradable Hydrogel in Microwells for Image-Based Cell Classification and Selective Photorelease of Cells. ACS APPLIED BIO MATERIALS 2020; 3:5887-5895. [PMID: 35021817 DOI: 10.1021/acsabm.0c00583] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Single cell arrays provide an accurate classification of analyte cells through an image-based analysis of cellular phenotypes. Light-guided cell retrieval from a single cell array is a promising approach for the rapid and simple sorting of difficult to distinguish cells. In this study, we developed a single cell array enclosed with a photodegradable hydrogel in microwells to enable both comprehensive image-based single cell analysis and light-guided cell retrieval. In this system, individual cells became trapped in the microwells together with the photodegradable hydrogel at a high cell density on a chip regardless of cell type, adhesiveness, and motility. Fluorescence-stained model cells and vaccinated dendritic cells were identified by microscopic imaging and then selectively released through the light-induced degradation of the cell-embedding hydrogels. The target cells were selectively retrieved with a purity of >95% from the cell mixture through rapid photorelease, and the retrieved cells were confirmed to grow normally. Our results provide proof-of-principle that the photoresponsive microwell array serves as a versatile tool for image-based cell sorting in cellular researches and the manufacturing processes of high-performance cells.
Collapse
Affiliation(s)
- Satoshi Yamaguchi
- Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8904, Japan.,PRESTO, Japan Science and Technology Agency, 4-1-8 Hon-cho, Kawaguchi, Saitama 351-0198, Japan
| | - Risa Takagi
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Tsuyoshi Hosogane
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Yuki Ohashi
- Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8904, Japan
| | - Yoko Sakai
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Shouichi Sakakihara
- The Institute of Scientific and Industrial Research, Osaka University, 8-1 Mihogaoka, Ibaraki, Osaka 567-0047, Japan
| | - Ryota Iino
- Institute for Molecular Science, 5-1 Higashiyama, Myodaiji, Okazaki 444-8787, Japan
| | - Kazuhito V Tabata
- Department of Applied Chemistry, Graduate School of Engineering, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Hiroyuki Noji
- Department of Applied Chemistry, Graduate School of Engineering, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Akimitsu Okamoto
- Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8904, Japan.,Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| |
Collapse
|
4
|
Yuan X, Yang M, Chen X, Zhang X, Sukhadia S, Musolino N, Bao H, Chen T, Xu C, Wang Q, Santoro S, Ricklin D, Hu J, Lin R, Yang W, Li Z, Qin W, Zhao A, Scholler N, Coukos G. Correction to: Characterization of the first fully human anti-TEM1 scFv in models of solid tumor imaging and immunotoxin-based therapy. Cancer Immunol Immunother 2018; 67:329-339. [PMID: 29313073 PMCID: PMC11028179 DOI: 10.1007/s00262-017-2101-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Tumor endothelial marker 1 (TEM1) has been identified as a novel surface marker upregulated on the blood vessels and stroma in many solid tumors. We previously isolated a novel single-chain variable fragment (scFv) 78 against TEM1 from a yeast display scFv library. Here we evaluated the potential applications of scFv78 as a tool for tumor molecular imaging, immunotoxin-based therapy and nanotherapy. Epitope mapping, three-dimensional (3D) structure docking and affinity measurements indicated that scFv78 could bind to both human and murine TEM1, with equivalent affinity, at a well-conserved conformational epitope. The rapid internalization of scFv78 and scFv78-labeled nanoparticles was triggered after specific TEM1 binding. The scFv78-saporin immunoconjugate also exerted dose-dependent cytotoxicity with high specificity to TEM1-positive cells in vitro. Finally, specific and sensitive tumor localization of scFv78 was confirmed with optical imaging in a mouse tumor model that has highly endogenous mTEM1 expression in the vasculature. Our data indicate that scFv78, the first fully human anti-TEM1 recombinant antibody, recognizes both human and mouse TEM1 and has unique and favorable features that are advantageous for the development of imaging probes or antibody-toxin conjugates for a large spectrum of human TEM1-positive solid tumors.
Collapse
Affiliation(s)
- Xiaopeng Yuan
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangdong, China
| | - Mingjuan Yang
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Xiang Chen
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Xuhua Zhang
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Shrey Sukhadia
- Department of Chemistry and Biochemistry, University of the Sciences in Philadelphia, Philadelphia, PA, USA
| | - Najia Musolino
- Ludwig Institute for Cancer Research at CHUV, Rue du Bugnon 46-BH09-701, 1011, Lausanne, Switzerland
| | - Huijing Bao
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Tingtao Chen
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Chen Xu
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Qirui Wang
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Stephen Santoro
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel Ricklin
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jia Hu
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Ruihe Lin
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Wei Yang
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhijun Li
- Department of Chemistry and Biochemistry, University of the Sciences in Philadelphia, Philadelphia, PA, USA
| | - Weijun Qin
- Department of Urology, Xijing Hospital, Fourth Military University, Xi'an, 710032, China.
| | - Aizhi Zhao
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Nathalie Scholler
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- SRI International, Menlo Park, CA, USA
| | - George Coukos
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA.
- Ludwig Institute for Cancer Research at CHUV, Rue du Bugnon 46-BH09-701, 1011, Lausanne, Switzerland.
| |
Collapse
|
5
|
Ma B, Ujjani C. The clinical development of obinutuzumab for the treatment of follicular lymphoma. Cancer Manag Res 2017; 9:103-113. [PMID: 28435325 PMCID: PMC5391868 DOI: 10.2147/cmar.s114526] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Impressive progress has been made in recent decades for advanced-stage follicular lymphoma with the availability of anti-CD20 monoclonal antibodies, initially rituximab and more recently obinutuzumab. Obinutuzumab is a unique, third-generation, fully humanized glycoengineered IgG1 type II anti-CD20 monoclonal antibody. It has been shown to have increased antitumor activity compared to rituximab in preclinical studies, including whole-blood B-cell depletion assays, xenograft models, and primate models. This has spurred on the development of obinutuzumab through Phase I/II trials as monotherapy and in combination with chemotherapeutic agents and other targeted therapies. Its efficacy compared to rituximab and in rituximab-refractory disease has led to its continued development and eventual approval for the treatment of follicular lymphoma. Here in this review, we highlight the design and development of obinutuzumab in the treatment of advanced stage grade 1-3A follicular lymphoma and its future directions.
Collapse
Affiliation(s)
| | - Chaitra Ujjani
- Lombardi Comprehensive Cancer Center, MedStar Georgetown University Hospital, Washington, DC, USA
| |
Collapse
|
6
|
Yuan X, Yang M, Chen X, Zhang X, Sukhadia S, Musolino N, Bao H, Chen T, Xu C, Wang Q, Santoro S, Ricklin D, Hu J, Lin R, Yang W, Li Z, Qin W, Zhao A. Characterization of the first fully human anti-TEM1 scFv in models of solid tumor imaging and immunotoxin-based therapy. Cancer Immunol Immunother 2017; 66:367-378. [PMID: 27933426 PMCID: PMC11029759 DOI: 10.1007/s00262-016-1937-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 11/19/2016] [Indexed: 10/20/2022]
Abstract
Tumor endothelial marker 1 (TEM1) has been identified as a novel surface marker upregulated on the blood vessels and stroma in many solid tumors. We previously isolated a novel single-chain variable fragment (scFv) 78 against TEM1 from a yeast display scFv library. Here, we evaluated the potential applications of scFv78 as a tool for tumor molecular imaging, immunotoxin-based therapy and nanotherapy. Epitope mapping, three-dimensional structure docking and affinity measurements indicated that scFv78 could bind to both human and murine TEM1, with equivalent affinity, at a well-conserved conformational epitope. The rapid internalization of scFv78 and scFv78-labeled nanoparticles was triggered after specific TEM1 binding. The scFv78-saporin immunoconjugate also exerted dose-dependent cytotoxicity with high specificity to TEM1-positive cells in vitro. Finally, specific and sensitive tumor localization of scFv78 was confirmed with optical imaging in a tumor mouse model that has highly endogenous mTEM1 expression in the vasculature. Our data indicated that scFv78, the first fully human anti-TEM1 recombinant antibody, recognizes both human and mouse TEM1 and has unique and favorable features that are advantageous for the development of imaging probes or antibody-toxin conjugates for a large spectrum of human TEM1-positive solid tumors.
Collapse
Affiliation(s)
- Xiaopeng Yuan
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangdong, China
| | - Mingjuan Yang
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Xiang Chen
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Xuhua Zhang
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Shrey Sukhadia
- Department of Chemistry and Biochemistry, University of the Sciences in Philadelphia, Philadelphia, PA, USA
| | | | - Huijing Bao
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Tingtao Chen
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Chen Xu
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Qirui Wang
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Stephen Santoro
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel Ricklin
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jia Hu
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Ruihe Lin
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Wei Yang
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhijun Li
- Department of Chemistry and Biochemistry, University of the Sciences in Philadelphia, Philadelphia, PA, USA
| | - Weijun Qin
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| | - Aizhi Zhao
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA.
- University City Science Center, Room 544, 3624 Market Street, Philadelphia, PA, 19104, USA.
| |
Collapse
|
7
|
Shinmi D, Nakano R, Mitamura K, Suzuki-Imaizumi M, Iwano J, Isoda Y, Enokizono J, Shiraishi Y, Arakawa E, Tomizuka K, Masuda K. Novel anticarcinoembryonic antigen antibody-drug conjugate has antitumor activity in the existence of soluble antigen. Cancer Med 2017; 6:798-808. [PMID: 28211613 PMCID: PMC5387159 DOI: 10.1002/cam4.1003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 11/16/2016] [Accepted: 12/09/2016] [Indexed: 01/21/2023] Open
Abstract
Carcinoembryonic antigen (CEA) is a classic tumor‐specific antigen that is overexpressed in several cancers, including gastric cancer. Although some anti‐CEA antibodies have been tested, to the best of our knowledge, there are currently no clinically approved anti‐CEA antibody therapies. Because of this, we have created the novel anti‐CEA antibody, 15‐1‐32, which exhibits stronger binding to membrane‐bound CEA on cancer cells than existing anti‐CEA antibodies. 15‐1‐32 also shows poor affinity for soluble CEA; thus, the binding activity of 15‐1‐32 to membrane‐bound CEA is not influenced by soluble CEA. In addition, we constructed a 15‐1‐32‐monomethyl auristatin E conjugate (15‐1‐32‐vcMMAE) to improve the therapeutic efficacy of 15‐1‐32. 15‐1‐32‐vcMMAE showed enhanced antitumor activity against gastric cancer cell lines. Unlike with existing anti‐CEA antibody therapies, antitumor activity of 15‐1‐32‐vcMMAE was retained in the presence of high concentrations of soluble CEA.
Collapse
Affiliation(s)
- Daisuke Shinmi
- Research Core Function Laboratories, Research Functions Unit, R&D Division, Kyowa Hakko Kirin Co., Ltd., Tokyo, Japan
| | - Ryosuke Nakano
- Research Core Function Laboratories, Research Functions Unit, R&D Division, Kyowa Hakko Kirin Co., Ltd., Tokyo, Japan
| | - Keisuke Mitamura
- Oncology Research Laboratories, Oncology R&D Unit, R&D Division, Kyowa Hakko Kirin Co., Ltd., Tokyo, Japan
| | | | - Junko Iwano
- Research Core Function Laboratories, Research Functions Unit, R&D Division, Kyowa Hakko Kirin Co., Ltd., Tokyo, Japan
| | - Yuya Isoda
- Innovative Technology Laboratories, Research Functions Unit, R&D Division, Kyowa Hakko Kirin Co., Ltd., Tokyo, Japan
| | - Junichi Enokizono
- Research Core Function Laboratories, Research Functions Unit, R&D Division, Kyowa Hakko Kirin Co., Ltd., Tokyo, Japan
| | - Yasuhisa Shiraishi
- R&D Planning Department, R&D Division, Kyowa Hakko Kirin Co., Ltd., Tokyo, Japan
| | - Emi Arakawa
- Fuji Research Park, R&D Division, Kyowa Hakko Kirin Co., Ltd., Tokyo, Japan
| | - Kazuma Tomizuka
- Innovative Technology Laboratories, Research Functions Unit, R&D Division, Kyowa Hakko Kirin Co., Ltd., Tokyo, Japan
| | - Kazuhiro Masuda
- Innovative Technology Laboratories, Research Functions Unit, R&D Division, Kyowa Hakko Kirin Co., Ltd., Tokyo, Japan
| |
Collapse
|
8
|
Klein C, Waldhauer I, Nicolini VG, Freimoser-Grundschober A, Nayak T, Vugts DJ, Dunn C, Bolijn M, Benz J, Stihle M, Lang S, Roemmele M, Hofer T, van Puijenbroek E, Wittig D, Moser S, Ast O, Brünker P, Gorr IH, Neumann S, de Vera Mudry MC, Hinton H, Crameri F, Saro J, Evers S, Gerdes C, Bacac M, van Dongen G, Moessner E, Umaña P. Cergutuzumab amunaleukin (CEA-IL2v), a CEA-targeted IL-2 variant-based immunocytokine for combination cancer immunotherapy: Overcoming limitations of aldesleukin and conventional IL-2-based immunocytokines. Oncoimmunology 2017; 6:e1277306. [PMID: 28405498 PMCID: PMC5384349 DOI: 10.1080/2162402x.2016.1277306] [Citation(s) in RCA: 208] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 12/16/2016] [Accepted: 12/21/2016] [Indexed: 12/11/2022] Open
Abstract
We developed cergutuzumab amunaleukin (CEA-IL2v, RG7813), a novel monomeric CEA-targeted immunocytokine, that comprises a single IL-2 variant (IL2v) moiety with abolished CD25 binding, fused to the C-terminus of a high affinity, bivalent carcinoembryonic antigen (CEA)-specific antibody devoid of Fc-mediated effector functions. Its molecular design aims to (i) avoid preferential activation of regulatory T-cells vs. immune effector cells by removing CD25 binding; (ii) increase the therapeutic index of IL-2 therapy by (a) preferential retention at the tumor by having a lower dissociation rate from CEA-expressing cancer cells vs. IL-2R-expressing cells, (b) avoiding any FcγR-binding and Fc effector functions and (c) reduced binding to endothelial cells expressing CD25; and (iii) improve the pharmacokinetics, and thus convenience of administration, of IL-2. The crystal structure of the IL2v-IL-2Rβγ complex was determined and CEA-IL2v activity was assessed using human immune effector cells. Tumor targeting was investigated in tumor-bearing mice using 89Zr-labeled CEA-IL2v. Efficacy studies were performed in (a) syngeneic mouse models as monotherapy and combined with anti-PD-L1, and in (b) xenograft mouse models in combination with ADCC-mediating antibodies. CEA-IL2v binds to CEA with pM avidity but not to CD25, and consequently did not preferentially activate Tregs. In vivo, CEA-IL2v demonstrated superior pharmacokinetics and tumor targeting compared with a wild-type IL-2-based CEA immunocytokine (CEA-IL2wt). CEA-IL2v strongly expanded NK and CD8+ T cells, skewing the CD8+:CD4+ ratio toward CD8+ T cells both in the periphery and in the tumor, and mediated single agent efficacy in syngeneic MC38-CEA and PancO2-CEA models. Combination with trastuzumab, cetuximab and imgatuzumab, all of human IgG1 isotype, resulted in superior efficacy compared with the monotherapies alone. Combined with anti-PD-L1, CEA-IL2v mediated superior efficacy over the respective monotherapies, and over the combination with an untargeted control immunocytokine. These preclinical data support the ongoing clinical investigation of the cergutuzumab amunaleukin immunocytokine with abolished CD25 binding for the treatment of CEA-positive solid tumors in combination with PD-L1 checkpoint blockade and ADCC competent antibodies.
Collapse
Affiliation(s)
- Christian Klein
- Roche Pharma Research & Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland
| | - Inja Waldhauer
- Roche Pharma Research & Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland
| | - Valeria G. Nicolini
- Roche Pharma Research & Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland
| | | | - Tapan Nayak
- Roche Pharma Research & Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Danielle J. Vugts
- Roche Pharma Research & Early Development, Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, the Netherlands
| | - Claire Dunn
- Roche Pharma Research & Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland
| | - Marije Bolijn
- Roche Pharma Research & Early Development, Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, the Netherlands
| | - Jörg Benz
- Roche Pharma Research & Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Martine Stihle
- Roche Pharma Research & Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Sabine Lang
- Roche Pharma Research & Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland
| | - Michaele Roemmele
- Roche Pharma Research & Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland
| | - Thomas Hofer
- Roche Pharma Research & Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland
| | - Erwin van Puijenbroek
- Roche Pharma Research & Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland
| | - David Wittig
- Roche Pharma Research & Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland
| | - Samuel Moser
- Roche Pharma Research & Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland
| | - Oliver Ast
- Roche Pharma Research & Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland
| | - Peter Brünker
- Roche Pharma Research & Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland
| | - Ingo H. Gorr
- Roche Pharma Research & Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Sebastian Neumann
- Roche Pharma Research & Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | | | - Heather Hinton
- Roche Pharma Research & Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland
| | - Flavio Crameri
- Roche Pharma Research & Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Jose Saro
- Roche Pharma Research & Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland
| | - Stefan Evers
- Roche Pharma Research & Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland
| | - Christian Gerdes
- Roche Pharma Research & Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland
| | - Marina Bacac
- Roche Pharma Research & Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland
| | - Guus van Dongen
- Roche Pharma Research & Early Development, Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, the Netherlands
| | - Ekkehard Moessner
- Roche Pharma Research & Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland
| | - Pablo Umaña
- Roche Pharma Research & Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland
| |
Collapse
|
9
|
McEnaney PJ, Fitzgerald KJ, Zhang AX, Douglass EF, Shan W, Balog A, Kolesnikova MD, Spiegel DA. Chemically synthesized molecules with the targeting and effector functions of antibodies. J Am Chem Soc 2014; 136:18034-43. [PMID: 25514603 PMCID: PMC4291750 DOI: 10.1021/ja509513c] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Indexed: 12/21/2022]
Abstract
This article reports the design, synthesis, and evaluation of a novel class of molecules of intermediate size (approximately 7000 Da), which possess both the targeting and effector functions of antibodies. These compounds—called synthetic antibody mimics targeting prostate cancer (SyAM-Ps)—bind simultaneously to prostate-specific membrane antigen and Fc gamma receptor I, thus eliciting highly selective cancer cell phagocytosis. SyAMs have the potential to combine the advantages of both small-molecule and biologic therapies, and may address many drawbacks associated with available treatments for cancer and other diseases.
Collapse
Affiliation(s)
- Patrick J McEnaney
- Department of Chemistry, Yale University , 225 Prospect Street, New Haven, Connecticut 06511, United States
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Singer J, Jensen‐Jarolim E. IgE-based immunotherapy of cancer: challenges and chances. Allergy 2014; 69:137-49. [PMID: 24117861 PMCID: PMC4022995 DOI: 10.1111/all.12276] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/27/2013] [Indexed: 12/16/2022]
Abstract
Passive immunotherapy with monoclonal antibodies is an indispensable cornerstone of clinical oncology. Notably, all FDA-approved antibodies comprise the IgG class, although numerous research articles proposed monoclonal antibodies of the IgM, IgG, IgA and IgE classes directed specifically against tumor-associated antigens. In particular, for the IgE isotype class, several recent studies could demonstrate high tumoricidic efficacy. Therefore, this review specifically highlights the latest developments toward IgE-based immunotherapy of cancer. Possible mechanisms and safety aspects of IgE-mediated tumor cell death are discussed with special focus on the attracted immune cells. An outlook is given on how especially comparative oncology could contribute to further developments. Humans and dogs have a highly comparable IgE biology, suggesting that translational AllergoOncology studies in patients with canine cancer could have predictive value for the potential of IgE-based anticancer immunotherapy in human clinical oncology.
Collapse
Affiliation(s)
- J. Singer
- Comparative Immunology and Oncology Institute of Pathophysiology and Allergy Research Medical University of Vienna Vienna Austria
| | - E. Jensen‐Jarolim
- Comparative Immunology and Oncology Institute of Pathophysiology and Allergy Research Medical University of Vienna Vienna Austria
- Comparative Medicine Messerli Research Institute of the University of Veterinary Medicine Vienna Medical University Vienna and University Vienna Vienna Austria
| |
Collapse
|
11
|
Herter S, Birk MC, Klein C, Gerdes C, Umana P, Bacac M. Glycoengineering of therapeutic antibodies enhances monocyte/macrophage-mediated phagocytosis and cytotoxicity. THE JOURNAL OF IMMUNOLOGY 2014; 192:2252-60. [PMID: 24489098 DOI: 10.4049/jimmunol.1301249] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Therapeutic Abs possess several clinically relevant mechanisms of action including perturbation of tumor cell signaling, activation of complement-dependent cytotoxicity, Ab-dependent cellular cytotoxicity (ADCC), Ab-dependent cellular phagocytosis (ADCP), and induction of adaptive immunity. In view of the important role of phagocytic lineage cells in the mechanism of action of therapeutic Abs, we analyzed FcγR receptor-dependent effector functions of monocytes and macrophages triggered by glycoengineered (GE) Abs (having enhanced FcγRIIIa [CD16a] binding affinity) versus their wild-type (WT) counterparts under different experimental conditions. We first defined the precise FcγR repertoire on classical and nonclassical intermediate monocytes--M1 and M2c macrophage populations. We further show that WT and GE Abs display comparable binding and induce similar effector functions (ADCC and ADCP) in the absence of nonspecific, endogenous IgGs. However, in the presence of these IgGs (i.e., in a situation that more closely mimics physiologic conditions), GE Abs display significantly superior binding and promote stronger monocyte and macrophage activity. These data show that in addition to enhancing CD16a-dependent NK cell cytotoxicity, glycoengineering also enhances monocyte and macrophage phagocytic and cytotoxic activities through enhanced binding to CD16a under conditions that more closely resemble the physiologic setting.
Collapse
Affiliation(s)
- Sylvia Herter
- Discovery Oncology, Pharma Research and Early Development (pRED), Roche Glycart AG, 8952 Schlieren, Zurich, Switzerland
| | | | | | | | | | | |
Collapse
|
12
|
Braster R, O’Toole T, van Egmond M. Myeloid cells as effector cells for monoclonal antibody therapy of cancer. Methods 2014; 65:28-37. [DOI: 10.1016/j.ymeth.2013.06.020] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Revised: 06/06/2013] [Accepted: 06/18/2013] [Indexed: 02/07/2023] Open
|
13
|
Bakema JE, van Egmond M. Fc receptor-dependent mechanisms of monoclonal antibody therapy of cancer. Curr Top Microbiol Immunol 2014; 382:373-92. [PMID: 25116109 DOI: 10.1007/978-3-319-07911-0_17] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Targeted therapies like treatment with monoclonal antibodies (mAbs) have entered the arsenal of modern anticancer drugs. mAbs combine specificity with multiple effector functions that can lead to reduction of tumour burden. Direct mechanisms of action, including induction of apoptosis or growth inhibition, depend on the biology of the target antigen. Fc tails of mAbs have furthermore the potential to initiate complement-dependent lysis as well as immune effector cell-mediated tumour cell killing via binding to Fc receptors. Natural killer cells can induce apoptosis via antibody-dependent cellular cytotoxicity (ADCC), whereas macrophages are able to phagocytose mAb-opsonized tumour cells (antibody-dependent cellular phagocytosis; ADCP). Finally, neutrophils can induce non-apoptotic tumour cell death, especially in the presence of immunoglobulin A (IgA) antitumour mAbs. In spite of promising clinical successes in some malignancies, improvement of mAb immunotherapy is required to achieve overall complete remission in cancer patients. New strategies to enhance Fc receptor-mediated mechanisms of action or to overcome the immunosuppressive microenvironment of the tumour in mAb therapy of cancer are therefore currently being explored and will be addressed in this chapter.
Collapse
Affiliation(s)
- Jantine E Bakema
- Tumor Biology Section, Department of Otolaryngology/Head-Neck Surgery, VU University Medical Center, De Boelelaan 1117, 1007 MB, Amsterdam, The Netherlands
| | | |
Collapse
|
14
|
Rafiq S, Siadak A, Butchar JP, Cheney C, Lozanski G, Jacob NK, Lapalombella R, McGourty J, Moledor M, Lowe R, Setter B, Jones J, Flynn JM, Andritsos L, Devine S, Mo X, Jarjoura D, Tridandapani S, Algate P, Byrd JC, Muthusamy N. Glycovariant anti-CD37 monospecific protein therapeutic exhibits enhanced effector cell-mediated cytotoxicity against chronic and acute B cell malignancies. MAbs 2013; 5:723-35. [PMID: 23883821 DOI: 10.4161/mabs.25282] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
TRU-016 is a SMIP(TM) (monospecific protein therapeutic) molecule against the tetraspanin transmembrane family protein CD37 that is currently in Phase 2 trials in Chronic Lymphocytic Leukemia (CLL) and Non-Hodgkin Lymphoma (NHL). In an attempt to enhance the ADCC function of SMIP-016, the chimeric version of TRU-016, SMIP-016(GV) was engineered with a modification in a glycosylation site in the Fc domain. The wild-type and glycovariant SMIP proteins mediate comparable Type I antibody-like direct cytotoxicity in the presence of anti-human Fc crosslinker and show a similar tyrosine phosphorylation pattern post-treatment. However, NK cells stimulated with the SMIP-016(GV) exhibit enhanced activation and release 3-fold more interferon-γ compared with SMIP-016. SMIP-016(GV) shows enhanced ADCC function against cells expressing CD37 with NK cell effectors derived from both normal and CLL-affected individuals. Enhanced ADCC is observed against CLL cells and is sustained at concentrations of SMIP-016(GV) as low at 5E(-6) µg/mL on cells expressing minimal CD37 antigen. In support of the biological relevance of this, SMIP-016(GV) mediates effective ADCC against primary acute lymphoblastic leukemia (ALL) cells with low surface expression of CD37. Collectively, these data suggest potential use of the novel therapeutic agent SMIP-016(GV) with enhanced effector function for B cell malignancies, including CLL and ALL therapy.
Collapse
Affiliation(s)
- Sarwish Rafiq
- Integrated Biomedical Science Graduate Program; The Ohio State University; Columbus, OH USA; Division of Hematology, Department of Internal Medicine; The Ohio State University; Columbus, OH USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Tomita U, Yamaguchi S, Maeda Y, Chujo K, Minamihata K, Nagamune T. Protein cell-surface display through in situ enzymatic modification of proteins with a poly(Ethylene glycol)-lipid. Biotechnol Bioeng 2013; 110:2785-9. [PMID: 23592269 DOI: 10.1002/bit.24933] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 03/27/2013] [Accepted: 04/01/2013] [Indexed: 01/24/2023]
Abstract
Cell-surface display of functional proteins is a powerful and useful tool for regulating and reinforcing cellular functions. Direct incorporation of site-specifically lipidated proteins from the extracellular medium is more rapid, easily controllable and reliable in displaying active proteins than expression through gene transfer. However, undesirable amphiphilic reagents such as organic co-solvents and detergents were required for suppressing aggregation of ordinary lipidated proteins in solution. We report here sortase A-catalyzed modification of proteins with a poly(ethylene glycol)(PEG)-lipid in situ on the surface of living cells. Proteins fused with a recognition tag were site-specifically ligated with the PEG-lipid which was preliminary incorporated into cell membranes. Accordingly, target proteins were successfully displayed on living cells without aggregation under an amphiphilic reagent-free condition. Furthermore, to demonstrate the availability of the present method, Fc domains of immunoglobulin G were displayed on cancer cells, and the phagocytosis of cancer cells with dendritic cells were enhanced through the Fc-Fc receptor interaction. Thus, the present facile chemoenzymatic method for protein display can be utilized for modulating cell-cell interactions in cell and tissue engineering fields.
Collapse
Affiliation(s)
- Urara Tomita
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo 113-8656, Japan
| | | | | | | | | | | |
Collapse
|
16
|
Merli M, Ferrario A, Basilico C, Maffioli M, Caramazza D, Appio L, Arcaini L, Passamonti F. Novel agents in indolent lymphomas. Ther Adv Hematol 2013; 4:133-48. [PMID: 23610620 PMCID: PMC3629754 DOI: 10.1177/2040620712466865] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Indolent non-Hodgkin's lymphomas (iNHLs) include follicular lymphomas (FL), marginal-zone lymphoma, lymphoplasmacytic lymphoma/Waldenström macroglobulinemia and small lymphocytic lymphoma. First-line standard therapy in advanced, symptomatic iNHL consists of rituximab-based immunochemotherapy. The recent rediscovery of the 'old' chemotherapeutic agent bendamustine, an alkylating agent with a peculiar mechanism of action, has added a new effective and well-tolerated option to the therapeutic armamentarium in iNHL, increasing response rates and duration. However, patients invariably relapse and subsequent active and well-tolerated agents are needed. In recent years a large number of new targeted agents have been tested in preclinical and clinical experimentation in FL and indolent nonfollicular lymphoma (iNFL), including the new monoclonal antibodies binding CD20 or other surface antigens, immunoconjugates and bispecific antibodies. Moreover novel agents directed against intracellular processes such as proteasome inhibitors, mTOR inhibitors and agents that target the tumour microenvironment, notably the immunomodulatory agent lenalidomide, are under active clinical investigation. The development of these new drugs may change in the near future the approach to iNHL patients, leading to better tolerated and effective therapy regimens.
Collapse
Affiliation(s)
- Michele Merli
- Division of Hematology, Department of Internal Medicine, Ospedale di Circolo and Fondazione Macchi, University of Insubria, Varese, Italy
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Direct and immune mediated antibody targeting of ERBB receptors in a colorectal cancer cell-line panel. Proc Natl Acad Sci U S A 2012; 109:21046-51. [PMID: 23213241 DOI: 10.1073/pnas.1218750110] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
A significant proportion of colorectal cancer (CRC) patients are resistant to anti-ERBB1 [avian erythroblastic leukemia viral (v-erb-b) oncogene homolog, receptor for EGF] monoclonal antibodies (Mabs). We evaluated both immune and nonimmune effects of cetuximab (anti-ERBB1 Mab), trastuzumab (anti-ERBB2 Mab), pertuzumab (anti-ERBB2 Mab), and lapatinib (dual ERBB1 and ERBB2 tyrosine kinase inhibitor) in a large well-characterized panel of 64 CRC cell lines to find response predictive tumor characteristics. There was a significant correlation between the direct effects of cetuximab and lapatinib. Both agents were associated (P = 0.0004) with "triple' wild-type status in KRAS, BRAF, and PIK3CA exon 20. Most cell lines were resistant to the direct effects of anti-ERBB2 Mabs, suggesting that the effects of lapatinib might mainly be through ERBB1. Microarray mRNA expression profiles of sensitive and resistant cell lines showed that although ERBB1 receptor or ligand levels did not associate with cetuximab sensitivity, high levels of ERBB2 (P = 0.036) and amphiregulin (P = 0.026) predicted sensitivity to lapatinib. However, higher ERBB1 expression predicted susceptibility to cetuximab-induced antibody-dependent cellular cytotoxicity and occurred independently of KRAS/BRAF/PIK3CA mutations (P = 0.69). Lapatinib may be an effective alternative therapy to cetuximab in triple wild-type tumors. Microarray analysis provides suggestive biomarkers for resistance. ERBB1 levels, independent of mutation status, predict immune killing. Therefore, anti-ERBB1 antibodies may be considered in CRC tumors with higher ERBB1 expression and favorable FcγR polymorphisms.
Collapse
|
18
|
Cérutti M, Golay J. Lepidopteran cells, an alternative for the production of recombinant antibodies? MAbs 2012; 4:294-309. [PMID: 22531440 DOI: 10.4161/mabs.19942] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Monoclonal antibodies are used with great success in many different therapeutic domains. In order to satisfy the growing demand and to lower the production cost of these molecules, many alternative systems have been explored. Among them, the baculovirus/insect cells system is a good candidate. This system is very safe, given that the baculoviruses have a highly restricted host range and they are not pathogenic to vertebrates or plants. But the major asset is the speed with which it is possible to obtain very stable recombinant viruses capable of producing fully active proteins whose glycosylation pattern can be modulated to make it similar to the human one. These features could ultimately make the difference by enabling the production of antibodies with very low costs. However, efforts are still needed, in particular to increase production rates and thus make this system commercially viable for the production of these therapeutic agents.
Collapse
Affiliation(s)
- Martine Cérutti
- CNRS UPS3044 Baculovirus et Thérapie, CNRS GDR3260, ACCITH Anticorps et Ciblage Thérapeutique and LabEx MabImprove, Saint Christol Lèz Alès, France.
| | | |
Collapse
|
19
|
Tomita U, Yamaguchi S, Sugimoto Y, Takamori S, Nagamune T. Poly(ethylene glycol)-Lipid-Conjugated Antibodies Enhance Dendritic Cell Phagocytosis of Apoptotic Cancer Cells. Pharmaceuticals (Basel) 2012; 5:405-16. [PMID: 24281554 PMCID: PMC3763647 DOI: 10.3390/ph5050405] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Revised: 03/27/2012] [Accepted: 04/17/2012] [Indexed: 11/25/2022] Open
Abstract
A simple method for attaching immunoglobulin G (IgG) on the cell surface was successfully developed for enhancing phagocytosis of apoptotic tumor cells (ATCs) by dendritic cells (DCs) ex vivo. By conjugating with a poly(ethylene glycol) (PEG)-lipid, named the biocompatible anchor for the membrane (BAM), arbitrary IgG could be incorporated into the cell membrane. In particular, when IgG-BAM conjugates were prepared at the optimal molar ratio of IgG to BAM (1 to 20), almost all cells were efficiently modified with IgG by treatment with IgG-BAM. This simple method was successfully applied to four types of mammalian cells. Furthermore, treatment of ATCs with the IgG-BAM conjugate increased the phagocytosis ratio of ATCs by DCs two-fold when compared to no treatment. This phagocytosis-enhancing effect was nearly identical to treatment with a tumor-specific IgG. Thus, without employing the tumor-specific IgG, which is difficult to obtain for any tumor cells and is expensive, the present method could opsonize ATC with the use of arbitrary IgG. The results strongly indicate that IgG-BAM treatment represents a promising method for opsonizing ATC with human serum IgG, and that this approach will lead to objective clinical responses in DC vaccines.
Collapse
Affiliation(s)
- Urara Tomita
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.
| | | | | | | | | |
Collapse
|
20
|
Dodson LF, Hawkins WG, Goedegebuure P. Potential targets for pancreatic cancer immunotherapeutics. Immunotherapy 2011; 3:517-37. [PMID: 21463193 DOI: 10.2217/imt.11.10] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Pancreatic adenocarcinoma is the fourth leading cause of cancer death with an overall 5-year survival of less than 5%. As there is ample evidence that pancreatic adenocarcinomas elicit antitumor immune responses, identification of pancreatic cancer-associated antigens has spurred the development of vaccination-based strategies for treatment. While promising results have been observed in animal tumor models, most clinical studies have found only limited success. As most trials were performed in patients with advanced pancreatic cancer, the contribution of immune suppressor mechanisms should be taken into account. In this article, we detail recent work in tumor antigen vaccination and the recently identified mechanisms of immune suppression in pancreatic cancer. We offer our perspective on how to increase the clinical efficacy of vaccines for pancreatic cancer.
Collapse
Affiliation(s)
- Lindzy F Dodson
- Washington University School of Medicine, Department of Surgery, Saint Louis, MO 63110, USA.
| | | | | |
Collapse
|
21
|
Abstract
INTRODUCTION The advent of anti-CD20 monoclonal antibody (mAb) rituximab heralded a new era in the treatment of non-Hodgkin's lymphoma leading to significant improvements in outcome for patients. This unprecedented success has changed the mindset of the clinical community and catalyzed the interest in the pharmaceutical industry to develop the next-generation of antibodies and antibody conjugates in cancer. AREAS COVERED There are an ever increasing number of newer generation anti-CD20 and rituximab 'bio-similars' undergoing early phase clinical development. In addition emerging novel therapies including antibody drug conjugates (brentuximab vedotin, SGN-35) and mAb against T-cell lymphomas antigens (e.g., zanolimumab) offer hope of improved outcome for other lymphomas. Bispecific T-cell-engaging antibodies and combination immunotherapy, also provide the promise of further improvements. Radiolabelled antibodies or radioimmunotherapy (RIT) has also demonstrated high clinical activity and two drugs namely 131I-tositumomab (Bexxar) and 90Y-ibritumomab (Zevalin) are licensed. EXPERT OPINION Despite the large numbers of new anti-CD20 mAb currently undergoing clinical testing, improving on clinical efficacy of rituximab is a substantial challenge. Further improvements in outcome for patients will require rigorous testing in well designed clinical trials alongside the translation of new insights into mechanism of mAb action that lead to improvements in clinical efficacy.
Collapse
Affiliation(s)
- Sam Mayes
- University of Manchester, Manchester Academic Health Science Centre, School of Cancer and Enabling Sciences, School of Medicine, Manchester, M20 4BX, UK
| | | | | |
Collapse
|
22
|
Bologna L, Gotti E, Manganini M, Rambaldi A, Intermesoli T, Introna M, Golay J. Mechanism of action of type II, glycoengineered, anti-CD20 monoclonal antibody GA101 in B-chronic lymphocytic leukemia whole blood assays in comparison with rituximab and alemtuzumab. THE JOURNAL OF IMMUNOLOGY 2011; 186:3762-9. [PMID: 21296976 DOI: 10.4049/jimmunol.1000303] [Citation(s) in RCA: 152] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
We analyzed in B-chronic lymphocytic leukemia (B-CLL) whole blood assays the activity of therapeutic mAbs alemtuzumab, rituximab, and type II glycoengineered anti-CD20 mAb GA101. Whole blood samples were treated with Abs, and death of CD19(+) B-CLL was measured by flow cytometry. Alemtuzumab efficiently lysed B-CLL targets with maximal lysis at 1-4 h (62%). In contrast, rituximab induced a more limited cell death (21%) that was maximal only at 24 h. GA101 killed B-CLL targets to a similar extent but more rapidly than rituximab, with 19.2 and 23.5% cell death at 4 and 24 h, respectively, compared with 7.9 and 21.4% for rituximab. Lysis by both rituximab and GA101 correlated directly with CD20 expression levels (r(2) = 0.88 and 0.85, respectively). Interestingly, lysis by all three Abs at high concentrations was mostly complement dependent, because it was blocked by the anti-C5 Ab eculizumab by 90% in the case of alemtuzumab and rituximab and by 64% in the case of GA101. Although GA101 caused homotypic adhesion, it induced only limited (3%) direct cell death of purified B-CLL cells. Both rituximab and GA101 showed the same efficiency in phagocytosis assays, but phagocytosis was not significant in whole blood due to excess Igs. Finally, GA101 at 1-100 μg/ml induced 2- to 3-fold more efficient NK cell degranulation than rituximab in isolated B-CLL or normal PBMCs. GA101, but not rituximab, also mediated significant NK cell degranulation in whole blood samples. Thus, complement and Ab-dependent cellular cytotoxicity are believed to be the major effector mechanisms of GA101 in whole blood assays.
Collapse
Affiliation(s)
- Luca Bologna
- Laboratory of Cellular Therapy G. Lanzani, Division of Hematology, Ospedali Riuniti, 24128 Bergamo, Italy
| | | | | | | | | | | | | |
Collapse
|
23
|
Immunotherapy for treating metastatic colorectal cancer. Surg Oncol 2011; 21:67-77. [PMID: 21292476 DOI: 10.1016/j.suronc.2010.10.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2010] [Revised: 10/10/2010] [Accepted: 10/14/2010] [Indexed: 01/17/2023]
Abstract
BACKGROUND Colorectal cancer remains one of the leading causes of death in the world. Surgery still remains the mainstay of treatment for primary and metastatic colorectal cancer. Immunotherapy used as an adjunct to surgery can play an important role in controlling the spread of tumour. METHODS The online databases PubMed, Medline, Scirus and Medscape Oncology were used to identify articles of relevance. Keywords included; "Immunotherapy", "Cellular Immunotherapy", "Metastatic Colorectal Cancer", "Monoclonal Antibody" "Tumour Vaccines" and "Adoptive Cell Therapy". The databases search was from the period of June 1995 until May 2010 inclusive. RESULTS Our understanding of tumour immunology has allowed the development of some successful therapies. Immunotherapy through the use of monoclonal antibodies is an effective adjunct to chemotherapy for metastatic colorectal cancer. Other modalities that are in the stages of development are cellular and conjugated vaccines. However, these vaccines are being experimented in advanced stages of colorectal tumours. CONCLUSION Colorectal cancer vaccines are being developed for advanced stages of colorectal tumour. However, their use as an early adjunct could potentially limit the spread of tumour or even result in cure. Further trials are required to ensure the safety and efficacy of cellular vaccines against colorectal tumours to allow their use on patients early in their disease presentation.
Collapse
|